Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 131(12): 1004-1017, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36321446

RESUMEN

BACKGROUND: Genome-wide association studies have discovered a link between genetic variants on human chromosome 15q26.1 and increased coronary artery disease (CAD) susceptibility; however, the underlying pathobiological mechanism is unclear. This genetic locus contains the FES (FES proto-oncogene, tyrosine kinase) gene encoding a cytoplasmic protein-tyrosine kinase involved in the regulation of cell behavior. We investigated the effect of the 15q26.1 variants on FES expression and whether FES plays a role in atherosclerosis. METHODS AND RESULTS: Analyses of isogenic monocytic cell lines generated by CRISPR (clustered regularly interspaced short palindromic repeats)-mediated genome editing showed that monocytes with an engineered 15q26.1 CAD risk genotype had reduced FES expression. Small-interfering-RNA-mediated knockdown of FES promoted migration of monocytes and vascular smooth muscle cells. A phosphoproteomics analysis showed that FES knockdown altered phosphorylation of a number of proteins known to regulate cell migration. Single-cell RNA-sequencing revealed that in human atherosclerotic plaques, cells that expressed FES were predominately monocytes/macrophages, although several other cell types including smooth muscle cells also expressed FES. There was an association between the 15q26.1 CAD risk genotype and greater numbers of monocytes/macrophage in human atherosclerotic plaques. An animal model study demonstrated that Fes knockout increased atherosclerotic plaque size and within-plaque content of monocytes/macrophages and smooth muscle cells, in apolipoprotein E-deficient mice fed a high fat diet. CONCLUSIONS: We provide substantial evidence that the CAD risk variants at the 15q26.1 locus reduce FES expression in monocytes and that FES depletion results in larger atherosclerotic plaques with more monocytes/macrophages and smooth muscle cells. This study is the first demonstration that FES plays a protective role against atherosclerosis and suggests that enhancing FES activity could be a potentially novel therapeutic approach for CAD intervention.


Asunto(s)
Aterosclerosis , Enfermedad de la Arteria Coronaria , Placa Aterosclerótica , Proteínas Proto-Oncogénicas c-fes , Animales , Humanos , Ratones , Arterias/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Estudio de Asociación del Genoma Completo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Proto-Oncogénicas c-fes/metabolismo
2.
J Gastroenterol Hepatol ; 34(10): 1869-1877, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31038805

RESUMEN

BACKGROUND AND AIM: DNA hypermethylation has emerged as a novel molecular biomarker for the diagnosis and prognosis prediction of many cancers. We aimed to identify clinically useful biomarkers regulated by DNA methylation in hepatocellular carcinoma (HCC). METHODS: Genome-wide methylation analysis in HCCs and paired noncancerous tissues was performed using an Illumina Infinium HumanMethylation 450K BeadChip array. Methylation-specific polymerase chain reaction and pyrosequencing were used to validate the methylation status of selected genes in 100 paired HCCs and noncancerous samples. RESULTS: A total of 97 027 (20.0%) out of 485 577 CpG sites significantly were differed between HCC and noncancerous tissues. Among all the significant CpG sites, 48.8% are hypermethylated and 51.2% are hypomethylated in HCCs. Multiple signaling pathways (AMP-activated protein kinase, estrogen, and adipocytokine) involved in gene methylation were identified in HCC. FES was selected for further analysis based on its high level of methylation confirmed by polymerase chain reaction and pyrosequencing. The result showed that FES hypermethylation was correlated with tumor size (0.001), serum alpha fetoprotein (0.023), and tumor differentiation (0.006). FES protein was significantly downregulated in 51/100 (51%) HCCs, and 94.12% (48/51) of them were due to promoter hypermethylation. Both FES hypermethylation and protein downregulation were associated with the progression-free survival and overall survival of HCC patients. Overexpressed and knockdown of FES confirmed its inhibitory effect on the proliferation and migration of HCC cells. CONCLUSIONS: We identified many new differentially methylated CpGs in HCCs and demonstrate that FES functions as a tumor suppressor gene in HCC and its methylation status could be used as an indicator for prognosis of HCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Metilación de ADN , Epigénesis Genética , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-fes/genética , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Islas de CpG , Detección Precoz del Cáncer/métodos , Femenino , Predisposición Genética a la Enfermedad , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular , Fenotipo , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fes/metabolismo , Factores de Tiempo , Carga Tumoral , alfa-Fetoproteínas/análisis
4.
Wei Sheng Wu Xue Bao ; 53(3): 299-305, 2013 Mar 04.
Artículo en Zh | MEDLINE | ID: mdl-23678577

RESUMEN

OBJECTIVE: To prepare anti-fps mono-specific serum, and detect the fps antigen in tumors induced by acute transforming avian leukosis/sarcoma virus containing v-fps oncogene. METHODS: Two part of v-fps gene was amplified by RT-PCR using the Fu-J viral RNA as the template. Mono-specific serum was prepared by immuning Kunming white mouse with both two recombinant infusion proteins expressed by the prokaryotic expression system. Indirect immunofluorescent assay was used to detect fps antigen in tumor tissue suspension cells and CEF infected by sarcoma supernatant. Immunohistochemical method was used to detect fps antigen in tumor tissue. RESULTS: The mouse mono-specific serum was specific as it had no cross reaction with classical ALV-J strains. The result reveals that the tumor tissue suspension cells, the CEF infected by sarcoma supernatant, and the slice immunohistochemistry of the sarcoma showed positive results. CONCLUSION: The anti-fps mono-specific serum was prepared, and the detection method was established, which laid the foundation for the study of viral biological characteristics and mechanism of tumourgenesis of acute transforming avian leukosis/sarcoma virus containing v-fps oncogene.


Asunto(s)
Virus de la Leucosis Aviar/inmunología , Virus del Sarcoma Aviar/inmunología , Pollos , Fibrosarcoma/inmunología , Enfermedades de las Aves de Corral/inmunología , Proteínas Proto-Oncogénicas c-fes/inmunología , Animales , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Leucosis Aviar/inmunología , Leucosis Aviar/virología , Transformación Celular Neoplásica , Fibrosarcoma/virología , Ratones , Enfermedades de las Aves de Corral/virología , Proteínas Proto-Oncogénicas c-fes/genética , ARN Viral/genética , Sarcoma Aviar/inmunología , Sarcoma Aviar/virología , Organismos Libres de Patógenos Específicos
5.
Prostate ; 72(2): 201-8, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21563194

RESUMEN

BACKGROUND: c-Fes is a proto-oncogene encoded non-receptor protein-tyrosine kinase (PTK). However, genetic studies have indicated that it has anti-tumorigenic effects in certain cancers. The pathological and clinical significance of c-Fes in prostate cancer are unknown. METHODS: Expression of c-Fes was evaluated in normal glands, prostatic intraepithelial neoplasia (PIN), cancer cells in tissues of knock-in mouse adenocarcinoma prostate (KIMAP) model, and prostate cancer patients free of metastasis. Expression of c-Fes was analyzed by immunohistochemistry, and quantified by using the immunoreactivity score (IRS) (staining intensity × percentage of positive cells). Relationships between c-Fes expression and pT stage, Gleason's score (GS), and biochemical recurrence in patients who underwent radical surgery were also investigated. RESULTS: In KIMAP, the percentage in normal glands, PIN and cancer cells positive for c-Fes expression were 0 (0/7), 25.0 (2/8), and 100% (7/7), respectively. In human tissues, c-Fes expression was also significantly higher in cancer cells than in normal cells and PIN, and it correlated with pT stage (P < 0.001) and GS (P = 0.047). Multivariate analysis showed that c-Fes expression was an independent predictor of poor outcome poor prognosis (hazard ratio = 3.21, 95% confidence interval = 1.11-9.37, P = 0.032). CONCLUSION: The results suggested that c-Fes expression is a useful predictor of biochemical recurrence after radical surgery. The results also suggested that c-Fes is a potentially useful therapeutic target in prostate cancer and a predictor of biochemical recurrence after radical prostatectomy.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Recurrencia Local de Neoplasia/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fes/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/cirugía , Animales , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Transgénicos , Recurrencia Local de Neoplasia/metabolismo , Valor Predictivo de las Pruebas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/cirugía , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fes/genética , Estudios Retrospectivos
6.
Mol Ther ; 19(1): 122-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20978475

RESUMEN

X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene encoding the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase catalytic subunit gp91(phox). A recent clinical trial for X-CGD using a spleen focus-forming virus (SFFV)-based γ-retroviral vector has demonstrated clear therapeutic benefits in several patients although complicated by enhancer-mediated mutagenesis and diminution of effectiveness over time due to silencing of the viral long terminal repeat (LTR). To improve safety and efficacy, we have designed a lentiviral vector that directs transgene expression primarily in myeloid cells. To this end, we created a synthetic chimeric promoter that contains binding sites for myeloid transcription factors CAAT box enhancer-binding family proteins (C/EBPs) and PU.1, which are highly expressed during granulocytic differentiation. As predicted, the chimeric promoter regulated higher reporter gene expression in myeloid than in nonmyeloid cells, and in human hematopoietic progenitors upon granulocytic differentiation. In a murine model of stem cell gene therapy for X-CGD, the chimeric vector resulted in high levels of gp91(phox) expression in committed myeloid cells and granulocytes, and restored normal NADPH-oxidase activity. These findings were recapitulated in human neutrophils derived from transduced X-CGD CD34(+) cells in vivo, and suggest that the chimeric promoter will have utility for gene therapy of myeloid lineage disorders such as CGD.


Asunto(s)
Catepsina G/genética , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/terapia , Células Mieloides/fisiología , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Recombinantes de Fusión/genética , Transgenes , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Genes Ligados a X , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Granulocitos/metabolismo , Enfermedad Granulomatosa Crónica/enzimología , Enfermedad Granulomatosa Crónica/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis/genética , Células Mieloides/metabolismo , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Virus Formadores de Foco en el Bazo/genética , Virus Formadores de Foco en el Bazo/metabolismo , Células Madre/metabolismo , Secuencias Repetidas Terminales , Transactivadores/metabolismo
7.
Biochem Biophys Res Commun ; 393(1): 174-8, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20117079

RESUMEN

FES is a cytoplasmic tyrosine kinase activated by several membrane receptors, originally identified as a viral oncogene product. We have recently identified FES as a crucial effector of oncogenic KIT mutant receptor. However, FES implication in wild-type KIT receptor function was not addressed. We report here that FES interacts with KIT and is phosphorylated following activation by its ligand SCF. Unlike in the context of oncogenic KIT mutant, FES is not involved in wild-type KIT proliferation signal, or in cell adhesion. Instead, FES is required for SCF-induced chemotaxis. In conclusion, FES kinase is a mediator of wild-type KIT signalling implicated in cell migration.


Asunto(s)
Quimiotaxis , Proteínas Proto-Oncogénicas c-fes/metabolismo , Factor de Células Madre/metabolismo , Dominios Homologos src , Adhesión Celular , Línea Celular Tumoral , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-fes/genética , Técnicas del Sistema de Dos Híbridos , Tirosina
8.
Exp Cell Res ; 315(17): 2929-40, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19732771

RESUMEN

The fps/fes proto-oncogene encodes a cytoplasmic protein-tyrosine kinase implicated in vesicular trafficking and cytokine and growth factor signaling in hematopoietic, neuronal, vascular endothelial and epithelial lineages. Genetic evidence has suggested a tumor suppressor role for Fps/Fes in breast and colon. Here we used fps/fes knockout mice to investigate potential roles for this kinase in development and function of the mammary gland. Fps/Fes expression was induced during pregnancy and lactation, and its kinase activity was dramatically enhanced. Milk protein and fat composition from nursing fps/fes-null mothers was normal; however, pups reared by them gained weight more slowly than pups reared by wild-type mothers. Fps/Fes displayed a predominantly dispersed punctate intracellular distribution which was consistent with vesicles within the luminal epithelial cells of lactating breast, while a small fraction co-localized with beta-catenin and E-cadherin on their basolateral surfaces. Fps/Fes was found to be a component of the E-cadherin adherens junction (AJ) complex; however, the phosphotyrosine status of beta-catenin and core AJ components in fps/fes-null breast tissue was unaltered, and epithelial cell AJs and gland morphology were intact. We conclude that Fps/Fes is not essential for the maintenance of epithelial cell AJs in the lactating breast but may instead play important roles in vesicular trafficking and milk secretion.


Asunto(s)
Cadherinas/fisiología , Células Epiteliales/fisiología , Lactancia/genética , Glándulas Mamarias Animales/fisiología , Proteínas Proto-Oncogénicas c-fes/deficiencia , Animales , Células Epiteliales/citología , Femenino , Immunoblotting , Glándulas Mamarias Animales/citología , Ratones , Ratones Noqueados , Leche/metabolismo , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Embarazo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fes/genética
9.
Genes Chromosomes Cancer ; 48(3): 272-84, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19051325

RESUMEN

The FES locus encodes a unique nonreceptor protein-tyrosine kinase (FES) traditionally viewed as a proto-oncogene but more recently implicated as a tumor suppressor in colorectal cancer (CRC). Recent studies have demonstrated that while FES is expressed in normal colonic epithelium, expression is lost in tumor tissue and colorectal cancer cell lines, a finding common among tumor suppressors. Here we provide compelling evidence that promoter methylation is an important mechanism responsible for downregulation of FES gene expression in colorectal cancer cells. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine resulted in the expression of functional FES transcripts in all CRC cell lines examined, including Caco-2, COLO 320, DLD-1, HCT 116, SNU-1040, SW-480, and HT-29. Bisulfite sequencing of genomic DNA isolated from 5-aza-2'-deoxycytidine-treated HT-29 cells identified methylated CpG dinucleotides immediately upstream from the FES transcription initiation sites. In contrast, this region of the FES promoter was hypomethylated in genomic DNA from normal colonic epithelium. In addition, methylation completely blocked the activity of the FES promoter in reporter gene assays. Promoter methylation is a previously unrecognized mechanism by which FES expression is suppressed in CRC cell lines, and is consistent with a tumor suppressor role for FES in this tumor site despite its tyrosine kinase activity.


Asunto(s)
Neoplasias Colorrectales/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fes/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Islas de CpG/fisiología , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación hacia Abajo , Humanos , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fes/metabolismo
10.
Nat Commun ; 11(1): 3216, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587248

RESUMEN

Chemical tools to monitor drug-target engagement of endogenously expressed protein kinases are highly desirable for preclinical target validation in drug discovery. Here, we describe a chemical genetics strategy to selectively study target engagement of endogenous kinases. By substituting a serine residue into cysteine at the DFG-1 position in the ATP-binding pocket, we sensitize the non-receptor tyrosine kinase FES towards covalent labeling by a complementary fluorescent chemical probe. This mutation is introduced in the endogenous FES gene of HL-60 cells using CRISPR/Cas9 gene editing. Leveraging the temporal and acute control offered by our strategy, we show that FES activity is dispensable for differentiation of HL-60 cells towards macrophages. Instead, FES plays a key role in neutrophil phagocytosis via SYK kinase activation. This chemical genetics strategy holds promise as a target validation method for kinases.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes , Proteínas Proto-Oncogénicas c-fes , Transportadoras de Casetes de Unión a ATP/química , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Edición Génica , Humanos , Macrófagos/metabolismo , Mutación , Neutrófilos , Fagocitosis , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fes/química , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Proto-Oncogénicas c-fes/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo
11.
Clin Cancer Res ; 26(1): 265-273, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573955

RESUMEN

PURPOSE: The identification of novel targets for developing synergistic drug-radiation combinations would pave the way to overcome tumor radioresistance. We conducted cell-based screening of a human kinome siRNA library to identify a radiation-specific kinase that has a synergistic toxic effect with radiation upon inhibition and is not essential for cell survival in the absence of radiation. EXPERIMENTAL DESIGN: Unbiased RNAi screening was performed by transfecting A549 cells with a human kinome siRNA library followed by irradiation. Radiosensitizing effects of a target gene and involved mechanisms were examined. RESULTS: We identified the nonreceptor protein tyrosine kinase FES (FEline Sarcoma oncogene) as a radiosensitizing target. The expression of FES was increased in response to irradiation. Cell viability and clonogenic survival after irradiation were significantly decreased by FES knockdown in lung and pancreatic cancer cell lines. In contrast, FES depletion alone did not significantly affect cell proliferation without irradiation. An inducible RNAi mouse xenograft model verified in vivo radiosensitizing effects. FES-depleted cells showed increased apoptosis, DNA damage, G2-M phase arrest, and mitotic catastrophe after irradiation. FES depletion promoted radiation-induced reactive oxygen species formation, which resulted in phosphorylation of S6K and MDM2. The radiosensitizing effect of FES knockdown was partially reversed by inhibition of S6K activity. Consistent with the increase in phosphorylated MDM2, an increase in nuclear p53 levels was observed, which appears to contribute increased radiosensitivity of FES-depleted cells. CONCLUSIONS: We uncovered that inhibition of FES could be a potential strategy for inducing radiosensitization in cancer. Our results provide the basis for developing novel radiosensitizers.


Asunto(s)
Apoptosis , Daño del ADN , Neoplasias Pulmonares/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-fes/antagonistas & inhibidores , Interferencia de ARN , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Ratones , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/radioterapia , Fosforilación/efectos de la radiación , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Proto-Oncogénicas c-fes/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Tolerancia a Radiación , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biochemistry ; 48(22): 4780-8, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19382747

RESUMEN

The c-fes proto-oncogene encodes a unique nonreceptor protein-tyrosine kinase (c-Fes) that contributes to the differentiation of myeloid hematopoietic, vascular endothelial, and some neuronal cell types. Although originally identified as the normal cellular homologue of the oncoproteins encoded by avian and feline transforming retroviruses, c-Fes has recently been implicated as a tumor suppressor in breast and colonic epithelial cells. Structurally, c-Fes consists of a unique N-terminal region harboring an FCH domain, two coiled-coil motifs, a central SH2 domain, and a C-terminal kinase domain. In living cells, c-Fes kinase activity is tightly regulated by a mechanism that remains unclear. Previous studies have established that c-Fes forms high molecular weight oligomers in vitro, suggesting that the dual coiled-coil motifs may regulate the interconversion of inactive monomeric and active oligomeric states. Here we show for the first time that c-Fes forms oligomers in live cells independently of its activation status using a YFP bimolecular fluorescence complementation assay. We also demonstrate that both N-terminal coiled-coil regions are essential for c-Fes oligomerization in transfected COS-7 cells as well as HCT 116 colorectal cancer and K-562 myeloid leukemia cell lines. Together, these data provide the first evidence that c-Fes, unlike c-Src, c-Abl, and other nonreceptor tyrosine kinases, is constitutively oligomeric in both its repressed and active states. This finding suggests that conformational changes, rather than oligomerization, may govern its kinase activity in vivo.


Asunto(s)
Prueba de Complementación Genética , Proteínas Proto-Oncogénicas c-fes/biosíntesis , Proteínas Proto-Oncogénicas c-fes/genética , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Células COS , Chlorocebus aethiops , Activación Enzimática/genética , Silenciador del Gen , Células HCT116 , Humanos , Immunoblotting , Células K562 , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fes/química , Transducción de Señal/genética
13.
Int J Oncol ; 34(1): 89-96, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19082481

RESUMEN

The c-Fes protein-tyrosine kinase is associated with growth and differentiation of hematopoietic, neuronal, vascular endothelial and epithelial cell types. In this study, we investigated whether small interfering RNA (siRNA)-mediated knockdown of c-Fes expression affected proliferation of the human renal carcinoma cell lines, ACHN and VMRC-RCW. Immunofluorescence microscopy showed that c-Fes was expressed in both the cytosol and nuclei of these cells, and siRNA treatment preferentially downregulated c-Fes expression in the cytosol. Knock-down of c-Fes inhibited cellular proliferation in a dose-dependent manner with minimal increase in cell death. c-Fes siRNA treatment also downregulated the phosphorylation of Akt1 on S473 and IKKalpha on T23, and cyclin D1 expression, enhanced the expression of IkappaBalpha, and prevented the nuclear localization of NFkappaB. Treatment with an NFkappaB inhibitory peptide (SN50) also blocked the proliferation and nuclear localization of NFkappaB in these cells. The effect of SN50 treatment was not enhanced by c-Fes siRNA, suggesting that downregulation of c-Fes expression inhibited cell cycle progression through the Akt1/NFkappaB pathway. In contrast to siRNA-mediated knockdown, ectopic expression of either wild-type or kinase-inactive c-Fes in renal carcinoma cells failed to alter their proliferation in vitro and in vivo. Thus, suppression of proliferation resulting from siRNA-mediated knockdown may depend upon an expression of c-Fes protein rather than its kinase activity. Taken together, our results indicate that downregulation of c-Fes expression may be a potential therapeutic strategy for advanced human renal cell carcinoma and inhibition of its kinase activity as an antiangiogenic therapy does not seem to induce the growth of human renal carcinoma cells.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Proteínas Proto-Oncogénicas c-fes/metabolismo , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Núcleo Celular , Proliferación Celular , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Proteínas I-kappa B/metabolismo , Immunoblotting , Técnicas para Inmunoenzimas , Inmunoprecipitación , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fes/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fes/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal , Células Tumorales Cultivadas
15.
J Cancer Res Clin Oncol ; 144(1): 21-31, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28952025

RESUMEN

PURPOSE: The feline sarcoma oncogene protein (FES) is a non-receptor tyrosine kinase implicated in both oncogenesis and tumor suppression. Here, cancer cell lines and human tissues were employed to clarify the pathological and prognostic significance of FES in bladder cancer. METHODS: The relationship between FES expression and cancer aggressiveness was investigated using 3 cell lines (T24: corresponding to grade 3, 5637: corresponding to grade 2, and RT4: corresponding to grade 1) and 203 tissues derived from human bladder malignancies. Proliferation, invasion, and migration of cancer cells were assessed following the knockdown (KD) of FES expression by the siRNA method. Relationships between FES expression and pathological features, aggressiveness, and outcome were investigated. RESULTS: FES-KD inhibited the proliferation, migration, and invasion of T24 cells but not of RT4 cells and 5637 cells. Considering all patients, FES expression demonstrated a negative relationship with grade but no association with muscle invasion or cancer cell proliferation. However, it was positively correlated with pT stage and cell proliferation in high-grade tumors (p = 0.002); no such association was found for low-grade tumors. In addition, elevated FES expression was a negative prognostic indicator of metastasis after radical surgery for patients with high-grade tumors (p = 0.021) but not for those with low-grade malignancies. CONCLUSIONS: FES appeared to act as a suppressor of carcinogenesis, being associated with low tumor grade in the overall patient group. However, its expression correlated with cancer aggressiveness and poor outcome in high-grade bladder cancer. FES, therefore, represents a potential therapeutic target and useful prognostic factor for such patients.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteínas Proto-Oncogénicas c-fes/biosíntesis , Neoplasias de la Vejiga Urinaria/metabolismo , Biomarcadores de Tumor/genética , Western Blotting , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-fes/genética , Tasa de Supervivencia , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
16.
Biochem J ; 399(1): 141-50, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16792528

RESUMEN

The c-Fes protein tyrosine kinase is implicated in the differentiation of a number of cell types including neuronal, endothelial and myeloid cells. Structurally, Fes consists of a unique N-terminal region, followed by SH2 (Src homology domain 2) and kinase domains. Two coiled-coil (CC) domains (CC1 and CC2) located within the unique N-terminal region are critical regulators of Fes activity in vivo and may function to recruit Fes activators and/or substrates. A yeast two-hybrid screen, utilizing a K-562 cell cDNA library and the Fes CC2 domain as bait, identified an interacting clone encoding the CC domain and B-box motifs (residues 114-357) of the transcriptional co-repressor KRAB-associated protein (KAP)-1. KAP-1(114-357) interacted with full-length Fes in yeast, and the KAP-1 CC domain was sufficient to bind the Fes N-terminal region in Sf-9 cells. Co-expression of Fes with full-length KAP-1 in human 293T cells stimulated Fes autophosphorylation and led to KAP-1 tyrosine phosphorylation. Association of endogenous Fes and KAP-1 was also observed in HL-60 myeloid leukaemia cells. Together, these data identify a novel Fes-KAP-1 interaction, and suggest a dual role for KAP-1 as both a Fes activator and downstream effector.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Insectos , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Represoras/genética , Saccharomyces cerevisiae , Especificidad por Sustrato , Proteína 28 que Contiene Motivos Tripartito
17.
J Clin Invest ; 127(6): 2310-2325, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28463229

RESUMEN

Identification and functional validation of oncogenic drivers are essential steps toward advancing cancer precision medicine. Here, we have presented a comprehensive analysis of the somatic genomic landscape of the widely used BRAFV600E- and NRASQ61K-driven mouse models of melanoma. By integrating the data with publically available genomic, epigenomic, and transcriptomic information from human clinical samples, we confirmed the importance of several genes and pathways previously implicated in human melanoma, including the tumor-suppressor genes phosphatase and tensin homolog (PTEN), cyclin dependent kinase inhibitor 2A (CDKN2A), LKB1, and others. Importantly, this approach also identified additional putative melanoma drivers with prognostic and therapeutic relevance. Surprisingly, one of these genes encodes the tyrosine kinase FES. Whereas FES is highly expressed in normal human melanocytes, FES expression is strongly decreased in over 30% of human melanomas. This downregulation correlates with poor overall survival. Correspondingly, engineered deletion of Fes accelerated tumor progression in a BRAFV600E-driven mouse model of melanoma. Together, these data implicate FES as a driver of melanoma progression and demonstrate the potential of cross-species oncogenomic approaches combined with mouse modeling to uncover impactful mutations and oncogenic driver alleles with clinical importance in the treatment of human cancer.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas c-fes/genética , Neoplasias Cutáneas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Variaciones en el Número de Copia de ADN , Genes Supresores de Tumor , Genómica , Humanos , Melanoma/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Oncogenes , Proteínas Proto-Oncogénicas c-fes/metabolismo , Neoplasias Cutáneas/metabolismo , Vía de Señalización Wnt
18.
Gene ; 620: 1-9, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28389358

RESUMEN

MicroRNA-125b (miR-125b) has been reported to be upregulated in several kinds of leukemia, suggesting that miR-125b plays a role in Leukemia development. In this study, it was shown that miR-125b expression level decreased in response to 1α, 25-dihydroxy-vitamin D3 (1,25D3) in a dose- and time-dependent manner and miR-125b blocked 1,25D3-induced monocytic differentiation of U937 cells. In addition, miR-125b decreased mRNA expression of myelomonocytic differentiation markers, including CD11c, CD18 and CD64 and arrested the cell cycle at the S phase in U937 and HL60 cells. Fes was identified as a novel direct target of miR-125b and miR-125b could also reduce the expression levels of PU.1 and macrophage colony-stimulating factor receptor (MCSFR). Furthermore, Fes was found to be involved in monocytic differentiation via upregulation of PU.1 and MCSFR and Fes siRNA could also inhibit 1,25D3-induced monocytic differentiation of U937 and HL60 cells and decrease mRNA expression of CD11c, CD18 and CD64. Importantly, the inhibition of Fes siRNA on 1,25D3-induced monocytic differentiation could be rescued by transfection with miR-125b inhibitor. Our data highlights an important role of miR-125b in AML progression, implying the potential application of miR-125b in AML therapy.


Asunto(s)
Diferenciación Celular , Leucemia Mieloide Aguda/metabolismo , MicroARNs/genética , Proteínas Proto-Oncogénicas c-fes/genética , Regiones no Traducidas 3' , Antígenos CD/genética , Antígenos CD/metabolismo , Calcitriol/farmacología , Ciclo Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Leucemia Mieloide Aguda/genética , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
19.
Sci Rep ; 7(1): 7595, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790351

RESUMEN

Analysis of single-cell RNA-Seq data can provide insights into the specific functions of individual cell types that compose complex tissues. Here, we examined gene expression in two distinct subpopulations of mouse taste cells: Tas1r3-expressing type II cells and physiologically identified type III cells. Our RNA-Seq libraries met high quality control standards and accurately captured differential expression of marker genes for type II (e.g. the Tas1r genes, Plcb2, Trpm5) and type III (e.g. Pkd2l1, Ncam, Snap25) taste cells. Bioinformatics analysis showed that genes regulating responses to stimuli were up-regulated in type II cells, while pathways related to neuronal function were up-regulated in type III cells. We also identified highly expressed genes and pathways associated with chemotaxis and axon guidance, providing new insights into the mechanisms underlying integration of new taste cells into the taste bud. We validated our results by immunohistochemically confirming expression of selected genes encoding synaptic (Cplx2 and Pclo) and semaphorin signalling pathway (Crmp2, PlexinB1, Fes and Sema4a) components. The approach described here could provide a comprehensive map of gene expression for all taste cell subpopulations and will be particularly relevant for cell types in taste buds and other tissues that can be identified only by physiological methods.


Asunto(s)
Regulación de la Expresión Génica , Papilas Gustativas/metabolismo , Gusto/fisiología , Transcriptoma , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígeno CD56/genética , Antígeno CD56/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Proto-Oncogénicas c-fes/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Transducción de Señal , Análisis de la Célula Individual/métodos , Transmisión Sináptica/genética , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , Papilas Gustativas/citología , Secuenciación del Exoma
20.
Hum Gene Ther Clin Dev ; 24(2): 86-98, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23845071

RESUMEN

Chronic granulomatous disease (CGD) is a primary immunodeficiency characterized by impaired antimicrobial activity in phagocytic cells. As a monogenic disease affecting the hematopoietic system, CGD is amenable to gene therapy. Indeed in a phase I/II clinical trial, we demonstrated a transient resolution of bacterial and fungal infections. However, the therapeutic benefit was compromised by the occurrence of clonal dominance and malignant transformation demanding alternative vectors with equal efficacy but safety-improved features. In this work we have developed and tested a self-inactivating (SIN) gammaretroviral vector (SINfes.gp91s) containing a codon-optimized transgene (gp91(phox)) under the transcriptional control of a myeloid promoter for the gene therapy of the X-linked form of CGD (X-CGD). Gene-corrected cells protected X-CGD mice from Aspergillus fumigatus challenge at low vector copy numbers. Moreover, the SINfes.gp91s vector generates substantial amounts of superoxide in human cells transplanted into immunodeficient mice. In vitro genotoxicity assays and longitudinal high-throughput integration site analysis in transplanted mice comprising primary and secondary animals for 11 months revealed a safe integration site profile with no signs of clonal dominance.


Asunto(s)
Gammaretrovirus/genética , Vectores Genéticos/metabolismo , Enfermedad Granulomatosa Crónica/terapia , Animales , Aspergillus fumigatus/patogenicidad , Células Cultivadas , Metilación de ADN , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Terapia Genética , Vectores Genéticos/genética , Humanos , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fes/genética , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA