Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogenesis ; 5: e209, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26974204

RESUMO

The sphingosine kinase-1/sphingosine 1-phosphate (SphK1/S1P) signaling pathway has been reported to modulate the expression of the canonical transcription factor hypoxia-inducible HIF-1α in multiple cell lineages. HIF-2α is also frequently overexpressed in solid tumors but its role has been mostly studied in clear cell renal cell carcinoma (ccRCC), the most common form of kidney cancer, where HIF-2α has been established as a driver of a more aggressive disease. In this study, the role of SphK1/S1P signaling with regard to HIF-2α was investigated in various cancer cell models including ccRCC cells. Under hypoxic conditions or in ccRCC lacking a functional von Hippel-Lindau (VHL) gene and expressing high levels of HIF-2α, SphK1 activity controls HIF-2α expression and transcriptional activity through a phospholipase D (PLD)-driven mechanism. SphK1 silencing promotes a VHL-independent HIF-2α loss of expression and activity and reduces cell proliferation in ccRCC. Importantly, downregulation of SphK1 is associated with impaired Akt and mTOR signaling in ccRCC. Taking advantage of a monoclonal antibody neutralizing extracellular S1P, we show that inhibition of S1P extracellular signaling blocks HIF-2α accumulation in ccRCC cell lines, an effect mimicked when the S1P transporter Spns2 or the S1P receptor 1 (S1P1) is silenced. Here, we report the first evidence that the SphK1/S1P signaling pathway regulates the transcription factor hypoxia-inducible HIF-2α in diverse cancer cell lineages notably ccRCC, where HIF-2α has been established as a driver of a more aggressive disease. These findings demonstrate that SphK1/S1P signaling may act as a canonical regulator of HIF-2α expression in ccRCC, giving support to its inhibition as a therapeutic strategy that could contribute to reduce HIF-2 activity in ccRCC.

2.
Oncogene ; 33(15): 1934-44, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23624916

RESUMO

Given the failure of chemo- and biotherapies to fight advanced pancreatic cancer, one major challenge is to identify critical events that initiate invasion. One priming step in epithelia carcinogenesis is the disruption of epithelial cell anchorage to the basement membrane which can be provided by hemidesmosomes (HDs). However, the existence of HDs in pancreatic ductal epithelium and their role in carcinogenesis remain unexplored. HDs have been explored in normal and cancer pancreatic cells, and patient samples. Unique cancer cell models where HD assembly can be pharmacologically manipulated by somatostatin/sst2 signaling have been then used to investigate the role and molecular mechanisms of dynamic HD during pancreatic carcinogenesis. We surprisingly report the presence of mature type-1 HDs comprising the integrin α6ß4 and bullous pemphigoid antigen BP180 in the human pancreatic ductal epithelium. Importantly, HDs are shown to disassemble during pancreatic carcinogenesis. HD breakdown requires phosphoinositide 3-kinase (PI3K)-dependent induction of the matrix-metalloprotease MMP-9, which cleaves BP180. Consequently, integrin α6ß4 delocalizes to the cell-leading edges where it paradoxically promotes cell migration and invasion through S100A4 activation. As S100A4 in turn stimulates MMP-9 expression, a vicious cycle maintains BP180 cleavage. Inactivation of this PI3K-MMP-9-S100A4 signaling loop conversely blocks BP180 cleavage, induces HD reassembly and inhibits cell invasion. We conclude that mature type-1 HDs are critical anchoring structures for the pancreatic ductal epithelium whose disruption, upon PI3K activation during carcinogenesis, provokes pancreatic cancer cell migration and invasion.


Assuntos
Carcinoma Ductal Pancreático/patologia , Hemidesmossomos/patologia , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Autoantígenos/metabolismo , Western Blotting , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Epitélio/metabolismo , Epitélio/patologia , Imunofluorescência , Hemidesmossomos/metabolismo , Humanos , Imuno-Histoquímica , Microscopia Confocal , Colágenos não Fibrilares/metabolismo , Neoplasias Pancreáticas/patologia , Interferência de RNA , Receptores de Somatostatina/metabolismo , Colágeno Tipo XVII
3.
Oncogene ; 33(11): 1367-74, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-23563181

RESUMO

The mRNA cap-binding protein eIF4E (eukaryotic translation initiation factor 4E) permits ribosome recruitment to capped mRNAs, and its phosphorylated form has an important role in cell transformation. The oncogenic function of eIF4E is, however, antagonised by the hypophosphorylated forms of the inhibitory eIF4E-binding proteins 1 and 2. eIF4E-binding protein 1 and 2 (4E-BP1 and 2) are two major targets of the protein kinase mTOR, and are essential for the antiproliferative effects of mTOR inhibitors. Herein, we report that pancreas expresses specifically and massively 4E-BP1 (4E-BP2 is nearly undetectable). However, 4E-BP1 expression is extinguished in more than half of the human pancreatic ductal adenocarcinomas (PDAC). 4E-BP1 shutoff is recapitulated in a mouse genetic model of PDAC, which is based on a pancreas-specific mutation of Kras, the more frequently mutated oncogene in human pancreatic tumours. 4E-BP1 downregulation enhances eIF4E phosphorylation and facilitates pancreatic cancer cell proliferation in vitro and tumour development in vivo. Furthermore, 4E-BP1 loss combined with the absence of 4E-BP2 renders eIF4E phosphorylation, protein synthesis and cell proliferation resistant to mTOR inhibition. However, proliferation can be better limited by a recently developed compound that mimics the function of 4E-BP1 and 2 independently of mTOR inhibition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Pancreáticas/patologia , Fosfoproteínas/genética , Biossíntese de Proteínas , Carcinogênese , Proteínas de Ciclo Celular , Humanos , Fosforilação
6.
Microvasc Res ; 89: 25-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23747987

RESUMO

Anti-angiogenic and anti-lymphangiogenic drugs slow tumor progression and dissemination. However, an important difficulty is that a tumor reacts and compensates to obtain the blood supply needed for tumor growth and lymphatic vessels to escape to distant loci. Therefore, there is a growing consensus on the requirement of multiple anti-(lymph)angiogenic molecules to stop cell invasion efficiently. Here we studied the cooperation between endogenous anti-angiogenic molecules, endostatin and fibstatin, and a chemokine, the Platelet Factor-4 variant 1, CXCL4L1. Anti-angiogenic factors were co-expressed by IRES-based bicistronic vectors and their cooperation was analyzed either by local delivery following transduction of pancreatic adenocarcinoma cells with lentivectors, or by distant delivery resulting from intramuscular administration in vivo of adeno-associated virus derived vectors followed by tumor subcutaneous injection. In this study, fibstatin and CXCL4L1 cooperate to inhibit endothelial cell proliferation, migration and tubulogenesis in vitro. No synergistic effect was found for fibstatin-endostatin combination. Importantly, we demonstrated for the first time that fibstatin and CXCL4L1 not only inhibit in vivo angiogenesis, but also lymphangiogenesis and tumor spread to the lymph nodes, whereas no beneficial effect was found on tumor growth inhibition using molecule combinations compared to molecules alone. These data reveal the synergy of CXCL4L1 and fibstatin in inhibition of tumor angiogenesis, lymphangiogenesis and metastasis and highlight the potential of IRES-based vectors to develop anti-metastasis combined gene therapies.


Assuntos
Proteínas de Transporte/metabolismo , Regulação Neoplásica da Expressão Gênica , Linfangiogênese/fisiologia , Proteínas de Membrana/metabolismo , Neovascularização Patológica , Fator Plaquetário 4/metabolismo , Animais , Movimento Celular , Proliferação de Células , Colágeno/química , DNA Complementar/metabolismo , Progressão da Doença , Combinação de Medicamentos , Endostatinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/química , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Proteoglicanas/química , Proteínas Recombinantes/metabolismo
7.
Oncogene ; 32(6): 671-7, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22508483

RESUMO

The eIF4E-binding proteins (4E-BPs) are inhibitors of protein synthesis that sequester the mRNA cap-binding protein eIF4E and consequently block cell growth and proliferation. In most tumors however, their inhibitory function is compromised by major oncogenic signaling pathways. Recently, thanks to the generation of mouse genetic models, considerable progress has been made in elucidating the involvement of 4E-BPs and their unique target, eIF4E, in the process of carcinogenesis. Increasing evidence indicates that an 'addiction' to protein synthesis emerges in cancer cells, highlighting the potential that 4E-BPs have as targets for therapeutics. In this review, we summarize the biochemical function, regulation and anti-oncogenic activity of the 4E-BPs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Transformação Celular Neoplásica , Fator de Iniciação 4E em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/fisiologia , Fosfoproteínas/fisiologia , Animais , Proteínas de Ciclo Celular , Proliferação de Células , Fator de Iniciação 4F em Eucariotos/metabolismo , Humanos , Fosforilação , Biossíntese de Proteínas , Proteínas Repressoras
8.
Cell Mol Life Sci ; 65(19): 3110-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18810319

RESUMO

The suppressor of translation initiation 4E-BP1 functions as a key regulator in cellular growth, differentiation, apoptosis and survival. While the control of 4E-BP1 activity via phosphorylation has been widely studied, the molecular mechanisms and the signaling pathways that govern 4E-BP1 gene expression are largely unknown. Here we show that inactivation of phosphatidylinositol 3-kinase (PI3K) consequent to stable expression of the antiproliferative somatostatin receptor 2 (sst2) in pancreatic cancer cells leads to transcriptional accumulation of the hypophosphorylated forms of 4E-BP1 protein. In cancer cells, while 4E-BP1 gene promoter is maintained repressed in a PI3K-dependent mechanism, sst2-dependent inactivation of the PI3K/Akt pathway releases 4E-BP1 gene transcription. Furthermore, the use of a pharmacological inhibitor and dominant-negative or -positive mutants of PI3K all affect 4E-BP1 protein expression and promoter activity in different cell lines. These data show that, in addition to inactivation of 4E-BP1 via hyperphosphorylation, signaling through the PI3K pathway silences 4E-BP1 gene transcription.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inativação Gênica , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas de Ciclo Celular , Linhagem Celular , Ativação Enzimática , Genes Reporter , Humanos , Dados de Sequência Molecular , Ornitina Descarboxilase/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfoproteínas/genética , Biossíntese de Proteínas , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia
9.
Cell Mol Life Sci ; 62(11): 1267-74, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15905964

RESUMO

Ornithine decarboxylase (ODC) is the ratelimiting enzyme in the biosynthesis of polyamines, which are required for optimal cell growth and proliferation. ODC is overexpressed in many tumors and, conversely, its overexpression induces transformation. We have previously reported that ODC mRNA alternative splicing relieves the translation repression normally imposed by a long and structured 5' untranslated region (UTR), and that the ODC 5' UTR contains an internal ribosome entry site (IRES). Here we show that ODC IRES activity is enhanced following inclusion of alternative sequences generated by splicing at cryptic acceptor sites. Furthermore, the alternative ODC IRES is more sensitive to cell-cycledependent changes in the rate of translation. These findings uncover a new biological property of differentially spliced transcripts. This is the first example of alternative splicing that modulates mRNA translation through the cell cycle in a cap-independent manner.


Assuntos
Processamento Alternativo , Ornitina Descarboxilase/genética , Ornitina Descarboxilase/metabolismo , RNA Mensageiro/metabolismo , Ribossomos/metabolismo , Regiões 5' não Traduzidas/genética , Regiões 5' não Traduzidas/metabolismo , Células HeLa , Humanos , RNA Mensageiro/genética
10.
J Endocrinol Invest ; 28(11 Suppl International): 5-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16625838

RESUMO

Since its discovery three decades ago as an inhibitor of GH release from the pituitary gland, somatostatin has attracted much attention because of its functional role in the regulation of a wide variety of physiological functions in the brain, pituitary, pancreas, gastrointestinal tract, adrenals, thyroid, kidney and immune system. Its actions include inhibition of endocrine and exocrine secretions, modulation of neurotransmission, motor and cognitive functions, inhibition of intestinal motility, absorption of nutrients and ions and vascular contractility. In addition, the peptide controls the proliferation of normal and tumor cells. Its action is mediated by a family of G protein-coupled receptors [somatostatin receptor (SSTR)1-SSTR5] that are widely distributed in normal and cancer cells. Direct antitumor activities, mediated through SSTR expressed in tumor cells, include blockade of autocrine/paracrine growth-promoting hormone and growth factor production, inhibition of growth factor-mediated mitogenic signals and induction of apoptosis. Indirect antitumor effects include inhibition of growth-promoting hormone and growth factor secretion, and antiangiogenic actions. Many human tumors express more than one SSTR subtype, with SSTR2 being predominant. These receptors represent the molecular basis for the clinical use of somatostatin analogs in the treatment of endocrine tumors and their in vivo localization. This review covers the present knowledge in SSTR biology and signaling.


Assuntos
Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Antineoplásicos , Divisão Celular , Cognição , Humanos , Atividade Motora , Neoplasias/patologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Transmissão Sináptica
11.
EMBO Rep ; 2(10): 893-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11600453

RESUMO

Studies on the control of eukaryotic translation initiation by a cap-independent recruitment of the 40S ribosomal subunit to internal messenger RNA sequences called internal ribosome entry sites (IRESs) have shown that these sequence elements are present in a growing list of viral and cellular RNAs. Here we discuss their prevalence, mechanisms whereby they may function and their uses in regulating gene expression.


Assuntos
Regulação da Expressão Gênica , Biossíntese de Proteínas , RNA/metabolismo , Ribossomos/química , Ribossomos/fisiologia , Regiões 5' não Traduzidas , Animais , Diferenciação Celular , Humanos , Mitose , Modelos Biológicos , RNA Mensageiro/metabolismo , Ribossomos/metabolismo
12.
Genes Dev ; 15(16): 2083-93, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11511540

RESUMO

Cap-dependent translation is mediated by eIF4F, a protein complex composed of three subunits as follows: eIF4E, which recognizes the mRNA 5' cap structure; eIF4A, an RNA-helicase; and eIF4G, a scaffolding protein that binds eIF4E, eIF4A, and the eIF4E-kinase Mnk1 simultaneously. eIF4E is hypophosphorylated and cap-dependent translation is reduced at mitosis. Here, we show that 4E-BP1, a suppressor of eIF4E function, is also hypophosphorylated in mitosis, resulting in disruption of the eIF4F complex. Consequently, eIF4E is sequestered from the eIF4G/Mnk1 complex. These results explain the specific inhibition of cap-dependent translation in mitosis and also explain how eIF4E is rendered hypophosphorylated during mitosis. Furthermore, eIF4E interaction with eIF4GII is strongly decreased coincident with hyperphosphorylation of eIF4GII. Thus, inhibition of cap-dependent translation in mitosis results from a combination of phosphorylation modifications leading to eIF4F complex disruption.


Assuntos
Proteínas de Transporte/metabolismo , Fator de Iniciação Eucariótico 4G , Mitose/genética , Proteínas de Transporte Nucleocitoplasmático , Fatores de Iniciação de Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Biossíntese de Proteínas/genética , Proteínas Adaptadoras de Transdução de Sinal , Transporte Biológico , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular , Fator de Iniciação 4F em Eucariotos , Células HeLa , Humanos , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas de Ligação ao Cap de RNA , Capuzes de RNA , Proteínas de Ligação a RNA/metabolismo
13.
Dev Biol ; 232(2): 275-83, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11401391

RESUMO

The eukaryotic translation initiation factor (eIF) 4F facilitates the recruitment of ribosomes to the mRNA 5' end. The 4E-BPs are small proteins with hypophosphorylated forms that interact with the cap binding protein eIF4E, preventing its interaction with eIF4G, thereby preventing ribosome interaction with mRNA. In sea urchin, fertilization triggers a rapid rise in protein synthesis. Here, we demonstrate that a 4E-BP homologue exists and is associated with eIF4E in unfertilized eggs. We also show that 4E-BP/eIF4E association diminishes a few minutes following fertilization. This decrease is correlated with a decrease in the total amount of 4E-BP in combination with an increase in the phosphorylation of the protein. We propose that 4E-BP acts as a repressor of protein synthesis in unfertilized sea urchin eggs and that 4E-BP/eIF4E dissociation plays an important role in the rise in protein synthesis that occurs shortly following fertilization.


Assuntos
Proteínas de Transporte/metabolismo , Fertilização/fisiologia , Fatores de Iniciação de Peptídeos/metabolismo , Ouriços-do-Mar/metabolismo , Animais , Fator de Iniciação 4E em Eucariotos , Feminino , Masculino , Óvulo/metabolismo , Iniciação Traducional da Cadeia Peptídica , Fosforilação , Testes de Precipitina , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Ribossomos/metabolismo
14.
RNA ; 7(3): 382-94, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11333019

RESUMO

Eukaryotic initiation factor (elF) 4A functions as a subunit of the initiation factor complex elF4F, which mediates the binding of mRNA to the ribosome. elF4A possesses ATPase and RNA helicase activities and is the prototype for a large family of putative RNA helicases (the DEAD box family). It is thought that the function of elF4A during translation initiation is to unwind the mRNA secondary structure in the 5' UTR to facilitate ribosome binding. However, the evidence to support this hypothesis is rather indirect, and it was reported that elF4A is also required for the translation of mRNAs possessing minimal 5' UTR secondary structure. Were this hypothesis correct, the requirement for elF4A should correlate with the degree of mRNA secondary structure. To test this hypothesis, the effect of a dominant-negative mutant of mammalian elF4A on translation of mRNAs with various degrees of secondary structure was studied in vitro. Here, we show that mRNAs containing stable secondary structure in the 5' untranslated region are more susceptible to inhibition by the elF4A mutant. The mutant protein also strongly inhibits translation from several picornavirus internal ribosome entry sites (IRES), although to different extents. UV crosslinking of elF4F subunits and elF4B to the mRNA cap structure is dramatically reduced by the elF4A mutant and RNA secondary structure. Finally, the elF4A mutant forms a more stable complex with elF4G, as compared to the wild-type elF4A, thus explaining the mechanism by which substoichiometric amounts of mutant elF4A inhibit translation.


Assuntos
Regiões 5' não Traduzidas/química , Complexo Proteico Nuclear de Ligação ao Cap , Conformação de Ácido Nucleico , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas , RNA Helicases/metabolismo , RNA Mensageiro/química , Proteínas de Saccharomyces cerevisiae , Animais , Sequência de Bases , Sequência Conservada , Fator de Iniciação 4A em Eucariotos , Camundongos , Dados de Sequência Molecular , Mutação , Fatores de Iniciação de Peptídeos/genética , Fosfoproteínas/metabolismo , Picornaviridae/genética , Ligação Proteica , Capuzes de RNA , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Ribossomos/metabolismo
15.
Curr Opin Genet Dev ; 11(1): 13-8, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11163145

RESUMO

Control of translation in eukaryotes occurs mainly at the initiation step. Translation rates in mammals are robust in the G1 phase of the cell cycle but are low during mitosis. These changes correlate with the activity of several canonical translation initiation factors, which is modulated during the cell cycle to regulate translation.


Assuntos
Regulação da Expressão Gênica , Interfase , Biossíntese de Proteínas , Animais , Mitose , Picornaviridae/genética , Picornaviridae/metabolismo , Capuzes de RNA , Transdução de Sinais , Leveduras/genética , Leveduras/metabolismo
16.
Biochem Pharmacol ; 60(8): 1237-43, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11007962

RESUMO

The purpose of this review is to summarize recent experimental data describing the regulation of the phosphorylation of eIF4E, the cap-binding protein, by the MAPK-activated protein kinase Mnk1. Mnk1 does not interact directly with eIF4E, but uses a docking site in eIF4G, a partner of eIF4E. Consequently, control of eIF4E phosphorylation may not strictly depend on changes in Mnk1 activity. The possibility that integrity of the eIF4E/eIF4G/Mnk1 complex also impinges upon eIF4E phosphorylation is discussed.


Assuntos
Fatores de Iniciação de Peptídeos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ciclo Celular/fisiologia , Fator de Iniciação 4E em Eucariotos , Fator de Iniciação Eucariótico 4G , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Fatores de Iniciação de Peptídeos/fisiologia , Fosforilação , Ligação Proteica , Proteínas de Ligação ao Cap de RNA
17.
Mol Cell ; 5(4): 607-16, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10882097

RESUMO

The eukaryotic mRNA 5' cap structure facilitates translation. However, cap-dependent translation is impaired at mitosis, suggesting a cap-independent mechanism for mRNAs translated during mitosis. Translation of ornithine decarboxylase (ODC), the rate-limiting enzyme in the biosynthesis of polyamines, peaks twice during the cell cycle, at the G1/S transition and at G2/M. Here, we describe a cap-independent internal ribosome entry site (IRES) in the ODC mRNA that functions exclusively at G2/M. This ensures elevated levels of polyamines, which are implicated in mitotic spindle formation and chromatin condensation. c-myc mRNA also contains an IRES that functions during mitosis. Thus, IRES-dependent translation is likely to be a general mechanism to synthesize short-lived proteins even at mitosis, when cap-dependent translation is interdicted.


Assuntos
Regiões 5' não Traduzidas , Interfase/fisiologia , Ornitina Descarboxilase/biossíntese , Iniciação Traducional da Cadeia Peptídica/genética , Capuzes de RNA , Códon de Iniciação , Indução Enzimática/efeitos dos fármacos , Fase G1/fisiologia , Fase G2/fisiologia , Células HeLa , Humanos , Picornaviridae/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , Sirolimo/farmacologia
18.
J Biol Chem ; 275(28): 21596-604, 2000 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-10764816

RESUMO

Accumulating evidence demonstrates that cytokine receptor signaling is negatively regulated by a family of Src homology 2 domain-containing adaptor molecules termed SOCS (suppressor of cytokine signaling). Previous studies have indicated that the expression of SOCS-related molecules is tightly controlled at the level of transcription. Furthermore, it has been reported that SOCS polypeptides are relatively unstable in cells, unless they are associated with elongins B and C. Herein, we document the existence of a third mechanism of regulation of SOCS function. Our data showed that expression of SOCS-1, a member of the SOCS family, is strongly repressed at the level of translation initiation. Structure-function analyses indicated that this effect is mediated by the 5' untranslated region of socs-1 and that it relates to the presence of two upstream AUGs in this region. Further studies revealed that socs-1 translation is cap-dependent and that it is modulated by eIF4E-binding proteins. In combination, these results uncover a novel level of regulation of SOCS-related molecules. Moreover, coupled with previous findings, they suggest that SOCS expression is tightly regulated through multiple mechanisms, in order to avoid inappropriate interference with cytokine-mediated effects.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular , Biossíntese de Proteínas , Proteínas Repressoras , Linfócitos T/metabolismo , Timo/metabolismo , Animais , Sequência de Bases , Células COS , Proteínas de Transporte/biossíntese , Linhagem Celular , Inibidores Enzimáticos , Éxons , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Recombinantes/biossíntese , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Transcrição Gênica , Transfecção , Domínios de Homologia de src
19.
Mol Cell Biol ; 19(6): 4302-10, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10330171

RESUMO

Eukaryotic translation initiation factor 4E (eIF4E) is the mRNA 5' cap binding protein, which plays an important role in the control of translation. The activity of eIF4E is regulated by a family of repressor proteins, the 4E-binding proteins (4E-BPs), whose binding to eIF4E is determined by their phosphorylation state. When hyperphosphorylated, 4E-BPs do not bind to eIF4E. Phosphorylation of the 4E-BPs is effected by the phosphatidylinositol (PI) 3-kinase signal transduction pathway and is inhibited by rapamycin through its binding to FRAP/mTOR (FK506 binding protein-rapamycin-associated protein or mammalian target of rapamycin). Phosphorylation of 4E-BPs can also be induced by protein synthesis inhibitors. These observations led to the proposal that FRAP/mTOR functions as a "sensor" of the translational apparatus (E. J. Brown and S. L. Schreiber, Cell 86:517-520, 1996). To test this model, we have employed the tetracycline-inducible system to increase eIF4E expression. Removal of tetracycline induced eIF4E expression up to fivefold over endogenous levels. Strikingly, upon induction of eIF4E, 4E-BP1 became dephosphorylated and the extent of dephosphorylation was proportional to the expression level of eIF4E. Dephosphorylation of p70(S6k) also occurred upon eIF4E induction. In contrast, the phosphorylation of Akt, an upstream effector of both p70(S6k) and 4E-BP phosphorylation, was not affected by eIF4E induction. We conclude that eIF4E engenders a negative feedback loop that targets a component of the PI 3-kinase signalling pathway which lies downstream of PI 3-kinase.


Assuntos
Proteínas de Transporte , Fosfoproteínas/fisiologia , Biossíntese de Proteínas , Proteínas Quinases S6 Ribossômicas/fisiologia , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anisomicina/farmacologia , Western Blotting , Proteínas de Ciclo Celular , Linhagem Celular Transformada , Cicloeximida/farmacologia , Fatores de Iniciação em Eucariotos , Immunoblotting , Camundongos , Modelos Biológicos , Proteína Oncogênica v-akt , Ornitina Descarboxilase/metabolismo , Fosforilação , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Oncogênicas de Retroviridae/metabolismo , Sirolimo/farmacologia , Tetraciclina/farmacologia , Fatores de Tempo
20.
EMBO J ; 18(1): 270-9, 1999 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-9878069

RESUMO

Human eukaryotic translation initiation factor 4E (eIF4E) binds to the mRNA cap structure and interacts with eIF4G, which serves as a scaffold protein for the assembly of eIF4E and eIF4A to form the eIF4F complex. eIF4E is an important modulator of cell growth and proliferation. It is the least abundant component of the translation initiation machinery and its activity is modulated by phosphorylation and interaction with eIF4E-binding proteins (4E-BPs). One strong candidate for the eIF4E kinase is the recently cloned MAPK-activated protein kinase, Mnk1, which phosphorylates eIF4E on its physiological site Ser209 in vitro. Here we report that Mnk1 is associated with the eIF4F complex via its interaction with the C-terminal region of eIF4G. Moreover, the phosphorylation of an eIF4E mutant lacking eIF4G-binding capability is severely impaired in cells. We propose a model whereby, in addition to its role in eIF4F assembly, eIF4G provides a docking site for Mnk1 to phosphorylate eIF4E. We also show that Mnk1 interacts with the C-terminal region of the translational inhibitor p97, an eIF4G-related protein that does not bind eIF4E, raising the possibility that p97 can block phosphorylation of eIF4E by sequestering Mnk1.


Assuntos
Fatores de Iniciação de Peptídeos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Fator de Iniciação 4E em Eucariotos , Fator de Iniciação Eucariótico 4G , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Biológicos , Fatores de Iniciação de Peptídeos/genética , Fosforilação , Mutação Puntual , Ligação Proteica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...