Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
EMBO J ; 33(10): 1089-90, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24711516

RESUMEN

Endothelial sprouting during angiogenesis is a highly coordinated morphogenetic process that involves polarized tip cells leading stalk cells to form new capillaries. While tip and stalk cells previously were thought to be stable and have static phenotypes within the sprout, it is becoming increasingly clear that endothelial cells undergo dynamic rearrangements. A new study using computer simulations, validated by in vitro and in vivo experimental data, now provides an explanation for these rearrangements, showing that sprouting cells are in a continuum of migratory states, regulated by differential cell-cell adhesions and protrusive activities to drive proper vascular organization.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/patología , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Femenino , Humanos , Masculino
2.
Nat Methods ; 11(12): 1229-32, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25306545

RESUMEN

We developed molecular tension probes (TPs) that report traction forces of adherent cells with high spatial resolution, can in principle be linked to virtually any surface, and obviate monitoring deformations of elastic substrates. TPs consist of DNA hairpins conjugated to fluorophore-quencher pairs that unfold and fluoresce when subjected to specific forces. We applied TPs to reveal that cellular traction forces are heterogeneous within focal adhesions and localized at their distal edges.


Asunto(s)
Adhesión Celular/fisiología , Sondas de ADN , Adhesiones Focales/fisiología , Mecanotransducción Celular/fisiología , Animales , Células Cultivadas , Sondas de ADN/química , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Microscopía Fluorescente
3.
Proc Natl Acad Sci U S A ; 111(22): 7968-73, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843171

RESUMEN

The density and architecture of capillary beds that form within a tissue depend on many factors, including local metabolic demand and blood flow. Here, using microfluidic control of local fluid mechanics, we show the existence of a previously unappreciated flow-induced shear stress threshold that triggers angiogenic sprouting. Both intraluminal shear stress over the endothelium and transmural flow through the endothelium above 10 dyn/cm(2) triggered endothelial cells to sprout and invade into the underlying matrix, and this threshold is not impacted by the maturation of cell-cell junctions or pressure gradient across the monolayer. Antagonizing VE-cadherin widened cell-cell junctions and reduced the applied shear stress for a given transmural flow rate, but did not affect the shear threshold for sprouting. Furthermore, both transmural and luminal flow induced expression of matrix metalloproteinase 1, and this up-regulation was required for the flow-induced sprouting. Once sprouting was initiated, continuous flow was needed to both sustain sprouting and prevent retraction. To explore the potential ramifications of a shear threshold on the spatial patterning of new sprouts, we used finite-element modeling to predict fluid shear in a variety of geometric settings and then experimentally demonstrated that transmural flow guided preferential sprouting toward paths of draining interstitial fluid flow as might occur to connect capillary beds to venules or lymphatics. In addition, we show that luminal shear increases in local narrowings of vessels to trigger sprouting, perhaps ultimately to normalize shear stress across the vasculature. Together, these studies highlight the role of shear stress in controlling angiogenic sprouting and offer a potential homeostatic mechanism for regulating vascular density.


Asunto(s)
Células Endoteliales/fisiología , Metaloproteinasa 1 de la Matriz/fisiología , Microfluídica/instrumentación , Microfluídica/métodos , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Adenoviridae/genética , Capilares/citología , Capilares/fisiología , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/ultraestructura , Análisis de Elementos Finitos , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metaloproteinasa 1 de la Matriz/genética , Mecanotransducción Celular/fisiología , Microscopía Electrónica de Transmisión , Estrés Mecánico
4.
Proc Natl Acad Sci U S A ; 110(3): 881-6, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23277584

RESUMEN

Recent methods have revealed that cells on planar substrates exert both shear (in-plane) and normal (out-of-plane) tractions against the extracellular matrix (ECM). However, the location and origin of the normal tractions with respect to the adhesive and cytoskeletal elements of cells have not been elucidated. We developed a high-spatiotemporal-resolution, multidimensional (2.5D) traction force microscopy to measure and model the full 3D nature of cellular forces on planar 2D surfaces. We show that shear tractions are centered under elongated focal adhesions whereas upward and downward normal tractions are detected on distal (toward the cell edge) and proximal (toward the cell body) ends of adhesions, respectively. Together, these forces produce significant rotational moments about focal adhesions in both protruding and retracting peripheral regions. Temporal 2.5D traction force microscopy analysis of migrating and spreading cells shows that these rotational moments are highly dynamic, propagating outward with the leading edge of the cell. Finally, we developed a finite element model to examine how rotational moments could be generated about focal adhesions in a thin lamella. Our model suggests that rotational moments can be generated largely via shear lag transfer to the underlying ECM from actomyosin contractility applied at the intracellular surface of a rigid adhesion of finite thickness. Together, these data demonstrate and probe the origin of a previously unappreciated multidimensional stress profile associated with adhesions and highlight the importance of new approaches to characterize cellular forces.


Asunto(s)
Adhesiones Focales/fisiología , Animales , Fenómenos Biofísicos , Fibroblastos/citología , Fibroblastos/fisiología , Análisis de Elementos Finitos , Proteínas Fluorescentes Verdes/metabolismo , Mecanotransducción Celular/fisiología , Ratones , Microscopía Fluorescente , Modelos Biológicos , Proteínas Recombinantes/metabolismo , Rotación
5.
Proc Natl Acad Sci U S A ; 110(17): 6712-7, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569284

RESUMEN

Angiogenesis is a complex morphogenetic process whereby endothelial cells from existing vessels invade as multicellular sprouts to form new vessels. Here, we have engineered a unique organotypic model of angiogenic sprouting and neovessel formation that originates from preformed artificial vessels fully encapsulated within a 3D extracellular matrix. Using this model, we screened the effects of angiogenic factors and identified two distinct cocktails that promoted robust multicellular endothelial sprouting. The angiogenic sprouts in our system exhibited hallmark structural features of in vivo angiogenesis, including directed invasion of leading cells that developed filopodia-like protrusions characteristic of tip cells, following stalk cells exhibiting apical-basal polarity, and lumens and branches connecting back to the parent vessels. Ultimately, sprouts bridged between preformed channels and formed perfusable neovessels. Using this model, we investigated the effects of angiogenic inhibitors on sprouting morphogenesis. Interestingly, the ability of VEGF receptor 2 inhibition to antagonize filopodia formation in tip cells was context-dependent, suggesting a mechanism by which vessels might be able to toggle between VEGF-dependent and VEGF-independent modes of angiogenesis. Like VEGF, sphingosine-1-phosphate also seemed to exert its proangiogenic effects by stimulating directional filopodial extension, whereas matrix metalloproteinase inhibitors prevented sprout extension but had no impact on filopodial formation. Together, these results demonstrate an in vitro 3D biomimetic model that reconstitutes the morphogenetic steps of angiogenic sprouting and highlight the potential utility of the model to elucidate the molecular mechanisms that coordinate the complex series of events involved in neovascularization.


Asunto(s)
Biomimética/métodos , Microfluídica/métodos , Modelos Biológicos , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Polaridad Celular/fisiología , Dimetilpolisiloxanos , Clorhidrato de Fingolimod , Técnica del Anticuerpo Fluorescente , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/farmacología , Lisofosfolípidos/metabolismo , Morfogénesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Glicoles de Propileno/farmacología , Seudópodos/fisiología , Pirroles/farmacología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
6.
J Cell Sci ; 126(Pt 18): 4121-35, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23843620

RESUMEN

Cell migration makes a fundamental contribution to both normal physiology and disease pathogenesis. Integrin engagement with extracellular ligands spatially controls, via the cyclical activation and deactivation of the small GTPase Rac1, the dynamic membrane protrusion and cytoskeletal reorganization events that are required for directional migration. Although the pathways that control integrin-mediated Rac1 activation are reasonably well defined, the mechanisms that are responsible for switching off activity are poorly understood. Here, proteomic analysis of activated integrin-associated complexes suggests filamin-A and IQ-motif-containing GTPase-activating protein 1 (IQGAP1) as candidates that link ß1 integrin to Rac1. siRNA-mediated knockdown of either filamin-A or IQGAP1 induced high, dysregulated Rac1 activity during cell spreading on fibronectin. Using immunoprecipitation and immunocytochemistry, filamin-A and IQGAP1 were shown to be part of a complex that is recruited to active ß1 integrin. Mass spectrometric analysis of individual filamin-A, IQGAP1 and Rac1 pull-downs and biochemical analysis, identified RacGAP1 as a novel IQGAP1 binding partner. Further immunoprecipitation and immunocytochemistry analyses demonstrated that RacGAP1 is recruited to IQGAP1 and active ß1 integrin, and that suppression of RacGAP1 expression triggered elevated Rac1 activity during spreading on fibronectin. Consistent with these findings, reduced expression of filamin-A, IQGAP1 or RacGAP1 triggered unconstrained membrane protrusion and disrupted directional cell migration on fibrillar extracellular matrices. These findings suggest a model whereby integrin engagement, followed by filamin-A, IQGAP1 and RacGAP1 recruitment, deactivates Rac1 to constrain its activity spatially and thereby coordinate directional cell migration.


Asunto(s)
Integrina beta1/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Movimiento Celular , Filaminas , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteómica , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Proteína de Unión al GTP rac1/genética , Proteínas Activadoras de ras GTPasa/genética
7.
Nat Cell Biol ; 10(9): 1039-50, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19160484

RESUMEN

Using two-colour imaging and high resolution TIRF microscopy, we investigated the assembly and maturation of nascent adhesions in migrating cells. We show that nascent adhesions assemble and are stable within the lamellipodium. The assembly is independent of myosin II but its rate is proportional to the protrusion rate and requires actin polymerization. At the lamellipodium back, the nascent adhesions either disassemble or mature through growth and elongation. Maturation occurs along an alpha-actinin-actin template that elongates centripetally from nascent adhesions. Alpha-Actinin mediates the formation of the template and organization of adhesions associated with actin filaments, suggesting that actin crosslinking has a major role in this process. Adhesion maturation also requires myosin II. Rescue of a myosin IIA knockdown with an actin-bound but motor-inhibited mutant of myosin IIA shows that the actin crosslinking function of myosin II mediates initial adhesion maturation. From these studies, we have developed a model for adhesion assembly that clarifies the relative contributions of myosin II and actin polymerization and organization.


Asunto(s)
Actinina/metabolismo , Actinas/metabolismo , Animales , Células CHO , Adhesión Celular , Polaridad Celular , Cricetinae , Cricetulus , Reactivos de Enlaces Cruzados/metabolismo , Humanos , Modelos Biológicos , Miosina Tipo IIA no Muscular/metabolismo , Seudópodos/metabolismo , Ratas
8.
J Med Chem ; 67(10): 8122-8140, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38712838

RESUMEN

Multiple sclerosis (MS) is a chronic disease with an underlying pathology characterized by inflammation-driven neuronal loss, axonal injury, and demyelination. Bruton's tyrosine kinase (BTK), a nonreceptor tyrosine kinase and member of the TEC family of kinases, is involved in the regulation, migration, and functional activation of B cells and myeloid cells in the periphery and the central nervous system (CNS), cell types which are deemed central to the pathology contributing to disease progression in MS patients. Herein, we describe the discovery of BIIB129 (25), a structurally distinct and brain-penetrant targeted covalent inhibitor (TCI) of BTK with an unprecedented binding mode responsible for its high kinome selectivity. BIIB129 (25) demonstrated efficacy in disease-relevant preclinical in vivo models of B cell proliferation in the CNS, exhibits a favorable safety profile suitable for clinical development as an immunomodulating therapy for MS, and has a low projected total human daily dose.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Encéfalo , Esclerosis Múltiple , Inhibidores de Proteínas Quinasas , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Humanos , Animales , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/química , Encéfalo/metabolismo , Ratones , Descubrimiento de Drogas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Ratas , Relación Estructura-Actividad , Proliferación Celular/efectos de los fármacos , Femenino
9.
ALTEX ; 40(3): 485-518, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36648096

RESUMEN

Disease models enable researchers to investigate, test, and identify therapeutic targets that would alter the patients' disease condition and improve quality of life. Advances in genetic alteration and analytical techniques have enabled rapid devel­opment of disease models using preclinical animals and cell cultures. However, success rates of drug development remain low due to limited recapitulation of clinical pathophysiology by these models. To resolve this challenge, the pharmaceutical industry has explored microphysiological system (MPS) disease models, which are complex in vitro systems that include but are not limited to organ-on-a-chip, organoids, spheroids, and 3D bioengineered tissues (e.g., 3D printing, hydrogels). Capable of integrating key in vivo properties, such as disease-relevant human cells, multi-cellularity/dimensionality of organs, and/or well-controlled physical and molecular cues, MPS disease models are being developed for a variety of indications. With on-going qualifications or validations for wide adoption within the pharmaceutical industry, MPS disease models hold exciting potential to enable in-depth investigation of in vivo pathophysiology and enhance drug discovery and development processes. To introduce the present status of MPS disease models, this paper describes notable examples in six disease areas: cancer, liver/kidney diseases, respiratory diseases/COVID-19, neurodegenerative diseases, gastrointestinal diseases, and select rare diseases. Additionally, we describe current technical limitations and provide recommendations for future development that would expand application opportunities within the pharmaceutical industry.


Asunto(s)
Productos Biológicos , COVID-19 , Animales , Humanos , Sistemas Microfisiológicos , Calidad de Vida , Hígado , Dispositivos Laboratorio en un Chip
10.
Adv Biol (Weinh) ; : e2300131, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37814378

RESUMEN

In May 2022, there is an International Regulatory and Pharmaceutical Industry (Innovation and Quality [IQ] Microphysiological Systems [MPS] Affiliate) Workshop on the standardization of complex in vitro models (CIVMs) in drug development. This manuscript summarizes the discussions and conclusions of this joint workshop organized and executed by the IQ MPS Affiliate and the United States Food and Drug Administration (FDA). A key objective of the workshop is to facilitate discussions around opportunities and/or needs for standardization of MPS and chart potential pathways to increase model utilization in the context of regulatory decision making. Participation in the workshop included 200 attendees from the FDA, IQ MPS Affiliate, and 26 global regulatory organizations and affiliated parties representing Europe, Japan, and Canada. It is agreed that understanding global perspectives regarding the readiness of CIVM/MPS models for regulatory decision making and potential pathways to gaining acceptance is useful to align on globally. The obstacles are currently too great to develop standards for every context of use (COU). Instead, it is suggested that a more tractable approach may be to think of broadly applicable standards that can be applied regardless of COU and/or organ system. Considerations and next steps for this effort are described.

11.
J Cell Biol ; 176(5): 573-80, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17312025

RESUMEN

We have used isoform-specific RNA interference knockdowns to investigate the roles of myosin IIA (MIIA) and MIIB in the component processes that drive cell migration. Both isoforms reside outside of protrusions and act at a distance to regulate cell protrusion, signaling, and maturation of nascent adhesions. MIIA also controls the dynamics and size of adhesions in central regions of the cell and contributes to retraction and adhesion disassembly at the rear. In contrast, MIIB establishes front-back polarity and centrosome, Golgi, and nuclear orientation. Using ATPase- and contraction-deficient mutants of both MIIA and MIIB, we show a role for MIIB-dependent actin cross-linking in establishing front-back polarity. From these studies, MII emerges as a master regulator and integrator of cell migration. It mediates each of the major component processes that drive migration, e.g., polarization, protrusion, adhesion assembly and turnover, polarity, signaling, and tail retraction, and it integrates spatially separated processes.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular , Miosina Tipo IIA no Muscular/fisiología , Miosina Tipo IIB no Muscular/fisiología , Actinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Células CHO , Adhesión Celular/fisiología , Cricetinae , Cricetulus , Miosina Tipo IIA no Muscular/antagonistas & inhibidores , Miosina Tipo IIB no Muscular/antagonistas & inhibidores , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/fisiología , Interferencia de ARN , Ratas , Transducción de Señal
12.
Biophys J ; 100(3): 583-592, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21281572

RESUMEN

We used correlation methods to detect and quantify interactions between paxillin and focal adhesion kinase (FAK) in migrating cells. Cross-correlation raster-scan image correlation spectroscopy revealed that wild-type paxillin and the phosphorylation-inhibiting paxillin mutant Y31F-Y118F do not interact with FAK in the cytosol but a phosphomimetic mutant of paxillin, Y31E-Y118E, does. By extending cross-correlation number and brightness analysis to the total internal reflection fluorescence modality, we were able to show that tetramers of paxillin and FAK form complexes in nascent adhesions with a 1:1 stoichiometry ratio. The phosphomimetic mutations on paxillin increase the size of the complex and the assembly rate of nascent adhesions, suggesting that the physical molecular aggregation of paxillin and FAK regulates adhesion formation. In contrast, when phosphorylation is inhibited, the interaction decreases and the adhesions tend to elongate rather than turn over. These direct in vivo data show that the phosphorylation of paxillin is specific to adhesions and leads to localized complex formation with FAK to regulate the dynamics of nascent adhesions.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Paxillin/metabolismo , Análisis Espectral/métodos , Animales , Células CHO , Adhesión Celular , Cricetinae , Cricetulus , Citosol/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica
13.
PLoS Comput Biol ; 6(2): e1000688, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195494

RESUMEN

Productive cell migration requires the spatiotemporal coordination of cell adhesion, membrane protrusion, and actomyosin-mediated contraction. Integrins, engaged by the extracellular matrix (ECM), nucleate the formation of adhesive contacts at the cell's leading edge(s), and maturation of nascent adhesions to form stable focal adhesions constitutes a functional switch between protrusive and contractile activities. To shed additional light on the coupling between integrin-mediated adhesion and membrane protrusion, we have formulated a quantitative model of leading edge dynamics combining mechanistic and phenomenological elements and studied its features through classical bifurcation analysis and stochastic simulation. The model describes in mathematical terms the feedback loops driving, on the one hand, Rac-mediated membrane protrusion and rapid turnover of nascent adhesions, and on the other, myosin-dependent maturation of adhesions that inhibit protrusion at high ECM density. Our results show that the qualitative behavior of the model is most sensitive to parameters characterizing the influence of stable adhesions and myosin. The major predictions of the model, which we subsequently confirmed, are that persistent leading edge protrusion is optimal at an intermediate ECM density, whereas depletion of myosin IIA relieves the repression of protrusion at higher ECM density.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Modelos Biológicos , Procesos Estocásticos , Animales , Células CHO , Extensiones de la Superficie Celular/metabolismo , Biología Computacional/métodos , Simulación por Computador , Cricetinae , Cricetulus , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrinas/metabolismo , Miosinas/metabolismo , Paxillin/genética , Paxillin/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rac/metabolismo
14.
J Biol Chem ; 284(50): 34713-22, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19826001

RESUMEN

Tensin is a family of multidomain scaffold proteins that bind the cytoplasmic tail of beta-integrins and localize to adhesions that anchor stress fibers in cells. Tensin expression is suppressed in cancer, especially metastatic cancer. The N-terminal domain of tensin1 associates with protein phosphatase-1alpha (PP1alpha) and mediates PP1alpha localization to adhesions. Here, we show F302A mutation in a KVXF motif of tensin1 abrogates binding to PP1alpha. The SH2 domain in tensin family member c-ten requires R474 to bind a RhoGAP called DLC-1 (deleted in liver cancer). We mutated the corresponding residue in tensin1, R1488A, and showed this reduces association with DLC-1. Unexpectedly, tensin1 F302A also had reduced association with DLC-1. Expression of tensin1 F302A or R1488A showed similar dominant phenotypes, with reduced cell polarization, lowered MLC20 phosphorylation and reduced levels of RhoA(GTP) compared with cells expressing tensin1 WT. However, migration and invasion of metastatic MDA MB 231 breast cancer cells were differentially affected by tensin1 mutated at F302A or R1488A. Cancer cells stably expressing F302A tensin1 showed increased migration and invasion compared with cells stably expressing either R1488A tensin1 or WT tensin1. This suggests that PP1alpha bound to tensin1 has additional effects in reducing migration and invasion that are not mediated through DLC-1. Our results show the importance of PP1alpha binding to tensin1 for the regulation of cell polarization, migration, and invasion.


Asunto(s)
Movimiento Celular/fisiología , Polaridad Celular , Proteínas de Microfilamentos/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/fisiología , Línea Celular , Forma de la Célula , Activación Enzimática , Proteínas Activadoras de GTPasa , Humanos , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Invasividad Neoplásica/fisiopatología , Neoplasias/metabolismo , Neoplasias/patología , Mutación Puntual , Proteína Fosfatasa 1/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tensinas , Proteínas Supresoras de Tumor/genética , Proteína de Unión al GTP rhoA/metabolismo
15.
APL Bioeng ; 2(4): 046107, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31069329

RESUMEN

Physical features of the extracellular matrix (ECM) heavily influence cell migration strategies and efficiency. Migration in and on fibrous ECMs is of significant physiologic importance, but limitations in the ability to experimentally define the diameter, density, and alignment of native ECMs in vitro have hampered our understanding of how these properties affect this basic cell function. Here, we designed a high-throughput in vitro platform that models fibrous ECM as collections of lines of cell-adhesive fibronectin on a flat surface to eliminate effects of dimensionality and topography. Using a microcontact printing approach to orthogonally vary line alignment, density, and size, we determined each factor's individual influence on NIH3T3 fibroblast migration. High content imaging and statistical analyses revealed that ECM alignment is the most critical parameter in influencing cell morphology, polarization, and migratory behavior. Specifically, increasing ECM alignment led cells to adopt an elongated uniaxial morphology and migrate with enhanced speed and persistence. Intriguingly, migration speeds were tightly correlated with the organization of focal adhesions, where cells with the most aligned adhesions migrated fastest. Highly organized focal adhesions and associated actin stress fibers appeared to define the number and location of protrusive fronts, suggesting that ECM alignment influences active Rac1 localization. Utilizing a novel microcontact-printing approach that lacks confounding influences of substrate dimensionality, mechanics, or differences in the adhesive area, this work highlights the effect of ECM alignment on orchestrating the cytoskeletal machinery that governs directed uniaxial cell migration.

16.
Nat Commun ; 8(1): 371, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851858

RESUMEN

A major challenge in tissue engineering is the development of materials that can support angiogenesis, wherein endothelial cells from existing vasculature invade the surrounding matrix to form new vascular structures. To identify material properties that impact angiogenesis, here we have developed an in vitro model whereby molded tubular channels inside a synthetic hydrogel are seeded with endothelial cells and subjected to chemokine gradients within a microfluidic device. To accomplish precision molding of hydrogels and successful integration with microfluidics, we developed a class of hydrogels that could be macromolded and micromolded with high shape and size fidelity by eliminating swelling after polymerization. Using this material, we demonstrate that matrix degradability switches three-dimensional endothelial cell invasion between two distinct modes: single-cell migration and the multicellular, strand-like invasion required for angiogenesis. The ability to incorporate these tunable hydrogels into geometrically constrained settings will enable a wide range of previously inaccessible biomedical applications.The fabrication of vascularized 3D tissues requires an understanding of how material properties govern endothelial cell invasion into the surrounding matrix. Here the authors integrate a non-swelling synthetic hydrogel with a microfluidic device to study chemokine gradient-driven angiogenic sprouting and find that matrix degradability modulates the collectivity of cell migration.


Asunto(s)
Movimiento Celular , Células Endoteliales de la Vena Umbilical Humana/citología , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Proliferación Celular , Humanos , Hidrogeles , Microfluídica/instrumentación , Neovascularización Fisiológica , Ingeniería de Tejidos/instrumentación
17.
Nat Commun ; 6: 6135, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25609142

RESUMEN

Integrin activation, which is regulated by allosteric changes in receptor conformation, enables cellular responses to the chemical, mechanical and topological features of the extracellular microenvironment. A global view of how activation state converts the molecular composition of the region proximal to integrins into functional readouts is, however, lacking. Here, using conformation-specific monoclonal antibodies, we report the isolation of integrin activation state-dependent complexes and their characterization by mass spectrometry. Quantitative comparisons, integrating network, clustering, pathway and image analyses, define multiple functional protein modules enriched in a conformation-specific manner. Notably, active integrin complexes are specifically enriched for proteins associated with microtubule-based functions. Visualization of microtubules on micropatterned surfaces and live cell imaging demonstrate that active integrins establish an environment that stabilizes microtubules at the cell periphery. These data provide a resource for the interrogation of the global molecular connections that link integrin activation to adhesion signalling.


Asunto(s)
Integrinas/metabolismo , Microtúbulos/metabolismo , Proteómica/métodos , Sitio Alostérico , Anticuerpos Monoclonales/química , Corteza Cerebral/metabolismo , Análisis por Conglomerados , Dimetilpolisiloxanos/química , Fibroblastos/metabolismo , Prepucio/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Integrina beta1/metabolismo , Células K562 , Masculino , Espectrometría de Masas , Microscopía Fluorescente , Unión Proteica , Conformación Proteica , Proteoma , Transducción de Señal
18.
Trends Cell Biol ; 20(12): 705-14, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20965727

RESUMEN

Cells constantly probe and respond to a myriad of cues that are present in their local surroundings. The effects of soluble cues are relatively straightforward to manipulate, yet teasing apart how cells transduce signals from the extracellular matrix and neighboring cells has proven to be challenging due to the spatially and mechanically complex adhesive interactions. Over the years, advances in the engineering of biocompatible materials have enabled innovative ways to study adhesion-mediated cell functions, and numerous insights have elucidated the significance of the cellular microenvironment. Here, we highlight some of the major approaches and discuss the potential for future advancement.


Asunto(s)
Materiales Biocompatibles/química , Bioingeniería , Adhesión Celular , Animales , Biología Celular , Matriz Extracelular/química , Humanos
19.
Mol Cell Biomech ; 5(4): 247-258, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20084179

RESUMEN

Hemodynamic shear stress guides a variety of endothelial phenotype characteristics, including cell morphology, cytoskeletal structure, and gene expression profile. The sensing and processing of extracellular fluid forces may be mediated by mechanotransmission through the actin cytoskeleton network to intracellular locations of signal initiation. In this study, we identify rapid actin-mediated morphological changes in living subconfluent and confluent bovine aortic endothelial cells (ECs) in response to onset of unidirectional steady fluid shear stress (15 dyn/cm(2)). After flow onset, subconfluent cells exhibited dynamic edge activity in lamellipodia and small ruffles in the downstream and side directions for the first 12 min; activity was minimal in the upstream direction. After 12 min, peripheral edge extension subsided. Confluent cell monolayers that were exposed to shear stress exhibited only subtle increases in edge fluctuations after flow onset. Addition of cytochalasin D to disrupt actin polymerization served to suppress the magnitude of flow-mediated actin remodeling in both subconfluent confluent EC monolayers. Interestingly, when subconfluent ECs were exposed to two sequential flow step increases (1 dyn/cm(2) followed by 15 dyn/cm(2) 12 min later), actin-mediated edge activity was not additionally increased after the second flow step. Thus, repeated flow increases served to desensitize mechanosensitive structural dynamics in the actin cytoskeleton.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA