Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
J Biol Chem ; 291(15): 8031-47, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26826125

RESUMEN

Herein, we have identified cross-talk between the Hippo and fibroblast growth factor receptor (FGFR) oncogenic signaling pathways in cholangiocarcinoma (CCA). Yes-associated protein (YAP) nuclear localization and up-regulation of canonical target genes was observed in CCA cell lines and a patient-derived xenograft (PDX). Expression of FGFR1, -2, and -4 was identified in human CCA cell lines, driven, in part, by YAP coactivation of TBX5. In turn, FGFR signaling in a cell line with minimal basal YAP expression induced its cellular protein expression and nuclear localization. Treatment of YAP-positive CCA cell lines with BGJ398, a pan-FGFR inhibitor, resulted in a decrease in YAP activation. FGFR activation of YAP appears to be driven largely by FGF5 activation of FGFR2, as siRNA silencing of this ligand or receptor, respectively, inhibited YAP nuclear localization. BGJ398 treatment of YAP-expressing cells induced cell death due to Mcl-1 depletion. In a YAP-associated mouse model of CCA, expression of FGFR 1, 2, and 4 was also significantly increased. Accordingly, BGJ398 treatment was tumor-suppressive in this model and in a YAP-positive PDX model. These preclinical data suggest not only that the YAP and Hippo signaling pathways culminate in an Mcl-1-regulated tumor survival pathway but also that nuclear YAP expression may be a biomarker to employ in FGFR-directed therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares/patología , Colangiocarcinoma/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares/metabolismo , Línea Celular Tumoral , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Ratones , Ratones SCID , Fosfoproteínas/análisis , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
2.
Dev Biol ; 351(1): 176-85, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21223962

RESUMEN

Previous studies suggested that FGF signaling is important for lens formation. However, the times at which FGFs act to promote lens formation, the FGFs that are involved, the cells that secrete them and the mechanisms by which FGF signaling may promote lens formation are not known. We found that transcripts encoding several FGF ligands and the four classical FGF receptors are detectable in the lens-forming ectoderm at the time of lens induction. Conditional deletion of Fgfr1 and Fgfr2 from this tissue resulted in the formation of small lens rudiments that soon degenerated. Lens placodes lacking Fgfr1 and 2 were thinner than in wild-type embryos. Deletion of Fgfr2 increased cell death from the initiation of placode formation and concurrent deletion of Fgfr1 enhanced this phenotype. Fgfr1/2 conditional knockout placode cells expressed lower levels of proteins known to be regulated by FGF receptor signaling, but proteins known to be important for lens formation were present at normal levels in the remaining placode cells, including the transcription factors Pax6, Sox2 and FoxE3 and the lens-preferred protein αA-crystallin. Previous studies identified a genetic interaction between BMP and FGF signaling in lens formation and conditional deletion of Bmpr1a caused increased cell death in the lens placode, resulting in the formation of smaller lenses. In the present study, conditional deletion of both Bmpr1a and Fgfr2 increased cell death beyond that seen in Fgfr2(CKO) placodes and prevented lens formation. These results suggest that the primary role of autocrine or paracrine FGF signaling is to provide essential survival signals to lens placode cells. Because apoptosis was already increased at the onset of placode formation in Fgfr1/2 conditional knockout placode cells, FGF signaling was functionally absent during the period of lens induction by the optic vesicle. Since the expression of proteins required for lens formation was not altered in the knockout placode cells, we can conclude that FGF signaling from the optic vesicle is not required for lens induction.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Cristalino/embriología , Transducción de Señal/fisiología , Animales , Receptores de Proteínas Morfogenéticas Óseas/fisiología , Ectodermo/química , Proteínas del Ojo/fisiología , Factores de Crecimiento de Fibroblastos/análisis , Mutación de Línea Germinal , Proteínas de Homeodominio/fisiología , Ratones , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Proteínas Represoras/fisiología
3.
Sci Rep ; 10(1): 17054, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051476

RESUMEN

We investigated whether or not nitric oxide synthase 3 (NOS3) rs2070744 genotypes can affect the response for lenvatinib treatment in patients with hepatocellular carcinoma (HCC). We evaluated the relation of the NOS3 rs2070744 genotypes to the tumor response, progression-free survival (PFS), and overall survival (OS) as the response for lenvatinib. We also examined the association between fibroblast growth factor receptor (FGFR) gene polymorphisms, a potential feature of lenvatinib, and the response. There were no significant differences between the studies for either PFS or OS, even though patients with the TT genotype had a longer mean PFS (hazard ratio [HR] 0.60; p = 0.069) and mean OS (HR 0.46; p = 0.075) than those with the TC/CC genotypes. However, patients with a single-nucleotide polymorphism (SNP) combination pattern of the NOS3 rs2070744 TC/CC and FGFR4 rs351855 CT/TT genotypes had a significantly shorter mean PFS (HR 2.56; p = 0.006) and mean OS (HR 3.36; p = 0.013) than those with the other genotypes. The NOS3 rs2070744 genotypes did not influence the clinical response. However, the SNP combination pattern of the NOS3 rs2070744 and FGFR4 rs351855 genotypes may be helpful as treatment effect predictors and prognostic factors for HCC patients treated with lenvatinib.


Asunto(s)
Carcinoma Hepatocelular/genética , Óxido Nítrico Sintasa de Tipo III/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores Farmacológicos , Carcinoma Hepatocelular/tratamiento farmacológico , Supervivencia sin Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Compuestos de Fenilurea/farmacología , Polimorfismo de Nucleótido Simple/genética , Pronóstico , Quinolinas/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética
4.
Neuron ; 15(5): 1017-28, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7576646

RESUMEN

We report that the growth cones of Xenopus retinal ganglion cells express fibroblast growth factor receptors (FGFRs) and that bFGF stimulates neurite extension from cultured retinal neurons. Furthermore, bFGF is abundant in the developing optic tract but is reduced in the optic tectum. To test whether FGF signaling plays a role in axonal guidance in vivo, bFGF was exogenously applied to the developing optic pathway in "exposed brain" preparations. FGF-treated retinal axons navigate normally through the optic tract, but the majority veer aberrantly at the tectal border and bypass the target. Our results implicate FGF signaling in target recognition and suggest that diminished levels of bFGF in the tectum cause arriving axons to slow their growth.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Ganglionares de la Retina/fisiología , Transducción de Señal , Colículos Superiores/embriología , Xenopus/embriología , Animales , Axones/fisiología , Axones/ultraestructura , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/análisis , Neuritas/química , Neuritas/ultraestructura , Quiasma Óptico/química , Quiasma Óptico/embriología , Quiasma Óptico/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Células Ganglionares de la Retina/ultraestructura , Colículos Superiores/química , Colículos Superiores/metabolismo , Colículos Superiores/ultraestructura
5.
Mol Cancer ; 7: 30, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18412956

RESUMEN

BACKGROUND: The t(6;8) translocation found in rare and agressive myeloproliferative disorders results in a chimeric gene encoding the FOP-FGFR1 fusion protein. This protein comprises the N-terminal region of the centrosomal protein FOP and the tyrosine kinase of the FGFR1 receptor. FOP-FGFR1 is localized at the centrosome where it exerts a constitutive kinase activity. RESULTS: We show that FOP-FGFR1 interacts with the large centrosomal protein CAP350 and that CAP350 is necessary for FOP-FGFR1 localisation at centrosome. FOP-FGFR1 activates the phosphoinositide-3 kinase (PI3K) pathway. We show that p85 interacts with tyrosine 475 of FOP-FGFR1, which is located in a YXXM consensus binding sequence for an SH2 domain of p85. This interaction is in part responsible for PI3K activation. Ba/F3 cells that express FOP-FGFR1 mutated at tyrosine 475 have reduced proliferative ability. Treatment with PI3K pathway inhibitors induces death of FOP-FGFR1 expressing cells. FOP-FGFR1 also recruits phospholipase Cgamma1 (PLCgamma1) at the centrosome. We show that this enzyme is recruited by FOP-FGFR1 at the centrosome during interphase. CONCLUSION: These results delineate a particular type of oncogenic mechanism by which an ectopic kinase recruits its substrates at the centrosome whence unappropriate signaling induces continuous cell growth and MPD.


Asunto(s)
Centrosoma/enzimología , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Células COS , Proliferación Celular , Supervivencia Celular , Chlorocebus aethiops , Células HeLa , Humanos , Interfase , Proteínas de Microtúbulos/metabolismo , Mutación , Trastornos Mieloproliferativos/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/genética , Dominios y Motivos de Interacción de Proteínas , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal
6.
Mech Dev ; 123(12): 925-40, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17011755

RESUMEN

The vertebrate craniofacial skeleton develops via a complex process involving signaling cascades in all three germ layers. Fibroblast growth factor (FGF) signaling is essential for several steps in pharyngeal arch development. In zebrafish, Fgf3 and Fgf8 in the mesoderm and hindbrain have an early role to pattern the pouch endoderm, influencing craniofacial integrity. Endodermal FGF signaling is required for the differentiation and survival of postmigratory neural crest cells that form the pharyngeal skeleton. We identify a novel role for zebrafish Fgf receptor-like 1a (Fgfrl1a) that is indispensable during gill cartilage development. We show that depletion of Fgfrl1a is sufficient to abolish cartilage derivatives of the ceratobranchials. Using an Fgfrl1a-deficient model, we analyzed expression of genes critical for chondrogenesis in the different compartments of the developing pharyngeal arch. Fgfrl1a-depleted animals demonstrate typical neural crest specification and migration to populate the arch primordia as well as normal pouch segmentation. However, in the absence of Fgfrl1a, larvae fail to express the transcription factor glial cells missing 2 (gcm2), a gene necessary for cartilage and gill filament formation, in the ectodermal lining of the branchial arches. In addition, two transcription factors essential for chondrogenesis, sox9a and runx2b, fail to express within the mesenchymal condensations of the branchial arches. A duplicate zebrafish gene, fgfrl1b, has now been identified. We show that Fgfrl1b is also required for proper formation of all ventral cartilage elements and acts cooperatively with Fgfrl1a during gill cartilage formation.


Asunto(s)
Cartílago/embriología , Branquias/embriología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Región Branquial/química , Región Branquial/embriología , Cartílago/química , Movimiento Celular/genética , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ectodermo/química , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Branquias/química , Proteínas HMGB/análisis , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Datos de Secuencia Molecular , Cresta Neural/citología , Filogenia , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Factor de Transcripción SOX9 , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/análisis , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Mol Cell Biol ; 17(10): 5739-47, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9315632

RESUMEN

Fibroblast growth factor (FGF) receptors (FGFRs) are membrane-spanning tyrosine kinase receptors that mediate regulatory signals for cell proliferation and differentiation in response to FGFs. We have previously determined that the Lys650-->Glu mutation in the activation loop of the kinase domain of FGFR3, which is responsible for the lethal skeletal dysplasia thanatophoric dyplasia type II (TDII), greatly enhances the ligand-independent kinase activity of the receptor. Here, we demonstrate that expression of this construct induces a c-fos promoter construct approximately 10-fold but does not lead to proliferation or morphological transformation of NIH 3T3 cells. In contrast, the isolated kinase domain of activated FGFR3, targeted to the plasma membrane by a myristylation signal, is able to stimulate c-fos expression by 40-fold, induce proliferation of quiescent cells, and morphologically transform fibroblasts. This result suggests that the extracellular and transmembrane domains of FGFRs exert a negative regulatory influence on the activity of the kinase domain. Targeting of the activated kinase domain to either the cytoplasm or the nucleus does not significantly affect biological signaling, suggesting that signals from FGFR3 resulting in mitogenesis originate exclusively from the plasma membrane. Furthermore, our novel observation that expression of a highly activated FGFR3 kinase domain is able to morphologically transform fibroblasts suggests that dysregulation of FGFR3 has the potential to play a role in human neoplasia.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Tirosina Quinasas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Células 3T3 , Animales , División Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Genes fos/genética , Humanos , Ratones , Ácido Mirístico , Ácidos Mirísticos/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes de Fusión
8.
Nucleic Acids Res ; 33(11): e102, 2005 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-15987787

RESUMEN

RNA interference (RNAi)-mediated gene knockdown is a potent approach for studying gene function. We have previously reported a plasmid-based, tamoxifen-inducible gene knockdown system in cultured cells using a combined RNAi and Cre-LoxP system. Here, we validate this system in mouse and show that it can be used to suppress the expression of an endogenous gene (Fgfr2) with high efficiency. We show that transgenic mice carrying the U6-ploxPneo-Fgfr2 RNAi construct are normal, displaying Fgfr2 transcripts equivalent to those of wild-type controls, indicating that the U6 promoter is inactive in vivo due to the presence of the neo in the promoter. After excision of the neo by crossing with transgenic mice that express Cre in the mouse germline, the U6 promoter is activated, leading to over 95% reduction of Fgfr2 transcripts, and consequently, embryonic lethality. On the other hand, activation of the U6 promoter using transgenic mice that express Cre in the progress zone of the limb results in live mice with malformation of digits of both the forelimbs and hindlimbs. This method provides a fast, yet efficient way to decipher gene functions in vivo in a tissue-specific manner.


Asunto(s)
Integrasas/metabolismo , Ratones Noqueados/genética , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Proteínas Virales/metabolismo , Animales , Genes Letales , Integrasas/genética , Deformidades Congénitas de las Extremidades/etiología , Mesodermo/metabolismo , Ratones , ARN Polimerasa III/genética , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Recombinación Genética , Proteínas Virales/genética
9.
Mol Biol Cell ; 15(7): 3106-13, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15107468

RESUMEN

The keratinocyte growth factor receptor (KGFR) is a member of the fibroblast growth factor receptor (FGFR) superfamily. The proximal signaling molecules of FGFRs are much less characterized compared with other growth factor receptors. Using the yeast two-hybrid assay, we have identified ribosomal S6 kinase (RSK) to be a protein that associates with the cytoplasmic domain of the KGFR. The RSK family of kinases controls multiple cellular processes, and our studies for the first time show association between the KGFR and RSK. Using a lung-specific inducible transgenic system we have recently demonstrated protective effects of KGF on the lung epithelium and have demonstrated KGF-induced activation of the prosurvival Akt pathway both in vivo and in vitro. Here we show that a kinase inactive RSK mutant blocks KGF-induced Akt activation and KGF-mediated inhibition of caspase 3 activation in epithelial cells subjected to oxidative stress. It was recently shown that RSK2 recruits PDK1, the kinase responsible for both Akt and RSK activation. When viewed collectively, it appears that the association between the KGFR and RSK plays an important role in KGF-induced Akt activation and consequently in the protective effects of KGF on epithelial cells.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Caspasa 3 , Inhibidores de Caspasas , Línea Celular , Activación Enzimática , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Factor 7 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/fisiología , Flavonoides/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Inmunoprecipitación , Ratones , Mutación/genética , Fosforilación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteínas Proto-Oncogénicas c-akt , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Proteínas Quinasas S6 Ribosómicas/análisis , Proteínas Quinasas S6 Ribosómicas/genética , Técnicas del Sistema de Dos Híbridos , Tirosina/metabolismo
10.
Mol Biol Cell ; 6(10): 1349-65, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8573791

RESUMEN

Extracellular matrix controls capillary endothelial cell sensitivity to soluble mitogens by binding integrin receptors and thereby activating a chemical signaling response that rapidly integrates with growth factor-induced signaling mechanisms. Here we report that in addition to integrins, growth factor receptors and multiple molecules that transduce signals conveyed by both types of receptors are immobilized on the cytoskeleton (CSK) and spatially integrated within the focal adhesion complex (FAC) at the site of integrin binding. FACs were rapidly induced in round cells and physically isolated from the remainder of the CSK after detergent-extraction using magnetic microbeads coated with fibronectin or a synthetic RGD-containing peptide. Immunofluorescence microscopy revealed that multiple signaling molecules (e.g., pp60c-src, pp125FAK, phosphatidylinositol-3-kinase, phospholipase C-gamma, and Na+/H+ antiporter) involved in both integrin and growth factor receptor signaling pathways became associated with the CSK framework of the FAC within 15 min after binding to beads coated with integrin ligands. Recruitment of tyrosine kinases to the FAC was also accompanied by a local increase in tyrosine phosphorylation, as indicated by enhanced phosphotyrosine staining at the site of integrin binding. In contrast, neither recruitment of signaling molecules nor increased phosphotyrosine staining was observed when cells bound to beads coated with a control ligand (acetylated low density lipoprotein) that ligates transmembrane scavenger receptors, but does not induce FAC formation. Western blot analysis confirmed that FACs isolated using RGD-beads were enriched for pp60c-src, pp125FAK, phospholipase C-gamma, and the Na+/H+ antiporter when compared with intact CSK or basal cell surface preparations that retained lipid bilayer. Isolated FACs were also greatly enriched for the high affinity fibroblast growth factor receptor flg. Most importantly, isolated FACs continued to exhibit multiple chemical signaling activities in vitro, including protein tyrosine kinase activities (pp60c-src and pp125FAK) as well as the ability to undergo multiple sequential steps in the inositol lipid synthesis cascade. These data suggest that many of the chemical signaling events that are induced by integrins and growth factor receptors in capillary cells may effectively function in a "solid-state" on insoluble CSK scaffolds within the FAC and that the FAC may represent a major site for signal integration between these two regulatory pathways. Future investigations into the biochemical and biophysical basis of signal transduction may be facilitated by this method, which results in isolation of FACs that retain the CSK framework as well as multiple associated chemical signaling activities.


Asunto(s)
Adhesión Celular/fisiología , Proteínas del Citoesqueleto/análisis , Integrinas/análisis , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Transducción de Señal/fisiología , Corteza Suprarrenal , Animales , Bovinos , Moléculas de Adhesión Celular/análisis , Membrana Celular/química , Endotelio Vascular , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Integrinas/metabolismo , Isoenzimas/análisis , Microesferas , Oligopéptidos/síntesis química , Oligopéptidos/metabolismo , Fosfatidilinositol 3-Quinasas , Fosfolipasa C gamma , Fosfotransferasas (Aceptor de Grupo Alcohol)/análisis , Fosfotirosina/análisis , Proteínas Tirosina Quinasas/análisis , Proteínas Proto-Oncogénicas pp60(c-src)/análisis , Receptores Inmunológicos/metabolismo , Intercambiadores de Sodio-Hidrógeno/análisis , Fosfolipasas de Tipo C/análisis
11.
Biochim Biophys Acta ; 1668(2): 240-7, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15737335

RESUMEN

Receptor Tyrosine Kinases (RTKs) conduct biochemical signals via lateral dimerization in the plasma membrane, and defects in their dimerization lead to unregulated signaling and disease. RTK transmembrane (TM) domains are proposed to play an important role in the process, underscored by the finding that single amino acids mutations in the TM domains can induce pathological phenotypes. Therefore, many important questions pertaining to the mode of signal transduction and the mechanism of pathology induction could be answered by studying the chemical-physical basis behind RTK TM domain dimerization and the interactions of RTK TM domains with lipids in model bilayer systems. As a first step towards this goal, here we report the synthesis of the TM domain of fibroblast growth factor receptor 3 (FGFR3), an RTK that is crucial for skeletal development. We have used solid phase peptide synthesis to produce two peptides: one corresponding to the membrane embedded segment and the naturally occurring flanking residues at the N- and C-termini (TMwt), and a second one in which the flanking residues have been substituted with diLysines at the termini (TMKK). We have demonstrated that the hydrophobic FGFR3 TM domain can be synthesized for biophysical studies with high yield. The protocol presented in the paper can be applied to the synthesis of other RTK TM domains. As expected, the Lys flanks decrease the hydrophobicity of the TM domain, such that TMKK elutes much earlier than TMwt during reverse phase HPLC purification. The Lysines have no effect on peptide solubility in SDS and on peptide secondary structure, but they abolish peptide dimerization on SDS gels. These results suggest that caution should be exercised when modifying RTK TM domains to render them more manageable for biophysical studies.


Asunto(s)
Lisina/química , Proteínas de la Membrana/química , Proteínas Tirosina Quinasas/análisis , Proteínas Tirosina Quinasas/química , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dimerización , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Relación Estructura-Actividad
12.
J Natl Cancer Inst ; 88(19): 1399-404, 1996 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-8827018

RESUMEN

BACKGROUND: Radiation at doses high enough to cure cancer also frequently destroys normal tissue. Development of agents that protect normal tissue without also protecting diseased tissue has been difficult. In vivo radioprotection of bone marrow by acidic and basic fibroblast growth factors (FGF1 and FGF2, respectively) has recently been demonstrated after whole-body irradiation of C3H/HeN mice. PURPOSE: Our purpose was to determine whether myeloprotective doses of those growth factors also protect malignant tumors. METHODS: First, we investigated the effects of exogenous FGF1 or FGF2 (FGF1/2) administration (treatment group receiving two intravenous injections of 3 micrograms FGF1/2 per mouse 24 hours and 4 hours before local irradiation of right hind leg and control group receiving two intravenous injections of 0.1 mL of saline) on growth and radiosensitivity of three transplantable murine tumors (one squamous cell carcinoma [SCC-VII] and two sarcomas [KHT and Rif-1]), all of which were grown in C3H/HeN mice. We then evaluated the effect of FGF1/2 on tumor cell proliferation, cell cycle distribution, and pulmonary metastatic frequency in the mice. Specifically, survival studies were performed in mice treated with 0, 6, 6.5, 7.5, 8.5, 9, or 10 Gy whole-body irradiation with or without FGF2 (n = 250). Rif-1 (n = 40), KHT (n = 40), and SCC-VII (n = 40) tumors were implanted in the hind leg of mice, and mice were treated with FGF2 or saline when their tumor-bearing thighs were 9 mm in diameter. In separate experiments (treatment group receiving two injections of 3 micrograms each of FGF2 [6 micrograms total] either intravenously or intratumorally 24 hours and 4 hours before local tumor irradiation and control group receiving 0.1 mL saline), tumor growth was followed, and mice were killed to count lung metastases and measure tumor proliferating cell nuclear antigen (PCNA) and bromodeoxyuridine labeling at various times thereafter (three to eight mice per group). Tumor growth curves of untreated and irradiated tumors were determined with and without intravenous or intratumoral FGF1/2 in SCC-VII tumors (n = 120). Radiation doses to the tumor-bearing leg were 15 and 30 Gy for SCC-VII, 30 Gy for Rif-1, and 15 Gy for KHT. From each experiment, the mean (+/- 1 standard error) was calculated from data obtained from three to 20 mice. Statistical tests used included two-tailed Student's t test, the chi-squared test, and Fisher's exact test. All P values represent two-tailed tests of statistical significance. RESULTS: There was no statistically significant difference in tumor growth rate between FGF2-treated and saline-treated mice when FGF2 was administered intravenously at doses and schedules found to be optimally myeloprotective in whole-body irradiation experiments. Intravenous administration of FGF2 did not induce lung metastases, and it did not augment the S-phase fraction of tumor cells. Likewise, there was no evidence of enhanced cell proliferation as measured by PCNA-labeling index. Intratumoral injection of FGF1/2 did increase the size of SCC-VII tumors (P < .05 [Student's t test] at 3 days after treatment); however, the radiation response after intratumoral injection of growth factor was not compromised. CONCLUSION: Low intravenous doses of FGF1 or FGF2 appear to protect bone marrow from the toxic effects of radiation without increasing the rates of tumor growth or metastases or decreasing the radiosensitivity of tumors.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Factor 2 de Crecimiento de Fibroblastos/farmacología , Protectores contra Radiación/farmacología , Sarcoma Experimental/radioterapia , Animales , Carcinoma de Células Escamosas/secundario , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Inmunohistoquímica/métodos , Inyecciones Intravenosas , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Antígeno Nuclear de Célula en Proliferación/análisis , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Proteínas Recombinantes/farmacología , Sarcoma Experimental/secundario , Factores de Tiempo
13.
Cancer Res ; 54(19): 5206-11, 1994 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-7923141

RESUMEN

Although several tyrosine kinase-type growth factor receptors have been demonstrated in human colonic epithelial cells, the full spectrum of growth factor receptors has not been identified. Low stringency screening of a complementary DNA library prepared from the human colon cancer-derived cell line HT-29 with a probe containing the tyrosine kinase domain of human c-src kinase led to the identification and isolation of a clone containing a receptor class tyrosine kinase. This putative receptor was found to be identical to the human fibroblast growth factor receptor 3 (FGFR3) except for a region of 150 nucleotides (50 amino acids) encoding the presumptive ligand-binding domain, where it exhibited only 32% homology with the previously described FGFR3. The variant domain corresponded precisely to the splicing junctions of the exon encoding the carboxyl terminal half of the third immunoglobulin-like domain, suggesting that two forms of FGFR3 result from splicing of alternate exons in a manner similar to that previously found for FGFR1 and FGFR2. By prior convention, the previously reported from of FGFR3 was designated IIIc due to its high degree of homology with the IIIc domain of FGFR1 (83% homology) and the IIIc domain of FGFR2 (81% homology). However, the ligand-binding domain of FGFR3 found in the HT-29 cell line was more highly divergent from all previously reported FGFR immunoglobulin-like domain IIIs than any other two members of this receptor family. Therefore, we propose to designate the newly reported form as the FGFR3 IIIb variant. Genomic polymerase chain reaction confirmed that the IIIb-containing exon occupies a position 5' relative to the IIIc-containing exon within the FGFR3 gene. Northern blot analysis using a probe encompassing sequences unique to the FGFR3 IIIb mRNA confirmed the expression of a 4.4-kilobase transcript in two colon cancer-derived cell lines as well as normal human colonic mucosa. Using a technique combining reverse transcriptase polymerase chain reaction with restriction endonuclease digestion, cell lines, primary cells, and tissues were assessed for IIIb and IIIc transcripts; expression of the IIIb variant was associated with an epithelial lineage, while the IIIc variant was expressed predominantly in nonepithelial cells and tissues.


Asunto(s)
Colon/química , Proteínas Tirosina Quinasas , Receptores de Factores de Crecimiento de Fibroblastos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Epitelio/química , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Ratas , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
14.
Cancer Res ; 54(10): 2794-9, 1994 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-8168112

RESUMEN

Glioblastomas were examined for abnormalities in fibroblast growth factor receptor (FGFR) expression by polymerase chain reaction and immunocytochemical analysis. Polymerase chain reaction analysis demonstrated that FGFR1 mRNA levels were significantly higher in glioblastomas than in normal brain adjacent to the tumor or in untransformed human brain. These results were consistent with immunocytochemical localization of FGFR1 protein in glioblastomas: glioblastoma cells exhibited intense FGFR1 immunoreactivity in frozen sections of tumor and low to undetectable FGFR1 immunoreactivity in adjacent normal brain or in normal white matter obtained from patients without neoplastic disease. Endothelial cells of capillaries and larger vessels within the tumor were devoid of FGFR1 immunoreactivity. All glioblastomas evaluated in the present study expressed FGFR1 mRNA and FGFR1 immunoreactivity. Examination of the FGFR1 gene by Southern blot analysis indicated that overexpression of FGFR1 mRNA in glioblastomas did not result from gene amplification. These results indicate that glioblastoma cells, in contrast to endothelial cells within the tumor, display increased levels of FGFR1. Therefore, FGFR1 signal transduction may be associated with increased autocrine growth activity of tumor cells and is probably not related to the increased endothelial cell proliferation associated with these tumors.


Asunto(s)
Química Encefálica , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/química , Glioblastoma/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Neoplásico/análisis , Receptores de Factores de Crecimiento de Fibroblastos/análisis
15.
Cancer Res ; 54(2): 518-22, 1994 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8275490

RESUMEN

Previously, we identified an amplified gene in a stomach cancer cell line, KATO-III, and designated it K-sam. This gene was later found to be identical with a gene for a receptor tyrosine kinase, bek/FGFR2. One of the characteristics of the K-sam gene is structural diversity of its transcripts; K-sam complementary DNA (cDNA) cloned from human brain (K-sam-I) has a completely different sequence at the third extracellular immunoglobulin-like domain as compared to that of the K-sam cDNA derived from KATO-III cells (K-sam-II). Recent study has revealed that this difference signifies a differential ligand affinity; the receptor encoded by the K-sam-I cDNA has a high affinity for basic fibroblast growth factor (bFGF), while the K-sam-II cDNA corresponds to a receptor with the high affinity for keratinocyte growth factor (KGF). Reverse transcription-polymerase chain reaction and RNA blot analysis showed that the K-sam-II-type transcript was present in carcinoma cell lines but not in any of the sarcoma cell lines examined. The K-sam-I-type transcript was expressed in both carcinoma and sarcoma cell lines. Furthermore, KGF enhanced the DNA synthesis of the esophageal cancer cells, TE-1, in a dose-dependent manner, while the effect of bFGF was not substantial. In contrast, the glioblastoma cell line, A-172, that expressed the bFGF receptor showed a mitogenic response to bFGF but not to KGF. These data suggest that KGF is a growth factor used preferentially in cancer cells, and this preference is based on the presence of the K-sam-II-type receptor in carcinoma cells but not in sarcoma cells due to alternative splicing.


Asunto(s)
Carcinoma/genética , ADN de Neoplasias/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos , Sustancias de Crecimiento/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Sarcoma/genética , Teratoma/genética , Secuencia de Bases , Carcinoma/química , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Sondas ARN , Sarcoma/química , Teratoma/química , Células Tumorales Cultivadas
16.
Cancer Res ; 54(2): 523-30, 1994 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-7506125

RESUMEN

Overexpression of some transmembrane tyrosine kinase growth factor receptors in breast and other tumors has been found to correlate with poor prognosis. Following the cloning of the first two members of the fibroblast growth factor family of receptors (FGFRs), amplification of these receptors in breast carcinomas was found. We have examined 23 breast carcinoma cell lines to determine the extent of expression of mRNA for fgfrs 1 through 4. All breast carcinoma cell lines examined expressed mRNA for at least one fgfr and several expressed high mRNA levels for a particular receptor. MDA-MB-134, an estrogen receptor-positive cell line, expressed very high levels of mRNA for fgfr-1 and elevated levels of mRNA for fgfr-4. This cell line was found to have an amplified fgfr-1 gene, but the gene for fgfr-4 was not amplified. MDA-MB-453 cells were found to express high levels of mRNA for fgfr-4 without amplification of the gene. MDA-MB-134 cells were examined for their response to FGF ligands. Tyrosine phosphorylation of a M(r) 150,000 protein resulted when MDA-MB-134 cells were treated with FGF-1 or FGF-2, implying the presence of a functional FGFR-1. MDA-MB-134 cells were growth-inhibited by picomolar concentrations of FGF-1 or FGF-2 in a dose-dependent manner under both anchorage-independent and anchorage-dependent conditions. These results may provide insight into the consequences of FGFR overexpression in breast tumors and the development of treatment modalities which use manipulation of growth factor responses.


Asunto(s)
Neoplasias de la Mama/química , ARN Mensajero/análisis , ARN Neoplásico/análisis , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Anticuerpos/análisis , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Fosforilación , Fosfotirosina , Células Tumorales Cultivadas , Tirosina/análogos & derivados , Tirosina/inmunología
17.
Cancer Res ; 54(24): 6571-7, 1994 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-7987857

RESUMEN

Angiogenesis of human melanomas has been the focus of intense interest since it was shown that the spread and prognosis of primary tumors is correlated with their vascularization (N. Weidner, J. P. Semple, W. R. Welch, and J. Folkman, N. Engl. J. Med., 324: 1-8, 1991). Basic fibroblast growth factor (bFGF) and its high-affinity receptor FGFR-1 have been implicated in melanoma growth and angiogenesis (R. Halaban, Y. Funasaka, J. Lee, J. Rubin, D. Ron, and D. Birnbaum, Fibroblast Growth Factors in Normal and Malignant Melanocytes, pp. 232-243. New York: The New York Academy of Sciences, 1991). We have studied the expression of the Tie endothelial cell receptor tyrosine kinase mRNA in skin and primary cutaneous melanomas as well as in their skin and brain metastases by in situ hybridization. The Tie probe hybridized very weakly with the vascular endothelium of capillaries of normal skin, while it was detected in larger arteries and veins as well as in capillaries around sweat glands. However, capillaries and medium-sized vessels within cutaneous and brain metastases of melanoma were strongly positive for Tie mRNA. In contrast, endothelial cells contained very little or no FGFR-1 transcripts, whereas abundant FGFR-1 mRNA was present in melanoma tumor cells and in fibrovascular stroma. In agreement with these findings, a Tie-specific amplified cDNA band was obtained by reverse transcription-polymerase chain reaction from melanoma metastases but not from normal skin. These results suggest a role for the Tie receptor in the angiogenesis associated with melanoma metastases.


Asunto(s)
Endotelio Vascular/química , Melanoma/química , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/análisis , Receptores de Superficie Celular/análisis , Neoplasias Cutáneas/química , Piel/química , Secuencia de Bases , Neoplasias Encefálicas/química , Neoplasias Encefálicas/secundario , Humanos , Hibridación in Situ , Melanoma/secundario , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Receptores TIE
18.
Cancer Res ; 53(20): 4741-4, 1993 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8402651

RESUMEN

Acidic and basic fibroblast growth factors are mitogenic polypeptides that are overexpressed in pancreatic cancer. To determine whether fibroblast growth factors may exert direct effects on pancreatic cancer cells in vivo, we compared the expression of the high-affinity type I fibroblast growth factor receptor (FGFR-1) in human pancreatic tissues. In the normal pancreas, FGFR-1 immunostaining was seen mainly in acinar cells. In pancreatic cancers, FGFR-1 was abundant in ductal-like cancer cells which also exhibited many FGFR-1 mRNA in situ hybridization grains. Analysis by the polymerase chain reaction and RNase protection revealed that the 2-immunoglobulin-like and the 3-immunoglobulin-like forms of FGFR-1 were expressed in all tissue samples, and that the 2-immunoglobulin-like form was overexpressed in the cancer tissues by comparison with the normal tissues. These findings suggest that the 2-immunoglobulin-like form of FGFR-1 may contribute to aberrant autocrine and paracrine pathways in pancreatic cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Adenocarcinoma/patología , Anticuerpos Monoclonales , Secuencia de Bases , Southern Blotting , ADN/análisis , Cartilla de ADN , ADN de Neoplasias/análisis , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hibridación in Situ , Datos de Secuencia Molecular , Páncreas/citología , Neoplasias Pancreáticas/patología , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis
19.
PLoS One ; 11(7): e0159263, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27414797

RESUMEN

We recently developed novel bioluminescent binding assays for several protein/peptide hormones to study their interactions with receptors using the so far brightest NanoLuc reporter. To validate the novel bioluminescent binding assay using a variety of protein/peptide hormones, in the present work we applied it to the fibroblast growth factor (FGF) family using the prototype member FGF2 as an example. A fully active recombinant FGF2 retaining a unique exposed cysteine (Cys) residue was chemically conjugated with an engineered NanoLuc carrying a unique exposed Cys residue at the C-terminus via formation of an intermolecular disulfide linkage. The NanoLuc-conjugated FGF2 (FGF2-Luc) retained high binding affinity to the overexpressed FGFR1 and the endogenous FGF receptor with the calculated dissociation constants of 161 ± 21 pM (n = 3) and 25 ± 4 pM (n = 3), respectively. In competition binding assays using FGF2-Luc as a tracer, receptor-binding potencies of wild-type or mutant FGF2s were accurately quantified. Thus, FGF2-Luc represents a novel non-radioactive tracer for the quantitative measurement of ligand-receptor interactions in the FGF family. These data suggest that the novel bioluminescent binding assay can be applied to a variety of protein/peptide hormones for ligand-receptor interaction studies.


Asunto(s)
Mediciones Luminiscentes/métodos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/análisis , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Ligandos , Unión Proteica , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Proteínas Recombinantes
20.
Placenta ; 41: 53-61, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27208408

RESUMEN

INTRODUCTION: α-klotho is an anti-aging protein, potentially important in preeclampsia (PE). Produced by kidney, brain and placenta, and by mRNA splicing is both a full-length membrane-bound and a truncated soluble protein in the circulation. The membrane-bound protein is an obligate co-receptor for fibroblast growth factor 23 (FGF23) and its action on receptor (FGFR), but ADAM proteinases also cause its shedding. The aims of this study were to investigate levels of maternal plasma, placental, and fetal membrane α-Klotho and their association with placental accelerated villous maturation (AVM) in PE. In addition, placental and membrane levels of ADAM17 and FGFR were measured in the same patients. METHODS: Maternal blood, placenta and fetal membranes from 61 women (31 with PE and 30 controls) between 32 and 40 weeks gestation were collected. Plasma α-klotho was measured by ELISA, and quantitative immunohistochemistry used for α-klotho, ADAM17 and FGFR1 in tissues. Placental AVM was histologically assessed. RESULTS: Maternal plasma levels of α-Klotho were higher in PE compared to controls (p = 0.01) and patients with the highest levels had significantly less AVM (p = 0.03). α-Klotho, ADAM17, and FGFR were all present in syncytiotrophoblast and cytotrophoblast of membranes. Between 32 and 40 weeks gestation, all placental levels decreased in controls respectively (p = 0.04, p = 0.004, p = 0.05), but not in PE. Fetal membrane levels were unchanged. DISCUSSION: Maternal plasma α-Klotho was increased in PE and its levels associated with reduced placental AVM. Changes in placental α-Klotho, ADAM17, and FGFR suggest their involvement in the pathophysiology of PE.


Asunto(s)
Edad Gestacional , Glucuronidasa/análisis , Placenta/química , Preeclampsia/fisiopatología , Proteína ADAM17/análisis , Adulto , Membranas Extraembrionarias/química , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/sangre , Humanos , Proteínas Klotho , Embarazo , Tercer Trimestre del Embarazo , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Trofoblastos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA