Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 435(2): 113930, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38237846

RESUMEN

The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/patología , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral/genética
2.
Development ; 146(9)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30967426

RESUMEN

Establishing apical-basal epithelial cell polarity is fundamental for mammary gland duct morphogenesis during mammalian development. While the focal adhesion adapter protein paxillin is a well-characterized regulator of mesenchymal cell adhesion signaling, F-actin cytoskeleton remodeling and single cell migration, its role in epithelial tissue organization and mammary gland morphogenesis in vivo has not been investigated. Here, using a newly developed paxillin conditional knockout mouse model with targeted ablation in the mammary epithelium, in combination with ex vivo three-dimensional organoid and acini cultures, we identify new roles for paxillin in the establishment of apical-basal epithelial cell polarity and lumen formation, as well as mammary gland duct diameter and branching. Paxillin is shown to be required for the integrity and apical positioning of the Golgi network, Par complex and the Rab11/MyoVb trafficking machinery. Paxillin depletion also resulted in reduced levels of apical acetylated microtubules, and rescue experiments with the HDAC6 inhibitor tubacin highlight the central role for paxillin-dependent regulation of HDAC6 activity and associated microtubule acetylation in controlling epithelial cell apical-basal polarity and tissue branching morphogenesis.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Paxillin/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Polaridad Celular/genética , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Ratones , Microtúbulos/metabolismo , Morfogénesis/genética , Morfogénesis/fisiología , Paxillin/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
3.
Development ; 144(21): 4002-4014, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935710

RESUMEN

Paxillin and Hic-5 are homologous focal adhesion adaptor proteins that coordinate cytoskeletal rearrangements in response to integrin signaling, but their role(s) in cortical development are unknown. Here, we find that Hic-5-deficient mice are postnatal viable with normal cortical layering. Mice with a neural-specific deletion of paxillin are also postnatal viable, but show evidence of a cortical neuron migration delay that is evident pre- and perinatally, but is not detected at postnatal day 35 (P35). This phenotype is not modified by Hic-5 deficiency (double knockout). Specific deletion of paxillin in postmitotic neurons using Nex-Cre-mediated recombination as well as in utero electroporation of a Cre-expression construct identified a cell-autonomous requirement for paxillin in migrating neurons. Paxillin-deficient neurons have shorter leading processes that exhibited multiple swellings in comparison with control. Multiphoton imaging revealed that paxillin-deficient neurons migrate ∼30% slower than control neurons. This phenotype is similar to that produced by deletion of focal adhesion kinase (FAK), a signaling partner of paxillin, and suggests that paxillin and FAK function cell-autonomously to control migrating neuron morphology and speed during cortical development.


Asunto(s)
Movimiento Celular , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Adhesiones Focales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Paxillin/metabolismo , Alelos , Animales , Movimiento Celular/genética , Forma de la Célula , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Integrasas/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones Noqueados , Células-Madre Neurales/metabolismo , Especificidad de Órganos , Paxillin/deficiencia , Paxillin/genética
4.
Dev Biol ; 425(1): 70-84, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28315297

RESUMEN

Paxillin (Pxn) is a key adapter protein and signaling regulator at sites of cell-extracellular matrix (ECM) adhesion. Here, we investigated the role of Pxn during vertebrate development using the zebrafish embryo as a model system. We have characterized two Pxn genes, pxna and pxnb, in zebrafish that are maternally supplied and expressed in multiple tissues. Gene editing and antisense gene knockdown approaches were used to uncover Pxn functions during zebrafish development. While mutation of either pxna or pxnb alone did not cause gross embryonic phenotypes, double mutants lacking maternally supplied pxna or pxnb displayed defects in cardiovascular, axial, and skeletal muscle development. Transient knockdown of Pxn proteins resulted in similar defects. Irregular myotome shape and ECM composition were observed, suggesting an "inside-out" signaling role for Paxillin genes in the development of myotendinous junctions. Inhibiting non-muscle Myosin-II during somitogenesis altered the subcellular localization of Pxn protein and phenocopied pxn gene loss-of-function. This indicates that Paxillin genes are effectors of actomyosin contractility-driven morphogenesis of trunk musculature in zebrafish. Together, these results reveal new functions for Pxn during muscle development and provide novel genetic models to elucidate Pxn functions.


Asunto(s)
Actomiosina/metabolismo , Morfogénesis , Músculo Esquelético/metabolismo , Paxillin/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Western Blotting , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Técnicas de Silenciamiento del Gen , Microscopía Confocal , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Mutación , Paxillin/genética , Isoformas de Proteínas/genética , Homología de Secuencia de Ácido Nucleico , Somitos/embriología , Somitos/metabolismo , Imagen de Lapso de Tiempo/métodos , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-38801098

RESUMEN

Focal adhesions serve as structural and signaling hubs, facilitating bidirectional communication at the cell-extracellular matrix interface. Paxillin and the related Hic-5 (TGFß1i1) are adaptor/scaffold proteins that recruit numerous structural and regulatory proteins to focal adhesions, where they perform both overlapping and discrete functions. In this study, paxillin and Hic-5 were expressed in U2OS osteosarcoma cells as biotin ligase (BioID2) fusion proteins and used as bait proteins for proximity-dependent biotinylation in order to directly compare their respective interactomes. The fusion proteins localized to both focal adhesions and the centrosome, resulting in biotinylation of components of each of these structures. Biotinylated proteins were purified and analyzed by mass spectrometry. The list of proximity interactors for paxillin and Hic-5 comprised numerous shared core focal adhesion proteins that likely contribute to their similar functions in cell adhesion and migration, as well as proteins unique to paxillin and Hic-5 that have been previously localized to focal adhesions, the centrosome, or the nucleus. Western blotting confirmed biotinylation and enrichment of FAK and vinculin, known interactors of Hic-5 and paxillin, as well as several potentially unique proximity interactors of Hic-5 and paxillin, including septin 7 and ponsin, respectively. Further investigation into the functional relationship between the unique interactors and Hic-5 or paxillin may yield novel insights into their distinct roles in cell migration.

6.
J Biol Chem ; 287(44): 37309-20, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22955285

RESUMEN

Dysregulation of cell adhesion and motility is known to be an important factor in the development of tumor malignancy. Actopaxin (α-parvin) is a paxillin, integrin-linked kinase, and F-actin binding focal adhesion protein with several serine phosphorylation sites in the amino terminus that contribute to the regulation of cell spreading and migration. Here, phosphorylation of actopaxin is shown to contribute to the regulation of matrix degradation and cell invasion. Osteosarcoma cells stably expressing wild type (WT), nonphosphorylatable (Quint), and phosphomimetic (S4D/S8D) actopaxin demonstrate that actopaxin phosphorylation is necessary for efficient Src and matrix metalloproteinase-driven degradation of extracellular matrix. Rac1 was found to be required for actopaxin-induced matrix degradation whereas inhibition of myosin contractility promoted degradation in the phosphomutant-expressing Quint cells, indicating that a balance of Rho GTPase signaling and regulation of cellular tension are important for the process. Furthermore, actopaxin forms a complex with the Rac1/Cdc42 GEF ß-PIX and Rac1/Cdc42 effector PAK1, to regulate actopaxin-dependent matrix degradation. Actopaxin phosphorylation is elevated in the invasive breast cancer cell line MDA-MB-231 compared with normal breast epithelial MCF10A cells. Expression of the nonphosphorylatable Quint actopaxin in MDA-MB-231 cells inhibits cell invasion whereas overexpression of WT actopaxin promotes invasion in MCF10A cells. Taken together, this study demonstrates a new role for actopaxin phosphorylation in matrix degradation and cell invasion via regulation of Rho GTPase signaling.


Asunto(s)
Matriz Extracelular/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias/patología , Procesamiento Proteico-Postraduccional , Proteolisis , Línea Celular Tumoral , Movimiento Celular , Inhibidores Enzimáticos/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Metaloproteinasas de la Matriz Secretadas/antagonistas & inhibidores , Metaloproteinasas de la Matriz Secretadas/metabolismo , Miosinas/metabolismo , Invasividad Neoplásica , Neoplasias/enzimología , Neoplasias/metabolismo , Fosforilación , Factores de Intercambio de Guanina Nucleótido Rho , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
7.
FASEB J ; 26(6): 2592-606, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22441983

RESUMEN

Focal adhesion (FA) formation and disassembly play an essential role in adherence and migration of endothelial cells. These processes are highly regulated and involve various signaling molecules that are not yet completely identified. Lnk [Src homology 2-B3 (SH2B3)] belongs to a family of SH2-containing proteins with important adaptor functions. In this study, we showed that Lnk distribution follows that of vinculin, localizing Lnk in FAs. Inhibition of Lnk by RNA interference resulted in decreased spreading, whereas sustained expression dramatically increases the number of focal and cell-matrix adhesions. We demonstrated that Lnk expression impairs FA turnover and cell migration and regulates ß1-integrin-mediated signaling via Akt and GSK3ß phosphorylation. Moreover, the α-parvin protein was identified as one of the molecular targets of Lnk responsible for impaired FA dynamics and cell migration. Finally, we established the ILK protein as a new molecular partner for Lnk and proposed a model in which Lnk regulates α-parvin expression through its interaction with ILK. Collectively, our results underline the adaptor Lnk as a novel and effective key regulator of integrin-mediated signaling controlling endothelial cell adhesion and migration.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Adhesiones Focales/fisiología , Proteínas de Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/fisiología , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Uniones Célula-Matriz/metabolismo , Adhesiones Focales/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta1/fisiología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
Mol Biol Cell ; 34(7): ar65, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37043310

RESUMEN

Rab GTPase-mediated vesicle trafficking of cell surface proteins, including integrins, through endocytic and recycling pathways is important in controlling cell-extracellular matrix interactions during cell migration. The focal adhesion adaptor protein, paxillin, plays a central role in regulating adhesion dynamics and was previously shown to promote anterograde vesicle trafficking through modulation of microtubule acetylation via its inhibition of the deacetylase HDAC6. The role of paxillin in retrograde trafficking is unknown. Herein, we identified a role for paxillin in the modulation of the Rab5 GTPase, which is necessary for regulating early endosome dynamics and focal adhesion turnover. Using MDA-MB-231 breast cancer cells and paxillin (-/-) fibroblasts, paxillin was shown to impact Rab5-associated vesicle size and distribution, as well as Rab5 GTPase activity, through its modulation of HDAC6. Using a combination of real-time imaging and particle tracking analysis, paxillin was shown to promote Rab5-associated vesicle motility through inhibition of HDAC6-mediated micro-tubule deacetylation, along with the localization of active integrin to focal adhesions.


Asunto(s)
Adhesiones Focales , Procesamiento Proteico-Postraduccional , Humanos , Paxillin/metabolismo , Acetilación , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Microtúbulos/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Adhesión Celular/fisiología
9.
Dev Biol ; 349(2): 225-37, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21034731

RESUMEN

Signaling through cell adhesion complexes plays a critical role in coordinating cytoskeletal remodeling necessary for efficient cell migration. During embryonic development, normal morphogenesis depends on a series of concerted cell movements; but the roles of cell adhesion signaling during these movements are poorly understood. The transparent zebrafish embryo provides an excellent system to study cell migration during development. Here, we have identified zebrafish git2a and git2b, two new members of the GIT family of genes that encode ArfGAP proteins associated with cell adhesions. Loss-of-function studies revealed an essential role for Git2a in zebrafish cell movements during gastrulation. Time-lapse microscopy analysis demonstrated that antisense depletion of Git2a greatly reduced or arrested cell migration towards the vegetal pole of the embryo. These defects were rescued by expression of chicken GIT2, indicating a specific and conserved role for Git2 in controlling embryonic cell movements. Git2a knockdown embryos showed defects in cell morphology that were associated with reduced cell contractility. We show that Git2a is required for phosphorylation of myosin light chain (MLC), which regulates myosin II-mediated cell contractility. Consistent with this, embryos treated with Blebbistatin-a small molecule inhibitor for myosin II activity-exhibited cell movement defects similar to git2a knockdown embryos. These observations provide in vivo evidence of a physiologic role for Git2a in regulating cell morphogenesis and directed cell migration via myosin II activation during zebrafish embryonic development.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Desarrollo Embrionario/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Morfogénesis/fisiología , Transducción de Señal/fisiología , Pez Cebra/embriología , Animales , Secuencia de Bases , Proteínas Activadoras de GTPasa/genética , Técnicas de Silenciamiento del Gen , Compuestos Heterocíclicos de 4 o más Anillos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Datos de Secuencia Molecular , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Filogenia , Análisis de Secuencia de ADN , Imagen de Lapso de Tiempo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Mol Biol Cell ; 33(2): ar14, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34851720

RESUMEN

Distant organ metastasis is linked to poor prognosis during cancer progression. The expression level of the focal adhesion adapter protein paxillin varies among different human cancers, but its role in tumor progression is unclear. Herein we utilize a newly generated PyMT mammary tumor mouse model with conditional paxillin ablation in breast tumor epithelial cells, combined with in vitro three-dimensional (3D) tumor organoids invasion analysis and 2D calcium switch assays, to assess the roles for paxillin in breast tumor cell invasion. Paxillin had little effect on primary tumor initiation and growth but is critical for the formation of distant lung metastasis. In paxillin-depleted 3D tumor organoids, collective cell invasion was substantially perturbed. The 2D cell culture revealed paxillin-dependent stabilization of adherens junctions (AJ). Mechanistically, paxillin is required for AJ assembly through facilitating E-cadherin endocytosis and recycling and HDAC6-mediated microtubule acetylation. Furthermore, Rho GTPase activity analysis and rescue experiments with a RhoA activator or Rac1 inhibitor suggest paxillin is potentially regulating the E-cadherin-dependent junction integrity and contractility through control of the balance of RhoA and Rac1 activities. Together, these data highlight new roles for paxillin in the regulation of cell-cell adhesion and collective tumor cell migration to promote the formation of distance organ metastases.


Asunto(s)
Uniones Adherentes/fisiología , Invasividad Neoplásica/genética , Paxillin/metabolismo , Uniones Adherentes/genética , Animales , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/metabolismo , Femenino , Adhesiones Focales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Paxillin/genética , Proteínas de Unión al GTP rho/metabolismo
11.
Nat Cell Biol ; 6(2): 154-61, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14743221

RESUMEN

Cell migration is a complex, highly regulated process that involves the continuous formation and disassembly of adhesions (adhesion turnover). Adhesion formation takes place at the leading edge of protrusions, whereas disassembly occurs both at the cell rear and at the base of protrusions. Despite the importance of these processes in migration, the mechanisms that regulate adhesion formation and disassembly remain largely unknown. Here we develop quantitative assays to measure the rate of incorporation of molecules into adhesions and the departure of these proteins from adhesions. Using these assays, we show that kinases and adaptor molecules, including focal adhesion kinase (FAK), Src, p130CAS, paxillin, extracellular signal-regulated kinase (ERK) and myosin light-chain kinase (MLCK) are critical for adhesion turnover at the cell front, a process central to migration.


Asunto(s)
Adhesión Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Movimiento Celular/fisiología , Proteína Sustrato Asociada a CrK , Proteínas del Citoesqueleto/genética , Fibroblastos/citología , Fibroblastos/fisiología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Quinasa de Cadena Ligera de Miosina/genética , Paxillin , Fosfoproteínas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Familia-src Quinasas/genética
12.
Int Rev Cell Mol Biol ; 355: 1-52, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32859368

RESUMEN

The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.


Asunto(s)
Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Invasividad Neoplásica , Neoplasias/metabolismo , Paxillin/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Neoplasias/patología , Paxillin/fisiología , Transducción de Señal
13.
Curr Biol ; 16(14): 1375-85, 2006 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-16860736

RESUMEN

BACKGROUND: Integrin signaling, stimulated by cell adhesion to the extracellular matrix, plays a critical role in coordinating changes in cell morphology and migration. The requisite remodeling of the cytoskeleton is controlled by the Rho family of small GTPases, which are, in turn, regulated via activation by guanine-nucleotide exchange factors (GEFs) and inactivation by GTPase-activating proteins (GAPs). However, the mechanisms contributing to the precise spatial and temporal regulation of these Rho GTPase modulators remain poorly understood. RESULTS: The Cdc42/Rac GAP CdGAP has previously been implicated as an inhibitor of growth-factor-induced lamellipodia formation. Herein, CdGAP is shown to localize to focal adhesions, potentially through its direct association with the amino terminus of actopaxin, a paxillin and actin binding protein. CdGAP activity is regulated in an adhesion-dependent manner and, through the overexpression of wild-type CdGAP and a GAP-deficient mutant, as well as RNA interference, is shown to be required for normal cell spreading, polarized lamellipodia formation, and cell migration. Introduction of an actopaxin mutant defective for CdGAP binding, or reduction of actopaxin by using RNAi, significantly attenuated these effects. CONCLUSIONS: We have established that CdGAP is an important regulator of integrin-induced Rho family signaling to the cytoskeleton and that its interaction with the focal-adhesion protein actopaxin is critical for the correct spatial and/or temporal regulation of CdGAP function. A complete understanding of the coordination of signaling events downstream of integrin engagement with the extracellular matrix will provide valuable insight into the regulation of cell migration during processes such as wound repair, development, and tumor cell metastasis.


Asunto(s)
Actinina/metabolismo , Movimiento Celular/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Integrinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Polaridad Celular/genética , Polaridad Celular/fisiología , Forma de la Célula/fisiología , Adhesiones Focales/metabolismo , Proteínas Activadoras de GTPasa/análisis , Proteínas Activadoras de GTPasa/genética , Proteínas Fluorescentes Verdes/análisis , Humanos , Proteínas Luminiscentes/análisis , Paxillin/genética , Paxillin/metabolismo , Proteína Fluorescente Roja
14.
J Cell Biol ; 166(6): 901-12, 2004 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-15353548

RESUMEN

Actopaxin is an actin and paxillin binding protein that localizes to focal adhesions. It regulates cell spreading and is phosphorylated during mitosis. Herein, we identify a role for actopaxin phosphorylation in cell spreading and migration. Stable clones of U2OS cells expressing actopaxin wild-type (WT), nonphosphorylatable, and phosphomimetic mutants were developed to evaluate actopaxin function. All proteins targeted to focal adhesions, however the nonphosphorylatable mutant inhibited spreading whereas the phosphomimetic mutant cells spread more efficiently than WT cells. Endogenous and WT actopaxin, but not the nonphosphorylatable mutant, were phosphorylated in vivo during cell adhesion/spreading. Expression of the nonphosphorylatable actopaxin mutant significantly reduced cell migration, whereas expression of the phosphomimetic increased cell migration in scrape wound and Boyden chamber migration assays. In vitro kinase assays demonstrate that extracellular signal-regulated protein kinase phosphorylates actopaxin, and treatment of U2OS cells with the MEK1 inhibitor UO126 inhibited adhesion-induced phosphorylation of actopaxin and also inhibited cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Actinina , Secuencia de Aminoácidos , Butadienos/farmacología , Línea Celular Tumoral , Tamaño de la Célula/genética , Células Clonales , Inhibidores Enzimáticos/farmacología , Adhesiones Focales/metabolismo , Humanos , Cinética , Proteínas de Microfilamentos/química , Mutación , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína
15.
J Cell Biol ; 165(3): 371-81, 2004 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15138291

RESUMEN

Cells lacking vinculin are highly metastatic and motile. The reasons for this finding have remained unclear. Both enhanced survival and motility are critical to metastasis. Here, we show that vinculin null (vin-/-) cells and cells expressing a vinculin Y822F mutant have increased survival due to up-regulated activity of extracellular signal-regulated kinase (ERK). This increase is shown to result from vinculin's modulation of paxillin-FAK interactions. A vinculin fragment (amino acids 811-1066) containing the paxillin binding site restored apoptosis and suppressed ERK activity in vin-/- cells. Both vinY822F and vin-/- cells exhibit increased interaction between paxillin and focal adhesion kinase (FAK) and increased paxillin and FAK phosphorylation. Transfection with paxillin Y31FY118F dominant-negative mutant in these cells inhibits ERK activation and restores apoptosis. The enhanced motility of vin-/- and vinY822F cells is also shown to be due to a similar mechanism. Thus, vinculin regulates survival and motility via ERK by controlling the accessibility of paxillin for FAK interaction.


Asunto(s)
Movimiento Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Vinculina/fisiología , Animales , Apoptosis/genética , Sitios de Unión/genética , Adhesión Celular/genética , Supervivencia Celular/genética , Proteínas del Citoesqueleto/genética , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Ratones , Mutación/genética , Metástasis de la Neoplasia/genética , Células Madre Neoplásicas , Paxillin , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfoproteínas/genética , Fosforilación , Transfección , Regulación hacia Arriba/genética , Vinculina/genética , Vinculina/metabolismo
16.
Mol Biol Cell ; 30(11): 1298-1313, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30893012

RESUMEN

Fibroblasts transformed by the proto-oncogene Src form individual invadopodia that can spontaneously self-organize into large matrix-degrading superstructures called rosettes. However, the mechanisms by which the invadopodia can spatiotemporally reorganize their architecture is not well understood. Here, we show that Hic-5, a close relative of the scaffold protein paxillin, is essential for the formation and organization of rosettes in active Src-transfected NIH3T3 fibroblasts and cancer-associated fibroblasts. Live cell imaging, combined with domain-mapping analysis of Hic-5, identified critical motifs as well as phosphorylation sites that are required for the formation and dynamics of rosettes. Using pharmacological inhibition and mutant expression, we show that FAK kinase activity, along with its proximity to and potential interaction with the LD2,3 motifs of Hic-5, is necessary for rosette formation. Invadopodia dynamics and their coalescence into rosettes were also dependent on Rac1, formin, and myosin II activity. Superresolution microscopy revealed the presence of formin FHOD1 and INF2-mediated unbranched radial F-actin fibers emanating from invadopodia and rosettes, which may facilitate rosette formation. Collectively, our data highlight a novel role for Hic-5 in orchestrating the organization of invadopodia into higher-order rosettes, which may promote the localized matrix degradation necessary for tumor cell invasion.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas con Dominio LIM/metabolismo , Podosomas/metabolismo , Procesamiento Proteico-Postraduccional , Familia-src Quinasas/genética , Actinas/metabolismo , Actinas/fisiología , Animales , Línea Celular Transformada , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Fetales/metabolismo , Proteínas Fetales/fisiología , Fibroblastos/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Forminas/metabolismo , Forminas/fisiología , Proteínas con Dominio LIM/fisiología , Ratones , Miosina Tipo II/metabolismo , Miosina Tipo II/fisiología , Células 3T3 NIH , Neuropéptidos/metabolismo , Neuropéptidos/fisiología , Fosforilación , Podosomas/fisiología , Formación de Roseta , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/fisiología
17.
Mol Biol Cell ; 30(25): 3037-3056, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31644368

RESUMEN

Focal adhesion (FA)-stimulated reorganization of the F-actin cytoskeleton regulates cellular size, shape, and mechanical properties. However, FA cross-talk with the intermediate filament cytoskeleton is poorly understood. Genetic ablation of the FA-associated scaffold protein Hic-5 in mouse cancer-associated fibroblasts (CAFs) promoted a dramatic collapse of the vimentin network, which was rescued following EGFP-Hic-5 expression. Vimentin collapse correlated with a loss of detergent-soluble vimentin filament precursors and decreased vimentin S72/S82 phosphorylation. Additionally, fluorescence recovery after photobleaching analysis indicated impaired vimentin dynamics. Microtubule (MT)-associated EB1 tracking and Western blotting of MT posttranslational modifications indicated no change in MT dynamics that could explain the vimentin collapse. However, pharmacological inhibition of the RhoGTPase Cdc42 in Hic-5 knockout CAFs rescued the vimentin collapse, while pan-formin inhibition with SMIFH2 promoted vimentin collapse in Hic-5 heterozygous CAFs. Our results reveal novel regulation of vimentin organization/dynamics by the FA scaffold protein Hic-5 via modulation of RhoGTPases and downstream formin activity.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Adhesiones Focales/metabolismo , Proteínas con Dominio LIM/metabolismo , Vimentina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/genética , Células Cultivadas , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Fosforilación , Factores de Transcripción/metabolismo
18.
PLoS One ; 14(2): e0211408, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30759123

RESUMEN

Cell motility is critical to biological processes from wound healing to cancer metastasis to embryonic development. The involvement of organelles in cell motility is well established, but the role of organelle positional reorganization in cell motility remains poorly understood. Here we present an automated image analysis technique for tracking the shape and motion of Golgi bodies and cell nuclei. We quantify the relationship between nuclear orientation and the orientation of the Golgi body relative to the nucleus before, during, and after exposure of mouse fibroblasts to a controlled change in cell substrate topography, from flat to wrinkles, designed to trigger polarized motility. We find that the cells alter their mean nuclei orientation, in terms of the nuclear major axis, to increasingly align with the wrinkle direction once the wrinkles form on the substrate surface. This change in alignment occurs within 8 hours of completion of the topographical transition. In contrast, the position of the Golgi body relative to the nucleus remains aligned with the pre-programmed wrinkle direction, regardless of whether it has been fully established. These findings indicate that intracellular positioning of the Golgi body precedes nuclear reorientation during mouse fibroblast directed migration on patterned substrates. We further show that both processes are Rho-associated kinase (ROCK) mediated as they are abolished by pharmacologic ROCK inhibition whereas mouse fibroblast motility is unaffected. The automated image analysis technique introduced could be broadly employed in the study of polarization and other cellular processes in diverse cell types and micro-environments. In addition, having found that the nuclei Golgi vector may be a more sensitive indicator of substrate features than the nuclei orientation, we anticipate the nuclei Golgi vector to be a useful metric for researchers studying the dynamics of cell polarity in response to different micro-environments.


Asunto(s)
Movimiento Celular , Núcleo Celular/ultraestructura , Aparato de Golgi/ultraestructura , Imagen de Lapso de Tiempo/métodos , Animales , Polaridad Celular , Células Cultivadas , Fibroblastos , Ratones
19.
Mol Biol Cell ; 16(9): 4316-28, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16000375

RESUMEN

The ArfGAP paxillin kinase linker (PKL)/G protein-coupled receptor kinase-interacting protein (GIT)2 has been implicated in regulating cell spreading and motility through its transient recruitment of the p21-activated kinase (PAK) to focal adhesions. The Nck-PAK-PIX-PKL protein complex is recruited to focal adhesions by paxillin upon integrin engagement and Rac activation. In this report, we identify tyrosine-phosphorylated PKL as a protein that associates with the SH3-SH2 adaptor Nck, in a Src-dependent manner, after cell adhesion to fibronectin. Both cell adhesion and Rac activation stimulated PKL tyrosine phosphorylation. PKL is phosphorylated on tyrosine residues 286/392/592 by Src and/or FAK and these sites are required for PKL localization to focal adhesions and for paxillin binding. The absence of either FAK or Src-family kinases prevents PKL phosphorylation and suppresses localization of PKL but not GIT1 to focal adhesions after Rac activation. Expression of an activated FAK mutant in the absence of Src-family kinases partially restores PKL localization, suggesting that Src activation of FAK is required for PKL phosphorylation and localization. Overexpression of the nonphosphorylated GFP-PKL Triple YF mutant stimulates cell spreading and protrusiveness, similar to overexpression of a paxillin mutant that does not bind PKL, suggesting that failure to recruit PKL to focal adhesions interferes with normal cell spreading and motility.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Adhesiones Focales/enzimología , Proteínas Activadoras de GTPasa/metabolismo , Fosfoproteínas/metabolismo , Familia-src Quinasas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Adhesiones Focales/metabolismo , Genes Reporteros , Péptidos y Proteínas de Señalización Intercelular , Ratones , Proteínas Oncogénicas/metabolismo , Paxillin/metabolismo , Fosforilación , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Tirosina/metabolismo , Familia-src Quinasas/genética
20.
Oncogene ; 37(13): 1699-1713, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29348458

RESUMEN

The linearization of the stromal extracellular matrix (ECM) by cancer-associated fibroblasts (CAFs) facilitates tumor cell growth and metastasis. However, the mechanism by which the ECM is remodeled is not fully understood. Hic-5 (TGFß1i1), a focal adhesion scaffold protein, has previously been reported to be crucial for stromal ECM deposition and remodeling in vivo. Herein we show that CAFs lacking Hic-5 exhibit a significant reduction in the ability to form fibrillar adhesions, a specialized form of focal adhesion that promote fibronectin fibrillogenesis. Hic-5 was found to promote fibrillar adhesion formation through a newly characterized interaction with tensin1. Furthermore, Src-dependent phosphorylation of Hic-5 facilitated the interaction with tensin1 to prevent ß1 integrin internalization and trafficking to the lysosome. The interaction between Hic-5 and tensin1 was mechanosensitive, promoting fibrillar adhesion formation and fibronectin fibrillogenesis in a rigidity-dependent fashion. Importantly, this Src-dependent mechanism was conserved in three-dimensional (3D) ECM environments. Immunohistochemistry of tensin1 showed enrichment in CAFs in vivo, which was abrogated upon deletion of Hic-5. Interestingly, elevated Hic-5 expression correlates with reduced distant metastasis-free survival in patients with basal-like, HER2+ and grade 3 tumors. Thus, we have identified Hic-5 as a crucial regulator of ECM remodeling in CAFs by promoting fibrillar adhesion formation through a novel interaction with tensin1.


Asunto(s)
Adhesión Celular , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas con Dominio LIM/fisiología , Neoplasias/metabolismo , Tensinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Adhesión Celular/genética , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/patología , Adhesiones Focales/genética , Técnicas de Silenciamiento del Gen , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neoplasias/genética , Neoplasias/patología , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA