Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Lab Chip ; 24(7): 1867-1874, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38487919

RESUMO

Microfluidic lab-on-a-chip technologies enable the analysis and manipulation of small fluid volumes and particles at small scales and the control of fluid flow and transport processes at the microscale, leading to the development of new methods to address a broad range of scientific and medical challenges. Microfluidic and lab-on-a-chip technologies have made a noteworthy impact in basic, preclinical, and clinical research, especially in hematology and vascular biology due to the inherent ability of microfluidics to mimic physiologic flow conditions in blood vessels and capillaries. With the potential to significantly impact translational research and clinical diagnostics, technical issues and incentive mismatches have stymied microfluidics from fulfilling this promise. We describe how accessibility, usability, and manufacturability of microfluidic technologies should be improved and how a shift in mindset and incentives within the field is also needed to address these issues. In this report, we discuss the state of the microfluidic field regarding current limitations and propose future directions and new approaches for the field to advance microfluidic technologies closer to translation and clinical use. While our report focuses on using blood as the prototypical biofluid sample, the proposed ideas and research directions can be extrapolated to other areas of hematology, oncology, biology, and medicine.


Assuntos
Técnicas Analíticas Microfluídicas , Microfluídica , Microfluídica/métodos , Técnicas Analíticas Microfluídicas/métodos , Dispositivos Lab-On-A-Chip , Pesquisa Translacional Biomédica
2.
Res Pract Thromb Haemost ; 8(1): 102291, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38222077

RESUMO

Background: During clotting, thrombin generates fibrin monomers and activates plasma-derived transglutaminase factor (F) XIIIa; collagen and thrombin-activated platelets offer thrombin-independent cellular FXIIIa (cFXIIIa) for clotting. Detecting fibrin on collagen and tissue factor surfaces in whole blood clotting typically uses complex reagents like fluorescent fibrinogen or antifibrin antibody. Objectives: We want to test whether the peptide using the α2- antiplasmin crosslinking mechanism by FXIIIa is a useful tool in both monitoring FXIIIa activity, and visualize and monitor fibrin formation, deposition, and extent of crosslinking within fibrin structures in whole blood clots formed under flow. Methods: We tested a fluorescent peptide derived from α2-antiplasmin sequence (Ac-GNQEQVSPLTLLKWC-fluorescein) to monitor the location of transglutaminase activity and fibrin during whole blood clotting under microfluidic flow (wall shear rate, 100 s-1). Results: The peptide rapidly colocated with accumulating fibrin due to transglutaminase activity, confirmed by Phe-Pro-Arg-chloromethylketone inhibiting fibrin and peptide labeling. The FXIIIa inhibitor T101 had no effect on fibrin generation but ablated the labeling of fibrin by the peptide. Similarly, Gly-Pro-Arg-Pro abated fibrin formation and thus strongly attenuated the peptide signal. At arterial wall shear rate (1000 s-1), less fibrin was formed, and consequently, less peptide labeling of fibrin was detected compared with venous conditions. The addition of tissue plasminogen activator caused a reduction of both fibrin and peptide signals. Also, the peptide strongly colocalized with fibrin (but not platelets) in clots from laser-injured mouse cremaster arterioles. For clotting under flow, FXIIIa activity was most likely plasma-derived since a RhoA inhibitor did not block α2-antiplasmin fragment cross-linking to fibrin. Conclusion: Under flow, the majority of FXIIIa-dependent fibrin labeling with peptide during clotting was distal of thrombin activity. The synthetic peptide provided a strong and sustained labeling of fibrin as it formed under flow.

3.
Biophys J ; 122(4): 697-712, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36635963

RESUMO

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.


Assuntos
Fibrina , Trombina , Trombose , Humanos , Fibrina/química , Fibrinogênio/metabolismo , Fluoresceína-5-Isotiocianato , Heparina , Microfluídica/métodos , Trombina/química , Trombose/metabolismo , Glicoproteínas da Membrana de Plaquetas/química
4.
TH Open ; 6(2): e135-e143, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35707619

RESUMO

Platelet contractility drives clot contraction to enhance clot density and stability. Clot contraction is typically studied under static conditions, with fewer studies of wall-adherent platelet clots formed under flow. We tested the effect of inhibitors of ADP and/or thromboxane A2 (TXA2) signaling on clot contraction. Using an eight-channel microfluidic device, we perfused PPACK-treated whole blood (WB) ± acetylsalicylic acid (ASA), 2-methylthioAMP (2-MeSAMP), and/or MRS-2179 over collagen (100/s) for 7.5 min, then stopped flow to observe contraction for 7.5 minutes. Two automated imaging methods scored fluorescent platelet percent contraction over the no-flow observation period: (1) "global" measurement of clot length and (2) "local" changes in surface area coverage of the numerous platelet aggregates within the clot. Total platelet fluorescence intensity (FI) decreased with concomitant decrease in global aggregate contraction when ASA, 2-MeSAMP, and/or MRS-2179 were present. Total platelet FI and global aggregate contraction were highly correlated ( R 2 = 0.87). In contrast, local aggregate contraction was more pronounced than global aggregate contraction across all inhibition conditions. However, ASA significantly reduced local aggregate contraction relative to conditions without TXA2 inhibition. P-selectin display was significantly reduced by ADP and TXA2 inhibition, but there was limited detection of global or local aggregate contraction in P-selectin-positive platelets across all conditions, as expected for densely packed "core" platelets. Our results demonstrate that global aggregate contraction is inhibited by ASA, 2-MeSAMP, and MRS-2179, while ASA more potently inhibited local aggregate contraction. These results help resolve how different platelet antagonists affect global and local clot structure and function.

5.
PLoS One ; 17(1): e0262523, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35045100

RESUMO

Risk quantification algorithms in the ICU can provide (1) an early alert to the clinician that a patient is at extreme risk and (2) help manage limited resources efficiently or remotely. With electronic health records, large data sets allow the training of predictive models to quantify patient risk. A gradient boosting classifier was trained to predict high-risk and low-risk trauma patients, where patients were labeled high-risk if they expired within the next 10 hours or within the last 10% of their ICU stay duration. The MIMIC-III database was filtered to extract 5,400 trauma patient records (526 non-survivors) each of which contained 5 static variables (age, gender, etc.) and 28 dynamic variables (e.g., vital signs and metabolic panel). Training data was also extracted from the dynamic variables using a 3-hour moving time window whereby each window was treated as a unique patient-time fragment. We extracted the mean, standard deviation, and skew from each of these 3-hour fragments and included them as inputs for training. Additionally, a survival metric upon admission was calculated for each patient using a previously developed National Trauma Data Bank (NTDB)-trained gradient booster model. The final model was able to distinguish between high-risk and low-risk patients to an AUROC of 92.9%, defined as the area under the receiver operator characteristic curve. Importantly, the dynamic survival probability plots for patients who die appear considerably different from those who survive, an example of reducing the high dimensionality of the patient record to a single trauma trajectory.


Assuntos
Mortalidade Hospitalar/tendências , Medição de Risco/métodos , Adulto , Idoso , Algoritmos , Gerenciamento de Dados/métodos , Bases de Dados Factuais , Registros Eletrônicos de Saúde , Feminino , Hospitalização/estatística & dados numéricos , Hospitalização/tendências , Humanos , Escala de Gravidade do Ferimento , Unidades de Terapia Intensiva/estatística & dados numéricos , Aprendizado de Máquina , Masculino , Pessoa de Meia-Idade , Probabilidade , Prognóstico , Curva ROC , Estudos Retrospectivos , Fatores de Risco
6.
PLoS Comput Biol ; 18(1): e1009850, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35089923

RESUMO

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.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas , Modelos Biológicos , Trombose/metabolismo , Animais , Plaquetas/citologia , Plaquetas/fisiologia , Biologia Computacional , Camundongos , Agregação Plaquetária/fisiologia , RNA-Seq , Análise de Célula Única
7.
Cardiovasc Eng Technol ; 13(2): 331-342, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34591275

RESUMO

PURPOSE: Coronary flow is affected by evolving events such as atherosclerotic plaque formation, rupture, and thrombosis, resulting in myocardial ischemia and infarction. Highly resolved 3D hemodynamic data at the stenosis is essential to model shear-sensitive thrombotic events in coronary artery disease. METHODS: We developed a hybrid 1D-3D simulation framework to compute patient-specific coronary hemodynamics efficiently. A 1D model of the coronary flow is coupled to an image-based 3D model of the region of interest. This framework affords the advantages of reduced-order modeling, decreasing the global computational cost, without sacrificing the accuracy of the quantities of interest. RESULTS: We validated our 1D-3D model against full 3D coronary simulations in healthy and diseased conditions. Our results showed good agreement between the 3D and the 1D-3D models while reducing the computational cost by 40-fold compared to the 3D simulation. The 1D-3D model predicted left/right coronary flow distribution within 3% and provided an accurate estimation of fractional flow reserve and wall shear stress distribution at the stenosis comparable to the 3D simulation. CONCLUSION: Savings in computational cost may be significant in situations with changing geometry, such as growing thrombosis. Also, this approach would allow quantifying the time-dependent effect of thrombotic growth and occlusion on the global coronary circulation.


Assuntos
Doença da Artéria Coronariana , Estenose Coronária , Reserva Fracionada de Fluxo Miocárdico , Constrição Patológica , Doença da Artéria Coronariana/diagnóstico por imagem , Circulação Coronária , Estenose Coronária/diagnóstico por imagem , Vasos Coronários/diagnóstico por imagem , Hemodinâmica , Humanos , Modelos Cardiovasculares , Modelagem Computacional Específica para o Paciente
8.
PLoS One ; 16(11): e0260366, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34813608

RESUMO

A highly reduced extrinsic pathway coagulation model (8 ODEs) under flow considered a thin 15-micron platelet layer where transport limitations were largely negligible (except for fibrinogen) and where cofactors (FVIIa, FV, FVIII) were not rate-limiting. By including thrombin feedback activation of FXI and the antithrombin-I activities of fibrin, the model accurately simulated measured fibrin formation and thrombin fluxes. Using this reduced model, we conducted 10,000 Monte Carlo (MC) simulations for ±50% variation of 5 plasma zymogens and 2 fibrin binding sites for thrombin. A sensitivity analysis of zymogen concentrations indicated that FIX activity most influenced thrombin generation, a result expected from hemophilia A and B. Averaging all MC simulations confirmed both the mean and standard deviation of measured fibrin generation on 1 tissue factor (TF) molecule per µm2. Across all simulations, free thrombin in the layer ranged from 20 to 300 nM (mean: 50 nM). The top 2% of simulations that produced maximal fibrin were dominated by conditions with low antithrombin-I activity (decreased weak and strong sites) and high FIX concentration. In contrast, the bottom 2% of simulations that produced minimal fibrin were dominated by low FIX and FX. The percent reduction of fibrin by an ideal FXIa inhibitor (FXI = 0) ranged from 71% fibrin reduction in the top 2% of MC simulations to only 34% fibrin reduction in the bottom 2% of MC simulations. Thus, the antithrombotic potency of FXIa inhibitors may vary depending on normal ranges of zymogen concentrations. This reduced model allowed efficient multivariable sensitivity analysis.


Assuntos
Fator IX/metabolismo , Fator XI/metabolismo , Fibrina/metabolismo , Fibrinogênio/metabolismo , Trombose/metabolismo , Processamento Alternativo , Circulação Sanguínea , Coagulação Sanguínea , Fibrina/genética , Fibrinogênio/genética , Humanos , Modelos Biológicos , Trombose/sangue , Trombose/genética , Trombose/fisiopatologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-34693101

RESUMO

Distinct from dilute, isotropic, and homogeneous reaction systems typically used in laboratory kinetic assays, blood is concentrated, two-phase, flowing, and highly anisotropic when clotting on a surface. This review focuses on spatial gradients that are generated and can dictate thrombus structure and function. Novel experimental and computational tools have recently emerged to explore reaction-transport coupling during clotting. Multiscale simulations help bridge tissue length scales (the coronary arteries) to millimeter scales of a growing clot to the microscopic scale of single-cell signaling and adhesion. Microfluidic devices help create and control pathological velocity profiles, albeit at a low Reynolds number. Since rate processes and force loading are often coupled, this review highlights prevailing convective-diffusive transport physics that modulate cellular and molecular processes during thrombus formation.

10.
Lab Chip ; 21(19): 3667-3674, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34476426

RESUMO

Point-of-care diagnostics of platelet and coagulation function present demanding challenges. Current clinical diagnostics often use centrifuged plasmas or platelets and frozen plasma standards, recombinant protein standards, or even venoms. Almost all commercialized tests of blood do not recreate the in vivo hemodynamics where platelets accumulate to high densities and thrombin is generated from a procoagulant surface. Despite numerous drugs that target platelets, insufficient coagulation, or excess coagulation, POC blood testing is essentially limited to viscoelastic methods that provide a clotting time, clot strength, and clot lysis, while used mostly in trauma centers with specialized capabilities. Microfluidics now allows small volumes of whole blood (<1 mL) to be tested under venous or arterial shear rates with multi-color readouts to follow platelet function, thrombin generation, fibrin production, and clot stability. Injection molded chips containing pre-patterned fibrillar collagen and lipidated tissue factor can be stored dry for 6 months at 4C, thus allowing rapid blood testing on single-use disposable chips. Using only a small imaging microscope and micropump, these microfluidic devices can detect platelet inhibitors, direct oral anticoagulants (DOACs) and their reversal agents. POC microfluidics are ideal for neonatal surgical applications that involve small blood samples, rapid DOAC testing in stroke or bleeding or emergency surgery situations with patients presenting high risk cofactors for either bleeding or thrombosis.


Assuntos
Microfluídica , Trombose , Coagulação Sanguínea , Plaquetas , Fibrina , Humanos , Recém-Nascido , Sistemas Automatizados de Assistência Junto ao Leito , Trombina , Trombose/diagnóstico
11.
Br J Haematol ; 193(5): 976-987, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33973229

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Neutralizantes/química , Inibidores dos Fatores de Coagulação Sanguínea/química , Fator VIII/química , Hemofilia A/sangue , Anticorpos Monoclonais/sangue , Anticorpos Neutralizantes/sangue , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Humanos , Proteínas Recombinantes/química
12.
Biophys J ; 120(11): 2102-2111, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33838138

RESUMO

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.


Assuntos
Adesivos , Migração e Rolagem de Leucócitos , Adesão Celular , DNA , Leucócitos , Microesferas
13.
Thromb Haemost ; 121(1): 46-57, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32961573

RESUMO

BACKGROUND: As thrombosis proceeds, certain platelets in a clot expose phosphatidylserine (PS) on their outer membrane. These PS+ platelets subsequently sort to the perimeter of the mass via platelet contraction. It remains unclear how thrombin and fibrin may alter PS+ platelet sorting within a clot. OBJECTIVE: We investigated the role of fibrin in PS+ platelet sorting. METHODS: We used an 8-channel microfluidic assay of clotting over collagen (±tissue factor) at 100 s-1 initial wall shear rate. Temporal PS+ platelet sorting was measured using a Pearson's correlation coefficient between the annexin V distribution in a clot at 9 versus 15 minutes. Spatial PS+ platelet sorting was measured using an autocorrelation metric of the final annexin V distribution. RESULTS: By 6 minutes, PS+ platelets were distributed throughout the platelet deposits and became highly spatially sorted by 15 minutes when thrombin and fibrin were blocked with Phe-Pro-Arg-chloromethylketone (PPACK). Fibrin polymerization (no PPACK) attenuated temporal and spatial PS sorting and clot contraction. With Gly-Pro-Arg-Pro (GPRP) added to block fibrin polymerization, PS sorting was prominent as was clot contraction. Exogenously added tissue plasminogen activator drove fibrinolysis that in turn promoted clot contraction and PS sorting, albeit to a lesser degree than the PPACK or GPRP conditions. Clots lacking fibrin displayed 3.6 times greater contraction than clots with fibrin. CONCLUSION: PS sorting correlated with clot contraction, as previously reported. However, fibrin inversely correlated with both percent contraction and PS sorting. Fibrin attenuated clot contraction and PS sorting relative to clots without fibrin.


Assuntos
Plaquetas/metabolismo , Fibrina/metabolismo , Fosfatidilserinas/metabolismo , Trombose/metabolismo , Coagulação Sanguínea , Plaquetas/citologia , Plaquetas/patologia , Humanos , Técnicas Analíticas Microfluídicas , Testes de Função Plaquetária , Trombina/metabolismo , Tromboplastina/metabolismo , Trombose/sangue
14.
PLoS One ; 15(11): e0242166, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33201935

RESUMO

A 400-estimator gradient boosting classifier was trained to predict survival probabilities of trauma patients. The National Trauma Data Bank (NTDB) provided 799233 complete patient records (778303 survivors and 20930 deaths) each containing 32 features, a number further reduced to only 8 features via the permutation importance method. Importantly, the 8 features can all be readily determined at admission: systolic blood pressure, heart rate, respiratory rate, temperature, oxygen saturation, gender, age and Glasgow coma score. Since death was rare, a rebalanced training set was used to train the model. The model is able to predict a survival probability for any trauma patient and accurately distinguish between a deceased and survived patient in 92.4% of all cases. Partial dependence curves (Psurvival vs. feature value) obtained from the trained model revealed the global importance of Glasgow coma score, age, and systolic blood pressure while pulse rate, respiratory rate, temperature, oxygen saturation, and gender had more subtle single variable influences. Shapley values, which measure the relative contribution of each of the 8 features to individual patient risk, were computed for several patients and were able to quantify patient-specific warning signs. Using the NTDB to sample across numerous patient traumas and hospital protocols, the trained model and Shapley values rapidly provides quantitative insight into which combination of variables in an 8-dimensional space contributed most to each trauma patient's predicted global risk of death upon emergency room admission.


Assuntos
Bases de Dados Factuais , Hospitalização , Aprendizado de Máquina , Ferimentos e Lesões/mortalidade , Algoritmos , Reações Falso-Positivas , Escala de Coma de Glasgow , Frequência Cardíaca , Humanos , Escala de Gravidade do Ferimento , Probabilidade , Curva ROC , Reprodutibilidade dos Testes , Risco , Fatores Sexuais
15.
Res Pract Thromb Haemost ; 4(7): 1158-1166, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33134782

RESUMO

BACKGROUND: Hemostatic clots have a P-selectin positive platelet core covered with a shell of P-selectin negative platelets. OBJECTIVE: To develop a new human blood microfluidic assay to interrogate core/shell mechanics. METHODS: A 2-stage assay perfused whole blood over collagen/± tissue factor (TF) for 180 seconds at 100 s-1 wall shear rate, followed by buffer perfusion at either 100 s-1 (venous) or 1000 s-1 (arterial). This microfluidic assay used an extended channel height (120 µm), allowing buffer perfusion well before occlusion. RESULTS: Clot growth on collagen stopped immediately with buffer exchange, revealing ~10% reduction in platelet fluorescence intensity (at 100 s-1) and ~30% (at 1000 s-1) by 1200 seconds. Thrombin generation (on collagen/TF) reduced erosion at either buffer flow rate. P-selectin-positive platelets were stable (no erosion) against 1000 s-1, in contrast to P-selectin negative platelets. Thrombin inhibition (with D-Phe-Pro-Arg-CMK) reduced the number of P-selectin-positive platelets and lowered thrombus stability through the reduction of P-selectin-positive platelets. Interestingly, fibrin inhibition (with H-Gly-Pro-Arg-Pro-OH acetate salt) increased the number of P-selectin-positive platelets but did not lower stability, suggesting that fibrin was only in the core region. Thromboxane inhibition reduced P-selectin-positive platelets and caused a nearly 60% reduction of the clot at arterial buffer flow. P2Y1 antagonism reduced clot size and the number of P-selectin-positive platelets and reduced the stability of P-selectin-negative platelets. CONCLUSION: The 2-stage assay (extended channel height plus buffer exchange) interrogated platelet stability using human blood. Under all conditions, P-selectin-positive platelets never left the clot.

16.
Biomicrofluidics ; 14(5): 054103, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33014235

RESUMO

Custom polydimethylsiloxane (PDMS) microfluidic devices allow for small-volume human blood research under hemodynamic conditions of bleeding and clotting. However, issues of PDMS molding/assembly, bio-coating, and sample preparation often limit their point-of-care use. We aim to develop a microfluidic device that has the same utility as previously established PDMS devices but which is more usable in point-of-care operation. We designed an injection-molded 1 × 3 in.2 device with eight flow paths crossing a bio-printed surface of a collagen/tissue factor. The device is rapidly primed and compatible with multi-channel pipetting (<0.5 ml blood) and operates under venous or arterial shear rates using constant flow rate or constant pressure modes. Platelet and fibrin deposition were monitored dynamically by the imaging of immunofluorescence. For whole blood clotting at a wall shear rate of 200 s-1, the intrachip CV at 400 s for platelet and fibrin deposition was 10% and the interdonor CV at 400 s was 30% for platelet and 22% for fibrin deposition (across 10 healthy donors). No significant difference was detected for samples tested on a new chip vs a chip stored for 6 months at 4 °C. Using the fibrin signal, dose-response testing of whole blood revealed IC50's of 120 nM for rivaroxaban and apixaban, and 60 nM for dabigatran. A complete reversal of apixaban inhibition was observed for an equimolar addition of Xa DOAC reversal agent Andexanet Alfa. We demonstrate the ability to manufacture single-use, storage-stable eight-channel chips. In clinical settings, such chips may help evaluate patient bleeding risk, therapy choice, drug activity, or reversal.

17.
J Appl Lab Med ; 5(6): 1253-1264, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32674119

RESUMO

BACKGROUND: Platelet dysfunction often accompanies trauma-induced coagulopathy. Because soluble fibrin impairs platelet glycoprotein VI (GPVI) signaling and platelets of trauma patients can display impaired calcium mobilization, we explored the role of fibrinolysis on platelet dysfunction during trauma. METHODS: Convulxin-induced GPVI calcium mobilization was investigated in healthy platelet-rich plasma (PRP) pretreated with thrombin and tissue plasminogen activator (tPA). Blood samples from healthy participants (n = 7) and trauma patients (n = 22) were tested for platelet calcium mobilization, plasma D-dimer, platelet D-dimer binding (via flow cytometry), and platelet lumi-aggregometry. RESULTS: For healthy platelets, maximal platelet dysfunction was observed when cross-linked soluble fibrin (no tPA) or cross-linked fibrin degradation products (FDPs) were generated in suspension before convulxin stimulation. Lack of fibrin polymerization (inhibited by Gly-Pro-Arg-Pro [GPRP]) or lack of factor XIIIa cross-linking (T101-inhibited) restored GPVI signaling, whereas non-cross-linked FDPs only partially blocked signaling induced by convulxin. In addition, D-dimer added to healthy PRP impaired platelet aggregation and dense granule release induced by various agonists. Plasma D-dimer level was strongly correlated (R = 0.8236) with platelet dysfunction as measured by platelet calcium mobilization induced with various agonists. By 48 to 120 h after trauma, plasma D-dimer levels declined, and platelet function increased significantly but not to healthy levels. Trauma platelets displayed elevated D-dimer binding that was only partially reduced by αIIbß3-inhibitor GR144053. After 60-minute incubation, washed healthy platelets resuspended in plasma from trauma patients captured approximately 10 000 D-dimer equivalents per platelet. CONCLUSIONS: During trauma, D-dimer and FDPs inhibit platelets, potentially via GPVI and integrin αIIbß3 engagement, contributing to a fibrinolysis-dependent platelet loss-of-function phenotype.


Assuntos
Produtos de Degradação da Fibrina e do Fibrinogênio , Ativador de Plasminogênio Tecidual , Plaquetas , Fibrina , Humanos
18.
Thromb Res ; 192: 141-151, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32480168

RESUMO

Kinase inhibitors can pose bleeding risks as platelet signaling evolves during clotting. Using microfluidics (200 s-1 wall shear rate) to perfuse Factor XIIa-inhibited or thrombin-inhibited whole blood (WB) over collagen ± tissue factor (TF), we explored the potency of the Src family kinase (SFK) inhibitor dasatinib or the spleen tyrosine kinase (Syk) inhibitor GS-9973 present at clot initiation or added after 90 s (via rapid switch to inhibitor-pretreated WB). When initially present, dasatinib potently inhibited platelet deposition on collagen (no TF). Furthermore, dasatinib immediately inhibited subsequent platelet deposition when introduced 90 s after clot initiation. However, when thrombin was generated, dasatinib was markedly less potent against platelet deposition on collagen/TF (but blocked fibrin deposition) and had no effect when added 90 s after clot initiation. Similarly, dasatinib added at 90 s had no effect on clotting on collagen/TF when fibrin was also blocked with Gly-Pro-Arg-Pro, indicating that strong thrombin-induced signaling (but not fibrin-induced signaling) can bypass the SFK inhibition at later times. The Syk inhibitor GS-9973 was less potent than dasatinib when present initially, but inhibited clot growth when added at 90 s, even in the presence of thrombin (±fibrin). Interestingly, the active form (R-406) of fostamatinib inhibits platelet function in only 2 0f 5 healthy blood samples. SFK-inhibitors may have reduced antithrombotic activity and reduced bleeding risks in settings of high TF and local thrombin generation. For oncology patients, SFK-inhibitors like dasatinib may have reduced antithrombotic activity and reduced bleeding risk in settings of local thrombin generation.


Assuntos
Trombina , Quinases da Família src , Plaquetas , Colágeno , Dasatinibe/farmacologia , Humanos
19.
Res Pract Thromb Haemost ; 4(1): 54-63, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31989085

RESUMO

BACKGROUND: Microfluidic clotting assays permit drug action studies for hemophilia therapeutics under flow. However, limited availability of patient samples and Inter-donor variability limit the application of such assays, especially with many patients on prophylaxis. OBJECTIVE: To develop approaches to phenocopy hemophilia using modified healthy blood in microfluidic assays. METHODS: Corn trypsin inhibitor (4 µg/mL)-treated healthy blood was dosed with either anti-factor VIII (FVIII; hemophilia A model) or a recombinant factor IX (FIX) missense variant (FIX-V181T; hemophilia B model). Treated blood was perfused at 100 s-1 wall shear rate over collagen/tissue factor (TF) or collagen/factor XIa (FXIa). RESULTS: Anti-FVIII partially blocked fibrin production on collagen/TF, but completely blocked fibrin production on collagen/FXIa, a phenotype reversed with 1 µmol/L bispecific antibody (emicizumab), which binds FIXa and factor X. As expected, emicizumab had no significant effect on healthy blood (no anti-FVIII present) perfused over collagen/FXIa. The efficacy of emicizumab in anti-FVIII-treated healthy blood phenocopied the action of emicizumab in the blood of a patient with hemophilia A perfused over collagen/FXIa. Interestingly, a patient-derived FVIII-neutralizing antibody reduced fibrin production when added to healthy blood perfused over collagen/FXIa. For low TF surfaces, reFIX-V181T (50 µg/mL) fully blocked platelet and fibrin deposition, a phenotype fully reversed with anti-TFPI. CONCLUSION: Two new microfluidic hemophilia A and B models demonstrate the potency of anti-TF pathway inhibitor, emicizumab, and a patient-derived inhibitory antibody. Using collagen/FXIa-coated surfaces resulted in reliable and highly sensitive hemophilia models.

20.
PLoS Comput Biol ; 15(8): e1007266, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31381558

RESUMO

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.


Assuntos
Coagulação Sanguínea/fisiologia , Modelos Biológicos , Trombose/sangue , Plaquetas/metabolismo , Colágeno/sangue , Biologia Computacional , Simulação por Computador , Cisteína Endopeptidases/sangue , Fator XIa/metabolismo , Fibrina/metabolismo , Humanos , Técnicas In Vitro , Cinética , Proteínas de Neoplasias/sangue , Fluxo Sanguíneo Regional/fisiologia , Trombina/metabolismo , Tromboplastina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...