Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.473
Filter
Add more filters

Publication year range
1.
Mol Cell ; 84(6): 995-997, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38518749

ABSTRACT

Chakrabarty et al.1 demonstrate that phospho-EIF2α (pEIF2α), the translation initiation factor that mediates the integrated stress response (ISR), is necessary and sufficient for the autophagic degradation of mitochondria following the addition of mitochondrial stressors.


Subject(s)
Mitochondria , Stress, Physiological , Phosphorylation , Mitochondria/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism
2.
Mol Cell ; 84(11): 2009-2010, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848688

ABSTRACT

In this issue, Diamond et al.1 and Kim et al.2 report that depletion of eIF4E leads to translational upregulation of GCN4, a key player in the integrated stress response, in an eIF2α phosphorylation-independent manner, suggesting a new mode of translational adaptation.


Subject(s)
Eukaryotic Initiation Factor-4E , Stress, Physiological , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Phosphorylation , Humans , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Protein Biosynthesis , Animals , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
3.
Mol Cell ; 84(3): 506-521.e11, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38159565

ABSTRACT

Regulated protein phosphorylation controls most cellular processes. The protein phosphatase PP1 is the catalytic subunit of many holoenzymes that dephosphorylate serine/threonine residues. How these enzymes recruit their substrates is largely unknown. Here, we integrated diverse approaches to elucidate how the PP1 non-catalytic subunit PPP1R15B (R15B) captures its full trimeric eIF2 substrate. We found that the substrate-recruitment module of R15B is largely disordered with three short helical elements, H1, H2, and H3. H1 and H2 form a clamp that grasps the substrate in a region remote from the phosphorylated residue. A homozygous N423D variant, adjacent to H1, reducing substrate binding and dephosphorylation was discovered in a rare syndrome with microcephaly, developmental delay, and intellectual disability. These findings explain how R15B captures its 125 kDa substrate by binding the far end of the complex relative to the phosphosite to present it for dephosphorylation by PP1, a paradigm of broad relevance.


Subject(s)
Catalytic Domain , Eukaryotic Initiation Factor-2 , Protein Phosphatase 1 , Humans , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism
4.
Mol Cell ; 84(11): 2135-2151.e7, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848692

ABSTRACT

In response to stress, eukaryotes activate the integrated stress response (ISR) via phosphorylation of eIF2α to promote the translation of pro-survival effector genes, such as GCN4 in yeast. Complementing the ISR is the target of rapamycin (TOR) pathway, which regulates eIF4E function. Here, we probe translational control in the absence of eIF4E in Saccharomyces cerevisiae. Intriguingly, we find that loss of eIF4E leads to de-repression of GCN4 translation. In addition, we find that de-repression of GCN4 translation is accompanied by neither eIF2α phosphorylation nor reduction in initiator ternary complex (TC). Our data suggest that when eIF4E levels are depleted, GCN4 translation is de-repressed via a unique mechanism that may involve faster scanning by the small ribosome subunit due to increased local concentration of eIF4A. Overall, our findings suggest that relative levels of eIF4F components are key to ribosome dynamics and may play important roles in translational control of gene expression.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Stress, Physiological , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Phosphorylation , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4F/genetics , Protein Biosynthesis , Gene Expression Regulation, Fungal , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/genetics , Signal Transduction , Ribosomes/metabolism , Ribosomes/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4A/genetics
5.
Annu Rev Biochem ; 85: 1-4, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27050288

ABSTRACT

Aging and longevity are controlled by a multiplicity of molecular and cellular signaling events that interface with environmental factors to maintain cellular homeostasis. Modulation of these pathways to extend life span, including insulin-like signaling and the response to dietary restriction, identified the cellular machineries and networks of protein homeostasis (proteostasis) and stress resistance pathways as critical players in the aging process. A decline of proteostasis capacity during aging leads to dysfunction of specific cell types and tissues, rendering the organism susceptible to a range of chronic diseases. This volume of the Annual Review of Biochemistry contains a set of two reviews addressing our current understanding of the molecular mechanisms underlying aging in model organisms and humans.


Subject(s)
Aging/genetics , Caenorhabditis elegans/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation, Developmental , Protein Serine-Threonine Kinases/genetics , Unfolded Protein Response , Aging/metabolism , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caloric Restriction , Eukaryotic Initiation Factor-2/metabolism , Homeostasis/genetics , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Proteolysis , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Annu Rev Biochem ; 85: 5-34, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145842

ABSTRACT

Dietary restriction (DR), a moderate reduction in food intake, improves health during aging and extends life span across multiple species. Specific nutrients, rather than overall calories, mediate the effects of DR, with protein and specific amino acids (AAs) playing a key role. Modulations of single dietary AAs affect traits including growth, reproduction, physiology, health, and longevity in animals. Epidemiological data in humans also link the quality and quantity of dietary proteins to long-term health. Intricate nutrient-sensing pathways fine tune the metabolic responses to dietary AAs in a highly conserved manner. In turn, these metabolic responses can affect the onset of insulin resistance, obesity, neurodegenerative disease, and other age-related diseases. In this review we discuss how AA requirements are shaped and how ingested AAs regulate a spectrum of homeostatic processes. Finally, we highlight the resulting opportunity to develop nutritional strategies to improve human health during aging.


Subject(s)
Aging/genetics , Amino Acids/metabolism , Diet, Protein-Restricted/methods , Dietary Proteins/metabolism , Gene Expression Regulation, Developmental , Protein Serine-Threonine Kinases/genetics , Aging/metabolism , Amino Acids/administration & dosage , Animals , Caloric Restriction , Dietary Proteins/administration & dosage , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Homeostasis/genetics , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/prevention & control , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Obesity/prevention & control , Protein Serine-Threonine Kinases/metabolism , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Proteostasis Deficiencies/prevention & control , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
Mol Cell ; 83(18): 3303-3313.e6, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37683648

ABSTRACT

Cells respond to intrinsic and extrinsic stresses by reducing global protein synthesis and activating gene programs necessary for survival. Here, we show that the integrated stress response (ISR) is driven by the non-canonical cap-binding protein eIF3d that acts as a critical effector to control core stress response orchestrators, the translation factor eIF2α and the transcription factor ATF4. We find that during persistent stress, eIF3d activates the translation of the kinase GCN2, inducing eIF2α phosphorylation and inhibiting general protein synthesis. In parallel, eIF3d upregulates the m6A demethylase ALKBH5 to drive 5' UTR-specific demethylation of stress response genes, including ATF4. Ultimately, this cascade converges on ATF4 expression by increasing mRNA engagement of translation machinery and enhancing ribosome bypass of upstream open reading frames (uORFs). Our results reveal that eIF3d acts in a life-or-death decision point during chronic stress and uncover a synergistic signaling mechanism in which translational cascades complement transcriptional amplification to control essential cellular processes.


Subject(s)
Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2 , 5' Untranslated Regions , Eukaryotic Initiation Factor-2/genetics , Open Reading Frames , Phosphorylation , RNA Cap-Binding Proteins , Humans
8.
Mol Cell ; 83(20): 3740-3753.e9, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37832546

ABSTRACT

Mitochondrial DNA double-strand breaks (mtDSBs) lead to the degradation of circular genomes and a reduction in copy number; yet, the cellular response in human cells remains elusive. Here, using mitochondrial-targeted restriction enzymes, we show that a subset of cells with mtDSBs exhibited defective mitochondrial protein import, reduced respiratory complexes, and loss of membrane potential. Electron microscopy confirmed the altered mitochondrial membrane and cristae ultrastructure. Intriguingly, mtDSBs triggered the integrated stress response (ISR) via the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by DELE1 and heme-regulated eIF2α kinase (HRI). When ISR was inhibited, the cells experienced intensified mitochondrial defects and slower mtDNA recovery post-breakage. Lastly, through proteomics, we identified ATAD3A-a membrane-bound protein interacting with nucleoids-as potentially pivotal in relaying signals from impaired genomes to the inner mitochondrial membrane. In summary, our study delineates the cascade connecting damaged mitochondrial genomes to the cytoplasm and highlights the significance of the ISR in maintaining mitochondrial homeostasis amid genome instability.


Subject(s)
Mitochondria , eIF-2 Kinase , Humans , Mitochondria/genetics , Mitochondria/metabolism , eIF-2 Kinase/metabolism , Cytoplasm/metabolism , Phosphorylation , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Eukaryotic Initiation Factor-2/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
9.
Cell ; 158(5): 1159-1172, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171414

ABSTRACT

In Alzheimer's disease (AD) brain, exposure of axons to Aß causes pathogenic changes that spread retrogradely by unknown mechanisms, affecting the entire neuron. We found that locally applied Aß1-42 initiates axonal synthesis of a defined set of proteins including the transcription factor ATF4. Inhibition of local translation and retrograde transport or knockdown of axonal Atf4 mRNA abolished Aß-induced ATF4 transcriptional activity and cell loss. Aß1-42 injection into the dentate gyrus (DG) of mice caused loss of forebrain neurons whose axons project to the DG. Protein synthesis and Atf4 mRNA were upregulated in these axons, and coinjection of Atf4 siRNA into the DG reduced the effects of Aß1-42 in the forebrain. ATF4 protein and transcripts were found with greater frequency in axons in the brain of AD patients. These results reveal an active role for intra-axonal translation in neurodegeneration and identify ATF4 as a mediator for the spread of AD pathology.


Subject(s)
Activating Transcription Factor 4/analysis , Alzheimer Disease/pathology , Brain/pathology , Activating Transcription Factor 4/metabolism , Amyloid beta-Peptides/genetics , Animals , Axons/metabolism , Brain/cytology , Brain Chemistry , Eukaryotic Initiation Factor-2/metabolism , Hippocampus , Humans , Mice, Inbred C57BL , Rats , Transcription Factor CHOP/metabolism
10.
Mol Cell ; 81(1): 88-103.e6, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33220178

ABSTRACT

The small molecule ISRIB antagonizes the activation of the integrated stress response (ISR) by phosphorylated translation initiation factor 2, eIF2(αP). ISRIB and eIF2(αP) bind distinct sites in their common target, eIF2B, a guanine nucleotide exchange factor for eIF2. We have found that ISRIB-mediated acceleration of eIF2B's nucleotide exchange activity in vitro is observed preferentially in the presence of eIF2(αP) and is attenuated by mutations that desensitize eIF2B to the inhibitory effect of eIF2(αP). ISRIB's efficacy as an ISR inhibitor in cells also depends on presence of eIF2(αP). Cryoelectron microscopy (cryo-EM) showed that engagement of both eIF2B regulatory sites by two eIF2(αP) molecules remodels both the ISRIB-binding pocket and the pockets that would engage eIF2α during active nucleotide exchange, thereby discouraging both binding events. In vitro, eIF2(αP) and ISRIB reciprocally opposed each other's binding to eIF2B. These findings point to antagonistic allostery in ISRIB action on eIF2B, culminating in inhibition of the ISR.


Subject(s)
Acetamides/chemistry , Cyclohexylamines/chemistry , Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2/chemistry , Allosteric Regulation , Animals , Binding Sites , CHO Cells , Cricetulus , Cryoelectron Microscopy , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , HeLa Cells , Humans , Phosphorylation
11.
Mol Cell ; 81(9): 1879-1889.e6, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33743194

ABSTRACT

The conserved Gcn2 protein kinase mediates cellular adaptations to amino acid limitation through translational control of gene expression that is exclusively executed by phosphorylation of the α-subunit of the eukaryotic translation initiation factor 2 (eIF2α). Using quantitative phosphoproteomics, however, we discovered that Gcn2 targets auxiliary effectors to modulate translation. Accordingly, Gcn2 also phosphorylates the ß-subunit of the trimeric eIF2 G protein complex to promote its association with eIF5, which prevents spontaneous nucleotide exchange on eIF2 and thereby restricts the recycling of the initiator methionyl-tRNA-bound eIF2-GDP ternary complex in amino-acid-starved cells. This mechanism contributes to the inhibition of translation initiation in parallel to the sequestration of the nucleotide exchange factor eIF2B by phosphorylated eIF2α. Gcn2 further phosphorylates Gcn20 to antagonize, in an inhibitory feedback loop, the formation of the Gcn2-stimulatory Gcn1-Gcn20 complex. Thus, Gcn2 plays a substantially more intricate role in controlling translation initiation than hitherto appreciated.


Subject(s)
Amino Acids/deficiency , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proteomics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Feedback, Physiological , Gene Expression Regulation, Fungal , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
12.
Mol Cell ; 81(3): 614-628.e4, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33338396

ABSTRACT

Stalling during translation triggers ribosome quality control (RQC) to maintain proteostasis. Recently, stalling has also been linked to the activation of integrated stress response (ISR) by Gcn2. How the two processes are coordinated is unclear. Here, we show that activation of RQC by Hel2 suppresses that of Gcn2. We further show that Hel2 and Gcn2 are activated by a similar set of agents that cause ribosome stalling, with maximal activation of Hel2 observed at a lower frequency of stalling. Interestingly, inactivation of one pathway was found to result in the overactivation of the other, suggesting that both are activated by the same signal of ribosome collisions. Notably, the processes do not appear to be in direct competition with each other; ISR prefers a vacant A site, whereas RQC displays no preference. Collectively, our findings provide important details about how multiple pathways that recognize stalled ribosomes coordinate to mount the appropriate response.


Subject(s)
Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Stress, Physiological , Ubiquitin-Protein Ligases/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Serine-Threonine Kinases/genetics , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Stress, Physiological/genetics , Time Factors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
13.
Cell ; 153(5): 1108-19, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706745

ABSTRACT

Eukaryotic translation initiation begins with assembly of a 43S preinitiation complex. First, methionylated initiator methionine transfer RNA (Met-tRNAi(Met)), eukaryotic initiation factor (eIF) 2, and guanosine triphosphate form a ternary complex (TC). The TC, eIF3, eIF1, and eIF1A cooperatively bind to the 40S subunit, yielding the 43S preinitiation complex, which is ready to attach to messenger RNA (mRNA) and start scanning to the initiation codon. Scanning on structured mRNAs additionally requires DHX29, a DExH-box protein that also binds directly to the 40S subunit. Here, we present a cryo-electron microscopy structure of the mammalian DHX29-bound 43S complex at 11.6 Å resolution. It reveals that eIF2 interacts with the 40S subunit via its α subunit and supports Met-tRNAi(Met) in an unexpected P/I orientation (eP/I). The structural core of eIF3 resides on the back of the 40S subunit, establishing two principal points of contact, whereas DHX29 binds around helix 16. The structure provides insights into eukaryote-specific aspects of translation, including the mechanism of action of DHX29.


Subject(s)
Mammals/metabolism , Peptide Chain Initiation, Translational , RNA Helicases/chemistry , RNA, Ribosomal/chemistry , Ribonucleoproteins/chemistry , Animals , Base Sequence , Cell-Free System , Cryoelectron Microscopy , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Humans , Mammals/genetics , Models, Molecular , Molecular Sequence Data , RNA Helicases/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal, 18S/chemistry , RNA, Ribosomal, 18S/metabolism , Rabbits , Ribonucleoproteins/metabolism
14.
Cell ; 155(2): 321-32, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120133

ABSTRACT

Olfactory receptor (OR) expression requires the transcriptional activation of 1 out of 1,000s of OR alleles and a feedback signal that preserves this transcriptional choice. The mechanism by which olfactory sensory neurons (OSNs) detect ORs to signal to the nucleus remains elusive. Here, we show that OR proteins generate this feedback by activating the unfolded protein response (UPR). OR expression induces Perk-mediated phosphorylation of the translation initiation factor eif2α causing selective translation of activating transcription factor 5 (ATF5). ATF5 induces the transcription of adenylyl cyclase 3 (Adcy3), which relieves the UPR. Our data provide a role for the UPR in defining neuronal identity and cell fate commitment and support a two-step model for the feedback signal: (1) OR protein, as a stress stimulus, alters the translational landscape of the OSN and induces Adcy3 expression; (2), Adcy3 relieves that stress, restores global translation, and makes OR choice permanent.


Subject(s)
Feedback, Physiological , Neurons/metabolism , Receptors, Odorant/metabolism , Unfolded Protein Response , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Adenylyl Cyclases/metabolism , Animals , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Mice , Mice, Knockout , Neurons/cytology , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , eIF-2 Kinase/metabolism
15.
Mol Cell ; 77(4): 913-925.e4, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31812349

ABSTRACT

Regulation of translation is essential during stress. However, the precise sets of proteins regulated by the key translational stress responses-the integrated stress response (ISR) and mTORC1-remain elusive. We developed multiplexed enhanced protein dynamics (mePROD) proteomics, adding signal amplification to dynamic-SILAC and multiplexing, to enable measuring acute changes in protein synthesis. Treating cells with ISR/mTORC1-modulating stressors, we showed extensive translatome modulation with ∼20% of proteins synthesized at highly reduced rates. Comparing translation-deficient sub-proteomes revealed an extensive overlap demonstrating that target specificity is achieved on protein level and not by pathway activation. Titrating cap-dependent translation inhibition confirmed that synthesis of individual proteins is controlled by intrinsic properties responding to global translation attenuation. This study reports a highly sensitive method to measure relative translation at the nascent chain level and provides insight into how the ISR and mTORC1, two key cellular pathways, regulate the translatome to guide cellular survival upon stress.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Biosynthesis , Proteomics/methods , HeLa Cells , Humans , Stress, Physiological/genetics , Unfolded Protein Response
16.
Mol Cell ; 79(4): 546-560.e7, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32589964

ABSTRACT

Translational control targeting the initiation phase is central to the regulation of gene expression. Understanding all of its aspects requires substantial technological advancements. Here we modified yeast translation complex profile sequencing (TCP-seq), related to ribosome profiling, and adapted it for mammalian cells. Human TCP-seq, capable of capturing footprints of 40S subunits (40Ss) in addition to 80S ribosomes (80Ss), revealed that mammalian and yeast 40Ss distribute similarly across 5'TRs, indicating considerable evolutionary conservation. We further developed yeast and human selective TCP-seq (Sel-TCP-seq), enabling selection of 40Ss and 80Ss associated with immuno-targeted factors. Sel-TCP-seq demonstrated that eIF2 and eIF3 travel along 5' UTRs with scanning 40Ss to successively dissociate upon AUG recognition; notably, a proportion of eIF3 lingers on during the initial elongation cycles. Highlighting Sel-TCP-seq versatility, we also identified four initiating 48S conformational intermediates, provided novel insights into ATF4 and GCN4 mRNA translational control, and demonstrated co-translational assembly of initiation factor complexes.


Subject(s)
Multiprotein Complexes/metabolism , Peptide Initiation Factors/metabolism , Protein Biosynthesis , Ribosomes/metabolism , 5' Untranslated Regions , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Codon, Initiator , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-3/genetics , Eukaryotic Initiation Factor-3/metabolism , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Peptide Initiation Factors/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
17.
Mol Cell ; 79(4): 588-602.e6, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32615089

ABSTRACT

The ribosome-associated protein quality control (RQC) system that resolves stalled translation events is activated when ribosomes collide and form disome, trisome, or higher-order complexes. However, it is unclear whether this system distinguishes collision complexes formed on defective mRNAs from those with functional roles on endogenous transcripts. Here, we performed disome and trisome footprint profiling in yeast and found collisions were enriched on diverse sequence motifs known to slow translation. When 60S recycling was inhibited, disomes accumulated at stop codons and could move into the 3' UTR to reinitiate translation. The ubiquitin ligase and RQC factor Hel2/ZNF598 generally recognized collisions but did not induce degradation of endogenous transcripts. However, loss of Hel2 triggered the integrated stress response, via phosphorylation of eIF2α, thus linking these pathways. Our results suggest that Hel2 has a role in sensing ribosome collisions on endogenous mRNAs, and such events may be important for cellular homeostasis.


Subject(s)
DNA Footprinting/methods , Genome, Fungal , Ribosomes/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Anisomycin/pharmacology , Codon, Terminator , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Mutation , Phosphorylation , RNA Stability , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics
18.
Mol Cell ; 77(4): 875-886.e7, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31836389

ABSTRACT

Dysregulation of cellular protein synthesis is linked to a variety of diseases. Mutations in EIF2S3, encoding the γ subunit of the heterotrimeric eukaryotic translation initiation factor eIF2, cause MEHMO syndrome, an X-linked intellectual disability disorder. Here, using patient-derived induced pluripotent stem cells, we show that a mutation at the C terminus of eIF2γ impairs CDC123 promotion of eIF2 complex formation and decreases the level of eIF2-GTP-Met-tRNAiMet ternary complexes. This reduction in eIF2 activity results in dysregulation of global and gene-specific protein synthesis and enhances cell death upon stress induction. Addition of the drug ISRIB, an activator of the eIF2 guanine nucleotide exchange factor, rescues the cell growth, translation, and neuronal differentiation defects associated with the EIF2S3 mutation, offering the possibility of therapeutic intervention for MEHMO syndrome.


Subject(s)
Acetamides/pharmacology , Cyclohexylamines/pharmacology , Epilepsy/genetics , Eukaryotic Initiation Factor-2/genetics , Genitalia/abnormalities , Hypogonadism/genetics , Mental Retardation, X-Linked/genetics , Microcephaly/genetics , Mutation , Obesity/genetics , Protein Biosynthesis/drug effects , Apoptosis , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology
19.
Annu Rev Neurosci ; 42: 227-247, 2019 07 08.
Article in English | MEDLINE | ID: mdl-30909783

ABSTRACT

Microsatellite mutations involving the expansion of tri-, tetra-, penta-, or hexanucleotide repeats cause more than 40 different neurological disorders. Although, traditionally, the position of the repeat within or outside of an open reading frame has been used to focus research on disease mechanisms involving protein loss of function, protein gain of function, or RNA gain of function, the discoveries of bidirectional transcription and repeat-associated non-ATG (RAN) have blurred these distinctions. Here we review what is known about RAN proteins in disease, the mechanisms by which they are produced, and the novel therapeutic opportunities they provide.


Subject(s)
DNA Repeat Expansion/genetics , Nerve Tissue Proteins/genetics , Nervous System Diseases/genetics , Protein Biosynthesis , Codon, Initiator/genetics , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/physiology , Gain of Function Mutation , Genetic Code , Humans , Loss of Function Mutation , Microsatellite Repeats/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Transcription, Genetic
20.
Mol Cell ; 73(3): 474-489.e5, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30595434

ABSTRACT

Local translation is rapidly regulated by extrinsic signals during neural wiring, but its control mechanisms remain elusive. Here we show that the extracellular cue Sema3A induces an initial burst in local translation that precisely controls phosphorylation of the translation initiation factor eIF2α via the unfolded protein response (UPR) kinase PERK. Strikingly, in contrast to canonical UPR signaling, Sema3A-induced eIF2α phosphorylation bypasses global translational repression and underlies an increase in local translation through differential activity of eIF2B mediated by protein phosphatase 1. Ultrasensitive proteomics analysis of axons reveals 75 proteins translationally controlled via the Sema3A-p-eIF2α pathway. These include proteostasis- and actin cytoskeleton-related proteins but not canonical stress markers. Finally, we show that PERK signaling is needed for directional axon migration and visual pathway development in vivo. Thus, our findings reveal a noncanonical eIF2 signaling pathway that controls selective changes in axon translation and is required for neural wiring.


Subject(s)
Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-2/metabolism , Neurogenesis , Retinal Ganglion Cells/metabolism , Xenopus Proteins/metabolism , Animals , Axons/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2B/genetics , Female , Male , Neurogenesis/drug effects , Phosphorylation , Protein Interaction Maps , Proteomics/methods , Retinal Ganglion Cells/drug effects , Semaphorin-3A/metabolism , Semaphorin-3A/pharmacology , Signal Transduction , Tissue Culture Techniques , Xenopus laevis/embryology , Xenopus laevis/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL