Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Exp Cell Res ; 330(2): 382-397, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25447308

RESUMEN

Cell migration is crucial in development, tissue repair and immunity and frequently aberrant in pathological processes including tumor metastasis. Focal adhesions (FAs) are integrin-based adhesion complexes that form the link between the cytoskeleton and the extracellular matrix and are thought to orchestrate cell migration. Understanding the regulation of FAs by (oncogenic) signaling pathways may identify strategies to target pathological cell migration. Here we describe the development of a robust FA tracker that enables the automatic, multi-parametric analysis of FA dynamics, morphology and composition from time-lapse image series generated by total internal reflection fluorescence (TIRF) microscopy. In control prostate carcinoma cells, this software recapitulates previous findings that relate morphological characteristics of FAs to their lifetime and their cellular location. We then investigated how FAs are altered when cell migration is induced by the metastasis-promoting hormone HGF and subsequently inhibited by activation of the small GTPase Rap1. We performed a detailed analysis of individual FA parameters, which identified FA size, sliding and intensity as primary targets of Rap1. HGF did not have strong effects on any of the FA parameters within the first hours of its addition. Subsequent Bayesian network inference (BNI), using all measured parameters as input, revealed little correlation between changes in cell migration and FA characteristics in this prostate carcinoma cell line. Instead BNI indicated a concerted coordination of cell size and FA parameters. Thus our results did not reveal a direct relation between the regulation of cell migration and the regulation of FA dynamics.


Asunto(s)
Adhesiones Focales/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Procesamiento de Imagen Asistido por Computador/métodos , Neoplasias de la Próstata/patología , Proteínas de Unión al GTP rap1/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Masculino , Microscopía Fluorescente , Metástasis de la Neoplasia , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Programas Informáticos
2.
Proc Natl Acad Sci U S A ; 109(10): 3814-9, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22343288

RESUMEN

Epac1 is a cAMP-regulated guanine nucleotide exchange factor for the small G protein Rap. Upon cAMP binding, Epac1 undergoes a conformational change that results in its release from autoinhibition. In addition, cAMP induces the translocation of Epac1 from the cytosol to the plasma membrane. This relocalization of Epac1 is required for efficient activation of plasma membrane-located Rap and for cAMP-induced cell adhesion. This translocation requires the Dishevelled, Egl-10, Pleckstrin (DEP) domain, but the molecular entity that serves as the plasma membrane anchor and the possible mechanism of regulated binding remains elusive. Here we show that Epac1 binds directly to phosphatidic acid. Similar to the cAMP-induced Epac1 translocation, this binding is regulated by cAMP and requires the DEP domain. Furthermore, depletion of phosphatidic acid by inhibition of phospholipase D1 prevents cAMP-induced translocation of Epac1 as well as the subsequent activation of Rap at the plasma membrane. Finally, mutation of a single basic residue within a polybasic stretch of the DEP domain, which abolishes translocation, also prevents binding to phosphatidic acid. From these results we conclude that cAMP induces a conformational change in Epac1 that enables DEP domain-mediated binding to phosphatidic acid, resulting in the tethering of Epac1 at the plasma membrane and subsequent activation of Rap.


Asunto(s)
Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ácidos Fosfatidicos/química , Factores de Intercambio de Guanina Nucleótido/química , Células HEK293 , Humanos , Lípidos/química , Liposomas/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes/química , Proteínas de Unión al GTP rap1/química
3.
Mol Oncol ; 9(1): 295-308, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25241147

RESUMEN

Prostate cancer is an androgen receptor (AR)-dependent malignancy at initiation and progression, therefore hormone therapy is the primary line of systemic treatment. Despite initial disease regression, tumours inevitably recur and progress to an advanced castration-resistant state a major feature of which is metastasis to the bone. Up-regulation of AR cofactors and chaperones that overcome low hormone conditions to maintain basal AR activity has been postulated as a mechanism of therapy relapse. p23, an essential component of the apo-AR complex, acts also after ligand binding to increase AR transcriptional activity and target gene expression, partly by increasing chromatin-loaded holo-receptor-complexes. Immunohistochemical studies have demonstrated increased p23 expression in advanced prostate cancer. Here, we further characterise p23 roles in AR signalling and show that it modulates cytosolic AR levels in the absence of hormone, confirming a chaperoning function in the aporeceptor complex and suggesting p23 upregulates AR signalling at multiple stages. Moreover, p23 protein levels significantly increased upon treatment with not only androgen but also clinically relevant anti-androgens. This was in contrast to the HSP90 inhibitor 17-AAG, which did not modulate expression of the cochaperone - important given the HSP90-independent roles we and others have previously described for p23. Further, we demonstrate p23 is implicated in prostate cancer cell motility and in acquisition of invasiveness capacity through the expression of specific genes known to participate in cancer progression. This may drive metastatic processes in vivo since analysis of prostate tumour biopsies revealed that high nuclear p23 significantly correlated with shorter survival times and with development of metastases in patients with lower grade tumours. We propose that increased p23 expression may allow cells to acquire a more aggressive phenotype, contributing to disease progression, and that p23 is a plausible secondary target in combination with HSP90 inhibition as a potential therapy for advanced prostate cancer.


Asunto(s)
Movimiento Celular , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias de la Próstata/metabolismo , Benzoquinonas/farmacología , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Oxidorreductasas Intramoleculares/genética , Lactamas Macrocíclicas/farmacología , Masculino , Metástasis de la Neoplasia , Prostaglandina-E Sintasas , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
4.
Curr Biol ; 23(4): R159-61, 2013 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-23428328

RESUMEN

By beautiful imaging and state-of-the-art experiments, vinculin is established to be a central switch in mechanotransduction at integrin-based focal adhesions. Cycles of tension-regulated vinculin switching control focal adhesion dynamics and signaling to enable polarized cell migration and alignment.


Asunto(s)
Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Vinculina/metabolismo , Animales
5.
Dev Cell ; 27(5): 574-85, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-24290981

RESUMEN

Epithelial cell migration is crucial for the development and regeneration of epithelial tissues. Aberrant regulation of epithelial cell migration has a major role in pathological processes such as the development of cancer metastasis and tissue fibrosis. Here, we report that in response to factors that promote cell motility, the Rap guanine exchange factor RAPGEF2 is rapidly phosphorylated by I-kappa-B-kinase-ß and casein kinase-1α and consequently degraded by the proteasome via the SCF(ßTrCP) ubiquitin ligase. Failure to degrade RAPGEF2 in epithelial cells results in sustained activity of Rap1 and inhibition of cell migration induced by HGF, a potent metastatic factor. Furthermore, expression of a degradation-resistant RAPGEF2 mutant greatly suppresses dissemination and metastasis of human breast cancer cells. These findings reveal a molecular mechanism regulating migration and invasion of epithelial cells and establish a key direct link between IKKß and cell motility controlled by Rap-integrin signaling.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Movimiento Celular/fisiología , Células Epiteliales/citología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Células HEK293 , Xenoinjertos , Humanos , Masculino , Fosforilación/fisiología , Proteínas Ligasas SKP Cullina F-box/metabolismo , Pez Cebra
6.
Sci Signal ; 5(231): rs5, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22763340

RESUMEN

The scattering of cultured epithelial cells in response to hepatocyte growth factor (HGF) is a model system that recapitulates key features of metastatic cell behavior in vitro, including disruption of cell-cell adhesions and induction of cell migration. We have developed image analysis tools that do not require fluorescence tagging and that automatically track and characterize three aspects of scattering in live cells: increase in cell motility, loss of cell-cell adhesion, and spatial dispersion of cells (the redistribution of cells during scattering). We used these tools to screen a library of drugs, and we identified several efficient inhibitors of scattering, which we classified as selective inhibitors of either motility or loss of cell-cell adhesion, or as nonselective inhibitors. We validated the inhibitors and putative targets from this screen in two unrelated model cell lines. Using pharmacological treatments and RNA interference (RNAi), we found that nonsteroidal anti-inflammatory drugs inhibited cell-cell dissociation, that indirubins inhibited cell motility, and that cyclin-dependent kinase 1 and ribosomal S6 kinase were signaling intermediates in HGF-induced cell scattering. This assay is suitable for larger-scale screenings of chemical compounds or RNAi libraries.


Asunto(s)
Adhesión Celular , Movimiento Celular , Línea Celular , Células Epiteliales/citología , Humanos , Interferencia de ARN
7.
J Cell Biol ; 196(5): 641-52, 2012 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-22391038

RESUMEN

To remodel endothelial cell-cell adhesion, inflammatory cytokine- and angiogenic growth factor-induced signals impinge on the vascular endothelial cadherin (VE-cadherin) complex, the central component of endothelial adherens junctions. This study demonstrates that junction remodeling takes place at a molecularly and phenotypically distinct subset of VE-cadherin adhesions, defined here as focal adherens junctions (FAJs). FAJs are attached to radial F-actin bundles and marked by the mechanosensory protein Vinculin. We show that endothelial hormones vascular endothelial growth factor, tumor necrosis factor α, and most prominently thrombin induced the transformation of stable junctions into FAJs. The actin cytoskeleton generated pulling forces specifically on FAJs, and inhibition of Rho-Rock-actomyosin contractility prevented the formation of FAJs and junction remodeling. FAJs formed normally in cells expressing a Vinculin binding-deficient mutant of α-catenin, showing that Vinculin recruitment is not required for adherens junction formation. Comparing Vinculin-devoid FAJs to wild-type FAJs revealed that Vinculin protects VE-cadherin junctions from opening during their force-dependent remodeling. These findings implicate Vinculin-dependent cadherin mechanosensing in endothelial processes such as leukocyte extravasation and angiogenesis.


Asunto(s)
Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Mecanotransducción Celular/fisiología , Estrés Mecánico , Vinculina/metabolismo , Actinas/genética , Actinas/metabolismo , Uniones Adherentes/efectos de los fármacos , Antígenos CD/genética , Cadherinas/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Vinculina/genética
8.
PLoS One ; 7(11): e50072, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209645

RESUMEN

We developed new image analysis tools to analyse quantitatively the extracellular-matrix-dependent cell spreading process imaged by live-cell epifluorescence microscopy. Using these tools, we investigated cell spreading induced by activation of the small GTPase, Rap1. After replating and initial adhesion, unstimulated cells exhibited extensive protrusion and retraction as their spread area increased, and displayed an angular shape that was remodelled over time. In contrast, activation of endogenous Rap1, via 007-mediated stimulation of Epac1, induced protrusion along the entire cell periphery, resulting in a rounder spread surface, an accelerated spreading rate and an increased spread area compared to control cells. Whereas basal, anisotropic, spreading was completely dependent on Src activity, Rap1-induced spreading was refractory to Src inhibition. Under Src inhibited conditions, the characteristic Src-induced tyrosine phosphorylations of FAK and paxillin did not occur, but Rap1 could induce the formation of actomyosin-connected adhesions, which contained vinculin at levels comparable to that found in unperturbed focal adhesions. From these results, we conclude that Rap1 can induce cell adhesion and stimulate an accelerated rate of cell spreading through mechanisms that bypass the canonical FAK-Src-Paxillin signalling cascade.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica , Paxillin/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Familia-src Quinasas/metabolismo , Actomiosina/farmacología , Anisotropía , Adhesión Celular , Línea Celular Tumoral , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Matriz Extracelular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Microscopía Fluorescente/métodos , ARN Interferente Pequeño/metabolismo , Complejo Shelterina , Transducción de Señal , Vinculina/metabolismo
9.
Cell Signal ; 23(12): 2056-64, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21840392

RESUMEN

Epac1 and its effector Rap1 are important mediators of cAMP induced tightening of endothelial junctions and consequential increased barrier function. We have investigated the involvement of Rap1 signalling in basal, unstimulated, barrier function of a confluent monolayer of HUVEC using real time Electric Cell-substrate Impedance Sensing. Depletion of Rap1, but not Epac1, results in a strong decrease in barrier function. This decrease is also observed when cells are depleted of the cAMP independent Rap exchange factors PDZ-GEF1 and 2, showing that PDZ-GEFs are responsible for Rap1 activity in control of basal barrier function. Monolayers of cells depleted of PDZ-GEF or Rap1 show an irregular, zipper-like organization of VE-cadherin and live imaging of VE-cadherin-GFP reveals enhanced junction motility upon depletion of PDZ-GEF or Rap1. Importantly, activation of Epac1 increases the formation of cortical actin bundles at the cell-cell junctions, inhibits junction motility and restores barrier function of PDZ-GEFs depleted, but not Rap1 depleted cells. We conclude that PDZ-GEF activates Rap1 under resting conditions to stabilize cell-cell junctions and maintain basal integrity. Activation of Rap1 by cAMP/Epac1 induces junctional actin to further tighten cell-cell contacts.


Asunto(s)
Uniones Adherentes/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión a Telómeros/metabolismo , ADP Ribosa Transferasas/farmacología , Actinas/metabolismo , Antígenos CD/metabolismo , Toxinas Botulínicas/farmacología , Cadherinas/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Impedancia Eléctrica , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/agonistas , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Microscopía Fluorescente , Proteínas del Tejido Nervioso/genética , Interferencia de ARN , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Imagen de Lapso de Tiempo , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA