Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32.844
Filter
1.
Proc Natl Acad Sci U S A ; 121(35): e2401743121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159370

ABSTRACT

While the centrality of posttranscriptional modifications to RNA biology has long been acknowledged, the function of the vast majority of modified sites remains to be discovered. Illustrative of this, there is not yet a discrete biological role assigned for one of the most highly conserved modifications, 5-methyluridine at position 54 in tRNAs (m5U54). Here, we uncover contributions of m5U54 to both tRNA maturation and protein synthesis. Our mass spectrometry analyses demonstrate that cells lacking the enzyme that installs m5U in the T-loop (TrmA in Escherichia coli, Trm2 in Saccharomyces cerevisiae) exhibit altered tRNA modification patterns. Furthermore, m5U54-deficient tRNAs are desensitized to small molecules that prevent translocation in vitro. This finding is consistent with our observations that relative to wild-type cells, trm2Δ cell growth and transcriptome-wide gene expression are less perturbed by translocation inhibitors. Together our data suggest a model in which m5U54 acts as an important modulator of tRNA maturation and translocation of the ribosome during protein synthesis.


Subject(s)
Escherichia coli , RNA, Transfer , Ribosomes , Saccharomyces cerevisiae , Uridine , RNA, Transfer/metabolism , RNA, Transfer/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Ribosomes/metabolism , Uridine/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , RNA Processing, Post-Transcriptional , Protein Biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , tRNA Methyltransferases/metabolism , tRNA Methyltransferases/genetics
2.
Sci Rep ; 14(1): 18149, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103467

ABSTRACT

Cryogenic electron microscopy (cryo-EM) has emerged as a powerful method for the determination of structures of complex biological molecules. The accurate characterisation of the dynamics of such systems, however, remains a challenge. To address this problem, we introduce cryoENsemble, a method that applies Bayesian reweighting to conformational ensembles derived from molecular dynamics simulations to improve their agreement with cryo-EM data, thus enabling the extraction of dynamics information. We illustrate the use of cryoENsemble to determine the dynamics of the ribosome-bound state of the co-translational chaperone trigger factor (TF). We also show that cryoENsemble can assist with the interpretation of low-resolution, noisy or unaccounted regions of cryo-EM maps. Notably, we are able to link an unaccounted part of the cryo-EM map to the presence of another protein (methionine aminopeptidase, or MetAP), rather than to the dynamics of TF, and model its TF-bound state. Based on these results, we anticipate that cryoENsemble will find use for challenging heterogeneous cryo-EM maps for biomolecular systems encompassing dynamic components.


Subject(s)
Bayes Theorem , Cryoelectron Microscopy , Molecular Dynamics Simulation , Cryoelectron Microscopy/methods , Ribosomes/ultrastructure , Ribosomes/chemistry , Ribosomes/metabolism , Protein Conformation
3.
Front Immunol ; 15: 1411957, 2024.
Article in English | MEDLINE | ID: mdl-39114656

ABSTRACT

Introduction: CD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide-MHC-I complex by the T-cell receptor (TCR) and co-stimulation. This cognate interaction promotes the organisation of intimate cell-cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell-cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 70S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs. Methods: Here, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 70S by puromycin. Results: We observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs. Discussion: Cytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs.


Subject(s)
CD8-Positive T-Lymphocytes , Cytosol , Lymphocyte Activation , Mitochondria , Protein Biosynthesis , Receptors, Antigen, T-Cell , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Lymphocyte Activation/immunology , Cytosol/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Mitochondria/immunology , Cytoskeleton/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Ribosomes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
4.
Sci Rep ; 14(1): 19421, 2024 08 21.
Article in English | MEDLINE | ID: mdl-39169208

ABSTRACT

Protein expression in vivo is predominately controlled via regulatory feedback mechanisms that adjust the level of mRNA transcription. However for positive sense single-stranded RNA viruses, protein expression is often controlled via secondary structural elements, such as internal ribosomal entry sites, that are encoded within the mRNA. The self-regulation of mRNA translation observed in this class of viruses suggests that it may be possible to design mRNAs that self-regulate their protein expression, enabling the creation of mRNAs for vaccines and other synthetic biology applications where protein levels in the cell can be tightly controlled without feedback to a transcriptional mechanism. As a proof of concept, I design a polycistronic mRNA based on bacteriophage MS2, where the upstream gene is capable of repressing synthesis of the downstream gene. Using a computational tool that simulates ribosome kinetics and the co-translational folding of the mRNA in response, I show that mutations to the mRNA can be identified which enhance the efficiency of the translation and the repression of the downstream gene. The results of this study open up the possibility of designing bespoke mRNA gene circuits in which the amount of protein synthesised in cells are self-regulated for therapeutic or antigenic purposes.


Subject(s)
Gene Regulatory Networks , RNA, Messenger , RNA, Messenger/genetics , RNA, Messenger/metabolism , Levivirus/genetics , Protein Biosynthesis , Ribosomes/metabolism , Ribosomes/genetics , Synthetic Biology/methods , Gene Expression Regulation
5.
Sci Signal ; 17(848): eadl1030, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39106320

ABSTRACT

Hexanucleotide repeat expansion in the C9ORF72 gene is the most frequent inherited cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The expansion results in multiple dipeptide repeat proteins, among which arginine-rich poly-GR proteins are highly toxic to neurons and decrease the rate of protein synthesis. We investigated whether the effect on protein synthesis contributes to neuronal dysfunction and degeneration. We found that the expression of poly-GR proteins inhibited global translation by perturbing translation elongation. In iPSC-differentiated neurons, the translation of transcripts with relatively slow elongation rates was further slowed, and stalled, by poly-GR. Elongation stalling increased ribosome collisions and induced a ribotoxic stress response (RSR) mediated by ZAKα that increased the phosphorylation of the kinase p38 and promoted cell death. Knockdown of ZAKα or pharmacological inhibition of p38 ameliorated poly-GR-induced toxicity and improved the survival of iPSC-derived neurons from patients with C9ORF72-ALS/FTD. Our findings suggest that targeting the RSR may be neuroprotective in patients with ALS/FTD caused by repeat expansion in C9ORF72.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , DNA Repeat Expansion , Frontotemporal Dementia , Induced Pluripotent Stem Cells , Neurons , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Humans , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Neurons/metabolism , Neurons/pathology , Induced Pluripotent Stem Cells/metabolism , DNA Repeat Expansion/genetics , Peptide Chain Elongation, Translational , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Stress, Physiological/genetics , Ribosomes/metabolism , Ribosomes/genetics
6.
Nat Commun ; 15(1): 6839, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122697

ABSTRACT

There has been a dramatic increase in the identification of non-canonical translation and a significant expansion of the protein-coding genome. Among the strategies used to identify unannotated small Open Reading Frames (smORFs) that encode microproteins, Ribosome profiling (Ribo-Seq) is the gold standard for the annotation of novel coding sequences by reporting on smORF translation. In Ribo-Seq, ribosome-protected footprints (RPFs) that map to multiple genomic sites are removed since they cannot be unambiguously assigned to a specific genomic location. Furthermore, RPFs necessarily result in short (25-34 nucleotides) reads, increasing the chance of multi-mapping alignments, such that smORFs residing in these regions cannot be identified by Ribo-Seq. Moreover, it has been challenging to identify protein evidence for Ribo-Seq. To solve this, we developed Rp3, a pipeline that integrates proteogenomics and Ribosome profiling to provide unambiguous evidence for a subset of microproteins missed by current Ribo-Seq pipelines. Here, we show that Rp3 maximizes proteomics detection and confidence of microprotein-encoding smORFs.


Subject(s)
Open Reading Frames , Proteogenomics , Ribosomes , Ribosomes/metabolism , Ribosomes/genetics , Proteogenomics/methods , Open Reading Frames/genetics , Protein Biosynthesis , Humans , Proteomics/methods , Proteins/genetics , Proteins/metabolism , Ribosome Profiling
7.
Nat Commun ; 15(1): 6579, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097616

ABSTRACT

Bacteria often evolve antibiotic resistance through mutagenesis. However, the processes causing the mutagenesis have not been fully resolved. Here, we find that a broad range of ribosome-targeting antibiotics cause mutations through an underexplored pathway. Focusing on the clinically important aminoglycoside gentamicin, we find that the translation inhibitor causes genome-wide premature stalling of RNA polymerase (RNAP) in a loci-dependent manner. Further analysis shows that the stalling is caused by the disruption of transcription-translation coupling. Anti-intuitively, the stalled RNAPs subsequently induce lesions to the DNA via transcription-coupled repair. While most of the bacteria are killed by genotoxicity, a small subpopulation acquires mutations via SOS-induced mutagenesis. Given that these processes are triggered shortly after antibiotic addition, resistance rapidly emerges in the population. Our work reveals a mechanism of action of ribosomal antibiotics, illustrates the importance of dissecting the complex interplay between multiple molecular processes in understanding antibiotic efficacy, and suggests new strategies for countering the development of resistance.


Subject(s)
Anti-Bacterial Agents , DNA-Directed RNA Polymerases , Drug Resistance, Bacterial , Genomic Instability , Gentamicins , Ribosomes , Anti-Bacterial Agents/pharmacology , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Ribosomes/metabolism , Ribosomes/drug effects , Gentamicins/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Mutation , Mutagenesis , Transcription, Genetic/drug effects , Protein Biosynthesis/drug effects
8.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125999

ABSTRACT

Elastin, a key structural protein essential for the elasticity of the skin and elastogenic tissues, degrades with age. Replenishing elastin holds promise for anti-aging cosmetics and the supplementation of elastic activities of the cardiovascular system. We employed RiboScreenTM, a technology for identifying molecules that enhance the production of specific proteins, to target the production of tropoelastin. We make use of RiboScreenTM in two crucial steps: first, to pinpoint a target ribosomal protein (TRP), which acts as a switch to increase the production of the protein of interest (POI), and second, to identify small molecules that activate this ribosomal protein switch. Using RiboScreenTM, we identified ribosomal protein L40, henceforth eL40, as a TRP switch to boost tropoelastin production. Drug discovery identified a small-molecule hit that binds to eL40. In-cell treatment demonstrated activity of the eL40 ligand and delivered increased tropoelastin production levels in a dose-dependent manner. Thus, we demonstrate that RiboScreenTM can successfully identify a small-molecule hit capable of selectively enhancing tropoelastin production. This compound has the potential to be developed for topical or systemic applications to promote skin rejuvenation and to supplement elastic functionality within the cardiovascular system.


Subject(s)
Elastin , Ribosomal Proteins , Ribosomes , Tropoelastin , Tropoelastin/metabolism , Tropoelastin/genetics , Humans , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Elastin/metabolism , Elastin/genetics , Ribosomes/metabolism , Ribosomes/drug effects , Ligands , Small Molecule Libraries/pharmacology
9.
Nat Commun ; 15(1): 6692, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107322

ABSTRACT

Translation initiation is a highly regulated step needed for protein synthesis. Most cell-based mechanistic work on translation initiation has been done using non-stressed cells growing in medium with sufficient nutrients and oxygen. This has yielded our current understanding of 'canonical' translation initiation, involving recognition of the mRNA cap by eIF4E1 followed by successive recruitment of initiation factors and the ribosome. Many cells, however, such as tumor cells, are exposed to stresses such as hypoxia, low nutrients or proteotoxic stress. This leads to inactivation of mTORC1 and thereby inactivation of eIF4E1. Hence the question arises how cells translate mRNAs under such stress conditions. We study here how mRNAs are translated in an eIF4E1-independent manner by blocking eIF4E1 using a constitutively active version of eIF4E-binding protein (4E-BP). Via ribosome profiling we identify a subset of mRNAs that are still efficiently translated when eIF4E1 is inactive. We find that these mRNAs preferentially release eIF4E1 when eIF4E1 is inactive and bind instead to eIF3d via its cap-binding pocket. eIF3d then enables these mRNAs to be efficiently translated due to its cap-binding activity. In sum, our work identifies eIF3d-dependent translation as a major mechanism enabling mRNA translation in an eIF4E-independent manner.


Subject(s)
Eukaryotic Initiation Factor-3 , Eukaryotic Initiation Factor-4E , Protein Biosynthesis , RNA, Messenger , Ribosomes , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-3/genetics , Humans , RNA, Messenger/metabolism , RNA, Messenger/genetics , Ribosomes/metabolism , Protein Binding , RNA Caps/metabolism , HEK293 Cells , Peptide Chain Initiation, Translational , Cell Cycle Proteins , Adaptor Proteins, Signal Transducing
10.
Nat Commun ; 15(1): 6873, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39127721

ABSTRACT

Ribosomes are regulated by evolutionarily conserved ubiquitination/deubiquitination events. We uncover the role of the deubiquitinase OTUD6 in regulating global protein translation through deubiquitination of the RPS7/eS7 subunit on the free 40 S ribosome in vivo in Drosophila. Coimmunoprecipitation and enrichment of monoubiquitinated proteins from catalytically inactive OTUD6 flies reveal RPS7 as the ribosomal substrate. The 40 S protein RACK1 and E3 ligases CNOT4 and RNF10 function upstream of OTUD6 to regulate alkylation stress. OTUD6 interacts with RPS7 specifically on the free 40 S, and not on 43 S/48 S initiation complexes or the translating ribosome. Global protein translation levels are bidirectionally regulated by OTUD6 protein abundance. OTUD6 protein abundance is physiologically regulated in aging and in response to translational and alkylation stress. Thus, OTUD6 may promote translation initiation, the rate limiting step in protein translation, by titering the amount of 40 S ribosome that recycles.


Subject(s)
Drosophila Proteins , Protein Biosynthesis , Ribosomal Proteins , Ubiquitination , Animals , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Ribosomes/metabolism , Stress, Physiological , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Genome Biol Evol ; 16(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39119893

ABSTRACT

Ribosomes are ribonucleoprotein complexes highly conserved across all domains of life. The size differences of ribosomal RNAs (rRNAs) can be mainly attributed to variable regions termed expansion segments (ESs) protruding out from the ribosomal surface. The ESs were found to be involved in a range of processes including ribosome biogenesis and maturation, translation, and co-translational protein modification. Here, we analyze the rRNAs of the yeasts from the Magnusiomyces/Saprochaete clade belonging to the basal lineages of the subphylum Saccharomycotina. We find that these yeasts are missing more than 400 nt from the 25S rRNA and 150 nt from the 18S rRNAs when compared to their canonical counterparts in Saccharomyces cerevisiae. The missing regions mostly map to ESs, thus representing a shift toward a minimal rRNA structure. Despite the structural changes in rRNAs, we did not identify dramatic alterations in the ribosomal protein inventories. We also show that the size-reduced rRNAs are not limited to the species of the Magnusiomyces/Saprochaete clade, indicating that the shortening of ESs happened independently in several other lineages of the subphylum Saccharomycotina.


Subject(s)
RNA, Ribosomal , Ribosomes , RNA, Ribosomal/genetics , Ribosomes/metabolism , Ribosomes/genetics , Phylogeny , Ribosomal Proteins/genetics , Saccharomycetales/genetics , Saccharomycetales/classification , Saccharomycetales/metabolism , RNA, Ribosomal, 18S/genetics , Saccharomyces cerevisiae/genetics , Evolution, Molecular
12.
J Med Virol ; 96(8): e29869, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39165093

ABSTRACT

Epstein-Barr virus (EBV) is a highly successful pathogen that infects ~95% of the adult population and is associated with diverse cancers and autoimmune diseases. The most abundant viral factor in latently infected cells is not a protein but a noncoding RNA called EBV-encoded RNA 1 (EBER1). Even though EBER1 is highly abundant and was discovered over forty years ago, the function of EBER1 has remained elusive. EBER1 interacts with the ribosomal protein L22, which normally suppresses the expression of its paralog L22-like 1 (L22L1). Here we show that when L22 binds EBER1, it cannot suppress L22L1, resulting in L22L1 being expressed and incorporated into ribosomes. We further show that L22L1-containing ribosomes preferentially translate mRNAs involved in the oxidative phosphorylation pathway. Moreover, upregulation of L22L1 is indispensable for growth transformation and immortalization of resting B cells upon EBV infection. Taken together, our results suggest that the function of EBER1 is to modulate host gene expression at the translational level, thus bypassing the need for dysregulating host gene transcription.


Subject(s)
Herpesvirus 4, Human , Oxidative Phosphorylation , RNA, Viral , Ribosomal Proteins , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Humans , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , B-Lymphocytes/virology , Host-Pathogen Interactions/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Ribosomes/metabolism , Ribosomes/genetics , RNA-Binding Proteins
13.
Nat Commun ; 15(1): 7188, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169056

ABSTRACT

The transcriptional control of sporulation in Bacillus subtilis is reasonably well understood, but its translational control is underexplored. Here, we use RNA-seq, ribosome profiling and fluorescence microscopy to study the translational dynamics of B. subtilis sporulation. We identify two events of translation silencing and describe spatiotemporal changes in subcellular localization of ribosomes during sporulation. We investigate the potential regulatory role of ribosomes during sporulation using a strain lacking zinc-independent paralogs of three zinc-dependent ribosomal proteins (L31, L33 and S14). The mutant strain exhibits delayed sporulation, reduced germination efficiency, dysregulated translation of metabolic and sporulation-related genes, and disruptions in translation silencing, particularly in late sporulation.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Gene Expression Regulation, Bacterial , Protein Biosynthesis , Ribosomal Proteins , Ribosomes , Spores, Bacterial , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacillus subtilis/physiology , Spores, Bacterial/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/growth & development , Ribosomes/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Mutation , Microscopy, Fluorescence
14.
Nat Commun ; 15(1): 6817, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122673

ABSTRACT

Arboviruses can be paternally transmitted by male insects to offspring for long-term persistence, but the mechanism remains largely unknown. Here, we use a model system of a destructive rice reovirus and its leafhopper vector to find that insect ribosome-rescuer Pelo-Hbs1 complex expressed on the sperm surface mediates paternal arbovirus transmission. This occurs through targeting virus-containing tubules constituted by viral nonstructural protein Pns11 to sperm surface via Pns11-Pelo interaction. Tubule assembly is dependent on Hsp70 activity, while Pelo-Hbs1 complex inhibits tubule assembly via suppressing Hsp70 activity. However, virus-activated ubiquitin ligase E3 mediates Pelo ubiquitinated degradation, synergistically causing Hbs1 degradation. Importantly, Pns11 effectively competes with Pelo for binding to E3, thus antagonizing E3-mediated Pelo-Hbs1 degradation. These processes cause a slight reduction of Pelo-Hbs1 complex in infected testes, promoting effective tubule assembly. Our findings provide insight into how insect sperm-specific Pelo-Hbs1 complex is modulated to promote paternal virus transmission without disrupting sperm function.


Subject(s)
Hemiptera , Insect Proteins , Spermatozoa , Animals , Male , Spermatozoa/metabolism , Spermatozoa/virology , Hemiptera/virology , Hemiptera/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics , Arboviruses , HSP70 Heat-Shock Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Reoviridae/physiology , Insect Vectors/virology , Insect Vectors/metabolism , Ribosomes/metabolism , Arbovirus Infections/transmission , Arbovirus Infections/metabolism , Arbovirus Infections/virology
15.
J Phys Chem B ; 128(29): 7002-7021, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39012038

ABSTRACT

Ribosomes bind to many metabolic enzymes and change their activity. A general mechanism for ribosome-mediated amplification of metabolic enzyme activity, RAMBO, was formulated and elucidated for the glycolytic enzyme triosephosphate isomerase, TPI. The RAMBO effect results from a ribosome-dependent electric field-substrate dipole interaction energy that can increase or decrease the ground state of the reactant and product to regulate catalytic rates. NMR spectroscopy was used to determine the interaction surface of TPI binding to ribosomes and to measure the corresponding kinetic rates in the absence and presence of intact ribosome particles. Chemical cross-linking and mass spectrometry revealed potential ribosomal protein binding partners of TPI. Structural results and related changes in TPI energetics and activity show that the interaction between TPI and ribosomal protein L11 mediate the RAMBO effect.


Subject(s)
Ribosomes , Triose-Phosphate Isomerase , Triose-Phosphate Isomerase/metabolism , Triose-Phosphate Isomerase/chemistry , Ribosomes/metabolism , Ribosomes/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Kinetics , Electricity , Protein Binding
16.
Biophys Chem ; 312: 107287, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981174

ABSTRACT

Although intracellular ultrastructures have typically been studied using microscopic techniques, it is difficult to observe ultrastructures at the submicron scale of living cells due to spatial resolution (fluorescence microscopy) or high vacuum environment (electron microscopy). We investigate the nanometer scale intracellular ultrastructures of living CHO cells in various osmolality using small-angle X-ray scattering (SAXS), and especially the structures of ribosomes, DNA double helix, and plasma membranes in-cell environment are observed. Ribosomes expand and contract in response to osmotic pressure, and the inter-ribosomal correlation occurs under isotonic and hyperosmolality. The DNA double helix is not dependent on the osmotic pressure. Under high osmotic pressure, the plasma membrane folds into form a multilamellar structure with a periodic length of about 6 nm. We also study the ultrastructural changes caused by formaldehyde fixation, freezing and heating.


Subject(s)
Cell Membrane , Cricetulus , Osmotic Pressure , Scattering, Small Angle , X-Ray Diffraction , Animals , CHO Cells , Cricetinae , Cell Membrane/chemistry , DNA/chemistry , Ribosomes/chemistry , Ribosomes/metabolism , Formaldehyde/chemistry , Freezing
17.
Elife ; 122024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008347

ABSTRACT

Previously, Tuller et al. found that the first 30-50 codons of the genes of yeast and other eukaryotes are slightly enriched for rare codons. They argued that this slowed translation, and was adaptive because it queued ribosomes to prevent collisions. Today, the translational speeds of different codons are known, and indeed rare codons are translated slowly. We re-examined this 5' slow translation 'ramp.' We confirm that 5' regions are slightly enriched for rare codons; in addition, they are depleted for downstream Start codons (which are fast), with both effects contributing to slow 5' translation. However, we also find that the 5' (and 3') ends of yeast genes are poorly conserved in evolution, suggesting that they are unstable and turnover relatively rapidly. When a new 5' end forms de novo, it is likely to include codons that would otherwise be rare. Because evolution has had a relatively short time to select against these codons, 5' ends are typically slightly enriched for rare, slow codons. Opposite to the expectation of Tuller et al., we show by direct experiment that genes with slowly translated codons at the 5' end are expressed relatively poorly, and that substituting faster synonymous codons improves expression. Direct experiment shows that slow codons do not prevent downstream ribosome collisions. Further informatic studies suggest that for natural genes, slow 5' ends are correlated with poor gene expression, opposite to the expectation of Tuller et al. Thus, we conclude that slow 5' translation is a 'spandrel'--a non-adaptive consequence of something else, in this case, the turnover of 5' ends in evolution, and it does not improve translation.


Subject(s)
Codon , Evolution, Molecular , Protein Biosynthesis , Saccharomyces cerevisiae , Protein Biosynthesis/genetics , Saccharomyces cerevisiae/genetics , Codon/genetics , Codon Usage , Ribosomes/metabolism , Ribosomes/genetics , 5' Untranslated Regions/genetics
18.
Elife ; 122024 Jul 16.
Article in English | MEDLINE | ID: mdl-39010741

ABSTRACT

Multicellular organisms are composed of specialized cell types with distinct proteomes. While recent advances in single-cell transcriptome analyses have revealed differential expression of mRNAs, cellular diversity in translational profiles remains underinvestigated. By performing RNA-seq and Ribo-seq in genetically defined cells in the Drosophila brain, we here revealed substantial post-transcriptional regulations that augment the cell-type distinctions at the level of protein expression. Specifically, we found that translational efficiency of proteins fundamental to neuronal functions, such as ion channels and neurotransmitter receptors, was maintained low in glia, leading to their preferential translation in neurons. Notably, distribution of ribosome footprints on these mRNAs exhibited a remarkable bias toward the 5' leaders in glia. Using transgenic reporter strains, we provide evidence that the small upstream open-reading frames in the 5' leader confer selective translational suppression in glia. Overall, these findings underscore the profound impact of translational regulation in shaping the proteomics for cell-type distinction and provide new insights into the molecular mechanisms driving cell-type diversity.


Subject(s)
Neuroglia , Protein Biosynthesis , Animals , Neuroglia/metabolism , Neurons/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Gene Expression Regulation , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Brain/metabolism , Brain/cytology , Ribosomes/metabolism , Drosophila/genetics
19.
Int J Mol Sci ; 25(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39063227

ABSTRACT

Regulation of translation is a crucial step in gene expression. Developmental signals and environmental stimuli dynamically regulate translation via upstream small open reading frames (uORFs) and ribosome pausing. Recent studies have revealed many plant genes that are specifically regulated by uORF translation following changes in growth conditions, but ribosome-pausing events are less well understood. In this study, we performed ribosome profiling (Ribo-seq) of etiolated maize (Zea mays) seedlings exposed to light for different durations, revealing hundreds of genes specifically regulated at the translation level during the early period of light exposure. We identified over 400 ribosome-pausing events in the dark that were rapidly released after illumination. These results suggested that ribosome pausing negatively regulates translation from specific genes, a conclusion that was supported by a non-targeted proteomics analysis. Importantly, we identified a conserved nucleotide motif downstream of the pausing sites. Our results elucidate the role of ribosome pausing in the control of gene expression in plants; the identification of the cis-element at the pausing sites provides insight into the mechanisms behind translation regulation and potential targets for artificial control of plant translation.


Subject(s)
Gene Expression Regulation, Plant , Open Reading Frames , Plant Proteins , Protein Biosynthesis , Ribosomes , Seedlings , Zea mays , Zea mays/genetics , Zea mays/metabolism , Ribosomes/metabolism , Seedlings/genetics , Seedlings/metabolism , Seedlings/radiation effects , Seedlings/growth & development , Plant Proteins/genetics , Plant Proteins/metabolism , Open Reading Frames/genetics , Light , Darkness , Proteomics/methods
20.
Biomolecules ; 14(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39062596

ABSTRACT

Ribosomes are not totally globular machines. Instead, they comprise prominent structural protrusions and a myriad of tentacle-like projections, which are frequently made up of ribosomal RNA expansion segments and N- or C-terminal extensions of ribosomal proteins. This is more evident in higher eukaryotic ribosomes. One of the most characteristic protrusions, present in small ribosomal subunits in all three domains of life, is the so-called beak, which is relevant for the function and regulation of the ribosome's activities. During evolution, the beak has transitioned from an all ribosomal RNA structure (helix h33 in 16S rRNA) in bacteria, to an arrangement formed by three ribosomal proteins, eS10, eS12 and eS31, and a smaller h33 ribosomal RNA in eukaryotes. In this review, we describe the different structural and functional properties of the eukaryotic beak. We discuss the state-of-the-art concerning its composition and functional significance, including other processes apparently not related to translation, and the dynamics of its assembly in yeast and human cells. Moreover, we outline the current view about the relevance of the beak's components in human diseases, especially in ribosomopathies and cancer.


Subject(s)
Ribosomes , Humans , Ribosomes/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Eukaryota/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Animals
SELECTION OF CITATIONS
SEARCH DETAIL