Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 416
Filter
1.
Mol Cell ; 81(12): 2566-2582.e6, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33878294

ABSTRACT

The mitochondrial translation system originates from a bacterial ancestor but has substantially diverged in the course of evolution. Here, we use single-particle cryo-electron microscopy (cryo-EM) as a screening tool to identify mitochondrial translation termination mechanisms and to describe them in molecular detail. We show how mitochondrial release factor 1a releases the nascent chain from the ribosome when it encounters the canonical stop codons UAA and UAG. Furthermore, we define how the peptidyl-tRNA hydrolase ICT1 acts as a rescue factor on mitoribosomes that have stalled on truncated messages to recover them for protein synthesis. Finally, we present structural models detailing the process of mitochondrial ribosome recycling to explain how a dedicated elongation factor, mitochondrial EFG2 (mtEFG2), has specialized for cooperation with the mitochondrial ribosome recycling factor to dissociate the mitoribosomal subunits at the end of the translation process.


Subject(s)
Mitochondria/physiology , Mitochondrial Ribosomes/metabolism , Peptide Chain Termination, Translational/physiology , Animals , Carboxylic Ester Hydrolases , Codon, Terminator , Cryoelectron Microscopy/methods , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Peptide Chain Termination, Translational/genetics , Peptide Elongation Factor G/metabolism , Peptide Termination Factors/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomal Proteins/physiology , Ribosomes/metabolism
2.
Mol Cell ; 80(3): 470-484.e8, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33053322

ABSTRACT

Cellular responses to environmental stress are frequently mediated by RNA-binding proteins (RBPs). Here, we examined global RBP dynamics in Saccharomyces cerevisiae in response to glucose starvation and heat shock. Each stress induced rapid remodeling of the RNA-protein interactome without corresponding changes in RBP abundance. Consistent with general translation shutdown, ribosomal proteins contacting the mRNA showed decreased RNA association. Among translation components, RNA association was most reduced for initiation factors involved in 40S scanning (eukaryotic initiation factor 4A [eIF4A], eIF4B, and Ded1), indicating a common mechanism of translational repression. In unstressed cells, eIF4A, eIF4B, and Ded1 primarily targeted the 5' ends of mRNAs. Following glucose withdrawal, 5' binding was abolished within 30 s, explaining the rapid translation shutdown, but mRNAs remained stable. Heat shock induced progressive loss of 5' RNA binding by initiation factors over ∼16 min and provoked mRNA degradation, particularly for translation-related factors, mediated by Xrn1. Taken together, these results reveal mechanisms underlying translational control of gene expression during stress.


Subject(s)
Peptide Initiation Factors/metabolism , Protein Biosynthesis/physiology , Stress, Physiological/physiology , 5' Untranslated Regions , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factors/metabolism , Glucose/metabolism , Heat-Shock Response/physiology , Peptide Initiation Factors/physiology , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
3.
Cell ; 133(2): 292-302, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423200

ABSTRACT

In nearly every organism studied, reduced caloric intake extends life span. In yeast, span extension from dietary restriction is thought to be mediated by the highly conserved, nutrient-responsive target of rapamycin (TOR), protein kinase A (PKA), and Sch9 kinases. These kinases coordinately regulate various cellular processes including stress responses, protein turnover, cell growth, and ribosome biogenesis. Here we show that a specific reduction of 60S ribosomal subunit levels slows aging in yeast. Deletion of genes encoding 60S subunit proteins or processing factors or treatment with a small molecule, which all inhibit 60S subunit biogenesis, are each sufficient to significantly increase replicative life span. One mechanism by which reduced 60S subunit levels leads to life span extension is through induction of Gcn4, a nutrient-responsive transcription factor. Genetic epistasis analyses suggest that dietary restriction, reduced 60S subunit abundance, and Gcn4 activation extend yeast life span by similar mechanisms.


Subject(s)
DNA-Binding Proteins/physiology , Ribosome Subunits, Large, Eukaryotic/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , Transcription Factors/physiology , Basic-Leucine Zipper Transcription Factors , Gene Deletion , Histone Deacetylases/physiology , Ribosomal Proteins/physiology , Silent Information Regulator Proteins, Saccharomyces cerevisiae/physiology , Sirtuin 2 , Sirtuins/physiology
4.
Nucleic Acids Res ; 49(1): 206-220, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33330942

ABSTRACT

Proteostasis needs to be tightly controlled to meet the cellular demand for correctly de novo folded proteins and to avoid protein aggregation. While a coupling between translation rate and co-translational folding, likely involving an interplay between the ribosome and its associated chaperones, clearly appears to exist, the underlying mechanisms and the contribution of ribosomal proteins remain to be explored. The ribosomal protein uL3 contains a long internal loop whose tip region is in close proximity to the ribosomal peptidyl transferase center. Intriguingly, the rpl3[W255C] allele, in which the residue making the closest contact to this catalytic site is mutated, affects diverse aspects of ribosome biogenesis and function. Here, we have uncovered, by performing a synthetic lethal screen with this allele, an unexpected link between translation and the folding of nascent proteins by the ribosome-associated Ssb-RAC chaperone system. Our results reveal that uL3 and Ssb-RAC cooperate to prevent 80S ribosomes from piling up within the 5' region of mRNAs early on during translation elongation. Together, our study provides compelling in vivo evidence for a functional connection between peptide bond formation at the peptidyl transferase center and chaperone-assisted de novo folding of nascent polypeptides at the solvent-side of the peptide exit tunnel.


Subject(s)
Molecular Chaperones/physiology , Multiprotein Complexes/physiology , Peptide Chain Elongation, Translational/physiology , Protein Folding , Proteostasis/physiology , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Alleles , Loss of Function Mutation , Molecular Chaperones/genetics , Mutation, Missense , Peptidyl Transferases/physiology , Point Mutation , Recombinant Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Ribosomes/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
5.
Nucleic Acids Res ; 49(4): 2226-2239, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33503254

ABSTRACT

Ribosome hibernation is a universal translation stress response found in bacteria as well as plant plastids. The term was coined almost two decades ago and despite recent insights including detailed cryo-EM structures, the physiological role and underlying molecular mechanism of ribosome hibernation has remained unclear. Here, we demonstrate that Escherichia coli hibernation factors RMF, HPF and RaiA (HFs) concurrently confer ribosome hibernation. In response to carbon starvation and resulting growth arrest, we observe that HFs protect ribosomes at the initial stage of starvation. Consistently, a deletion mutant lacking all three factors (ΔHF) is severely inhibited in regrowth from starvation. ΔHF cells increasingly accumulate 70S ribosomes harbouring fragmented rRNA, while rRNA in wild-type 100S dimers is intact. RNA fragmentation is observed to specifically occur at HF-associated sites in 16S rRNA of assembled 70S ribosomes. Surprisingly, degradation of the 16S rRNA 3'-end is decreased in cells lacking conserved endoribonuclease YbeY and exoribonuclease RNase R suggesting that HFs directly block these ribonucleases from accessing target sites in the ribosome.


Subject(s)
Escherichia coli Proteins/physiology , Ribonucleases/metabolism , Ribosomal Proteins/physiology , Ribosomes/metabolism , Carbon/physiology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Mutation , Protein Biosynthesis , RNA, Ribosomal, 16S/metabolism , Ribosomal Proteins/genetics , Stress, Physiological/genetics
6.
Blood ; 136(11): 1262-1273, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32702755

ABSTRACT

Diamond-Blackfan anemia (DBA) was the first ribosomopathy described and is a constitutional inherited bone marrow failure syndrome. Erythroblastopenia is the major characteristic of the disease, which is a model for ribosomal diseases, related to a heterozygous allelic variation in 1 of the 20 ribosomal protein genes of either the small or large ribosomal subunit. The salient feature of classical DBA is a defect in ribosomal RNA maturation that generates nucleolar stress, leading to stabilization of p53 and activation of its targets, resulting in cell-cycle arrest and apoptosis. Although activation of p53 may not explain all aspects of DBA erythroid tropism, involvement of GATA1/HSP70 and globin/heme imbalance, with an excess of the toxic free heme leading to reactive oxygen species production, account for defective erythropoiesis in DBA. Despite significant progress in defining the molecular basis of DBA and increased understanding of the mechanistic basis for DBA pathophysiology, progress in developing new therapeutic options has been limited. However, recent advances in gene therapy, better outcomes with stem cell transplantation, and discoveries of putative new drugs through systematic drug screening using large chemical libraries provide hope for improvement.


Subject(s)
Anemia, Diamond-Blackfan , Abnormalities, Multiple/genetics , Adenosine Deaminase/blood , Adenosine Deaminase/genetics , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/therapy , Child, Preschool , Congenital Abnormalities/genetics , Diagnosis, Differential , Disease Management , Drug Resistance , Erythrocytes/enzymology , Fetal Growth Retardation/etiology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/physiology , Genetic Heterogeneity , Genetic Therapy , Glucocorticoids/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Infant , Infant, Newborn , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Models, Biological , Mutation , Neoplastic Syndromes, Hereditary/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Tumor Suppressor Protein p53/physiology
7.
Nucleic Acids Res ; 48(3): 1068-1083, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31777928

ABSTRACT

Engineering the process of molecular translation, or protein biosynthesis, has emerged as a major opportunity in synthetic and chemical biology to generate novel biological insights and enable new applications (e.g. designer protein therapeutics). Here, we review methods for engineering the process of translation in vitro. We discuss the advantages and drawbacks of the two major strategies-purified and extract-based systems-and how they may be used to manipulate and study translation. Techniques to engineer each component of the translation machinery are covered in turn, including transfer RNAs, translation factors, and the ribosome. Finally, future directions and enabling technological advances for the field are discussed.


Subject(s)
Bioengineering , Protein Biosynthesis , Amino Acids/metabolism , RNA, Transfer/biosynthesis , RNA, Transfer/metabolism , Ribosomal Proteins/physiology , Ribosomes/chemistry , Ribosomes/metabolism
8.
Mol Microbiol ; 114(3): 391-408, 2020 09.
Article in English | MEDLINE | ID: mdl-32291821

ABSTRACT

Many bacterial small RNAs (sRNAs) efficiently inhibit translation of target mRNAs by forming a duplex that sequesters the Shine-Dalgarno (SD) sequence or start codon and prevents formation of the translation initiation complex. There are a growing number of examples of sRNA-mRNA binding interactions distant from the SD region, but how these mediate translational regulation remains unclear. Our previous work in Escherichia coli and Salmonella identified a mechanism of translational repression of manY mRNA by the sRNA SgrS through a binding interaction upstream of the manY SD. Here, we report that SgrS forms a duplex with a uridine-rich translation-enhancing element in the manY 5' untranslated region. Notably, we show that the enhancer is ribosome-dependent and that the small ribosomal subunit protein S1 interacts with the enhancer to promote translation of manY. In collaboration with the chaperone protein Hfq, SgrS interferes with the interaction between the translation enhancer and ribosomal protein S1 to repress translation of manY mRNA. Since bacterial translation is often modulated by enhancer-like elements upstream of the SD, sRNA-mediated enhancer silencing could be a common mode of gene regulation.


Subject(s)
Enhancer Elements, Genetic , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Host Factor 1 Protein/genetics , Peptide Chain Initiation, Translational , RNA, Small Untranslated/genetics , Ribosomal Proteins/physiology , 5' Untranslated Regions/genetics , Base Pairing , Binding Sites , Gene Expression Regulation, Bacterial , Protein Biosynthesis , RNA Interference , RNA, Bacterial/genetics , Ribosomes/physiology
9.
Nucleic Acids Res ; 47(21): 11326-11343, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31642471

ABSTRACT

Ribosome was long considered as a critical yet passive player in protein synthesis. Only recently the role of its basic components, ribosomal RNAs and proteins, in translational control has begun to emerge. Here we examined function of the small ribosomal protein uS3/Rps3, earlier shown to interact with eukaryotic translation initiation factor eIF3, in termination. We identified two residues in consecutive helices occurring in the mRNA entry pore, whose mutations to the opposite charge either reduced (K108E) or increased (R116D) stop codon readthrough. Whereas the latter increased overall levels of eIF3-containing terminating ribosomes in heavy polysomes in vivo indicating slower termination rates, the former specifically reduced eIF3 amounts in termination complexes. Combining these two mutations with the readthrough-reducing mutations at the extreme C-terminus of the a/Tif32 subunit of eIF3 either suppressed (R116D) or exacerbated (K108E) the readthrough phenotypes, and partially corrected or exacerbated the defects in the composition of termination complexes. In addition, we found that K108 affects efficiency of termination in the termination context-specific manner by promoting incorporation of readthrough-inducing tRNAs. Together with the multiple binding sites that we identified between these two proteins, we suggest that Rps3 and eIF3 closely co-operate to control translation termination and stop codon readthrough.


Subject(s)
Codon, Terminator/metabolism , Eukaryotic Initiation Factor-3/metabolism , Peptide Chain Termination, Translational , Ribosomal Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Binding Sites/genetics , Eukaryotic Initiation Factor-3/genetics , Organisms, Genetically Modified , Peptide Chain Termination, Translational/genetics , Protein Binding , Protein Biosynthesis/genetics , RNA, Transfer/metabolism , Ribosomal Proteins/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
10.
Int J Mol Sci ; 22(11)2021 May 23.
Article in English | MEDLINE | ID: mdl-34071057

ABSTRACT

Cytosolic ribosomes (cytoribosomes) are macromolecular ribonucleoprotein complexes that are assembled from ribosomal RNA and ribosomal proteins, which are essential for protein biosynthesis. Mitochondrial ribosomes (mitoribosomes) perform translation of the proteins essential for the oxidative phosphorylation system. The biogenesis of cytoribosomes and mitoribosomes includes ribosomal RNA processing, modification and binding to ribosomal proteins and is assisted by numerous biogenesis factors. This is a major energy-consuming process in the cell and, therefore, is highly coordinated and sensitive to several cellular stressors. In mitochondria, the regulation of mitoribosome biogenesis is essential for cellular respiration, a process linked to cell growth and proliferation. This review briefly overviews the key stages of cytosolic and mitochondrial ribosome biogenesis; summarizes the main steps of ribosome biogenesis alterations occurring during tumorigenesis, highlighting the changes in the expression level of cytosolic ribosomal proteins (CRPs) and mitochondrial ribosomal proteins (MRPs) in different types of tumors; focuses on the currently available information regarding the extra-ribosomal functions of CRPs and MRPs correlated to cancer; and discusses the role of CRPs and MRPs as biomarkers and/or molecular targets in cancer treatment.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms/metabolism , Organelle Biogenesis , Ribosomes , Animals , Apoptosis , Autophagy , Cell Cycle , Cell Movement , Cell Nucleolus/metabolism , Cytosol/metabolism , DNA Repair , Endoplasmic Reticulum Stress , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Mitochondrial/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/physiology , Ribosomes/physiology
11.
BMC Plant Biol ; 20(1): 463, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032526

ABSTRACT

BACKGROUND: In plants, each ribosomal protein (RP) is encoded by a small gene family but it is largely unknown whether the family members are functionally diversified. There are two RPL23a paralogous genes (RPL23aA and RPL23aB) encoding cytoplasmic ribosomal proteins in Arabidopsis thaliana. Knock-down of RPL23aA using RNAi impeded growth and led to morphological abnormalities, whereas knock-out of RPL23aB had no observable phenotype, thus these two RPL23a paralogous proteins have been used as examples of ribosomal protein paralogues with functional divergence in many published papers. RESULTS: In this study, we characterized T-DNA insertion mutants of RPL23aA and RPL23aB. A rare non-allelic non-complementation phenomenon was found in the F1 progeny of the rpl23aa X rpl23ab cross, which revealed a dosage effect of these two genes. Both RPL23aA and RPL23aB were found to be expressed almost in all examined tissues as revealed by GUS reporter analysis. Expression of RPL23aB driven by the RPL23aA promoter can rescue the phenotype of rpl23aa, indicating these two proteins are actually equivalent in function. Interestingly, based on the publicly available RNA-seq data, we found that these two RPL23a paralogues were expressed in a concerted manner and the expression level of RPL23aA was much higher than that of RPL23aB at different developmental stages and in different tissues. CONCLUSIONS: Our findings suggest that the two RPL23a paralogous proteins are functionally equivalent but the two genes are not. RPL23aA plays a predominant role due to its higher expression levels. RPL23aB plays a lesser role due to its lower expression. The presence of paralogous genes for the RPL23a protein in plants might be necessary to maintain its adequate dosage.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Genes, Plant , Ribosomal Proteins/genetics , Arabidopsis/physiology , Arabidopsis Proteins/physiology , DNA, Bacterial , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Plant , Mutation , Promoter Regions, Genetic , Ribosomal Proteins/physiology
12.
Mol Biol Rep ; 47(8): 6083-6090, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32748020

ABSTRACT

Ribosomal protein S3 (RPS3) is a component of the 40S ribosomal subunit. It is known to function in ribosome biogenesis and as an endonuclease. RPS3 has been shown to be over expressed in colon adenocarcinoma but its role in colon cancer is still unknown. In this study, we aim at determining the expression levels of RPS3 in a colon cancer cell line Caco-2 compared to a normal colon mucosa cell line NCM-460 and study the effects of targeting this protein by siRNA on cellular behavior. RPS3 was found to be expressed in both cell lines. However, siRNA treatment showed a more protruding effect on Caco-2 cells compared to NCM-460 cells. RPS3 knockdown led to a significant decrease in the proliferation, survival, migration and invasion and an increase in the apoptosis of Caco-2 cells. Western blot analysis demonstrated that these effects correlated with an increase in the level of the tumor suppressor p53 and a decrease in the level and activity of lactate dehydrogenase (LDH), an enzyme involved in the metabolism of cancer cells. No significant effect was shown in normal colon NCM-460 cells. Targeting p53 by siRNA did not affect RPS3 levels indicating that p53 may be a downstream target of RPS3. However, the concurrent knockdown of RPS3 and p53 showed no change in LDH level in Caco-2 cells suggesting an interesting interplay among the three proteins. These findings might present RPS3 as a selective molecular marker in colon cancer and an attractive target for colon cancer therapy.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , L-Lactate Dehydrogenase/biosynthesis , Neoplasm Proteins/physiology , Ribosomal Proteins/physiology , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/genetics , Apoptosis , Cell Line, Tumor , Colon/metabolism , Colonic Neoplasms/genetics , Gene Knockdown Techniques , Humans , Intestinal Mucosa/metabolism , L-Lactate Dehydrogenase/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/genetics
13.
Mol Cell ; 46(2): 171-86, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541556

ABSTRACT

MicroRNAs (miRNAs) regulate physiological and pathological processes by inducing posttranscriptional repression of target messenger RNAs (mRNAs) via incompletely understood mechanisms. To discover factors required for human miRNA activity, we performed an RNAi screen using a reporter cell line of miRNA-mediated repression of translation initiation. We report that reduced expression of ribosomal protein genes (RPGs) dissociated miRNA complexes from target mRNAs, leading to increased polysome association, translation, and stability of miRNA-targeted mRNAs relative to untargeted mRNAs. RNA sequencing of polysomes indicated substantial overlap in sets of genes exhibiting increased or decreased polysomal association after Argonaute or RPG knockdowns, suggesting similarity in affected pathways. miRNA profiling of monosomes and polysomes demonstrated that miRNAs cosediment with ribosomes. RPG knockdowns decreased miRNAs in monosomes and increased their target mRNAs in polysomes. Our data show that most miRNAs repress translation and that the levels of RPGs modulate miRNA-mediated repression of translation initiation.


Subject(s)
MicroRNAs/physiology , Peptide Chain Initiation, Translational/genetics , Ribosomal Proteins/genetics , HeLa Cells , Humans , MicroRNAs/genetics , RNA Interference , Ribosomal Proteins/metabolism , Ribosomal Proteins/physiology , Tumor Suppressor Protein p53/genetics
14.
Nucleic Acids Res ; 46(4): 2096-2106, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29361028

ABSTRACT

Ribosome biogenesis in eukaryotes is a complicated process that involves association and dissociation of numerous assembly factors and snoRNAs. The yeast small ribosomal subunit is first assembled into 90S pre-ribosomes in an ordered and dynamic manner. Efg1 is a protein with no recognizable domain that is associated with early 90S particles. Here, we determine the crystal structure of Efg1 from Chaetomium thermophilum at 3.3 Å resolution, revealing a novel elongated all-helical structure. Efg1 is not located in recently determined cryo-EM densities of 90S likely due to its low abundance in mature 90S. Genetic analysis in Saccharomyces cerevisiae shows that the functional core of Efg1 contains two helical hairpins composed of highly conserved residues. Depletion of Efg1 blocks 18S rRNA processing at sites A1 and A2, but not at site A0, and production of small ribosomal subunits. Efg1 is initially recruited by the 5' domain of 18S rRNA. Its absence disturbs the assembly of the 5' domain and inhibits release of U14 snoRNA from 90S. Our study shows that Efg1 is required for early assembly and reorganization of the 5' domain of 18S rRNA.


Subject(s)
Chaetomium , Fungal Proteins/chemistry , Fungal Proteins/physiology , Ribosomal Proteins/chemistry , Ribosomal Proteins/physiology , Crystallography, X-Ray , Models, Molecular , RNA Processing, Post-Transcriptional , RNA, Ribosomal, 18S/metabolism , RNA, Small Nucleolar/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosomes/metabolism , Sequence Alignment
15.
Nucleic Acids Res ; 46(9): 4715-4732, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29788267

ABSTRACT

The contribution of most ribosomal proteins to ribosome synthesis has been quite well analysed in Saccharomyces cerevisiae. However, few yeast ribosomal proteins still await characterization. Herein, we show that L14, an essential 60S ribosomal protein, assembles in the nucleolus at an early stage into pre-60S particles. Depletion of L14 results in a deficit in 60S subunits and defective processing of 27SA2 and 27SA3 to 27SB pre-rRNAs. As a result, 27S pre-rRNAs are subjected to turnover and export of pre-60S particles is blocked. These phenotypes likely appear as the direct consequence of the reduced pre-60S particle association not only of L14 upon its depletion but also of a set of neighboring ribosomal proteins located at the solvent interface of 60S subunits and the adjacent region surrounding the polypeptide exit tunnel. These pre-60S intermediates also lack some essential trans-acting factors required for 27SB pre-rRNA processing but accumulate practically all factors required for processing of 27SA3 pre-rRNA. We have also analysed the functional interaction between the eukaryote-specific carboxy-terminal extensions of the neighboring L14 and L16 proteins. Our results indicate that removal of the most distal parts of these extensions cause slight translation alterations in mature 60S subunits.


Subject(s)
Ribosomal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Saccharomyces cerevisiae Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , RNA, Ribosomal, 5.8S/metabolism , Ribosomal Proteins/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/physiology
16.
Carcinogenesis ; 40(1): 52-60, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30247545

ABSTRACT

Ribosomal stress is known to increase cancer risk; however, the molecular mechanism underlying its various effects on cancer remains unclear. To decipher this puzzle, we investigated the upstream signaling pathway that might be involved in promoting ribosomal stress that leads to tumor progression. Our results suggested that inhibition of kinase PIM1 attenuated PC3 cell growth and motility following the condensed cellular body and decreased protein translation in PIM1-inhibited cells. In addition, PIM1 was found to be a component of the small 40S ribosomal subunit and could regulate the expression of ribosomal small subunit protein 7 (RPS7). Our investigation also revealed that PIM1 enhanced the protein stability of c-Myc. Furthermore, a functional E-box motif was found upstream of the transcription start site in RPS7, and RPS7 has been proven to be a transcriptional target of c-Myc. Additionally, knocking down RPS7 dramatically reduced cell growth in vitro and in vivo, whereas enhancing RPS7 expression reversed the condensed cellular body and decreased protein translation resulted from PIM1 inhibition. Finally, biochemical recurrence-free survival and overall survival analysis indicated that the concomitant upregulation of PIM1 and RPS7 correlated with the worst prognosis of prostate cancer (PCa). Overall, our results demonstrated that kinase PIM1 promotes cell growth through c-Myc-RPS7-induced ribosomal stress in PCa. These findings substantially expanded our understanding on the molecular mechanism of PIM1-promoted abnormal ribosomal biosynthesis in tumorigenesis and tumor progression in PCa. Therapies that target molecules involved in PIM1-RPS7-induced ribosomal stress could provide a promising approach to treating PCa.


Subject(s)
Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/physiology , Proto-Oncogene Proteins c-pim-1/physiology , Ribosomal Proteins/physiology , Ribosomes/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Ribosome Subunits, Small, Eukaryotic/physiology
17.
J Biol Chem ; 293(34): 13284-13296, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29980600

ABSTRACT

Small nucleolar RNAs (snoRNAs) are noncoding RNAs that guide chemical modifications of structural RNAs. Whereas snoRNAs primarily localize in the nucleolus, where their canonical function is to target nascent ribosomal RNAs for 2'-O-methylation, recent studies provide evidence that snoRNAs traffic out of the nucleus. Furthermore, RNA-Seq data indicate that extracellular vesicles released from cells contain snoRNAs. However, it is not known whether snoRNA secretion is regulated or whether secreted snoRNAs are functional. Here, we show that inflammation stimulates secretion of Rpl13a snoRNAs U32a (SNORD32a), U33 (SNORD33), U34 (SNORD34), and U35a (SNORD35a) from cultured macrophages, in mice, and in human subjects. Secreted snoRNAs co-fractionate with extracellular vesicles and are taken up by recipient cells. In a murine parabiosis model, we demonstrate that snoRNAs travel through the circulation to function in distant tissues. These findings support a previously unappreciated link between inflammation and snoRNA secretion in mice and humans and uncover a potential role for secreted snoRNAs in cell-cell communication.


Subject(s)
Cell Nucleolus/metabolism , Cell Nucleus/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/chemistry , RNA, Small Nucleolar/metabolism , Ribosomal Proteins/physiology , Animals , Biological Transport , Cell Nucleolus/genetics , Cell Nucleus/genetics , Female , Humans , Male , Methylation , Mice , Mice, Knockout , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/genetics
18.
J Biol Chem ; 293(33): 12770-12780, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29959229

ABSTRACT

Set7/9 (also known as Set7, Set9, Setd7, and Kmt7) is a lysine methyltransferase that catalyzes the methylation of multiple substrates, including histone H3 and non-histone proteins. Although not essential for normal development and physiology, Set7/9-mediated methylation events play important roles in regulating cellular pathways involved in various human diseases, making Set7/9 a promising therapeutic target. Multiple Set7/9 inhibitors have been developed, which exhibit varying degrees of potency and selectivity in vitro However, validation of these compounds in vivo has been hampered by the lack of a reliable cellular biomarker for Set7/9 activity. Here, we report the identification of Rpl29, a ribosomal protein abundantly expressed in all cell types, as a major substrate of Set7/9. We show that Rpl29 lysine 5 (Rpl29K5) is methylated exclusively by Set7/9 and can be demethylated by Lsd1 (also known as Kdm1a). Rpl29 is not a core component of the ribosome translational machinery and plays a regulatory role in translation efficiency. Our results indicate that Rpl29 methylation has no effect on global protein synthesis but affects Rpl29 subcellular localization. Using an Rpl29 methylation-specific antibody, we demonstrate that Rpl29K5 methylation is present ubiquitously and validate that (R)-PFI-2, a Set7/9 inhibitor, efficiently reduces Rpl29K5 methylation in cell lines. Thus, Rpl29 methylation can serve as a specific cellular biomarker for measuring Set7/9 activity.


Subject(s)
Blood Coagulation Factors/genetics , DNA Methylation , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/chemistry , Ribosomal Proteins/physiology , Animals , Blood Coagulation Factors/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Male , Mice, Knockout , Protein Processing, Post-Translational , RNA-Binding Proteins , Transcription, Genetic
19.
Cell Biol Int ; 42(1): 121-129, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28949066

ABSTRACT

In previous work, we presented experimental and theoretical evidence that D-3F or 4-N-(2-Amino-3-fluoropyridine)-4-deoxidation-4'-demethylepipofophyllotoxin induced G2 /M phase arrest and apoptosis, purportedly by increasing the expression of P53. However, the precise mechanism of D-3F action is currently unknown. Here, we investigated the mechanism by which D-3F treatment induces increased expression of P53. This study showed that D-3F definitively inhibited the activity of topoisomerase II in a dose-dependent manner and resulted in DNA damage. The results were in overall agreement with modeling and docking studies performed on D-3F. In addition, D-3F increased the levels of P53 and P21 in HeLa cells in a dose-dependent manner, this in turn prolonged the half-life of P53. Taken together, these data suggested that D-3F-mediated transient enhancement of P53 stabilization may be critical for the P53/P21 signalling pathway leading to G2 /M phase arrest on HeLa cells. Furthermore, D-3F downregulated the phosphorylation of E3 ubiquitin-protein ligase murine double minute 2 (Mdm2) at Ser166, inhibited Mdm2-mediated ubiquitination of P53, and released 60S ribosomal protein L11 (RPL11) from the nucleolus into the nucleoplasm. To conclude, the topoisomerase II inhibitor D-3F causes P53 to accumulate in HeLa cell lines by enhancing its stability as a result of DNA-damage induced RPL11 relocalization and subsequent blocking of the P53-Mdm2 feedback loop.


Subject(s)
Ribosomal Proteins/physiology , Topoisomerase II Inhibitors/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Nucleolus , DNA Damage , Genes, p53/drug effects , Genes, p53/physiology , HeLa Cells , Humans , Phosphorylation , Podophyllum/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction , Topoisomerase II Inhibitors/pharmacology
20.
J Immunol ; 197(6): 2280-9, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27489283

ABSTRACT

Although ribosomal proteins (RP) are thought to primarily facilitate biogenesis of the ribosome and its ability to synthesize protein, emerging evidence suggests that individual RP can perform critical regulatory functions that control developmental processes. We showed previously that despite the ubiquitous expression of the RP ribosomal protein L22 (Rpl22), germline ablation of Rpl22 in mice causes a selective, p53-dependent block in the development of αß, but not γδ, T cell progenitors. Nevertheless, the basis by which Rpl22 loss selectively induces p53 in αß T cell progenitors remained unclear. We show in this study that Rpl22 regulates the development of αß T cells by restraining endoplasmic reticulum (ER) stress responses. In the absence of Rpl22, ER stress is exacerbated in αß, but not γδ, T cell progenitors. The exacerbated ER stress in Rpl22-deficient αß T lineage progenitors is responsible for selective induction of p53 and their arrest, as pharmacological induction of stress is sufficient to induce p53 and replicate the selective block of αß T cells, and attenuation of ER stress signaling by knockdown of protein kinase R-like ER kinase, an ER stress sensor, blunts p53 induction and rescues development of Rpl22-deficient αß T cell progenitors. Rpl22 deficiency appears to exacerbate ER stress by interfering with the ability of ER stress signals to block new protein synthesis. Our finding that Rpl22 deficiency exacerbates ER stress responses and induces p53 in αß T cell progenitors provides insight into how a ubiquitously expressed RP can perform regulatory functions that are selectively required by some cell lineages but not others.


Subject(s)
Endoplasmic Reticulum Stress , Gene Expression Regulation , Precursor Cells, T-Lymphoid/physiology , RNA-Binding Proteins/physiology , Receptors, Antigen, T-Cell, alpha-beta , Ribosomal Proteins/physiology , Signal Transduction , T-Lymphocyte Subsets/physiology , Animals , Cell Differentiation , Cell Lineage/physiology , Mice , Ribosomal Proteins/deficiency , T-Lymphocyte Subsets/immunology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL