Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Lab Chip ; 23(13): 3092-3105, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37305977

ABSTRACT

Hydrodynamic trapping of a particle or cluster of particles based on contact and non-contact approaches has brought prominent insights to micro-nano scale applications. Of the non-contact methods, image-based real-time control in cross-slot microfluidic devices is one of the most promising potential platform for single cellular assays. Here, we report results from experiments conducted in two cross-slot microfluidic channels of different widths, with varying real-time delay of the control algorithm and different magnification. Sustained trapping of 5 µm diameter particles was achieved with high strain rates, of order 102 s-1, higher than in any previous studies. Our experiments show that the maximum attainable strain rate is a function of the real-time delay of the control algorithm and the particle resolution (pixel/µm). Therefore, we anticipate that with further reduced time delays and enhanced particle resolution, considerably higher strain rates can be attained, opening the platform to single cellular assay studies where very high strain rates are required.

2.
Res Pract Thromb Haemost ; 7(1): 100037, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36846647

ABSTRACT

Background: Blood platelets have evolved a complex mechanotransduction machinery to rapidly respond to hemodynamic conditions. A variety of microfluidic flow-based approaches have been developed to explore platelet mechanotransduction; however, these experimental models primarily focus on the effects of increased wall shear stress on platelet adhesion events and do not consider the critical effects of extensional strain on platelet activation in free flow. Objectives: We report the development and application of a hyperbolic microfluidic assay that allows for investigation of platelet mechanotransduction under quasi-homogenous extensional strain rates in the absence of surface adhesions. Methods: Using a combined computational fluid dynamic and experimental microfluidic approach, we explore 5 extensional strain regimes (geometries) and their effect on platelet calcium signal transduction. Results: We demonstrate that in the absence of canonical adhesion, receptor engagement platelets are highly sensitive to both initial increase and subsequent decrease in extensional strain rates within the range of 747 to 3319/s. Furthermore, we demonstrate that platelets rapidly respond to the rate of change in extensional strain and define a threshold of ≥7.33 × 106/s/m, with an optimal range of 9.21 × 107 to 1.32 × 108/s/m. In addition, we demonstrate a key role of both the actin-based cytoskeleton and annular microtubules in the modulation of extensional strain-mediated platelet mechanotransduction. Conclusion: This method opens a window onto a novel platelet signal transduction mechanism and may have potential diagnostic utility in the identification of patients who are prone to thromboembolic complications associated with high-grade arterial stenosis or are on mechanical circulatory support systems, for which the extensional strain rate is a predominant hemodynamic driver.

3.
BMC Biol ; 20(1): 73, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35331224

ABSTRACT

BACKGROUND: Supraphysiological hemodynamics are a recognized driver of platelet activation and thrombosis at high-grade stenosis and in blood contacting circulatory support devices. However, whether platelets mechano-sense hemodynamic parameters directly in free flow (in the absence of adhesion receptor engagement), the specific hemodynamic parameters at play, the precise timing of activation, and the signaling mechanism(s) involved remain poorly elucidated. RESULTS: Using a generalized Newtonian computational model in combination with microfluidic models of flow acceleration and quasi-homogenous extensional strain, we demonstrate that platelets directly mechano-sense acute changes in free-flow extensional strain independent of shear strain, platelet amplification loops, von Willebrand factor, and canonical adhesion receptor engagement. We define an extensional strain sensing "mechanosome" in platelets involving cooperative Ca2+ signaling driven by the mechanosensitive channel Piezo1 (as the primary strain sensor) and the fast ATP gated channel P2X1 (as the secondary signal amplifier). We demonstrate that type II PI3 kinase C2α activity (acting as a "clutch") couples extensional strain to the mechanosome. CONCLUSIONS: Our findings suggest that platelets are adapted to rapidly respond to supraphysiological extensional strain dynamics, rather than the peak magnitude of imposed wall shear stress. In the context of overall platelet activation and thrombosis, we posit that "extensional strain sensing" acts as a priming mechanism in response to threshold levels of extensional strain allowing platelets to form downstream adhesive interactions more rapidly under the limiting effects of supraphysiological hemodynamics.


Subject(s)
Platelet Activation , Thrombosis , Blood Platelets/metabolism , Hemodynamics , Humans , Ion Channels , Stress, Mechanical , von Willebrand Factor/metabolism
4.
Sci Transl Med ; 12(553)2020 07 22.
Article in English | MEDLINE | ID: mdl-32718993

ABSTRACT

Arterial thrombosis causes heart attacks and most strokes and is the most common cause of death in the world. Platelets are the cells that form arterial thrombi, and antiplatelet drugs are the mainstay of heart attack and stroke prevention. Yet, current drugs have limited efficacy, preventing fewer than 25% of lethal cardiovascular events without clinically relevant effects on bleeding. The key limitation on the ability of all current drugs to impair thrombosis without causing bleeding is that they block global platelet activation, thereby indiscriminately preventing platelet function in hemostasis and thrombosis. Here, we identify an approach with the potential to overcome this limitation by preventing platelet function independently of canonical platelet activation and in a manner that appears specifically relevant in the setting of thrombosis. Genetic or pharmacological targeting of the class II phosphoinositide 3-kinase (PI3KC2α) dilates the internal membrane reserve of platelets but does not affect activation-dependent platelet function in standard tests. Despite this, inhibition of PI3KC2α is potently antithrombotic in human blood ex vivo and mice in vivo and does not affect hemostasis. Mechanistic studies reveal this antithrombotic effect to be the result of impaired platelet adhesion driven by pronounced hemodynamic shear stress gradients. These findings demonstrate an important role for PI3KC2α in regulating platelet structure and function via a membrane-dependent mechanism and suggest that drugs targeting the platelet internal membrane may be a suitable approach for antithrombotic therapies with an improved therapeutic window.


Subject(s)
Blood Platelets , Thrombosis , Animals , Hemostasis , Mice , Phosphatidylinositol 3-Kinases , Platelet Activation , Platelet Aggregation , Thrombosis/drug therapy
5.
Semin Thromb Hemost ; 46(5): 622-636, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32604421

ABSTRACT

The manipulation of blood within in vitro environments presents a persistent challenge, due to the highly reactive nature of blood, and its multifaceted response to material contact, changes in environmental conditions, and stimulation during handling. Microfluidic Lab-on-Chip systems offer the promise of robust point-of-care diagnostic tools and sophisticated research platforms. The capacity for precise control of environmental and experimental conditions afforded by microfluidic technologies presents unique opportunities that are particularly relevant to research and clinical applications requiring the controlled manipulation of blood. A critical bottleneck impeding the translation of existing Lab-on-Chip technology from laboratory bench to the clinic is the ability to reliably handle relatively small blood samples without negatively impacting blood composition or function. This review explores design considerations critical to the development of microfluidic systems intended for use with whole blood from an engineering perspective. Material hemocompatibility is briefly explored, encompassing common microfluidic device materials, as well as surface modification strategies intended to improve hemocompatibility. Operational hemocompatibility, including shear-induced effects, temperature dependence, and gas interactions are explored, microfluidic sample preparation methodologies are introduced, as well as current techniques for on-chip manipulation of the whole blood. Finally, methods of assessing hemocompatibility are briefly introduced, with an emphasis on primary hemostasis and platelet function.


Subject(s)
Biocompatible Materials/standards , Microfluidics/methods , Humans
6.
J Thromb Haemost ; 18(3): 748-752, 2020 03.
Article in English | MEDLINE | ID: mdl-32112535

ABSTRACT

Experimental videomicroscopic in vitro assays of thrombus formation based on blood perfusion are instrumental in a wide range of basic studies in thrombosis, screening for hereditary or acquired plateletrelated pathologies, and assessing the effectiveness of novel anti-platelet therapies. Here, we discuss application of the broadly used "in vitro thrombosis model": a frequently used assay to study the formation of 3D aggregates under flow, which involves perfusing anticoagulated whole blood over fibrillar collagen in a flow geometry of rectangular cross-section, such as glass microcapillaries or parallel-plate flow chambers. Major advantaged of this assay are simplicity and ability to reproduce the four main stages of platelet thrombus formation, i.e. platelet tethering, adhesion, activation and aggregation under a wide range of hemodynamic conditions. On the other hand, these devices represent, at best, simple reductive models of thrombosis. We also describe how blood flow assays can be used to study various aspects of platelet function on adhesive proteins and discuss the relevance of such flow models. Finally, we propose recommendations for standardization related to the use of this assay that cover collagen source, coating methods, micropatterning, sample composition, anticoagulation, choice of flow device, hemodynamic conditions, quantification challenges, variability, pre-analytical conditions and other issues.


Subject(s)
Thrombosis , Blood Platelets , Communication , Hemostasis , Humans , Platelet Function Tests , Reference Standards
7.
Anal Chem ; 91(16): 10830-10839, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31343155

ABSTRACT

There is a need for scalable automated lab-on-chip systems incorporating precise hemodynamic control that can be applied to high-content screening of new more efficacious antiplatelet therapies. This paper reports on the development and characterization of a novel active micropump-mixer microfluidic to address this need. Using a novel reciprocating elastomeric micropump design, we take advantage of the flexible structural and actuation properties of this framework to manage the hemodynamics for on-chip platelet thrombosis assay on type 1 fibrillar collagen, using whole blood. By characterizing and harnessing the complex three-dimensional hemodynamics of the micropump operation in conjunction with a microvalve controlled reagent injection system we demonstrate that this prototype can act as a real-time assay of antiplatelet drug pharmacokinetics. In a proof-of-concept preclinical application, we utilize this system to investigate the way in which rapid dosing of human whole blood with isoform selective inhibitors of phosphatidylinositol 3-kinase dose dependently modulate platelet thrombus dynamics. This modular system exhibits utility as an automated multiplexable assay system with applications to high-content chemical library screening of new antiplatelet therapies.


Subject(s)
Indomethacin/blood , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Platelet Aggregation Inhibitors/blood , Blood Platelets/drug effects , Hemodynamics , Humans , Indomethacin/pharmacokinetics , Microfluidic Analytical Techniques/instrumentation , Platelet Aggregation Inhibitors/pharmacokinetics
8.
Res Pract Thromb Haemost ; 3(2): 197-206, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31011704

ABSTRACT

BACKGROUND: Arterial thrombosis models are important for preclinical evaluation of antithrombotics but how anesthetic protocol can influence experimental results is not studied. OBJECTIVES: We studied how three most commonly used rodent anesthetics affect the induction of thrombosis and thrombus resolution with antiplatelet agent integrilin (Eptifibatide). METHODS: The Folts, electrolytic, and FeCl3 models of carotid artery thrombosis were evaluated. The extent of blood flow reduction required to elicit cyclic flow reductions (CFR) was examined in the Folts model. The occlusion time and stability following electrolytic or FeCl3 injury was assessed. The efficacy of Eptifibatide was studied in each cohort and clot composition following FeCl3 application was assessed histologically. RESULTS: Isoflurane and ketamine-xylazine (ket-x) elicited higher basal blood flow velocities. For reliable CFR in the Folts model, a higher degree of blood flow reduction was required under ket-x and isoflurane. For the FeCl3 and electrolytic models, injury severity had to be increased in mice under ket-x anesthesia to achieve rapid occlusion. FeCl3-injured artery sections from ket-x and isoflurane-treated mice showed vessel dilatation and clots that were more fibrin/red-cell rich compared to pentobarbitone. Integrilin led to cycle abolishment for all three Folts-injury cohorts but for the electrolytic model a 2.5-fold higher dose was required to restore blood flow under pentobarbitone. Integrilin after FeCl3 arterial injury was partially ineffective in isoflurane-treated mice. CONCLUSIONS: Anesthesia impacts rodent carotid artery occlusion experiments and alters integrilin efficacy. It is important to consider anesthetic protocols in animal experiments involving pharmacological agents for treatment of atherothrombosis.

9.
FEBS Lett ; 593(1): 88-96, 2019 01.
Article in English | MEDLINE | ID: mdl-30421419

ABSTRACT

PI3KC2α is a phosphoinositide 3-kinase with a recently reported function in platelets; PI3KC2α-deficient mouse platelets have altered membrane structure and impaired function. Yet, how these membrane changes cause platelet dysfunction remains unknown. Here, focused ion beam-scanning electron microscopy of PI3KC2α-deficient platelet ultrastructure reveals a specific effect on the internal membrane structure, while liquid chromatography-tandem mass spectrometry profiling of 294 lipid species shows unaltered lipid composition. Functionally, PI3KC2α-deficient platelets exhibit impaired thrombosis specifically under conditions involving membrane tethering. These studies indicate that the structural changes in PI3KC2α-deficient platelets are limited to the membrane, occur without major changes in lipid composition, and selectively impair cell function during thrombus formation. These findings illustrate a unique mechanism that may be targetable for anti-thrombotic benefit.


Subject(s)
Blood Platelets/cytology , Cell Membrane/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blood Platelets/chemistry , Chromatography, Liquid , Gene Knockout Techniques , Membrane Lipids/chemistry , Mice , Microscopy, Electron, Scanning , Tandem Mass Spectrometry
10.
Lab Chip ; 18(12): 1778-1792, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29789838

ABSTRACT

This paper reports on the parameters that determine the haemocompatibility of elastomeric microvalves for blood handling in microfluidic systems. Using a comprehensive investigation of blood function, we describe a hierarchy of haemocompatibility as a function of microvalve geometry and identify a "normally-closed" v-gate pneumatic microvalve design that minimally affects blood plasma fibrinogen and von Willebrand factor composition, minimises effects on erythrocyte structure and function, and limits effects on platelet activation and aggregation, while facilitating rapid switching control for blood sample delivery. We propose that the haemodynamic profile of valve gate geometries is a significant determinant of platelet-dependent biofouling and haemocompatibility. Overall our findings suggest that modification of microvalve gate geometry and consequently haemodynamic profile can improve haemocompatibility, while minimising the requirement for chemical or protein modification of microfluidic surfaces. This biological insight and approach may be harnessed to inform future haemocompatible microfluidic valve and component design, and is an advance towards lab-on-chip automation for blood based diagnostic systems.


Subject(s)
Blood Transfusion/instrumentation , Elastomers/chemistry , Microfluidic Analytical Techniques/instrumentation , Blood Platelets/cytology , Blood Platelets/physiology , Equipment Design , Erythrocytes/cytology , Erythrocytes/physiology , Humans , Materials Testing , Stress, Mechanical
11.
Lab Chip ; 17(15): 2595-2608, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28660968

ABSTRACT

Von Willebrand's disease (VWD) is the most common inherited bleeding disorder caused by either quantitative or qualitative defects of von Willebrand factor (VWF). Current tests for VWD require relatively large blood volumes, have low throughput, are time-consuming, and do not incorporate the physiologically relevant effects of haemodynamic forces. We developed a microfluidic device incorporating micro-contractions that harnesses well-defined haemodynamic strain gradients to initiate platelet aggregation in citrated whole blood. The microchannel architecture has been specifically designed to allow for continuous real-time imaging of platelet aggregation dynamics. Subjects aged ≥18 years with previously diagnosed VWD or who presented for evaluation of a bleeding disorder, where the possible diagnosis included VWD, were tested. Samples were obtained for device characterization as well as for pathology-based testing. Platelet aggregation in the microfluidic device is independent of platelet amplification loops but dependent on low-level platelet activation, GPIb/IX/V and integrin αIIbß3 engagement. Microfluidic output directly correlates with VWF antigen levels and is able to sensitively detect aggregation defects associated with VWD subtypes. Testing demonstrated a strong correlation with standard clinical laboratory-based tests. Head-to-head comparison with PFA100® demonstrated equivalent, if not improved, sensitivity for screening aggregation defects associated with VWD. This strain rate gradient microfluidic prototype has the potential to be a clinically useful, rapid and high throughput-screening tool for VWD as well as other strain-dependent platelet disorders. In addition, the microfluidic device represents a novel approach to examine the effects of high magnitude/short duration (ms) strain rate gradients on platelet function.


Subject(s)
Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Platelet Aggregation/physiology , Platelet Function Tests/instrumentation , von Willebrand Diseases/diagnosis , Adolescent , Adult , Deamino Arginine Vasopressin/administration & dosage , Deamino Arginine Vasopressin/pharmacology , Equipment Design , Female , Hematocrit , Humans , Male , Microfluidic Analytical Techniques/methods , Middle Aged , Platelet Aggregation/drug effects , Platelet Function Tests/methods , Young Adult , von Willebrand Factor
12.
IEEE Trans Biomed Eng ; 62(7): 1718-27, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25706500

ABSTRACT

The recent application of new microfluidic technologies and methods has facilitated significant progress in the understanding of the fundamental mechanisms governing blood platelet function and how these parameters affect pathological thrombus formation. In-line with these new bioengineering approaches, the application of nonlinear dynamic systems analysis holds particular potential to extend our understanding of the complex interplay between mechanical and biochemical factors that underlie this complex biological phenomenon. In this paper we propose a simple mathematical model of the main dynamics of platelet aggregation/disaggregation observed experimentally in a novel microfluidic device that approximates a severe arterial stenosis. We apply dynamic systems theory (system identification) to explore the dynamics of the biomechanical platelet aggregation response to a range of shear stress rates, inhibiting blood-born chemical pathways of platelet activation (ADP, TXA2, and thrombin). We demonstrate that the proposed model is able to replicate experimental results with low variation, and suggest that the reduced set of model parameters has the potential to be used as a simplified way to evaluate the biomechanical dynamics of platelet aggregation. The proposed model has application to the development of automatic controllers within the context of microfluidic systems that may show great utility in the clinical assessment of platelet hyperfunction.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Platelet Aggregation/physiology , Biomechanical Phenomena , Blood Physiological Phenomena , Equipment Design , Humans , Nonlinear Dynamics
13.
J Biol Chem ; 289(8): 5051-60, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24385425

ABSTRACT

The Dok proteins are a family of adaptor molecules that have a well defined role in regulating cellular migration, immune responses, and tumor progression. Previous studies have demonstrated that Doks-1 to 3 are expressed in platelets and that Dok-2 is tyrosine-phosphorylated downstream of integrin αIIbß3, raising the possibility that it participates in integrin αIIbß3 outside-in signaling. We demonstrate that Dok-2 in platelets is primarily phosphorylated by Lyn kinase. Moreover, deficiency of Dok-2 leads to dysregulated integrin αIIbß3-dependent cytosolic calcium flux and phosphatidylinositol(3,4)P2 accumulation. Although agonist-induced integrin αIIbß3 affinity regulation was unaltered in Dok-2(-/-) platelets, Dok-2 deficiency was associated with a shear-dependent increase in integrin αIIbß3 adhesive function, resulting in enhanced platelet-fibrinogen and platelet-platelet adhesive interactions under flow. This increase in adhesion was restricted to discoid platelets and involved the shear-dependent regulation of membrane tethers. Dok-2 deficiency was associated with an increased rate of platelet aggregate formation on thrombogenic surfaces, leading to accelerated thrombus growth in vivo. Overall, this study defines an important role for Dok-2 in regulating biomechanical adhesive function of discoid platelets. Moreover, they define a previously unrecognized prothrombotic mechanism that is not detected by conventional platelet function assays.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Shear Strength , Adaptor Proteins, Signal Transducing/deficiency , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Fibrinogen/pharmacology , Hemorheology/drug effects , Humans , Immobilized Proteins/pharmacology , Mice , Mice, Inbred C57BL , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/deficiency , Platelet Adhesiveness/drug effects , Shear Strength/drug effects , Thrombosis/metabolism , Thrombosis/pathology , Thrombosis/physiopathology , Time Factors
14.
PLoS One ; 8(10): e74123, 2013.
Article in English | MEDLINE | ID: mdl-24194822

ABSTRACT

This paper reports on an investigation of mass transport of blood cells at micro-scale stenosis where local strain-rate micro-gradients trigger platelet aggregation. Using a microfluidic flow focusing platform we investigate the blood flow streams that principally contribute to platelet aggregation under shear micro-gradient conditions. We demonstrate that relatively thin surface streams located at the channel wall are the primary contributor of platelets to the developing aggregate under shear gradient conditions. Furthermore we delineate a role for red blood cell hydrodynamic lift forces in driving enhanced advection of platelets to the stenosis wall and surface of developing aggregates. We show that this novel microfluidic platform can be effectively used to study the role of mass transport phenomena driving platelet recruitment and aggregate formation and believe that this approach will lead to a greater understanding of the mechanisms underlying shear-gradient dependent discoid platelet aggregation in the context of cardiovascular diseases such as acute coronary syndromes and ischemic stroke.


Subject(s)
Constriction, Pathologic/physiopathology , Platelet Aggregation/physiology , Thrombosis/physiopathology , Biological Transport/physiology , Blood Platelets/physiology , Humans , Hydrodynamics , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
15.
Methods Mol Biol ; 1046: 39-58, 2013.
Article in English | MEDLINE | ID: mdl-23868581

ABSTRACT

Fluorescence microscopy techniques have provided important insights into the structural and signalling events occurring during platelet adhesion under both static and blood flow conditions. However, due to limitations in sectioning ability and sensitivity these techniques are restricted in their capacity to precisely image the adhesion footprint of spreading platelets. In particular, investigation of platelet adhesion under hemodynamic shear stress requires an imaging platform with high spatial discrimination and sensitivity and rapid temporal resolution. This chapter describes in detail a multimode imaging approach combining total internal reflection fluorescence microscopy (TIRFM) with high speed epifluorescence and differential interference contrast (DIC) microscopy along with a novel microfluidic perfusion system developed in our laboratory to examine platelet membrane adhesion dynamics under static and flow conditions.


Subject(s)
Microfluidic Analytical Techniques/methods , Molecular Biology/methods , Platelet Adhesiveness/genetics , Blood Platelets/metabolism , Hemodynamics , Humans , Microscopy, Fluorescence , Stress, Mechanical
16.
Methods Mol Biol ; 788: 73-89, 2012.
Article in English | MEDLINE | ID: mdl-22130701

ABSTRACT

The platelet is a specialized adhesive cell that plays a key role in thrombus formation under both physiological and pathological blood flow conditions. Platelet adhesion and activation are dynamic processes associated with rapid morphological and functional changes, with the earliest signaling events occurring over a subsecond time-scale. The relatively small size of platelets combined with the dynamic nature of platelet adhesion under blood flow means that the investigation of platelet signaling events requires techniques with both high spatial discrimination and rapid temporal resolution. Unraveling the complex signaling processes governing platelet adhesive function under conditions of hemodynamic shear stress has been a longstanding goal in platelet research and has been greatly influenced by the development and application of microimaging-based techniques. Advances in the area of epi-fluorescence and confocal-based platelet calcium (Ca(2+)) imaging have facilitated the in vitro and in vivo elucidation of the early signaling events regulating platelet adhesion and activation. These studies have identified distinct Ca(2+) signaling mechanisms that serve to dynamically regulate activation of the major platelet integrin α(IIb)ß(3) and associated adhesion and aggregation processes under flow. This chapter describes in detail a ratiometric calcium imaging protocol and associated troubleshooting procedures developed in our laboratory to examine live platelet Ca(2+) signaling dynamics. This technique provides a method for high-resolution imaging of the Ca(2+) dynamics underpinning platelet adhesion and thrombus formation under conditions of pathophysiological shear stress.


Subject(s)
Blood Circulation/physiology , Blood Platelets/cytology , Blood Platelets/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Animals , Cell Adhesion , Cell Survival , Humans , Mice
17.
J Biomech ; 44(6): 1031-9, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21392770

ABSTRACT

Platelet activation under blood flow is thought to be critically dependent on the autologous secretion of soluble platelet agonists (chemical activators) such as ADP and thromboxane. However, recent evidence challenging this model suggests that platelet activation can occur independent of soluble agonist signalling, in response to the mechanical effects of micro-scale shear gradients. A key experimental tool utilized to define the effect of shear gradients on platelet aggregation is the murine intravital microscopy model of platelet thrombosis under conditions of acute controlled arteriolar stenosis. This paper presents a computational structural and hydrodynamic simulation of acute stenotic blood flow in the small bowel mesenteric vessels of mice. Using a homogeneous fluid at low Reynolds number (0.45) we investigated the relationship between the local hydrodynamic strain-rates and the severity of arteriolar stensosis. We conclude that the critical rates of blood flow acceleration and deceleration at sites of artificially induced stenosis (vessel side-wall compression or ligation) are a function of tissue elasticity. By implementing a structural simulation of arteriolar side wall compression, we present a mechanistic model that provides accurate simulations of stenosis in vivo and allows for predictions of the effects on local haemodynamics in the murine small bowel mesenteric thrombosis model.


Subject(s)
Blood Platelets , Constriction, Pathologic/physiopathology , Mesenteric Arteries/physiopathology , Models, Cardiovascular , Platelet Activation , Thrombosis/physiopathology , Animals , Constriction, Pathologic/complications , Disease Models, Animal , Mice , Thrombosis/etiology
18.
Lab Chip ; 10(3): 291-302, 2010 Feb 07.
Article in English | MEDLINE | ID: mdl-20091000

ABSTRACT

This paper reports the development of a platform technology for measuring platelet function and aggregation based on localized strain rate micro-gradients. Recent experimental findings within our laboratories have identified a key role for strain rate micro-gradients in focally triggering initial recruitment and subsequent aggregation of discoid platelets at sites of blood vessel injury. We present the design justification, hydrodynamic characterization and experimental validation of a microfluidic device incorporating contraction-expansion geometries that generate strain rate conditions mimicking the effects of pathological changes in blood vessel geometry. Blood perfusion through this device supports our published findings of both in vivo and in vitro platelet aggregation and confirms a critical requirement for the coupling of blood flow acceleration to downstream deceleration for the initiation and stabilization of platelet aggregation, in the absence of soluble platelet agonists. The microfluidics platform presented will facilitate the detailed analysis of the effects of hemodynamic parameters on the rate and extent of platelet aggregation and will be a useful tool to elucidate the hemodynamic and platelet mechano-transduction mechanisms, underlying this shear-dependent process.


Subject(s)
Blood Flow Velocity/physiology , Blood Platelets/physiology , Mechanotransduction, Cellular/physiology , Microfluidic Analytical Techniques/instrumentation , Platelet Activation/physiology , Biomimetic Materials , Blood Platelets/cytology , Cells, Cultured , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Humans , Reproducibility of Results , Sensitivity and Specificity
19.
J Biol Chem ; 285(4): 2886-96, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19940148

ABSTRACT

Phosphoinositide (PI) 3-kinase (PI3K) signaling processes play an important role in regulating the adhesive function of integrin alpha(IIb)beta(3), necessary for platelet spreading and sustained platelet aggregation. PI3K inhibitors are effective at reducing platelet aggregation and thrombus formation in vivo and as a consequence are currently being evaluated as novel antithrombotic agents. PI3K regulation of integrin alpha(IIb)beta(3) activation (affinity modulation) primarily occurs downstream of G(i)-coupled and tyrosine kinase-linked receptors linked to the activation of Rap1b, AKT, and phospholipase C. In the present study, we demonstrate an important role for PI3Ks in regulating the avidity (strength of adhesion) of high affinity integrin alpha(IIb)beta(3) bonds, necessary for the cellular transmission of contractile forces. Using knock-out mouse models and isoform-selective PI3K inhibitors, we demonstrate that the Type Ia p110 beta isoform plays a major role in regulating thrombin-stimulated fibrin clot retraction in vitro. Reduced clot retraction induced by PI3K inhibitors was not associated with defects in integrin alpha(IIb)beta(3) activation, actin polymerization, or actomyosin contractility but was associated with a defect in integrin alpha(IIb)beta(3) association with the contractile cytoskeleton. Analysis of integrin alpha(IIb)beta(3) adhesion contacts using total internal reflection fluorescence microscopy revealed an important role for PI3Ks in regulating the stability of high affinity integrin alpha(IIb)beta(3) bonds. These studies demonstrate an important role for PI3K p110 beta in regulating the avidity of high affinity integrin alpha(IIb)beta(3) receptors, necessary for the cellular transmission of contractile forces. These findings may provide new insight into the potential antithrombotic properties of PI3K p110 beta inhibitors.


Subject(s)
Blood Platelets/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/drug effects , Class I Phosphatidylinositol 3-Kinases , Clot Retraction/drug effects , Clot Retraction/physiology , Cytoskeleton/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Stress, Mechanical , Thrombin/pharmacology , Thromboxane A2/metabolism
20.
Nat Med ; 15(6): 665-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465929

ABSTRACT

Platelet aggregation at sites of vascular injury is essential for hemostasis and arterial thrombosis. It has long been assumed that platelet aggregation and thrombus growth are initiated by soluble agonists generated at sites of vascular injury. By using high-resolution intravital imaging techniques and hydrodynamic analyses, we show that platelet aggregation is primarily driven by changes in blood flow parameters (rheology), with soluble agonists having a secondary role, stabilizing formed aggregates. We find that in response to vascular injury, thrombi initially develop through the progressive stabilization of discoid platelet aggregates. Analysis of blood flow dynamics revealed that discoid platelets preferentially adhere in low-shear zones at the downstream face of forming thrombi, with stabilization of aggregates dependent on the dynamic restructuring of membrane tethers. These findings provide insight into the prothrombotic effects of disturbed blood flow parameters and suggest a fundamental reinterpretation of the mechanisms driving platelet aggregation and thrombus growth.


Subject(s)
Platelet Aggregation , Thrombosis/pathology , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Adhesion , Hemodynamics , Image Processing, Computer-Assisted , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...