Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 2359, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883558

RESUMEN

How adhesive forces are transduced and integrated into biochemical signals at focal adhesions (FAs) is poorly understood. Using cells adhering to deformable micropillar arrays, we demonstrate that traction force and FAK localization as well as traction force and Y397-FAK phosphorylation are linearly coupled at individual FAs on stiff, but not soft, substrates. Similarly, FAK phosphorylation increases linearly with external forces applied to FAs using magnetic beads. This mechanosignaling coupling requires actomyosin contractility, talin-FAK binding, and full-length vinculin that binds talin and actin. Using an in vitro 3D biomimetic wound healing model, we show that force-FAK signaling coupling coordinates cell migration and tissue-scale forces to promote microtissue repair. A simple kinetic binding model of talin-FAK interactions under force can recapitulate the experimental observations. This study provides insights on how talin and vinculin convert forces into FAK signaling events regulating cell migration and tissue repair.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Modelos Biológicos , Actomiosina/metabolismo , Animales , Fenómenos Biomecánicos , Biomimética , Movimiento Celular/fisiología , Células Cultivadas , Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Mecanotransducción Celular , Ratones , Ratones Noqueados , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Talina/antagonistas & inhibidores , Talina/genética , Talina/metabolismo , Cicatrización de Heridas/fisiología
2.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182805

RESUMEN

Lipid catabolism and anabolism changes play a role in stemness acquisition by cancer cells, and cancer stem cells (CSCs) are particularly dependent on the activity of the enzymes involved in these processes. Lipidomic changes could play a role in CSCs' ability to cause disease relapse and chemoresistance. The exploration of lipid composition and metabolism changes in CSCs in the context of hepatocellular cancer (HCC) is still incomplete and their lipidomic scenario continues to be elusive. We aimed to evaluate through high-throughput mass spectrometry (MS)-based lipidomics the levels of the members of the six major classes of sphingolipids and phospholipids in two HCC cell lines (HepG2 and Huh-7) silenced for the expression of histone variant macroH2A1 (favoring stemness acquisition), or silenced for the expression of focal adhesion tyrosine kinase (FAK) (hindering aggressiveness and stemness). Transcriptomic changes were evaluated by RNA sequencing as well. We found definite lipidomic and transcriptomic changes in the HCC lines upon knockdown (KD) of macroH2A1 or FAK, in line with the acquisition or loss of stemness features. In particular, macroH2A1 KD increased total sphingomyelin (SM) levels and decreased total lysophosphatidylcholine (LPC) levels, while FAK KD decreased total phosphatidylcholine (PC) levels. In conclusion, in HCC cell lines knocked down for specific signaling/epigenetic processes driving opposite stemness potential, we defined a lipidomic signature that hallmarks hepatic CSCs to be exploited for therapeutic strategies.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Metabolismo de los Lípidos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Histonas/antagonistas & inhibidores , Histonas/deficiencia , Histonas/genética , Humanos , Metabolismo de los Lípidos/genética , Lipidómica , Neoplasias Hepáticas/genética , Lisofosfatidilcolinas/metabolismo , Fosfatidilcolinas/metabolismo , RNA-Seq , Esfingomielinas/metabolismo
3.
J Clin Invest ; 130(10): 5444-5460, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32673290

RESUMEN

Cells sense the extracellular environment and mechanical stimuli and translate these signals into intracellular responses through mechanotransduction, which alters cell maintenance, proliferation, and differentiation. Here we use a mouse model of trauma-induced heterotopic ossification (HO) to examine how cell-extrinsic forces impact mesenchymal progenitor cell (MPC) fate. After injury, single-cell (sc) RNA sequencing of the injury site reveals an early increase in MPC genes associated with pathways of cell adhesion and ECM-receptor interactions, and MPC trajectories to cartilage and bone. Immunostaining uncovers active mechanotransduction after injury with increased focal adhesion kinase signaling and nuclear translocation of transcriptional coactivator TAZ, inhibition of which mitigates HO. Similarly, joint immobilization decreases mechanotransductive signaling, and completely inhibits HO. Joint immobilization decreases collagen alignment and increases adipogenesis. Further, scRNA sequencing of the HO site after injury with or without immobilization identifies gene signatures in mobile MPCs correlating with osteogenesis, and signatures from immobile MPCs with adipogenesis. scATAC-seq in these same MPCs confirm that in mobile MPCs, chromatin regions around osteogenic genes are open, whereas in immobile MPCs, regions around adipogenic genes are open. Together these data suggest that joint immobilization after injury results in decreased ECM alignment, altered MPC mechanotransduction, and changes in genomic architecture favoring adipogenesis over osteogenesis, resulting in decreased formation of HO.


Asunto(s)
Extremidades/lesiones , Células Madre Mesenquimatosas/patología , Células Madre Mesenquimatosas/fisiología , Osificación Heterotópica/etiología , Restricción Física , Aciltransferasas , Adipogénesis/genética , Animales , Diferenciación Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Masculino , Mecanotransducción Celular/genética , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osificación Heterotópica/patología , Osificación Heterotópica/fisiopatología , Osteogénesis/genética , Restricción Física/efectos adversos , Restricción Física/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Sci Rep ; 6: 27029, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27244251

RESUMEN

Arteriogenesis, or the lumenal expansion of pre-existing arterioles in the presence of an upstream occlusion, is a fundamental vascular growth response. Though alterations in shear stress stimulate arteriogenesis, the migration of monocytes into the perivascular space surrounding collateral arteries and their differentiation into macrophages is critical for this vascular growth response to occur. Focal adhesion kinase's (FAK) role in regulating cell migration has recently been expanded to primary macrophages. We therefore investigated the effect of the myeloid-specific conditional deletion of FAK on vascular remodeling in the mouse femoral arterial ligation (FAL) model. Using laser Doppler perfusion imaging, whole mount imaging of vascular casted gracilis muscles, and immunostaining for CD31 in gastrocnemius muscles cross-sections, we found that there were no statistical differences in perfusion recovery, arteriogenesis, or angiogenesis 28 days after FAL. We therefore sought to determine FAK expression in different myeloid cell populations. We found that FAK is expressed at equally low levels in Ly6C(hi) and Ly6C(lo) blood monocytes, however expression is increased over 2-fold in bone marrow derived macrophages. Ultimately, these results suggest that FAK is not required for monocyte migration to the perivascular space and that vascular remodeling following arterial occlusion occurs independently of myeloid specific FAK.


Asunto(s)
Arteriopatías Oclusivas/genética , Quinasa 1 de Adhesión Focal/genética , Eliminación de Gen , Músculo Esquelético/metabolismo , Neovascularización Fisiológica/genética , Remodelación Vascular/genética , Animales , Arteriopatías Oclusivas/metabolismo , Arteriopatías Oclusivas/patología , Movimiento Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Arteria Femoral/metabolismo , Arteria Femoral/patología , Arteria Femoral/cirugía , Quinasa 1 de Adhesión Focal/deficiencia , Expresión Génica , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
5.
Oncotarget ; 6(26): 22214-26, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26084289

RESUMEN

The involvement of ErbB family members in breast cancer progression and metastasis has been demonstrated by many studies. However, the downstream effectors that mediate their migratory and invasive responses have not been fully explored. In this study, we show that the non-receptor tyrosine kinase PYK2 is a key effector of EGFR and HER2 signaling in human breast carcinoma. We found that PYK2 is activated by both EGF and heregulin (HRG) in breast cancer cells, and positively regulates EGF/HRG-induced cell spreading, migration and invasion. PYK2 depletion markedly affects ERK1/2 and STAT3 phosphorylation in response to EGF/HRG as well as to IL8 treatment. Importantly, PYK2 depletion also reduced EGF/HRG-induced MMP9 and IL8 transcription, while IL8 inhibition abrogated EGF-induced MMP9 transcription and attenuated cell invasion. IL8, which is transcriptionally regulated by STAT3 and induces PYK2 activation, prolonged EGF-induced PYK2, STAT3 and ERK1/2 phosphorylation suggesting that IL8 acts through an autocrine loop to reinforce EGF-induced signals. Collectively our studies suggest that PYK2 is a common downstream effector of ErbB and IL8 receptors, and that PYK2 integrates their signaling pathways through a positive feedback loop to potentiate breast cancer invasion. Hence, PYK2 could be a potential therapeutic target for a subset of breast cancer patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Receptores de Interleucina-8/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Femenino , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Invasividad Neoplásica , Neurregulina-1/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
6.
Int J Cancer ; 137(7): 1549-59, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25809490

RESUMEN

Human conventional renal cell carcinoma (CCC) remains resistant to current therapies. Focal Adhesion Kinase (FAK) is upregulated in many epithelial tumors and clearly implicated in nearly all facets of cancer. However, only few reports have assessed whether FAK may be associated with renal tumorigenesis. In this study, we investigated the potential role of FAK in the growth of human CCC using a panel of CCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene as well as normal/tumoral renal tissue pairs. FAK was found constitutively expressed in human CCC both in culture cells and freshly harvested tumors obtained from patients. We showed that CCC cell growth was dramatically reduced in FAK-depleted cells or after FAK inhibition with various inhibitors and this effect was obtained through inhibition of cell proliferation and induction of cell apoptosis. Additionally, our results indicated that FAK knockdown decreased CCC cell migration and invasion. More importantly, depletion or pharmacological inhibition of FAK substantially inhibited tumor growth in vivo. Interestingly, investigations of the molecular mechanism revealed loss of FAK phosphorylation during renal tumorigenesis impacting multiple signaling pathways. Taken together, our findings reveal a previously uncharacterized role of FAK in CCC whereby FAK exerts oncogenic properties through a non canonical signaling pathway involving its scaffolding kinase-independent properties. Therefore, targeting the FAK scaffold may represent a promising approach for developing innovative and highly specific therapies in human CCC.


Asunto(s)
Carcinoma de Células Renales/terapia , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/deficiencia , Neoplasias Renales/terapia , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Xenoinjertos , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Ratones , Fosforilación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/biosíntesis , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
7.
Nat Commun ; 5: 5054, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25270220

RESUMEN

Metastasis is the main cause of cancer-related death and thus understanding the molecular and cellular mechanisms underlying this process is critical. Here, our data demonstrate, contrary to established dogma, that loss of haematopoietic-derived focal adhesion kinase (FAK) is sufficient to enhance tumour metastasis. Using both experimental and spontaneous metastasis models, we show that genetic ablation of haematopoietic FAK does not affect primary tumour growth but enhances the incidence of metastasis significantly. At a molecular level, haematopoietic FAK deletion results in an increase in PU-1 levels and decrease in GATA-1 levels causing a shift of hematopoietic homeostasis towards a myeloid commitment. The subsequent increase in circulating granulocyte number, with an increase in serum CXCL12 and granulocyte CXCR4 levels, was required for augmented metastasis in mice lacking haematopoietic FAK. Overall our findings provide a mechanism by which haematopoietic FAK controls cancer metastasis.


Asunto(s)
Quinasa 1 de Adhesión Focal/deficiencia , Sistema Hematopoyético/enzimología , Neoplasias/enzimología , Neoplasias/patología , Animales , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quinasa 1 de Adhesión Focal/genética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/fisiopatología , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
8.
Br J Cancer ; 110(11): 2747-55, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24809783

RESUMEN

BACKGROUND: Elevated expression of focal adhesion kinase (FAK) occurs in numerous human cancers including colon-, cervix- and breast cancer. Although several studies have implicated FAK in mammary tumour formation induced by ectopic oncogene expression, evidence supporting a role for FAK in spontaneous mammary tumour development caused by loss of tumour suppressor genes such as p53 is lacking. Alterations in the tumour suppressor gene p53 have been implicated in over 50% of human breast cancers. Given that elevated FAK expression highly correlates with p53 mutation status in human breast cancer, we set out to investigate the importance of FAK in p53-mediated spontaneous mammary tumour development. METHODS: To directly assess the role of FAK, we generated mice with conditional inactivation of FAK and p53. We generated female p53(lox/lox)/FAK(+/+)/WapCre, p53(lox/lox)/FAK(flox/+)/WapCre and p53(lox/lox)/FAK(flox/-)/WapCre mice, and mice with WapCre-mediated conditional expression of p53(R270H), the mouse equivalent of human p53(R273H) hot spot mutation, together with conditional deletion of FAK, P53(R270H/+)/FAK(lox/+)/WapCre and p53(R270H/+)/FAK(flox/-)/WapCre mice. All mice were subjected to one pregnancy to induce WapCre-mediated deletion of p53 or expression of p53 R270H, and Fak genes flanked by two loxP sites, and subsequently followed the development of mammary tumours. RESULTS: Using this approach, we show that FAK is important for p53-induced mammary tumour development. In addition, mice with the mammary gland-specific conditional expression of p53 point mutation R270H, the mouse equivalent to human R273H, in combination with conditional deletion of Fak showed reduced incidence of p53(R270H)-induced mammary tumours. In both models these effects of FAK were related to reduced proliferation in preneoplastic lesions in the mammary gland ductal structures. CONCLUSIONS: Mammary gland-specific ablation of FAK hampers p53-regulated spontaneous mammary tumour formation. Focal adhesion kinase deletion reduced proliferative capacity of p53 null and p53(R270H) mammary epithelial cells but did not lead to increased apoptosis in vivo. Our data identify FAK as an important regulator in mammary epithelial cell proliferation in p53-mediated and p53(R270H)-induced mammary tumour development.


Asunto(s)
Carcinoma/enzimología , Carcinosarcoma/enzimología , Quinasa 1 de Adhesión Focal/genética , Neoplasias Mamarias Experimentales/enzimología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinoma/genética , Carcinoma/patología , Carcinosarcoma/genética , Carcinosarcoma/patología , Proliferación Celular , Células Epiteliales/enzimología , Femenino , Quinasa 1 de Adhesión Focal/deficiencia , Humanos , Incidencia , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Carga Tumoral , Proteína p53 Supresora de Tumor/genética
9.
J Immunol ; 191(12): 6208-21, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24227778

RESUMEN

Focal adhesion kinase (FAK) is a critical regulator of signal transduction in multiple cell types. Although this protein is activated upon TCR engagement, the cellular function that FAK plays in mature human T cells is unknown. By suppressing the function of FAK, we revealed that FAK inhibits TCR-mediated signaling by recruiting C-terminal Src kinase to the membrane and/or receptor complex following TCR activation. Thus, in the absence of FAK, the inhibitory phosphorylation of Lck and/or Fyn is impaired. Together, these data highlight a novel role for FAK as a negative regulator TCR function in human T cells. These results also suggest that changes in FAK expression could modulate sensitivity to TCR stimulation and contribute to the progression of T cell malignancies and autoimmune diseases.


Asunto(s)
Quinasa 1 de Adhesión Focal/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/fisiología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Adolescente , Adulto , Sustitución de Aminoácidos , Linfocitos T CD4-Positivos/enzimología , Proteína Tirosina Quinasa CSK , Activación Enzimática/fisiología , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Humanos , Células Jurkat , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Masculino , MicroARNs/genética , Persona de Mediana Edad , Complejos Multienzimáticos , Fosforilación , Fosfotirosina/fisiología , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-fyn/fisiología , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Adulto Joven , Familia-src Quinasas/metabolismo
10.
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
11.
PLoS One ; 7(6): e39613, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768099

RESUMEN

Entry of Neisseria meningitidis (the meningococcus) into human brain microvascular endothelial cells (HBMEC) is mediated by fibronectin or vitronectin bound to the surface protein Opc forming a bridge to the respective integrins. This interaction leads to cytoskeletal rearrangement and uptake of meningococci. In this study, we determined that the focal adhesion kinase (FAK), which directly associates with integrins, is involved in integrin-mediated internalization of N. meningitidis in HBMEC. Inhibition of FAK activity by the specific FAK inhibitor PF 573882 reduced Opc-mediated invasion of HBMEC more than 90%. Moreover, overexpression of FAK mutants that were either impaired in the kinase activity or were not capable of autophosphorylation or overexpression of the dominant-negative version of FAK (FRNK) blocked integrin-mediated internalization of N. meningitidis. Importantly, FAK-deficient fibroblasts were significantly less invaded by N. meningitidis. Furthermore, N. meningitidis induced tyrosine phosphorylation of several host proteins including the FAK/Src complex substrate cortactin. Inhibition of cortactin expression by siRNA silencing and mutation of critical amino acid residues within cortactin, that encompass Arp2/3 association and dynamin binding, significantly reduced meningococcal invasion into eukaryotic cells suggesting that both domains are critical for efficient uptake of N. meningitidis into eukaryotic cells. Together, these results indicate that N. meningitidis exploits the integrin signal pathway for its entry and that FAK mediates the transfer of signals from activated integrins to the cytoskeleton. A cooperative interplay between FAK, Src and cortactin then enables endocytosis of N. meningitidis into host cells.


Asunto(s)
Cortactina/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/microbiología , Quinasa 1 de Adhesión Focal/metabolismo , Neisseria meningitidis/fisiología , Familia-src Quinasas/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Encéfalo/patología , Endocitosis , Fibroblastos/enzimología , Fibroblastos/microbiología , Fibroblastos/patología , Quinasa 1 de Adhesión Focal/deficiencia , Interacciones Huésped-Patógeno , Humanos , Integrinas/metabolismo , Ratones , Microvasos/patología , Neisseria meningitidis/citología , Fosforilación , Fosfotirosina/metabolismo
12.
EMBO Rep ; 13(8): 733-40, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22732841

RESUMEN

We have recently described that autophagic targeting of Src maintains cancer cell viability when FAK signalling is defective. Here, we show that the Ret tyrosine kinase is also degraded by autophagy in cancer cells with altered/reduced FAK signalling, preventing its binding to FAK at integrin adhesions. Inhibition of autophagy restores Ret localization to focal adhesions. Importantly, Src kinase activity is required to target Ret to autophagosomes and enhance Ret degradation. Src is thus a general mediator of selective autophagic targeting of adhesion-linked kinases, and Ret a second FAK-binding tyrosine kinase degraded through autophagy in cancer cells under adhesion stress. Src--by controlling not only its own degradation but also that of other FAK-binding partners--allows cancer cell survival, suggesting a new therapeutic strategy.


Asunto(s)
Autofagia , Carcinoma de Células Escamosas/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Adhesión Celular , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/deficiencia , Humanos , Espacio Intracelular/metabolismo , Ratones , Fagosomas/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo
13.
Breast Cancer Res ; 14(1): R36, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22373082

RESUMEN

INTRODUCTION: Activation of focal adhesion kinase (FAK) is hypothesized to play an important role in the pathogenesis of human breast cancer. METHODS: To directly evaluate the role of FAK in mammary tumour progression, we have used a conditional FAK mouse model and mouse mammary tumour virus (MMTV)-driven Cre recombinase strain to inactivate FAK in the mammary epithelium of a transgenic mouse model of ErbB2 breast cancer. RESULTS: Although mammary epithelial disruption of FAK in this model resulted in both a delay in onset and a decrease in the number of neoplastic lesions, mammary tumours occurred in 100% of virgin female mice. All of the tumours and derived metastases that developed were proficient for FAK due to the absence of Cre recombinase expression. The hyperplastic epithelia where Cre-mediated recombination of FAK could be detected exhibited a profound proliferative defect. Consistent with these observations, disruption of FAK in established tumour cells resulted in reduced tumour growth that was associated with impaired proliferation. To avoid the selection for FAK-proficient ErbB2 tumour epithelia through escape of Cre-mediated recombination, we next intercrossed the FAK conditional mice with a separate MMTV-driven ErbB2 strain that co-expressed ErbB2 and Cre recombinase on the same transcriptional unit. CONCLUSIONS: While a delay in tumour induction was noted, FAK-deficient tumours arose in 100% of female animals indicating that FAK is dispensable for ErbB2 tumour initiation. In addition, the FAK-null ErbB2 tumours retained their metastatic potential. We further demonstrated that the FAK-related Pyk2 kinase is still expressed in these tumours and is associated with its downstream regulator p130Cas. These observations indicate that Pyk2 can functionally substitute for FAK in ErbB2 mammary tumour progression.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Quinasa 1 de Adhesión Focal/deficiencia , Neoplasias Pulmonares/enzimología , Neoplasias Mamarias Experimentales/enzimología , Receptor ErbB-2/metabolismo , Animales , Apoptosis , Movimiento Celular , Proteína Sustrato Asociada a CrK/metabolismo , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/fisiología , Quinasa 2 de Adhesión Focal/metabolismo , Eliminación de Gen , Integrasas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Paxillin/metabolismo , Fosforilación , Carga Tumoral
14.
Exp Hematol ; 40(4): 307-17.e3, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22155722

RESUMEN

Hematopoietic stem cells (HSCs) reside in complex bone marrow microenvironments, where niche-induced signals regulate hematopoiesis. Focal adhesion kinase (Fak) is a nonreceptor protein tyrosine kinase that plays an essential role in many cell types, where its activation controls adhesion, motility, and survival. Fak expression is relatively increased in HSCs compared to progenitors and mature blood cells. Therefore, we explored its role in HSC homeostasis. We have used the Mx1-Cre-inducible conditional knockout mouse model to investigate the effects of Fak deletion in bone marrow compartments. The total number as well as the fraction of cycling Lin(-)Sca-1(+)c-kit(+) (LSK) cells is increased in Fak(-/-) mice compared to controls, while hematopoietic progenitors and mature blood cells are unaffected. Bone marrow cells from Fak(-/-) mice exhibit enhanced, long-term (i.e., 20-week duration) engraftment in competitive transplantation assays. Intrinsic Fak function was assessed in serial transplantation assays, which showed that HSCs (Lin(-)Sca-1(+)c-kit(+)CD34(-)Flk-2(-) cells) sorted from Fak(-/-) mice have similar self-renewal and engraftment ability on a per-cell basis as wild-type HSCs. When Fak deletion is induced after engraftment of Fak(fl/fl)Mx1-Cre(+) bone marrow cells into wild-type recipient mice, the number of LSKs is unchanged. In conclusion, Fak inactivation does not intrinsically regulate HSC behavior and is not essential for steady-state hematopoiesis. However, widespread Fak inactivation in the hematopoietic system induces an increased and activated HSC pool size, potentially as a result of altered reciprocal interactions between HSCs and their microenvironment.


Asunto(s)
Quinasa 1 de Adhesión Focal/deficiencia , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/fisiología , Animales , Animales Congénicos , Antígenos de Diferenciación/análisis , Trasplante de Médula Ósea , Comunicación Celular , Ciclo Celular , Linaje de la Célula , Separación Celular , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/fisiología , Genes Reporteros , Hematopoyesis/genética , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/enzimología , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quimera por Radiación , Células del Estroma/fisiología
15.
Mol Cell Biol ; 31(21): 4258-69, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21876001

RESUMEN

Protein tyrosine phosphatase (PTP)-PEST is a critical regulator of cell adhesion and migration. However, the mechanism by which PTP-PEST is regulated in response to oncogenic signaling to dephosphorylate its substrates remains unclear. Here, we demonstrate that activated Ras induces extracellular signal-regulated kinase 1 and 2-dependent phosphorylation of PTP-PEST at S571, which recruits PIN1 to bind to PTP-PEST. Isomerization of the phosphorylated PTP-PEST by PIN1 increases the interaction between PTP-PEST and FAK, which leads to the dephosphorylation of FAK Y397 and the promotion of migration, invasion, and metastasis of v-H-Ras-transformed cells. These findings uncover an important mechanism for the regulation of PTP-PEST in activated Ras-induced tumor progression.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Isomerasa de Peptidilprolil/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Transformada , Movimiento Celular , Progresión de la Enfermedad , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Adhesiones Focales , Genes ras , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Modelos Biológicos , Peptidilprolil Isomerasa de Interacción con NIMA , Invasividad Neoplásica , Neoplasias/etiología , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética
16.
Arterioscler Thromb Vasc Biol ; 31(10): 2193-202, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21757658

RESUMEN

OBJECTIVE: The investment of newly formed endothelial cell tubes with differentiated smooth muscle cells (SMC) is critical for appropriate vessel formation, but the underlying mechanisms remain unknown. We previously showed that depletion of focal adhesion kinase (FAK) in the nkx2.5 expression domain led to aberrant outflow tract (OFT) morphogenesis and strove herein to determine the cell types and mechanisms involved. METHODS AND RESULTS: We crossed fak(loxp) targeted mice with available Cre drivers to deplete FAK in OFT SMC (FAK(wnt) and FAK(nk)) or coronary SMC (FAK(cSMC)). In each case, depletion of FAK led to defective vasculogenesis that was incompatible with postnatal life. Immunohistochemical analysis of the mutant vascular structures revealed that FAK was not required for progenitor cell proliferation, survival, or differentiation into SMC but was necessary for subsequent SMC recruitment to developing vasculature. Using a novel FAK-null SMC culture model, we found that depletion of FAK did not influence SMC growth or survival, but blocked directional SMC motility and invasion toward the potent endothelial-derived chemokine, platelet-derived growth factor PDGFBB. FAK depletion resulted in unstable lamellipodial protrusions due to defective spatial-temporal activation of the small GTPase, Rac-1, and lack of Rac1-dependent recruitment of cortactin (an actin stabilizing protein) to the leading edge. Moreover, FAK null SMC exhibited a significant reduction in stimulated extracellular matrix degradation. CONCLUSIONS: FAK drives PDGFBB-stimulated SMC chemotaxis/invasion and is essential for SMC to appropriately populate the aorticopulmonary septum and the coronary vascular plexus.


Asunto(s)
Quimiotaxis , Quinasa 1 de Adhesión Focal/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neovascularización Fisiológica , Animales , Aorta/embriología , Aorta/enzimología , Apoptosis , Becaplermina , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Quimiotaxis/genética , Vasos Coronarios/embriología , Vasos Coronarios/enzimología , Cortactina/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Regulación del Desarrollo de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/embriología , Neovascularización Fisiológica/genética , Neuropéptidos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Seudópodos/enzimología , Arteria Pulmonar/embriología , Arteria Pulmonar/enzimología , Codorniz/embriología , Interferencia de ARN , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
17.
Artículo en Inglés | MEDLINE | ID: mdl-22919583

RESUMEN

Host cell entry by the food-borne pathogen Campylobacter jejuni has been reported as one of the primary reasons of tissue damage in infected humans, however, molecular invasion mechanisms and cellular factors involved in this process are widely unclear. Here we used knockout cell lines derived from fibronectin(-/-), integrin beta1(-/-), and focal adhesion kinase (FAK)(-/-) deficient mice and corresponding wild-type (WT) controls, to study C. jejuni-induced signaling cascades involved in the bacterial invasion process. Using high resolution scanning electron microscopy, GTPase pull-downs, G-LISA, and gentamicin protection assays we found that each of these host cell factors is indeed required for activation of the small Rho GTPase member Rac1 and maximal host cell invasion of this pathogen. Interestingly, membrane ruffling, tight engulfment of bacteria and invasion were only seen during infection of WT control cells, but not in fibronectin(-/-), integrin beta1(-/-), and FAK(-/-) knockout cell lines. We also demonstrate that C. jejuni activates FAK autophosphorylation activity at Y-397 and phosphorylation of Y-925, which is required for stimulating two downstream guanine exchange factors, DOCK180 and Tiam-1, which are upstream of Rac1. Small interfering (si) RNA studies further show that DOCK180 and Tiam-1 act cooperatively to trigger Rac1 activation and C. jejuni invasion. Moreover, mutagenesis data indicate that the bacterial fibronectin-binding protein CadF and the intact flagellum are involved in Rho GTPase activation and host cell invasion. Collectively, our results suggest that C. jejuni infection of host epithelial target cells hijacks a major fibronectin → integrin beta1 → FAK → DOCK180/Tiam-1 signaling cascade, which has a crucial role for Rac1 GTPase activity and bacterial entry into host target cells.


Asunto(s)
Campylobacter jejuni/patogenicidad , Interacciones Huésped-Patógeno/fisiología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/fisiología , Secuencia de Bases , Infecciones por Campylobacter/etiología , Campylobacter jejuni/genética , Campylobacter jejuni/ultraestructura , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Línea Celular , Activación Enzimática , Células Epiteliales/microbiología , Células Epiteliales/ultraestructura , Fibronectinas/deficiencia , Fibronectinas/genética , Fibronectinas/fisiología , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/fisiología , Genes Bacterianos , Factores de Intercambio de Guanina Nucleótido/fisiología , Interacciones Huésped-Patógeno/genética , Humanos , Integrina beta1/genética , Integrina beta1/fisiología , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Modelos Biológicos , Mutación , Neuropéptidos/fisiología , ARN Interferente Pequeño/genética , Transducción de Señal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Virulencia/genética , Virulencia/fisiología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/fisiología
18.
J Leukoc Biol ; 89(2): 251-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21084629

RESUMEN

Macrophages function as key inflammatory mediators at sites of infection and tissue damage. Integrin and growth factor receptors facilitate recruitment of monocytes/macrophages to sites of inflammation in response to numerous extracellular stimuli. We have shown recently that FAK plays a role in regulating macrophage chemotaxis and invasion. As FAK is an established downstream mediator of integrin signaling, we sought to define the molecular circuitry involving FAK and the predominant ß1 integrin heterodimers expressed in these cells-α4ß1 and α5ß1. We show that α4ß1 and α5ß1 integrins are required for efficient haptotactic and chemotactic invasion and that stimulation of these integrin receptors leads to the adoption of distinct morphologies associated with motility. FAK is required downstream of α5ß1 for haptotaxis toward FN and chemotaxis toward M-CSF-1 and downstream of α4ß1 for the adoption of a polarized phenotype. The scaffolding molecule paxillin functions independently of FAK to promote chemotaxis downstream of α4ß1. These studies expand our understanding of ß1 integrin signaling networks that regulate motility and invasion in macrophages and thus, provide important new insights into mechanisms by which macrophages perform their diverse functions.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Quinasa 1 de Adhesión Focal/fisiología , Integrina alfa4beta1/fisiología , Integrina alfa5beta1/fisiología , Macrófagos/inmunología , Paxillin/fisiología , Transducción de Señal/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Polaridad Celular/genética , Polaridad Celular/inmunología , Quimiotaxis de Leucocito/genética , Quinasa 1 de Adhesión Focal/deficiencia , Quinasa 1 de Adhesión Focal/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Transducción de Señal/genética
19.
Cancer Res ; 69(2): 466-74, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19147559

RESUMEN

Focal adhesion kinase (FAK) has been implicated in the development of cancers, including those of the breast. Nevertheless, the molecular and cellular mechanisms by which FAK promotes mammary tumorigenesis in vivo are not well understood. Here, we show that targeted deletion of FAK in mouse mammary epithelium significantly suppresses mammary tumorigenesis in a well-characterized breast cancer model. Ablation of FAK leads to the depletion of a subset of bipotent cells in the tumor that express both luminal marker keratin 8/18 and basal marker keratin 5. Using mammary stem/progenitor markers, including aldehyde dehydrogenase, CD24, CD29, and CD61, we further revealed that ablation of FAK reduced the pool of cancer stem/progenitor cells in primary tumors of FAK-targeted mice and impaired their self-renewal and migration in vitro. Finally, through transplantation in NOD-SCID mice, we found that cancer stem/progenitor cells isolated from FAK-targeted mice have compromised tumorigenicity and impaired maintenance in vivo. Together, these results show a novel function of FAK in maintaining the mammary cancer stem/progenitor cell population and provide a novel mechanism by which FAK may promote breast cancer development and progression.


Asunto(s)
Quinasa 1 de Adhesión Focal/deficiencia , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Aldehído Deshidrogenasa/biosíntesis , Animales , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Femenino , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...