Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 7(1): 16499, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29184125

RESUMEN

Information in the microenvironment guides complex cellular decisions such as whether or not to proliferate and migrate. The effects of soluble extracellular signals on these cellular functions are fairly well understood, but relatively little is known about how the extracellular matrix (ECM), and particularly the mechanical information in the ECM, guides these cellular decisions. Here, we show that CD44, a major receptor for the glycosaminoglycan ECM component hyaluronan, coordinates the motility and proliferative responses to ECM stiffening. We analyzed these cellular responses on fibronectin-coated polyacrylamide hydrogels prepared at a physiologic range of ECM stiffness and found that stiffening of the ECM leads to both cell cycling and cell motility in serum-stimulated primary mouse dermal fibroblasts. Remarkably, deletion of CD44 impaired stiffness-stimulated motility of the primary cells without affecting other hallmark cellular responses to ECM stiffening including cell spread area, stress fiber formation, focal adhesion maturation, and intracellular stiffening. Even stiffness-mediated cell proliferation was unaffected by deletion of CD44. Our results reveal a novel effect of CD44, which is imposed downstream of ECM-mechanosensing and determines if cells couple or uncouple their proliferative and motility responses to ECM stiffness.


Asunto(s)
Movimiento Celular/genética , Microambiente Celular , Eliminación de Gen , Receptores de Hialuranos/genética , Animales , Biomarcadores , Proliferación Celular , Forma de la Célula , Matriz Extracelular/metabolismo , Fibroblastos , Masculino , Mecanotransducción Celular , Ratones , Ratones Noqueados , Fosforilación
2.
Sci Signal ; 7(330): ra57, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24939893

RESUMEN

Tissue and extracellular matrix (ECM) stiffness is transduced into intracellular stiffness, signaling, and changes in cellular behavior. Integrins and several of their associated focal adhesion proteins have been implicated in sensing ECM stiffness. We investigated how an initial sensing event is translated into intracellular stiffness and a biologically interpretable signal. We found that a pathway consisting of focal adhesion kinase (FAK), the adaptor protein p130Cas (Cas), and the guanosine triphosphatase Rac selectively transduced ECM stiffness into stable intracellular stiffness, increased the abundance of the cell cycle protein cyclin D1, and promoted S-phase entry. Rac-dependent intracellular stiffening involved its binding partner lamellipodin, a protein that transmits Rac signals to the cytoskeleton during cell migration. Our findings establish that mechanotransduction by a FAK-Cas-Rac-lamellipodin signaling module converts the external information encoded by ECM stiffness into stable intracellular stiffness and mechanosensitive cell cycling. Thus, lamellipodin is important not only in controlling cellular migration but also for regulating the cell cycle in response to mechanical signals.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular , Proteína Sustrato Asociada a CrK/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Transducción de Señal , Animales , Mecanotransducción Celular , Ratones
3.
J Biol Chem ; 288(44): 32093-105, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24052262

RESUMEN

Filamins are actin-binding and cross-linking proteins that organize the actin cytoskeleton and anchor transmembrane proteins to the cytoskeleton and scaffold signaling pathways. During hematopoietic cell differentiation, transient expression of ASB2α, the specificity subunit of an E3-ubiquitin ligase complex, triggers acute proteasomal degradation of filamins. This led to the proposal that ASB2α regulates hematopoietic cell differentiation by modulating cell adhesion, spreading, and actin remodeling through targeted degradation of filamins. Here, we show that the calponin homology domain 1 (CH1), within the filamin A (FLNa) actin-binding domain, is the minimal fragment sufficient for ASB2α-mediated degradation. Combining an in-depth flow cytometry analysis with mutagenesis of lysine residues within CH1, we find that arginine substitution at each of a cluster of three lysines (Lys-42, Lys-43, and Lys-135) renders FLNa resistant to ASB2α-mediated degradation without altering ASB2α binding. These lysines lie within previously predicted actin-binding sites, and the ASB2α-resistant filamin mutant is defective in targeting to F-actin-rich structures in cells. However, by swapping CH1 with that of α-actinin1, which is resistant to ASB2α-mediated degradation, we generated an ASB2α-resistant chimeric FLNa with normal subcellular localization. Notably, this chimera fully rescues the impaired cell spreading induced by ASB2α expression. Our data therefore reveal ubiquitin acceptor sites in FLNa and establish that ASB2α-mediated effects on cell spreading are due to loss of filamins.


Asunto(s)
Filaminas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Filaminas/genética , Humanos , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/fisiología
4.
J Cell Sci ; 125(Pt 16): 3858-69, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22595522

RESUMEN

Filamins are an important family of actin-binding proteins that, in addition to bundling actin filaments, link cell surface adhesion proteins, signaling receptors and channels to the actin cytoskeleton, and serve as scaffolds for an array of intracellular signaling proteins. Filamins are known to regulate the actin cytoskeleton, act as mechanosensors that modulate tissue responses to matrix density, control cell motility and inhibit activation of integrin adhesion receptors. In this study, we extend the repertoire of filamin activities to include control of extracellular matrix (ECM) degradation. We show that knockdown of filamin increases matrix metalloproteinase (MMP) activity and induces MMP2 activation, enhancing the ability of cells to remodel the ECM and increasing their invasive potential, without significantly altering two-dimensional random cell migration. We further show that within filamin A, the actin-binding domain is necessary, but not sufficient, to suppress the ECM degradation seen in filamin-A-knockdown cells and that dimerization and integrin binding are not required. Filamin mutations are associated with neuronal migration disorders and a range of congenital malformations characterized by skeletal dysplasia and various combinations of cardiac, craniofacial and intestinal anomalies. Furthermore, in breast cancers loss of filamin A has been correlated with increased metastatic potential. Our data suggest that effects on ECM remodeling and cell invasion should be considered when attempting to provide cellular explanations for the physiological and pathological effects of altered filamin expression or filamin mutations.


Asunto(s)
Proteínas Contráctiles/metabolismo , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de Microfilamentos/metabolismo , Actinas/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proteínas Contráctiles/deficiencia , Proteínas Contráctiles/genética , Activación Enzimática , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosarcoma/enzimología , Fibrosarcoma/genética , Filaminas , Técnicas de Silenciamiento del Gen , Humanos , Integrinas/metabolismo , Metaloproteinasa 14 de la Matriz , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Invasividad Neoplásica , Fenotipo , Estructura Terciaria de Proteína
5.
Annu Rev Biophys ; 41: 227-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404683

RESUMEN

Filamins are essential, evolutionarily conserved, modular, multidomain, actin-binding proteins that organize the actin cytoskeleton and maintain extracellular matrix connections by anchoring actin filaments to transmembrane receptors. By cross-linking and anchoring actin filaments, filamins stabilize the plasma membrane, provide cellular cortical rigidity, and contribute to the mechanical stability of the plasma membrane and the cell cortex. In addition to binding actin, filamins interact with more than 90 other binding partners including intracellular signaling molecules, receptors, ion channels, transcription factors, and cytoskeletal and adhesion proteins. Thus, filamins scaffold a wide range of signaling pathways and are implicated in the regulation of a diverse array of cellular functions including motility, maintenance of cell shape, and differentiation. Here, we review emerging structural and functional evidence that filamins are mechanosensors and/or mechanotransducers playing essential roles in helping cells detect and respond to physical forces in their local environment.


Asunto(s)
Movimiento Celular , Proteínas Contráctiles/química , Mecanotransducción Celular , Proteínas de Microfilamentos/química , Transducción de Señal , Animales , Membrana Celular/metabolismo , Forma de la Célula , Proteínas Contráctiles/metabolismo , Citoesqueleto/metabolismo , Filaminas , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/fisiología , Unión Proteica , Estructura Terciaria de Proteína
6.
J Cell Sci ; 124(Pt 15): 2631-41, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21750192

RESUMEN

Filamins are an important family of actin-binding and crosslinking proteins that mediate remodeling of the actin cytoskeleton and maintain extracellular matrix connections by anchoring transmembrane proteins to actin filaments and linking them to intracellular signaling cascades. We recently found that filamins are targeted for proteasomal degradation by the E3 ubiquitin ligase specificity subunit ASBα and that acute degradation of filamins through this ubiquitin-proteasome pathway correlates with cell differentiation. Specifically, in myeloid leukemia cells retinoic-acid-induced expression of ASB2α triggers filamin degradation and recapitulates early events crucial for cell differentiation. ASB2α is thought to link substrates to the ubiquitin transferase machinery; however, the mechanism by which ASB2α interacts with filamin to induce degradation remained unknown. Here, we use cell-based and biochemical assays to show that the subcellular localization of ASB2α to actin-rich structures is dependent on filamin and that the actin-binding domain (ABD) of filamin mediates the interaction with ASB2α. Furthermore, we show that the ABD is necessary and sufficient for ASB2α-mediated filamin degradation. We propose that ASB2α exerts its effect by binding the ABD and mediating its polyubiquitylation, so targeting filamins for degradation. These studies provide the molecular basis for ASB2α-mediated filamin degradation and unravel an important mechanism by which filamin levels can be acutely regulated.


Asunto(s)
Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Células Cultivadas , Proteínas Contráctiles/genética , Cricetinae , Cricetulus , Filaminas , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Immunoblotting , Ratones , Proteínas de Microfilamentos/genética , Unión Proteica , Ubiquitina-Proteína Ligasas/genética
7.
J Biol Chem ; 286(35): 30571-30581, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21737450

RESUMEN

By providing contacts between hematopoietic cells and the bone marrow microenvironment, integrins are implicated in cell adhesion and thereby in control of cell fate of normal and leukemia cells. The ASB2 gene, initially identified as a retinoic acid responsive gene and a target of the promyelocytic leukemia retinoic acid receptor α oncoprotein in acute promyelocytic leukemia cells, encodes two isoforms, a hematopoietic-type (ASB2α) and a muscle-type (ASB2ß) that are involved in hematopoietic and myogenic differentiation, respectively. ASB2α is the specificity subunit of an E3 ubiquitin ligase complex that targets filamins to proteasomal degradation. To examine the relationship of the ASB2α structure to E3 ubiquitin ligase function, functional assays and molecular modeling were performed. We show that ASB2α, through filamin A degradation, enhances adhesion of hematopoietic cells to fibronectin, the main ligand of ß1 integrins. Furthermore, we demonstrate that a short N-terminal region specific to ASB2α, together with ankyrin repeats 1 to 10, is necessary for association of ASB2α with filamin A. Importantly, the ASB2α N-terminal region comprises a 9-residue segment with predicted structural homology to the filamin-binding motifs of migfilin and ß integrins. Together, these data provide new insights into the molecular mechanisms of ASB2α binding to filamin.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Integrinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Animales , Adhesión Celular , Fibronectinas/metabolismo , Células HeLa , Humanos , Ratones , Músculos/metabolismo , Células 3T3 NIH , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato
8.
Zygote ; 18(1): 17-26, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19500445

RESUMEN

In Strongylocentrotus purpuratus the hyalins are a set of three to four rather large glycoproteins (hereafter referred to as 'hyalin'), which are the major constituents of the hyaline layer, the developing sea urchin embryo's extracellular matrix. Recent research from our laboratories has shown that hyalin is a cell adhesion molecule involved in sea urchin embryo-specific cellular interactions. Other laboratories have shown it to consist of 2-3% carbohydrate and a cloned, sequenced fragment demonstrated repeat domains (HYR) and non-repeat regions. Interest in this molecule has increased because HYR has been identified in organisms as diverse as bacteria, flies, worms, mice and humans, as well as sea urchins. Our laboratories have shown that hyalin appears to mediate a specific cellular interaction that has interested investigators for over a century, archenteron elongation/attachment to the blastocoel roof. We have shown this finding by localizing hyalin on the two components of the cellular interaction and by showing that hyalin and anti-hyalin antibody block the cellular interaction using a quantitative microplate assay. The microplate assay, however, has limitations because it does not directly assess hyalin's effects on the adhesion of the two components of the interaction. Here we have used an elegant direct assay that avoids the limitations, in which we microdissected the two components of the adhesive interaction and tested their re-adhesion to each other, thereby avoiding possible factors in the whole embryos that could confound or confuse results. Using both assays, we found that mild periodate treatment (6 h to 24 h in sodium acetate buffer with 0.2 M sodium periodate at 4 degrees C in the dark) of hyalin eliminates its ability to block the cellular interaction, suggesting that the carbohydrate component(s) may be involved in hyalin's specific adhesive function. This first step is important in identifying the molecular mechanisms of a well known cellular interaction in the NIH-designated sea urchin embryo model, a system that has led to the discovery of scores of physiological mechanisms, including those involved in human health and disease.


Asunto(s)
Gastrulación , Hialina/química , Erizos de Mar/química , Erizos de Mar/embriología , Animales , Adhesión Celular , Erizos de Mar/citología
9.
J Struct Biol ; 170(1): 157-63, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19963065

RESUMEN

Formation of a heterotrimeric IPP complex composed of integrin-linked kinase (ILK), the LIM domain protein PINCH, and parvin is important for signaling through integrin adhesion receptors. Mammals possess two PINCH genes that are expressed simultaneously in many tissues. PINCH1 and PINCH2 have overlapping functions and can compensate for one another in many settings; however, isoform-specific functions have been reported and it is proposed that association with a PINCH1- or PINCH2-containing IPP complex may provide a bifurcation point in integrin signaling promoting different cellular responses. Here we report that the LIM1 domains of PINCH1 and PINCH2 directly compete for the same binding site on the ankyrin repeat domain (ARD) of ILK. We determined the 1.9A crystal structure of the PINCH2 LIM1 domain complexed with the ARD of ILK, and show that disruption of this interface by point mutagenesis reduces binding in vitro and alters localization of PINCH2 in cells. These studies provide further evidence for the role of the PINCH LIM1 domain in association with ILK and highlight direct competition as one mechanism for regulating which PINCH isoform predominates in IPP complexes. Differential regulation of PINCH1 and PINCH2 expression may therefore provide a means for altering cellular integrin signaling pathways.


Asunto(s)
Unión Competitiva , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Repetición de Anquirina/genética , Cristalización , Regulación de la Expresión Génica/genética , Proteínas con Dominio LIM , Proteínas de la Membrana , Datos de Secuencia Molecular , Mutagénesis , Proteínas Serina-Treonina Quinasas/genética
10.
PLoS One ; 4(11): e7830, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19915675

RESUMEN

Mammalian filamins (FLNs) are a family of three large actin-binding proteins. FLNa, the founding member of the family, was implicated in migration by cell biological analyses and the identification of FLNA mutations in the neuronal migration disorder periventricular heterotopia. However, recent knockout studies have questioned the relevance of FLNa to cell migration. Here we have used shRNA-mediated knockdown of FLNa, FLNb or FLNa and FLNb, or, alternatively, acute proteasomal degradation of all three FLNs, to generate FLN-deficient cells and assess their ability to migrate. We report that loss of FLNa or FLNb has little effect on migration but that knockdown of FLNa and FLNb, or proteolysis of all three FLNs, impairs migration. The observed defect is primarily a deficiency in initiation of motility rather than a problem with maintenance of locomotion speed. FLN-deficient cells are also impaired in spreading. Re-expression of full length FLNa, but not re-expression of a mutated FLNa lacking immunoglobulin domains 19 to 21, reverts both the spreading and the inhibition of initiation of migration.Our results establish a role for FLNs in cell migration and spreading and suggest that compensation by other FLNs may mask phenotypes in single knockout or knockdown cells. We propose that interactions between FLNs and transmembrane or signalling proteins, mediated at least in part by immunoglobulin domains 19 to 21 are important for both cell spreading and initiation of migration.


Asunto(s)
Proteínas Contráctiles/fisiología , Proteínas de Microfilamentos/fisiología , Actinas/química , Animales , Línea Celular Tumoral , Movimiento Celular , Proteínas Contráctiles/metabolismo , Filaminas , Humanos , Inmunoglobulinas/química , Células Jurkat , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Mutación , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo
11.
Blood ; 112(13): 5130-40, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18799729

RESUMEN

The ordered series of proliferation and differentiation from hematopoietic progenitor cells is disrupted in leukemia, resulting in arrest of differentiation at immature proliferative stages. Characterizing the molecular basis of hematopoietic differentiation is therefore important for understanding and treating disease. Retinoic acid induces expression of ankyrin repeat-containing protein with a suppressor of cytokine signaling box 2 (ASB2) in acute promyelocytic leukemia cells, and ASB2 expression inhibits growth and promotes commitment, recapitulating an early step critical for differentiation. ASB2 is the specificity subunit of an E3 ubiquitin ligase complex and is proposed to exert its effects by regulating the turnover of specific proteins; however, no ASB2 substrates had been identified. Here, we report that ASB2 targets the actin-binding proteins filamin A and B for proteasomal degradation. Knockdown of endogenous ASB2 in leukemia cells delays retinoic acid-induced differentiation and filamin degradation; conversely, ASB2 expression in leukemia cells induces filamin degradation. ASB2 expression inhibits cell spreading, and this effect is recapitulated by knocking down both filamin A and filamin B. Thus, we suggest that ASB2 may regulate hematopoietic cell differentiation by modulating cell spreading and actin remodeling through targeting of filamins for degradation.


Asunto(s)
Proteínas Contráctiles/metabolismo , Leucemia/patología , Proteínas de Microfilamentos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Actinas/metabolismo , Adhesión Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proteínas Contráctiles/genética , Filaminas , Humanos , Leucemia/tratamiento farmacológico , Proteínas de Microfilamentos/genética , ARN Interferente Pequeño/farmacología , Proteínas Supresoras de la Señalización de Citocinas/genética , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...