Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 41(5): e109386, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35112724

RESUMO

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.


Assuntos
Bactérias/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-fes/metabolismo , Receptores Toll-Like/metabolismo , Animais , Citocinas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia
2.
Circ Res ; 131(12): 1004-1017, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36321446

RESUMO

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.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Placa Aterosclerótica , Proteínas Proto-Oncogênicas c-fes , Animais , Humanos , Camundongos , Artérias/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Estudo de Associação Genômica Ampla , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Proto-Oncogênicas c-fes/metabolismo
3.
Cell ; 134(5): 793-803, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18775312

RESUMO

The SH2 domain of cytoplasmic tyrosine kinases can enhance catalytic activity and substrate recognition, but the molecular mechanisms by which this is achieved are poorly understood. We have solved the structure of the prototypic SH2-kinase unit of the human Fes tyrosine kinase, which appears specialized for positive signaling. In its active conformation, the SH2 domain tightly interacts with the kinase N-terminal lobe and positions the kinase alphaC helix in an active configuration through essential packing and electrostatic interactions. This interaction is stabilized by ligand binding to the SH2 domain. Our data indicate that Fes kinase activation is closely coupled to substrate recognition through cooperative SH2-kinase-substrate interactions. Similarly, we find that the SH2 domain of the active Abl kinase stimulates catalytic activity and substrate phosphorylation through a distinct SH2-kinase interface. Thus, the SH2 and catalytic domains of active Fes and Abl pro-oncogenic kinases form integrated structures essential for effective tyrosine kinase signaling.


Assuntos
Proteínas Proto-Oncogênicas c-abl/química , Proteínas Proto-Oncogênicas c-fes/química , Sequência de Aminoácidos , Cristalografia por Raios X , Ativação Enzimática , Humanos , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-fes/metabolismo
4.
J Gastroenterol Hepatol ; 34(10): 1869-1877, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31038805

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Metilação de DNA , Epigênese Genética , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogênicas c-fes/genética , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Ilhas de CpG , Detecção Precoce de Câncer/métodos , Feminino , Predisposição Genética para Doença , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Fenótipo , Valor Preditivo dos Testes , Intervalo Livre de Progressão , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fes/metabolismo , Fatores de Tempo , Carga Tumoral , alfa-Fetoproteínas/análise
5.
J Biol Chem ; 288(14): 9881-9891, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23404507

RESUMO

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.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Janus Quinase 1/metabolismo , Fosfolipase D/fisiologia , Proteínas Proto-Oncogênicas c-fes/metabolismo , Animais , Neoplasias da Mama/metabolismo , Células COS , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , Células HL-60 , Humanos , Invasividade Neoplásica , Neoplasias/metabolismo , Fenótipo , Ácidos Fosfatídicos/química , Fosfolipase D/química , Plasmídeos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais
6.
J Biol Chem ; 287(1): 393-407, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22094461

RESUMO

Cell differentiation is compromised in acute leukemias. We report that mammalian target of rapamycin (mTOR) and S6 kinase (S6K) are highly expressed in the undifferentiated promyelomonocytic leukemic HL-60 cell line, whereas PLD2 expression is minimal. The expression ratio of PLD2 to mTOR (or to S6K) is gradually inverted upon in vitro induction of differentiation toward the neutrophilic phenotype. We present three ways that profoundly affect the kinetics of differentiation as follows: (i) simultaneous overexpression of mTOR (or S6K), (ii) silencing of mTOR via dsRNA-mediated interference or inhibition with rapamycin, and (iii) PLD2 overexpression. The last two methods shortened the time required for differentiation. By determining how PLD2 participates in cell differentiation, we found that PLD2 interacts with and activates the oncogene Fes/Fps, a protein-tyrosine kinase known to be involved in myeloid cell development. Fes activity is elevated with PLD2 overexpression, phosphatidic acid or phosphatidylinositol bisphosphate. Co-immunoprecipitation indicates a close PLD2-Fes physical interaction that is negated by a Fes-R483K mutant that incapacitates its Src homology 2 domain. All these suggest for the first time the following mechanism: mTOR/S6K down-regulation→PLD2 overexpression→PLD2/Fes association→phosphatidic acid-led activation of Fes kinase→granulocytic differentiation. Differentiation shortening could have a clinical impact on reducing the time of return to normalcy of the white cell counts after chemotherapy in patients with acute promyelocytic leukemia.


Assuntos
Diferenciação Celular , Leucemia Mieloide/patologia , Fosfolipase D/metabolismo , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inativação Gênica , Células HL-60 , Humanos , Cinética , Dados de Sequência Molecular , Fosfolipase D/genética , Proteínas Proto-Oncogênicas c-fes/química , Proteínas Proto-Oncogênicas c-fes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/deficiência , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Domínios de Homologia de src
7.
EMBO J ; 27(1): 38-50, 2008 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-18046454

RESUMO

The remodeling of epithelial monolayers induced by hepatocyte growth factor (HGF) results in the reorganization of actin cytoskeleton and cellular junctions. We previously showed that the membrane-cytoskeleton linker ezrin plays a major role in HGF-induced morphogenic effects. Here we identified a novel partner of phosphorylated ezrin, the Fes kinase, that acts downstream of ezrin in HGF-mediated cell scattering. We found that Fes interacts directly, through its SH2 domain, with ezrin phosphorylated at tyrosine 477. We show that in epithelial cells, activated Fes localizes either to focal adhesions or cell-cell contacts depending on cell confluency. The recruitment and the activation of Fes to the cell-cell contacts in confluent cells depend on its interaction with ezrin. When this interaction is impaired, Fes remains in focal adhesions and as a consequence the cells show defective spreading and scattering in response to HGF stimulation. Altogether, these results provide a novel mechanism whereby ezrin/Fes interaction at cell-cell contacts plays an essential role in HGF-induced cell scattering and implicates Fes in the cross-talk between cell-cell and cell-matrix adhesion.


Assuntos
Movimento Celular/fisiologia , Proteínas do Citoesqueleto/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Proteínas Proto-Oncogênicas c-fes/metabolismo , Animais , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Junções Célula-Matriz/fisiologia , Ativação Enzimática/fisiologia , Células LLC-PK1 , Suínos
8.
Physiol Genomics ; 43(8): 417-37, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21303932

RESUMO

Pulmonary arterial hypertension (PAH) is up to threefold more prevalent in women than men. Female mice overexpressing the serotonin transporter (SERT; SERT+ mice) exhibit PAH and exaggerated hypoxia-induced PAH, whereas male SERT+ mice remain unaffected. To further investigate these sex differences, microarray analysis was performed in the pulmonary arteries of normoxic and chronically hypoxic female and male SERT+ mice. Quantitative RT-PCR analysis was employed for validation of the microarray data. In relevant groups, immunoblotting was performed for genes of interest (CEBPß, CYP1B1, and FOS). To translate clinical relevance to our findings, CEBPß, CYP1B1, and FOS mRNA and protein expression was assessed in pulmonary artery smooth muscle cells (PASMCs) derived from idiopathic PAH (IPAH) patients and controls. In female SERT+ mice, multiple pathways with relevance to PAH were altered. This was also observed in chronically hypoxic female SERT+ mice. We selected 10 genes of interest for qRT-PCR analysis (FOS, CEBPß, CYP1B1, MYL3, HAMP2, LTF, PLN, NPPA, UCP1, and C1S), and 100% concordance was reported. Protein expression of three selected genes, CEBPß, CYP1B1, FOS, was also upregulated in female SERT+ mice. Serotonin and 17ß-estradiol increased CEBPß, CYP1B1, and FOS protein expression in PASMCs. In addition, CEBPß, CYP1B1, and FOS mRNA and protein expression was also increased in PASMCs derived from IPAH patients. Here, we have identified a number of genes that may predispose female SERT+ mice to PAH, and these findings may also be relevant to human PAH.


Assuntos
Hipóxia/complicações , Análise em Microsséries/métodos , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Animais , Animais Geneticamente Modificados/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Técnicas de Cultura de Células , Doença Hepática Crônica Induzida por Substâncias e Drogas , Citocromo P-450 CYP1B1 , Estrogênios/genética , Hipertensão Pulmonar Primária Familiar , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Hipertensão Pulmonar/genética , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-fes/metabolismo , Sexo
9.
Biochem Biophys Res Commun ; 393(1): 174-8, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20117079

RESUMO

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.


Assuntos
Quimiotaxia , Proteínas Proto-Oncogênicas c-fes/metabolismo , Fator de Células-Tronco/metabolismo , Domínios de Homologia de src , Adesão Celular , Linhagem Celular Tumoral , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-fes/genética , Técnicas do Sistema de Duplo-Híbrido , Tirosina
10.
Genes Chromosomes Cancer ; 48(3): 272-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19051325

RESUMO

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.


Assuntos
Neoplasias Colorretais/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fes/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Ilhas de CpG/fisiologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação para Baixo , Humanos , Dados de Sequência Molecular , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-fes/metabolismo
11.
Nat Commun ; 11(1): 3216, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587248

RESUMO

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.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes , Proteínas Proto-Oncogênicas c-fes , Transportadores de Cassetes de Ligação de ATP/química , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem Celular , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Edição de Genes , Humanos , Macrófagos/metabolismo , Mutação , Neutrófilos , Fagocitose , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fes/química , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Proto-Oncogênicas c-fes/metabolismo , Transdução de Sinais , Quinase Syk/metabolismo
12.
Clin Cancer Res ; 26(1): 265-273, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31573955

RESUMO

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.


Assuntos
Apoptose , Dano ao DNA , Neoplasias Pulmonares/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-fes/antagonistas & inibidores , Interferência de RNA , Radiossensibilizantes/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Camundongos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/radioterapia , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Proto-Oncogênicas c-fes/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Tolerância a Radiação , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Oncol ; 34(1): 89-96, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19082481

RESUMO

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.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Proteínas Proto-Oncogênicas c-fes/metabolismo , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Núcleo Celular , Proliferação de Células , Regulação para Baixo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Proteínas I-kappa B/metabolismo , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-fes/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fes/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas
15.
Int J Mol Med ; 20(1): 113-21, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17549397

RESUMO

Antiangiogenic therapy, including blockade of vascular endothelial growth factor (VEGF) signaling, was highly anticipated to improve the prognosis for patients with advanced cancers following the success of preclinical animal models. However, antiangiogenic monotherapy with VEGF antagonists has produced disappointing results in clinical trials to date. One of the reasons for this poor outcome is that angiogenesis is not solely regulated by VEGF. Inhibition of VEGF signaling, therefore, may select for tumor cell populations that stimulate angiogenesis through VEGF-independent pathways. Successful antiangiogenic therapy, therefore, may require simultaneous blockade of signaling downstream from multiple proangiogenic factor receptors. Recently, we found that non-receptor protein-tyrosine kinases, including members of the Src and Fes families, play vital roles in the responses of cultured endothelial cells to several proangiogenic factors. In this review, we summarize the contributions of these kinase families to angiogenic pathways in endothelial cells, and discuss the potential of these kinases as new targets for antiangiogenic drug discovery.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endotélio Vascular/metabolismo , Proteínas Proto-Oncogênicas c-fes/fisiologia , Quinases da Família src/fisiologia , Inibidores da Angiogênese/metabolismo , Animais , Capilares/metabolismo , Linhagem Celular , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Modelos Biológicos , Fosforilação , Proteínas Proto-Oncogênicas c-fes/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
16.
J Leukoc Biol ; 80(6): 1522-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16959897

RESUMO

Fps/Fes and Fer are members of a distinct subfamily of cytoplasmic protein tyrosine kinases that have recently been implicated in the regulation of innate immunity. Previous studies showed that mice lacking Fps/Fes are hypersensitive to systemic LPS challenge, and Fer-deficient mice displayed enhanced recruitment of leukocytes in response to local LPS challenge. This study identifies physiological, cellular, and molecular defects that contribute to the hyperinflammatory phenotype in Fps/Fes null mice. Plasma TNF-alpha levels were elevated in LPS challenged Fps/Fes null mice as compared with wild-type mice and cultured Fps/Fes null peritoneal macrophages treated with LPS showed increased TNF-alpha production. Cultured Fps/Fes null macrophages also displayed prolonged LPS-induced degradation of IkappaB-alpha, increased phosphorylation of the p65 subunit of NF-kappaB, and defective TLR4 internalization, compared with wild-type macrophages. Together, these observations provide a likely mechanistic basis for elevated proinflammatory cytokine secretion by Fps/Fes null macrophages and the increased sensitivity of Fps/Fes null mice to endotoxin. We posit that Fps/Fes modulates the innate immune response of macrophages to LPS, in part, by regulating internalization and down-regulation of the TLR4 receptor complex.


Assuntos
Regulação para Baixo/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/imunologia , Proteínas Proto-Oncogênicas c-fes/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Transcrição RelA/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Proteínas I-kappa B/imunologia , Proteínas I-kappa B/metabolismo , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Inibidor de NF-kappaB alfa , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Proto-Oncogênicas c-fes/deficiência , Proteínas Proto-Oncogênicas c-fes/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
17.
Biochem J ; 399(1): 141-50, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16792528

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas c-fes/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Humanos , Insetos , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Repressoras/genética , Saccharomyces cerevisiae , Especificidade por Substrato , Proteína 28 com Motivo Tripartido
18.
J Clin Invest ; 127(6): 2310-2325, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28463229

RESUMO

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.


Assuntos
Melanoma/genética , Proteínas Proto-Oncogênicas c-fes/genética , Neoplasias Cutâneas/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Variações do Número de Cópias de DNA , Genes Supressores de Tumor , Genômica , Humanos , Melanoma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Transplante de Neoplasias , Oncogenes , Proteínas Proto-Oncogênicas c-fes/metabolismo , Neoplasias Cutâneas/metabolismo , Via de Sinalização Wnt
19.
Gene ; 620: 1-9, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28389358

RESUMO

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.


Assuntos
Diferenciação Celular , Leucemia Mieloide Aguda/metabolismo , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-fes/genética , Regiões 3' não Traduzidas , Antígenos CD/genética , Antígenos CD/metabolismo , Calcitriol/farmacologia , Ciclo Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Leucemia Mieloide Aguda/genética , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Proteínas Proto-Oncogênicas c-fes/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
20.
Sci Rep ; 7(1): 7595, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28790351

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Papilas Gustativas/metabolismo , Paladar/fisiologia , Transcriptoma , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antígeno CD56/genética , Antígeno CD56/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Proto-Oncogênicas c-fes/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Transdução de Sinais , Análise de Célula Única/métodos , Transmissão Sináptica/genética , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Papilas Gustativas/citologia , Sequenciamento do Exoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA