Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 416
Filter
1.
Mol Biol Cell ; 33(1): ar7, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34731012

ABSTRACT

We demonstrate here that mitoribosomal protein synthesis, responsible for the synthesis of oxidative phosphorylation (OXPHOS) subunits encoded by the mitochondrial genome, occurs at high levels during glycolysis fermentation and in a manner uncoupled from OXPHOS complex assembly regulation. Furthermore, we provide evidence that the mitospecific domain of Mrp7 (bL27), a mitoribosomal component, is required to maintain mitochondrial protein synthesis during fermentation but is not required under respiration growth conditions. Maintaining mitotranslation under high-glucose-fermentation conditions also involves Mam33 (p32/gC1qR homologue), a binding partner of Mrp7's mitospecific domain, and together they confer a competitive advantage for a cell's ability to adapt to respiration-based metabolism when glucose becomes limiting. Furthermore, our findings support that the mitoribosome, and specifically the central protuberance region, may be differentially regulated and/or assembled, under the different metabolic conditions of fermentation and respiration. On the basis of our findings, we propose that the purpose of mitotranslation is not limited to the assembly of OXPHOS complexes, but also plays a role in mitochondrial signaling critical for switching cellular metabolism from a glycolysis- to a respiration-based state.


Subject(s)
Cell Respiration/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Fermentation/physiology , Glucose/metabolism , Glycolysis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Mitochondrial Ribosomes/metabolism , Oxidative Phosphorylation , Protein Biosynthesis , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology
2.
Genes (Basel) ; 12(12)2021 12 16.
Article in English | MEDLINE | ID: mdl-34946947

ABSTRACT

Chromatin is a highly dynamic biological entity that allows for both the control of gene expression and the stabilization of chromosomal domains. Given the high degree of plasticity observed in model and non-model organisms, it is not surprising that new chromatin components are frequently described. In this work, we tested the hypothesis that the remnants of the Doc5 transposable element, which retains a heterochromatin insertion pattern in the melanogaster species complex, can be bound by chromatin proteins, and thus be involved in the organization of heterochromatic domains. Using the Yeast One Hybrid approach, we found Rpl22 as a potential interacting protein of Doc5. We further tested in vitro the observed interaction through Electrophoretic Mobility Shift Assay, uncovering that the N-terminal portion of the protein is sufficient to interact with Doc5. However, in situ localization of the native protein failed to detect Rpl22 association with chromatin. The results obtained are discussed in the light of the current knowledge on the extra-ribosomal role of ribosomal protein in eukaryotes, which suggests a possible role of Rpl22 in the determination of the heterochromatin in Drosophila.


Subject(s)
DNA Transposable Elements/genetics , Drosophila Proteins/genetics , Heterochromatin/genetics , RNA-Binding Proteins/genetics , Ribosomal Proteins/genetics , Animals , Chromatin/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , RNA-Binding Proteins/physiology , Ribosomal Proteins/physiology , Ribosomes/metabolism
3.
Dev Cell ; 56(14): 2089-2102.e11, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34242585

ABSTRACT

In ribosomopathies, perturbed expression of ribosome components leads to tissue-specific phenotypes. What accounts for such tissue-selective manifestations as a result of mutations in the ribosome, a ubiquitous cellular machine, has remained a mystery. Combining mouse genetics and in vivo ribosome profiling, we observe limb-patterning phenotypes in ribosomal protein (RP) haploinsufficient embryos, and we uncover selective translational changes of transcripts that controlling limb development. Surprisingly, both loss of p53, which is activated by RP haploinsufficiency, and augmented protein synthesis rescue these phenotypes. These findings are explained by the finding that p53 functions as a master regulator of protein synthesis, at least in part, through transcriptional activation of 4E-BP1. 4E-BP1, a key translational regulator, in turn, facilitates selective changes in the translatome downstream of p53, and this thereby explains how RP haploinsufficiency may elicit specificity to gene expression. These results provide an integrative model to help understand how in vivo tissue-specific phenotypes emerge in ribosomopathies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Extremities/embryology , Haploinsufficiency , Protein Biosynthesis , Protein Processing, Post-Translational , Ribosomal Proteins/physiology , Tumor Suppressor Protein p53/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Body Patterning , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Phenotype , Ribosomes/metabolism
4.
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
5.
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
6.
Article in English | MEDLINE | ID: mdl-33549829

ABSTRACT

Hydrogen sulfide is a natural, widely distributed, poisonous substance and sulfide: quinone oxidoreductase (SQR) is responsible for oxidizing hydrogen sulfide to less toxic sulfur compounds. The increase of SQR mRNA level is an important mechanism for organisms to adapt to hydrogen sulfide-rich environments. However, its transcriptional regulation mechanism is not very clear. In this study, a mitochondrial 28S ribosomal protein S27 (MRPS27), which has never been reported as a transcription factor, was screened by yeast one-hybrid experiment from the echiuran worm Urechis unicinctus, a benthic organism living in marine sediments. Western blotting indicated that UuMRPS27 contents increased significantly in the nuclear extract of hindgut under exposed to 150 µM sulfide. ChIP and EMSA assays demonstrated that UuMRPS27 did bind to the sqr proximal promoter, the key binding sequence was CTAGAG (+12 to +17 of the promoter) detected by DNase I footprinting assay as well as transient transfection experiments. Furthermore, UuMRPS27, as a transcription activator, exhibited the highest transcription activity compared with other reported sqr transcription factors. Our data revealed for the first time the role of MRPS27 acting as a transcription factor which expanded the understanding of sqr transcriptional regulation in sulfide metabolism mechanism.


Subject(s)
Mitochondrial Proteins/physiology , Polychaeta/metabolism , Quinone Reductases/metabolism , Ribosomal Proteins/physiology , Sulfides/metabolism , Transcription Factors/physiology , Animals , Gene Expression Regulation , Polychaeta/genetics , Quinone Reductases/genetics , Transcriptional Activation
7.
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
8.
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
9.
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
10.
Cells ; 9(10)2020 10 15.
Article in English | MEDLINE | ID: mdl-33076379

ABSTRACT

A number of different defects in the process of ribosome production can lead to a diversified spectrum of disorders that are collectively identified as ribosomopathies. The specific factors involved may either play a role only in ribosome biogenesis or have additional extra-ribosomal functions, making it difficult to ascribe the pathogenesis of the disease specifically to an altered ribosome biogenesis, even if the latter is clearly affected. We reviewed the available literature in the field from this point of view with the aim of distinguishing, among ribosomopathies, the ones due to specific alterations in the process of ribosome production from those characterized by a multifactorial pathogenesis.


Subject(s)
RNA, Ribosomal/physiology , Rare Diseases/etiology , Ribosomal Proteins/physiology , Ribosomes/genetics , Ribosomes/pathology , Anemia, Diamond-Blackfan , Anemia, Macrocytic , Chromosome Deletion , Chromosomes, Human, Pair 5 , Dyskeratosis Congenita , Fetal Growth Retardation , Genetic Predisposition to Disease , Hair/abnormalities , Hirschsprung Disease , Humans , Mandibulofacial Dysostosis , Osteochondrodysplasias/congenital , Primary Immunodeficiency Diseases , Psychomotor Disorders , Shwachman-Diamond Syndrome
11.
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
12.
Biochimie ; 177: 87-97, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32828823

ABSTRACT

Emerging observations suggest that ribosomal proteins (RPs) play important extra-ribosomal roles in maintenance of cellular homeostasis. However, the mechanistic insights into these processes have not been extensively explored, especially in pathogenic bacteria. Here, we present our findings on potential extra-ribosomal functions of Mycobacterium tuberculosis (Mtb) RPs. We observed that Mtb RpsB and RpsQ are differentially localized to cell wall fraction in M. tuberculosis (H37Rv), while their M. smegmatis (Msm) homologs are primarily cytosolic. Cellular fractionation of ectopically expressed Mtb RPs in surrogate host (M. smegmatis) also shows their association with cell membrane/cell wall without any gross changes in cell morphology. M. smegmatis expressing Mtb RpsB exhibited altered redox homeostasis, decreased drug-induced ROS, reduced cell wall permeability and increased tolerance to various proteotoxic stress (oxidative stress, SDS and starvation). Mtb RpsB expression was also associated with increased resistance specifically towards Isoniazid, Ethionamide and Streptomycin. The enhanced drug tolerance was specific to Mtb RpsB and not observed upon ectopic expression of M. smegmatis homolog (Msm RpsB). Interestingly, C-terminus deletion in Mtb RpsB affected its localization and reversed the stress-resilient phenotypes. We also observed that M. tuberculosis (H37Rv) with upregulated RpsB levels had higher intracellular survival in macrophage. All these observations hint towards existence of moonlighting roles of Mtb RpsB in imparting stress resilience to mycobacteria. This work open avenues for further exploration of alternative pathways associated with fitness and drug tolerance in mycobacteria.


Subject(s)
Bacterial Proteins/physiology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Ribosomal Proteins/physiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Cell Membrane/metabolism , Cell Wall/metabolism , Cytosol/metabolism , Drug Tolerance/genetics , Humans , Lipids/analysis , Macrophages/metabolism , Macrophages/microbiology , Mutant Proteins/chemistry , Mutant Proteins/physiology , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Oxidation-Reduction , Oxidative Stress/genetics , Permeability , Reactive Oxygen Species/metabolism , Ribosomal Proteins/chemistry , Ribosomes/chemistry , THP-1 Cells
13.
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
14.
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
15.
Biomed Pharmacother ; 127: 110219, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32559850

ABSTRACT

BACKGROUND: Increasing studies have revealed that circular RNAs (circRNAs) contribute to gastric cancer (GC) progression. The circular RNA ribosomal protein L15 (circ-RPL15) is involved in chronic lymphocytic leukemia. However, its expression and functions in GC remain elusive. METHODS: The expression of circ-RPL15 in human GC tissues and adjacent normal tissues, human gastric cancer cell lines (MGC-803, BGC-823, MGN-28, SGC-7901, AGS) and normal gastric mucosal epithelial cell line (GES-1) were detected by RT-PCR. The relationship between circ-RPL15 level and clinical-pathological indicators were also analyzed. Gain- of function experiments of circ-RPL15 and miR-502-3p were conducted to verify their roles in mediating GC cell proliferation, apoptosis and metastasis. Also, the downstream mechanisms of circ-RPL15 were predicted by bioinformatics analysis, and the interactions between circ-RPL15 and miR-502-3p, miR-502-3p and OLFM4 were verified by dual luciferase reporter gene assay and RNA FISH. RESULTS: circ-RPL15 was upregulated in GC tissues and cell lines, and the overexpressed circ-RPL15 was correlated with poorer survival of GC patients. Functionally, circ-RPL15 upregulation distinctly promoted the proliferation, migration and invasion of GC cells and inhibited apoptosis. Mechanistically, circ-RPL15 functioned as a competitive endogenous RNA via sponging miR-502-3p and activated OLFM4/STAT3 pathway. CONCLUSION: circ-RPL15 promotes GC progression and predicts poor prognosis of GC patients, and regulates the malignant phenotypes of GC cells by mediating the miR-502-3p/OLFM4/STAT3 axis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Granulocyte Colony-Stimulating Factor/metabolism , MicroRNAs/metabolism , Ribosomal Proteins/physiology , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/physiopathology , Apoptosis/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Disease Progression , Female , Humans , Male , Middle Aged , Ribosomal Proteins/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Survival Analysis , Up-Regulation
16.
Elife ; 92020 05 20.
Article in English | MEDLINE | ID: mdl-32432546

ABSTRACT

A long-standing problem is how cells that lack one of the highly similar ribosomal proteins (RPs) often display distinct phenotypes. Yeast and other organisms live longer when they lack specific ribosomal proteins, especially of the large 60S subunit of the ribosome. However, longevity is neither associated with the generation time of RP deletion mutants nor with bulk inhibition of protein synthesis. Here, we queried actively dividing RP mutants through the cell cycle. Our data link transcriptional, translational, and metabolic changes to phenotypes associated with the loss of paralogous RPs. We uncovered translational control of transcripts encoding enzymes of methionine and serine metabolism, which are part of one-carbon (1C) pathways. Cells lacking Rpl22Ap, which are long-lived, have lower levels of metabolites associated with 1C metabolism. Loss of 1C enzymes increased the longevity of wild type cells. 1C pathways exist in all organisms and targeting the relevant enzymes could represent longevity interventions.


Subject(s)
Carbon/metabolism , Cell Division/physiology , Cellular Senescence/physiology , Gene Expression Regulation , Protein Biosynthesis , RNA-Binding Proteins/physiology , Ribosomal Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Cell Cycle/genetics , Cell Division/genetics , Cellular Senescence/genetics , Gene Library , Loss of Function Mutation , Methionine/metabolism , Phenotype , RNA, Fungal , RNA-Binding Proteins/genetics , RNA-Seq , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Serine/metabolism
17.
FEBS J ; 287(17): 3794-3813, 2020 09.
Article in English | MEDLINE | ID: mdl-32383535

ABSTRACT

Monocyte infiltration to the site of pathogenic invasion is critical for inflammatory response and host defence. However, this process demands precise regulation as uncontrolled migration of monocytes to the site delays resolution of inflammation and ultimately promotes chronic inflammation. C-C motif chemokine ligand 2 (CCL2) plays a key role in monocyte migration, and hence, its expression should be tightly regulated. Here, we report a post-transcriptional regulation of CCL2 involving the large ribosomal subunit protein L22 (RPL22) in LPS-activated, differentiated THP-1 cells. Early events following LPS treatment include transcriptional upregulation of RPL22 and its nuclear accumulation. The protein binds to the first 20 nt sequence of the 5'UTR of ccl2 mRNA. Simultaneous nuclear translocation of up-frameshift-1 protein and its interaction with RPL22 results in cytoplasmic degradation of the ccl2 mRNA at a later stage. Removal of RPL22 from cells results in increased expression of CCL2 in response to LPS causing disproportionate migration of monocytes. We propose that post-transcriptional regulation of CCL2 by RPL22 fine-tunes monocyte infiltration during a pathogenic insult and maintains homeostasis of the immune response critical to resolution of inflammation. DATABASES: Microarray data are available in NCBI GEO database (Accession No GSE126525).


Subject(s)
Chemokine CCL2/biosynthesis , Inflammation/genetics , Lipopolysaccharides/toxicity , Protein Processing, Post-Translational , RNA-Binding Proteins/physiology , Ribosomal Proteins/physiology , 5' Untranslated Regions , Active Transport, Cell Nucleus , Base Sequence , CRISPR-Cas Systems , Cell Movement , Chemokine CCL2/genetics , Humans , Inflammation/chemically induced , Inflammation/metabolism , MCF-7 Cells , Models, Molecular , Neoplasm Proteins/metabolism , Protein Conformation , Protein Interaction Mapping , RNA Helicases/metabolism , RNA Stability , RNA, Messenger/metabolism , Ribosomal Proteins/deficiency , Sequence Alignment , Sequence Homology, Nucleic Acid , THP-1 Cells , Trans-Activators/metabolism
18.
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
19.
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
20.
Cells ; 8(11)2019 10 24.
Article in English | MEDLINE | ID: mdl-31653044

ABSTRACT

Many facets of ribosome biogenesis and function, including ribosomal RNA (rRNA) transcription, 70S assembly and protein translation, are negatively impacted upon induction of a nutrient stress-sensing signalling pathway termed the stringent response. This stress response is mediated by the alarmones guanosine tetra- and penta-phosphate ((p)ppGpp), the accumulation of which leads to a massive cellular response that slows growth and aids survival. The 70S bacterial ribosome is an intricate structure, with assembly both complex and highly modular. Presiding over the assembly process is a group of P-loop GTPases within the TRAFAC (Translation Factor Association) superclass that are crucial for correct positioning of both early and late stage ribosomal proteins (r-proteins) onto the rRNA. Often described as 'molecular switches', members of this GTPase superfamily readily bind and hydrolyse GTP to GDP in a cyclic manner that alters the propensity of the GTPase to carry out a function. TRAFAC GTPases are considered to act as checkpoints to ribosome assembly, involved in binding to immature sections in the GTP-bound state, preventing further r-protein association until maturation is complete. Here we review our current understanding of the impact of the stringent response and (p)ppGpp production on ribosome maturation in prokaryotic cells, focusing on the inhibition of (p)ppGpp on GTPase-mediated subunit assembly, but also touching upon the inhibition of rRNA transcription and protein translation.


Subject(s)
Ribosomal Proteins/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , GTP Phosphohydrolases/metabolism , Guanosine Pentaphosphate/metabolism , Guanosine Tetraphosphate/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Prokaryotic Cells/metabolism , Protein Binding , Protein Biosynthesis , Pyrophosphatases/metabolism , Ribosomal Proteins/physiology , Ribosomes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL