Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arthritis Res Ther ; 16(5): 464, 2014 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-25280866

RESUMEN

INTRODUCTION: Synovial fibroblasts invade cartilage and bone, leading to joint destruction in rheumatoid arthritis. However, the mechanisms that regulate synovial fibroblast invasion are not well understood. Focal adhesion kinase (FAK) has been implicated in cellular invasion in several cell types, and FAK inhibitors are in clinical trials for cancer treatment. Little is known about the role of FAK in inflammatory arthritis, but, given its expression in synovial tissue, its known role in invasion in other cells and the potential clinical availability of FAK inhibitors, it is important to determine if FAK contributes to synovial fibroblast invasion and inflammatory arthritis. METHODS: After treatment with FAK inhibitors, invasiveness of human rheumatoid synovial fibroblasts was determined with Matrigel invasion chambers. Migration and focal matrix degradation, two components of cellular invasion, were assessed in FAK-inhibited rheumatoid synovial fibroblasts by transwell assay and microscopic examination of fluorescent gelatin degradation, respectively. Using mice with tumor necrosis factor α (TNFα)-induced arthritis in which fak could be inducibly deleted, invasion and migration by FAK-deficient murine arthritic synovial fibroblasts were determined as described above and arthritis was clinically and pathologically scored in FAK-deficient mice. RESULTS: Inhibition of FAK in human rheumatoid synovial fibroblasts impaired cellular invasion and migration. Focal matrix degradation occurred both centrally and at focal adhesions, the latter being a novel site for matrix degradation in synovial fibroblasts, but degradation was unaltered with FAK inhibitors. Loss of FAK reduced invasion in murine arthritic synovial fibroblasts, but not migration or TNFα-induced arthritis severity and joint erosions. CONCLUSIONS: FAK inhibitors reduce synovial fibroblast invasion and migration, but synovial fibroblast migration and TNFα-induced arthritis do not rely on FAK itself. Thus, inhibition of FAK alone is unlikely to be sufficient to treat inflammatory arthritis, but current drugs that inhibit FAK may inhibit multiple factors, which could increase their efficacy in rheumatoid arthritis.


Asunto(s)
Artritis/enzimología , Fibroblastos/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Membrana Sinovial/metabolismo , Animales , Artritis/genética , Artritis/patología , Artritis Reumatoide/enzimología , Artritis Reumatoide/patología , Western Blotting , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Indoles/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Quinolonas/farmacología , Sulfonamidas/farmacología , Sulfonas/farmacología , Membrana Sinovial/patología , Factores de Tiempo
2.
Biochem Biophys Res Commun ; 450(3): 1212-7, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24667605

RESUMEN

Fertilization triggers rapid changes in intracellular free calcium that serve to activate multiple signaling events critical to the initiation of successful development. Among the pathways downstream of the fertilization-induced calcium transient is the calcium-calmodulin dependent protein tyrosine kinase PTK2b or PYK2 kinase. PTK2b plays an important role in fertilization of the zebrafish oocyte and the objective of the present study was to establish whether PTK2b also functions in mammalian fertilization. PTK2b was activated during the first few hours after fertilization of the mouse oocyte during the period when anaphase resumption was underway and prior to the pronuclear stage. Suppression of PTK2b kinase activity in oocytes blocked sperm incorporation and egg activation although sperm-oocyte binding was not affected. Oocytes that failed to incorporate sperm after inhibitor treatment showed no evidence of a calcium transient and no evidence of anaphase resumption suggesting that egg activation did not occur. The results indicate that PTK2b functions during the sperm-egg fusion process or during the physical incorporation of sperm into the egg cytoplasm and is therefore critical for successful development.


Asunto(s)
Fertilización/fisiología , Quinasa 2 de Adhesión Focal/metabolismo , Oocitos/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Activación Enzimática , Femenino , Fertilización/efectos de los fármacos , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oocitos/efectos de los fármacos , Oocitos/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Interacciones Espermatozoide-Óvulo/efectos de los fármacos , Interacciones Espermatozoide-Óvulo/fisiología , Cigoto/efectos de los fármacos , Cigoto/enzimología , Cigoto/fisiología
3.
Biochem Pharmacol ; 86(6): 770-81, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23928188

RESUMEN

The focal adhesion kinase inhibitor, PF-562,271, is currently in clinical development for cancer, however it is not known how PF-562,271 affects T cell function. Here, we demonstrate inhibitory effects of PF-562,271 on the activation of primary human and mouse T cells. PF-562,271 inhibits T cell receptor signaling-induced T cell adhesion to intercellular adhesion molecule-1 and T cell interactions with antigen-presenting cells. An additional focal adhesion kinase inhibitor, PF-573,228, and genetic depletion of focal adhesion kinase also impair T cell conjugation with antigen-presenting cells. PF-562,271 blocks phosphorylation of the signaling molecules zeta chain associate protein of 70 kDa, linker of activated T cells, and extracellular signal-regulated kinase, and impairs T cell proliferation. The effects observed on T cell proliferation cannot solely be attributed to focal adhesion kinase inhibition, as genetic depletion did not alter proliferation. The effect of PF-562,271 on T cell proliferation is not rescued when proximal T cell receptor signaling is bypassed by stimulation with phorbol-12-myristate-13-acetate and ionomycin. Taken together, our findings demonstrate that focal adhesion kinase regulates integrin-mediated T cell adhesion following T cell receptor activation. Moreover, our findings suggest that PF-562,271 may have immunomodulatory effects that could impact its therapeutic applications.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Indoles/farmacología , Activación de Linfocitos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Comunicación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular , Ionomicina/farmacología , Ratones , Ratones Transgénicos , Fosforilación , Cultivo Primario de Células , Quinolonas/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Sulfonas/farmacología , Acetato de Tetradecanoilforbol/farmacología
4.
J Med Genet ; 50(8): 543-51, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23749989

RESUMEN

BACKGROUND AND AIM: We identified a balanced de novo translocation involving chromosomes Xq25 and 8q24 in an eight year-old girl with a non-progressive form of congenital ataxia, cognitive impairment and cerebellar hypoplasia. METHODS AND RESULTS: Breakpoint definition showed that the promoter of the Protein Tyrosine Kinase 2 (PTK2, also known as Focal Adhesion Kinase, FAK) gene on chromosome 8q24.3 is translocated 2 kb upstream of the THO complex subunit 2 (THOC2) gene on chromosome Xq25. PTK2 is a well-known non-receptor tyrosine kinase whereas THOC2 encodes a component of the evolutionarily conserved multiprotein THO complex, involved in mRNA export from nucleus. The translocation generated a sterile fusion transcript under the control of the PTK2 promoter, affecting expression of both PTK2 and THOC2 genes. PTK2 is involved in cell adhesion and, in neurons, plays a role in axonal guidance, and neurite growth and attraction. However, PTK2 haploinsufficiency alone is unlikely to be associated with human disease. Therefore, we studied the role of THOC2 in the CNS using three models: 1) THOC2 ortholog knockout in C.elegans which produced functional defects in specific sensory neurons; 2) Thoc2 knockdown in primary rat hippocampal neurons which increased neurite extension; 3) Thoc2 knockdown in neuronal stem cells (LC1) which increased their in vitro growth rate without modifying apoptosis levels. CONCLUSION: We suggest that THOC2 can play specific roles in neuronal cells and, possibly in combination with PTK2 reduction, may affect normal neural network formation, leading to cognitive impairment and cerebellar congenital hypoplasia.


Asunto(s)
Cerebelo/anomalías , Cromosomas Humanos Par 8/genética , Quinasa 1 de Adhesión Focal/genética , Malformaciones del Sistema Nervioso/genética , Trastornos Psicomotores/genética , Proteínas de Unión al ARN/genética , Translocación Genética , Animales , Caenorhabditis elegans/genética , Línea Celular Transformada , Niño , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Femenino , Fusión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Malformaciones del Sistema Nervioso/complicaciones , Trastornos Psicomotores/complicaciones , Ratas
5.
J Immunol ; 190(3): 1094-102, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23264658

RESUMEN

Progenitor B cells reside in complex bone marrow (BM) microenvironments where they receive signals for growth and maturation. We reported previously that the CXCL12-focal adhesion kinase (FAK)-VLA4 pathway plays an important role in progenitor B cell adhesion and migration. In this study, we have conditionally targeted in B cells FAK, and found that the numbers of progenitor pro-B, pre-B, and immature B cells are reduced by 30-40% in B cell-specific FAK knockout mice. When cultured in methylcellulose with IL-7 ± CXCL12, Fak-deleted pro-B cells yield significantly fewer cells and colonies. Using in situ quantitative imaging cytometry, we establish that in longitudinal femoral BM sections, pro-B cells are preferentially localized in close proximity to the endosteum of the metaphyses and the diaphysis. Fak deletion disrupts the nonrandom distribution of pro-B cells and induces the mobilization of pro-B cells to the periphery in vivo. These effects of Fak deletion on pro-B cell mobilization and localization in BM are amplified under inflammatory stress, that is, after immunization with nitrophenol-conjugated chicken γ-globulin in alum. Collectively, these studies suggest the importance of FAK in regulating pro-B cell homeostasis and maintenance of their spatial distribution in BM niches.


Asunto(s)
Linfocitos B/citología , Médula Ósea/ultraestructura , Quinasa 1 de Adhesión Focal/fisiología , Células Madre Hematopoyéticas/enzimología , Linfopoyesis/fisiología , Animales , Apoptosis , Linfocitos B/trasplante , Médula Ósea/inmunología , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Microambiente Celular , Quimiocina CXCL12/fisiología , Quimiotaxis de Leucocito/fisiología , Ensayo de Unidades Formadoras de Colonias , Femenino , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Homeostasis , Integrina alfa4beta1/fisiología , Interleucina-7/farmacología , Linfopenia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Invest Ophthalmol Vis Sci ; 53(6): 3067-81, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22491404

RESUMEN

PURPOSE: The lens is a powerful model system to study integrin-mediated cell-matrix interaction in an in vivo context, as it is surrounded by a true basement membrane, the lens capsule. To characterize better the function of integrin-linked kinase (ILK), we examined the phenotypic consequences of its deletion in the developing mouse lens. METHODS: ILK was deleted from the embryonic lens either at the time of placode invagination using the Le-Cre line or after initial lens formation using the Nestin-Cre line. RESULTS: Early deletion of ILK leads to defects in extracellular matrix deposition that result in lens capsule rupture at the lens vesicle stage (E13.5). If ILK was deleted at a later time-point after initial establishment of the lens capsule, rupture was prevented. Instead, ILK deletion resulted in secondary fiber migration defects and, most notably, in cell death of the anterior epithelium (E18.5-P0). Remarkably, dying cells did not stain positively for terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) or activated-caspase 3, suggesting that they were dying from a non-apoptotic mechanism. Moreover, cross to a Bax(fl/fl)/Bak⁻/⁻ mouse line that is resistant to most forms of apoptosis failed to promote cell survival in the ILK-deleted lens epithelium. Electron microscopy revealed the presence of numerous membranous vacuoles containing degrading cellular material. CONCLUSIONS. Our study reveals a role for ILK in extracellular matrix organization, fiber migration, and cell survival. Furthermore, to our knowledge we show for the first time that ILK disruption results in non-apoptotic cell death in vivo.


Asunto(s)
Células Epiteliales/patología , Eliminación de Gen , Cápsula del Cristalino/embriología , Cápsula del Cristalino/patología , Proteínas Serina-Treonina Quinasas/genética , Animales , Cápsula Anterior del Cristalino/lesiones , Cápsula Anterior del Cristalino/patología , Cadherinas/metabolismo , Muerte Celular/genética , Muerte Celular/fisiología , Movimiento Celular/fisiología , Colágeno Tipo IV/metabolismo , Epitelio/metabolismo , Proteínas del Ojo/metabolismo , Fibronectinas/metabolismo , Proteínas de Homeodominio/metabolismo , Laminina/metabolismo , Cápsula del Cristalino/lesiones , Ratones , Ratones Transgénicos , Microscopía Electrónica , Microscopía Electrónica de Transmisión , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Rotura , Regulación hacia Arriba , Vacuolas/patología
7.
Blood ; 116(20): 4103-15, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20664055

RESUMEN

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that has been extensively studied in fibroblasts; however its function in hematopoiesis remains an enigma. FAK is thought to be expressed in myeloid and erythroid progenitors, and its expression is enhanced in response to cytokines such as granu-locyte macrophage colony-stimulating factor. Furthermore, bone marrow cells cultured in granulocyte macrophage colony-stimulating factor show active migration and chemoattractant-induced polarization, which correlates with FAK induction. While loss of FAK in mice results in embryonic lethality, we have deleted FAK in the adult bone marrow. We show an essential role for FAK in regulating hemolytic, myelotoxic, as well as acute inflammatory stress responses in vivo. In vitro, loss of FAK in erythroid and myeloid progenitor's results in impaired cytokine induced growth and survival, as well as defects in the activation and expression of antiapoptotic proteins caspase 3 and Bcl-x(L). Additionally, reduced migration and adhesion of myeloid cells on extracellular matrix proteins, as well as impaired activation of Rac GTPase is also observed in the absence of FAK. Our studies reveal an essential role for FAK in integrating growth/survival and adhesion based functions in myeloid and erythroid cells predominantly under conditions of stress.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Hematopoyesis , Estrés Fisiológico , Actinas/metabolismo , Enfermedad Aguda , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/farmacología , Modelos Animales de Enfermedad , Eritropoyesis/efectos de los fármacos , Femenino , Fluorouracilo/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Eliminación de Gen , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Células Mieloides/efectos de los fármacos , Células Mieloides/enzimología , Células Mieloides/patología , Mielopoyesis/efectos de los fármacos , Peritonitis/inmunología , Peritonitis/patología , Fenilhidrazinas/farmacología , Estrés Fisiológico/efectos de los fármacos
8.
Development ; 136(21): 3657-67, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19820184

RESUMEN

The vertebrate lens provides an excellent model with which to study the mechanisms required for epithelial invagination. In the mouse, the lens forms from the head surface ectoderm. A domain of ectoderm first thickens to form the lens placode and then invaginates to form the lens pit. The epithelium of the lens placode remains in close apposition to the epithelium of the presumptive retina as these structures undergo a coordinated invagination. Here, we show that F-actin-rich basal filopodia that link adjacent presumptive lens and retinal epithelia function as physical tethers that coordinate invagination. The filopodia, most of which originate in the presumptive lens, form at E9.5 when presumptive lens and retinal epithelia first come into close contact, and have retracted by E11.5 when invagination is complete. At E10.5--the lens pit stage--there is approximately one filopodium per epithelial cell. Formation of filopodia is dependent on the Rho family GTPase Cdc42 and the Cdc42 effector IRSp53 (Baiap2). Loss of filopodia results in reduced lens pit invagination. Pharmacological manipulation of the actin-myosin contraction pathway showed that the filopodia can respond rapidly in length to change inter-epithelial distance. These data suggest that the lens-retina inter-epithelial filopodia are a fine-tuning mechanism to assist in lens pit invagination by transmitting the forces between presumptive lens and retina. Although invagination of the archenteron in sea urchins and dorsal closure in Drosophila are known to be partly dependent on filopodia, this mechanism of morphogenesis has not previously been identified in vertebrates.


Asunto(s)
Cristalino/embriología , Seudópodos/metabolismo , Retina/embriología , Actinas/metabolismo , Animales , Quinasa 1 de Adhesión Focal/metabolismo , Cristalino/citología , Ratones , Miosinas/metabolismo , Retina/citología , Organismos Libres de Patógenos Específicos
9.
J Neurosci Res ; 87(15): 3456-64, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19224576

RESUMEN

The formation of the myelin sheath is a crucial step during development because it enables fast and efficient propagation of signals within the limited space of the mammalian central nervous system (CNS). During the process of myelination, oligodendrocytes actively interact with the extracellular matrix (ECM). These interactions are considered crucial for proper and timely completion of the myelin sheath. However, the exact regulatory circuits involved in the signaling events that occur between the ECM and oligodendrocytes are currently not fully understood. Therefore, in the present study we investigated the role of a known integrator of cell-ECM signaling, namely, focal adhesion kinase (FAK), in CNS myelination via the use of conditional (oligodendrocyte-specific) and inducible FAK-knockout mice (Fak(flox/flox): PLP/CreER(T) mice). When inducing FAK knockout just prior to and during active myelination of the optic nerve, we observed a significant reduction in the number of myelinated fibers on postnatal day 14. In addition, our data revealed a decreased number of primary processes extending from oligodendrocyte cell bodies at this postnatal age and on induction of FAK knockout. In contrast, myelination appeared normal on postnatal day 28. Thus, our data suggest that FAK controls the efficiency and timing of CNS myelination during its initial stages, at least in part, by regulating oligodendrocyte process outgrowth and/or remodeling.


Asunto(s)
Diferenciación Celular/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Nervio Óptico/enzimología , Nervio Óptico/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Forma de la Célula/genética , Señales (Psicología) , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Vaina de Mielina/ultraestructura , Fibras Nerviosas Mielínicas/ultraestructura , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nervio Óptico/citología , Factores de Tiempo
10.
J Clin Invest ; 119(2): 252-66, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19147981

RESUMEN

Cancer cells require sustained oncogenic signaling in order to maintain their malignant properties. It is, however, unclear whether they possess other dependencies that can be exploited therapeutically. We report here that in a large fraction of human breast cancers, the gene encoding focal adhesion kinase (FAK), a core component of integrin signaling, was amplified and FAK mRNA was overexpressed. A mammary gland-specific deletion of Fak in mice did not seem to affect normal mammary epithelial cells, and these mice were protected from tumors initiated by the polyoma middle T oncoprotein (PyMT), which activates Ras and PI3K. FAK-deficient PyMT-transformed cells displayed both growth arrest and apoptosis, as well as diminished invasive and metastatic capacity. Upon silencing of Fak, mouse mammary tumor cells transformed by activated Ras became senescent and lost their invasive ability. Further, Neu-transformed cells also underwent growth arrest and apoptosis if integrin beta4-dependent signaling was simultaneously inactivated. Human breast cancer cells carrying oncogenic mutations that activate Ras or PI3K signaling displayed similar responses upon silencing of FAK. Mechanistic studies indicated that FAK sustains tumorigenesis by promoting Src-mediated phosphorylation of p130Cas. These results suggest that FAK supports Ras- and PI3K-dependent mammary tumor initiation, maintenance, and progression to metastasis by orchestrating multiple core cellular functions, including proliferation, survival, and avoidance of senescence.


Asunto(s)
Neoplasias de la Mama/etiología , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Genes ras , Neoplasias Mamarias Experimentales/etiología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/fisiología , Animales , Antígenos Transformadores de Poliomavirus/toxicidad , Neoplasias de la Mama/enzimología , Senescencia Celular , Proteína Sustrato Asociada a CrK/fisiología , Humanos , Neoplasias Pulmonares/secundario , Ratones , Invasividad Neoplásica
11.
Am J Physiol Heart Circ Physiol ; 296(3): H627-38, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19136609

RESUMEN

Migration and proliferation of cardiac fibroblasts (CFs) play an important role in the myocardial remodeling process. While many factors have been identified that regulate CF growth and migration, less is known about the signaling mechanisms involved in these processes. Here, we utilized Cre-LoxP technology to obtain focal adhesion kinase (FAK)-deficient adult mouse CFs and studied how FAK functioned in modulating cell adhesion, proliferation, and migration of these cells. Treatment of FAK(flox/flox) CFs with Ad/Cre virus caused over 70% reduction of FAK protein levels within a cell population. FAK-deficient CFs showed no changes in focal adhesions, cell morphology, or protein expression levels of vinculin, talin, or paxillin; proline-rich tyrosine kinase 2 (Pyk2) expression and activity were increased. Knockdown of FAK protein in CFs increased PDGF-BB-induced proliferation, while it reduced PDGF-BB-induced migration. Adhesion to fibronectin was not altered. To distinguish between the function of FAK and Pyk2, FAK function was inhibited via adenoviral-mediated overexpression of the natural FAK inhibitor FAK-related nonkinase (FRNK). Ad/FRNK had no effect on Pyk2 expression, inhibited the PDGF-BB-induced migration, but did not change the PDGF-BB-induced proliferation. FAK deficiency had only modest effects on increasing PDGF-BB activation of p38 and JNK MAPKs, with no alteration in the ERK response vs. control cells. These results demonstrate that FAK is required for the PDGF-BB-induced migratory response of adult mouse CFs and suggest that FAK could play an essential role in the wound-healing response that occurs in numerous cardiac pathologies.


Asunto(s)
Movimiento Celular , Proliferación Celular , Fibroblastos/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Miocardio/enzimología , Animales , Becaplermina , Adhesión Celular , Forma de la Célula , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/citología , Paxillin/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Talina/metabolismo , Factores de Tiempo , Vinculina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Am J Pathol ; 173(5): 1551-65, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18845837

RESUMEN

Focal adhesion kinase (FAK) is a central regulator of the focal adhesion, influencing cell proliferation, survival, and migration. Despite evidence demonstrating FAK overexpression in human cancer, its role in tumor initiation and progression is not well understood. Using Cre/LoxP technology to specifically knockout FAK in the mammary epithelium, we showed that FAK is not required for tumor initiation but is required for tumor progression. The mechanistic underpinnings of these results suggested that FAK regulates clinically relevant gene signatures and multiple signaling complexes associated with tumor progression and metastasis, such as Src, ERK, and p130Cas. Furthermore, a systems-level analysis identified FAK as a major regulator of the tumor transcriptome, influencing genes associated with adhesion and growth factor signaling pathways, and their cross talk. Additionally, FAK was shown to down-regulate the expression of clinically relevant proliferation- and metastasis-associated gene signatures, as well as an enriched group of genes associated with the G(2) and G(2)/M phases of the cell cycle. Computational analysis of transcription factor-binding sites within ontology-enriched or clustered gene sets suggested that the differentially expressed proliferation- and metastasis-associated genes in FAK-null cells were regulated through a common set of transcription factors, including p53. Therefore, FAK acts as a primary node in the activated signaling network in transformed motile cells and is a prime candidate for novel therapeutic interventions to treat aggressive human breast cancers.


Asunto(s)
Neoplasias de la Mama/patología , Epitelio/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Neoplasias Pulmonares/secundario , Glándulas Mamarias Animales/enzimología , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Animales , Neoplasias de la Mama/enzimología , Movimiento Celular/genética , Proliferación Celular , Proteína Sustrato Asociada a CrK/metabolismo , Modelos Animales de Enfermedad , Epitelio/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Adhesiones Focales/genética , Fase G2/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Noqueados , Mitosis/genética , Especificidad de Órganos , Fosforilación , Receptores de Factores de Crecimiento/genética , Familia-src Quinasas/metabolismo
13.
J Cell Biol ; 179(6): 1275-87, 2007 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-18070912

RESUMEN

Macrophages are a key component of the innate immune system. In this study, we investigate how focal adhesion kinase (FAK) and the related kinase Pyk2 integrate adhesion signaling and growth factor receptor signaling to regulate diverse macrophage functions. Primary bone marrow macrophages isolated from mice in which FAK is conditionally deleted from cells of the myeloid lineage exhibited elevated protrusive activity, altered adhesion dynamics, impaired chemotaxis, elevated basal Rac1 activity, and a marked inability to form stable lamellipodia necessary for directional locomotion. The contribution of FAK to macrophage function in vitro was substantiated in vivo by the finding that recruitment of monocytes to sites of inflammation was impaired in the absence of FAK. Decreased Pyk2 expression in primary macrophages also resulted in a diminution of invasive capacity. However, the combined loss of FAK and Pyk2 had no greater effect than the loss of either molecule alone, indicating that both kinases function within the same pathway to promote invasion.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quinasa 1 de Adhesión Focal/fisiología , Macrófagos/fisiología , Seudópodos/fisiología , Animales , Adhesión Celular/genética , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/fisiología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neuropéptidos/metabolismo , Seudópodos/genética , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
14.
Mol Cell Biol ; 27(15): 5352-64, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17526730

RESUMEN

To examine a role for focal adhesion kinase (FAK) in cardiac morphogenesis, we generated a line of mice with a conditional deletion of FAK in nkx2-5-expressing cells (herein termed FAKnk mice). FAKnk mice died shortly after birth, likely resulting from a profound subaortic ventricular septal defect and associated malalignment of the outflow tract. Additional less penetrant phenotypes included persistent truncus arteriosus and thickened valve leaflets. Thus, conditional inactivation of FAK in nkx2-5-expressing cells leads to the most common congenital heart defect that is also a subset of abnormalities associated with tetralogy of Fallot and the DiGeorge syndrome. No significant differences in proliferation or apoptosis between control and FAKnk hearts were observed. However, decreased myocardialization was observed for the conal ridges of the proximal outflow tract in FAKnk hearts. Interestingly, chemotaxis was significantly attenuated in isolated FAK-null cardiomyocytes in comparison to genetic controls, and these effects were concomitant with reduced tyrosine phosphorylation of Crk-associated substrate (CAS). Thus, it is possible that ventricular septation and appropriate outflow tract alignment is dependent, at least in part, upon FAK-dependent CAS activation and subsequent induction of polarized myocyte movement into the conal ridges. Future studies will be necessary to determine the precise contributions of the additional nkx2-5-derived lineages to the phenotypes observed.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Eliminación de Gen , Cardiopatías Congénitas/enzimología , Ventrículos Cardíacos/anomalías , Ventrículos Cardíacos/anatomía & histología , Animales , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Proteína Sustrato Asociada a CrK/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/enzimología , Femenino , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/enzimología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Morfogénesis , Miocitos Cardíacos/patología , Miofibrillas/patología , Fenotipo , Fosforilación , Factores de Transcripción/metabolismo
15.
Bone ; 41(1): 39-51, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17459803

RESUMEN

Integrins link the inside of a cell with its outside environment and in doing so regulate a wide variety of cell behaviors. Integrins are well known for their roles in angiogenesis and cell migration but their functions in bone formation are less clear. The majority of integrin signaling proceeds through focal adhesion kinase (FAK), an essential component of the focal adhesion complex. We generated transgenic mice in which FAK was deleted in osteoblasts and uncovered a previously unknown role in osteoblast differentiation associated with bone healing. FAK mutant cells migrated to the site of skeletal injury and angiogenesis was unaffected yet the transgenic mice still exhibited numerous defects in reparative bone formation. Osteoblast differentiation itself was unperturbed by the loss of FAK, whereas the attachment of osteoclasts to the bone matrix was disrupted in vivo. We postulate that defective bi-directional integrin signaling affects the organization of the collagen matrix. Finally, we present a compensatory candidate molecule, Pyk2, which localized to the focal adhesions in osteoblasts that were lacking FAK.


Asunto(s)
Regeneración Ósea/fisiología , Remodelación Ósea/fisiología , Quinasa 1 de Adhesión Focal/fisiología , Osteoblastos/citología , Animales , Secuencia de Bases , Matriz Ósea/citología , Regeneración Ósea/genética , Remodelación Ósea/genética , Adhesión Celular , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Cartilla de ADN/genética , Femenino , Desarrollo Fetal , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/fisiología , Heterocigoto , Técnicas In Vitro , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoclastos/citología , Embarazo , Transducción de Señal
16.
J Cell Biol ; 176(5): 667-80, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17325207

RESUMEN

In response to alphabeta1 integrin signaling, transducers such as focal adhesion kinase (FAK) become activated, relaying to specific machineries and triggering distinct cellular responses. By conditionally ablating Fak in skin epidermis and culturing Fak-null keratinocytes, we show that FAK is dispensable for epidermal adhesion and basement membrane assembly, both of which require alphabeta1 integrins. FAK is also dispensible for proliferation/survival in enriched medium. In contrast, FAK functions downstream of alphabeta1 integrin in regulating cytoskeletal dynamics and orchestrating polarized keratinocyte migration out of epidermal explants. Fak-null keratinocytes display an aberrant actin cytoskeleton, which is tightly associated with robust, peripheral focal adhesions and microtubules. We find that without FAK, Src, p190RhoGAP, and PKL-PIX-PAK, localization and/or activation at focal adhesions are impaired, leading to elevated Rho activity, phosphorylation of myosin light chain kinase, and enhanced tensile stress fibers. We show that, together, these FAK-dependent activities are critical to control the turnover of focal adhesions, which is perturbed in the absence of FAK.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Adhesiones Focales/enzimología , Citoesqueleto de Actina/ultraestructura , Animales , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula , Movimiento Celular/fisiología , Forma de la Célula , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Quinasa 2 de Adhesión Focal/análisis , Quinasa 2 de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Integrina beta1/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/ultraestructura , Ratones , Microtúbulos/metabolismo , Fosforilación , Proteínas Represoras/metabolismo , Transducción de Señal
17.
Circ Res ; 99(6): 636-45, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16902179

RESUMEN

Focal adhesion kinase (FAK) is a ubiquitously expressed cytoplasmic tyrosine kinase strongly activated by integrins and neurohumoral factors. Previous studies have shown that cardiac FAK activity is enhanced by hypertrophic stimuli before the onset of overt hypertrophy. Herein, we report that conditional deletion of FAK from the myocardium of adult mice did not affect basal cardiac performance, myocyte viability, or myofibrillar architecture. However, deletion of FAK abolished the increase in left ventricular posterior wall thickness, myocyte cross-sectional area, and hypertrophy-associated atrial natriuretic factor induction following pressure overload. Myocyte-restricted deletion of FAK attenuated the initial wave of extracellular signal-regulated kinase activation and cFos expression induced by adrenergic agonists and biomechanical stress. In addition, we found that persistent challenge of mice with myocyte-restricted FAK inactivation leads to enhanced cardiac fibrosis and cardiac dysfunction in comparison to challenged genetic controls. These studies show that loss of FAK impairs normal compensatory hypertrophic remodeling without a concomitant increase in apoptosis in response to cardiac pressure overload and highlight the possibility that FAK activation may be a common requirement for the initiation of this compensatory response.


Asunto(s)
Cardiomegalia/prevención & control , Quinasa 1 de Adhesión Focal/fisiología , Miocitos Cardíacos/citología , Animales , Apoptosis , Cardiomegalia/etiología , Fibrosis Endomiocárdica/etiología , Quinasa 1 de Adhesión Focal/deficiencia , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-fos/genética , Estrés Mecánico , Disfunción Ventricular Izquierda/terapia
18.
J Cell Biol ; 172(1): 151-62, 2006 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-16391003

RESUMEN

Morphogenesis of a vascular network requires dynamic vessel growth and regression. To investigate the cellular mechanism underlying this process, we deleted focal adhesion kinase (FAK), a key signaling mediator, in endothelial cells (ECs) using Tie2-Cre mice. Targeted FAK depletion occurred efficiently early in development, where mutants exhibited a distinctive and irregular vasculature, resulting in hemorrhage and lethality between embryonic day (e) 10.5 and 11.5. Capillaries and intercapillary spaces in yolk sacs were dilated before any other detectable abnormalities at e9.5, and explants demonstrate that the defects resulted from the loss of FAK and not from organ failure. Time-lapse microscopy monitoring EC behavior during vascular formation in explants revealed no apparent decrease in proliferation or migration but revealed increases in cell retraction and death leading to reduced vessel growth and increased vessel regression. Consistent with this phenotype, ECs derived from mutant embryos exhibited aberrant lamellipodial extensions, altered actin cytoskeleton, and nonpolarized cell movement. This study reveals that FAK is crucial for vascular morphogenesis and the regulation of EC survival and morphology.


Asunto(s)
Vasos Sanguíneos/anomalías , Anomalías Cardiovasculares/enzimología , Anomalías Cardiovasculares/genética , Endotelio Vascular/enzimología , Quinasa 1 de Adhesión Focal/deficiencia , Seudópodos/genética , Animales , Vasos Sanguíneos/patología , Capilares/anomalías , Capilares/patología , Anomalías Cardiovasculares/patología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/patología , Endotelio Vascular/patología , Quinasa 1 de Adhesión Focal/genética , Integrasas/genética , Ratones , Ratones Transgénicos , Mutación , Neovascularización Fisiológica/genética , Seudópodos/patología , Receptor TIE-2/genética
19.
J Cell Sci ; 118(Pt 12): 2613-23, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15914540

RESUMEN

The process of cell migration is initiated by protrusion at the leading edge of the cell, the formation of peripheral adhesions, the exertion of force on these adhesions, and finally the release of the adhesions at the rear of the cell. Focal adhesion kinase (FAK) is intimately involved in the regulation of this process, although the precise mechanism(s) whereby FAK regulates cell migration is unclear. We have used two approaches to reduce FAK expression in fibroblasts. Treatment of cells with FAK-specific siRNAs substantially reduced FAK expression and inhibited the spreading of fibroblasts in serum-free conditions, but did not affect the rate of spreading in the presence of serum. In contrast with the wild-type cells, the FAK siRNA-treated cells exhibited multiple extensions during cell spreading. The extensions appeared to be inappropriately formed lamellipodia as evidenced by the localization of cortactin to lamellipodial structures and the inhibition of such structures by expression of dominant-negative Rac. The wild-type phenotype was restored by reexpressing wild-type FAK in the knockdown cells, but not by expression of FAK containing a point mutation at the autophosphorylation site (FAK Y397F). In wound-healing assays, FAK knockdown cells failed to form broad lamellipodia, instead forming multiple leading edges. Similar results were obtained using primary mouse embryo fibroblasts from FAK-flox mice in which Cre-mediated excision was used to ablate the expression of FAK. These data are consistent with a role for FAK in regulating the formation of a leading edge during cell migration by coordinating integrin signaling to direct the correct spatial activation of membrane protrusion.


Asunto(s)
Movimiento Celular , Adhesiones Focales , Proteínas Tirosina Quinasas/metabolismo , Animales , Línea Celular , Polaridad Celular/genética , Forma de la Célula , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Eliminación de Gen , Regulación de la Expresión Génica , Aparato de Golgi/metabolismo , Ratones , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Seudópodos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Factores de Tiempo
20.
Nat Neurosci ; 7(10): 1059-69, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15378065

RESUMEN

The formation of neuronal networks in the central nervous system (CNS) requires precise control of axonal branch development and stabilization. Here we show that cell-specific ablation of the murine gene Ptk2 (more commonly known as fak), encoding focal adhesion kinase (FAK), increases the number of axonal terminals and synapses formed by neurons in vivo. Consistent with this, fak mutant neurons also form greater numbers of axonal branches in culture because they have increased branch formation and reduced branch retraction. Expression of wild-type FAK, but not that of several FAK variants that prevent interactions with regulators of Rho family GTPases including the p190 Rho guanine nuclear exchange factor (p190RhoGEF), rescues the axonal arborization phenotype observed in fak mutant neurons. In addition, expression of a mutant p190RhoGEF that cannot associate with FAK results in a phenotype very similar to that of neurons lacking FAK. Thus, FAK functions as a negative regulator of axonal branching and synapse formation, and it seems to exert its actions, in part, through Rho family GTPases.


Asunto(s)
Axones/metabolismo , Encéfalo/anomalías , Diferenciación Celular/genética , Proteínas Tirosina Quinasas/metabolismo , Sinapsis/metabolismo , Animales , Axones/ultraestructura , Encéfalo/metabolismo , Encéfalo/ultraestructura , Células Cultivadas , Corteza Cerebelosa/anomalías , Corteza Cerebelosa/metabolismo , Corteza Cerebelosa/ultraestructura , Proteínas de Unión al ADN , Regulación hacia Abajo/genética , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Proteínas Activadoras de GTPasa , Regulación del Desarrollo de la Expresión Génica/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipocampo/anomalías , Hipocampo/metabolismo , Hipocampo/ultraestructura , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinasas/genética , Ratas , Proteínas Represoras , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...