Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 5078, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35332242

RESUMO

Microscale bots intended for targeted drug delivery must move through three-dimensional (3D) environments that include bifurcations, inclined surfaces, and curvature. In previous studies, we have shown that magnetically actuated colloidal microwheels (µwheels) reversibly assembled from superparamagnetic beads can translate rapidly and be readily directed. Here we show that, at high concentrations, µwheels assemble into swarms that, depending on applied magnetic field actuation patterns, can be designed to transport cargo, climb steep inclines, spread over large areas, or provide mechanical action. We test the ability of these multimodal swarms to navigate through complex, inclined microenvironments by characterizing the translation and dispersion of individual µwheels and swarms of µwheels on steeply inclined and flat surfaces. Swarms are then studied within branching 3D vascular models with multiple turns where good targeting efficiencies are achieved over centimeter length scales. With this approach, we present a readily reconfigurable swarm platform capable of navigating through 3D microenvironments.


Assuntos
Sistemas de Liberação de Medicamentos , Campos Magnéticos
2.
Rev Sci Instrum ; 91(9): 093701, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-33003773

RESUMO

Superparamagnetic colloidal particles can be reversibly assembled into wheel-like structures called microwheels (µwheels), which roll on surfaces due to friction and can be driven at user-controlled speeds and directions using rotating magnetic fields. Here, we describe the hardware and software to create and control the magnetic fields that assemble and direct µwheel motion and the optics to visualize them. Motivated by portability, adaptability, and low-cost, an extruded aluminum heat-dissipating frame incorporating open optics and audio speaker coils outfitted with high magnetic permeability cores was constructed. Open-source software was developed to define the magnitude, frequency, and orientation of the magnetic field, allowing for real-time joystick control of µwheels through two-dimensional (2D) and three-dimensional (3D) fluidic environments. With this combination of hardware and software, µwheels translate at speeds up to 50 µm/s through sample sizes up to 5 × 5 × 5 cm3 using 0.75 mT-2.5 mT magnetic fields with rotation frequencies of 5 Hz-40 Hz. Heat dissipation by aluminum coil clamps maintained sample temperatures within 3 °C of ambient temperature, a range conducive for biological applications. With this design, µwheels can be manipulated and imaged in 2D and 3D networks at length scales of micrometers to centimeters.

3.
J Thromb Haemost ; 16(1): 104-115, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29064615

RESUMO

Essentials von Willebrand factor (VWF) function is shear stress dependent. Platelet accumulation in a microfluidic assay correlates with VWF levels. The microfluidic assay discriminates type 1 von Willebrand disease from healthy controls. The microfluidic flow assay detects responses to therapeutic intervention (DDAVP). SUMMARY: Background von Willebrand disease (VWD) is a mucocutaneous bleeding disorder with a reported prevalence of 1 in 10 000. von Willebrand factor (VWF) function and platelet adhesion are regulated by hemodynamic forces that are not integrated into most current clinical assays. Objective We evaluated whether a custom microfluidic flow assay (MFA) can screen for deficiencies in VWF in patients presenting with mucocutaneous bleeding. Methods Whole blood from individuals with mucocutaneous bleeding was assayed in a custom MFA. Results Thirty-two patients with type 1 VWD (10/32) or reported mucocutaneous bleeding were enrolled. The platelet adhesion velocity (r = 0.5978 for 750 s-1 and 0.6895 for 1500 s-1 ) and the maximum platelet surface area coverage (r = 0.5719 for 750 s-1 and 0.6633 for 1500 s-1 ) in the MFA correlated with VWF levels. Furthermore, the platelet adhesion velocity at 750 s-1 (type 1 VWD, mean 0.0009761, 95% confidence interval [CI] 0.0003404-0.001612; control, mean 0.003587, 95% CI 0.002455-0.004719) and at 1500 s-1 (type 1 VWD, mean 0.0003585, 95% CI 0.00003914-0.0006778; control, mean 0.003132, 95% CI 0.001565-0.004699) differentiated type 1 VWD from controls. Maximum platelet surface area coverage at 750 s-1 (type 1 VWD, mean 0.1831, 95% CI 0.03816-0.3281; control, mean 0.6755, 95% CI 0.471-0.88) and at 1500 s-1 (type 1 VWD, mean 0.07873, 95% CI 0.01689-0.1406; control, mean 0.6432, 95% CI 0.3607-0.9257) also differentiated type 1 VWD from controls. We also observed an improvement in platelet accumulation after 1-desamino-8-d-arginine vasopressin (DDAVP) treatment at 1500 s-1 (pre-DDAVP, mean 0.4784, 95% CI 0.1777-0.7791; post-DDAVP, mean 0.8444, 95% CI 0.7162-0.9726). Conclusions These data suggest that this approach can be used as a screening tool for VWD.


Assuntos
Plaquetas/metabolismo , Hemorreologia , Técnicas Analíticas Microfluídicas , Adesividade Plaquetária , Agregação Plaquetária , Testes de Função Plaquetária/métodos , Doença de von Willebrand Tipo 1/diagnóstico , Fator de von Willebrand/análise , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Velocidade do Fluxo Sanguíneo , Plaquetas/efeitos dos fármacos , Estudos de Casos e Controles , Criança , Pré-Escolar , Desamino Arginina Vasopressina/uso terapêutico , Diagnóstico Diferencial , Regulação para Baixo , Hemorreologia/efeitos dos fármacos , Hemostáticos/uso terapêutico , Humanos , Pessoa de Meia-Idade , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Valor Preditivo dos Testes , Fluxo Sanguíneo Regional , Estresse Mecânico , Resultado do Tratamento , Adulto Jovem , Doença de von Willebrand Tipo 1/sangue , Doença de von Willebrand Tipo 1/tratamento farmacológico , Doença de von Willebrand Tipo 1/fisiopatologia
4.
J Thromb Haemost ; 16(2): 352-363, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29045015

RESUMO

Essentials Signaling by Gas6 through Tyro3/Axl/Mer receptors is essential for stable platelet aggregation. UNC2025 is a small molecule inhibitor of the Mer tyrosine kinase. UNC2025 decreases platelet activation in vitro and thrombus formation in vivo. UNC2025's anti-platelet effect is synergistic with inhibition of the ADP receptor, P2Y12 . SUMMARY: Background Growth arrest-specific protein 6 signals through the TAM (TYRO-3-AXL-MERTK) receptor family, mediating platelet activation and thrombus formation via activation of the aggregate-stabilizing αIIb ß3 integrin. Objective To describe the antithrombotic effects mediated by UNC2025, a small-molecule MERTK tyrosine kinase inhibitor. Methods MERTK phosphorylation and downstream signaling were assessed by immunoblotting. Light transmission aggregometry, flow cytometry and microfluidic analysis were used to evaluate the impact of MERTK inhibition on platelet activation and stability of aggregates in vitro. The effects of MERTK inhibition on arterial and venous thrombosis, platelet accumulation at microvascular injury sites and tail bleeding times were determined with murine models. The effects of combined treatment with ADP-P2Y1&12 pathway antagonists and UNC2025 were also evaluated. Results and Conclusions Treatment with UNC2025 inhibited MERTK phosphorylation and downstream activation of AKT and SRC, decreased platelet activation, and protected animals from pulmonary embolism and arterial thrombosis without increasing bleeding times. The antiplatelet effect of UNC2025 was enhanced in combination with ADP-P2Y1&12 pathway antagonists, and a greater than additive effect was observed when these two agents with different mechanisms of inhibition were coadministered. TAM kinase signaling represents a potential therapeutic target, as inhibition of this axis, especially in combination with ADP-P2Y pathway antagonism, mediates decreased platelet activation, aggregate stability, and thrombus formation, with less hemorrhagic potential than current treatment strategies. The data presented here also demonstrate antithrombotic activity mediated by UNC2025, a novel translational agent, and support the development of TAM kinase inhibitors for clinical applications.


Assuntos
Adenina/análogos & derivados , Plaquetas/efeitos dos fármacos , Piperazinas/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Embolia Pulmonar/prevenção & controle , Trombose/prevenção & controle , c-Mer Tirosina Quinase/antagonistas & inibidores , Adenina/farmacocinética , Adenina/farmacologia , Animais , Plaquetas/enzimologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Piperazinas/farmacocinética , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacocinética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Proto-Oncogênicas/metabolismo , Embolia Pulmonar/sangue , Embolia Pulmonar/enzimologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombose/sangue , Trombose/enzimologia , c-Mer Tirosina Quinase/metabolismo , Receptor Tirosina Quinase Axl
5.
Cell Mol Bioeng ; 10(1): 3-15, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28529666

RESUMO

Hemostasis is the process of sealing a vascular injury with a thrombus to arrest bleeding. The type of thrombus that forms depends on the nature of the injury and hemodynamics. There are many models of intravascular thrombus formation whereby blood is exposed to prothrombotic molecules on a solid substrate. However, there are few models of extravascular thrombus formation whereby blood escapes into the extravascular space through a hole in the vessel wall. Here, we describe a microfluidic model of hemostasis that includes vascular, vessel wall, and extravascular compartments. Type I collagen and tissue factor, which support platelet adhesion and initiate coagulation, respectively, were adsorbed to the wall of the injury channel and act synergistically to yield a stable thrombus that stops blood loss into the extravascular compartment in ~7.5 min. Inhibiting factor VIII to mimic hemophilia A results in an unstable thrombus that was unable to close the injury. Treatment with a P2Y12 antagonist to reduce platelet activation prolonged the closure time two-fold compared to controls. Taken together, these data demonstrate a hemostatic model that is sensitive to both coagulation and platelet function and can be used to study coagulopathies and platelet dysfunction that result in excessive blood loss.

7.
Nat Commun ; 7: 10225, 2016 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-26725747

RESUMO

Propulsion at the microscale requires unique strategies such as the undulating or rotating filaments that microorganisms have evolved to swim. These features however can be difficult to artificially replicate and control, limiting the ability to actuate and direct engineered microdevices to targeted locations within practical timeframes. An alternative propulsion strategy to swimming is rolling. Here we report that low-strength magnetic fields can reversibly assemble wheel-shaped devices in situ from individual colloidal building blocks and also drive, rotate and direct them along surfaces at velocities faster than most other microscale propulsion schemes. By varying spin frequency and angle relative to the surface, we demonstrate that microwheels can be directed rapidly and precisely along user-defined paths. Such in situ assembly of readily modified colloidal devices capable of targeted movements provides a practical transport and delivery tool for microscale applications, especially those in complex or tortuous geometries.


Assuntos
Coloides/química , Campos Magnéticos , Movimento/fisiologia , Nanoestruturas/química
8.
Integr Biol (Camb) ; 7(7): 801-14, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26087748

RESUMO

Endothelial cells (EC) both inhibit and promote platelet function depending on their activation state. Quiescent EC inhibit platelet activation by constitutive secretion of platelet inhibitors. Activated EC promote platelet adhesion by secretion of von Willebrand factor (vWF). EC also secrete an extracellular matrix that support platelet adhesion when exposed following vascular injury. Previous studies of EC-platelet interactions under flow activate entire monolayers of cells by chemical activation. In this study, EC cultured in microfluidic channels were focally activated by heat from an underlying microelectrode. Based on finite element modeling, microelectrodes induced peak temperature increases of 10-40 °C above 37 °C after applying 5-9 V for 30 s resulting in three zones: (1) a quiescent zone corresponded to peak temperatures of less than 15 °C characterized by no EC activation or platelet accumulation. (2) An activation zone corresponding to an increase of 16-22 °C yielded EC that were viable, secreted elevated levels of vWF, and were P-selectin positive. Platelets accumulated in the retracted spaces between EC in the activation zone at a wall shear rate of 150 and 1500 s(-1). Experiments with blocking antibodies show that platelets adhere via GPIbα-vWF and α6ß1-laminin interactions. (3) A kill zone corresponded to peak temperatures of greater than 23 °C where EC were not viable and did not support platelet adhesion. These data define heating conditions for the activation of EC, causing the secretion of vWF and the exposure of a subendothelial matrix that support platelet adhesion and aggregation. This model provides for spatially defined zones of EC activation that could be a useful tool for measuring the relative roles of anti- and prothrombotic roles of EC at the site of vascular injury.


Assuntos
Células Endoteliais/fisiologia , Calefação/instrumentação , Calefação/métodos , Adesividade Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Lesões do Sistema Vascular/fisiopatologia , Células Cultivadas , Eletrodos , Células Endoteliais/citologia , Humanos , Lesões do Sistema Vascular/patologia
9.
Biomed Opt Express ; 6(3): 807-14, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25798305

RESUMO

We present a simplified approach for imaging a linear diode bar laser for application as an optical stretcher within a microfluidic geometry. We have recently shown that these linear sources can be used to measure cell mechanical properties; however, the source geometry creates imaging challenges. To minimize intensity losses and simplify implementation within microfluidic systems without the use of expensive objectives, we combine aspheric and cylindrical lenses to create a 1:1 image of the source at the stretcher focal plane and demonstrate effectiveness by measuring the deformation of human red blood cells and neutrophils.

12.
J Thromb Haemost ; 12(3): 373-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24345079

RESUMO

BACKGROUND: Blood flow regulates coagulation and fibrin assembly by controlling the rate of transport of zymogens, enzymes and plasma proteins to and from the site of an injury. OBJECTIVE: The objective of this work was to define the hemodynamic conditions under which fibrin can form under flow on tissue factor (TF)-rich substrates. METHODS: TF-coated silica beads (~ 800 nm) were patterned into 18-85-µm spots. Normal pooled plasma and factors VIII, IX and XI deficient plasmas were perfused over the beads coated with 0.08, 0.8 and 8 molecules-TF µm(-2) at shear rates of 50-1000 s(-1) . Fibrin deposition and thrombin generation were measured by fluorescence microscopy in a hydrodynamic focusing microfluidic device. RESULTS AND CONCLUSIONS: Fibrin deposition was supported on patterned bead spots, but not planar TF substrates at the same surface TF concentration. There was a threshold spot size and a shear rate dependent TF concentration that was necessary to support fibrin polymerization. FVIII and FIX had minor effects on fibrin dynamics at 8 molecules-TF µm(-2) , but were essential at 0.8 molecules-TF µm(-2) . The absence of FXI influenced thrombin generation and fibrin deposition at both 0.8 and 8 molecules-TF µm(-2) . These results show that fibrin deposition requires perturbations in the flow field that protect reactions from dilution by flow under venous and arterial conditions. FVIII and FIX have a modest effect on fibrin deposition at high TF concentrations, but are necessary for fibrin deposition at low TF concentrations. FXI amplifies thrombin generation under flow at both low and high TF concentrations.


Assuntos
Biomimética/métodos , Coagulação Sanguínea , Fibrina/química , Trombina/química , Adesão Celular , Precursores Enzimáticos/química , Produtos de Degradação da Fibrina e do Fibrinogênio/química , Hemodinâmica , Humanos , Hidrodinâmica , Processamento de Imagem Assistida por Computador , Bicamadas Lipídicas/química , Técnicas Analíticas Microfluídicas , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Resistência ao Cisalhamento , Dióxido de Silício/química , Tromboplastina/química
13.
Ann Biomed Eng ; 41(10): 2193-205, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23563992

RESUMO

Nitric oxide (NO) inhibits platelet aggregation at and near the site of a vascular injury by upregulation of cyclic guanosine monophosphate, which reduces the dimerization of the integrin α(IIb)ß3. The magnitude of NO flux from the vessel wall and the NO concentration that is necessary to inhibit platelet aggregation under physiological flow conditions is unknown. In this study, a NO releasing polymer, diazeniumdiolated dibutylhexanediamine, was integrated into a microfluidic flow assay to determine the relationship between NO wall flux and collagen mediated platelet adhesion, activation and aggregation. A NO flux equal to or greater than 2.5 × 10⁻¹° mol cm⁻² min⁻¹ was found to abrogate aggregation, but not initial platelet adhesion, on collagen at 200 and 500 s⁻¹ as effectively as the α(IIb)ß3 antagonist abciximab. The dynamic range of NO fluxes found to induce measurable inhibition of platelet aggregation spanned from 0.33 × 10⁻¹° to 2.5 × 10⁻¹° mol cm⁻² min⁻¹ at 200-500 s⁻¹. These fluxes correspond to near-wall NO concentrations of 3-90 nM based on a computational model of NO transport. The model predicts that NO concentration in the platelet rich layer near the wall is kinetically limited, while NO penetration into the lumen is mass transfer limited.


Assuntos
Plaquetas/metabolismo , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Óxido Nítrico/farmacocinética , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Compostos Azo/farmacocinética , Compostos Azo/farmacologia , Transporte Biológico/fisiologia , Velocidade do Fluxo Sanguíneo , Plaquetas/citologia , Feminino , Humanos , Masculino , Óxido Nítrico/farmacologia
14.
Biophys J ; 104(8): 1812-23, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23601328

RESUMO

Interstitial fluid flow within blood clots is a biophysical mechanism that regulates clot growth and dissolution. Assuming that a clot can be modeled as a porous medium, the physical property that dictates interstitial fluid flow is the hydraulic permeability. The objective of this study was to bound the possible values of the hydraulic permeability in clots formed in vivo and present relationships that can be used to estimate clot permeability as a function of composition. A series of clots with known densities of fibrin and platelets, the two major components of a clot, were formed under static conditions. The permeability was calculated by measuring the interstitial fluid velocity through the clots at a constant pressure gradient. Fibrin gels formed with a fiber volume fraction of 0.02-0.54 had permeabilities of 1.2 × 10(-1)-1.5 × 10(-4)µm(2). Platelet-rich clots with a platelet volume fraction of 0.01-0.61 and a fibrin volume fraction of 0.03 had permeabilities over a range of 1.1 × 10(-2)-1.5 × 10(-5)µm(2). The permeability of fibrin gels and of clots with platelet volume fraction of <0.2 were modeled as an array of disordered cylinders with uniform diameters. Clots with a platelet volume fraction of >0.2 were modeled as a Brinkman medium of coarse solids (platelets) embedded in a mesh of fine fibers (fibrin). Our data suggest that the permeability of clots formed in vivo can vary by up to five orders of magnitude, with pore sizes that range from 4 to 350 nm. These findings have important implications for the transport of coagulation zymogens/enzymes in the interstitial spaces during clot formation, as well as the design of fibrinolytic drug delivery strategies.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fibrina/metabolismo , Líquido Extracelular/metabolismo , Fibrina/química , Humanos , Cinética , Modelos Biológicos , Permeabilidade , Polimerização
15.
Biophys J ; 103(1): 137-45, 2012 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-22828340

RESUMO

Single-molecule force spectroscopy is used to probe the kinetics of receptor-ligand bonds by applying mechanical forces to an intermediate media on which the molecules reside. When this intermediate media is a live cell, the viscoelastic properties can affect the calculation of rate constants. We theoretically investigate the effect of media viscoelasticity on the common assumption that the bond force is equal to the instantaneous applied force. Dynamic force spectroscopy is simulated between two cells of varying micromechanical properties adhered by a single bond with a constant kinetic off-rate. We show that cell and microvilli deformation, and hydrodynamic drag contribute to bond forces that can be 28-90% lower than the applied force for loading rates of 10(3)-10(7) pN/s, resulting in longer bond lifetimes. These longer bond lifetimes are not caused by changes in bond kinetics; rather, they are due to the mechanical response of the intermediate media on which the bonds reside. Under the assumption that the instantaneous bond force is equal to the applied force--thereby ignoring viscoelasticity--leads to 14-39% error in the determination of off-rates. We present an approach that incorporates viscoelastic properties in calculating the instantaneous bond force and kinetic dissociation parameter of the intermolecular bond.


Assuntos
Microvilosidades/química , Espectrofotometria/métodos , Elasticidade , Hidrodinâmica , Microvilosidades/ultraestrutura , Simulação de Dinâmica Molecular , Viscosidade
16.
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
17.
J Thromb Haemost ; 6(12): 2193-201, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18983510

RESUMO

BACKGROUND: Flow chambers allow the ex vivo study of platelet response to defined surfaces at controlled wall shear stresses. However, most assays require 1-10 mL of blood and are poorly suited for murine whole blood experiments. OBJECTIVE: To measure murine platelet deposition and stability in response to focal zones of prothrombotic stimuli using 100 microL of whole blood and controlled flow exposure. METHODS: Microfluidic methods were used for patterning acid-soluble collagen in 100 microm x 100 microm patches and creating flow channels with a volume of 150 nL. Within 1 min of collection into PPACK and fluorescent anti-mouse CD41 mAb, whole blood from normal mice or from mice deficient in the integrin alpha(2) subunit was perfused for 5 min over the patterned collagen. Platelet accumulation was measured at venous and arterial wall shear rates. After 5 min, thrombus stability was measured with a 'shear step-up' to 8000 s(-1). RESULTS: Wild-type murine platelets adhered and aggregated on collagen in a biphasic shear-dependent manner with increased deposition from 100 to 400 s(-1), but decreased deposition at 1000 s(-1). Adhesion to patterned collagen was severely diminished for platelets lacking a functional alpha(2)beta(1) integrin. Those integrin alpha(2)-deficient platelets that did adhere were removed from the surface when challenged to shear step-up. PAR4 agonist (AYPGKF) treatment of the thrombus at 5 min enhanced aggregate stability during the shear step-up. CONCLUSIONS: PAR4 signaling enhances aggregate stability by mechanisms independent of other thrombin-dependent pathways such as fibrin formation.


Assuntos
Microfluídica , Adesividade Plaquetária , Receptores de Trombina/fisiologia , Trombose/patologia , Animais , Colágeno/metabolismo , Modelos Animais de Doenças , Integrina alfa2beta1 , Camundongos , Agregação Plaquetária , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transdução de Sinais , Estresse Mecânico
18.
J Control Release ; 111(3): 252-62, 2006 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-16476500

RESUMO

Convection enhanced drug delivery (CED) is a promising therapeutic method for treating diseases of the brain by enhancing the penetration of drugs. Most controlled release delivery methods rely on diffusion from a source to transport drugs throughout tissue. CED relies on direct infusion of drugs into tissue at a sufficiently high rate so that convective transport of drug is at least as important as diffusive transport. This work describes the fabrication and characterization of microfluidic probes for CED protocols and the role diffusion plays in determining penetration. Microfluidic channels were formed on silicon substrates by employing a sacrificial photoresist layer encased in a parylene structural layer. Flow in the microchannels was characterized by applying constant upstream pressures from 35 to 310 kPa, which resulted in flow rates of 0.5-4.5 microL/min. The devices were used to infuse Evans Blue and albumin in hydrogel brain phantoms. The results of these infusions were compared to a simple convection-diffusion model for infusions into porous media. In vivo infusions of albumin were performed in the gray matter of rats at upstream pressures of 35, 70, and 140 kPa. The microfabricated probes show reduced evidence of backflow along the device-tissue interface when compared with conventional needles used for CED.


Assuntos
Sistemas de Liberação de Medicamentos , Microfluídica/instrumentação , Preparações Farmacêuticas/administração & dosagem , Albuminas/administração & dosagem , Animais , Núcleo Caudado/metabolismo , Convecção , Difusão , Desenho de Equipamento , Azul Evans/administração & dosagem , Infusões Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley , Reologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...