Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Hematol ; 99(6): 1103-1107, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38572662

RESUMEN

Hyperleukocytosis is an emergency of acute leukemia leading to blood hyperviscosity, potentially resulting in life-threatening microvascular obstruction, or leukostasis. Due to the high number of red cells in the circulation, hematocrit/hemoglobin levels (Hct/Hgb) are major drivers of blood viscosity, but how Hct/Hgb mediates hyperviscosity in acute leukemia remains unknown. In vivo hemorheological studies are difficult to conduct and interpret due to issues related to visualizing and manipulating the microvasculature. To that end, a multi-vessel microfluidic device recapitulating the size-scale and geometry of the microvasculature was designed to investigate how Hct/Hgb interacts with acute leukemia to induce "in vitro" leukostasis. Using patient samples and cell lines, the degree of leukostasis was different among leukemia immunophenotypes with respect to white blood cell (WBC) count and Hct/Hgb. Among lymphoid immunophenotypes, severe anemia is protective against in vitro leukostasis and Hct/Hgb thresholds became apparent above which in vitro leukostasis significantly increased, to a greater extent with B-cell acute lymphoblastic leukemia (ALL) versus T-cell ALL. In vitro leukostasis in acute myeloid leukemia was primarily driven by WBC with little interaction with Hct/Hgb. This sets the stage for prospective clinical studies assessing how red cell transfusion may affect leukostasis risk in immunophenotypically different acute leukemia patients.


Asunto(s)
Viscosidad Sanguínea , Transfusión de Eritrocitos , Humanos , Microvasos , Leucostasis/etiología , Hematócrito , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/sangre , Femenino , Masculino , Hemoglobinas/análisis
2.
Nat Mater ; 16(2): 230-235, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27723740

RESUMEN

Haemostasis occurs at sites of vascular injury, where flowing blood forms a clot, a dynamic and heterogeneous fibrin-based biomaterial. Paramount in the clot's capability to stem haemorrhage are its changing mechanical properties, the major drivers of which are the contractile forces exerted by platelets against the fibrin scaffold. However, how platelets transduce microenvironmental cues to mediate contraction and alter clot mechanics is unknown. This is clinically relevant, as overly softened and stiffened clots are associated with bleeding and thrombotic disorders. Here, we report a high-throughput hydrogel-based platelet-contraction cytometer that quantifies single-platelet contraction forces in different clot microenvironments. We also show that platelets, via the Rho/ROCK pathway, synergistically couple mechanical and biochemical inputs to mediate contraction. Moreover, highly contractile platelet subpopulations present in healthy controls are conspicuously absent in a subset of patients with undiagnosed bleeding disorders, and therefore may function as a clinical diagnostic biophysical biomarker.


Asunto(s)
Coagulación Sanguínea/fisiología , Velocidad del Flujo Sanguíneo/fisiología , Plaquetas/fisiología , Citometría de Flujo/métodos , Mecanotransducción Celular/fisiología , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Células Cultivadas , Módulo de Elasticidad/fisiología , Dureza/fisiología , Humanos , Nanopartículas/química
3.
Mol Ther ; 25(10): 2372-2382, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28780274

RESUMEN

Ex vivo gene therapy using lentiviral vectors (LVs) is a proven approach to treat and potentially cure many hematologic disorders and malignancies but remains stymied by cumbersome, cost-prohibitive, and scale-limited production processes that cannot meet the demands of current clinical protocols for widespread clinical utilization. However, limitations in LV manufacture coupled with inefficient transduction protocols requiring significant excess amounts of vector currently limit widespread implementation. Herein, we describe a microfluidic, mass transport-based approach that overcomes the diffusion limitations of current transduction platforms to enhance LV gene transfer kinetics and efficiency. This novel ex vivo LV transduction platform is flexible in design, easy to use, scalable, and compatible with standard cell transduction reagents and LV preparations. Using hematopoietic cell lines, primary human T cells, primary hematopoietic stem and progenitor cells (HSPCs) of both murine (Sca-1+) and human (CD34+) origin, microfluidic transduction using clinically processed LVs occurs up to 5-fold faster and requires as little as one-twentieth of LV. As an in vivo validation of the microfluidic-based transduction technology, HSPC gene therapy was performed in hemophilia A mice using limiting amounts of LV. Compared to the standard static well-based transduction protocols, only animals transplanted with microfluidic-transduced cells displayed clotting levels restored to normal.


Asunto(s)
Microfluídica/métodos , Animales , Línea Celular , Células Cultivadas , Terapia Genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Lentivirus/genética , Ratones , Transducción Genética
4.
Blood ; 126(4): 531-8, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25964667

RESUMEN

Although the biology of platelet adhesion on subendothelial matrix after vascular injury is well characterized, how the matrix biophysical properties affect platelet physiology is unknown. Here we demonstrate that geometric orientation of the matrix itself regulates platelet α-granule secretion, a key component of platelet activation. Using protein microcontact printing, we show that platelets spread beyond the geometric constraints of fibrinogen or collagen micropatterns with <5-µm features. Interestingly, α-granule exocytosis and deposition of the α-granule contents such as fibrinogen and fibronectin were primarily observed in those areas of platelet extension beyond the matrix protein micropatterns. This enables platelets to "self-deposit" additional matrix, provide more cellular membrane to extend spreading, and reinforce platelet-platelet connections. Mechanistically, this phenomenon is mediated by actin polymerization, Rac1 activation, and αIIbß3 integrin redistribution and activation, and is attenuated in gray platelet syndrome platelets, which lack α-granules, and Wiskott-Aldrich syndrome platelets, which have cytoskeletal defects. Overall, these studies demonstrate how platelets transduce geometric cues of the underlying matrix geometry into intracellular signals to extend spreading, which endows platelets spatial flexibility when spreading onto small sites of exposed subendothelium.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Exocitosis/fisiología , Síndrome de Plaquetas Grises/patología , Adhesividad Plaquetaria/fisiología , Síndrome de Wiskott-Aldrich/patología , Citoesqueleto de Actina/metabolismo , Estudios de Casos y Controles , Membrana Celular/metabolismo , Células Cultivadas , Fibrinógeno/metabolismo , Fibronectinas/metabolismo , Síndrome de Plaquetas Grises/metabolismo , Humanos , Técnicas para Inmunoenzimas , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Seudópodos , Síndrome de Wiskott-Aldrich/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(40): 14430-5, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246564

RESUMEN

As platelets aggregate and activate at the site of vascular injury to stem bleeding, they are subjected to a myriad of biochemical and biophysical signals and cues. As clot formation ensues, platelets interact with polymerizing fibrin scaffolds, exposing platelets to a large range of mechanical microenvironments. Here, we show for the first time (to our knowledge) that platelets, which are anucleate cellular fragments, sense microenvironmental mechanical properties, such as substrate stiffness, and transduce those cues into differential biological signals. Specifically, as platelets mechanosense the stiffness of the underlying fibrin/fibrinogen substrate, increasing substrate stiffness leads to increased platelet adhesion and spreading. Importantly, adhesion on stiffer substrates also leads to higher levels of platelet activation, as measured by integrin αIIbß3 activation, α-granule secretion, and procoagulant activity. Mechanistically, we determined that Rac1 and actomyosin activity mediate substrate stiffness-dependent platelet adhesion, spreading, and activation to different degrees. This capability of platelets to mechanosense microenvironmental cues in a growing thrombus or hemostatic plug and then mechanotransduce those cues into differential levels of platelet adhesion, spreading, and activation provides biophysical insight into the underlying mechanisms of platelet aggregation and platelet activation heterogeneity during thrombus formation.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/citología , Movimiento Celular/fisiología , Mecanotransducción Celular/fisiología , Activación Plaquetaria/fisiología , Adhesividad Plaquetaria/fisiología , Resinas Acrílicas/metabolismo , Plaquetas/metabolismo , Microambiente Celular/fisiología , Fibrina/metabolismo , Fibrinógeno/metabolismo , Humanos , Proteínas Inmovilizadas/metabolismo , Microscopía Confocal , Selectina-P/metabolismo , Fosfatidilserinas/metabolismo , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Estrés Mecánico , Trombosis/fisiopatología , Proteína de Unión al GTP rac1/metabolismo
6.
Blood ; 122(10): 1695-706, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23881916

RESUMEN

Megakaryocytes generate platelets through extensive reorganization of the cytoskeleton and plasma membrane. Cdc42 interacting protein 4 (CIP4) is an F-BAR protein that localizes to membrane phospholipids through its BAR domain and interacts with Wiskott-Aldrich Syndrome Protein (WASP) via its SRC homology 3 domain. F-BAR proteins promote actin polymerization and membrane tubulation. To study its function, we generated CIP4-null mice that displayed thrombocytopenia similar to that of WAS(-) mice. The number of megakaryocytes and their progenitors was not affected. However, the number of proplatelet protrusions was reduced in CIP4-null, but not WAS(-), megakaryocytes. Electron micrographs of CIP4-null megakaryocytes showed an altered demarcation membrane system. Silencing of CIP4, not WASP, expression resulted in fewer proplatelet-like extensions. Fluorescence anisotropy studies showed that loss of CIP4 resulted in a more rigid membrane. Micropipette aspiration demonstrated decreased cortical actin tension in megakaryocytic cells with reduced CIP4 or WASP protein. These studies support a new biophysical mechanism for platelet biogenesis whereby CIP4 enhances the complex, dynamic reorganization of the plasma membrane (WASP independent) and actin cortex network (as known for WASP and cortical actin) to reduce the work required for generating proplatelets. CIP4 is a new component in the highly coordinated system of megakaryocytic membrane and cytoskeletal remodeling affecting platelet production.


Asunto(s)
Plaquetas/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Fenómenos Biomecánicos , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Megacariocitos/ultraestructura , Fluidez de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Antígenos de Histocompatibilidad Menor , Ploidias , Transporte de Proteínas , Células Madre/metabolismo , Células Madre/patología , Trombocitopenia/metabolismo , Trombocitopenia/patología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo
7.
J Biomech Eng ; 136(7)2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24441718

RESUMEN

Stroke is the most devastating complication after ventricular assist device (VAD) implantation, with an incidence of 14%-47% despite improvements in device design and anticoagulation. This complication continues to limit the widespread implementation of VAD therapy. Patient-specific computational fluid dynamics (CFD) analysis may elucidate ways to reduce this risk. A patient-specific three-dimensional model of the aortic arch was generated from computed tomography. A 12 mm VAD outflow-graft (VAD-OG) "anastomosed" to the aorta was rendered. CFD was applied to study blood flow patterns. Particle tracks, originating from the VAD, were computed with a Lagrangian phase model and percentage of particles entering the cerebral vessels was calculated. Twelve implantation configurations of the VAD-OG and three particle sizes (2, 4, and 5 mm) were considered. Percentage of particles entering the cerebral vessels ranged from 6% for the descending aorta VAD-OG anastomosis, to 14% for the ascending aorta at 90 deg VAD-OG anastomosis. Values were significantly different among all configurations (X(2) = 3925, p < 0.0001). Shallower and more cephalad anastomoses prevented formation of zones of recirculation in the ascending aorta. In this computational model and within the range of anatomic parameters considered, the percentage of particles entering the cerebral vessels from a VAD-OG is reduced by nearly 60% by optimizing outflow-graft configuration. Ascending aorta recirculation zones, which may be thrombogenic, can also be eliminated. CFD methods coupled with patient-specific anatomy may aid in identifying the optimal location and angle for VAD-OG anastomosis to minimize stroke risk.


Asunto(s)
Vasos Sanguíneos , Corazón Auxiliar/efectos adversos , Embolia Intracraneal/etiología , Embolia Intracraneal/prevención & control , Modelación Específica para el Paciente , Prótesis e Implantes , Aorta Torácica/fisiopatología , Vasos Sanguíneos/fisiopatología , Humanos , Hidrodinámica , Tamaño de la Partícula , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control
8.
bioRxiv ; 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38644993

RESUMEN

Multiple myeloma (MM), a cancer of bone marrow plasma cells, is the second-most common hematological malignancy. However, despite immunotherapies like chimeric antigen receptor (CAR)-T cells, relapse is nearly universal. The bone marrow (BM) microenvironment influences how MM cells survive, proliferate, and resist treatment. Yet, it is unclear which BM niches give rise to MM pathophysiology. Here, we present a 3D microvascularized culture system, which models the endosteal and perivascular bone marrow niches, allowing us to study MM-stroma interactions in the BM niche and model responses to therapeutic CAR-T cells. We demonstrated the prolonged survival of cell line-based and patient-derived multiple myeloma cells within our in vitro system and successfully flowed in donor-matched CAR-T cells. We then measured T cell survival, differentiation, and cytotoxicity against MM cells using a variety of analysis techniques. Our MM-on-a-chip system could elucidate the role of the BM microenvironment in MM survival and therapeutic evasion and inform the rational design of next-generation therapeutics. TEASER: A multiple myeloma model can study why the disease is still challenging to treat despite options that work well in other cancers.

9.
J Cell Mol Med ; 17(5): 579-96, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23490277

RESUMEN

Although the processes of haemostasis and thrombosis have been studied extensively in the past several decades, much of the effort has been spent characterizing the biological and biochemical aspects of clotting. More recently, researchers have discovered that the function and physiology of blood cells and plasma proteins relevant in haematologic processes are mechanically, as well as biologically, regulated. This is not entirely surprising considering the extremely dynamic fluidic environment that these blood components exist in. Other cells in the body such as fibroblasts and endothelial cells have been found to biologically respond to their physical and mechanical environments, affecting aspects of cellular physiology as diverse as cytoskeletal architecture to gene expression to alterations of vital signalling pathways. In the circulation, blood cells and plasma proteins are constantly exposed to forces while they, in turn, also exert forces to regulate clot formation. These mechanical factors lead to biochemical and biomechanical changes on the macro- to molecular scale. Likewise, biochemical and biomechanical alterations in the microenvironment can ultimately impact the mechanical regulation of clot formation. The ways in which these factors all balance each other can be the difference between haemostasis and thrombosis. Here, we review how the biomechanics of blood cells intimately interact with the cellular and molecular biology to regulate haemostasis and thrombosis in the context of health and disease from the macro- to molecular scale. We will also show how these biomechanical forces in the context of haemostasis and thrombosis have been replicated or measured in vitro.


Asunto(s)
Salud , Hemostasis , Trombosis/fisiopatología , Animales , Fenómenos Biomecánicos , Eritrocitos/metabolismo , Humanos , Trombosis/sangre
10.
Methods Mol Biol ; 2097: 55-65, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31776918

RESUMEN

Lentiviral vectors enable gene transfer into target cells, but manufacturing is complex, scale-limited, and costly. Here, we describe the use of microfluidic devices for efficient ex vivo gene transfer. Up to four- to fivefold reductions in viral vector usage and two- to fourfold reductions in transduction times can be obtained by using this method.


Asunto(s)
Microfluídica/métodos , Transducción Genética , Virus/metabolismo , Adsorción , Dimetilpolisiloxanos/química , Fibronectinas , Células HEK293 , Humanos , Células Jurkat , Proteínas Recombinantes , Silicio/química
11.
Nat Biomed Eng ; 2: 453-463, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30533277

RESUMEN

Alterations in the mechanical properties of erythrocytes occurring in inflammatory and hematologic disorders such as sickle cell disease (SCD) and malaria often lead to increased endothelial permeability, haemolysis, and microvascular obstruction. However, the associations among these pathological phenomena remain unknown. Here, we report a perfusable, endothelialized microvasculature-on-a-chip featuring an interpenetrating-polymer-network hydrogel that recapitulates the stiffness of blood-vessel intima, basement membrane self-deposition and self-healing endothelial barrier function for longer than 1 month. The microsystem enables the real-time visualization, with high spatiotemporal resolution, of microvascular obstruction and endothelial permeability under physiological flow conditions. We found how extracellular heme, a hemolytic byproduct, induces delayed but reversible endothelial permeability in a dose-dependent manner, and demonstrate that endothelial interactions with SCD or malaria-infected erythrocytes cause reversible microchannel occlusion and increased in situ endothelial permeability. The microvasculature-on-a-chip enables mechanistic insight into the endothelial barrier dysfunction associated with SCD, malaria and other inflammatory and haematological diseases.

12.
Nat Commun ; 9(1): 509, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29410404

RESUMEN

Hemostasis encompasses an ensemble of interactions among platelets, coagulation factors, blood cells, endothelium, and hemodynamic forces, but current assays assess only isolated aspects of this complex process. Accordingly, here we develop a comprehensive in vitro mechanical injury bleeding model comprising an "endothelialized" microfluidic system coupled with a microengineered pneumatic valve that induces a vascular "injury". With perfusion of whole blood, hemostatic plug formation is visualized and "in vitro bleeding time" is measured. We investigate the interaction of different components of hemostasis, gaining insight into several unresolved hematologic issues. Specifically, we visualize and quantitatively demonstrate: the effect of anti-platelet agent on clot contraction and hemostatic plug formation, that von Willebrand factor is essential for hemostasis at high shear, that hemophilia A blood confers unstable hemostatic plug formation and altered fibrin architecture, and the importance of endothelial phosphatidylserine in hemostasis. These results establish the versatility and clinical utility of our microfluidic bleeding model.


Asunto(s)
Tiempo de Sangría , Pruebas de Coagulación Sanguínea , Hemorragia , Hemostasis , Microfluídica , Coagulación Sanguínea , Plaquetas/metabolismo , Membrana Celular/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Adhesividad Plaquetaria , Resistencia al Corte , Estrés Mecánico
13.
Blood Rev ; 29(6): 377-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26005062

RESUMEN

During clot formation, platelets are subjected to various different signals and cues as they dynamically interact with extracellular matrix proteins such as von Willebrand factor (vWF), fibrin(ogen) and collagen. While the downstream signaling of platelet-ligand interactions is well-characterized, biophysical cues, such as hydrodynamic forces and mechanical stiffness of the underlying substrate, also mediate these interactions and affect the binding kinetics of platelets to these proteins. Recent studies have observed that, similar to nucleated cells, platelets mechanosense their microenvironment and exhibit dynamic physiologic responses to biophysical cues. This review discusses how platelet mechanosensing is affected by the hydrodynamic forces that dictate vWF-platelet interactions and fibrin polymerization and network formation. The similarities and differences in mechanosensing between platelets and nucleated cells and integrin-mediated platelet mechanosensing on both fibrin(ogen) and collagen are then reviewed. Further studies investigating how platelets interact with the mechanical microenvironment will improve our overall understanding of the hemostatic process.


Asunto(s)
Plaquetas/química , Citoesqueleto/metabolismo , Mecanotransducción Celular , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Factor de von Willebrand/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Plaquetas/metabolismo , Citoesqueleto/ultraestructura , Fibrina/genética , Fibrina/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Regulación de la Expresión Génica , Humanos , Activación Plaquetaria , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
14.
Thromb Res ; 133(4): 532-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24440140

RESUMEN

While the role of platelets in hemostasis is well characterized from a biological perspective, the biophysical interactions between platelets and their mechanical microenvironment are relatively unstudied. The field of cellular mechanics has developed a number of approaches to study the effects of extracellular matrix (ECM)-derived mechanical forces on various cells, and has elucidated that integrin-cytoskeleton-mediated force transduction governs many cellular processes. As platelets adhere and spread via molecular machinery that is similar to that which enables other cells to mechanosense and mechanotransduce forces from their biophysical microenvironment, platelets too are likely governed by the same overarching mechanisms. Indeed, recent platelet mechanobiology studies have revealed that key aspects of platelet physiology and activation are regulated by the mechanical and spatial properties of the ECM microenvironment. At the same time, there are also key differences that make platelets unique in the world of cells-- their size, origin as megakaryocyte fragments, and unique αIIbß3 integrin-- render their mechanosensing activities particularly interesting. The structurally "simple," anucleate nature of platelets coupled with their high actin concentration (20% of total protein) and integrin density [1] seem to make them ideal for mechanical force generation and transmission. Further studies will enhance our understanding of the role of platelet mechanobiology in hemostasis and thrombosis, potentially leading to new categories of diagnostics that investigate the mechanical properties of clots to determine bleeding risk, as well as therapies that target the mechanotransduction signaling pathway to alter the stability of clots.


Asunto(s)
Plaquetas/fisiología , Mecanotransducción Celular/fisiología , Fenómenos Biofísicos , Plaquetas/citología , Hemostasis , Humanos , Microscopía de Fuerza Atómica
15.
Biomicrofluidics ; 8(4): 046501, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25379106

RESUMEN

Cell culture in microfluidic systems has primarily been conducted in devices comprised of polydimethylsiloxane (PDMS) or other elastomers. As polystyrene (PS) is the most characterized and commonly used substrate material for cell culture, microfluidic cell culture would ideally be conducted in PS-based microsystems that also enable tight control of perfusion and hydrodynamic conditions, which are especially important for culture of vascular cell types. Here, we report a simple method to prototype perfusable PS microfluidics for endothelial cell culture under flow that can be fabricated using standard lithography and wet laboratory equipment to enable stable perfusion at shear stresses up to 300 dyn/cm(2) and pumping pressures up to 26 kPa for at least 100 h. This technique can also be extended to fabricate perfusable hybrid PS-PDMS microfluidics of which one application is for increased efficiency of viral transduction in non-adherent suspension cells by leveraging the high surface area to volume ratio of microfluidics and adhesion molecules that are optimized for PS substrates. These biologically compatible microfluidic devices can be made more accessible to biological-based laboratories through the outsourcing of lithography to various available microfluidic foundries.

16.
J Clin Invest ; 124(8): 3590-600, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24983320

RESUMEN

Venous thrombi, fibrin- and rbc-rich clots triggered by inflammation and blood stasis, underlie devastating, and sometimes fatal, occlusive events. During intravascular fibrin deposition, rbc are thought to become passively trapped in thrombi and therefore have not been considered a modifiable thrombus component. In the present study, we determined that activity of the transglutaminase factor XIII (FXIII) is critical for rbc retention within clots and directly affects thrombus size. Compared with WT mice, mice carrying a homozygous mutation in the fibrinogen γ chain (Fibγ390-396A) had a striking 50% reduction in thrombus weight due to reduced rbc content. Fibrinogen from mice harboring the Fibγ390-396A mutation exhibited reduced binding to FXIII, and plasma from these mice exhibited delayed FXIII activation and fibrin crosslinking, indicating these residues mediate FXIII binding and activation. FXIII-deficient mice phenocopied mice carrying Fibγ390-396A and produced smaller thrombi with fewer rbc than WT mice. Importantly, FXIII-deficient human clots also exhibited reduced rbc retention. The addition of FXIII to FXIII-deficient clots increased rbc retention, while inhibition of FXIII activity in normal blood reduced rbc retention and produced smaller clots. These findings establish the FXIII-fibrinogen axis as a central determinant in venous thrombogenesis and identify FXIII as a potential therapeutic target for limiting venous thrombosis.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/patología , Factor XIII/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Trombosis de la Vena/sangre , Animales , Coagulación Sanguínea , Modelos Animales de Enfermedad , Deficiencia del Factor XIII/sangre , Deficiencia del Factor XIII/genética , Factor XIIIa/genética , Factor XIIIa/metabolismo , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteínas Mutantes/genética , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Trombosis de la Vena/genética
17.
Artículo en Inglés | MEDLINE | ID: mdl-22185643

RESUMEN

BACKGROUND: Currently, mechanical support is the most promising alternative to cardiac transplantation. Ventricular assist devices (VADs) were originally used to provide mechanical circulatory support in patients awaiting planned heart transplantation ('bridge-to-transplantation' therapy). The success of short-term bridge devices led to clinical trials evaluating the clinical suitability of long-term support ('destination' therapy) with left ventricular assist devices (LVADs). The first larger scale, randomised trial that tested long-term support with an LVAD reported a 44% reduction in the risk of stroke or death in patients with an LVAD. In spite of the success of LVADs as bridge-to-transplantation and long-term support, patients managed by these devices are still at risk of several adverse events. The most devastating complication is caused by embolisation of thrombi formed within the LVAD or inside the heart into the brain. Prevention of thrombi formation is attempted through anticoagulation management and by improving LVADs design; however, there is still significant occurrence of thromboembolic events in patients. Investigators have reported that the incidence of thromboembolic cerebral events ranges from 14% to 47% over a period of 6-12 months. METHODS AND APPROACH: An alternative method to reduce the incidence of cerebral embolisation is proposed by the co-authors, and the hypothesis is that it is possible to minimise the number of thrombi flowing into the carotid and vertebral arteries by an optimal placement of the LVAD outflow conduit, with or without the addition of aortic bypass connecting the ascending aorta and the innominate artery (IA), or left carotid artery. This paper presents the computational fluid dynamics (CFD) analysis of the aortic arch haemodynamics using a representative geometry of the human aortic arch with or without an alternative aortic bypass. In order to study the trajectory of the thrombi within the aortic arch bed, the CFD code, Fluent 6.3, is utilised to resolve the flow field and to solve the Lagrangian particle tracking of thrombi released randomly at the inlet of the LVAD cannula. RESULTS: Results are presented for simulations of thrombi in the range of 2-5 mm. The percentage of individual diameter as well as aggregate diameter thrombi flowing to the carotid and vertebral arteries as a function of LVAD conduit placement and aortic bypass implantation is reported. The influence of the LVAD conduit implantation and bypass reveals a nearly 50% variation in predicted cerebral embolism rates. CONCLUSIONS: The adjustment of the location of the anastomosis of the LVAD outflow cannula as well as its angle of incidence plays a significant role in the level of thromboembolisms. By proper adjustment in this CFD study of a synthetic model of an aortic arch bed, we found that nearly a 50% reduction in cerebral embolism could be achieved for a configuration consisting of a shallow angle of implantation over a baseline normal incidence of the LVAD cannula. Within the limitations of our model, we have established that the LVAD implantation geometry is an important factor and should be taken into consideration when implanting an LVAD. It is possible that other parameters such as distance of the LVAD outflow cannula to the root of the IA could affect the thrombi embolisation probabilities. However, the results of this study suggest that the risk of stroke may be significantly reduced by as much as 50% by tailoring the VAD implantation by a simple surgical manoeuvre. The results of this line of research may ultimately lead to techniques that can be used to estimate the optimal LVAD configuration in a patient-specific manner by pre-operative imaging.


Asunto(s)
Aorta Torácica , Procedimientos Quirúrgicos Cardíacos/métodos , Ventrículos Cardíacos/cirugía , Corazón Auxiliar/efectos adversos , Accidente Cerebrovascular/prevención & control , Aorta Torácica/anatomía & histología , Aorta Torácica/fisiología , Aorta Torácica/cirugía , Simulación por Computador , Femenino , Hemodinámica , Humanos , Embolia Intracraneal/etiología , Embolia Intracraneal/prevención & control , Masculino , Modelos Anatómicos , Accidente Cerebrovascular/etiología , Tromboembolia/etiología , Tromboembolia/prevención & control
18.
J Vis Exp ; (64)2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22760254

RESUMEN

Advances in microfabrication techniques have enabled the production of inexpensive and reproducible microfluidic systems for conducting biological and biochemical experiments at the micro- and nanoscales (1,2). In addition, microfluidics have also been specifically used to quantitatively analyze hematologic and microvascular processes, because of their ability to easily control the dynamic fluidic environment and biological conditions(3-6). As such, researchers have more recently used microfluidic systems to study blood cell deformability, blood cell aggregation, microvascular blood flow, and blood cell-endothelial cell interactions(6-13).However, these microfluidic systems either did not include cultured endothelial cells or were larger than the sizescale relevant to microvascular pathologic processes. A microfluidic platform with cultured endothelial cells that accurately recapitulates the cellular, physical, and hemodynamic environment of the microcirculation is needed to further our understanding of the underlying biophysical pathophysiology of hematologic diseases that involve the microvasculature. Here, we report a method to create an "endothelialized" in vitro model of the microvasculature, using a simple, single mask microfabrication process in conjunction with standard endothelial cell culture techniques, to study pathologic biophysical microvascular interactions that occur in hematologic disease. This "microvasculature-on-a-chip" provides the researcher with a robust assay that tightly controls biological as well as biophysical conditions and is operated using a standard syringe pump and brightfield/fluorescence microscopy. Parameters such as microcirculatory hemodynamic conditions, endothelial cell type, blood cell type(s) and concentration(s), drug/inhibitory concentration etc., can all be easily controlled. As such, our microsystem provides a method to quantitatively investigate disease processes in which microvascular flow is impaired due to alterations in cell adhesion, aggregation, and deformability, a capability unavailable with existing assays.


Asunto(s)
Células Endoteliales/patología , Endotelio Vascular/patología , Enfermedades Hematológicas/patología , Técnicas Analíticas Microfluídicas/métodos , Dimetilpolisiloxanos/química , Humanos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Microvasos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA