Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 5: e209, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26974204

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-23624916

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Hemidesmosomes/pathology , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Autoantigens/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/physiology , Epithelium/metabolism , Epithelium/pathology , Fluorescent Antibody Technique , Hemidesmosomes/metabolism , Humans , Immunohistochemistry , Microscopy, Confocal , Non-Fibrillar Collagens/metabolism , Pancreatic Neoplasms/pathology , RNA Interference , Receptors, Somatostatin/metabolism , Collagen Type XVII
3.
Oncogene ; 33(11): 1367-74, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-23563181

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Pancreatic Neoplasms/pathology , Phosphoproteins/genetics , Protein Biosynthesis , Carcinogenesis , Cell Cycle Proteins , Humans , Phosphorylation
6.
Microvasc Res ; 89: 25-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23747987

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lymphangiogenesis/physiology , Membrane Proteins/metabolism , Neovascularization, Pathologic , Platelet Factor 4/metabolism , Animals , Cell Movement , Cell Proliferation , Collagen/chemistry , DNA, Complementary/metabolism , Disease Progression , Drug Combinations , Endostatins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Laminin/chemistry , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/blood supply , Proteoglycans/chemistry , Recombinant Proteins/metabolism
7.
Oncogene ; 32(6): 671-7, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-22508483

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Transformation, Neoplastic , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factors/physiology , Phosphoproteins/physiology , Animals , Cell Cycle Proteins , Cell Proliferation , Eukaryotic Initiation Factor-4F/metabolism , Humans , Phosphorylation , Protein Biosynthesis , Repressor Proteins
8.
Cell Mol Life Sci ; 65(19): 3110-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18810319

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Silencing , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Cell Cycle Proteins , Cell Line , Enzyme Activation , Genes, Reporter , Humans , Molecular Sequence Data , Ornithine Decarboxylase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/genetics , Protein Biosynthesis , Repressor Proteins/genetics , Signal Transduction/physiology
9.
Cell Mol Life Sci ; 62(11): 1267-74, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15905964

ABSTRACT

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.


Subject(s)
Alternative Splicing , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , 5' Untranslated Regions/genetics , 5' Untranslated Regions/metabolism , HeLa Cells , Humans , RNA, Messenger/genetics
10.
J Endocrinol Invest ; 28(11 Suppl International): 5-9, 2005.
Article in English | MEDLINE | ID: mdl-16625838

ABSTRACT

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.


Subject(s)
Receptors, Somatostatin/physiology , Somatostatin/physiology , Antineoplastic Agents , Cell Division , Cognition , Humans , Motor Activity , Neoplasms/pathology , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Synaptic Transmission
11.
EMBO Rep ; 2(10): 893-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11600453

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis , RNA/metabolism , Ribosomes/chemistry , Ribosomes/physiology , 5' Untranslated Regions , Animals , Cell Differentiation , Humans , Mitosis , Models, Biological , RNA, Messenger/metabolism , Ribosomes/metabolism
12.
Genes Dev ; 15(16): 2083-93, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11511540

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4G , Mitosis/genetics , Nucleocytoplasmic Transport Proteins , Peptide Initiation Factors/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/genetics , Adaptor Proteins, Signal Transducing , Biological Transport , Cell Cycle/physiology , Cell Cycle Proteins , Eukaryotic Initiation Factor-4F , HeLa Cells , Humans , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Phosphorylation , RNA Cap-Binding Proteins , RNA Caps , RNA-Binding Proteins/metabolism
13.
Dev Biol ; 232(2): 275-83, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11401391

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Fertilization/physiology , Peptide Initiation Factors/metabolism , Sea Urchins/metabolism , Animals , Eukaryotic Initiation Factor-4E , Female , Male , Ovum/metabolism , Peptide Chain Initiation, Translational , Phosphorylation , Precipitin Tests , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism
14.
RNA ; 7(3): 382-94, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11333019

ABSTRACT

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.


Subject(s)
5' Untranslated Regions/chemistry , Nuclear Cap-Binding Protein Complex , Nucleic Acid Conformation , Peptide Initiation Factors/metabolism , Protein Biosynthesis , RNA Helicases/metabolism , RNA, Messenger/chemistry , Saccharomyces cerevisiae Proteins , Animals , Base Sequence , Conserved Sequence , Eukaryotic Initiation Factor-4A , Mice , Molecular Sequence Data , Mutation , Peptide Initiation Factors/genetics , Phosphoproteins/metabolism , Picornaviridae/genetics , Protein Binding , RNA Caps , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Ribosomes/metabolism
15.
Curr Opin Genet Dev ; 11(1): 13-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11163145

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Interphase , Protein Biosynthesis , Animals , Mitosis , Picornaviridae/genetics , Picornaviridae/metabolism , RNA Caps , Signal Transduction , Yeasts/genetics , Yeasts/metabolism
16.
Biochem Pharmacol ; 60(8): 1237-43, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11007962

ABSTRACT

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.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Cell Cycle/physiology , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Humans , Intracellular Signaling Peptides and Proteins , Peptide Initiation Factors/physiology , Phosphorylation , Protein Binding , RNA Cap-Binding Proteins
17.
Mol Cell ; 5(4): 607-16, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10882097

ABSTRACT

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.


Subject(s)
5' Untranslated Regions , Interphase/physiology , Ornithine Decarboxylase/biosynthesis , Peptide Chain Initiation, Translational/genetics , RNA Caps , Codon, Initiator , Enzyme Induction/drug effects , G1 Phase/physiology , G2 Phase/physiology , HeLa Cells , Humans , Picornaviridae/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Sirolimus/pharmacology
18.
J Biol Chem ; 275(28): 21596-604, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10764816

ABSTRACT

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.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Protein Biosynthesis , Repressor Proteins , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Base Sequence , COS Cells , Carrier Proteins/biosynthesis , Cell Line , Enzyme Inhibitors , Exons , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Sequence Alignment , Sequence Homology, Nucleic Acid , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Transcription, Genetic , Transfection , src Homology Domains
19.
Mol Cell Biol ; 19(6): 4302-10, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330171

ABSTRACT

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.


Subject(s)
Carrier Proteins , Phosphoproteins/physiology , Protein Biosynthesis , Ribosomal Protein S6 Kinases/physiology , 3T3 Cells , Adaptor Proteins, Signal Transducing , Animals , Anisomycin/pharmacology , Blotting, Western , Cell Cycle Proteins , Cell Line, Transformed , Cycloheximide/pharmacology , Eukaryotic Initiation Factors , Immunoblotting , Mice , Models, Biological , Oncogene Protein v-akt , Ornithine Decarboxylase/metabolism , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Retroviridae Proteins, Oncogenic/metabolism , Sirolimus/pharmacology , Tetracycline/pharmacology , Time Factors
20.
EMBO J ; 18(1): 270-9, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9878069

ABSTRACT

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.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Line , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Mice , Models, Biological , Peptide Initiation Factors/genetics , Phosphorylation , Point Mutation , Protein Binding , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...