Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunol ; 190(3): 1094-102, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264658

RESUMO

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.


Assuntos
Linfócitos B/citologia , Medula Óssea/ultraestrutura , Quinase 1 de Adesão Focal/fisiologia , Células-Tronco Hematopoéticas/enzimologia , Linfopoese/fisiologia , Animais , Apoptose , Linfócitos B/transplante , Medula Óssea/imunologia , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Microambiente Celular , Quimiocina CXCL12/fisiologia , Quimiotaxia de Leucócito/fisiologia , Ensaio de Unidades Formadoras de Colônias , Feminino , Quinase 1 de Adesão Focal/deficiência , Quinase 1 de Adesão Focal/genética , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Homeostase , Integrina alfa4beta1/fisiologia , Interleucina-7/farmacologia , Linfopenia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Biochem Biophys Res Commun ; 450(3): 1212-7, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24667605

RESUMO

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.


Assuntos
Fertilização/fisiologia , Quinase 2 de Adesão Focal/metabolismo , Oócitos/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Ativação Enzimática , Feminino , Fertilização/efeitos dos fármacos , Quinase 2 de Adesão Focal/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oócitos/efeitos dos fármacos , Oócitos/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Interações Espermatozoide-Óvulo/fisiologia , Zigoto/efeitos dos fármacos , Zigoto/enzimologia , Zigoto/fisiologia
3.
J Med Genet ; 50(8): 543-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23749989

RESUMO

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.


Assuntos
Cerebelo/anormalidades , Cromossomos Humanos Par 8/genética , Quinase 1 de Adesão Focal/genética , Malformações do Sistema Nervoso/genética , Transtornos Psicomotores/genética , Proteínas de Ligação a RNA/genética , Translocação Genética , Animais , Caenorhabditis elegans/genética , Linhagem Celular Transformada , Criança , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Feminino , Fusão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Malformações do Sistema Nervoso/complicações , Transtornos Psicomotores/complicações , Ratos
4.
J Clin Invest ; 119(2): 252-66, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19147981

RESUMO

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.


Assuntos
Neoplasias da Mama/etiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Genes ras , Neoplasias Mamárias Experimentais/etiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Animais , Antígenos Transformantes de Poliomavirus/toxicidade , Neoplasias da Mama/enzimologia , Senescência Celular , Proteína Substrato Associada a Crk/fisiologia , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Invasividade Neoplásica
5.
Development ; 136(21): 3657-67, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19820184

RESUMO

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.


Assuntos
Cristalino/embriologia , Pseudópodes/metabolismo , Retina/embriologia , Actinas/metabolismo , Animais , Quinase 1 de Adesão Focal/metabolismo , Cristalino/citologia , Camundongos , Miosinas/metabolismo , Retina/citologia , Organismos Livres de Patógenos Específicos
6.
Blood ; 116(20): 4103-15, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-20664055

RESUMO

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.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Hematopoese , Estresse Fisiológico , Actinas/metabolismo , Doença Aguda , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/enzimologia , Células da Medula Óssea/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Modelos Animais de Doenças , Eritropoese/efeitos dos fármacos , Feminino , Fluoruracila/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Deleção de Genes , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/enzimologia , Células Mieloides/patologia , Mielopoese/efeitos dos fármacos , Peritonite/imunologia , Peritonite/patologia , Fenil-Hidrazinas/farmacologia , Estresse Fisiológico/efeitos dos fármacos
7.
J Cell Biol ; 176(5): 667-80, 2007 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-17325207

RESUMO

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.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Adesões Focais/enzimologia , Citoesqueleto de Actina/ultraestrutura , Animais , Adesão Celular/fisiologia , Técnicas de Cultura de Células , Movimento Celular/fisiologia , Forma Celular , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Quinase 2 de Adesão Focal/análise , Quinase 2 de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Integrina beta1/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/ultraestrutura , Camundongos , Microtúbulos/metabolismo , Fosforilação , Proteínas Repressoras/metabolismo , Transdução de Sinais
8.
J Cell Biol ; 172(1): 151-62, 2006 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-16391003

RESUMO

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.


Assuntos
Vasos Sanguíneos/anormalidades , Anormalidades Cardiovasculares/enzimologia , Anormalidades Cardiovasculares/genética , Endotélio Vascular/enzimologia , Quinase 1 de Adesão Focal/deficiência , Pseudópodes/genética , Animais , Vasos Sanguíneos/patologia , Capilares/anormalidades , Capilares/patologia , Anormalidades Cardiovasculares/patologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/patologia , Endotélio Vascular/patologia , Quinase 1 de Adesão Focal/genética , Integrases/genética , Camundongos , Camundongos Transgênicos , Mutação , Neovascularização Fisiológica/genética , Pseudópodes/patologia , Receptor TIE-2/genética
9.
Mol Cell Biol ; 27(15): 5352-64, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17526730

RESUMO

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.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/deficiência , Deleção de Genes , Cardiopatias Congênitas/enzimologia , Ventrículos do Coração/anormalidades , Ventrículos do Coração/anatomia & histologia , Animais , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Proteína Substrato Associada a Crk/metabolismo , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/enzimologia , Feminino , Ventrículos do Coração/embriologia , Ventrículos do Coração/enzimologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Morfogênese , Miócitos Cardíacos/patologia , Miofibrilas/patologia , Fenótipo , Fosforilação , Fatores de Transcrição/metabolismo
10.
J Neurosci Res ; 87(15): 3456-64, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19224576

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Bainha de Mielina/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Nervo Óptico/enzimologia , Nervo Óptico/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Forma Celular/genética , Sinais (Psicologia) , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/ultraestrutura , Fibras Nervosas Mielinizadas/ultraestrutura , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Nervo Óptico/citologia , Fatores de Tempo
11.
Am J Pathol ; 173(5): 1551-65, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18845837

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Epitélio/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Neoplasias Pulmonares/secundário , Glândulas Mamárias Animais/enzimologia , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Animais , Neoplasias da Mama/enzimologia , Movimento Celular/genética , Proliferação de Células , Proteína Substrato Associada a Crk/metabolismo , Modelos Animais de Doenças , Epitélio/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Adesões Focais/genética , Fase G2/genética , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Knockout , Mitose/genética , Especificidade de Órgãos , Fosforilação , Receptores de Fatores de Crescimento/genética , Quinases da Família src/metabolismo
12.
Neuron ; 40(3): 501-14, 2003 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-14642275

RESUMO

Targeted deletion of focal adhesion kinase (fak) in the developing dorsal forebrain resulted in local disruptions of the cortical basement membrane located between the neuroepithelium and pia-meninges. At disruption sites, clusters of neurons invaded the marginal zone. Retraction of radial glial endfeet, midline fusion of brain hemispheres, and gliosis also occurred, similar to type II cobblestone lissencephaly as seen in congenital muscular dystrophy. Interestingly, targeted deletion of fak in neurons alone did not result in cortical ectopias, indicating that fak deletion from glia is required for neuronal mislocalization. Unexpectedly, fak deletion specifically from meningeal fibroblasts elicited similar cortical ectopias in vivo and altered laminin organization in vitro. These observations provide compelling evidence that FAK plays a key signaling role in cortical basement membrane assembly and/or remodeling. In addition, FAK is required within neurons during development because neuron-specific fak deletion alters dendritic morphology in the absence of lamination defects.


Assuntos
Membrana Basal/metabolismo , Córtex Cerebral/anormalidades , Distrofias Musculares/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Proteínas de Bactérias/metabolismo , Membrana Basal/patologia , Western Blotting , Calbindina 2 , Calbindinas , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/ultraestrutura , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Dura-Máter , Distroglicanas , Embrião de Mamíferos , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteína Glial Fibrilar Ácida/metabolismo , Heterozigoto , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Infecções , Peptídeos e Proteínas de Sinalização Intracelular , Laminas/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Distrofias Musculares/congênito , Distrofias Musculares/genética , Distrofias Musculares/patologia , Mutação , Proteínas do Tecido Nervoso , Neurônios/patologia , Fatores de Transcrição Otx , Fosfopiruvato Hidratase/metabolismo , Fosfotirosina/metabolismo , Testes de Precipitina , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteína Reelina
13.
Circ Res ; 99(6): 636-45, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16902179

RESUMO

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.


Assuntos
Cardiomegalia/prevenção & controle , Quinase 1 de Adesão Focal/fisiologia , Miócitos Cardíacos/citologia , Animais , Apoptose , Cardiomegalia/etiologia , Fibrose Endomiocárdica/etiologia , Quinase 1 de Adesão Focal/deficiência , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-fos/genética , Estresse Mecânico , Disfunção Ventricular Esquerda/terapia
14.
Nat Neurosci ; 7(10): 1059-69, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15378065

RESUMO

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.


Assuntos
Axônios/metabolismo , Encéfalo/anormalidades , Diferenciação Celular/genética , Proteínas Tirosina Quinases/metabolismo , Sinapses/metabolismo , Animais , Axônios/ultraestrutura , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Células Cultivadas , Córtex Cerebelar/anormalidades , Córtex Cerebelar/metabolismo , Córtex Cerebelar/ultraestrutura , Proteínas de Ligação a DNA , Regulação para Baixo/genética , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteínas Ativadoras de GTPase , Regulação da Expressão Gênica no Desenvolvimento/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipocampo/anormalidades , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinases/genética , Ratos , Proteínas Repressoras , Sinapses/ultraestrutura
15.
Bone ; 41(1): 39-51, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17459803

RESUMO

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.


Assuntos
Regeneração Óssea/fisiologia , Remodelação Óssea/fisiologia , Quinase 1 de Adesão Focal/fisiologia , Osteoblastos/citologia , Animais , Sequência de Bases , Matriz Óssea/citologia , Regeneração Óssea/genética , Remodelação Óssea/genética , Adesão Celular , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Primers do DNA/genética , Feminino , Desenvolvimento Fetal , Quinase 1 de Adesão Focal/deficiência , Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/fisiologia , Heterozigoto , Técnicas In Vitro , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Osteoclastos/citologia , Gravidez , Transdução de Sinais
16.
Arthritis Res Ther ; 16(5): 464, 2014 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-25280866

RESUMO

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.


Assuntos
Artrite/enzimologia , Fibroblastos/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Membrana Sinovial/metabolismo , Animais , Artrite/genética , Artrite/patologia , Artrite Reumatoide/enzimologia , Artrite Reumatoide/patologia , Western Blotting , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Indóis/farmacologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Quinolonas/farmacologia , Sulfonamidas/farmacologia , Sulfonas/farmacologia , Membrana Sinovial/patologia , Fatores de Tempo
17.
Biochem Pharmacol ; 86(6): 770-81, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23928188

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Indóis/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Sulfonamidas/farmacologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular , Ionomicina/farmacologia , Camundongos , Camundongos Transgênicos , Fosforilação , Cultura Primária de Células , Quinolonas/farmacologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
18.
Invest Ophthalmol Vis Sci ; 53(6): 3067-81, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22491404

RESUMO

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.


Assuntos
Células Epiteliais/patologia , Deleção de Genes , Cápsula do Cristalino/embriologia , Cápsula do Cristalino/patologia , Proteínas Serina-Treonina Quinases/genética , Animais , Cápsula Anterior do Cristalino/lesões , Cápsula Anterior do Cristalino/patologia , Caderinas/metabolismo , Morte Celular/genética , Morte Celular/fisiologia , Movimento Celular/fisiologia , Colágeno Tipo IV/metabolismo , Epitélio/metabolismo , Proteínas do Olho/metabolismo , Fibronectinas/metabolismo , Proteínas de Homeodomínio/metabolismo , Laminina/metabolismo , Cápsula do Cristalino/lesões , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Microscopia Eletrônica de Transmissão , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Ruptura , Regulação para Cima , Vacúolos/patologia
19.
Am J Physiol Heart Circ Physiol ; 296(3): H627-38, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19136609

RESUMO

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.


Assuntos
Movimento Celular , Proliferação de Células , Fibroblastos/enzimologia , Quinase 1 de Adesão Focal/metabolismo , Miocárdio/enzimologia , Animais , Becaplermina , Adesão Celular , Forma Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/deficiência , Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/citologia , Paxilina/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-sis , Talina/metabolismo , Fatores de Tempo , Vinculina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
J Cell Biol ; 179(6): 1275-87, 2007 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-18070912

RESUMO

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.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/fisiologia , Macrófagos/fisiologia , Pseudópodes/fisiologia , Animais , Adesão Celular/genética , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/fisiologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeos/metabolismo , Pseudópodes/genética , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA