Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Cell ; 77(6): 1176-1192.e16, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31999954

ABSTRACT

Microexons represent the most highly conserved class of alternative splicing, yet their functions are poorly understood. Here, we focus on closely related neuronal microexons overlapping prion-like domains in the translation initiation factors, eIF4G1 and eIF4G3, the splicing of which is activity dependent and frequently disrupted in autism. CRISPR-Cas9 deletion of these microexons selectively upregulates synaptic proteins that control neuronal activity and plasticity and further triggers a gene expression program mirroring that of activated neurons. Mice lacking the Eif4g1 microexon display social behavior, learning, and memory deficits, accompanied by altered hippocampal synaptic plasticity. We provide evidence that the eIF4G microexons function as a translational brake by causing ribosome stalling, through their propensity to promote the coalescence of cytoplasmic granule components associated with translation repression, including the fragile X mental retardation protein FMRP. The results thus reveal an autism-disrupted mechanism by which alternative splicing specializes neuronal translation to control higher order cognitive functioning.


Subject(s)
Autistic Disorder/physiopathology , Cognitive Dysfunction/pathology , Eukaryotic Initiation Factor-4G/physiology , Exons/genetics , Fragile X Mental Retardation Protein/metabolism , Neuroblastoma/pathology , Neurons/pathology , Animals , Behavior, Animal , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, Inbred C57BL , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neurogenesis , Neurons/metabolism , Protein Biosynthesis , RNA Splicing , Tumor Cells, Cultured
2.
Plant Biotechnol J ; 16(11): 1918-1927, 2018 11.
Article in English | MEDLINE | ID: mdl-29604159

ABSTRACT

Rice tungro disease (RTD) is a serious constraint in rice production across tropical Asia. RTD is caused by the interaction between Rice tungro spherical virus (RTSV) and Rice tungro bacilliform virus. RTSV resistance found in traditional cultivars has contributed to a reduction in the incidence of RTD in the field. Natural RTSV resistance is a recessive trait controlled by the translation initiation factor 4 gamma gene (eIF4G). The Y1059 V1060 V1061 residues of eIF4G are known to be associated with the reactions to RTSV. To develop new sources of resistance to RTD, mutations in eIF4G were generated using the CRISPR/Cas9 system in the RTSV-susceptible variety IR64, widely grown across tropical Asia. The mutation rates ranged from 36.0% to 86.6%, depending on the target site, and the mutations were successfully transmitted to the next generations. Among various mutated eIF4G alleles examined, only those resulting in in-frame mutations in SVLFPNLAGKS residues (mainly NL), adjacent to the YVV residues, conferred resistance. Furthermore, our data suggest that eIF4G is essential for normal development, as alleles resulting in truncated eIF4G could not be maintained in homozygous state. The final products with RTSV resistance and enhanced yield under glasshouse conditions were found to no longer contain the Cas9 sequence. Hence, the RTSV-resistant plants with the novel eIF4G alleles represent a valuable material to develop more diverse RTSV-resistant varieties.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Disease Resistance/genetics , Eukaryotic Initiation Factor-4G/genetics , Gene Editing/methods , Oryza/genetics , Plant Diseases/virology , Tungrovirus , Alleles , Eukaryotic Initiation Factor-4G/physiology , Genes, Plant/genetics , Genes, Plant/physiology , Oryza/virology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/virology
3.
J Neurosci ; 34(27): 9034-9, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24990923

ABSTRACT

Memory retrieval, often termed reconsolidation, can render previously consolidated memories susceptible to manipulation that can lead to alterations in memory strength. Although it is known that reconsolidation requires mammalian target of rapamycin complex 1 (mTORC1)-dependent translation, the specific contributions of its downstream effectors in reconsolidation are unclear. Using auditory fear conditioning in mice, we investigated the role of eukaryotic translation initiation factor 4E (eIF4E)-eIF4G interactions and p70 S6 kinase polypeptide 1 (S6K1) in reconsolidation. We found that neither 4EGI-1 (2-[(4-(3,4-dichlorophenyl)-thiazol-2-ylhydrazono)-3-(2-nitrophenyl)]propionic acid), an inhibitor of eFI4E-eIF4G interactions, nor PF-4708671 [2-((4-(5-ethylpyrimidin-4-yl)piperazin-1-yl)methyl)-5-(trifluoromethyl)-1H-benzo[d]imidazole], an inhibitor of S6K1, alone blocked the reconsolidation of auditory fear memory. In contrast, using these drugs in concert to simultaneously block eIF4E-eIF4G interactions and S6K1 immediately after memory reactivation significantly attenuated fear memory reconsolidation. Moreover, the combination of 4EGI-1 and PF-4708671 further destabilized fear memory 10 d after memory reactivation, which was consistent with experiments using rapamycin, an mTORC1 inhibitor. Furthermore, inhibition of S6K1 immediately after retrieval resulted in memory destabilization 10 d after reactivation, whereas inhibition of eIF4E-eIF4G interactions did not. These results indicate that the reconsolidation of fear memory requires concomitant association of eIF4E to eIF4G as well as S6K1 activity and that the persistence of memory at longer intervals after memory reactivation also requires mTORC1-dependent processes that involve S6K1. These findings suggest a potential mechanism for how mTORC1-dependent translation is fine tuned to alter memory persistence.


Subject(s)
Avoidance Learning/physiology , Conditioning, Classical/physiology , Fear/physiology , Mental Recall/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Acoustic Stimulation , Animals , Avoidance Learning/drug effects , Conditioning, Classical/drug effects , Cues , Electroshock , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4E/physiology , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/physiology , Hydrazones , Imidazoles/pharmacology , Male , Mechanistic Target of Rapamycin Complex 1 , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Mental Recall/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitro Compounds/pharmacology , Piperazines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Sirolimus/pharmacology , Thiazoles/pharmacology
4.
PLoS One ; 8(9): e74085, 2013.
Article in English | MEDLINE | ID: mdl-24019950

ABSTRACT

The rate-limiting step(s) of translation in the nervous system have not been clearly identified. We have been examining this question in the cell body of the Aplysia sensory neuron, where translational regulation is important for the regulation of synaptic strength. In the present study, we examined the role of the adaptor protein eIF4G. We cloned Aplysia eIF4G (Ap4G) and Ap4G contains all the standard metazoan eIF4G protein-protein interaction domains. Overexpressing Ap4G in Aplysia sensory neurons caused an increase in both cap-dependent and internal ribosome entry site (IRES)-dependent translation using a previously characterized bicistronic fluorescent reporter. Unexpectedly, measurement of overall translation using the methionine analog, L-azidohomoalanine, revealed that overexpression of Ap4G did not lead to an increase in overall translation rates. Indeed, the effect of Ap4G on the bicistronic reporter depended on the presence of an upstream open reading frame (uORF) in the 5' UTR encoded by the vector. We have previously shown that Mnk strongly decreased cap-dependent translation and this depended on a putative 4G binding domain. Here we extend these results showing that even in the absence of the uORF, overexpression of Mnk strongly decreases cap-dependent translation and this depends on the Mnk binding site in eIF4G. Similarly, an increase in cap-dependent translation seen with overexpression of elongation factor 2 kinase did not depend on the uORF. Overall, we show that eIF4G is rate limiting for translation of an mRNA encoding an uORF, but is not generally a rate-limiting step for translation.


Subject(s)
Eukaryotic Initiation Factor-4G/physiology , Protein Biosynthesis , RNA Caps , Ribosomes/metabolism , Sensory Receptor Cells/metabolism , 5' Untranslated Regions , Animals , Aplysia , Cells, Cultured , Cloning, Molecular , Eukaryotic Initiation Factor-4G/genetics , Open Reading Frames
5.
J Neurosci ; 33(17): 7165-74, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23616526

ABSTRACT

Axonal protein synthesis is a complex process involving selective mRNA localization and translational regulation. In this study, using in situ hybridization and metabolic labeling, we show that the mRNAs encoding eukaryotic translation initiation factors eIF2B2 and eIF4G2 are present in the axons of rat sympathetic neurons and are locally translated. We also report that a noncoding microRNA, miR16, modulates the axonal expression of eIF2B2 and eIF4G2. Transfection of axons with precursor miR16 and anti-miR16 showed that local miR16 levels modulated axonal eIF2B2 and eIF4G2 mRNA and protein levels, as well as axon outgrowth. siRNA-mediated knock-down of axonal eIF2B2 and eIF4G2 mRNA also resulted in a significant decrease in axonal eIF2B2 and eIF4G2 protein. Moreover, results of metabolic labeling studies showed that downregulation of axonal eIF2B2 and eIF4G2 expression also inhibited local protein synthesis and axon growth. Together, these data provide evidence that miR16 mediates axonal growth, at least in part, by regulating the local protein synthesis of eukaryotic translation initiation factors eIF2B2 and eIF4G2 in the axon.


Subject(s)
Adrenergic Fibers/metabolism , Axons/metabolism , Eukaryotic Initiation Factor-2B/biosynthesis , Eukaryotic Initiation Factor-4G/biosynthesis , Protein Biosynthesis/physiology , Adrenergic Fibers/physiology , Animals , Axons/physiology , Cells, Cultured , Down-Regulation/physiology , Eukaryotic Initiation Factor-2B/antagonists & inhibitors , Eukaryotic Initiation Factor-2B/physiology , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/physiology , Female , Male , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/physiology
6.
Radiat Oncol ; 8: 35, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23402580

ABSTRACT

BACKGROUND: Proliferating tumor cells require continuous protein synthesis. De novo synthesis of most proteins is regulated through cap-dependent translation. Cellular stress such as ionizing radiation (IR) blocks cap-dependent translation resulting in shut-down of global protein translation which saves resources and energy needed for the stress response. At the same time, levels of proteins required for stress response are maintained or even increased. The study aimed to analyze the regulation of signaling pathways controlling protein translation in response to IR and the impact on Mcl-1, an anti-apoptotic and radioprotective protein, which levels rapidly decline upon IR. METHODS: Protein levels and processing were analyzed by Western blot. The assembly of the translational pre-initiation complex was examined by Immunoprecipitation and pull-down experiments with 7-methyl GTP agarose. To analyze IR-induced cell death, dissipation of the mitochondrial membrane potential and DNA fragmentation were determined by flow cytometry. Protein levels of the different initiation factors were down-regulated using RNA interference approach. RESULTS: IR induced caspase-dependent cleavage of the translational initiation factors eIF4G1, eIF3A, and eIF4B resulting in disassembly of the cap-dependent initiation complex. In addition, DAP5-dependent initiation complex that regulates IRES-dependent translation was disassembled in response to IR. Moreover, IR resulted in dephosphorylation of 4EBP1, an inhibitor of cap-dependent translation upstream of caspase activation. However, knock-down of eIF4G1, eIF4B, DAP5, or 4EBP1 did not affect IR-induced decline of the anti-apoptotic protein Mcl-1. CONCLUSION: Our data shows that cap-dependent translation is regulated at several levels in response to IR. However, the experiments indicate that IR-induced Mcl-1 decline is not a consequence of translational inhibition in Jurkat cells.


Subject(s)
Peptide Chain Initiation, Translational/radiation effects , Radiation, Ionizing , Adaptor Proteins, Signal Transducing/metabolism , Caspases/physiology , Cell Cycle Proteins , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-4G/physiology , Humans , Jurkat Cells , Myeloid Cell Leukemia Sequence 1 Protein , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
Nucleic Acids Res ; 41(1): 586-98, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23161679

ABSTRACT

microRNAs (miRNAs) regulate gene expression at multiple levels by repressing translation, stimulating deadenylation and inducing the premature decay of target messenger RNAs (mRNAs). Although the mechanism by which miRNAs repress translation has been widely studied, the precise step targeted and the molecular insights of such repression are still evasive. Here, we have used our newly designed in vitro system, which allows to study miRNA effect on translation independently of deadenylation. By using specific inhibitors of various stages of protein synthesis, we first show that miRNAs target exclusively the early steps of translation with no effect on 60S ribosomal subunit joining, elongation or termination. Then, by using viral proteases and IRES-driven mRNA constructs, we found that translational inhibition takes place during 43S ribosomal scanning and requires both the poly(A) binding protein and eIF4G independently from their physical interaction.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Peptide Chain Initiation, Translational , 5' Untranslated Regions , Eukaryotic Initiation Factor-4G/physiology , Hepacivirus/genetics , Peptides/metabolism , Poly(A)-Binding Proteins/physiology , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism
8.
Mol Cell ; 45(2): 244-54, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22284680

ABSTRACT

The formation of mRNPs controls the interaction of the translation and degradation machinery with individual mRNAs. The yeast Scd6 protein and its orthologs regulate translation and mRNA degradation in yeast, C. elegans, D. melanogaster, and humans by an unknown mechanism. We demonstrate that Scd6 represses translation by binding the eIF4G subunit of eIF4F in a manner dependent on its RGG domain, thereby forming an mRNP repressed for translation initiation. Strikingly, several other RGG domain-containing proteins in yeast copurify with eIF4E/G and we demonstrate that two such proteins, Npl3 and Sbp1, also directly bind eIF4G and repress translation in a manner dependent on their RGG motifs. These observations identify the mechanism of Scd6 function through its RGG motif and indicate that eIF4G plays an important role as a scaffolding protein for the recruitment of translation repressors.


Subject(s)
Eukaryotic Initiation Factor-4G/physiology , Fungal Proteins/physiology , Protein Biosynthesis , Amino Acid Motifs , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/physiology , Eukaryotic Initiation Factor-4G/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Models, Genetic , RNA, Messenger/metabolism
9.
Biol Reprod ; 86(3): 95, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190698

ABSTRACT

During spermiogenesis, expression of the specific proteins needed for proper differentiation of male germ cells is under translational control. We have shown that PAIP2A is a major translational regulator involved in the maturation of male germ cells and male fertility. To identify the proteins controlled by PAIP2A during spermiogenesis, we characterized the proteomic profiles of elongated spermatids from wild-type (WT) mice and mice that were Paip2a/Paip2b double-null mutants (DKO). Elongated spermatid populations were obtained and proteins were extracted and separated on gradient polyacrylamide gels. The gels were digested with trypsin and peptides were identified by mass spectrometry. We identified 632 proteins with at least two unique peptides and a confidence level of 95%. Only 209 proteins were consistently detected in WT or DKO replicates with more than five spectra. Twenty-nine proteins were differentially expressed with at least a 1.5-fold change; 10 and 19 proteins were down- and up-regulated, respectively, in DKO compared to WT mice. We confirmed the significantly different expression levels of three proteins, EIF4G1, AKAP4, and HK1, by Western blot analysis. We have characterized novel proteins that have their expression controlled by PAIP2A; of these, 50% are involved in flagellar structure and sperm motility. Although several proteins affected by abrogation of Paip2a have established roles in reproduction, the roles of many others remain to be determined.


Subject(s)
A Kinase Anchor Proteins/physiology , Carrier Proteins/physiology , Eukaryotic Initiation Factor-4G/physiology , Hexokinase/physiology , Spermatogenesis/physiology , A Kinase Anchor Proteins/genetics , Animals , Carrier Proteins/genetics , Eukaryotic Initiation Factor-4G/genetics , Fertility/genetics , Fertility/physiology , Flagella/physiology , Gene Expression Profiling , Hexokinase/genetics , Male , Mice , Mice, Knockout , Models, Animal , Proteomics , RNA-Binding Proteins , Repressor Proteins , Sperm Motility/physiology , Spermatogenesis/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
10.
J Mol Biol ; 412(4): 674-87, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21840318

ABSTRACT

Eukaryotic initiation factor (eIF) 4A is a DEAD-box helicase that stimulates translation initiation by unwinding mRNA secondary structure. The accessory proteins eIF4G, eIF4B, and eIF4H enhance the duplex unwinding activity of eIF4A, but the extent to which they modulate eIF4A activity is poorly understood. Here, we use real-time fluorescence assays to determine the kinetic parameters of duplex unwinding and ATP hydrolysis by these initiation factors. To ensure efficient duplex unwinding, eIF4B and eIF4G cooperatively activate the duplex unwinding activity of eIF4A. Our data reveal that eIF4H is much less efficient at stimulating eIF4A unwinding activity than eIF4B, implying that eIF4H is not able to completely substitute for eIF4B in duplex unwinding. By monitoring unwinding and ATPase assays under identical conditions, we demonstrate that eIF4B couples the ATP hydrolysis cycle of eIF4A with strand separation, thereby minimizing nonproductive unwinding events. Using duplex substrates with altered GC contents but similar predicted thermal stabilities, we further show that the rate of formation of productive unwinding complexes is strongly influenced by the local stability per base pair, in addition to the stability of the entire duplex. This finding explains how a change in the GC content of a hairpin is able to influence translation initiation while maintaining the overall predicted thermal stability.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/physiology , Eukaryotic Initiation Factors/physiology , Nucleic Acid Heteroduplexes/metabolism , Adenosine Triphosphatases/physiology , Base Composition/physiology , Base Pairing/physiology , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , DNA Helicases/physiology , Enzyme Activation/physiology , Eukaryotic Initiation Factor-4A/chemistry , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/metabolism , Humans , Hydrolysis , Models, Biological , Models, Molecular , Nucleic Acid Heteroduplexes/chemistry , Protein Structure, Tertiary
11.
Mol Cell ; 43(4): 613-23, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855800

ABSTRACT

Angiogenin is a stress-activated ribonuclease that cleaves tRNA within anticodon loops to produce tRNA-derived stress-induced fragments (tiRNAs). Transfection of natural or synthetic tiRNAs inhibits protein synthesis and triggers the phospho-eIF2α-independent assembly of stress granules (SGs), essential components of the stress response program. We show that selected tiRNAs inhibit protein synthesis by displacing eIF4G/eIF4A from uncapped > capped RNAs. tiRNAs also displace eIF4F, but not eIF4E:4EBP1, from isolated m(7)G cap. We identify a terminal oligoguanine motif that is required to displace the eIF4F complex, inhibit translation, and induce SG assembly. We show that the tiRNA-associated translational silencer YB-1 contributes to angiogenin-, tiRNA-, and oxidative stress-induced translational repression. Our data reveal some of the mechanisms by which stress-induced tRNA cleavage inhibits protein synthesis and activates a cytoprotective stress response program.


Subject(s)
Peptide Chain Initiation, Translational , RNA, Transfer/physiology , Ribonuclease, Pancreatic/physiology , Cell Line , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-4G/physiology , Humans , RNA, Transfer/chemistry , Stress, Physiological , Y-Box-Binding Protein 1/metabolism , Y-Box-Binding Protein 1/physiology
12.
Development ; 137(10): 1699-707, 2010 May.
Article in English | MEDLINE | ID: mdl-20430745

ABSTRACT

The ENU-induced repro8 mutation was identified in a screen to uncover genes that control mouse gametogenesis. repro8 causes male-limited infertility, with failure of spermatocytes to exit meiotic prophase via the G2/MI transition. The repro8 mutation is in the Eif4g3 gene, encoding eukaryotic translation initiation factor 4, gamma 3. Mutant germ cells appear to execute events of meiotic prophase normally, and many proteins characteristic of the prophase-to-metaphase transition are not obviously depleted. However, activity of CDC2A (CDK1) kinase is dramatically reduced in mutant spermatocytes. Strikingly, HSPA2, a chaperone protein for CDC2A kinase, is absent in mutant spermatocytes in spite of the presence of Hspa2 transcript, consistent with the observation that the repro8 phenotype is markedly similar to the phenotype of the Hspa2 knockout. Thus, EIF4G3 is required for HSPA2 translation in spermatocytes, a finding that provides the first genetic evidence for selective translational control of meiotic exit in mammalian spermatocytes.


Subject(s)
Eukaryotic Initiation Factor-4G/genetics , Infertility, Male/genetics , Meiosis/genetics , Mutation, Missense/physiology , Spermatocytes/metabolism , Animals , Cell Division/genetics , Cell Proliferation , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-4G/physiology , Gene Expression Regulation, Developmental , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Spermatocytes/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology
13.
PLoS One ; 5(2): e9114, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20161741

ABSTRACT

Initiation factor eIF4G is a key regulator of eukaryotic protein synthesis, recognizing proteins bound at both ends of an mRNA to help recruit messages to the small (40S) ribosomal subunit. Notably, the genomes of a wide variety of eukaryotes encode multiple distinct variants of eIF4G. We found that deletion of eIF4G1, but not eIF4G2, impairs growth and global translation initiation rates in budding yeast under standard laboratory conditions. Not all mRNAs are equally sensitive to loss of eIF4G1; genes that encode messages with longer poly(A) tails are preferentially affected. However, eIF4G1-deletion strains contain significantly lower levels of total eIF4G, relative to eIF4G2-delete or wild type strains. Homogenic strains, which encode two copies of either eIF4G1 or eIF4G2 under native promoter control, express a single isoform at levels similar to the total amount of eIF4G in a wild type cell and have a similar capacity to support normal translation initiation rates. Polysome microarray analysis of these strains and the wild type parent showed that translationally active mRNAs are similar. These results suggest that total eIF4G levels, but not isoform-specific functions, determine mRNA-specific translational efficiency.


Subject(s)
Eukaryotic Initiation Factor-4G/physiology , Protein Biosynthesis , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , Base Sequence , Cell Division/genetics , Cell Division/physiology , Eukaryotic Initiation Factor-4F/genetics , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4F/physiology , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Gene Expression Profiling , Gene Expression Regulation, Fungal , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Polyribosomes/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion
14.
Nat Cell Biol ; 11(7): 903-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19525934

ABSTRACT

Inflammatory breast cancer (IBC) is the most lethal form of primary breast cancer. IBC lethality derives from generation of tumour emboli, which are non-adherent cell clusters that rapidly spread by a form of continuous invasion known as passive metastasis. In most cancers, expression of E-cadherin, an epithelial marker, is indicative of low metastatic potential. In IBC, E-cadherin is overexpressed and supports formation of tumour emboli by promoting tumour cell interactions rather than adherence to stroma. E-cadherin, a surface component of adherens junctions, is anchored by interaction with p120 catenin (p120). We show that the unique pathogenic properties of IBC result in part from overexpression of the translation initiation factor eIF4GI in most IBCs. eIF4GI reprograms the protein synthetic machinery for increased translation of mRNAs with internal ribosome entry sites (IRESs) that promote IBC tumour cell survival and formation of tumour emboli. Overexpression of eIF4GI promotes formation of IBC tumour emboli by enhancing translation of IRES-containing p120 mRNAs. These findings provide a new understanding of translational control in the development of advanced breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Eukaryotic Initiation Factor-4G/physiology , Animals , Biological Transport/genetics , Biological Transport/physiology , Breast Neoplasms/genetics , Cadherins/genetics , Cadherins/metabolism , Catenins , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cell Line, Tumor , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Confocal , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Delta Catenin
15.
Mol Nutr Food Res ; 52(4): 393-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18306429

ABSTRACT

Mammalian target of rapamycin (mTOR) is a protein serine-threonine kinase that functions as a central element in signaling pathway involved in control of cell growth and proliferation. mTOR exists in at least two distinct multi-protein complexes, mTORC1 and mTORC2. mTOR kinase controls the translation machinery, in response to nutrients and growth factors, via activation of p70 ribosomal S6 kinase and inhibition of eukaryotic initiation factor-4E-binding protein. In this report, we review the mTOR signaling pathway and its interaction with food intake, insulin resistance, lifespan and adipogenic regulation during the molecular nutrition regulation.


Subject(s)
Protein Kinases/physiology , Animals , Energy Intake , Eukaryotic Initiation Factor-4G/physiology , Glucose/metabolism , Humans , Insulin Resistance , Mammals , Protein Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
16.
Development ; 134(15): 2863-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17611220

ABSTRACT

Translational control is crucial for proper timing of developmental events that take place in the absence of transcription, as in meiotic activation in oocytes, early embryogenesis in many organisms, and spermatogenesis. Here we show that a novel form of the translation initiation complex component eIF4G in Drosophila, eIF4G2, is required specifically for male germ cells to undergo meiotic division and proper spermatid differentiation. Flies mutant for eIF4G2 are viable and female fertile but male sterile. Spermatocytes form, but the germ cells in mutant males skip the major events of the meiotic divisions and form aberrant spermatids with large nuclei. Consistent with the failure to undergo the meiotic divisions, function of eIF4G2 is required post-transcriptionally for normal accumulation of the core cell cycle regulatory proteins Twine and CycB in mature spermatocytes. Loss of eIF4G2 function also causes widespread defects in spermatid differentiation. Although differentiation markers Dj and Fzo are expressed in late-stage eIF4G2 mutant germ cells, several key steps of spermatid differentiation fail, including formation of a compact mitochondrial derivative and full elongation. Our results suggest that an alternate form of the translation initiation machinery may be required for regulation and execution of key steps in male germ cell differentiation.


Subject(s)
Cell Differentiation , Drosophila Proteins/physiology , Eukaryotic Initiation Factor-4G/physiology , Gene Expression Regulation, Developmental , Meiosis/genetics , Protein Biosynthesis , Spermatids/cytology , Spermatozoa/cytology , Animals , Animals, Genetically Modified , Cell Cycle/genetics , Cell Differentiation/genetics , Cells, Cultured , Drosophila/genetics , Drosophila Proteins/genetics , Eukaryotic Initiation Factor-4G/genetics , Fertility/genetics , Male , Sequence Homology, Amino Acid , Testis/growth & development
17.
Development ; 134(15): 2851-61, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17611222

ABSTRACT

During spermatogenesis, cells coordinate differentiation with the meiotic cell cycle to generate functional gametes. We identified a novel gene, which we named off-schedule (ofs), as being essential for this coordinated control. During the meiotic G(2) phase, Drosophila ofs mutant germ cells do not reach their proper size and fail to execute meiosis or significant differentiation. The accumulation of four cell cycle regulators--Cyclin A, Boule, Twine and Roughex--is altered in these mutants, indicating that ofs reveals a novel branch of the pathway controlling meiosis and differentiation. Ofs is homologous to eukaryotic translation initiation factor eIF4G. The level of ofs expression in spermatocytes is much higher than for the known eIF4G ortholog (known as eIF-4G or eIF4G), suggesting that Ofs substitutes for this protein. Consistent with this, assays for association with mRNA cap complexes, as well as RNA-interference and phenotypic-rescue experiments, demonstrate that Ofs has eIF4G activity. Based on these studies, we speculate that spermatocytes monitor G(2) growth as one means to coordinate the initiation of meiotic division and differentiation.


Subject(s)
Cell Differentiation/genetics , Drosophila Proteins/physiology , Drosophila/genetics , Eukaryotic Initiation Factor-4G/physiology , Meiosis/genetics , Protein Biosynthesis , Spermatocytes/cytology , Animals , Animals, Genetically Modified , Cells, Cultured , Cyclin A/metabolism , Drosophila Proteins/genetics , Eukaryotic Initiation Factor-4G/genetics , Eye Proteins/genetics , Gene Expression Regulation , Gene Expression Regulation, Developmental , Male , Sequence Homology, Amino Acid , Spermatids/cytology , Spermatocytes/growth & development , Spermatocytes/metabolism , Tissue Distribution
18.
RNA ; 13(3): 374-84, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17237356

ABSTRACT

The eukaryotic initiation factor (eIF) 4G family plays a central role during translation initiation, bridging between the 5' and 3' ends of the mRNA via its N-terminal third while recruiting other factors and ribosomes through its central and C-terminal third. The protein p97/NAT1/DAP5 is homologous to the central and C-terminal thirds of eIF4G. p97 has long been considered to be a translational repressor under normal cellular conditions. Further, caspase cleavage liberates a p86 fragment that is thought to mediate cap-independent translation in apoptotic cells. We report here that, surprisingly, human p97 is polysome associated in proliferating cells and moves to stress granules in stressed, nonapoptotic cells. Tethered-function studies in living cells show that human p97 and p86 both can activate translation; however, we were unable to detect polysome association of p86 in apoptotic cells. We further characterized the zebrafish orthologs of p97, and found both to be expressed throughout embryonic development. Their simultaneous knockdown by morpholino injection led to impaired mesoderm formation and early embryonic lethality, indicating conservation of embryonic p97 function from fish to mammals. These data indicate that full-length p97 is a translational activator with essential role(s) in unstressed cells, suggesting a reassessment of current models of p97 function.


Subject(s)
Caspases/metabolism , Eukaryotic Initiation Factor-4G/physiology , Polyribosomes/metabolism , Protein Biosynthesis/genetics , Repressor Proteins/physiology , Amino Acid Sequence , Animals , Cytoplasmic Granules/chemistry , Eukaryotic Initiation Factor-4G/analysis , Eukaryotic Initiation Factor-4G/genetics , Humans , Molecular Sequence Data , Polyribosomes/chemistry , RNA, Messenger/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
19.
Mol Cell Biol ; 27(1): 147-56, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17060447

ABSTRACT

The tumor suppressor function of Programmed Cell Death 4 (Pdcd4) is achieved through interactions between Pdcd4 and components of the translation initiation complex, namely, the RNA helicase eIF4A and the scaffolding protein eIF4G. These interactions are mediated through two MA3 domains on the Pdcd4 molecule and result in inhibition of protein synthesis. We have solved the high-resolution crystal structure of the C-terminal MA3 (cMA3) domain of Pdcd4 in several crystal forms and demonstrated its similarity to the MA3 domain of eIF4G. As predicted by the structure, the cMA3 domain competes with eIF4Gc for binding to eIF4A and surprisingly is sufficient to inhibit translation initiation. Mutations that abolish eIF4A binding negate both functions of the cMA3. Interestingly mutations in the Akt phosphorylation site influenced neither cMA3 binding to eIF4A nor its ability to inhibit translation initiation. Finally, our structural analysis reveals MA3 domains to be a novel subfamily of VHS domains.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/physiology , Gene Expression Regulation , Protein Biosynthesis , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Amino Acid Sequence , Eukaryotic Initiation Factor-4A/physiology , Eukaryotic Initiation Factor-4G/physiology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
20.
EMBO J ; 25(17): 4008-19, 2006 Sep 06.
Article in English | MEDLINE | ID: mdl-16932749

ABSTRACT

p97 (also referred to as DAP5, NAT1 or eIF4G2) has been proposed to act as a repressor of protein synthesis. However, we found that p97 is abundantly expressed in proliferating cells and p97 is recruited to ribosomes following growth factor stimulation. We also report that p97 binds eIF2beta through its C-terminal domain and localizes to ribosome through its N-terminal MIF4G domain. When overexpressed, p97 increases reporter luciferase activity. In contrast, overexpression of the C-terminal two-thirds of eukaryotic initiation factor 4GI (eIF4GI), a region that shares significant homology with p97, or the N-terminal MIF4G domain of p97 markedly inhibits reporter activity, the rate of global translation and cell proliferation. Conversely, downregulation of p97 levels by RNA interference also decreases the rate of global translation and inhibits cell proliferation. This coincides with an increase in p27/Kip1 protein levels and a marked decrease in CDK2 kinase activity. Taken together, our results demonstrate that p97 is functionally different from the closely related C-terminal two-thirds of eIF4GI and it can positively promote protein synthesis and cell proliferation.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Proliferation , Eukaryotic Initiation Factor-4G/physiology , Ribosomes/physiology , Cell Line , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4G/genetics , Genes, Reporter , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Protein Binding , Protein Biosynthesis , RNA Interference , Ribosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...