Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell ; 133(2): 314-27, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423202

ABSTRACT

In mammalian cells, nonsense-mediated mRNA decay (NMD) generally requires that translation terminates sufficiently upstream of a post-splicing exon junction complex (EJC) during a pioneer round of translation. The subsequent binding of Upf1 to the EJC triggers Upf1 phosphorylation. We provide evidence that phospho-Upf1 functions after nonsense codon recognition during steps that involve the translation initiation factor eIF3 and mRNA decay factors. Phospho-Upf1 interacts directly with eIF3 and inhibits the eIF3-dependent conversion of 40S/Met-tRNA(i)(Met)/mRNA to translationally competent 80S/Met-tRNA(i)(Met)/mRNA initiation complexes to repress continued translation initiation. Consistent with phospho-Upf1 impairing eIF3 function, NMD fails to detectably target nonsense-containing transcripts that initiate translation independently of eIF3 from the CrPV IRES. There is growing evidence that translational repression is a key transition that precedes mRNA delivery to the degradation machinery. Our results uncover a critical step during NMD that converts a pioneer translation initiation complex to a translationally compromised mRNP.


Subject(s)
Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , Trans-Activators/metabolism , Animals , COS Cells , Chlorocebus aethiops , Codon, Nonsense , HeLa Cells , Hepacivirus/metabolism , Humans , Phosphorylation , RNA Helicases , Ribonucleoproteins/metabolism
2.
Mol Cell Proteomics ; 6(7): 1135-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17322308

ABSTRACT

Protein synthesis in mammalian cells requires initiation factor eIF3, an approximately 800-kDa protein complex that plays a central role in binding of initiator methionyl-tRNA and mRNA to the 40 S ribosomal subunit to form the 48 S initiation complex. The eIF3 complex also prevents premature association of the 40 and 60 S ribosomal subunits and interacts with other initiation factors involved in start codon selection. The molecular mechanisms by which eIF3 exerts these functions are poorly understood. Since its initial characterization in the 1970s, the exact size, composition, and post-translational modifications of mammalian eIF3 have not been rigorously determined. Two powerful mass spectrometric approaches were used in the present study to determine post-translational modifications that may regulate the activity of eIF3 during the translation initiation process and to characterize the molecular structure of the human eIF3 protein complex purified from HeLa cells. In the first approach, the bottom-up analysis of eIF3 allowed for the identification of a total of 13 protein components (eIF3a-m) with a sequence coverage of approximately 79%. Furthermore 29 phosphorylation sites and several other post-translational modifications were unambiguously identified within the eIF3 complex. The second mass spectrometric approach, involving analysis of intact eIF3, allowed the detection of a complex with each of the 13 subunits present in stoichiometric amounts. Using tandem mass spectrometry four eIF3 subunits (h, i, k, and m) were found to be most easily dissociated and therefore likely to be on the periphery of the complex. It is noteworthy that none of these four subunits were found to be phosphorylated. These data raise interesting questions about the function of phosphorylation as it relates to the core subunits of the complex.


Subject(s)
Eukaryotic Initiation Factor-3/chemistry , Protein Biosynthesis , Protein Processing, Post-Translational , Chromatography, Liquid , Eukaryotic Initiation Factor-3/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Subunits/chemistry , Protein Subunits/metabolism , Tandem Mass Spectrometry/methods
3.
J Biol Chem ; 280(11): 10827-33, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15640154

ABSTRACT

The androgen receptor (AR) dynamically assembles and disassembles multicomponent receptor complexes in order to respond rapidly and reversibly to fluctuations in androgen levels. We are interested in identifying the basal factors that compose the AR aporeceptor and holoreceptor complexes and impact the transcriptional process. Using tandem mass spectroscopy analysis, we identified the trimeric DNA-dependent protein kinase (DNA-PK) complex as the major AR-interacting proteins. AR directly interacts with both Ku70 and Ku80 in vivo and in vitro, as shown by co-immunoprecipitation, glutathione S-transferase pull-down, and Sf9 cell/baculovirus expression. The interaction was localized to the androgen receptor ligand binding domain and is independent of DNA interactions. Ku interacts with AR in the cytoplasm and nucleus regardless of the presence or absence of androgen. Ku acts as a coactivator of AR activity in a luciferase reporter assay employing both Ku-defective cells and Ku small interfering RNA knock-down in a prostate cancer cell line. DNA-PK catalytic subunit (DNA-PKcs) also acts as a coactivator of androgen receptor activity in a luciferase reporter assay employing DNA-PKcs defective cells. AR nuclear translocation is not affected in Ku defective cells, implying Ku functionality may be mainly nuclear. Chromatin immunoprecipitation experiments demonstrated that both Ku70 and Ku80 interact with the prostate-specific antigen promoter in an androgen-dependant manner. Finally, in vitro transcription assays demonstrated Ku involvement in transcriptional recycling with androgen dependent promoters.


Subject(s)
Antigens, Nuclear/physiology , DNA-Binding Proteins/physiology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic , Androgens/metabolism , Animals , Antigens, Nuclear/chemistry , Antigens, Nuclear/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin Immunoprecipitation , Cytoplasm/metabolism , DNA/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Genes, Reporter , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Insecta , Ku Autoantigen , Ligands , Luciferases/metabolism , Male , Mass Spectrometry , Models, Genetic , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
4.
EMBO J ; 23(8): 1761-9, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15071500

ABSTRACT

The eucaryotic translation initiation factor 4B (eIF4B) stimulates the helicase activity of the DEAD box protein eIF4A to unwind inhibitory secondary structure in the 5' untranslated region of eucaryotic mRNAs. Here, using phosphopeptide mapping and a phosphospecific antiserum, we identify a serum-responsive eIF4B phosphorylation site, Ser422, located in an RNA-binding region required for eIF4A helicase-promoting activity. Ser422 phosphorylation appears to be regulated by the S6Ks: (a) Ser422 phosphorylation is sensitive to pharmacological inhibitors of phosphoinositide-3 kinase and the mammalian target of rapamycin; (b) S6K1/S6K2 specifically phosphorylate Ser422 in vitro; and (c) rapamycin-resistant S6Ks confer rapamycin resistance upon Ser422 phosphorylation in vivo. Substitution of Ser422 with Ala results in a loss of activity in an in vivo translation assay, indicating that phosphorylation of this site plays an important role in eIF4B function. We therefore propose that eIF4B may mediate some of the effects of the S6Ks on translation.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Phosphoserine/metabolism , Ribosomal Protein S6 Kinases/metabolism , Cell Line , Drug Resistance , Eukaryotic Initiation Factors/genetics , Humans , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinases/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
5.
J Biol Chem ; 279(10): 8946-56, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14688252

ABSTRACT

Eukaryotic initiation factor 3 (eIF3) is a 12-subunit protein complex that plays a central role in binding of initiator methionyl-tRNA and mRNA to the 40 S ribosomal subunit to form the 40 S initiation complex. The molecular mechanisms by which eIF3 exerts these functions are poorly understood. To learn more about the structure and function of eIF3 we have expressed and purified individual human eIF3 subunits or complexes of eIF3 subunits using baculovirus-infected Sf9 cells. The results indicate that the subunits of human eIF3 that have homologs in Saccharomyces cerevisiae form subcomplexes that reflect the subunit interactions seen in the yeast eIF3 core complex. In addition, we have used an in vitro 40 S ribosomal subunit binding assay to investigate subunit requirements for efficient association of the eIF3 subcomplexes to the 40 S ribosomal subunit. eIF3j alone binds to the 40 S ribosomal subunit, and its presence is required for stable 40 S binding of an eIF3bgi subcomplex. Furthermore, purified eIF3 lacking eIF3j binds 40 S ribosomal subunits weakly, but binds tightly when eIF3j is added. Cleavage of a 16-residue C-terminal peptide from eIF3j by caspase-3 significantly reduces the affinity of eIF3j for the 40 S ribosomal subunit, and the cleaved form provides substantially less stabilization of purified eIF3-40S complexes. These results indicate that eIF3j, and especially its C terminus, play an important role in the recruitment of eIF3 to the 40 S ribosomal subunit.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Ribosomes/metabolism , Animals , Binding Sites , Cell Line , Eukaryotic Initiation Factor-3/analysis , Humans , Protein Binding , Protein Subunits
6.
Eur J Biochem ; 270(20): 4133-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519125

ABSTRACT

Mammalian translation initiation factor 3 (eIF3) is a multisubunit complex containing at least 12 subunits with an apparent aggregate mass of approximately 700 kDa. eIF3 binds to the 40S ribosomal subunit, promotes the binding of methionyl-tRNAi and mRNA, and interacts with several other initiation factors to form the 40S initiation complex. Human cDNAs encoding 11 of the 12 subunits have been isolated previously; here we report the cloning and characterization of a twelfth subunit, a 28-kDa protein named eIF3k. Evidence that eIF3k is present in the eIF3 complex was obtained. A monoclonal anti-eIF3a (p170) Ig coimmunoprecipitates eIF3k with the eIF3 complex. Affinity purification of histidine-tagged eIF3k from transiently transfected COS cells copurifies other eIF3 subunits. eIF3k colocalizes with eIF3 on 40S ribosomal subunits. eIF3k coexpressed with five other 'core' eIF3 subunits in baculovirus-infected insect cells, forms a stable, immunoprecipitatable, complex with the 'core'. eIF3k interacts directly with eIF3c, eIF3g and eIF3j by glutathione S-transferase pull-down assays. Sequences homologous with eIF3k are found in the genomes of Caenorhabitis elegans, Arabidopsis thaliana and Drosophila melanogaster, and a homologous protein has been reported to be present in wheat eIF3. Its ubiquitous expression in human tissues, yet its apparent absence in Saccharomyces cerevisiae and Schizosaccharomyces pombe, suggest a unique regulatory role for eIF3k in higher organisms. The studies of eIF3k complete the characterization of mammalian eIF3 subunits.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , DNA, Complementary/metabolism , Eukaryotic Initiation Factor-3/genetics , Eukaryotic Initiation Factor-3/isolation & purification , HeLa Cells , Humans , Protein Structure, Quaternary , Ribosomes
7.
J Virol ; 77(17): 9652-61, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12915577

ABSTRACT

In order to cope with hostile host environments, many viruses have developed strategies to perturb the cellular machinery to suit their replication needs. Some herpesvirus genes protect cells from undergoing apoptosis to prolong the lives of infected cells, while others, such as Epstein-Barr virus Zta, slow down the G(1)/S transition phase to allow ample opportunity for transcription and translation of viral genes before the onset of cellular genomic replication. In this study, we investigated whether Kaposi's sarcoma-associated herpesvirus (KSHV) K-bZIP, a homologue of the Epstein-Barr virus transcription factor BZLF1 (Zta), plays a role in cell cycle regulation. Here we show that K-bZIP physically associates with cyclin-CDK2 and downmodulates its kinase activity. The association can be detected in the natural environment of KSHV-infected cells without artificial overexpression of either component. With purified protein, it can be shown that the interaction between K-bZIP and cyclin-CDK2 is direct and that K-bZIP alone is sufficient to inhibit CDK2 activity. The interacting domain of K-bZIP has been mapped to the basic region. The result of these associations is a prolonged G(1) phase, accompanied by the induction of p21 and p27 in a naturally infected B-cell line. Thus, in addition to the previously described transcription and genome replication functions, a new role of K-bZIP in KSHV replication is identified in this report.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins/physiology , Cell Cycle/physiology , Cyclin-Dependent Kinases/physiology , Herpesvirus 8, Human/physiology , Protein Serine-Threonine Kinases/physiology , Viral Proteins/physiology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Basic-Leucine Zipper Transcription Factors , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cyclin-Dependent Kinase 2 , G1 Phase/physiology , Gene Expression , Genes, Viral , HeLa Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins , Sequence Deletion , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/physiology
8.
FEBS Lett ; 514(1): 49-54, 2002 Mar 06.
Article in English | MEDLINE | ID: mdl-11904180

ABSTRACT

Several components of translation, e.g. eIF4E and PKR, are implicated in cancer. The e-subunit (p48) of mammalian initiation factor 3 is encoded by the Int6 gene, a common site for integration of the mouse mammary tumor virus genome, leading to the production of a truncated eukaryotic initiation factor-3e (eIF3e). Stable expression of a truncated eIF3e in NIH 3T3 cells causes malignant transformation by four criteria: foci formation; anchorage independent growth; accelerated growth; and lack of contact inhibition. Stable expression of full-length eIF3e does not cause transformation. The truncated eIF3e also inhibits the onset of apoptosis caused by serum starvation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Peptide Initiation Factors/genetics , Proto-Oncogene Proteins/genetics , 3T3 Cells , Animals , Eukaryotic Initiation Factor-3 , Gene Deletion , Mice
SELECTION OF CITATIONS
SEARCH DETAIL