Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 817
Filter
1.
Nature ; 610(7930): 205-211, 2022 10.
Article in English | MEDLINE | ID: mdl-36171285

ABSTRACT

Translation is the fundamental process of protein synthesis and is catalysed by the ribosome in all living cells1. Here we use advances in cryo-electron tomography and sub-tomogram analysis2,3 to visualize the structural dynamics of translation inside the bacterium Mycoplasma pneumoniae. To interpret the functional states in detail, we first obtain a high-resolution in-cell average map of all translating ribosomes and build an atomic model for the M. pneumoniae ribosome that reveals distinct extensions of ribosomal proteins. Classification then resolves 13 ribosome states that differ in their conformation and composition. These recapitulate major states that were previously resolved in vitro, and reflect intermediates during active translation. On the basis of these states, we animate translation elongation inside native cells and show how antibiotics reshape the cellular translation landscapes. During translation elongation, ribosomes often assemble in defined three-dimensional arrangements to form polysomes4. By mapping the intracellular organization of translating ribosomes, we show that their association into polysomes involves a local coordination mechanism that is mediated by the ribosomal protein L9. We propose that an extended conformation of L9 within polysomes mitigates collisions to facilitate translation fidelity. Our work thus demonstrates the feasibility of visualizing molecular processes at atomic detail inside cells.


Subject(s)
Cryoelectron Microscopy , Mycoplasma pneumoniae , Protein Biosynthesis , Ribosomal Proteins , Ribosomes , Anti-Bacterial Agents/pharmacology , Mycoplasma pneumoniae/cytology , Mycoplasma pneumoniae/drug effects , Mycoplasma pneumoniae/metabolism , Mycoplasma pneumoniae/ultrastructure , Peptide Chain Elongation, Translational/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Polyribosomes/ultrastructure , Protein Biosynthesis/drug effects , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/drug effects , Ribosomes/metabolism , Ribosomes/ultrastructure
2.
Biochem Biophys Res Commun ; 588: 125-132, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34953209

ABSTRACT

Stress granules (SGs) are cytoplasmic biomolecular condensates that are formed against a variety of stress conditions when translation initiation is perturbed. SGs form through the weak protein-protein, protein-RNA, and RNA-RNA interactions, as well as through the intrinsically disordered domains and post-translation modifications within RNA binding proteins (RBPs). SGs are known to contribute to cell survivability by minimizing the stress-induced damage to the cells by delaying the activation of apoptosis. Here, we find that dihydrocapsaicin (DHC), an analogue of capsaicin, is a SG inducer that promotes polysome disassembly and reduces global protein translation via phosphorylation of eIF2α. DHC-mediated SG assembly is controlled by the phosphorylation of eIF2α at serine 51 position and is controlled by all four eIF2α stress kinases (i.e., HRI, PKR, PERK, and GCN2) with HRI showing maximal effect. We demonstrate that DHC is a bonafide compound that induces SG assembly, disassembles polysome, phosphorylates eIF2α in an HRI dependent manner, and thereby arrest global translation. Together, our results suggest that DHC is a novel SG inducer and an alternate to sodium arsenite to study SG dynamics.


Subject(s)
Capsaicin/analogs & derivatives , Eukaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Stress Granules/metabolism , eIF-2 Kinase/metabolism , Animals , Capsaicin/pharmacology , Cell Line , Enzyme Activation/drug effects , Humans , Mice , Oxidative Stress/drug effects , Phosphorylation/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Stress Granules/drug effects
3.
Nat Commun ; 12(1): 4920, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389715

ABSTRACT

Malignant mesothelioma (MpM) is an aggressive, invariably fatal tumour that is causally linked with asbestos exposure. The disease primarily results from loss of tumour suppressor gene function and there are no 'druggable' driver oncogenes associated with MpM. To identify opportunities for management of this disease we have carried out polysome profiling to define the MpM translatome. We show that in MpM there is a selective increase in the translation of mRNAs encoding proteins required for ribosome assembly and mitochondrial biogenesis. This results in an enhanced rate of mRNA translation, abnormal mitochondrial morphology and oxygen consumption, and a reprogramming of metabolic outputs. These alterations delimit the cellular capacity for protein biosynthesis, accelerate growth and drive disease progression. Importantly, we show that inhibition of mRNA translation, particularly through combined pharmacological targeting of mTORC1 and 2, reverses these changes and inhibits malignant cell growth in vitro and in ex-vivo tumour tissue from patients with end-stage disease. Critically, we show that these pharmacological interventions prolong survival in animal models of asbestos-induced mesothelioma, providing the basis for a targeted, viable therapeutic option for patients with this incurable disease.


Subject(s)
Mesothelioma, Malignant/genetics , Oncogenes/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Animals , Asbestos , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/metabolism , Mesothelioma, Malignant/chemically induced , Mesothelioma, Malignant/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Naphthyridines/pharmacology , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Tumor Cells, Cultured
4.
Nat Commun ; 12(1): 1352, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649340

ABSTRACT

Local translation allows for a spatial control of gene expression. Here, we use high-throughput smFISH to screen centrosomal protein-coding genes, and we describe 8 human mRNAs accumulating at centrosomes. These mRNAs localize at different stages during cell cycle with a remarkable choreography, indicating a finely regulated translational program at centrosomes. Interestingly, drug treatments and reporter analyses reveal a common translation-dependent localization mechanism requiring the nascent protein. Using ASPM and NUMA1 as models, single mRNA and polysome imaging reveals active movements of endogenous polysomes towards the centrosome at the onset of mitosis, when these mRNAs start localizing. ASPM polysomes associate with microtubules and localize by either motor-driven transport or microtubule pulling. Remarkably, the Drosophila orthologs of the human centrosomal mRNAs also localize to centrosomes and also require translation. These data identify a conserved family of centrosomal mRNAs that localize by active polysome transport mediated by nascent proteins.


Subject(s)
Centrosome/metabolism , Polyribosomes/metabolism , RNA Transport , Animals , Cell Cycle Proteins/metabolism , Centrosome/drug effects , Cycloheximide/pharmacology , Drosophila/genetics , HeLa Cells , Humans , Mitosis/drug effects , Open Reading Frames/genetics , Polyribosomes/drug effects , Puromycin/pharmacology , RNA Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
5.
Cell Rep ; 30(13): 4355-4369.e6, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32234473

ABSTRACT

Activation of p53 by the small molecule Nutlin can result in a combination of cell cycle arrest and apoptosis. The relative strength of these events is difficult to predict by classical gene expression analysis, leaving uncertainty as to the therapeutic benefits. In this study, we report a translational control mechanism shaping p53-dependent apoptosis. Using polysome profiling, we establish Nutlin-induced apoptosis to associate with the enhanced translation of mRNAs carrying multiple copies of an identified 3' UTR CG-rich motif mediating p53-dependent death (CGPD-motif). We identify PCBP2 and DHX30 as CGPD-motif interactors. We find that in cells undergoing persistent cell cycle arrest in response to Nutlin, CGPD-motif mRNAs are repressed by the PCBP2-dependent binding of DHX30 to the motif. Upon DHX30 depletion in these cells, the translation of CGPD-motif mRNAs increases, and the response to Nutlin shifts toward apoptosis. Instead, DHX30 inducible overexpression in SJSA1 cells leads to decreased translation of CGPD-motif mRNAs.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Piperazines/pharmacology , Protein Biosynthesis/drug effects , RNA Helicases/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Cell Cycle Checkpoints/drug effects , Cell Line , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Humans , Neoplasm Proteins/metabolism , Nucleotide Motifs/genetics , Phenotype , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Cells ; 9(1)2020 01 10.
Article in English | MEDLINE | ID: mdl-31936702

ABSTRACT

Eukaryotic initiation factor 6 (eIF6) is necessary for the nucleolar biogenesis of 60S ribosomes. However, most of eIF6 resides in the cytoplasm, where it acts as an initiation factor. eIF6 is necessary for maximal protein synthesis downstream of growth factor stimulation. eIF6 is an antiassociation factor that binds 60S subunits, in turn preventing premature 40S joining and thus the formation of inactive 80S subunits. It is widely thought that eIF6 antiassociation activity is critical for its function. Here, we exploited and improved our assay for eIF6 binding to ribosomes (iRIA) in order to screen for modulators of eIF6 binding to the 60S. Three compounds, eIFsixty-1 (clofazimine), eIFsixty-4, and eIFsixty-6 were identified and characterized. All three inhibit the binding of eIF6 to the 60S in the micromolar range. eIFsixty-4 robustly inhibits cell growth, whereas eIFsixty-1 and eIFsixty-6 might have dose- and cell-specific effects. Puromycin labeling shows that eIF6ixty-4 is a strong global translational inhibitor, whereas the other two are mild modulators. Polysome profiling and RT-qPCR show that all three inhibitors reduce the specific translation of well-known eIF6 targets. In contrast, none of them affect the nucleolar localization of eIF6. These data provide proof of principle that the generation of eIF6 translational modulators is feasible.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Biosynthesis , Ribosome Subunits, Large, Eukaryotic/metabolism , Cell Line , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Cell Survival , Enzyme-Linked Immunosorbent Assay , Humans , Peptide Chain Initiation, Translational/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
7.
Nucleic Acids Res ; 48(1): 116-129, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31701124

ABSTRACT

Upon detection of viral infections, cells activate the expression of type I interferons (IFNs) and pro-inflammatory cytokines to control viral dissemination. As part of their antiviral response, cells also trigger the translational shutoff response which prevents translation of viral mRNAs and cellular mRNAs in a non-selective manner. Intriguingly, mRNAs encoding for antiviral factors bypass this translational shutoff, suggesting the presence of additional regulatory mechanisms enabling expression of the self-defence genes. Here, we identified the dsRNA binding protein ILF3 as an essential host factor required for efficient translation of the central antiviral cytokine, IFNB1, and a subset of interferon-stimulated genes. By combining polysome profiling and next-generation sequencing, ILF3 was also found to be necessary to establish the dsRNA-induced transcriptional and translational programs. We propose a central role for the host factor ILF3 in enhancing expression of the antiviral defence mRNAs in cellular conditions where cap-dependent translation is compromised.


Subject(s)
Interferon-beta/genetics , Nuclear Factor 90 Proteins/genetics , Protein Biosynthesis , RNA, Double-Stranded/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , A549 Cells , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Cytokines/genetics , Cytokines/immunology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Gene Expression Regulation , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon-beta/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Nuclear Factor 90 Proteins/immunology , Poly I-C/pharmacology , Polyribosomes/drug effects , Polyribosomes/genetics , Polyribosomes/immunology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Double-Stranded/metabolism , RNA, Messenger/immunology , RNA, Viral/antagonists & inhibitors , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic , Signal Transduction , Ubiquitins/genetics , Ubiquitins/immunology , Virus Replication
8.
Cell Chem Biol ; 26(2): 179-190.e12, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30503283

ABSTRACT

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is an expanded G4C2 repeat [(G4C2)exp] in C9ORF72. ALS/FTD-associated toxicity has been traced to the RNA transcribed from the repeat expansion [r(G4C2)exp], which sequesters RNA-binding proteins (RBPs) and undergoes repeat-associated non-ATG (RAN) translation to generate toxic dipeptide repeats. Using in vitro and cell-based assays, we identified a small molecule (4) that selectively bound r(G4C2)exp, prevented sequestration of an RBP, and inhibited RAN translation. Indeed, biophysical characterization showed that 4 selectively bound the hairpin form of r(G4C2)exp, and nuclear magnetic resonance spectroscopy studies and molecular dynamics simulations defined this molecular recognition event. Cellular imaging revealed that 4 localized to r(G4C2)exp cytoplasmic foci, the putative sites of RAN translation. Collectively, these studies highlight that the hairpin structure of r(G4C2)exp is a therapeutically relevant target and small molecules that bind it can ameliorate c9ALS/FTD-associated toxicity.


Subject(s)
C9orf72 Protein/genetics , DNA Repeat Expansion/genetics , Small Molecule Libraries/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Binding Sites , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Humans , Kinetics , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Thermodynamics
9.
New Phytol ; 217(3): 1077-1085, 2018 02.
Article in English | MEDLINE | ID: mdl-29139127

ABSTRACT

The importance of translational regulation during Arabidopsis seed germination has been shown previously. Here the role of transcriptional and translational regulation during seed imbibition of the very dormant DELAY OF GERMINATION 1 (DOG1) near-isogenic line was investigated. Polysome profiling was performed on dormant and after-ripened seeds imbibed for 6 and 24 h in water and in the transcription inhibitor cordycepin. Transcriptome and translatome changes were investigated. Ribosomal profiles of after-ripened seeds imbibed in cordycepin mimic those of dormant seeds. The polysome occupancy of mRNA species is not affected by germination inhibition, either as a result of seed dormancy or as a result of cordycepin treatment, indicating the importance of the regulation of transcript abundance. The expression of auxin metabolism genes is discriminative during the imbibition of after-ripened and dormant seeds, which is confirmed by altered concentrations of indole-3-acetic acid conjugates and precursors.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Biosynthetic Pathways , Indoleacetic Acids/metabolism , Plant Dormancy , Protein Biosynthesis , Transcriptome/genetics , Tryptophan/metabolism , Arabidopsis/drug effects , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Biosynthetic Pathways/drug effects , Deoxyadenosines/pharmacology , Gene Expression Regulation, Plant/drug effects , Germination/drug effects , Plant Dormancy/drug effects , Plant Dormancy/genetics , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seeds/drug effects , Seeds/physiology , Transcription, Genetic/drug effects , Transcriptome/drug effects
10.
Invest New Drugs ; 36(2): 217-229, 2018 04.
Article in English | MEDLINE | ID: mdl-29116477

ABSTRACT

Deregulation of cap-dependent translation has been implicated in the malignant transformation of numerous human tissues. 4EGI-1, a novel small-molecule inhibitor of cap-dependent translation, disrupts formation of the eukaryotic initiation factor 4F (eIF4F) complex. The effects of 4EGI-1-mediated inhibition of translation initiation in malignant pleural mesothelioma (MPM) were examined. 4EGI-1 preferentially inhibited cell viability and induced apoptosis in MPM cells compared to normal mesothelial (LP9) cells. This effect was associated with hypophosphorylation of 4E-binding protein 1 (4E-BP1) and decreased protein levels of the cancer-related genes, c-myc and osteopontin. 4EGI-1 showed enhanced cytotoxicity in combination with pemetrexed or gemcitabine. Translatome-wide polysome microarray analysis revealed a large cohort of genes that were translationally regulated upon treatment with 4EGI-1. The 4EGI-1-regulated translatome was negatively correlated to a previously published translatome regulated by eIF4E overexpression in human mammary epithelial cells, which is in agreement with the notion that 4EGI-1 inhibits the eIF4F complex. These data indicate that inhibition of the eIF4F complex by 4EGI-1 or similar translation inhibitors could be a strategy for treating mesothelioma. Genome wide translational profiling identified a large cohort of promising target genes that should be further evaluated for their potential significance in the treatment of MPM.


Subject(s)
Genome, Human , Hydrazones/pharmacology , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Protein Biosynthesis/drug effects , RNA Caps/metabolism , Thiazoles/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Down-Regulation/drug effects , Eukaryotic Initiation Factor-4E/deficiency , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Humans , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Pemetrexed/pharmacology , Pemetrexed/therapeutic use , Phosphoproteins/metabolism , Phosphorylation/drug effects , Pleural Neoplasms/pathology , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding , Proteome/metabolism , Reproducibility of Results , Gemcitabine
11.
J Biol Chem ; 292(45): 18469-18485, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28939771

ABSTRACT

The ability to detect and respond to oxidative stress is crucial to the survival of living organisms. In cells, sensing of increased levels of reactive oxygen species (ROS) activates many defensive mechanisms that limit or repair damage to cell components. The ROS-signaling responses necessary for cell survival under oxidative stress conditions remain incompletely understood, especially for the translational machinery. Here, we found that drug treatments or a genetic deficiency in the thioredoxin system that increase levels of endogenous hydrogen peroxide in the yeast Saccharomyces cerevisiae promote site-specific endonucleolytic cleavage in 25S ribosomal RNA (rRNA) adjacent to the c loop of the expansion segment 7 (ES7), a putative regulatory region located on the surface of the 60S ribosomal subunit. Our data also show that ES7c is cleaved at early stages of the gene expression program that enables cells to successfully counteract oxidative stress and is not a prerequisite or consequence of apoptosis. Moreover, the 60S subunits containing ES7c-cleaved rRNA cofractionate with intact subunits in sucrose gradients and repopulate polysomes after a short starvation-induced translational block, indicating their active role in translation. These results demonstrate that ES7c cleavage in rRNA is an early and sensitive marker of increased ROS levels in yeast cells and suggest that changes in ribosomes may be involved in the adaptive response to oxidative stress.


Subject(s)
Gene Expression Regulation, Fungal , Oxidative Stress , Polyribosomes/enzymology , RNA, Fungal/metabolism , RNA, Ribosomal/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/enzymology , Apoptosis/drug effects , Biomarkers/metabolism , Gene Deletion , Gene Expression Regulation, Fungal/drug effects , Hormesis , Kinetics , Nucleic Acid Conformation , Oxidants/pharmacology , Oxidative Stress/drug effects , Peroxidases/genetics , Peroxidases/metabolism , Polyribosomes/drug effects , Polyribosomes/metabolism , RNA Cleavage/drug effects , RNA Stability/drug effects , RNA, Fungal/chemistry , RNA, Ribosomal/chemistry , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reducing Agents/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spheroplasts/drug effects , Spheroplasts/enzymology , Spheroplasts/growth & development , Spheroplasts/physiology , Unfolded Protein Response/drug effects
12.
J Neurosci ; 37(7): 1862-1872, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087764

ABSTRACT

Translation in dendrites is believed to support synaptic changes during memory consolidation. Although translational control mechanisms are fundamental mediators of memory, little is known about their role in local translation. We previously found that polyribosomes accumulate in dendritic spines of the adult rat lateral amygdala (LA) during consolidation of aversive pavlovian conditioning and that this memory requires cap-dependent initiation, a primary point of translational control in eukaryotic cells. Here we used serial electron microscopy reconstructions to quantify polyribosomes in LA dendrites when consolidation was blocked by the cap-dependent initiation inhibitor 4EGI-1. We found that 4EGI-1 depleted polyribosomes in dendritic shafts and selectively prevented their upregulation in spine heads, but not bases and necks, during consolidation. Cap-independent upregulation was specific to spines with small, astrocyte-associated synapses. Our results reveal that cap-dependent initiation is involved in local translation during learning and that local translational control varies with synapse type.SIGNIFICANCE STATEMENT Translation initiation is a central regulator of long-term memory formation. Local translation in dendrites supports memory by providing necessary proteins at synaptic sites, but it is unknown whether this requires initiation or bypasses it. We used serial electron microscopy reconstructions to examine polyribosomes in dendrites when memory formation was blocked by an inhibitor of translation initiation. This revealed two major pools of polyribosomes that were upregulated during memory formation: one pool in dendritic spine heads that was initiation dependent and another pool in the bases and necks of small spines that was initiation independent. Thus, translation regulation differs between spine types and locations, and translation that occurs closest to individual synapses during memory formation is initiation dependent.


Subject(s)
Basolateral Nuclear Complex/cytology , Dendritic Spines/metabolism , Gene Expression Regulation/physiology , Memory Consolidation/physiology , Neurons/ultrastructure , Protein Biosynthesis/physiology , Analysis of Variance , Animals , Association Learning/drug effects , Association Learning/physiology , Basolateral Nuclear Complex/diagnostic imaging , Basolateral Nuclear Complex/drug effects , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Gene Expression Regulation/drug effects , Hydrazones/pharmacology , Image Processing, Computer-Assisted , Male , Memory Consolidation/drug effects , Microscopy, Electron, Transmission , Models, Animal , Neuroimaging , Neurons/drug effects , Polyribosomes/drug effects , Polyribosomes/ultrastructure , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Thiazoles/pharmacology
13.
J Cell Physiol ; 232(3): 576-584, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27292014

ABSTRACT

Expression of the mu-opioid receptor (MOR) protein is controlled by extensive transcriptional and post-transcriptional processing. MOR gene expression has previously been shown to be altered by a post-transcriptional mechanism involving the MOR mRNA untranslated region (UTR). Here, we demonstrate for the first time the role of heterogeneous nuclear ribonucleic acids (hnRNA)-binding protein (hnRNP) K and poly(C)-binding protein 1 (PCBP1) as post-transcriptional inducers in MOR gene regulation. In the absence of morphine, a significant level of MOR mRNA is sustained in its resting state and partitions in the translationally inactive polysomal fraction. Morphine stimulation activates the downstream targets hnRNP K and PCPB1 and induces partitioning of the MOR mRNA to the translationally active fraction. Using reporter and ligand binding assays, as well as RNA EMSA, we reveal potential RNP binding sites located in the 5'-untranslated region of human MOR mRNA. In addition, we also found that morphine-induced RNPs could regulate MOR expression. Our results establish the role of hnRNP K and PCPB1 in the translational control of morphine-induced MOR expression in human neuroblastoma (NMB) cells as well as cells stably expressing MOR (NMB1). J. Cell. Physiol. 232: 576-584, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation/drug effects , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Transcription, Genetic/drug effects , 5' Untranslated Regions/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Immunoprecipitation , Mice , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Receptors, Opioid, mu/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
14.
Nat Commun ; 7: 12125, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381497

ABSTRACT

Stress granules (SGs) harbour translationally stalled messenger ribonucleoproteins and play important roles in regulating gene expression and cell fate. Here we show that neddylation promotes SG assembly in response to arsenite-induced oxidative stress. Inhibition or depletion of key components of the neddylation machinery concomitantly inhibits stress-induced polysome disassembly and SG assembly. Affinity purification and subsequent mass-spectrometric analysis of Nedd8-conjugated proteins from translationally stalled ribosomal fractions identified ribosomal proteins, translation factors and RNA-binding proteins (RBPs), including SRSF3, a previously known SG regulator. We show that SRSF3 is selectively neddylated at Lys85 in response to arsenite. A non-neddylatable SRSF3 (K85R) mutant do not prevent arsenite-induced polysome disassembly, but fails to support the SG assembly, suggesting that the neddylation pathway plays an important role in SG assembly.


Subject(s)
Arsenites/toxicity , Cytoplasmic Granules/drug effects , NEDD8 Protein/genetics , Protein Biosynthesis/drug effects , Serine-Arginine Splicing Factors/genetics , Cell Line, Tumor , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysine/metabolism , NEDD8 Protein/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Oxidative Stress , Polyribosomes/drug effects , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
15.
Nucleic Acids Res ; 44(16): 7777-91, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27422873

ABSTRACT

The archaea-/eukaryote-specific 40S-ribosomal-subunit protein S31 is expressed as an ubiquitin fusion protein in eukaryotes and consists of a conserved body and a eukaryote-specific N-terminal extension. In yeast, S31 is a practically essential protein, which is required for cytoplasmic 20S pre-rRNA maturation. Here, we have studied the role of the N-terminal extension of the yeast S31 protein. We show that deletion of this extension partially impairs cell growth and 40S subunit biogenesis and confers hypersensitivity to aminoglycoside antibiotics. Moreover, the extension harbours a nuclear localization signal that promotes active nuclear import of S31, which associates with pre-ribosomal particles in the nucleus. In the absence of the extension, truncated S31 inefficiently assembles into pre-40S particles and two subpopulations of mature small subunits, one lacking and another one containing truncated S31, can be identified. Plasmid-driven overexpression of truncated S31 partially suppresses the growth and ribosome biogenesis defects but, conversely, slightly enhances the hypersensitivity to aminoglycosides. Altogether, these results indicate that the N-terminal extension facilitates the assembly of S31 into pre-40S particles and contributes to the optimal translational activity of mature 40S subunits but has only a minor role in cytoplasmic cleavage of 20S pre-rRNA at site D.


Subject(s)
Ribosomal Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Alleles , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Mutation/genetics , Nuclear Localization Signals/metabolism , Phenotype , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , RNA Processing, Post-Transcriptional/drug effects , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development
16.
BMB Rep ; 49(8): 449-54, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27345716

ABSTRACT

Stress Granules (SGs) are microscopically visible, phase dense aggregates of translationally stalled messenger ribonucleoprotein (mRNP) complexes formed in response to distinct stress conditions. It is generally considered that SG formation is induced to protect cells from conditions of stress. The precise constituents of SGs and the mechanism through which SGs are dynamically regulated in response to stress are not completely understood. Hence, it is important to identify proteins which regulate SG assembly and disassembly. In the present study, we report Neuregulin-2 (NRG2) as a novel component of SGs; furthermore, depletion of NRG2 potently inhibits SG formation. We also demonstrate that NRG2 specifically localizes to SGs under various stress conditions. Knockdown of NRG2 has no effect on stress-induced polysome disassembly, suggesting that the component does not influence early step of SG formation. It was also observed that reduced expression of NRG2 led to marginal increase in cell survival under arsenite-induced stress. [BMB Reports 2016; 49(8): 449-454].


Subject(s)
Cytoplasmic Granules/metabolism , Nerve Growth Factors/metabolism , Stress, Physiological , Arsenites/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cytoplasmic Granules/drug effects , Gene Deletion , Gene Knockdown Techniques , HEK293 Cells , Humans , Oxidative Stress/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , RNA, Small Interfering/metabolism , Stress, Physiological/drug effects
17.
J Biol Chem ; 291(1): 291-302, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26527679

ABSTRACT

Drug resistance of cancer cells to various therapeutic agents and molecular targets is a major problem facing current cancer research. The tumor suppressor gene Scribble encodes a polarity protein that is conserved between Drosophila and mammals; loss of the locus disrupts cell polarity, inhibits apoptosis, and mediates cancer process. However, the role of Scribble in drug resistance remains unknown. We show here that knockdown of Scribble enhances drug resistance by permitting accumulation of Snail, which functions as a transcription factor during the epithelial-mesenchymal transition. Then, loss of Scribble activates the mRNA-binding protein human antigen R (HuR) by facilitating translocation of HuR from the nucleus to the cytoplasm. Furthermore, we demonstrate HuR can recognize AU-rich elements of the Snail-encoding mRNA, thereby regulating Snail translation. Moreover, loss of Scribble-induced HuR translocation mediates the accumulation of Snail via activation of the p38 MAPK pathway. Thus, this work clarifies the role of polarity protein Scribble, which is directly implicated in the regulation of developmental transcription factor Snail, and suggesting a mechanism for Scribble mediating cancer drug resistance.


Subject(s)
Drug Resistance, Neoplasm , ELAV-Like Protein 1/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Protein Biosynthesis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mice, Nude , Models, Biological , Neoplasms/genetics , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
PLoS One ; 10(7): e0132090, 2015.
Article in English | MEDLINE | ID: mdl-26186436

ABSTRACT

Interest in mRNA methylation has exploded in recent years. The sudden interest in a 40 year old discovery was due in part to the finding of FTO's (Fat Mass Obesity) N6-methyl-adenosine (m6A) deaminase activity, thus suggesting a link between obesity-associated diseases and the presence of m6A in mRNA. Another catalyst of the sudden rise in mRNA methylation research was the release of mRNA methylomes for human, mouse and Saccharomyces cerevisiae. However, the molecular function, or functions of this mRNA 'epimark' remain to be discovered. There is supportive evidence that m6A could be a mark for mRNA degradation due to its binding to YTH domain proteins, and consequently being chaperoned to P bodies. Nonetheless, only a subpopulation of the methylome was found binding to YTHDF2 in HeLa cells.The model organism Saccharomyces cerevisiae, has only one YTH domain protein (Pho92, Mrb1), which targets PHO4 transcripts for degradation under phosphate starvation. However, mRNA methylation is only found under meiosis inducing conditions, and PHO4 transcripts are apparently non-methylated. In this paper we set out to investigate if m6A could function alternatively to being a degradation mark in S. cerevisiae; we also sought to test whether it can be induced under non-standard sporulation conditions. We find a positive association between the presence of m6A and message translatability. We also find m6A induction following prolonged rapamycin treatment.


Subject(s)
Meiosis/drug effects , Protein Biosynthesis/drug effects , Ribosomes/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Sirolimus/pharmacology , Cluster Analysis , Gene Knockout Techniques , HeLa Cells , Humans , Methylation/drug effects , Phenotype , Polyribosomes/drug effects , Polyribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/drug effects , Saccharomyces cerevisiae/drug effects , Spores, Fungal/drug effects , Spores, Fungal/physiology
19.
J Am Chem Soc ; 137(16): 5336-45, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25825793

ABSTRACT

One major class of disease-causing RNAs is expanded repeating transcripts. These RNAs cause diseases via multiple mechanisms, including: (i) gain-of-function, in which repeating RNAs bind and sequester proteins involved in RNA biogenesis and (ii) repeat associated non-ATG (RAN) translation, in which repeating transcripts are translated into toxic proteins without use of a canonical, AUG, start codon. Herein, we develop and study chemical probes that bind and react with an expanded r(CGG) repeat (r(CGG)(exp)) present in a 5' untranslated region that causes fragile X-associated tremor/ataxia syndrome (FXTAS). Reactive compounds bind to r(CGG)(exp) in cellulo as shown with Chem-CLIP-Map, an approach to map small molecule binding sites within RNAs in cells. Compounds also potently improve FXTAS-associated pre-mRNA splicing and RAN translational defects, while not affecting translation of the downstream open reading frame. In contrast, oligonucleotides affect both RAN and canonical translation when they bind to r(CGG)(exp), which is mechanistically traced to a decrease in polysome loading. Thus, designer small molecules that react with RNA targets can be used to profile the RNAs to which they bind in cells, including identification of binding sites, and can modulate several aspects of RNA-mediated disease pathology in a manner that may be more beneficial than oligonucleotides.


Subject(s)
Biotin/analogs & derivatives , Biotin/pharmacology , Protein Biosynthesis/drug effects , RNA/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Ataxia/genetics , Ataxia/metabolism , COS Cells , Chlorocebus aethiops , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Humans , Polyribosomes/drug effects , Polyribosomes/genetics , Polyribosomes/metabolism , RNA/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/drug effects , Tremor/genetics , Tremor/metabolism , Trinucleotide Repeat Expansion/drug effects
20.
Aging Cell ; 14(4): 547-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25807975

ABSTRACT

Calorie restriction (CR) and rapamycin (RP) extend lifespan and improve health across model organisms. Both treatments inhibit mammalian target of rapamycin (mTOR) signaling, a conserved longevity pathway and a key regulator of protein homeostasis, yet their effects on proteome homeostasis are relatively unknown. To comprehensively study the effects of aging, CR, and RP on protein homeostasis, we performed the first simultaneous measurement of mRNA translation, protein turnover, and abundance in livers of young (3 month) and old (25 month) mice subjected to 10-week RP or 40% CR. Protein abundance and turnover were measured in vivo using (2) H3 -leucine heavy isotope labeling followed by LC-MS/MS, and translation was assessed by polysome profiling. We observed 35-60% increased protein half-lives after CR and 15% increased half-lives after RP compared to age-matched controls. Surprisingly, the effects of RP and CR on protein turnover and abundance differed greatly between canonical pathways, with opposite effects in mitochondrial (mt) dysfunction and eIF2 signaling pathways. CR most closely recapitulated the young phenotype in the top pathways. Polysome profiles indicated that CR reduced polysome loading while RP increased polysome loading in young and old mice, suggesting distinct mechanisms of reduced protein synthesis. CR and RP both attenuated protein oxidative damage. Our findings collectively suggest that CR and RP extend lifespan in part through the reduction of protein synthetic burden and damage and a concomitant increase in protein quality. However, these results challenge the notion that RP is a faithful CR mimetic and highlight mechanistic differences between the two interventions.


Subject(s)
Aging/genetics , Caloric Restriction , Liver/drug effects , Proteome/genetics , Sirolimus/pharmacology , Aging/metabolism , Animals , Deuterium , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Regulation , Half-Life , Homeostasis , Isotope Labeling , Leucine/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Protein Stability , Proteolysis , Proteome/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL