Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Immunol Res ; 2023: 1884439, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36703865

RESUMO

Platelets have toll-like receptors (TLRs); however, their function in thrombosis or hemostasis under flow conditions is not fully known. Thrombin-inhibited anticoagulated whole blood was treated with various TLR agonists and then perfused over fibrillar collagen using microfluidic assay at venous wall shear rate (100 s-1). Platelet deposition was imaged with fluorescent anti-CD61. For perfusion of whole blood without TLR agonist addition, platelets rapidly accumulated on collagen and eventually occluded the microchannels. Interestingly, most of the tested TLR agonists (Pam3CKS4, MALP-2, polyinosinic-polycytidylic acid HMW, imiquimod, and CpG oligodeoxynucleotides) strongly reduced platelet deposition on collagen, while only the TLR4 agonist endotoxin lipopolysaccharide (LPS) enhanced deposition. Following 90 sec of deposition under flow of untreated blood, the addition of various TLR-7 agonists (imiquimod, vesatolimod, and GSK2245035) all caused immediate blockade of further platelet deposition. Since TLR signaling can activate nuclear factor-kappaB (NF-κB), the IKK-inhibitor (IKK inhibitor VII) and NF-κB inhibitor (Bay 11-7082) were tested. The IKK/NF-κB inhibitors strongly inhibited platelet deposition under flow. Furthermore, addition of Pam3CSK4 (TLR1/2 ligand), MALP-2 (TLR2/6 ligand), and Imquimod (TLR7 ligand) reduced phosphotidylserine (PS) exposure. Activation of TLR1/2, TLR2/6, TLR3, TLR7, and TLR9 in whole blood reduced platelet deposition under flow on collagen; however, LPS (major Gram negative bacterial pathogenic component) activation of LTR4 was clearly prothrombotic.


Assuntos
NF-kappa B , Receptor 2 Toll-Like , Humanos , Receptor 2 Toll-Like/agonistas , Receptor 7 Toll-Like , Receptor 1 Toll-Like , Ligantes , Lipopolissacarídeos/farmacologia , Imiquimode , Receptores Toll-Like/agonistas , Colágeno
2.
Thromb Res ; 218: 112-129, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36037547

RESUMO

The collagen receptor glycoprotein VI (GPVI) drives strong platelet activation, however its role at later stages of clotting remains less clear. Controlled timing of addition of anti-human GPVI Fab (clone E12) with microfluidic venous whole blood flow over collagen (± lipidated tissue factor, TF) produced distinct effects on platelets, fibrin, P-selectin exposure, and phosphatidylserine (PS) exposure. On collagen alone, Fab present initially potently reduced platelet deposition on collagen, while Fab added 90 s after initial platelet deposition, stopped subsequent platelet accumulation (despite the absence of fibrin). With thrombin generation via TF, Fab added at either t = 0 or 90 s had no effect on platelet deposition. However, Fab added initially, but not at 90-s, blocked fibrin formation. Gly-Pro-Arg-Pro ablated fibrin formation without effect on platelet accumulation (regardless of Fab added at t = 0 or 90 s), indicating thrombin signaling can suffice over GPVI signaling. Still, Fab moderately reduced P-selectin exposure with thrombin present and fibrin absent. On collagen/TF, Fab present initially ablated PS exposure, but had no effect when added 30 to 90-s later. The thrombin generated via PS exposure had an important role in driving platelet deposition in the presence of Fab, since inhibition of PS via annexin V binding in the presence of Fab significantly inhibited platelet deposition. We conclude GPVI signaling in the first platelet layer on collagen dictates thrombin and fibrin production, but the role of GPVI at subsequent times after formation of the first monolayer is obscured by thrombin-induced signaling.


Assuntos
Trombina , Tromboplastina , Anexina A5 , Colágeno/metabolismo , Colágeno/farmacologia , Fibrina/metabolismo , Humanos , Microfluídica , Selectina-P/metabolismo , Fosfatidilserinas , Glicoproteínas da Membrana de Plaquetas , Receptores de Colágeno/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo
3.
Biochim Biophys Acta Gen Subj ; 1865(2): 129805, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33276061

RESUMO

BACKGROUND: Thrombin activates fibrinogen and binds the fibrin E-domain (Kd ~ 2.8 µM) and the splice variant γ'-domain (Kd ~ 0.1 µM). We investigated if the loading of D-Phe-Pro-Arg-chloromethylketone inhibited thrombin (PPACK-thrombin) onto fibrin could enhance fibrin stability. METHODS: A 384-well plate thermal shift assay (TSA) with SYPRO-orange provided melting temperatures (Tm) of thrombin, PPACK-thrombin, fibrinogen, fibrin monomer, and fibrin. RESULTS: Large increases in Tm indicated that calcium led to protein stabilization (0 vs. 2 mM Ca2+) for fibrinogen (54.0 vs. 62.3 °C) and fibrin (62.3 vs. 72.2 °C). Additionally, active site inhibition with PPACK dramatically increased the Tm of thrombin (58.3 vs. 78.3 °C). Treatment of fibrinogen with fibrin polymerization inhibitor GPRP increased fibrinogen stability by ΔTm = 9.3 °C, similar to the ΔTm when fibrinogen was converted to fibrin monomer (ΔTm = 8.8 °C) or to fibrin (ΔTm = 10.4 °C). Addition of PPACK-thrombin at high 5:1 M ratio to fibrin(ogen) had little effect on fibrin(ogen) Tm values, indicating that thrombin binding does not detectably stabilize fibrin via a putative bivalent E-domain to γ'-domain interaction. CONCLUSIONS: TSA was a sensitive assay of protein stability and detected: (1) the effects of calcium-stabilization, (2) thrombin active site labeling, (3) fibrinogen conversion to fibrin, and (4) GPRP induced changes in fibrinogen stability being essentially equivalent to that of fibrin monomer or polymerized fibrin. SIGNIFICANCE: The low volume, high throughput assay has potential for use in understanding interactions with rare or mutant fibrin(ogen) variants.


Assuntos
Produtos de Degradação da Fibrina e do Fibrinogênio/química , Fibrina/química , Fibrinogênio/química , Trombina/química , Clorometilcetonas de Aminoácidos/química , Fibrinogênios Anormais/química , Humanos , Oligopeptídeos/química , Estabilidade Proteica , Temperatura de Transição
4.
J Thromb Haemost ; 16(5): 973-983, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29488682

RESUMO

Essentials Platelet packing density in a hemostatic plug limits molecular movement to diffusion. A diffusion-dependent steep thrombin gradient forms radiating outwards from the injury site. Clot retraction affects the steepness of the gradient by increasing platelet packing density. Together, these effects promote hemostatic plug core formation and inhibit unnecessary growth. SUMMARY: Background Hemostasis studies performed in vivo have shown that hemostatic plugs formed after penetrating injuries are characterized by a core of highly activated, densely packed platelets near the injury site, covered by a shell of less activated and loosely packed platelets. Thrombin production occurs near the injury site, further activating platelets and starting the process of platelet mass retraction. Tightening of interplatelet gaps may then prevent the escape and exchange of solutes. Objectives To reconstruct the hemostatic plug macro- and micro-architecture and examine how platelet mass contraction regulates solute transport and solute concentration in the gaps between platelets. Methods Our approach consisted of three parts. First, platelet aggregates formed in vitro under flow were analyzed using scanning electron microscopy to extract data on porosity and gap size distribution. Second, a three-dimensional (3-D) model was constructed with features matching the platelet aggregates formed in vitro. Finally, the 3-D model was integrated with volume and morphology measurements of hemostatic plugs formed in vivo to determine how solutes move within the platelet plug microenvironment. Results The results show that the hemostatic mass is characterized by extremely narrow gaps, porosity values even smaller than previously estimated and stagnant plasma velocity. Importantly, the concentration of a chemical species released within the platelet mass increases as the gaps between platelets shrink. Conclusions Platelet mass retraction provides a physical mechanism to establish steep chemical concentration gradients that determine the extent of platelet activation and account for the core-and-shell architecture observed in vivo.


Assuntos
Músculos Abdominais/irrigação sanguínea , Arteríolas/lesões , Plaquetas/metabolismo , Hemostasia , Agregação Plaquetária , Trombina/metabolismo , Trombose/sangue , Lesões do Sistema Vascular/sangue , Animais , Arteríolas/patologia , Arteríolas/fisiopatologia , Velocidade do Fluxo Sanguíneo , Plaquetas/patologia , Retração do Coágulo , Simulação por Computador , Difusão , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Microcirculação , Modelos Biológicos , Porosidade , Trombose/patologia , Trombose/fisiopatologia , Fatores de Tempo , Lesões do Sistema Vascular/patologia , Lesões do Sistema Vascular/fisiopatologia
5.
J Thromb Haemost ; 16(2): 316-329, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29156107

RESUMO

Essentials Neutrophil extracellular traps (NETs) are generated during thrombosis and sepsis. The effect of hemodynamics on NETosis during sterile thrombosis was studied using microfluidics. Pressure gradients > 70 mmHg per mm-clot across sterile occlusions drive shear-induced NETosis. High interstitial hemodynamic forces trigger rapid NET release. SUMMARY: Background Neutrophil extracellular traps (NETs) are released when neutrophils encounter infectious pathogens, especially during sepsis. Additionally, NETosis occurs during venous and arterial thrombosis, disseminated intravascular coagulation, and trauma. Objective To determine whether hemodynamic forces trigger NETosis during sterile thrombosis. Methods NETs were imaged with Sytox Green during microfluidic perfusion of activated factor XII-inhibited or thrombin-inhibited human whole blood over fibrillar collagen (with or without tissue factor). Results For perfusions at initial inlet venous or arterial wall shear rates (100 s-1 or 1000 s-1 ), platelets rapidly accumulated and occluded microchannels with subsequent neutrophil infiltration under either flow condition; however, NETosis was detected only in the arterial condition. The level of shear-induced NETs (SINs) at 30 min was > 150-fold higher in the arterial condition in the absence of thrombin and > 80-fold greater in the presence of thrombin than the level in the venous condition. With or without thrombin, venous perfusion for 15 min generated no NETs, but an abrupt shift-up to arterial perfusion triggered NETosis within 2 min, NETs eventually reaching levels 15 min later that were 60-fold greater than that in microchannels without perfusion shift-up. SINs contained citrullinated histone H3 and myeloperoxidase, and were DNase-sensitive, but were not blocked by inhibitors of platelet-neutrophil adhesion, high-mobility group protein box 1-receptor for advanced glycation end products interaction, cyclooxygenase, ATP/ADP, or peptidylarginine deiminase 4. For measured pressure gradients exceeding 70 mmHg per millimeter of clot across NET-generating occlusions to drive interstitial flow, the calculated fluid shear stress on neutrophils exceeded the known lytic value of 150 dyne cm-2 . Conclusions High interstitial hemodynamic forces can drive physically entrapped neutrophils to rapidly release NETs during sterile occlusive thrombosis.


Assuntos
Armadilhas Extracelulares/metabolismo , Hemodinâmica , Ativação de Neutrófilo , Neutrófilos/metabolismo , Trombose/sangue , Trombose/fisiopatologia , Plaquetas/metabolismo , Pressão Sanguínea , Citrulinação , Simulação por Computador , Histonas/sangue , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Peroxidase/sangue , Transdução de Sinais , Estresse Mecânico
6.
J Thromb Haemost ; 15(12): 2396-2407, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28981200

RESUMO

Essentials Collagen and thrombin when used simultaneously generate highly activated platelets. The effect of thrombin stimulation on subsequent glycoprotein VI (GPVI) function was observed. Soluble fibrin, but not protease activated receptor (PAR) activation, prevented GPVI activation. Circulating soluble fibrin in coagulopathic blood may cause an acquired GPVI signaling defect. SUMMARY: Background In coagulopathic blood, circulating thrombin may drive platelet dysfunction. Methods/Results Using calcium dye-loaded platelets, the effect of thrombin exposure and soluble fibrin generation on subsequent platelet GPVI function was investigated. Exposure of apixaban-treated platelet-rich plasma (12% PRP) to thrombin (1-10 nm), but not ADP or thromboxane mimetic U46619 exposure, dramatically blocked subsequent GPVI activation by convulxin, collagen-related peptide or fibrillar collagen. Consistent with soluble fibrin multimerizing and binding GPVI, the onset of convulxin insensitivity required 200-500 s of thrombin exposure, was not mimicked by exposure to PAR-1/4 activating peptides, was not observed with washed platelets, and was blocked by fibrin polymerization inhibitor (GPRP) or factor XIIIa inhibitor (T101). PAR-1 signaling through Gαq was not required because vorapaxar blocked thrombin-induced calcium mobilization but had no effect on the ability of thrombin to impair GPVI-signaling. Convulxin insensitivity was unaffected by the metalloprotease inhibitor GM6001 or the αIIb ß3 antagonist GR144053, indicating negligible roles for GPVI shedding or αIIb ß3 binding of fibrin. Thrombin treatment of washed platelets resuspended in purified fibrinogen also produced convulxin insensitivity that was prevented by GPRP. Exposure of apixaban/PPACK-treated whole blood to thrombin-treated fibrinogen resulted in > 50% decrease in platelet deposition in a collagen microfluidic assay that required soluble fibrin assembly. Conclusions Conversion of only 1% plasma fibrinogen in coagulopathic blood would generate 90 nm soluble fibrin, far exceeding ~1 nmGPVI in blood. Soluble fibrin, rather than thrombin-induced platelet activation throuh PAR-1 and PAR-4, downregulated GPVI-signaling in response to stimuli, and may lead to subsequent hypofunction of endogenous or transfused platelets.


Assuntos
Transtornos da Coagulação Sanguínea/sangue , Fibrina/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/sangue , Difosfato de Adenosina/farmacologia , Plaquetas/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Colágeno/metabolismo , Venenos de Crotalídeos/farmacologia , Humanos , Técnicas In Vitro , Lectinas Tipo C , Oligopeptídeos/sangue , Ativação Plaquetária/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Trombina/metabolismo
7.
Haemophilia ; 23(5): 759-768, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28475272

RESUMO

INTRODUCTION: Factor VIII (FVIII) or factor IX (FIX)-deficient haemophilic patients display deficits in platelet and fibrin deposition under flow detectable in microfluidics. Compared to fibrin generation, decreased platelet deposition in haemophilic blood flow is more easily rescued with recombinant factor VIIa (rFVIIa), whereas rFVIIa requires FXIIa participation to generate fibrin when tissue factor (TF) is absent. AIMS: Perfusion of haemophilic whole blood (WB) over collagen/TF surfaces was used to determine whether rFVIIa/TF was sufficient to bypass poor FIXa/FVIIIa function in blood from patients with haemophilia A and B. METHODS: Whole blood treated with high-dose corn trypsin inhibitor (40 µg mL-1 ) from seven healthy donors and 10 patients was perfused over fibrillar collagen presenting low or high TF (TFlow or TFhigh ) at wall shear rate of 100 s-1 . RESULTS: With WB from healthy controls, platelet deposition and fibrin accumulation increased as TF increased. Factor-deficient WB (1-3% of normal) displayed striking deficits in platelet deposition and fibrin formation at either TFlow or TFhigh . In contrast, mildly factor-deficient WB (14-32%) supported fibrin formation under flow on TFhigh /collagen. With either TFlow or TFhigh , exogenously added rFVIIa (20 nm) increased platelet deposition and fibrin accumulation in WB from factor-deficient patients (1-3% of normal) to levels commensurate with untreated healthy WB. CONCLUSION: The absence of FIXa/FVIIIa in patients with severe haemophilia results in deficits in fibrin formation that cannot be rescued by wall-derived TF ex vivo. The effects of rFVIIa on platelet adhesion and rFVIIa/TF can act together to reinforce thrombin generation, platelet deposition and fibrin formation under flow.


Assuntos
Colágeno/administração & dosagem , Fator VIIa/administração & dosagem , Fibrina/biossíntese , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Hemofilia B/sangue , Hemofilia B/tratamento farmacológico , Tromboplastina/administração & dosagem , Coagulação Sanguínea/efeitos dos fármacos , Testes de Coagulação Sanguínea , Plaquetas/metabolismo , Colágeno/metabolismo , Hemofilia A/diagnóstico , Hemofilia B/diagnóstico , Humanos , Modelos Biológicos , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Ligação Proteica , Proteínas Recombinantes/administração & dosagem , Transdução de Sinais , Tromboplastina/metabolismo
8.
J Thromb Haemost ; 15(3): 526-537, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27992950

RESUMO

Essentials Methods were developed to image the hemostatic response in mouse femoral arteries in real time. Penetrating injuries produced thrombi consisting primarily of platelets. Similar to arterioles, a core-shell architecture of platelet activation occurs in the femoral artery. Differences from arterioles included slower platelet activation and reduced thrombin dependence. SUMMARY: Background Intravital studies performed in the mouse microcirculation show that hemostatic thrombi formed after penetrating injuries develop a characteristic architecture in which a core of fully activated, densely packed platelets is overlaid with a shell of less activated platelets. Objective Large differences in hemodynamics and vessel wall biology distinguish arteries from arterioles. Here we asked whether these differences affect the hemostatic response and alter the impact of anticoagulants and antiplatelet agents. Methods Approaches previously developed for intravital imaging in the mouse microcirculation were adapted to the femoral artery, enabling real-time fluorescence imaging despite the markedly thicker vessel wall. Results Arterial thrombi initiated by penetrating injuries developed the core-and-shell architecture previously observed in the microcirculation. However, although platelet accumulation was greater in arterial thrombi, the kinetics of platelet activation were slower. Inhibiting platelet ADP P2Y12 receptors destabilized the shell and reduced thrombus size without affecting the core. Inhibiting thrombin with hirudin suppressed fibrin accumulation, but had little impact on thrombus size. Removing the platelet collagen receptor, glycoprotein VI, had no effect. Conclusions These results (i) demonstrate the feasibility of performing high-speed fluorescence imaging in larger vessels and (ii) highlight differences as well as similarities in the hemostatic response in the macro- and microcirculation. Similarities include the overall core-and-shell architecture. Differences include the slower kinetics of platelet activation and a smaller contribution from thrombin, which may be due in part to the greater thickness of the arterial wall and the correspondingly greater separation of tissue factor from the vessel lumen.


Assuntos
Artéria Femoral/diagnóstico por imagem , Hemostasia , Microcirculação , Ferimentos Penetrantes/terapia , Difosfato de Adenosina/metabolismo , Animais , Anticoagulantes/farmacologia , Arteríolas/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Artéria Femoral/lesões , Fibrina/metabolismo , Hemodinâmica , Microscopia Intravital , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Transdução de Sinais , Trombina/antagonistas & inibidores , Trombina/metabolismo , Tromboplastina/metabolismo , Trombose/diagnóstico por imagem , Trombose/tratamento farmacológico
9.
J Thromb Haemost ; 14(5): 906-17, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26848552

RESUMO

The biophysics of blood flow can dictate the function of molecules and cells in the vasculature with consequent effects on hemostasis, thrombosis, embolism, and fibrinolysis. Flow and transport dynamics are distinct for (i) hemostasis vs. thrombosis and (ii) venous vs. arterial episodes. Intraclot transport changes dramatically the moment hemostasis is achieved or the moment a thrombus becomes fully occlusive. With platelet concentrations that are 50- to 200-fold greater than platelet-rich plasma, clots formed under flow have a different composition and structure compared with blood clotted statically in a tube. The platelet-rich, core/shell architecture is a prominent feature of self-limiting hemostatic clots formed under flow. Importantly, a critical threshold concentration of surface tissue factor is required for fibrin generation under flow. Once initiated by wall-derived tissue factor, thrombin generation and its spatial propagation within a clot can be modulated by γ'-fibrinogen incorporated into fibrin, engageability of activated factor (FIXa)/activated FVIIIa tenase within the clot, platelet-derived polyphosphate, transclot permeation, and reduction of porosity via platelet retraction. Fibrin imparts tremendous strength to a thrombus to resist embolism up to wall shear stresses of 2400 dyne cm(-2) . Extreme flows, as found in severe vessel stenosis or in mechanical assist devices, can cause von Willebrand factor self-association into massive fibers along with shear-induced platelet activation. Pathological von Willebrand factor fibers are A Disintegrin And Metalloprotease with ThromboSpondin-1 domain 13 resistant but are a substrate for fibrin generation due to FXIIa capture. Recently, microfluidic technologies have enhanced the ability to interrogate blood in the context of stenotic flows, acquired von Willebrand disease, hemophilia, traumatic bleeding, and drug action.


Assuntos
Hemostasia , Reologia , Trombose/fisiopatologia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo , Plaquetas/efeitos dos fármacos , Constrição Patológica , Difusão , Fator IXa/química , Fator VIIIa/química , Fibrina/química , Fibrinólise , Humanos , Camundongos , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Plasma Rico em Plaquetas/metabolismo , Polifosfatos/química , Porosidade , Estresse Mecânico , Trombina/farmacologia , Tromboplastina/farmacologia , Fator de von Willebrand/química
10.
J Thromb Haemost ; 14(5): 1070-81, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26725377

RESUMO

UNLABELLED: Essentials Protein disulfide isomerases may have an essential role in thrombus formation. A platelet-binding sensor (PDI-sAb) was developed to detect thiol reductase activity under flow. Primary human platelet adhesion to collagen at 200 s(-1) was correlated with the PDI-sAb signal. Detected thiol reductase activity was localized in the core of growing thrombi at the site of injury in mice. SUMMARY: Background Protein disulfide isomerases (PDIs) may regulate thrombus formation in vivo, although the sources and targets of PDIs are not fully understood. Methods and results Using click chemistry to link anti-CD61 and a C-terminal azido disulfide-linked peptide construct with a quenched reporter, we developed a fluorogenic platelet-targeting antibody (PDI-sAb) for thiol reductase activity detection in whole blood under flow conditions. PDI-sAb was highly responsive to various exogenous reducing agents (dithiothreitol, glutathione and recombinant PDI) and detected thiol reductase activity on P-selectin/phosphatidylserine-positive platelets activated with convulxin/PAR1 agonist peptide, a signal partially blocked by PDI inhibitors and antibody. In a microfluidic thrombosis model using 4 µg mL(-1) corn trypsin inhibitor-treated human blood perfused over collagen (wall shear rate = 100 s(-1) ), the PDI-sAb signal increased mostly over the first 200 s, whereas platelets continually accumulated for over 500 s, indicating that primary adhesion to collagen, but not secondary aggregation, was correlated with the PDI-sAb signal. Rutin and the PDI blocking antibody RL90 reduced platelet adhesion and the PDI-sAb signal only when thrombin production was inhibited with PPACK, suggesting limited effects of platelet thiol isomerase activity on platelet aggregation on collagen in the presence of thrombin. With anti-mouse CD41 PDI-sAb used in an arteriolar laser injury model, thiol reductase activity was localized in the core of growing thrombi where platelets displayed P-selectin and were in close proximity to disrupted endothelium. Conclusion PDI-sAb is a sensitive and real-time reporter of platelet- and vascular-derived disulfide reduction that targets clots as they form under flow to reveal spatial gradients.


Assuntos
Plaquetas/enzimologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Compostos de Sulfidrila/química , Animais , Anticorpos/química , Velocidade do Fluxo Sanguíneo , Transtornos Plaquetários/metabolismo , Fibrina/química , Hemodinâmica , Humanos , Integrina beta3/química , Microscopia Intravital , Camundongos , Microfluídica , Peptídeos/química , Adesividade Plaquetária , Trombina/química , Trombose/metabolismo
11.
J Thromb Haemost ; 13(9): 1699-708, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26178390

RESUMO

BACKGROUND: Under severe stenotic conditions, von Willebrand factor (VWF) multimerizes into large insoluble fibers at pathological shear rates. OBJECTIVE: Evaluate the mechanics and biology of VWF fibers without the confounding effects of endothelium or collagen. METHODS: Within a micropost-impingement microfluidic device, > 100-µm long VWF fibers multimerized on the post within 10 min using EDTA-treated platelet-free plasma (PFP) perfused at wall shear rates > 5000 s(-1) . RESULTS: von Willebrand factor fiber thickness increased to > 10 µm as a result of increasing the shear rate to 10,000 s(-1) . In a stress-strain test, fibrous VWF had an elastic modulus of ~50 MPa. The insoluble VWF fibers were non-amyloid because they rapidly dissolved in trypsin, plasmin or 2% SDS, but were resistant to 50 nm ADAMTS13 or 100 nm tissue plasminogen activator in plasma. Following fiber formation, perfusion of low corn trypsin inhibitor (CTI)-treated (4 µg mL(-1) ), recalcified citrated plasma at 1500 s(-1) caused fibrin formation on the VWF fibers, a result not observed with purified type 1 collagen or a naked micropost. During VWF fiber formation, contact pathway factors accumulated on VWF because the use of EDTA/D-Phe-Pro-Arg chloromethylketone (PPACK)/apixaban/high CTI-treated PFP during VWF fiber formation prevented the subsequent fibrin production from low-CTI, recalcified citrated PFP. VWF fibers displayed FXIIa-immunostaining. When PPACK-inhibited whole blood was perfused over VWF fibers, platelets rolled and arrested on the surface of VWF, but only displayed P-selectin if prevailing shear rates were pathological. Platelet arrest on VWF fibers was blocked with αIIb ß3 antagonist GR144053. CONCLUSIONS: We report VWF fiber-contact pathway crosstalk and mechanisms of thrombolytic resistance in hemodynamic settings of myocardial infarction.


Assuntos
Proteínas ADAM/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Hemorreologia , Ativação Plaquetária/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/farmacologia , Fator de von Willebrand/química , Proteína ADAMTS13 , Biopolímeros , Módulo de Elasticidade , Fibrinolisina/farmacologia , Humanos , Técnicas In Vitro , Dispositivos Lab-On-A-Chip , Selectina-P/sangue , Piperazinas/farmacologia , Piperidinas/farmacologia , Adesividade Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Subunidades Proteicas , Solubilidade , Fator de von Willebrand/fisiologia , Fator de von Willebrand/ultraestrutura
12.
Haemophilia ; 21(2): 266-274, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25311576

RESUMO

In prior microfluidic studies with haemophilic blood perfused over collagen, we found that a severe deficiency (<1% factor level) reduced platelet and fibrin deposition, while a moderate deficiency (1-5%) only reduced fibrin deposition. We investigated: (i) the differential effect of rFVIIa (0.04-20 nm) on platelet and fibrin deposition, and (ii) the contribution of the contact pathway to rFVIIa-induced haemophilic blood clotting. Haemophilic or healthy blood with low and high corn trypsin inhibitor (CTI, 4 or 40 µg mL(-1) ) was perfused over collagen at an initial venous wall shear rate of 100 s(-1) . At 100 s(-1) wall shear rate, where FXIIa leads to thrombin production without added tissue factor, FXI-deficient blood (3%) or severely FVIII-deficient blood (<1%) produced no fibrin at either CTI level. Whereas rFVIIa potently enhanced platelet deposition, fibrin generation was not rescued. Distinct from the high CTI condition, engagement of the contact pathway (low CTI) in moderately FVIII-deficient (3%) or moderately FIX-deficient blood (5%) resulted in enhanced platelet and fibrin deposition following 4 nm rFVIIa supplementation. In mildly FVIII-deficient blood (15%) at <24 h since haemostatic therapy, rFVIIa enhanced both platelet and fibrin generation in either CTI condition although fibrin was produced more quickly and abundantly in low CTI. For tissue factor-free conditions of severe haemophilic blood clotting, we conclude that rFVIIa reliably generates low levels of 'signaling' thrombin sufficient to enhance platelet deposition on collagen, but is insufficient to drive fibrin polymerization unless potentiated by the contact pathway.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Fator VIIa/farmacologia , Fibrina/metabolismo , Hemofilia A/sangue , Hemofilia B/sangue , Testes de Coagulação Sanguínea , Estudos de Casos e Controles , Colágeno/metabolismo , Deficiência do Fator XI/sangue , Humanos , Técnicas Analíticas Microfluídicas , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Ligação Proteica , Proteínas Recombinantes/farmacologia
13.
J Thromb Haemost ; 12(2): 147-58, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24261634

RESUMO

BACKGROUND: Coagulation factor deficiencies create a range of bleeding phenotypes. Microfluidic devices offer controlled hemodynamics and defined procoagulant triggers for measurement of clotting under flow. OBJECTIVES: We tested a flow assay of contact pathway-triggered clotting to quantify platelet and fibrin deposition distal of dysfunctional thrombin production. Microfluidic metrics were then compared with PTT or % factor activity assays. METHODS: Whole blood (WB) treated with low level corn trypsin inhibitor (4 µg mL⁻¹) from nine healthy donors and 27 patients (deficient in factor [F] VIII, 19 patients; FIX, one patient; FXI, one patient; VWF, six patients) was perfused over fibrillar collagen at wall shear rate = 100 s⁻¹. RESULTS: Using healthy WB, platelets deposited within 30 s, while fibrin appeared within 6 min. Compared with healthy controls, WB from patients displayed a 50% reduction in platelet deposition only at < 1% factor activity. In contrast, striking defects in fibrin deposition occurred for patients with < 13% factor activity (or PTT > 40 s). Full occlusion of the 60-µm high channel was completely absent over the 15-min test in patients with < 1% factor activity, while an intermediate defect was present in patients with > 1% factor. CONCLUSION: Spontaneous bleeding in patients with < 1% factor activity may be linked to deficits in both platelet and fibrin deposition, a risk known to be mitigated when factor levels are raised to > 1% activity (PTT of ~40-60 s), a level that does not necessarily rescue fibrin formation under flow.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fibrina/metabolismo , Hemofilia A/fisiopatologia , Microfluídica/métodos , Humanos
14.
J Thromb Haemost ; 11 Suppl 1: 224-32, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23809126

RESUMO

Accurate computer simulation of blood function can inform drug target selection, patient-specific dosing, clinical trial design, biomedical device design, as well as the scoring of patient-specific disease risk and severity. These large-scale simulations rely on hundreds of independently measured physical parameters and kinetic rate constants. However, the models can be validated against large-scale, patient-specific laboratory measurements. By validation with high-dimensional data, modeling becomes a powerful tool to predict clinically complex scenarios. Currently, it is possible to accurately predict the clotting rate of plasma or blood in a tube as it is activated with a dose of tissue factor, even as numerous coagulation factors are altered by exogenous attenuation or potentiation. Similarly, the dynamics of platelet activation, as indicated by calcium mobilization or inside-out signaling, can now be numerically simulated with accuracy in cases where platelets are exposed to combinations of agonists. Multiscale models have emerged to combine platelet function and coagulation kinetics into complete physics-based descriptions of thrombosis under flow. Blood flow controls platelet fluxes, delivery and removal of coagulation factors, adhesive bonding, and von Willebrand factor conformation. The field of blood systems biology has now reached a stage that anticipates the inclusion of contact, complement, and fibrinolytic pathways along with models of neutrophil and endothelial activation. Along with '-omics' data sets, such advanced models seek to predict the multifactorial range of healthy responses and diverse bleeding and clotting scenarios, ultimately to understand and improve patient outcomes.


Assuntos
Coagulação Sanguínea , Biologia de Sistemas , Plaquetas/fisiologia , Humanos , Modelos Biológicos , Transdução de Sinais
15.
J Thromb Haemost ; 10(11): 2344-53, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22978514

RESUMO

BACKGROUND: Thrombin undergoes convective and diffusive transport, making it difficult to visualize during thrombosis. We developed the first sensor capable of revealing inner clot thrombin dynamics. METHODS AND RESULTS: An N-terminal-azido thrombin-sensitive fluorescent peptide (ThS-P) with a thrombin-releasable quencher was linked to anti-CD41 using click chemistry to generate a thrombin-sensitive platelet binding sensor (ThS-Ab). Rapid thrombin cleavage of ThS-P (K(m) = 40.3 µm, k(cat) = 1.5 s(-1) ) allowed thrombin monitoring by ThS-P or ThS-Ab in blood treated with 2-25 pm tissue factor (TF). Individual platelets had > 20-fold more ThS-Ab fluorescence after clotting. In a microfluidic assay of whole blood perfusion over collagen ± linked TF (wall shear rate = 100 s(-1) ), ThS-Ab fluorescence increased between 90 and 450 s for 0.1-1 molecule-TF µm(-2) and co-localized with platelets near fibrin. Without TF, neither thrombin nor fibrin was detected on the platelet deposits by 450 s. Using a microfluidic device to control the pressure drop across a thrombus forming on a porous collagen/TF plug (521 s(-1) ), thrombin and fibrin were detected at the thrombus-collagen interface at a zero pressure drop, whereas 80% less thrombin was detected at 3200 Pa in concert with fibrin polymerizing within the collagen. With anti-mouse CD41 ThS-Ab deployed in a mouse laser injury model, the highest levels of thrombin arose between 40 and 160 s nearest the injury site where fibrin co-localized and where the thrombus was most mechanically stable. CONCLUSION: ThS-Ab reveals thrombin locality, which depends on surface TF, flow and intrathrombus pressure gradients.


Assuntos
Plaquetas/citologia , Trombina/química , Animais , Anticorpos/química , Coagulação Sanguínea , Colágeno/química , Fibrina/química , Hemostasia , Humanos , Cinética , Lasers , Camundongos , Técnicas Analíticas Microfluídicas , Microfluídica , Peptídeos/química , Glicoproteína IIb da Membrana de Plaquetas/química , Pressão , Transporte Proteico , Tromboplastina/química , Trombose/metabolismo , Fatores de Tempo
16.
Ann Biomed Eng ; 40(11): 2355-64, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22460075

RESUMO

Blood clotting under hemodynamic conditions involves numerous multiscale interactions from the molecular scale to macroscopic vessel and systemic circulation scales. Transmission of shear forces to platelet receptors such as GPIbα, P-selectin, α(2)ß(1), and α(2b)ß(3) controls adhesion dynamics. These forces also drive membrane tether formation, cellular deformation, and mechanosignaling in blood cells. Blood flow results in red blood cell (RBC) drift towards the center of the vessel along with a near-wall plasma layer enriched with platelets. RBC motions also dramatically enhance platelet dispersion. Trajectories of individual platelets near a thrombotic deposit dictate capture-activation-arrest dynamics as these newly arriving platelets are exposed to chemical gradients of ADP, thromboxane, and thrombin within a micron-scale boundary layer formed around the deposit. If shear forces are sufficiently elevated (>50 dyne/cm(2)), the largest polymers of von Willebrand Factor may elongate with concomitant shear-induced platelet activation. Finally, thrombin generation enhances platelet recruitment and clot strength via fibrin polymerization. By combination of coarse-graining, continuum, and stochastic algorithms, the numerical simulation of the growth rate, composition, and occlusive/embolic potential of a thrombus now spans multiscale phenomena. These simulations accommodate particular flow geometries, blood phenotype, pharmacological regimen, and reactive surfaces to help predict disease risk or response to therapy.


Assuntos
Modelos Biológicos , Trombose/fisiopatologia , Circulação Sanguínea , Coagulação Sanguínea/fisiologia , Plaquetas/fisiologia , Humanos , Biologia de Sistemas
17.
Biorheology ; 48(1): 49-64, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21515936

RESUMO

Using microfluidic assays at a 100 s⁻¹ wall shear rate, we examined the effects of ethanol on cholesterol-loaded neutrophils with respect to: (1) collision efficiency and membrane tethering to P-selectin-coated microbeads, (2) rolling on P-selectin-coated surfaces, and (3) primary and secondary interactions with neutrophils pre-adhered to intercellular adhesion molecule-1 (ICAM-1). Using methyl-ß-cyclodextrin:cholesterol complexes, membrane cholesterol was increased over control by 4.6-fold (no ethanol), 3.6-fold (0.3% ethanol pre-loading), and 1.6-fold (0.3% ethanol post-loading). These treatments did not alter CD11b expression; however, PSGL-1 and L-selectin were lowered by cholesterol enrichment (±ethanol). Cholesterol enrichment enhanced microbead collision efficiency, which was abrogated by ethanol. Ethanol had no effect on elevation of tethering fraction by cholesterol enrichment. Incubation of cholesterol-loaded neutrophils with ethanol resulted in significantly longer membrane tethers, due to tether lifetime enhancement. On P-selectin-coated surfaces, cholesterol-enriched neutrophils exposed to ethanol rolled faster and with more variability than cholesterol-enriched neutrophils. Ethanol reduced homotypic collision efficiency of cholesterol-loaded neutrophils without effect on tethering fraction or secondary collision efficiency. Tether length during cholesterol-loaded neutrophil homotypic collisions was enhanced by ethanol, in part due to increased L-selectin/PSGL-1 bond tether lifetime. Overall, ethanol attenuated cholesterol-induced adhesion increases while increasing membrane fluidity as indicated by tether length.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Etanol/farmacologia , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Fenômenos Mecânicos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Adulto , Fenômenos Biomecânicos , Adesão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Colesterol/farmacologia , Humanos , Microesferas , Neutrófilos/metabolismo , Selectina-P/metabolismo
18.
Ann Biomed Eng ; 39(2): 922-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20949319

RESUMO

In a focal injury model, platelets adhere and activate under flow on a collagen-coated surface, creating a field of individual platelet aggregates. These aggregates exhibit distinct structural characteristics that are linked to the local flow conditions. By combining image analysis techniques and epifluorescence microscopy, we developed a robust strategy for quantifying the characteristic instantaneous width and length of a growing platelet deposit. We have confirmed the technique using model images consisting of ellipsoid objects and quantified the shear rate-dependent nature of aggregate morphology. Venous wall shear rate conditions (100 s(-1)) generated small, circular platelet deposits, whereas elevated arterial shear rates (500 and 1000 s(-1)) generated platelet masses elongated twofold in the direction of flow. At 2000 s(-1), an important regime for von Willebrand Factor (vWF)-mediated recruitment, we observed sporadic platelet capture events on collagen that led to rapidly growing deposits. Furthermore, inter-donor differences were investigated with respect to aggregate growth rate. After perfusion at elevated shear rates (1000 s(-1)) for 5 min, we identified a twofold increase in aggregate size (81.5 ± 24.6 µm; p < 0.1) and a threefold increase in growth rate parallel to the flow (0.40 ± 0.09 µm/s; p < 0.01) for an individual donor. Suspecting a role for vWF, we found that this donor had a twofold increase in soluble vWF relative to the other donors and pooled plasma. Microfluidic devices in combination with automated morphology analysis offer new tools for characterizing clot development under flow.


Assuntos
Plaquetas/citologia , Plaquetas/fisiologia , Colágeno/metabolismo , Mecanotransdução Celular/fisiologia , Agregação Plaquetária/fisiologia , Velocidade do Fluxo Sanguíneo , Tamanho Celular , Células Cultivadas , Humanos
19.
Integr Biol (Camb) ; 2(4): 183-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20473398

RESUMO

Determination of the patient-specific response to antiplatelet agents facilitates proper dosing for both acute and chronic prophylaxis. "Closed" systems (with or without flow) may fail to predict pharmacological potency in situations where platelets rapidly accumulate under flow conditions at a site of thrombosis ("Open" systems). Using an 8-channel microfluidic flow assay of human whole blood with corn trypsin inhibitor (+/- PPACK) perfused over focal zones of collagen, dose-response curves were measured for pharmacological agents at a wall shear rate of 210 s(-1). The P2Y(1) inhibitor MRS 2179 (IC(50) = 0.233 +/- 0.132 microM) and P2Y(12) inhibitor 2-MeSAMP (IC(50) = 2.558 +/- 0.799 microM) were potent blockers of secondary platelet accumulation under flow, while the P2X(1) inhibitor (NF 449) and apyrase failed to reduce platelet accumulation. MRS 2179 and 2-MeSAMP had undetectable effects on initial platelet adhesion to collagen. Numerical simulation of convective-diffusive transport and apyrase-mediated catalytic degradation of ADP indicated that ultra-high concentrations of apyrase ( approximately 2000 U mL(-1)) would be required to have the same effect under flow as much lower concentrations (1 U mL(-1)) currently used in closed systems (aggregometry or cone-and-plate viscometer). This is the first evaluation of IC(50) values for P2Y(12) and P2Y(1) antagonists under controlled flow conditions. Evaluation of antiplatelet agents in open flow systems demonstrates that inhibition of either ADP by apyrase or antagonism of P2X(1) signaling had no inhibitory effect on platelet accumulation. This technique provides a platform for rapidly investigating effects of antithrombotic therapies simultaneously in a model injury system.


Assuntos
Clorometilcetonas de Aminoácidos/administração & dosagem , Apirase/administração & dosagem , Adesividade Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2 , Trombose/patologia , Trombose/fisiopatologia , Células Cultivadas , Humanos , Técnicas Analíticas Microfluídicas/métodos , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12
20.
Biophys J ; 98(7): 1344-52, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20371335

RESUMO

Thrombin is released as a soluble enzyme from the surface of platelets and tissue-factor-bearing cells to trigger fibrin polymerization during thrombosis under flow conditions. Although isotropic fibrin polymerization under static conditions involves protofibril extension and lateral aggregation leading to a gel, factors regulating fiber growth are poorly quantified under hemodynamic flow due to the difficulty of setting thrombin fluxes. A membrane microfluidic device allowed combined control of both thrombin wall flux (10(-13) to 10(-11) nmol/mum(2) s) and the wall shear rate (10-100 s(-1)) of a flowing fibrinogen solution. At a thrombin flux of 10(-12) nmol/mum(2) s, both fibrin deposition and fiber thickness decreased as the wall shear rate increased from 10 to 100 s(-1). Direct measurement and transport-reaction simulations at 12 different thrombin flux-wall shear rate conditions demonstrated that two dimensionless numbers, the Peclet number (Pe) and the Damkohler number (Da), defined a state diagram to predict fibrin morphology. For Da < 10, we only observed thin films at all Pe. For 10 < Da < 900, we observed either mat fibers or gels, depending on the Pe. For Da > 900 and Pe < 100, we observed three-dimensional gels. These results indicate that increases in wall shear rate quench first lateral aggregation and then protofibril extension.


Assuntos
Biofísica/métodos , Fibrina/química , Fibrinogênio/química , Polímeros/química , Trombina/química , Plaquetas/metabolismo , Membrana Celular/metabolismo , Simulação por Computador , Géis , Humanos , Microfluídica , Microscopia Eletrônica de Varredura/métodos , Estresse Mecânico , Trombose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA