Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cell ; 73(5): 959-970.e5, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30686592

ABSTRACT

Ribosomes undergo substantial conformational changes during translation elongation to accommodate incoming aminoacyl-tRNAs and translocate along the mRNA template. We used multiple elongation inhibitors and chemical probing to define ribosome conformational states corresponding to differently sized ribosome-protected mRNA fragments (RPFs) generated by ribosome profiling. We show, using various genetic and environmental perturbations, that short 20-22 or classical 27-29 nucleotide RPFs correspond to ribosomes with open or occupied ribosomal A sites, respectively. These distinct states of translation elongation are readily discerned by ribosome profiling in all eukaryotes we tested, including fungi, worms, and mammals. This high-resolution ribosome profiling approach reveals mechanisms of translation-elongation arrest during distinct stress conditions. Hyperosmotic stress inhibits translocation through Rck2-dependent eEF2 phosphorylation, whereas oxidative stress traps ribosomes in a pre-translocation state, independent of Rck2-driven eEF2 phosphorylation. These results provide insights and approaches for defining the molecular events that impact translation elongation throughout biology.


Subject(s)
Gene Expression Profiling/methods , Peptide Chain Elongation, Translational , Ribosomal Proteins/genetics , Ribosomes/genetics , Stress, Physiological , Transcriptome , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Codon , HeLa Cells , Humans , Nucleic Acid Conformation , Osmotic Pressure , Oxidative Stress , Peptide Elongation Factor 2/genetics , Peptide Elongation Factor 2/metabolism , Phosphorylation , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , Transfer RNA Aminoacylation
2.
J Biol Chem ; 288(41): 29530-8, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23963452

ABSTRACT

Eukaryotic peptide release factor 3 (eRF3) is a conserved, essential gene in eukaryotes implicated in translation termination. We have systematically measured the contribution of eRF3 to the rates of peptide release with both saturating and limiting levels of eukaryotic release factor 1 (eRF1). Although eRF3 modestly stimulates the absolute rate of peptide release (∼5-fold), it strongly increases the rate of peptide release when eRF1 is limiting (>20-fold). This effect was generalizable across all stop codons and in a variety of contexts. Further investigation revealed that eRF1 remains associated with ribosomal complexes after peptide release and subunit dissociation and that eRF3 promotes the dissociation of eRF1 from these post-termination complexes. These data are consistent with models where eRF3 principally affects binding interactions between eRF1 and the ribosome, either prior to or subsequent to peptide release. A role for eRF3 as an escort for eRF1 into its fully accommodated state is easily reconciled with its close sequence similarity to the translational GTPase EFTu.


Subject(s)
Peptide Termination Factors/metabolism , Peptides/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Blotting, Western , Catalysis , Codon, Terminator/genetics , Guanosine Triphosphate/metabolism , Kinetics , Models, Genetic , Mutation , Peptide Chain Termination, Translational/genetics , Peptide Termination Factors/genetics , Peptides/genetics , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Binding , Protein Biosynthesis/genetics , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
Development ; 139(17): 3232-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22872088

ABSTRACT

Mutations in the human Shwachman-Bodian-Diamond syndrome (SBDS) gene cause defective ribosome assembly and are associated with exocrine pancreatic insufficiency, chronic neutropenia and skeletal defects. However, the mechanism underlying these phenotypes remains unclear. Here we show that knockdown of the zebrafish sbds ortholog fully recapitulates the spectrum of developmental abnormalities observed in the human syndrome, and further implicate impaired proliferation of ptf1a-expressing pancreatic progenitor cells as the basis for the observed pancreatic phenotype. It is thought that diseases of ribosome assembly share a p53-dependent mechanism. However, loss of p53 did not rescue the developmental defects associated with loss of zebrafish sbds. To clarify the molecular mechanisms underlying the observed organogenesis defects, we performed transcriptional profiling to identify candidate downstream mediators of the sbds phenotype. Among transcripts displaying differential expression, functional group analysis revealed marked enrichment of genes related to ribosome biogenesis, rRNA processing and translational initiation. Among these, ribosomal protein L3 (rpl3) and pescadillo (pes) were selected for additional analysis. Similar to knockdown of sbds, knockdown or mutation of either rpl3 or pes resulted in impaired expansion of pancreatic progenitor cells. The pancreatic phenotypes observed in rpl3- and pes-deficient embryos were also independent of p53. Together, these data suggest novel p53-independent roles for ribosomal biogenesis genes in zebrafish pancreas development.


Subject(s)
Bone Marrow Diseases/genetics , Disease Models, Animal , Exocrine Pancreatic Insufficiency/genetics , Lipomatosis/genetics , Nuclear Proteins/genetics , Pancreas/embryology , Ribosomal Proteins/genetics , Ribosomes/genetics , Zebrafish Proteins/genetics , Zebrafish , Alcian Blue , Animals , Anthraquinones , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Knockdown Techniques , In Situ Hybridization , Nuclear Proteins/deficiency , Oligonucleotide Array Sequence Analysis , Pancreas/metabolism , Ribosomal Protein L3 , Ribosomal Proteins/deficiency , Ribosomes/metabolism , Shwachman-Diamond Syndrome , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/deficiency
4.
J Mol Biol ; 410(1): 118-30, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21570405

ABSTRACT

Ribosomes exist as a heterogenous pool of macromolecular complexes composed of ribosomal RNA molecules, ribosomal proteins, and numerous associated "nonribosomal" proteins. To identify nonribosomal proteins that may modulate ribosome activity, we examined the composition of translationally active and inactive ribosomes using a proteomic multidimensional protein identification technology. Notably, the phosphorylated isoform of glycogen synthase, glycogen synthase 1 (GYS1), was preferentially associated with elongating ribosomes. Depletion of GYS1 affected the translation of a subset of cellular mRNAs, some of which encode proteins that modulate protein biosynthesis. These findings argue that GYS1 abundance, by virtue of its ribosomal association, provides a feedback loop between the energy state of the cells and the translation machinery.


Subject(s)
Glycogen Synthase/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Biomarkers/metabolism , Blotting, Northern , Blotting, Western , Gene Expression Profiling , Glycogen Synthase/antagonists & inhibitors , Glycogen Synthase/genetics , HeLa Cells , Humans , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
Mol Cell Biol ; 30(8): 2006-16, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20154146

ABSTRACT

Cells possess mechanisms that permit survival and recovery from stress, several of which regulate the phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha). We identified the human OGFOD1 protein as a novel stress granule component that regulates the phosphorylation of eIF2alpha and the resumption of translation in cells recovering from arsenite-induced stress. Coimmunoprecipitation studies revealed that OGFOD1 associates with a small subset of stress granule proteins (G3BP1, USP10, Caprin1, and YB-1) and the ribosome in both unstressed and stressed cells. Overexpression of OGFOD1 led to increased abundance of phosphorylated eIF2alpha, both in unstressed cells and in cells exposed to arsenite-induced stress, and to accelerated apoptosis during stress. Conversely, knockdown of OGFOD1 resulted in smaller amounts of phosphorylated eIF2alpha and a faster accumulation of polyribosomes in cells recovering from stress. Finally, OGFOD1 interacted with both eIF2alpha and the eIF2alpha kinase heme-regulated inhibitor (HRI), which was identified as a novel stress granule resident. These findings argue that OGFOD1 plays important proapoptotic roles in the regulation of translation and HRI-mediated phosphorylation of eIF2alpha in cells subjected to arsenite-induced stress.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Nuclear Proteins/metabolism , Oxidative Stress , Animals , Apoptosis , Arsenites/pharmacology , Carrier Proteins/genetics , DNA Helicases , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/genetics , HeLa Cells/drug effects , Humans , Phosphorylation , Poly-ADP-Ribose Binding Proteins , Protein Biosynthesis/drug effects , RNA Helicases , RNA Recognition Motif Proteins , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Teratogens/pharmacology , Thapsigargin/pharmacology , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
6.
Prog Mol Biol Transl Sci ; 90: 187-210, 2009.
Article in English | MEDLINE | ID: mdl-20374742

ABSTRACT

MicroRNAs are 20-21 nucleotides-long noncoding RNAs that function as posttranscriptional regulators of gene expression in a variety of organisms ranging from plants to mammalian cells. These regulators are encoded by approximately 800 genes in the mammalian genome and target half of the mRNAs in mammalian cells. While the biogenesis of microRNAs is fairly well understood, the mechanism by which target genes are regulated remains controversial. The recent discoveries that viruses encode microRNAs or subvert host cell microRNAs has enhanced our knowledge about biological functions of microRNAs during disease and has suggested that microRNAs could be used as targets in antiviral gene therapy. This review will provide a brief history of microRNA research, discuss the biogenesis and mechanisms of microRNAs, and summarize findings that have employed inhibitors of microRNA miR-122 to treat hepatitis C virus-induced liver disease.


Subject(s)
Gene Silencing , MicroRNAs/metabolism , Animals , Gene Expression Regulation , Hepacivirus/metabolism , Humans , Liver/metabolism , Liver/virology , MicroRNAs/genetics , Organ Specificity/genetics
7.
Nat Rev Microbiol ; 4(9): 651-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16912711

ABSTRACT

MicroRNAs (miRNAs), which can be expressed in a cell-type and tissue-specific manner, can influence the activities of genes that control cell growth and differentiation. Viruses often have clear tissue tropisms, raising the possibility that cellular miRNAs might modulate their pathogenesis. In this Review, we discuss recent findings that some vertebrate viruses either encode miRNAs or subvert cellular miRNAs, and that these miRNAs participate in both the infectious and the latent phase of the viral life cycle.


Subject(s)
DNA Viruses/genetics , DNA Viruses/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , DNA Viruses/growth & development , Down-Regulation , Genome, Viral , Humans , Up-Regulation , Vertebrates/virology
8.
Mol Cell Biol ; 22(20): 7258-67, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12242301

ABSTRACT

The SSU processome is required for production of the small ribosomal subunit RNA, the 18S rRNA. Specifically, the U3 small nucleolar RNA (snoRNA) component of the SSU processome is essential for the formation of the conserved central pseudoknot and for cleavages of the pre-rRNA, both of which are required for 18S maturation. To further elucidate how these events are mediated, we examined the regulatory and mechanistic roles of the U3 specific proteins: Imp3p, Imp4p, and Mpp10p. We found that these proteins demonstrated an interdependence with respect to their stability and to their association with the U3 snoRNA. Because mutations in the U3 snoRNA that disrupt pre-rRNA processing confer similar defects on growth and pre-rRNA processing as do carboxy-terminal truncations of Mpp10p, we hypothesized that Mpp10p may be involved in maintaining U3 snoRNA-pre-rRNA base pairing. However, combining the two mutations resulted in a more pronounced cleavage defect at site A(2), suggesting that Mpp10p is also required at an additional mechanistic step. Furthermore, heterologous complementation experiments demonstrate that the last 95 amino acids of yeast Mpp10p are specifically required for growth and pre-rRNA processing at low temperatures.


Subject(s)
Fungal Proteins/metabolism , Phosphoproteins/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Fungal/metabolism , RNA, Ribosomal, 18S/metabolism , RNA, Small Nucleolar/metabolism , Ribonucleoproteins/metabolism , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Fungal Proteins/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Phosphoproteins/genetics , RNA Precursors/chemistry , RNA, Fungal/chemistry , RNA, Ribosomal, 18S/chemistry , RNA, Small Nucleolar/chemistry , Ribonucleoproteins/genetics , Ribosomal Proteins/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
9.
Brain Res ; 945(2): 160-73, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12126878

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited motor neuron disease caused by mutations in the survival motor neuron gene (SMN1). While it has been shown that the SMN protein is involved in spliceosome biogenesis and pre-mRNA splicing, there is increasing evidence indicating that SMN may also perform important functions in the nucleolus. We demonstrate here through the use of a previously characterized polyclonal anti-SMN antibody, abSMN, that the SMN protein shows a striking colocalization with the nucleolar protein, fibrillarin, in both nucleoli and Cajal bodies/gems of primary neurons. Immunoblot analysis with antifibrillarin and two different anti-SMN antibodies reveals that SMN and fibrillarin also cofractionate in the insoluble protein fraction of cultured cell lysates. Immunoprecipitation experiments using whole cell extracts of HeLa cells and cultured neurons revealed that abSMN coprecipitated small amounts of the U3 small nucleolar RNA (snoRNA) previously shown to be associated with fibrillarin in vivo. These studies raise the possibility that SMN may serve a function in rRNA maturation/ribosome synthesis similar to its role in spliceosome biogenesis.


Subject(s)
Cell Nucleolus/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Brain Neoplasms/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein , HeLa Cells , Humans , In Situ Nick-End Labeling , Mice , Microscopy, Fluorescence , Motor Neurons/ultrastructure , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Neuroblastoma/metabolism , Precipitin Tests , RNA-Binding Proteins , Rabbits , Rats , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins , Survival of Motor Neuron 1 Protein , Tumor Cells, Cultured
10.
Nature ; 417(6892): 967-70, 2002 Jun 27.
Article in English | MEDLINE | ID: mdl-12068309

ABSTRACT

Although the U3 small nucleolar RNA (snoRNA), a member of the box C/D class of snoRNAs, was identified with the spliceosomal small nuclear RNAs (snRNAs) over 30 years ago, its function and its associated protein components have remained more elusive. The U3 snoRNA is ubiquitous in eukaryotes and is required for nucleolar processing of pre-18S ribosomal RNA in all organisms where it has been tested. Biochemical and genetic analyses suggest that U3 pre-rRNA base-pairing interactions mediate endonucleolytic pre-rRNA cleavages. Here we have purified a large ribonucleoprotein (RNP) complex from Saccharomyces cerevisiae that contains the U3 snoRNA and 28 proteins. Seventeen new proteins (Utp1 17) and Rrp5 were present, as were ten known components. The Utp proteins are nucleolar and specifically associated with the U3 snoRNA. Depletion of the Utp proteins impedes production of the 18S rRNA, indicating that they are part of the active pre-rRNA processing complex. On the basis of its large size (80S; calculated relative molecular mass of at least 2,200,000) and function, this complex may correspond to the terminal knobs present at the 5' ends of nascent pre-rRNAs. We have termed this large RNP the small subunit (SSU) processome.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Fungal/metabolism , RNA, Ribosomal, 18S/metabolism , RNA, Small Nucleolar/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Chromatin/genetics , Chromatin/metabolism , Macromolecular Substances , Microscopy, Electron , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Fungal/biosynthesis , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Ribosomal, 18S/biosynthesis , RNA, Ribosomal, 18S/chemistry , RNA, Ribosomal, 18S/genetics , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/chemistry , Ribonucleoproteins, Small Nucleolar/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
11.
Mol Cell ; 9(2): 329-39, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11864606

ABSTRACT

Little is understood about the role of nucleolar RNA binding proteins in ribosome biogenesis, although there is a clear need for them based on the strict folding requirements of the pre-rRNA. We have identified a superfamily of RNA binding proteins whose members are required for different stages of ribosome biogenesis. The Imp4 superfamily is composed of five individual families (Imp4, Rpf1, Rpf2, Brx1, and Ssf) that all possess the sigma(70)-like motif, a eukaryotic RNA binding domain with prokaryotic origins. The Imp4 superfamily members associate with RNAs that are consistent with their distinct roles in ribosome biogenesis and suggest the mechanisms by which they function.


Subject(s)
DNA-Directed RNA Polymerases/physiology , Eukaryotic Cells/metabolism , Multigene Family , RNA, Ribosomal/metabolism , RNA-Binding Proteins/physiology , Ribosomes/metabolism , Sigma Factor/physiology , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Nucleolus/metabolism , DNA-Directed RNA Polymerases/chemistry , Models, Biological , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phylogeny , Protein Binding , RNA/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nucleolar/chemistry , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Sigma Factor/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...