Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
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.
EMBO J ; 41(5): e109386, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35112724

RESUMEN

The mechanisms whereby neutrophils respond differentially to live and dead organisms are unknown. We show here that neutrophils produce 5- to 30-fold higher levels of the Cxcl2 chemokine in response to live bacteria, compared with killed bacteria or isolated bacterial components, despite producing similar levels of Cxcl1 or pro-inflammatory cytokines. Secretion of high levels of Cxcl2, which potently activates neutrophils by an autocrine mechanism, requires three signals. The first two signals are provided by two different sets of signal peptides released by live bacteria, which selectively activate formylated peptide receptor 1 (Fpr1) and Fpr2, respectively. Signal 3 originates from Toll-like receptor activation by microbial components present in both live and killed bacteria. Mechanistically, these signaling pathways converge at the level of the p38 MAP kinase, leading to activation of the AP-1 transcription factor and to Cxcl2 induction. Collectively, our data demonstrate that the simultaneous presence of agonists for Fpr1, Fpr2, and Toll-like receptors represents a unique signature associated with viable bacteria, which is sensed by neutrophils and induces Cxcl2-dependent autocrine cell activation.


Asunto(s)
Bacterias/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Receptores Toll-Like/metabolismo , Animales , Citocinas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
3.
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
4.
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
6.
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
7.
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
8.
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
9.
PLoS One ; 12(7): e0181178, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28727840

RESUMEN

Acute myelogenous leukemia (AML) is often associated with activating mutations in the receptor tyrosine kinase, Flt3, including internal tandem duplications (ITDs) within the regulatory juxtamembrane region. Previous studies have linked Flt3-ITD to the activation of the Fes protein tyrosine kinase in AML, and RNAi-knockdown studies suggest that Fes may be required for Flt3 function. In this study, we tested Fes inhibitors from three different chemical classes for their growth-suppressive activity against Flt3-ITD+ myeloid leukemia cell lines (MV4-11, MOLM-13 and MOLM-14) vs. myeloid cells with wild-type Flt3 (THP-1). All Fes inhibitors selectively inhibited the growth of Flt3-ITD+ AML cells, with IC50 values for diaminopyrimidine and pyrrolopyridine inhibitors ranging from 19 to 166 nM. In contrast, a pyrazolopyrimidine inhibitor was less potent in Flt3-ITD+ AML cells, with IC50 values in the 1.0 µM range. In vitro kinase assays showed that the most potent inhibitors of Flt3-ITD+ AML cell proliferation blocked both Fes and Flt3-ITD kinase activity, while the pyrazolopyrimidine was more selective for Fes vs. Flt3-ITD. All three inhibitors induced significant apoptosis in Flt3-ITD+ AML cells, with potency equivalent to or greater than the established Flt3-ITD inhibitor, tandutinib. Transformation of TF-1 cells with Flt3-ITD resulted in constitutive activation of endogenous Fes, and rendered the cells highly sensitive to all three Fes inhibitors with IC50 values in the 30-500 nM range. The pyrrolopyridine compound also induced apoptotic responses in patient-derived Flt3-ITD+ AML bone marrow cells but not in normal bone marrow mononuclear cells. These results demonstrate that Fes kinase activity contributes to Flt3-ITD signaling in AML, and suggests that dual inhibition of both Flt3 and Fes may provide a therapeutic advantage for the treatment of Flt3-ITD+ AML.


Asunto(s)
Leucemia Mieloide Aguda/patología , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fes/antagonistas & inhibidores , Pirazoles/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Concentración 50 Inhibidora , Leucemia Mieloide Aguda/enzimología , Masculino , Persona de Mediana Edad , Mutación , Transducción de Señal , Secuencias Repetidas en Tándem
10.
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
11.
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
12.
J Hematol Oncol ; 8: 47, 2015 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-25956236

RESUMEN

Eukaryotic cell membrane dynamics change in curvature during physiological and pathological processes. In the past ten years, a novel protein family, Fes/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain proteins, has been identified to be the most important coordinators in membrane curvature regulation. The F-BAR domain family is a member of the Bin/Amphiphysin/Rvs (BAR) domain superfamily that is associated with dynamic changes in cell membrane. However, the molecular basis in membrane structure regulation and the biological functions of F-BAR protein are unclear. The pathophysiological role of F-BAR protein is unknown. This review summarizes the current understanding of structure and function in the BAR domain superfamily, classifies F-BAR family proteins into nine subfamilies based on domain structure, and characterizes F-BAR protein structure, domain interaction, and functional relevance. In general, F-BAR protein binds to cell membrane via F-BAR domain association with membrane phospholipids and initiates membrane curvature and scission via Src homology-3 (SH3) domain interaction with its partner proteins. This process causes membrane dynamic changes and leads to seven important cellular biological functions, which include endocytosis, phagocytosis, filopodium, lamellipodium, cytokinesis, adhesion, and podosome formation, via distinct signaling pathways determined by specific domain-binding partners. These cellular functions play important roles in many physiological and pathophysiological processes. We further summarize F-BAR protein expression and mutation changes observed in various diseases and developmental disorders. Considering the structure feature and functional implication of F-BAR proteins, we anticipate that F-BAR proteins modulate physiological and pathophysiological processes via transferring extracellular materials, regulating cell trafficking and mobility, presenting antigens, mediating extracellular matrix degradation, and transmitting signaling for cell proliferation.


Asunto(s)
Membrana Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Humanos , Antígenos de Histocompatibilidad Menor
14.
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
15.
Biotechnol Lett ; 35(11): 1799-806, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23801117

RESUMEN

MiR-424 plays an important role via promoting the monocytic differentiation in many human leukemia cell lines. Here, we report that miR-424 decreased miR-125b expression to 36 % by directly targeting caudal type homeobox 2. However, miR-424 also decreased expression of Fes, PU.1 and colony-stimulating factor receptor (MCSFR). As Fes, PU.1 and MCSFR were down-regulated by over-expression of miR-125b (unpublished work), a similar effect of miR-424 and Fes siRNA on CD64, Egr-1, Egr-2 and CEBPA indicates that Fes may be an important downstream target of miR-424. We hypothesize that miR-424 promotes monocytic differentiation by regulating other critical factors and miR-424 has high affinity for these factors. For the first time, the molecular mechanism of miR-424 during monocytic differentiation of U937 cells has been elucidated in this study.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Monocitos/efectos de los fármacos , Factor de Transcripción CDX2 , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/biosíntesis , Monocitos/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-fes/biosíntesis , Receptores del Factor Estimulante de Colonias/biosíntesis , Transactivadores/biosíntesis , Células U937
16.
Wei Sheng Wu Xue Bao ; 53(3): 299-305, 2013 Mar 04.
Artículo en Chino | 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
17.
FEBS J ; 280(21): 5198-212, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23668323

RESUMEN

Syndapin [also called PACSIN (protein kinase C and casein kinase II interacting protein)] is an Fes-CIP4 homology Bin-amphiphysin-Rvs161/167 (F-BAR) and Src-homology 3 domain-containing protein. Three genes give rise to three main isoforms in mammalian cells. They each function in different endocytic and vesicle trafficking pathways and provide critical links between the cytoskeletal network in different cellular processes, such as neuronal morphogenesis and cell migration. The membrane remodelling activity of syndapin via its F-BAR domain and its interaction partners, such as dynamin and neural Wiskott-Aldrich syndrome protein binding to its Src-homology 3 domain, are important with respect to its function. Its various partner proteins provide insights into its mechanism of action, as well as its differential roles in these cellular processes. Signalling pathways leading to the regulation of syndapin function by phosphorylation are now contributing to our understanding of the broader functions of this family of proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endocitosis/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Animales , Humanos , Dominios y Motivos de Interacción de Proteínas
18.
J Biol Chem ; 288(14): 9881-9891, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23404507

RESUMEN

The products of the oncogene Fes and JAK3 are tyrosine kinases, whose expressions are elevated in tumor growth, angiogenesis, and metastasis. Phosphatidic acid, as synthesized by phospholipase D (PLD), enhances cancer cell survival. We report a new signaling pathway that integrates the two kinases with the lipase. A new JAK3-Fes-PLD2 axis is responsible for the highly proliferative phenotype of MDA-MB-231 breast cancer cells. Conversely, this pathway is maintained at a low rate of expression and activity levels in untransformed cells such as MCF10A. We also deciphered the inter-regulation that exists between the two kinases (JAK3 and the oncogene Fes) and between these two kinases and the lipase (PLD2). Whereas JAK3 and Fes marginally activate PLD2 in non-transformed cells, these kinases greatly enhance (>200%) PLD activity following protein-protein interaction through the SH2 domain and the Tyr-415 residue of PLD2. We also found that phosphatidic acid enhances Fes activity in MDA-MB-231 cells providing a positive activation loop between Fes and PLD2. In summary, the JAK3, Fes and PLD2 interactions in transformed cells maintain PLD2 at an enhanced level that leads to abnormal cell growth. Modulating this new JAK3-Fes-PLD2 pathway could be important to control the highly invasive phenotype of breast cancer cells.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Janus Quinasa 1/metabolismo , Fosfolipasa D/fisiología , Proteínas Proto-Oncogénicas c-fes/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Células COS , Línea Celular Tumoral , Proliferación Celular , Chlorocebus aethiops , Células HL-60 , Humanos , Invasividad Neoplásica , Neoplasias/metabolismo , Fenotipo , Ácidos Fosfatidicos/química , Fosfolipasa D/química , Plásmidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
20.
Mol Cancer Res ; 10(7): 881-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22589410

RESUMEN

KIT receptor is required for mast cell development, survival, and migration toward its ligand stem cell factor (SCF). Many solid tumors express SCF and this leads to mast cell recruitment to tumors and release of mediators linked to tumor angiogenesis, growth, and metastasis. Here, we investigate whether FES protein-tyrosine kinase, a downstream effector of KIT signaling in mast cells, is required for migration of mast cells toward SCF-expressing mammary tumors. Using a novel agarose drop assay for chemotaxis of bone marrow-derived mast cells (BMMC) toward SCF, we found that defects in chemotaxis of fes-null BMMCs correlated with disorganized microtubule networks in polarized cells. FES displayed partial colocalization with microtubules in polarized BMMCs and has at least two direct microtubule binding sites within its N-terminal F-BAR and SH2 domains. An oligomerization-disrupting mutation within the Fer/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain had no effect on microtubule binding, whereas microtubule binding to the SH2 domain was dependent on the phosphotyrosine-binding pocket. FES involvement in mast cell recruitment to tumors was tested using the AC2M2 mouse mammary carcinoma model. These tumor cells expressed SCF and promoted BMMC recruitment in a KIT- and FES-dependent manner. Engraftment of AC2M2 orthotopic and subcutaneous tumors in control or fes-null mice, revealed a key role for FES in recruitment of mast cells to the tumor periphery. This may contribute to the reduced tumor growth and metastases observed in fes-null mice compared with control mice. Taken together, FES is a potential therapeutic target to limit the progression of tumors with stromal mast cell involvement.


Asunto(s)
Células de la Médula Ósea , Neoplasias Mamarias Experimentales , Mastocitos , Proteínas Proto-Oncogénicas c-fes , Proteínas Proto-Oncogénicas c-kit , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/terapia , Mastocitos/citología , Mastocitos/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-fes/genética , Proteínas Proto-Oncogénicas c-fes/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...