Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
STAR Protoc ; 3(4): 101739, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36181679

ABSTRACT

eIF5-mimic protein (5MP) controls translation through binding to the ribosomal pre-initiation complex (PIC) and alters non-AUG translation rates for cancer oncogenes and repeat-expansions in neurodegenerative diseases. Here, we describe a semi-quantitative protocol for detecting 5MP-associated proteins in cultured human and fly cells. We detail one-step anti-FLAG affinity purification and whole-lane mass spectrometry analysis of samples resolved by SDS-PAGE. This protocol allows for quantitative evaluation of the effect of 5MP mutations on its molecular interactions, to elucidate translational control by 5MP. For complete details on the use and execution of this protocol, please refer to Singh et al. (2021).


Subject(s)
Protein Biosynthesis , Ribosomes , Humans , Ribosomes/metabolism , Proteins/metabolism , Cytosol/metabolism , Mass Spectrometry
2.
STAR Protoc ; 3(3): 101615, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36035794

ABSTRACT

eIF5-mimic protein (5MP) controls translation through its interaction with eukaryotic translation initiation factor (eIF) 2 and eIF3 and alters non-AUG translation rates for oncogenes in cancer and repeat expansions in neurodegenerative disease. To precisely evaluate the effect of 5MP mutations on binding affinity against eIFs, here we describe two label-free protocols of affinity measurement for 5MP binding to eIF2 or eIF3 protein segments, termed isothermal titration calorimetry (ITC) and bio-layer interferometry (BLI), starting with how to purify proteins used. For complete details on the use and execution of this protocol, please refer to Singh et al. (2021).


Subject(s)
Eukaryotic Initiation Factor-5 , Neurodegenerative Diseases , Calorimetry , Eukaryotic Initiation Factor-2 , Eukaryotic Initiation Factor-3 , Humans , Interferometry
3.
Sci Adv ; 8(14): eabm8501, 2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35394828

ABSTRACT

In contrast to prokaryotes wherein GUG and UUG are permissive start codons, initiation frequencies from non-AUG codons are generally low in eukaryotes, with CUG being considered as strongest. Here, we report that combined 5-cytosine methylation (5mC) and pseudouridylation (Ψ) of near-cognate non-AUG start codons convert GUG and UUG initiation strongly favored over CUG initiation in eukaryotic translation under a certain context. This prokaryotic-like preference is attributed to enhanced NUG initiation by Ψ in the second base and reduced CUG initiation by 5mC in the first base. Molecular dynamics simulation analysis of tRNAiMet anticodon base pairing to the modified codons demonstrates that Ψ universally raises the affinity of codon:anticodon pairing within the ribosomal preinitiation complex through partially mitigating discrimination against non-AUG codons imposed by eukaryotic initiation factor 1. We propose that translational control by chemical modifications of start codon bases can offer a new layer of proteome diversity regulation and therapeutic mRNA technology.

4.
Cell Rep ; 36(2): 109376, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260931

ABSTRACT

eIF5-mimic protein (5MP) is a translational regulatory protein that binds the small ribosomal subunit and modulates its activity. 5MP is proposed to reprogram non-AUG translation rates for oncogenes in cancer, but its role in controlling non-AUG initiated synthesis of deleterious repeat-peptide products, such as FMRpolyG observed in fragile-X-associated tremor ataxia syndrome (FXTAS), is unknown. Here, we show that 5MP can suppress both general and repeat-associated non-AUG (RAN) translation by a common mechanism in a manner dependent on its interaction with eIF3. Essentially, 5MP displaces eIF5 through the eIF3c subunit within the preinitiation complex (PIC), thereby increasing the accuracy of initiation. In Drosophila, 5MP/Kra represses neuronal toxicity and enhances the lifespan in an FXTAS disease model. These results implicate 5MP in protecting cells from unwanted byproducts of non-AUG translation in neurodegeneration.


Subject(s)
Codon, Initiator/genetics , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-3/metabolism , Protein Biosynthesis/genetics , Trinucleotide Repeat Expansion/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Differentiation/metabolism , DNA-Binding Proteins/chemistry , Drosophila/metabolism , Drosophila Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-3/chemistry , HEK293 Cells , Humans , Male , Models, Biological , Models, Molecular , Mutation/genetics , Peptide Chain Initiation, Translational , Protein Binding , Protein Domains , Receptors, Immunologic/metabolism
5.
Nucleic Acids Res ; 48(16): 8977-8992, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32710633

ABSTRACT

The protein kinase Gcn2 is a central transducer of nutritional stress signaling important for stress adaptation by normal cells and the survival of cancer cells. In response to nutrient deprivation, Gcn2 phosphorylates eIF2α, thereby repressing general translation while enhancing translation of specific mRNAs with upstream ORFs (uORFs) situated in their 5'-leader regions. Here we performed genome-wide measurements of mRNA translation during histidine starvation in fission yeast Schizosaccharomyces pombe. Polysome analyses were combined with microarray measurements to identify gene transcripts whose translation was up-regulated in response to the stress in a Gcn2-dependent manner. We determined that translation is reprogrammed to enhance RNA metabolism and chromatin regulation and repress ribosome synthesis. Interestingly, translation of intron-containing mRNAs was up-regulated. The products of the regulated genes include additional eIF2α kinase Hri2 amplifying the stress signaling and Gcn5 histone acetyl transferase and transcription factors, together altering genome-wide transcription. Unique dipeptide-coding uORFs and nucleotide motifs, such as '5'-UGA(C/G)GG-3', are found in 5' leader regions of regulated genes and shown to be responsible for translational control.


Subject(s)
Nucleotide Motifs , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Acetyltransferases/metabolism , Gene Expression Regulation, Fungal , Histidine/metabolism , Open Reading Frames , Protein Processing, Post-Translational , Schizosaccharomyces/genetics , eIF-2 Kinase/metabolism
6.
Mol Cell Biol ; 38(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29844065

ABSTRACT

Ribosomal stalk proteins recruit translation elongation GTPases to the factor-binding center of the ribosome. Initiation factor 5B (eIF5B in eukaryotes and aIF5B in archaea) is a universally conserved GTPase that promotes the joining of the large and small ribosomal subunits during translation initiation. Here we show that aIF5B binds to the C-terminal tail of the stalk protein. In the cocrystal structure, the interaction occurs between the hydrophobic amino acids of the stalk C-terminal tail and a small hydrophobic pocket on the surface of the GTP-binding domain (domain I) of aIF5B. A substitution mutation altering the hydrophobic pocket of yeast eIF5B resulted in a marked reduction in ribosome-dependent eIF5B GTPase activity in vitro In yeast cells, the eIF5B mutation affected growth and impaired GCN4 expression during amino acid starvation via a defect in start site selection for the first upstream open reading frame in GCN4 mRNA, as observed with the eIF5B deletion mutant. The deletion of two of the four stalk proteins diminished polyribosome levels (indicating defective translation initiation) and starvation-induced GCN4 expression, both of which were suppressible by eIF5B overexpression. Thus, the mutual interaction between a/eIF5B and the ribosomal stalk plays an important role in subunit joining during translation initiation in vivo.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Ribosomal Proteins/metabolism , Aeropyrum/genetics , Aeropyrum/metabolism , Amino Acid Substitution , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , Models, Molecular , Mutation , Peptide Chain Initiation, Translational , Phenotype , Protein Interaction Domains and Motifs , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Nucleic Acids Res ; 45(20): 11941-11953, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28981728

ABSTRACT

In the human genome, translation initiation from non-AUG codons plays an important role in various gene regulation programs. However, mechanisms regulating the non-AUG initiation rate remain poorly understood. Here, we show that the non-AUG initiation rate is nearly consistent under a fixed nucleotide context in various human and insect cells. Yet, it ranges from <1% to nearly 100% compared to AUG translation, depending on surrounding sequences, including Kozak, and possibly additional nucleotide contexts. Mechanistically, this range of non-AUG initiation is controlled in part, by the eIF5-mimic protein (5MP). 5MP represses non-AUG translation by competing with eIF5 for the Met-tRNAi-binding factor eIF2. Consistently, eIF5 increases, whereas 5MP decreases translation of NAT1/EIF4G2/DAP5, whose sole start codon is GUG. By modulating eIF5 and 5MP1 expression in combination with ribosome profiling we identified a handful of previously unknown non-AUG initiation sites, some of which serve as the exclusive start codons. If the initiation rate for these codons is low, then an AUG-initiated downstream ORF prevents the generation of shorter, AUG-initiated isoforms. We propose that the homeostasis of the non-AUG translatome is maintained through balanced expression of eIF5 and 5MP.


Subject(s)
Codon, Initiator/genetics , DNA-Binding Proteins/genetics , Eukaryotic Initiation Factor-5/genetics , Genome, Human , Animals , Binding, Competitive , Cell Line , Cell Line, Tumor , Codon, Initiator/metabolism , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-5/metabolism , Gene Expression Regulation , HEK293 Cells , Homeostasis/genetics , Humans , Protein Binding , Protein Biosynthesis/genetics , Ribosomes/genetics , Ribosomes/metabolism
8.
Cell Rep ; 18(11): 2651-2663, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297669

ABSTRACT

During eukaryotic translation initiation, eIF3 binds the solvent-accessible side of the 40S ribosome and recruits the gate-keeper protein eIF1 and eIF5 to the decoding center. This is largely mediated by the N-terminal domain (NTD) of eIF3c, which can be divided into three parts: 3c0, 3c1, and 3c2. The N-terminal part, 3c0, binds eIF5 strongly but only weakly to the ribosome-binding surface of eIF1, whereas 3c1 and 3c2 form a stoichiometric complex with eIF1. 3c1 contacts eIF1 through Arg-53 and Leu-96, while 3c2 faces 40S protein uS15/S13, to anchor eIF1 to the scanning pre-initiation complex (PIC). We propose that the 3c0:eIF1 interaction diminishes eIF1 binding to the 40S, whereas 3c0:eIF5 interaction stabilizes the scanning PIC by precluding this inhibitory interaction. Upon start codon recognition, interactions involving eIF5, and ultimately 3c0:eIF1 association, facilitate eIF1 release. Our results reveal intricate molecular interactions within the PIC, programmed for rapid scanning-arrest at the start codon.


Subject(s)
Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-5/metabolism , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Binding Sites , Eukaryotic Initiation Factor-1/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Mutation/genetics , Protein Binding , Protein Subunits/metabolism , RNA, Messenger/genetics , Saccharomyces cerevisiae Proteins/metabolism
9.
Nucleic Acids Res ; 44(18): 8704-8713, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27325740

ABSTRACT

ATF4 is a pro-oncogenic transcription factor whose translation is activated by eIF2 phosphorylation through delayed re-initiation involving two uORFs in the mRNA leader. However, in yeast, the effect of eIF2 phosphorylation can be mimicked by eIF5 overexpression, which turns eIF5 into translational inhibitor, thereby promoting translation of GCN4, the yeast ATF4 equivalent. Furthermore, regulatory protein termed eIF5-mimic protein (5MP) can bind eIF2 and inhibit general translation. Here, we show that 5MP1 overexpression in human cells leads to strong formation of 5MP1:eIF2 complex, nearly comparable to that of eIF5:eIF2 complex produced by eIF5 overexpression. Overexpression of eIF5, 5MP1 and 5MP2, the second human paralog, promotes ATF4 expression in certain types of human cells including fibrosarcoma. 5MP overexpression also induces ATF4 expression in Drosophila The knockdown of 5MP1 in fibrosarcoma attenuates ATF4 expression and its tumor formation on nude mice. Since 5MP2 is overproduced in salivary mucoepidermoid carcinoma, we propose that overexpression of eIF5 and 5MP induces translation of ATF4 and potentially other genes with uORFs in their mRNA leaders through delayed re-initiation, thereby enhancing the survival of normal and cancer cells under stress conditions.


Subject(s)
Activating Transcription Factor 4/metabolism , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-5/metabolism , Peptide Chain Initiation, Translational , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Drosophila melanogaster/metabolism , Eukaryotic Initiation Factor-3 , Fibrosarcoma/pathology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Male , Mass Spectrometry , Mice, Nude
10.
Anticancer Res ; 34(4): 1643-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24692693

ABSTRACT

BACKGROUND: Synthetic 6,7-annulated-4-substituted indole compounds, which elicit interesting antitumor effects in murine L1210 leukemia cells, were tested for their ability to inhibit human HL-60 tumor cell proliferation, disrupt mitosis and cytokinesis, and interfere with tubulin and actin polymerization in vitro. MATERIALS AND METHODS: Various markers of metabolic activity, mitotic disruption and cytokinesis were used to assess the effectiveness of the drugs in the HL-60 tumor cell system. The ability of annulated indoles to alter the polymerizations of purified tubulin and actin were monitored in cell-free assays and were compared to the effects of drugs known to disrupt the dynamic structures of the mitotic spindle and cleavage furrow. RESULTS: With one exception, annulated indoles inhibited the metabolic activity of HL-60 tumor cells in the low-micromolar range after two and four days in culture but these anti-proliferative effects were weaker than those of jasplakinolide, a known actin binder that blocks cytokinesis. After 24-48 h, antiproliferative concentrations of annulated indoles increased the mitotic index of HL-60 cells similarly to vincristine and stimulated the formation of many bi-nucleated cells, multi-nucleated cells and micronuclei, similarly to taxol and jasplakinolide, suggesting that these antitumor compounds might increase mitotic abnormality, induce chromosomal damage or missegregation, and block cytokinesis. Since annulated indoles mimicked the effect of vincristine on tubulin polymerization, but not that of taxol, these compounds might represent a new class of microtubule de-stabilizing agents that inhibit tubulin polymerization. Moreover, annulated indoles remarkably increased the rate and level of actin polymerization similarly to jasplakinolide, suggesting that they might also stabilize the cleavage furrow to block cytokinesis. CONCLUSION: Although novel derivatives with different substitutions must be synthesized to elucidate structure-activity relationships, identify more potent antitumor compounds and investigate different molecular targets, annulated indoles appear to interact with both tubulin to reduce microtubule assembly and actin to block cytokinesis, thereby inducing bi- and multinucleation, resulting in genomic instability and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Cytokinesis/drug effects , Indoles/pharmacology , Mitosis/drug effects , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Indoles/chemistry , Protein Multimerization/drug effects , Tubulin/metabolism
11.
J Biol Chem ; 288(41): 29467-81, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23995839

ABSTRACT

Several proteasome-associated proteins regulate degradation by the 26 S proteasome using the ubiquitin chains that mark most substrates for degradation. The proteasome-associated protein Ecm29, however, has no ubiquitin-binding or modifying activity, and its direct effect on substrate degradation is unclear. Here, we show that Ecm29 acts as a proteasome inhibitor. Besides inhibiting the proteolytic cleavage of peptide substrates in vitro, it inhibits the degradation of ubiquitin-dependent and -independent substrates in vivo. Binding of Ecm29 to the proteasome induces a closed conformation of the substrate entry channel of the core particle. Furthermore, Ecm29 inhibits proteasomal ATPase activity, suggesting that the mechanism of inhibition and gate regulation by Ecm29 is through regulation of the proteasomal ATPases. Consistent with this, we identified through chemical cross-linking that Ecm29 binds to, or in close proximity to, the proteasomal ATPase subunit Rpt5. Additionally, we show that Ecm29 preferentially associates with both mutant and nucleotide depleted proteasomes. We propose that the inhibitory ability of Ecm29 is important for its function as a proteasome quality control factor by ensuring that aberrant proteasomes recognized by Ecm29 are inactive.


Subject(s)
Adenosine Triphosphatases/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/genetics , Immunoblotting , Mutation , Proteasome Endopeptidase Complex/genetics , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Ubiquitinated Proteins/metabolism
12.
Nature ; 497(7450): 512-6, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23644457

ABSTRACT

The proteasomal ATPase ring, comprising Rpt1-Rpt6, associates with the heptameric α-ring of the proteasome core particle (CP) in the mature proteasome, with the Rpt carboxy-terminal tails inserting into pockets of the α-ring. Rpt ring assembly is mediated by four chaperones, each binding a distinct Rpt subunit. Here we report that the base subassembly of the Saccharomyces cerevisiae proteasome, which includes the Rpt ring, forms a high-affinity complex with the CP. This complex is subject to active dissociation by the chaperones Hsm3, Nas6 and Rpn14. Chaperone-mediated dissociation was abrogated by a non-hydrolysable ATP analogue, indicating that chaperone action is coupled to nucleotide hydrolysis by the Rpt ring. Unexpectedly, synthetic Rpt tail peptides bound α-pockets with poor specificity, except for Rpt6, which uniquely bound the α2/α3-pocket. Although the Rpt6 tail is not visualized within an α-pocket in mature proteasomes, it inserts into the α2/α3-pocket in the base-CP complex and is important for complex formation. Thus, the Rpt-CP interface is reconfigured when the lid complex joins the nascent proteasome to form the mature holoenzyme.


Subject(s)
Molecular Chaperones/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Carrier Proteins/metabolism , Cryoelectron Microscopy , Holoenzymes/chemistry , Holoenzymes/metabolism , Models, Molecular , Proteasome Endopeptidase Complex/genetics , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
13.
Translation (Austin) ; 1(2): e26402, 2013.
Article in English | MEDLINE | ID: mdl-26824023

ABSTRACT

In yeast Saccharomyces cerevisiae, 25S rRNA makes up the major mass and shape of the 60S ribosomal subunit. During translation initiation, the 60S subunit joins the 40S initiation complex, producing the 80S initiation complex. During elongation, the 60S subunit binds the CCA-ends of aminoacyl- and peptidyl-tRNAs at the A-loop and P-loop, respectively, transferring the peptide onto the α-amino group of the aminoacyl-tRNA. To study the role of 25S rRNA in translation in vivo, we randomly mutated 25S rRNA and isolated and characterized seven point mutations that affected yeast cell growth and polysome profiles. Four of these mutations, G651A, A1435U, A1446G and A1587G, change a base involved in base triples crucial for structural integrity. Three other mutations change bases near the ribosomal surface: C2879U and U2408C alter the A-loop and P-loop, respectively, and G1735A maps near a Eukarya-specific bridge to the 40S subunit. By polysome profiling in mmslΔ mutants defective in nonfunctional 25S rRNA decay, we show that some of these mutations are defective in both the initiation and elongation phases of translation. Of the mutants characterized, C2879U displays the strongest defect in translation initiation. The ribosome transit-time assay directly shows that this mutation is also defective in peptide elongation/termination. Thus, our genetic analysis not only identifies bases critical for structural integrity of the 60S subunit, but also suggests a role for bases near the peptidyl transferase center in translation initiation.

14.
Mol Cell Biol ; 32(19): 3978-89, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851688

ABSTRACT

During translation initiation in Saccharomyces cerevisiae, an Arg- and Ser-rich segment (RS1 domain) of eukaryotic translation initiation factor 4G (eIF4G) and the Lys-rich segment (K-boxes) of eIF2ß bind three common partners, eIF5, eIF1, and mRNA. Here, we report that both of these segments are involved in mRNA recruitment and AUG recognition by distinct mechanisms. First, the eIF4G-RS1 interaction with the eIF5 C-terminal domain (eIF5-CTD) directly links eIF4G to the preinitiation complex (PIC) and enhances mRNA binding. Second, eIF2ß-K-boxes increase mRNA binding to the 40S subunit in vitro in a manner reversed by the eIF5-CTD. Third, mutations altering eIF4G-RS1, eIF2ß-K-boxes, and eIF5-CTD restore the accuracy of start codon selection impaired by an eIF2ß mutation in vivo, suggesting that the mutual interactions of the eIF segments within the PIC prime the ribosome for initiation in response to start codon selection. We propose that the rearrangement of interactions involving the eIF5-CTD promotes mRNA recruitment through mRNA binding by eIF4G and eIF2ß and assists the start codon-induced release of eIF1, the major antagonist of establishing tRNA(i)(Met):mRNA binding to the P site.


Subject(s)
Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-5/metabolism , RNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-5/chemistry , Eukaryotic Initiation Factor-5/genetics , Gene Expression Regulation, Fungal , Mutation , Protein Structure, Tertiary , RNA, Fungal/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
15.
Nucleic Acids Res ; 39(19): 8314-28, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745818

ABSTRACT

The translation factor eIF5 is an important partner of eIF2, directly modulating its function in several critical steps. First, eIF5 binds eIF2/GTP/Met-tRNA(i)(Met) ternary complex (TC), promoting its recruitment to 40S ribosomal subunits. Secondly, its GTPase activating function promotes eIF2 dissociation for ribosomal subunit joining. Finally, eIF5 GDP dissociation inhibition (GDI) activity can antagonize eIF2 reactivation by competing with the eIF2 guanine exchange factor (GEF), eIF2B. The C-terminal domain (CTD) of eIF5, a W2-type HEAT domain, mediates its interaction with eIF2. Here, we characterize a related human protein containing MA3- and W2-type HEAT domains, previously termed BZW2 and renamed here as eIF5-mimic protein 1 (5MP1). Human 5MP1 interacts with eIF2 and eIF3 and inhibits general and gene-specific translation in mammalian systems. We further test whether 5MP1 is a mimic or competitor of the GEF catalytic subunit eIF2Bε or eIF5, using yeast as a model. Our results suggest that 5MP1 interacts with yeast eIF2 and promotes TC formation, but inhibits TC binding to the ribosome. Moreover, 5MP1 is not a GEF but a weak GDI for yeast eIF2. We propose that 5MP1 is a partial mimic and competitor of eIF5, interfering with the key steps by which eIF5 regulates eIF2 function.


Subject(s)
DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Protein Biosynthesis , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-2/analysis , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-3/metabolism , Guanosine Diphosphate/metabolism , HeLa Cells , Humans , Mice , Molecular Mimicry , Peptide Initiation Factors/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
16.
J Biol Chem ; 285(42): 32200-12, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20699223

ABSTRACT

In eukaryotes, the 40 S ribosomal subunit serves as the platform of initiation factor assembly, to place itself precisely on the AUG start codon. Structural arrangement of the 18 S rRNA determines the overall shape of the 40 S subunit. Here, we present genetic evaluation of yeast 18 S rRNA function using 10 point mutations altering the polysome profile. All the mutants reduce the abundance of the mutant 40 S, making it limiting for translation initiation. Two of the isolated mutations, G875A, altering the core of the platform domain that binds eIF1 and eIF2, and A1193U, changing the h31 loop located below the P-site tRNA(i)(Met), show phenotypes indicating defective regulation of AUG selection. Evidence is provided that these mutations reduce the interaction with the components of the preinitiation complex, thereby inhibiting its function at different steps. These results indicate that the 18 S rRNA mutations impair the integrity of scanning-competent preinitiation complex, thereby altering the 40 S subunit response to stringent AUG selection. Interestingly, nine of the mutations alter the body/platform domains of 18 S rRNA, potentially affecting the bridges to the 60 S subunit, but they do not change the level of 18 S rRNA intermediates. Based on these results, we also discuss the mechanism of the selective degradation of the mutant 40 S subunits.


Subject(s)
Codon, Initiator/metabolism , Nucleic Acid Conformation , Protein Biosynthesis , Protein Subunits/metabolism , RNA, Fungal , RNA, Ribosomal, 18S , Ribosome Subunits, Small, Eukaryotic , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Point Mutation , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , RNA, Ribosomal, 18S/chemistry , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
17.
J Biol Chem ; 285(29): 21922-33, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20463023

ABSTRACT

Translation re-initiation provides the molecular basis for translational control of mammalian ATF4 and yeast GCN4 mediated by short upstream open reading (uORFs) in response to eIF2 phosphorylation. eIF4G is the major adaptor subunit of eIF4F that binds the cap-binding subunit eIF4E and the mRNA helicase eIF4A and is also required for re-initiation in mammals. Here we show that the yeast eIF4G2 mutations altering eIF4E- and eIF4A-binding sites increase re-initiation at GCN4 and impair recognition of the start codons of uORF1 or uORF4 located after uORF1. The increase in re-initiation at GCN4 was partially suppressed by increasing the distance between uORF1 and GCN4, suggesting that the mutations decrease the migration rate of the scanning ribosome in the GCN4 leader. Interestingly, eIF4E overexpression suppressed both the phenotypes caused by the mutation altering eIF4E-binding site. Thus, eIF4F is required for accurate AUG selection and re-initiation also in yeast, and the eIF4G interaction with the mRNA-cap appears to promote eIF4F re-acquisition by the re-initiating 40 S subunit. However, eIF4A overexpression suppressed the impaired AUG recognition but not the increase in re-initiation caused by the mutations altering eIF4A-binding site. These results not only provide evidence that mRNA unwinding by eIF4A stimulates start codon recognition, but also suggest that the eIF4A-binding site on eIF4G made of the HEAT domain stimulates the ribosomal scanning independent of eIF4A. Based on the RNA-binding activities identified within the unstructured segments flanking the eIF4G2 HEAT domain, we discuss the role of the HEAT domain in scanning beyond loading eIF4A onto the pre-initiation complex.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Peptide Chain Initiation, Translational , Saccharomyces cerevisiae/enzymology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Codon, Initiator/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Fungal , Models, Biological , Mutation/genetics , Open Reading Frames/genetics , Peptides , Protein Binding , Protein Structure, Tertiary , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
18.
J Biol Chem ; 283(2): 1094-103, 2008 Jan 11.
Article in English | MEDLINE | ID: mdl-17974565

ABSTRACT

Eukaryotic initiation factor (eIF) 1 is a small protein (12 kDa) governing fidelity in translation initiation. It is recruited to the 40 S subunit in a multifactor complex with Met-tRNA(i)(Met), eIF2, eIF3, and eIF5 and binds near the P-site. eIF1 release in response to start codon recognition is an important signal to produce an 80 S initiation complex. Although the ribosome-binding face of eIF1 was identified, interfaces to other preinitiation complex components and their relevance to eIF1 function have not been determined. Exploiting the solution structure of yeast eIF1, here we locate the binding site for eIF5 in its N-terminal tail and at a basic/hydrophobic surface area termed KH, distinct from the ribosome-binding face. Genetic and biochemical studies indicate that the eIF1 N-terminal tail plays a stimulatory role in cooperative multifactor assembly. A mutation altering the basic part of eIF1-KH is lethal and shows a dominant phenotype indicating relaxed start codon selection. Cheung et al. recently demonstrated that the alteration of hydrophobic residues of eIF1 disrupts a critical link to the preinitiation complex that suppresses eIF1 release before start codon selection (Cheung, Y.-N., Maag, D., Mitchell, S. F., Fekete, C. A., Algire, M. A., Takacs, J. E., Shirokikh, N., Pestova, T., Lorsch, J. R., and Hinnebusch, A. (2007) Genes Dev. 21, 1217-1230 ). Interestingly, eIF1-KH includes the altered hydrophobic residues. Thus, eIF5 is an excellent candidate for the direct partner of eIF1-KH that mediates the critical link. The direct interaction at eIF1-KH also places eIF5 near the decoding site of the 40 S subunit.


Subject(s)
Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factor-5/chemistry , Eukaryotic Initiation Factor-5/metabolism , Amino Acid Sequence , Binding Sites , Conserved Sequence , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-5/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Plasmids , Protein Conformation , Saccharomyces cerevisiae/genetics , Solutions
19.
Methods Enzymol ; 429: 105-37, 2007.
Article in English | MEDLINE | ID: mdl-17913621

ABSTRACT

This chapter describes phenotypic assays on specific and general aspects of translation using yeast Saccharomyces cerevisiae as a model eukaryote. To study the effect on start codon selection stringency, a his4(-) or his4-lacZ allele altering the first AUG to AUU is employed. Mutations relaxing the stringent selection confer the His(+) phenotype in the his4(-) strain background or increase expression from his4-lacZ compared to that from wild-type HIS4-lacZ (Sui(-) phenotype). Translation of the Gcn4p transcription activator is strictly regulated by amino acid availability depending on upstream ORF (uORF) elements in the GCN4 mRNA leader. Mutations reducing the eIF2/GTP/Met-tRNA(i)(Met) complex level or the rate of its binding to the 40S subunit derepress GCN4 translation by allowing ribosomes to bypass inhibitory uORFs in the absence of the starvation signal (Gcd(-) phenotype). Mutations impairing scanning or AUG recognition generally impair translational GCN4 induction during amino acid starvation (Gcn(-) phenotype). Different amino acid analogs or amino acid enzyme inhibitors are used to study Gcd(-) or Gcn(-) phenotypes. The method of polysome profiling is also described to gain an ultimate "phenotypic" proof for translation defects.


Subject(s)
Peptide Chain Initiation, Translational/physiology , Protein Biosynthesis/physiology , Saccharomyces cerevisiae/metabolism , Basic-Leucine Zipper Transcription Factors , Centrifugation, Density Gradient , Codon, Initiator , DNA-Binding Proteins/physiology , Genetic Vectors , Phenotype , Polyribosomes/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/physiology , Transformation, Genetic
20.
Methods Enzymol ; 429: 139-61, 2007.
Article in English | MEDLINE | ID: mdl-17913622

ABSTRACT

This chapter aims to describe methods to identify and characterize protein-protein interactions that were developed during our studies on translation initiation factor complexes. Methods include the two-hybrid assay, the GST pull-down assay, and the coimmunoprecipitation (co-IP) assay. The two-hybrid assay provides for a convenient start to find the minimal interaction domains, which generally produce well-behaved recombinant proteins suited for various in vitro interaction assays. Emphasis is placed on demonstrating physiological relevance of identified interactions. The effective strategy is to find mutations that reduce the interaction by genetic or site-directed mutational approaches and obtain correlations between their effects in vitro (GST pull down) and effects in vivo (co-IP).


Subject(s)
Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Chromatography, Affinity , Eukaryotic Initiation Factors/physiology , Glutathione Transferase/genetics , Immunoprecipitation , Mutagenesis, Site-Directed , Recombinant Fusion Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/physiology , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...