Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Biol Chem ; 300(3): 105703, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301895

ABSTRACT

Tandem GGGGCC repeat expansion in C9orf72 is a genetic cause of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Transcribed repeats are translated into dipeptide repeat proteins via repeat-associated non-AUG (RAN) translation. However, the regulatory mechanism of RAN translation remains unclear. Here, we reveal a GTPase-activating protein, eukaryotic initiation factor 5 (eIF5), which allosterically facilitates the conversion of eIF2-bound GTP into GDP upon start codon recognition, as a novel modifier of C9orf72 RAN translation. Compared to global translation, eIF5, but not its inactive mutants, preferentially stimulates poly-GA RAN translation. RAN translation is increased during integrated stress response, but the stimulatory effect of eIF5 on poly-GA RAN translation was additive to the increase of RAN translation during integrated stress response, with no further increase in phosphorylated eIF2α. Moreover, an alteration of the CUG near cognate codon to CCG or AUG in the poly-GA reading frame abolished the stimulatory effects, indicating that eIF5 primarily acts through the CUG-dependent initiation. Lastly, in a Drosophila model of C9orf72 FTLD/ALS that expresses GGGGCC repeats in the eye, knockdown of endogenous eIF5 by two independent RNAi strains significantly reduced poly-GA expressions, confirming in vivo effect of eIF5 on poly-GA RAN translation. Together, eIF5 stimulates the CUG initiation of poly-GA RAN translation in cellular and Drosophila disease models of C9orf72 FTLD/ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , DNA Repeat Expansion , Eukaryotic Initiation Factor-5 , Frontotemporal Lobar Degeneration , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , C9orf72 Protein/genetics , Dipeptides/genetics , DNA Repeat Expansion/genetics , Drosophila/genetics , Drosophila/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/physiopathology , HeLa Cells , Humans , Disease Models, Animal
2.
FEBS Lett ; 596(14): 1809-1826, 2022 07.
Article in English | MEDLINE | ID: mdl-35490374

ABSTRACT

Mitochondrial activity adapts to cellular energetic and metabolic demands; its dysfunction is a hallmark of ageing and many human diseases. The evolutionarily conserved translation elongation factor eIF5A is involved in maintaining mitochondrial function. In humans, eIF5A is encoded by two highly homologous but differentially expressed genes; in yeast, these are TIF51A and TIF51B. We show that yeast transcription factor Hap1 constitutively binds to the TIF51A promoter to activate its expression under respiration, but represses its expression under nonrespiration conditions by recruiting the corepressor Tup1. Hap1 indirectly regulates TIF51B expression by binding to and activating the TIF51B repressor genes ROX1 and MOT3 under respiration and repressing them under nonrespiration. Thus, the levels of eIF5A isoforms are adapted to the mitochondrial functional status.


Subject(s)
DNA-Binding Proteins , Eukaryotic Initiation Factor-5 , Mitochondria , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Functional Status , Humans , Mitochondria/genetics , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Mol Biol ; 434(10): 167564, 2022 05 30.
Article in English | MEDLINE | ID: mdl-35358571

ABSTRACT

Translation factors are essential for regulation of protein synthesis. The eukaryotic translation initiation factor 5A (eIF5A) family is made up of two paralogues - eIF5A1 and eIF5A2 - which display high sequence homology but distinct tissue tropism. While eIF5A1 directly binds to the ribosome and regulates translation initiation, elongation, and termination, the molecular function of eIF5A2 remains poorly understood. Here, we engineer an eIF5A2 knockout allele in the SW480 colon cancer cell line. Using ribosome profiling and RNA-Sequencing, we reveal that eIF5A2 is functionally distinct from eIF5A1 and does not regulate transcript-specific or global protein synthesis. Instead, eIF5A2 knockout leads to decreased intrinsic antiviral gene expression, including members of the IFITM and APOBEC3 family. Furthermore, cells lacking eIF5A2 display increased permissiveness to virus infection. Our results uncover eIF5A2 as a factor involved regulating the antiviral transcriptome, and reveal an example of how gene duplications of translation factors can result in proteins with distinct functions.


Subject(s)
Eukaryotic Initiation Factor-5 , Gene Expression Regulation , Peptide Initiation Factors , RNA-Binding Proteins , Virus Diseases , APOBEC Deaminases/genetics , Cell Line, Tumor , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Gene Knockout Techniques , Humans , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcriptome , Virus Diseases/genetics , Eukaryotic Translation Initiation Factor 5A
4.
Cell Rep ; 36(9): 109633, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469733

ABSTRACT

In this work, we show that Not4 and Not5 from the Ccr4-Not complex modulate translation elongation dynamics and change ribosome A-site dwelling occupancy in a codon-dependent fashion. These codon-specific changes in not5Δ cells are very robust and independent of codon position within the mRNA, the overall mRNA codon composition, or changes of mRNA expression levels. They inversely correlate with codon-specific changes in cells depleted for eIF5A and positively correlate with those in cells depleted for ribosome-recycling factor Rli1. Not5 resides in punctate loci, co-purifies with ribosomes and Rli1, but not with eIF5A, and limits mRNA solubility. Overexpression of wild-type or non-complementing Rli1 and loss of Rps7A ubiquitination enable Not4 E3 ligase-dependent translation of polyarginine stretches. We propose that Not4 and Not5 modulate translation elongation dynamics to produce a soluble proteome by Rps7A ubiquitination, dynamic condensates that limit mRNA solubility and exclude eIF5A, and a moonlighting function of Rli1.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Eukaryotic Initiation Factor-5/metabolism , Peptide Chain Elongation, Translational , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Ribosome Subunits, Small/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , ATP-Binding Cassette Transporters/genetics , Eukaryotic Initiation Factor-5/genetics , Gene Expression Regulation, Fungal , Peptide Initiation Factors/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Ribosome Subunits, Small/genetics , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Eukaryotic Translation Initiation Factor 5A
5.
Curr Med Sci ; 41(4): 764-769, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34403101

ABSTRACT

OBJECTIVE: The expression levels of histone deacetylase 2 (HDAC2), eukaryotic initiation factor 5 (eIF5), and eukaryotic initiation factor 6 (eIF6), and relationship between HDAC2 and eIF5 or eIF6 in lung cancer tissues were investigated, in order to charify the relationship between HDAC2 and the prognosis of lung cancer patients and its influence on the expression of eIF5 and eIF6. METHODS: The expression of HDAC2, eIF5, and eIF6 in lung cancer tissues was detected by quantitative reverse transcription polymerase chain reaction. The expression correlation between HDAC2 and eIF5 or eIF6 was tested using a t test. The correlation between HDAC2 and eIF5 or eIF6 was analyzed using the TCGA database. The identified cells were constructed with small interfering siRNA and HDAC2 overexpression plasmid. The proliferation and migration ability of the identified cells was investigated by CCK8 and Transwell assays, respectively. RESULTS: HDAC2, eIF5, and eIF6 were overexpressed in lung cancer tissues, and HDAC2 expression level was negatively correlated with the prognosis of lung cancer patients. HDAC2 expression level was positively correlated with eIF5 and eIF6 expression levels. HDAC2 could regulate the expression of eIF5 and eIF6. The regulation of proliferation and invasion of lung cancer cells by HDAC2 depended on eIF5 and eIF6. CONCLUSION: HDAC2, eIF5, and eIF6 were closely related with lung cancer tumorigenesis, which might be potential biological markers and therapeutic targets for lung cancer.


Subject(s)
Carcinogenesis/genetics , Eukaryotic Initiation Factor-5/genetics , Histone Deacetylase 2/genetics , Lung Neoplasms/genetics , Peptide Initiation Factors/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Progression-Free Survival , RNA Interference , RNA, Small Interfering/genetics
6.
Nucleic Acids Res ; 47(15): 8282-8300, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31291455

ABSTRACT

eIF3 is a large multiprotein complex serving as an essential scaffold promoting binding of other eIFs to the 40S subunit, where it coordinates their actions during translation initiation. Perhaps due to a high degree of flexibility of multiple eIF3 subunits, a high-resolution structure of free eIF3 from any organism has never been solved. Employing genetics and biochemistry, we previously built a 2D interaction map of all five yeast eIF3 subunits. Here we further improved the previously reported in vitro reconstitution protocol of yeast eIF3, which we cross-linked and trypsin-digested to determine its overall shape in 3D by advanced mass-spectrometry. The obtained cross-links support our 2D subunit interaction map and reveal that eIF3 is tightly packed with its WD40 and RRM domains exposed. This contrasts with reported cryo-EM structures depicting eIF3 as a molecular embracer of the 40S subunit. Since the binding of eIF1 and eIF5 further fortified the compact architecture of eIF3, we suggest that its initial contact with the 40S solvent-exposed side makes eIF3 to open up and wrap around the 40S head with its extended arms. In addition, we mapped the position of eIF5 to the region below the P- and E-sites of the 40S subunit.


Subject(s)
Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-5/chemistry , Peptide Chain Initiation, Translational , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Binding Sites/genetics , Cryoelectron Microscopy , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factor-3/genetics , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Models, Molecular , Protein Binding , Protein Domains , Ribosome Subunits, Small, Eukaryotic/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Biochem Biophys Res Commun ; 516(3): 753-759, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31255281

ABSTRACT

The GAIT (gamma-interferon-activated inhibitor of translation) complex or miR-297-RISC (RNA-induced silencing complex), together with hnRNP L or hnRNP L-bearing complex, operates an RNA switch in myeloid cells that regulates stress-dependent expression of vascular endothelial growth factor-A (VEGFA). Here, we have shown that hnRNP L directs multiple hypoxia-inducible RNA switches simultaneously and regulates expression of these oncogenic genes in addition to VEGFA. Bioinformatic and polysome profiling-microarray screens have identified DNM1L (Dynamin 1-like) and PHF21A (PHD finger protein 21A) mRNAs as regulated at the translational level by GAIT-dependent, hnRNP L-directed RNA switches. We have also uncovered CDK6 (Cyclin dependent kinase 6), MKLN1 (Muskelin 1) and EIF5 (Eukaryotic initiation factor 5) as novel miR-297-dependent, hnRNP L-directed RNA switch transcripts. Src Kinase is required for the phosphorylation of hnRNP L and activation of the RNA switch pathway. Knockdown of hnRNP L sensitizes the human U937 monocytic cells under hypoxia stress but not in normoxia via inducing cell apoptosis partially due to the reduced translation of hnRNP L target mRNAs. Collectively, our findings suggest that commonly controlled genes by the hnRNP L-directed RNA switches form a translational regulon that promotes hypoxia resistance and cell survival.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Protein Biosynthesis , RNA/metabolism , Regulon , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Hypoxia , Dynamins/genetics , Dynamins/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Gene Expression Profiling/methods , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , RNA/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , U937 Cells , Vascular Endothelial Growth Factor A/genetics
8.
J Cell Biochem ; 119(12): 9730-9741, 2018 12.
Article in English | MEDLINE | ID: mdl-30132996

ABSTRACT

The aberrant expression of RNA-binding proteins (RBPs) plays a crucial role in the occurrence and progression of human cancer. However, the key functions of RBPs in the metastasis of colorectal cancer have not yet been fully elucidated. Here, we integrated multi-omics data and identified four differentially expressed RBPs (APOBEC3G, EEF1A2, EIF5AL1 and CELF3) in patients with colorectal cancer metastasis. To clarify the underlying molecular mechanisms, we systematically analyzed the genomic features and downstream regulatory relationships of the four RBPs. In a genomic level, the copy number variations of APOBEC3G, EEF1A2, and CELF3 demonstrated significantly differential distributions between metastatic and nonmetastatic patients. Besides that, combining sequence and expression information, we identified 436 putative RNA targets regulated by the four RBPs through strict multistep bioinformatics screening. For the downstream analysis, the evidence from functional enrichment analysis and public literature indicated the roles of these target genes in the carcinogenesis and progression of colorectal cancer. Furthermore, through the machine learning algorithm and statistical analysis, we obtained two gene candidates that had obvious effects on the metastasis and overall survival status of patients with colorectal cancer. In summary, our study comprehensively explored the influence of APOBEC3G, EEF1A2, EIF5AL1, and CELF3 in colorectal cancer metastasis, which may offer favorable perspectives for clinical diagnosis and therapy.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , RNA-Binding Proteins/genetics , APOBEC-3G Deaminase/genetics , Algorithms , CELF Proteins/genetics , Colorectal Neoplasms/mortality , Computational Biology/methods , DNA Copy Number Variations , Eukaryotic Initiation Factor-5/genetics , Gene Expression Regulation, Neoplastic , Humans , Machine Learning , Peptide Elongation Factor 1/genetics , Peptide Initiation Factors/genetics , Eukaryotic Translation Initiation Factor 5A
9.
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
10.
Proc Natl Acad Sci U S A ; 114(11): E2126-E2135, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28223523

ABSTRACT

The eukaryotic 43S preinitiation complex (PIC) bearing Met-tRNAiMet in a ternary complex (TC) with eukaryotic initiation factor (eIF)2-GTP scans the mRNA leader for an AUG codon in favorable "Kozak" context. AUG recognition provokes rearrangement from an open PIC conformation with TC bound in a state not fully engaged with the P site ("POUT") to a closed, arrested conformation with TC tightly bound in the "PIN" state. Yeast ribosomal protein Rps3/uS3 resides in the mRNA entry channel of the 40S subunit and contacts mRNA via conserved residues whose functional importance was unknown. We show that substitutions of these residues reduce bulk translation initiation and diminish initiation at near-cognate UUG start codons in yeast mutants in which UUG selection is abnormally high. Two such substitutions-R116D and R117D-also increase discrimination against an AUG codon in suboptimal Kozak context. Consistently, the Arg116 and Arg117 substitutions destabilize TC binding to 48S PICs reconstituted in vitro with mRNA harboring a UUG start codon, indicating destabilization of the closed PIN state with a UUG-anticodon mismatch. Using model mRNAs lacking contacts with either the mRNA entry or exit channels of the 40S subunit, we demonstrate that Arg116/Arg117 are crucial for stabilizing PIC-mRNA contacts at the entry channel, augmenting the function of eIF3 at both entry and exit channels. The corresponding residues in bacterial uS3 promote the helicase activity of the elongating ribosome, suggesting that uS3 contacts with mRNA enhance multiple phases of translation across different domains of life.


Subject(s)
Codon, Initiator , Multiprotein Complexes/metabolism , Peptide Chain Initiation, Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Alleles , Amino Acid Substitution , Eukaryotic Initiation Factor-5/chemistry , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Mutation , Phenotype , Protein Binding , Protein Conformation , Protein Stability , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosome Subunits, Small, Eukaryotic/chemistry
11.
FEBS J ; 282(3): 504-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25417541

ABSTRACT

Cells respond to changes in their environment through mechanisms that often necessitate reprogramming of the translation machinery. The fastest and strongest of all tested responses is the translation inhibition observed following abrupt depletion of glucose from the media of yeast cells. The speed of the response suggests a post-translational modification of a key component of the translation machinery. This translation factor is as yet unknown. A cAMP-dependent protein kinase mutant yeast strain (tpk1(w)) that does not respond properly to glucose depletion and maintains translation was described previously. We hypothesized that the inability of tpk1(w) to arrest translation results from abnormal expression of key translation mediators. Genome-wide analysis of steady-state mRNA levels in tpk1(w) revealed underexpression of several candidates. Elevating the cellular levels of eukaryotic initiation factor (eIF) 5 by overexpression rescued the translational defect of tpk1(w). Restoring ribosomal dissociation by eIF5 necessitated an active GAP domain and multiple regions throughout this protein. Phosphoproteomics analysis of wild-type cells overexpressing eIF5 revealed increased phosphorylation in a novel site (Thr191) upon glucose depletion. Mutating this residue and introducing it into tpk1(w) abolished the ability of eIF5 to rescue the translational defect. Intriguingly, introducing this mutation into the wild-type strain did not hamper its translational response. We further show that Thr191 is phosphorylated in vitro by Casein Kinase II (CKII), and yeast cells with a mutated CKII have a reduced response to glucose depletion. These results implicate phosphorylation of eIF5 at Thr191 by CKII as one of the pathways for regulating translation upon glucose depletion.


Subject(s)
Eukaryotic Initiation Factor-5/metabolism , Polyribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Eukaryotic Initiation Factor-5/genetics , Phosphoproteins/metabolism , Phosphorylation , Proteomics , Saccharomyces cerevisiae Proteins/genetics
12.
Nucleic Acids Res ; 42(19): 12052-69, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25260592

ABSTRACT

48S initiation complex (48S IC) formation is the first stage in the eukaryotic translation process. According to the canonical mechanism, 40S ribosomal subunit binds to the 5'-end of messenger RNA (mRNA) and scans its 5'-untranslated region (5'-UTR) to the initiation codon where it forms the 48S IC. Entire process is mediated by initiation factors. Here we show that eIF5 and eIF5B together stimulate 48S IC formation influencing initiation codon selection during ribosomal scanning. Initiation on non-optimal start codons--following structured 5'-UTRs, in bad AUG context, within few nucleotides from 5'-end of mRNA and CUG start codon--is the most affected. eIF5-induced hydrolysis of eIF2-bound GTP is essential for stimulation. GTP hydrolysis increases the probability that scanning ribosomal complexes will recognize and arrest scanning at a non-optimal initiation codon. Such 48S ICs are less stable owing to dissociation of eIF2*GDP from initiator tRNA, and eIF5B is then required to stabilize the initiator tRNA in the P site of 40S subunit. Alternative model that eIF5 and eIF5B cause 43S pre-initiation complex rearrangement favoring more efficient initiation codon recognition during ribosomal scanning is equally possible. Mutational analysis of eIF1A and eIF5B revealed distinct functions of eIF5B in 48S IC formation and subunit joining.


Subject(s)
Eukaryotic Initiation Factor-5/metabolism , Eukaryotic Initiation Factors/metabolism , Peptide Chain Initiation, Translational , Ribosomes/metabolism , 5' Untranslated Regions , Codon, Initiator , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factors/genetics , Guanosine Triphosphate/metabolism , Mutation , RNA, Transfer, Met/metabolism
13.
Nucleic Acids Res ; 42(15): 9623-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25114053

ABSTRACT

eIF5 is the GTPase activating protein (GAP) for the eIF2 · GTP · Met-tRNAi (Met) ternary complex with a critical role in initiation codon selection. Previous work suggested that the eIF5 mutation G31R/SUI5 elevates initiation at UUG codons by increasing GAP function. Subsequent work implicated eIF5 in rearrangement of the preinitiation complex (PIC) from an open, scanning conformation to a closed state at AUG codons, from which Pi is released from eIF2 · GDP · Pi. To identify eIF5 functions crucial for accurate initiation, we investigated the consequences of G31R on GTP hydrolysis and Pi release, and the effects of intragenic G31R suppressors on these reactions, and on the partitioning of PICs between open and closed states. eIF5-G31R altered regulation of Pi release, accelerating it at UUG while decreasing it at AUG codons, consistent with its ability to stabilize the closed complex at UUG. Suppressor G62S mitigates both defects of G31R, accounting for its efficient suppression of UUG initiation in G31R,G62S cells; however suppressor M18V impairs GTP hydrolysis with little effect on PIC conformation. The strong defect in GTP hydrolysis conferred by M18V likely explains its broad suppression of Sui(-) mutations in numerous factors. We conclude that both of eIF5's functions, regulating Pi release and stabilizing the closed PIC conformation, contribute to stringent AUG selection in vivo.


Subject(s)
Codon, Initiator , Eukaryotic Initiation Factor-5/metabolism , Guanosine Triphosphate/metabolism , Peptide Chain Initiation, Translational , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-5/chemistry , Eukaryotic Initiation Factor-5/genetics , Mutation , Phosphates/metabolism , Suppression, Genetic
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.
Amino Acids ; 42(5): 1651-9, 2012 May.
Article in English | MEDLINE | ID: mdl-21360085

ABSTRACT

Eukaryotic initiation factor 5A (eIF5A) has recently been identified as a biomarker of prognostic significance and therapeutic potential for the treatment in hepatocellular carcinoma. This prompted us to establish a rapid and robust assay to determine deoxyhypusine and hypusine formed with the purified enzymes deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH) from Plasmodium to develop a rapid screening assay for antimalarial drugs. The peptide hydrolysate obtained from hypusinylated eIF5A was analyzed by ultra performance liquid chromatography (UPLC) with retention times for deoxyhypusine of 7.44 min and for hypusine of 7.30 min, respectively. The limit of detection for both compounds was 0.144 ng/µl. Determination of the specific activity of Plasmodium DOHH resulted in a twofold higher specific activity than its human counterpart. Following the iron-complexing strategy of the ferrous iron which is present in the active site of Plasmodium DOHH, a series of iron chelating compounds was tested. 2,2'-Dipyridyl and mimosine abolished DOHH activity completely while 4-oxo-piperidine-carboxylates i.e. the nitrophenylether JK8-2 and EHW 437, the oxime ether of the piperidine aldehyde, showed no inhibition although they were highly active in in vitro cultures of Plasmodium and in vivo in a rodent mouse model. The method allows a high-throughput screening (HPTS) of antimalarial drugs and the evaluation of eIF5A as a biomarker.


Subject(s)
Antimalarials/isolation & purification , Eukaryotic Initiation Factor-5/metabolism , Lysine/analogs & derivatives , Malaria/drug therapy , Plasmodium/enzymology , Animals , Antimalarials/chemistry , Antimalarials/therapeutic use , Catalytic Domain , Chromatography, High Pressure Liquid , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/isolation & purification , Humans , Lysine/isolation & purification , Lysine/metabolism , Malaria/diagnosis , Mice , Mixed Function Oxygenases/isolation & purification , Mixed Function Oxygenases/metabolism , Oxidoreductases Acting on CH-NH Group Donors/isolation & purification , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Protein Hydrolysates/chemistry
16.
Biochem Biophys Res Commun ; 414(2): 390-6, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21964295

ABSTRACT

We earlier documented the structural and functional characterization of PeIF5B factor from Pisum sativum that shows strong homology to the universal translation initiation factor eIF5B (Rasheedi et al., 2007, 2010 [12,13]). We now show that PeIF5B is an unusually thermo-stable protein resisting temperatures up to 95 °C. PeIF5B prevents thermal aggregation of heat labile proteins, such as citrate synthase (CS) and NdeI, under heat stress or chemical denaturation conditions and promotes their functional folding. It also prevents the aggregation of DTT induced insulin reduction. GTP appears to stimulate PeIF5B-mediated chaperone activity. In-vivo, PeIF5B over expression significantly enhances, the viability of Escherichia coli cells after heat stress (50 °C). These observations lead us to conclude that PeIF5B, in addition to its role in protein translation, has chaperone like activity and could be likely involved in protein folding and protection from stress.


Subject(s)
Eukaryotic Initiation Factor-5/metabolism , Molecular Chaperones/metabolism , Pisum sativum/metabolism , Plant Proteins/metabolism , Escherichia coli , Eukaryotic Initiation Factor-5/chemistry , Eukaryotic Initiation Factor-5/genetics , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Heat-Shock Response , Hot Temperature , Hydrophobic and Hydrophilic Interactions , Insulin/chemistry , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Oxidation-Reduction , Peptide Chain Initiation, Translational , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Folding , Protein Stability
17.
Nucleic Acids Res ; 39(14): 6148-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21459853

ABSTRACT

The EKC/KEOPS complex is universally conserved in Archaea and Eukarya and has been implicated in several cellular processes, including transcription, telomere homeostasis and genomic instability. However, the molecular function of the complex has remained elusive so far. We analyzed the transcriptome of EKC/KEOPS mutants and observed a specific profile that is highly enriched in targets of the Gcn4p transcriptional activator. GCN4 expression was found to be activated at the translational level in mutants via the defective recognition of the inhibitory upstream ORFs (uORFs) present in its leader. We show that EKC/KEOPS mutants are defective for the N6-threonylcarbamoyl adenosine modification at position 37 (t(6)A(37)) of tRNAs decoding ANN codons, which affects initiation at the inhibitory uORFs and provokes Gcn4 de-repression. Structural modeling reveals similarities between Kae1 and bacterial enzymes involved in carbamoylation reactions analogous to t(6)A(37) formation, supporting a direct role for the EKC in tRNA modification. These findings are further supported by strong genetic interactions of EKC mutants with a translation initiation factor and with threonine biosynthesis genes. Overall, our data provide a novel twist to understanding the primary function of the EKC/KEOPS and its impact on several essential cellular functions like transcription and telomere homeostasis.


Subject(s)
Adenosine/analogs & derivatives , Basic-Leucine Zipper Transcription Factors/genetics , RNA, Transfer/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Adenosine/metabolism , Basic-Leucine Zipper Transcription Factors/biosynthesis , Codon, Initiator , Eukaryotic Initiation Factor-5/genetics , Evolution, Molecular , Gene Expression Profiling , Gene Expression Regulation, Fungal , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Mutation , Phylogeny , Protein Biosynthesis , RNA, Transfer/chemistry , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/genetics
18.
Neuron ; 67(6): 1021-33, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20869598

ABSTRACT

Local interneurons in the Drosophila antennal lobe are thought to play important roles in shaping odor responses. However, the physiological properties of excitatory local interneurons (eLNs) and their connectivity in the antennal lobe remain unclear. We first characterized the firing patterns of krasavietz-Gal4-labeled eLNs (krasavietz eLNs) in response to depolarizing currents. Paired recordings of krasavietz eLNs and PNs showed reciprocal excitatory connections mediated by dendrodendritic cholinergic synapses and gap junctions. Reciprocal connections were also found between two krasavietz eLNs but were rare between krasavietz eLNs and inhibitory LNs. Analysis of response onset latencies showed that krasavietz eLNs received monosynaptic inputs from ORNs. Furthermore, each eLN responded with distinct patterns to different odors, and each odor elicited distinct responses in different eLNs, with specific temporal patterns of spiking, indicating that eLNs serve specific coding functions in addition to global excitation in Drosophila olfactory processing.


Subject(s)
Interneurons/physiology , Nerve Net/physiology , Odorants , Sense Organs/cytology , Smell/physiology , Action Potentials/drug effects , Action Potentials/genetics , Adenosine Triphosphate/pharmacology , Animals , Animals, Genetically Modified , Bungarotoxins/pharmacology , Cholinergic Antagonists/pharmacology , Drosophila , Drosophila Proteins/genetics , Electric Stimulation/methods , Eukaryotic Initiation Factor-5/genetics , Female , GABA Antagonists/pharmacology , Gap Junctions/drug effects , Gap Junctions/genetics , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Interneurons/drug effects , Lysine/analogs & derivatives , Lysine/metabolism , Mecamylamine/pharmacology , Nerve Net/cytology , Nerve Net/drug effects , Nicotinic Antagonists/pharmacology , Olfactory Pathways/physiology , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques , Picrotoxin/pharmacology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X2
19.
Neuron ; 67(6): 1034-47, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20869599

ABSTRACT

In the Drosophila antennal lobe, excitation can spread between glomerular processing channels. In this study, we investigated the mechanism of lateral excitation. Dual recordings from excitatory local neurons (eLNs) and projection neurons (PNs) showed that eLN-to-PN synapses transmit both hyperpolarization and depolarization, are not diminished by blocking chemical neurotransmission, and are abolished by a gap-junction mutation. This mutation eliminates odor-evoked lateral excitation in PNs and diminishes some PN odor responses. This implies that lateral excitation is mediated by electrical synapses from eLNs onto PNs. In addition, eLNs form synapses onto inhibitory LNs. Eliminating these synapses boosts some PN odor responses and reduces the disinhibitory effect of GABA receptor antagonists on PNs. Thus, eLNs have two opposing effects on PNs, driving both direct excitation and indirect inhibition. We propose that when stimuli are weak, lateral excitation promotes sensitivity, whereas when stimuli are strong, lateral excitation helps recruit inhibitory gain control.


Subject(s)
Electrical Synapses/physiology , Neuropil/physiology , Olfactory Receptor Neurons/physiology , Smell/physiology , Animals , Animals, Genetically Modified , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cadmium Chloride/pharmacology , Connexins/genetics , Drosophila , Drosophila Proteins/genetics , Electric Stimulation/methods , Eukaryotic Initiation Factor-5/genetics , Female , Light , Luminescent Proteins/genetics , Models, Biological , Mutation/genetics , Nerve Tissue Proteins/genetics , Neural Inhibition/genetics , Neuropil/cytology , Neuropil/drug effects , Odorants , Olfactory Receptor Neurons/drug effects , Patch-Clamp Techniques/methods , Rhodopsin/genetics , Rhodopsin/metabolism , Sense Organs/cytology , Smell/drug effects , Smell/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , gamma-Aminobutyric Acid/metabolism
20.
Curr Biol ; 19(24): 2126-32, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-20005106

ABSTRACT

Alcohol addiction is a common affliction with a strong genetic component [1]. Although mammalian studies have provided significant insight into the molecular mechanisms underlying ethanol consumption [2], other organisms such as Drosophila melanogaster are better suited for unbiased, forward genetic approaches to identify novel genes. Behavioral responses to ethanol, such as hyperactivity, sedation, and tolerance, are conserved between flies and mammals [3, 4], as are the underlying molecular pathways [5-9]. However, few studies have investigated ethanol self-administration in flies [10]. Here we characterize ethanol consumption and preference in Drosophila. Flies prefer to consume ethanol-containing food over regular food, and this preference increases over time. Flies are attracted to the smell of ethanol, which partially mediates ethanol preference, but are averse to its taste. Preference for consuming ethanol is not entirely explained by attraction to either its sensory or caloric properties. We demonstrate that flies can exhibit features of alcohol addiction. First, flies self-administer ethanol to pharmacologically relevant concentrations. Second, flies will overcome an aversive stimulus in order to consume ethanol. Third, flies rapidly return to high levels of ethanol consumption after a period of imposed abstinence. Thus, ethanol preference in Drosophila provides a new model for studying aspects of addiction.


Subject(s)
Alcohol Drinking/physiopathology , Alcoholism/physiopathology , Disease Models, Animal , Drosophila melanogaster , Ethanol/chemistry , Food Preferences/physiology , Alcohol Drinking/genetics , Alcoholism/genetics , Analysis of Variance , Animals , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/physiology , Smell/physiology , Taste/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...