RESUMEN
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is overexpressed and activated in many cancer types. FAK regulates diverse cellular processes, including growth factor signaling, cell cycle progression, cell survival, cell motility, angiogenesis, and the establishment of immunosuppressive tumor microenvironments through kinase-dependent and kinase-independent scaffolding functions in the cytoplasm and nucleus. Mounting evidence has indicated that targeting FAK, either alone or in combination with other agents, may represent a promising therapeutic strategy for various cancers. In this review, we summarize the mechanisms underlying FAK-mediated signaling networks during tumor development. We also summarize the recent progress of FAK-targeted small-molecule compounds for anticancer activity from preclinical and clinical evidence.
Asunto(s)
Quinasa 1 de Adhesión Focal/fisiología , Neoplasias , Animales , Antineoplásicos/farmacología , Humanos , Neoplasias/metabolismo , Neoplasias/terapia , Microambiente TumoralRESUMEN
BACKGROUND: Hepatitis B virus (HBV) has a crucial role in the progression of hepatocellular carcinoma (HCC). Tumour cells must develop anoikis resistance in order to survive before metastasis. This study aimed to investigate the mechanism of IQGAP1 in HBV-mediated anoikis evasion and metastasis in HCC cells. METHODS: IQGAP1 expression was detected by immunohistochemistry, real-time PCR and immunoblot analysis. Lentiviral-mediated stable upregulation or knockdown of IGAQP1, immunoprecipitation, etc. were used in function and mechanism study. RESULTS: IQGAP1 was markedly upregulated in HBV-positive compared with HBV-negative HCC cells and tissues. IQGAP1 was positively correlated to poor prognosis of HBV-associated HCC patients. IQGAP1 overexpression significantly enhanced the anchorage-independent growth and metastasis, whereas IQGAP1-deficient HCC cells are more sensitive to anoikis. Mechanistically, we found that HBV-induced ROS enhanced the association of IQGAP1 and Rac1 that activated Rac1, leading to phosphorylation of Src/FAK pathway. Antioxidants efficiently inhibited IQGAP1-mediated anoikis resistance and metastasis. CONCLUSIONS: Our study indicated an important mechanism by which upregulated IQGAP1 by HBV promoted anoikis resistance, migration and invasion of HCC cells through Rac1-dependent ROS accumulation and activation of Src/FAK signalling, suggesting IQGAP1 as a prognostic indicator and a novel therapeutic target in HCC patients with HBV infection.
Asunto(s)
Carcinoma Hepatocelular/patología , Quinasa 1 de Adhesión Focal/fisiología , Neoplasias Hepáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP rac1/fisiología , Proteínas Activadoras de ras GTPasa/fisiología , Familia-src Quinasas/fisiología , Animales , Anoicis , Línea Celular Tumoral , Femenino , Hepatitis B/complicaciones , Humanos , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Transducción de Señal/fisiologíaRESUMEN
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of children and adolescents. The fusion-positive (FP)-RMS variant expressing chimeric oncoproteins such as PAX3-FOXO1 and PAX7-FOXO1 is at high risk. The fusion negative subgroup, FN-RMS, has a good prognosis when non-metastatic. Despite a multimodal therapeutic approach, FP-RMS and metastatic FN-RMS often show a dismal prognosis with 5-year survival of less than 30%. Therefore, novel targets need to be discovered to develop therapies that halt tumor progression, reducing long-term side effects in young patients. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that regulates focal contacts at the cellular edges. It plays a role in cell motility, survival, and proliferation in response to integrin and growth factor receptors' activation. FAK is often dysregulated in cancer, being upregulated and/or overactivated in several adult and pediatric tumor types. In RMS, both in vitro and preclinical studies point to a role of FAK in tumor cell motility/invasion and proliferation, which is inhibited by FAK inhibitors. In this review, we summarize the data on FAK expression and modulation in RMS. Moreover, we give an overview of the approaches to inhibit FAK in both preclinical and clinical cancer settings.
Asunto(s)
Quinasa 1 de Adhesión Focal/fisiología , Rabdomiosarcoma/fisiopatología , Neoplasias de los Tejidos Blandos/fisiopatología , Animales , Carcinogénesis , Niño , Ensayos Clínicos como Asunto , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Desarrollo de Músculos , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Rabdomiosarcoma/genética , Rabdomiosarcoma/terapia , Transducción de Señal , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/terapiaRESUMEN
Focal adhesion kinase (FAK) controls adhesion-dependent cell motility, survival, and proliferation. FAK has kinase-dependent and kinase-independent functions, both of which play major roles in embryogenesis and tumor invasiveness. The precise mechanisms of FAK activation are not known. Using x-ray crystallography, small angle x-ray scattering, and biochemical and functional analyses, we show that the key step for activation of FAK's kinase-dependent functions--autophosphorylation of tyrosine-397--requires site-specific dimerization of FAK. The dimers form via the association of the N-terminal FERM domain of FAK and are stabilized by an interaction between FERM and the C-terminal FAT domain. FAT binds to a basic motif on FERM that regulates co-activation and nuclear localization. FAK dimerization requires local enrichment, which occurs specifically at focal adhesions. Paxillin plays a dual role, by recruiting FAK to focal adhesions and by reinforcing the FAT:FERM interaction. Our results provide a structural and mechanistic framework to explain how FAK combines multiple stimuli into a site-specific function. The dimer interfaces we describe are promising targets for blocking FAK activation.
Asunto(s)
Quinasa 1 de Adhesión Focal/química , Secuencias de Aminoácidos , Animales , Cristalografía por Rayos X , Dimerización , Activación Enzimática , Quinasa 1 de Adhesión Focal/fisiología , Adhesiones Focales , Células HEK293 , Humanos , Modelos Moleculares , Fosforilación , Fosfotirosina/fisiología , Conformación Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Dispersión de RadiaciónRESUMEN
Objectives: The aim was to explore the function of the T-cell cytokine IFNγ for mesenchymal tissue remodelling in RA and to determine whether IFNγ signalling controls the invasive potential of fibroblast-like synoviocytes (FLS). Methods: To assess architectural responses, FLS were cultured in three-dimensional micromasses. FLS motility was analysed in migration and invasion assays. Signalling events relevant to cellular motility were defined by western blots. Baricitinib and small interfering RNA pools were used to suppress Janus kinase (JAK) functions. Results: Histological analyses of micromasses revealed unique effects of IFNγ on FLS shape and tissue organization. This was consistent with accelerated migration upon IFNγ stimulation. Given that cell shape and cell motility are under the control of the focal adhesion kinase (FAK), we next analysed its activity. Indeed, IFNγ stimulation induced the phosphorylation of FAK-Y925, a phosphosite implicated in FAK-mediated cell migration. Small interfering RNA knockdown of JAK2, but not JAK1, substantially abrogated FAK activation by IFNγ. Correspondingly, IFNγ-induced FAK activation and invasion of FLS was abrogated by the JAK inhibitor, baricitinib. Conclusion: Our study contributes insight into the synovial response to IFNγ and reveals JAK2 as a potential therapeutic target for FLS-mediated joint destruction in arthritis, especially in RA.
Asunto(s)
Artritis Reumatoide/metabolismo , Fibroblastos/metabolismo , Interferón gamma/fisiología , Janus Quinasa 2/antagonistas & inhibidores , Sinoviocitos/metabolismo , Adulto , Artritis Reumatoide/tratamiento farmacológico , Azetidinas/farmacología , Técnicas de Cultivo de Célula , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Masculino , Persona de Mediana Edad , Purinas , Pirazoles , ARN Interferente Pequeño/farmacología , Sulfonamidas/farmacologíaRESUMEN
The growth arrest-specific gene 7 (GAS7), a member of the growth-arrest-specific family, encodes three protein isoforms (GAS7A, GAS7B, and GAS7C) and plays a potential role in lung cancer as a tumor suppressor gene. In the present study, we found low endogenous expressions of GAS7C mRNA and protein in hepatocellular carcinoma (HCC) cell lines compared with normal liver cells, and that there was a distinct increase of GAS7C expression in HCC cells treated with oxaliplatin. CCK8, apoptosis, and Transwell migration assays showed that cell proliferation and motility of HepG2 and MHCC-97 H cells were inhibited by oxaliplatin, while apoptosis was increased. Interestingly, western blot analysis showed that treatment with oxaliplatin increased GAS7C and N-WASP protein levels and decreased the levels of proteins involved in the fibronectin/integrin/FAK pathway, such as FAK, in both HCC cell lines. In addition, ectopically overexpressed GAS7C obviously inhibited cell proliferation and cell motility. Flow cytometry results showed that overexpression of GAS7C induced apoptosis of HepG2 and MHCC-97 H cells. We further confirmed the correlation between GAS7C and the N-WASP/FAK/F-actin pathway by q-PCR and western blot analysis of in GAS7C-overexpressing HepG2 and MHCC-97 H cells. Inhibition of GAS7C substantially reversed the anti-cancer effect of oxaliplatin and blocked the activity of the N-WASP/FAK/F-actin pathway. Taken together, our results showed that oxaliplatin inhibits HCC cell proliferation and migration ability by up-regulating GAS7C and activating the N-WASP/FAK/F-actin pathway.
Asunto(s)
Actinas/fisiología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Quinasa 1 de Adhesión Focal/fisiología , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas del Tejido Nervioso/fisiología , Compuestos Organoplatinos/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Neuronal del Síndrome de Wiskott-Aldrich/fisiología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Hepáticas/patología , OxaliplatinoRESUMEN
OBJECTIVE: The objective of this study was to investigate the roles of dickkopf-1 (DKK-1) and integrin-related focal adhesion kinase (FAK) by TNF-α on the migration of fibroblast-like synoviocytes (FLSs) in RA. METHODS: Wound scratch assays were performed to assess FLS migration. Western blotting was used to measure the levels of DKK-1, Wnt signalling molecules and FAK signalling molecules. Quantitative real-time PCR was used to measure the expression levels of DKK-1, integrin αv, laminin, fibronectin, E-cadherin, MMP-8 and MMP-13. The concentrations of DKK-1, TNF-α and GSK-3ß were measured by ELISA. Genetic silencing of TNF-α was achieved by the transfection of small interfering RNA into cells. RESULTS: Migrating RA FLSs exhibited higher levels of DKK-1 and TNF-α expression compared with those in OA FLSs and/or stationary RA FLSs. Moreover, migrating FLSs exhibited significantly higher levels of FAK, p-JNK, paxillin and cdc42 expression, whereas the level of cytosolic ß-catenin was lower. WAY-262611, Wnt pathway agonist via inhibition of DKK-1, markedly inhibited cell migration of RA FLSs through the accumulation of cytosolic ß-catenin and suppression of FAK-related signalling pathways. TNF-α treatment to RA FLSs up-regulated expression of DKK-1, integrin αv, fibronectin, laminin and MMP-13. TNF-α stimulation also suppressed cytosolic ß-catenin and E-cadherin expression in a time-dependent manner. Moreover, TNF-α small interfering RNA-transfected migrating FLSs exhibited decreased activation of integrin-related FAK, paxillin, p-JNK and cdc42 signalling pathways. CONCLUSION: This study demonstrates that the activation of DKK-1 and the integrin-related FAK signalling pathway stimulated by TNF-α induces the dissociation of ß-catenin/E-cadherin, thus promoting RA FLS migration.
Asunto(s)
Artritis Reumatoide/patología , Quinasa 1 de Adhesión Focal/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/fisiología , Anciano , Artritis Reumatoide/metabolismo , Cadherinas/metabolismo , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Fibroblastos/fisiología , Quinasa 1 de Adhesión Focal/genética , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Persona de Mediana Edad , Osteoartritis/metabolismo , Osteoartritis/patología , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/genética , beta Catenina/metabolismoRESUMEN
Coumarins, identified as plant secondary metabolites possess diverse biological activities including anti-angiogenic properties. Daphnetin (DAP), a plant derived dihydroxylated derivative of coumarin has shown significant pharmacological properties such as anticancer, anti-arthritic and anti-inflammatory. The present study was performed to investigate the anti-angiogenic potential of DAP, focusing on the mechanism of action. The in vivo anti-angiogenic potential of DAP was evaluated by vascular endothelial growth factor (VEGF)-induced rat aortic ring (RAR) assay and chick chorioallantoic membrane (CAM) assay. For in vitro evaluation, wounding migration, transwell invasion, tube formation and apoptosis assays were performed on VEGF (8 ng/mL)-induced human umbilical vein endothelial cells (HUVECs). The cellular mechanism of DAP was examined on TNFα (10 ng/mL) and VEGF-induced HUVECs by extracting the mRNA and protein levels using RT-qPCR and western blotting. Our data demonstrated that DAP inhibited the in vivo angiogenesis in the RAR and CAM assay. DAP also inhibited the different steps of angiogenesis, such as migration, invasion, and tube formation in HUVECs. DAP inhibited nuclear factor-κB signalling together including TNF-α induced IκBα degradation; phosphorylation of IκB kinase (IKKα/ß) and translocation of the NF-κB-p65 protein. Furthermore, western blotting revealed that DAP significantly down-regulated the VEGF-induced signalling such as c-Src, FAK, ERK1/2 and the related phosphorylation of protein kinase B (Akt) and VEGFR2 expressions. DAP reduced the elevated mRNA expression of iNOS, MMP2 and also, induced apoptosis in VEGF-stimulated HUVECs by the caspase-3 dependent pathway. Taken together, this study reveals that DAP may have novel prospective as a new multi-targeted medication for the anti-angiogenesis and cancer therapy.
Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Umbeliferonas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inductores de la Angiogénesis/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/fisiología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , FN-kappa B/fisiología , Técnicas de Cultivo de Órganos , Proteínas Proto-Oncogénicas c-akt/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacologíaRESUMEN
Cells are mechanosensitive to extracellular matrix (ECM) deformation, which can be caused by muscle contraction or changes in hydrostatic pressure. Focal adhesions (FAs) mediate the linkage between the cell and the ECM and initiate mechanically stimulated signaling events. We developed a stretching apparatus in which cells grown on fibronectin-coated elastic substrates can be stretched and imaged live to study how FAs dynamically respond to ECM deformation. Human bone osteosarcoma epithelial cell line U2OS was transfected with GFP-paxillin as an FA marker and subjected to sustained uniaxial stretching. Two responses at different timescales were observed: rapid FA growth within seconds after stretching, and delayed FA disassembly and loss of cell polarity that occurred over tens of minutes. Rapid FA growth occurred in all cells; however, delayed responses to stretch occurred in an orientation-specific manner, specifically in cells with their long axes perpendicular to the stretching direction, but not in cells with their long axes parallel to stretch. Pharmacological treatments demonstrated that FA kinase (FAK) promotes but Src inhibits rapid FA growth, whereas FAK, Src, and calpain 2 all contribute to delayed FA disassembly and loss of polarity in cells perpendicular to stretching. Immunostaining for phospho-FAK after stretching revealed that FAK activation was maximal at 5 s after stretching, specifically in FAs oriented perpendicular to stretch. We hypothesize that orientation-specific activation of strain/stress-sensitive proteins in FAs upstream to FAK and Src promote orientation-specific responses in FA growth and disassembly that mediate polarity rearrangement in response to sustained stretch.
Asunto(s)
Polaridad Celular/fisiología , Adhesiones Focales/fisiología , Mecanotransducción Celular/fisiología , Calpaína/fisiología , Línea Celular Tumoral , Matriz Extracelular/fisiología , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Miosina Tipo II/fisiología , Estrés Mecánico , Familia-src Quinasas/fisiologíaRESUMEN
One of the key features of keloid is its fibroblasts migrating beyond the original wound border. During migration, cells not only undergo molecular changes but also mechanical modulation. This process is led by actin filaments serving as the backbone of intra-cellular force and transduces external mechanical signal via focal adhesion complex into the cell. Here, we focus on determining the mechanical changes of actin filaments and the spatial distribution of forces in response to changing chemical stimulations and during cell migration. Atomic force microscopy and micropost array detector are used to determine and compare the magnitude and distribution of filament elasticity and force generation in fibroblasts and keloid fibroblasts. We found both filament elasticity and force generation show spatial distribution in a polarized and migrating cell. Such spatial distribution is disrupted when mechano-signalling is perturbed by focal adhesion kinase inhibitor and in keloid fibroblasts. The demonstration of keloid pathology at the nanoscale highlights the coupling of cytoskeletal function with physical characters at the subcellular level and provides new research directions for migration-related disease such as keloid.
Asunto(s)
Citoesqueleto/fisiología , Fibroblastos/fisiología , Queloide/patología , Citoesqueleto de Actina/fisiología , Animales , Movimiento Celular , Polaridad Celular , Elasticidad , Fibroblastos/ultraestructura , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/fisiología , Adhesiones Focales/fisiología , Humanos , Ratones , Microscopía de Fuerza Atómica , Células 3T3 NIH , Quinolonas/farmacología , Estrés Mecánico , Sulfonas/farmacología , Cicatrización de HeridasRESUMEN
Tendon regeneration and healing requires tenocytes to move to the repair site followed by proliferation and synthesis of the extracellular matrix. A novel synthetic growth factor, mechano-growth factor (MGF), has been discovered to have positive roles in tissue repair through the improvement of cell proliferation and migration and the protection of cells against injury-induced apoptosis. However, it remains unclear whether MGF has the potential to accelerate tendon repair. In this study, using a transwell system, we found that MGF-C25E (a synthetic mechano-growth factor E peptide) significantly promotes tenocyte invasion, which was accompanied by the increased phosphorylation of focal adhesion kinase (FAK) and extracellular signal regulated kinase1/2 (ERK1/2) as well as the increased activity of matrix metalloproteinases-2 (MMP-2). The MMP-2 inhibitor OA-Hy blocked MGF-C25E-promoted tenocyte invasion. Inhibitors of FAK or ERK1/2 blocked MGF-C25E-promoted tenocyte invasion and MMP-2 activity as well. These results indicate that MGF-C25E promotes tenocyte invasion by increasing MMP-2 activity via the FAK-ERK1/2 signaling pathway. Taken together, our findings provide the first evidence that MGF-C25E enhances tenocyte invasion and indicate that it may serve as a potential repair material for promoting the healing and regeneration of injured tendons.
Asunto(s)
Tendón Calcáneo/patología , Quinasa 1 de Adhesión Focal/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Metaloproteinasa 2 de la Matriz/metabolismo , Cicatrización de Heridas , Tendón Calcáneo/citología , Tendón Calcáneo/lesiones , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , RegeneraciónRESUMEN
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/metabolismoRESUMEN
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 NoqueadosRESUMEN
UNLABELLED: Hepatocellular carcinoma (HCC) is one of the most common cancers and shows a propensity to metastasize and infiltrate adjacent and more distant tissues. However, the mechanisms that contribute to tumor metastasis remain unclear. Here we evaluate the effect of Argonaute2 (Ago2), a member of the Ago gene family that plays a role in short interfering RNA-mediated gene silencing, on HCC tumorigenesis, and metastasis. We found that Ago2 was frequently up-regulated in HCC specimens compared to that in corresponding adjacent nontumor liver. Interestingly, Ago2 overexpression can promote proliferation, colony formation in an anchor-independent manner, migration, tumorigenicity, and metastasis of HCC cells in vivo; in contrast, Ago2 knockdown can restrict anchor-independent colony formation, migration, and tumor metastasis of HCC cells in vivo. However, known microRNAs related to tumor metastasis appeared not be deregulated with Ago2 overexpression in HCC cells; even the knockdown of Dicer, which is responsible for microRNA biosynthesis, did not abolish the actions of Ago2 in HCC cells. Significantly, focal adhesion kinase (FAK), a well-known molecule associated with tumor metastasis, was up-regulated as a result of Ago2 overexpression. Chromatin immunoprecipitation assay showed that Ago2 can bind to the FAK promoter and then trigger its transcription. Moreover, an increased DNA copy number of Ago2 on chromosome 8q24, one of the most frequent DNA amplified regions, was validated and shown by way of fluorescence in situ hybridization. CONCLUSION: Our data demonstrate that Ago2 overexpression, as a result of genomic DNA amplification, promotes HCC tumorigenesis and metastasis by way of up-regulation of FAK transcription, thereby providing new insight into HCC progression and Ago2 function.
Asunto(s)
Proteínas Argonautas/fisiología , Carcinoma Hepatocelular/fisiopatología , Quinasa 1 de Adhesión Focal/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Hepáticas/fisiopatología , Metástasis de la Neoplasia/fisiopatología , Regulación hacia Arriba/fisiología , Animales , Proteínas Argonautas/antagonistas & inhibidores , Proteínas Argonautas/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Técnicas de Amplificación de Ácido Nucleico , ARN Interferente Pequeño/farmacología , Trasplante HeterólogoRESUMEN
The lymphatic system in skin plays important roles in drainage of wastes and in the afferent phase of immune response. We previously showed that activation of vascular endothelial growth factor receptor (VEGFR), specifically the VEGFC/VEGFR-3 pathway, attenuates oedema and inflammation by promoting lymphangiogenesis, suggesting a protective role of lymphatic vessels against skin inflammation. However, it remains unknown how physical stimuli promote lymphatic function. Here, we show that lymphatic endothelial cells (LECs) are activated by direct-current (DC) electrical stimulation, which induced extension of actin filaments of LECs, increased calcium influx into LECs, and increased phosphorylation of p38 mitogen-activated protein kinase (MAPK). An inhibitor of focal adhesion kinase, which plays a role in cellular adhesion and motility, diminished the DC-induced extension of F-actin and abrogated p38 phosphorylation. Time-lapse imaging revealed that pulsed-DC stimulation promoted proliferation and migration of LECs. Overall, these results indicate that electro-stimulation activates lymphatic function by activating p38 MAPK.
Asunto(s)
Células Endoteliales/fisiología , Señalización del Calcio , Movimiento Celular , Proliferación Celular , Células Cultivadas , Citoesqueleto/fisiología , Estimulación Eléctrica , Células Endoteliales/citología , Quinasa 1 de Adhesión Focal/fisiología , HumanosRESUMEN
The activation of signaling pathways involving protein tyrosine kinases (PTKs) has been demonstrated during Trypanosoma cruzi invasion. Herein, we describe the participation of FAK/Src in the invasion of cardiomyocytes by T. cruzi. The treatment of cardiomyocytes with genistein, a PTK inhibitor, significantly reduced T. cruzi invasion. Also, PP1, a potent Src-family protein inhibitor, and PF573228, a specific FAK inhibitor, also inhibited T. cruzi entry; maximal inhibition was achieved at concentrations of 25µM PP1 (53% inhibition) and 40µM PF573228 (50% inhibition). The suppression of FAK expression in siRNA-treated cells and tetracycline-uninduced Tet-FAK(WT)-46 cells significantly reduced T. cruzi invasion. The entry of T. cruzi is accompanied by changes in FAK and c-Src expression and phosphorylation. An enhancement of FAK activation occurs during the initial stages of T. cruzi-cardiomyocyte interaction (30 and 60min), with a concomitant increase in the level of c-Src expression and phosphorylation, suggesting that FAK/Src act as an integrated signaling pathway that coordinates parasite entry. These data provide novel insights into the signaling pathways that are involved in cardiomyocyte invasion by T. cruzi. A better understanding of the signal transduction networks involved in T. cruzi invasion may contribute to the development of more effective therapies for the treatment of Chagas' disease.
Asunto(s)
Quinasa 1 de Adhesión Focal/fisiología , Miocitos Cardíacos/parasitología , Transducción de Señal/fisiología , Trypanosoma cruzi/fisiología , Familia-src Quinasas/fisiología , Animales , Proteína Tirosina Quinasa CSK , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Fosforilación , Pirazoles/farmacología , Pirimidinas/farmacología , Quinolonas/farmacología , ARN Interferente Pequeño/fisiología , Sulfonas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismoRESUMEN
Interstitial flow is the convective transport of fluid through tissue extracellular matrix. This creeping fluid flow has been shown to affect the morphology and migration of cells such as fibroblasts, cancer cells, endothelial cells, and mesenchymal stem cells. A microfluidic cell culture system was designed to apply stable pressure gradients and fluid flow and allow direct visualization of transient responses of cells seeded in a 3D collagen type I scaffold. We used this system to examine the effects of interstitial flow on cancer cell morphology and migration and to extend previous studies showing that interstitial flow increases the metastatic potential of MDA-MB-435S melanoma cells [Shields J, et al. (2007) Cancer Cell 11:526-538]. Using a breast carcinoma line (MDA-MB-231) we also observed cell migration along streamlines in the presence of flow; however, we further demonstrated that the strength of the flow as well as the cell density determined directional bias of migration along the streamline. In particular, we found that cells either at high seeding density or with the CCR-7 receptor inhibited migration against, rather than with the flow. We provide further evidence that CCR7-dependent autologous chemotaxis is the mechanism that leads to migration with the flow, but also demonstrate a competing CCR7-independent mechanism that causes migration against the flow. Data from experiments investigating the effects of cell concentration, interstitial flow rate, receptor activity, and focal adhesion kinase phosphorylation support our hypothesis that the competing stimulus is integrin mediated. This mechanism may play an important role in development of metastatic disease.
Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Movimiento Celular/fisiología , Ingeniería Biomédica , Neoplasias de la Mama/secundario , Recuento de Células , Línea Celular Tumoral , Quimiotaxis/fisiología , Líquido Extracelular/fisiología , Femenino , Análisis de Elementos Finitos , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Receptores CCR7/antagonistas & inhibidores , Receptores CCR7/fisiología , Transducción de SeñalRESUMEN
Neurofibromatosis type I (NF1), caused by the mutation in the NF1 gene, is characterized by multiple pathological symptoms. Importantly, ~50% of NF1 patients also suffer learning difficulty. Although downstream pathways are well studied, regulation of the NF1-encoded neurofibromin protein is less clear. Here, we focused on the pathophysiology of Drosophila NF1 mutants in synaptic growth at neuromuscular junctions. Our analysis suggests that the Drosophila neurofibromin protein NF1 is required to constrain synaptic growth and transmission. NF1 functions downstream of the Drosophila focal adhesion kinase (FAK) Fak56 and physically interacts with Fak56. The N-terminal region of NF1 mediates the interaction with Fak56 and is required for the signaling activity and presynaptic localization of NF1. In presynapses, NF1 acts via the cAMP pathway, but independent of its GAP activity, to restrain synaptic growth. Thus, presynaptic FAK signaling may be disrupted, causing abnormal synaptic growth and transmission in the NF1 genetic disorder.
Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/fisiología , Quinasa 1 de Adhesión Focal/fisiología , Proteínas del Tejido Nervioso/fisiología , Unión Neuromuscular/fisiología , Sinapsis/fisiología , Proteínas Activadoras de ras GTPasa/fisiología , Adenilil Ciclasas/fisiología , Animales , AMP Cíclico/fisiología , Fenómenos Electrofisiológicos/fisiología , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Larva , Masculino , Microscopía Electrónica , Mutación/fisiología , Receptores Presinapticos/fisiología , Transducción de Señal/fisiología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiologíaRESUMEN
BACKGROUND: Alternative splicing of EDA fibronectin (FN) region is a cell type- and development-regulated mechanism controlled by pathological processes, growth factors and extracellular matrix (ECM). Classic and vascular Ehlers-Danlos syndrome (cEDS and vEDS) are connective tissue disorders caused by COL5A1/COL5A2 and COL3A1 gene mutations, leading to an in vivo abnormal collagen fibrillogenesis and to an in vitro defective organisation in the ECM of type V (COLLV) and type III collagen (COLLIII). These defects induce the FN-ECM disarray and the decrease of COLLs and FN receptors, the α2ß1 and α5ß1 integrins. Purified COLLV and COLLIII restore the COLL-FN-ECMs in both EDS cell strains. METHODS: Real-time PCR, immunofluorescence microscopy, and Western blotting were used to investigate the effects of COLLs on FN1 gene expression, EDA region alternative splicing, EDA(+)-FN-ECM assembly, α5ß1 integrin and EDA(+)-FN-specific α9 integrin subunit organisation, α5ß1 integrin and FAK co-regulation in EDS fibroblasts. RESULTS: COLLV-treated cEDS and COLLIII-treated vEDS fibroblasts up-regulate the FN1 gene expression, modulate the EDA(+) mRNA maturation and increase the EDA(+)-FN levels, thus restoring a control-like FN-ECM, which elicits the EDA(+)-FN-specific α9ß1 integrin organisation, recruits the α5ß1 integrin and switches on the FAK binding and phosphorylation. CONCLUSION: COLLs regulate the EDA(+)-FN-ECM organisation at transcriptional and post-transcriptional level and activate the α5ß1-FAK complexes. COLLs also recruit the α9ß1 integrin involved in the assembly of the EDA(+)-FN-ECM in EDS cells. GENERAL SIGNIFICANCE: The knowledge of the COLLs-ECM role in FN isotype expression and in EDA(+)-FN-ECM-mediated signal transduction adds insights in the ECM remodelling mechanisms in EDS cells.
Asunto(s)
Colágeno Tipo III/farmacología , Colágeno Tipo V/farmacología , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Síndrome de Ehlers-Danlos/metabolismo , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrina alfa5beta1/metabolismo , Integrinas/metabolismo , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerización de Proteína/efectos de los fármacosRESUMEN
PURPOSE: The requirement of integrins for mechanotransduction has been recognized for some time. We investigated the role of integrin subunits and their pathway in the physiological stretch induced contractility and proliferation of human bladder smooth muscle cells. MATERIALS AND METHODS: Human bladder smooth muscle cells were seeded on silicone membrane and subjected to stretch, simulating bladder cycles of various stretches and times, as controlled by customized software on a modified BioDynamic bioreactor. Cell proliferation, viability and cycle were determined by BrdU incorporation assay, the Cell Counting Kit-8 (Beyotime Institute of Biotechnology, Haimen, People's Republic of China) and flow cytometry, respectively. Cell contractility was determined using a collagen gel contraction assay. RESULTS: Physiological stretch increased cell contractility, proliferation and viability. Knockdown of integrin αv but not α4 in the cells disrupted the enhanced contractility induced by stretch. Under physiological stretch conditions, the integrin αv level and phospho-FAK/FAK ratio correlated positively with cell stretch induced enhanced contractility. Further examination revealed that contractile marker expression was associated with integrin αv activation through the FAK pathway. At the same time integrin α4 but not integrin αv mediated stretch induced cell proliferation and viability. CONCLUSIONS: These data revealed that different integrins have different roles in the contractility and proliferation of human bladder smooth muscle cells under physiological stretch. This suggests that different integrins may become specific therapeutic targets in patients with voiding dysfunction. They may also be used to design a specific microenvironment for optimal bladder tissue regeneration.