Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 159(5): 1200-1211, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416955

ABSTRACT

Ribosomes elongate at a nonuniform rate during translation. Theoretical models and experiments disagree on the in vivo determinants of elongation rate and the mechanism by which elongation rate affects protein levels. To resolve this conflict, we measured transcriptome-wide ribosome occupancy under multiple conditions and used it to formulate a whole-cell model of translation in E. coli. Our model predicts that elongation rates at most codons during nutrient-rich growth are not limited by the intracellular concentrations of aminoacyl-tRNAs. However, elongation pausing during starvation for single amino acids is highly sensitive to the kinetics of tRNA aminoacylation. We further show that translation abortion upon pausing accounts for the observed ribosome occupancy along mRNAs during starvation. Abortion reduces global protein synthesis, but it enhances the translation of a subset of mRNAs. These results suggest a regulatory role for aminoacylation and abortion during stress, and our study provides an experimentally constrained framework for modeling translation.


Subject(s)
Escherichia coli/physiology , Peptide Chain Elongation, Translational , Amino Acids/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Models, Biological , Protein Biosynthesis , Ribosomes/metabolism
2.
Mol Cell ; 71(2): 229-243.e11, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029003

ABSTRACT

Limitation for amino acids is thought to regulate translation in mammalian cells primarily by signaling through the kinases mTORC1 and GCN2. We find that a selective loss of arginine tRNA charging during limitation for arginine regulates translation through ribosome pausing at two of six arginine codons. Surprisingly, limitation for leucine, an essential and abundant amino acid in protein, results in little or no ribosome pausing. Chemical and genetic perturbation of mTORC1 and GCN2 signaling revealed that their robust response to leucine limitation prevents ribosome pausing, while an insufficient response to arginine limitation leads to loss of tRNA charging and ribosome pausing. Ribosome pausing decreases protein production and triggers premature ribosome termination without reducing mRNA levels. Together, our results suggest that amino acids that are not optimally sensed by the mTORC1 and GCN2 pathways still regulate translation through an evolutionarily conserved mechanism based on codon-specific ribosome pausing.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Protein Biosynthesis/physiology , Amino Acids/metabolism , Animals , Arginine/metabolism , Codon/metabolism , Leucine/metabolism , Mammals/genetics , Peptide Chain Elongation, Translational/genetics , Peptide Chain Elongation, Translational/physiology , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
Blood ; 139(13): 2038-2049, 2022 03 31.
Article in English | MEDLINE | ID: mdl-34861039

ABSTRACT

SF3B1 splicing factor mutations are near-universally found in myelodysplastic syndromes (MDS) with ring sideroblasts (RS), a clonal hematopoietic disorder characterized by abnormal erythroid cells with iron-loaded mitochondria. Despite this remarkably strong genotype-to-phenotype correlation, the mechanism by which mutant SF3B1 dysregulates iron metabolism to cause RS remains unclear due to an absence of physiological models of RS formation. Here, we report an induced pluripotent stem cell model of SF3B1-mutant MDS that for the first time recapitulates robust RS formation during in vitro erythroid differentiation. Mutant SF3B1 induces missplicing of ∼100 genes throughout erythroid differentiation, including proposed RS driver genes TMEM14C, PPOX, and ABCB7. All 3 missplicing events reduce protein expression, notably occurring via 5' UTR alteration, and reduced translation efficiency for TMEM14C. Functional rescue of TMEM14C and ABCB7, but not the non-rate-limiting enzyme PPOX, markedly decreased RS, and their combined rescue nearly abolished RS formation. Our study demonstrates that coordinated missplicing of mitochondrial transporters TMEM14C and ABCB7 by mutant SF3B1 sequesters iron in mitochondria, causing RS formation.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Myelodysplastic Syndromes , Phosphoproteins , ATP-Binding Cassette Transporters , Cell Differentiation/genetics , Flavoproteins/genetics , Flavoproteins/metabolism , Humans , Mitochondrial Proteins/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Phosphoproteins/genetics , Protoporphyrinogen Oxidase/genetics , Protoporphyrinogen Oxidase/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
4.
J Biol Chem ; 298(9): 102277, 2022 09.
Article in English | MEDLINE | ID: mdl-35863436

ABSTRACT

La-related protein 1 (LARP1) has been identified as a key translational inhibitor of terminal oligopyrimidine (TOP) mRNAs downstream of the nutrient sensing protein kinase complex, mTORC1. LARP1 exerts this inhibitory effect on TOP mRNA translation by binding to the mRNA cap and the adjacent 5'TOP motif, resulting in the displacement of the cap-binding protein eIF4E from TOP mRNAs. However, the involvement of additional signaling pathway in regulating LARP1-mediated inhibition of TOP mRNA translation is largely unexplored. In the present study, we identify a second nutrient sensing kinase GCN2 that converges on LARP1 to control TOP mRNA translation. Using chromatin-immunoprecipitation followed by massive parallel sequencing (ChIP-seq) analysis of activating transcription factor 4 (ATF4), an effector of GCN2 in nutrient stress conditions, in WT and GCN2 KO mouse embryonic fibroblasts, we determined that LARP1 is a GCN2-dependent transcriptional target of ATF4. Moreover, we identified GCN1, a GCN2 activator, participates in a complex with LARP1 on stalled ribosomes, suggesting a role for GCN1 in LARP1-mediated translation inhibition in response to ribosome stalling. Therefore, our data suggest that the GCN2 pathway controls LARP1 activity via two mechanisms: ATF4-dependent transcriptional induction of LARP1 mRNA and GCN1-mediated recruitment of LARP1 to stalled ribosomes.


Subject(s)
Amino Acids , Protein Biosynthesis , Protein Serine-Threonine Kinases , RNA 5' Terminal Oligopyrimidine Sequence , RNA, Messenger , RNA-Binding Proteins , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Animals , Cell Culture Techniques , Chromatin Immunoprecipitation , Eukaryotic Initiation Factor-4E/metabolism , Fibroblasts , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
PLoS Biol ; 18(8): e3000757, 2020 08.
Article in English | MEDLINE | ID: mdl-32833957

ABSTRACT

In eukaryotes, conserved mechanisms ensure that cell growth is coordinated with nutrient availability. Overactive growth during nutrient limitation ("nutrient-growth dysregulation") can lead to rapid cell death. Here, we demonstrate that cells can adapt to nutrient-growth dysregulation by evolving major metabolic defects. Specifically, when yeast lysine-auxotrophic mutant lys- encountered lysine limitation, an evolutionarily novel stress, cells suffered nutrient-growth dysregulation. A subpopulation repeatedly evolved to lose the ability to synthesize organosulfurs (lys-orgS-). Organosulfurs, mainly reduced glutathione (GSH) and GSH conjugates, were released by lys- cells during lysine limitation when growth was dysregulated, but not during glucose limitation when growth was regulated. Limiting organosulfurs conferred a frequency-dependent fitness advantage to lys-orgS- by eliciting a proper slow growth program, including autophagy. Thus, nutrient-growth dysregulation is associated with rapid organosulfur release, which enables the selection of organosulfur auxotrophy to better tune cell growth to the metabolic environment. We speculate that evolutionarily novel stresses can trigger atypical release of certain metabolites, setting the stage for the evolution of new ecological interactions.


Subject(s)
Adaptation, Physiological/genetics , Lysine/pharmacology , Metabolic Networks and Pathways/drug effects , Nutrients/pharmacology , Saccharomyces cerevisiae/metabolism , Autophagy/drug effects , Autophagy/genetics , Biological Evolution , Glucose/metabolism , Glucose/pharmacology , Lysine/deficiency , Metabolic Networks and Pathways/genetics , Nitrogen/metabolism , Nitrogen/pharmacology , Nutrients/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sirolimus/pharmacology , Stress, Physiological
6.
PLoS Biol ; 17(9): e3000396, 2019 09.
Article in English | MEDLINE | ID: mdl-31532761

ABSTRACT

The canonical model of eukaryotic translation posits that efficient translation initiation increases protein expression and mRNA stability. Contrary to this model, we find that increasing initiation rate can decrease both protein expression and stability of certain mRNAs in the budding yeast Saccharomyces cerevisiae. These mRNAs encode a stretch of polybasic residues that cause ribosome stalling. Our computational modeling predicts that the observed decrease in gene expression at high initiation rates occurs when ribosome collisions at stalls stimulate abortive termination of the leading ribosome or cause endonucleolytic mRNA cleavage. Consistent with this prediction, the collision-associated quality-control factors Asc1 and Hel2 (orthologs of human RACK1 and ZNF598, respectively) decrease gene expression from stall-containing mRNAs only at high initiation rates. Remarkably, hundreds of S. cerevisiae mRNAs that contain ribosome stall sequences also exhibit lower translation efficiency. We propose that inefficient translation initiation allows these stall-containing endogenous mRNAs to escape collision-stimulated reduction in gene expression.


Subject(s)
Peptide Chain Initiation, Translational , RNA, Messenger/physiology , Ribosomes/physiology , Adaptor Proteins, Signal Transducing/physiology , GTP-Binding Proteins/physiology , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/physiology , Ubiquitin-Protein Ligases/physiology
7.
PLoS Pathog ; 15(1): e1007518, 2019 01.
Article in English | MEDLINE | ID: mdl-30673779

ABSTRACT

Translation can initiate at alternate, non-canonical start codons in response to stressful stimuli in mammalian cells. Recent studies suggest that viral infection and anti-viral responses alter sites of translation initiation, and in some cases, lead to production of novel immune epitopes. Here we systematically investigate the extent and impact of alternate translation initiation in cells infected with influenza virus. We perform evolutionary analyses that suggest selection against non-canonical initiation at CUG codons in influenza virus lineages that have adapted to mammalian hosts. We then use ribosome profiling with the initiation inhibitor lactimidomycin to experimentally delineate translation initiation sites in a human lung epithelial cell line infected with influenza virus. We identify several candidate sites of alternate initiation in influenza mRNAs, all of which occur at AUG codons that are downstream of canonical initiation codons. One of these candidate downstream start sites truncates 14 amino acids from the N-terminus of the N1 neuraminidase protein, resulting in loss of its cytoplasmic tail and a portion of the transmembrane domain. This truncated neuraminidase protein is expressed on the cell surface during influenza virus infection, is enzymatically active, and is conserved in most N1 viral lineages. We do not detect globally higher levels of alternate translation initiation on host transcripts upon influenza infection or during the anti-viral response, but the subset of host transcripts induced by the anti-viral response is enriched for alternate initiation sites. Together, our results systematically map the landscape of translation initiation during influenza virus infection, and shed light on the evolutionary forces shaping this landscape.


Subject(s)
Orthomyxoviridae Infections/genetics , Orthomyxoviridae/genetics , Peptide Chain Initiation, Translational/genetics , Animals , Birds/genetics , Codon/genetics , Codon, Initiator/genetics , Codon, Initiator/metabolism , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/genetics , Influenza, Human/genetics , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational/genetics , Proteins/metabolism , Proteomics/methods , RNA, Messenger/metabolism , Ribosomes/metabolism , Sequence Homology, Amino Acid , Swine/virology , Transcriptome/genetics
8.
Proc Natl Acad Sci U S A ; 110(6): 2419-24, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23277573

ABSTRACT

The genetic code underlying protein synthesis is a canonical example of a degenerate biological system. Degeneracies in physical and biological systems can be lifted by external perturbations, thus allowing degenerate systems to exhibit a wide range of behaviors. Here we show that the degeneracy of the genetic code is lifted by environmental perturbations to regulate protein levels in living cells. By measuring protein synthesis rates from a synthetic reporter library in Escherichia coli, we find that environmental perturbations, such as reduction of cognate amino acid supply, lift the degeneracy of the genetic code by splitting codon families into a hierarchy of robust and sensitive synonymous codons. Rates of protein synthesis associated with robust codons are up to 100-fold higher than those associated with sensitive codons under these conditions. We find that the observed hierarchy between synonymous codons is not determined by usual rules associated with tRNA abundance and codon usage. Rather, competition among tRNA isoacceptors for aminoacylation underlies the robustness of protein synthesis. Remarkably, the hierarchy established using the synthetic library also explains the measured robustness of synthesis for endogenous proteins in E. coli. We further found that the same hierarchy is reflected in the fitness cost of synonymous mutations in amino acid biosynthesis genes and in the transcriptional control of σ-factor genes. Our study suggests that organisms can exploit degeneracy lifting as a general strategy to adapt protein synthesis to their environment.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Amino Acids/metabolism , Bacterial Proteins/genetics , Base Sequence , Codon/genetics , Environment , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Code , Kinetics , Luminescent Proteins/genetics , Models, Genetic , Open Reading Frames , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
Cancer Cell ; 41(5): 853-870.e13, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37084735

ABSTRACT

We uncover a tumor-suppressive process in urothelium called transcriptional-translational conflict caused by deregulation of the central chromatin remodeling component ARID1A. Loss of Arid1a triggers an increase in a nexus of pro-proliferation transcripts, but a simultaneous inhibition of the eukaryotic elongation factor 2 (eEF2), which results in tumor suppression. Resolution of this conflict through enhancing translation elongation speed enables the efficient and precise synthesis of a network of poised mRNAs resulting in uncontrolled proliferation, clonogenic growth, and bladder cancer progression. We observe a similar phenomenon in patients with ARID1A-low tumors, which also exhibit increased translation elongation activity through eEF2. These findings have important clinical implications because ARID1A-deficient, but not ARID1A-proficient, tumors are sensitive to pharmacologic inhibition of protein synthesis. These discoveries reveal an oncogenic stress created by transcriptional-translational conflict and provide a unified gene expression model that unveils the importance of the crosstalk between transcription and translation in promoting cancer.


Subject(s)
Chromatin , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics
10.
Microbiologyopen ; 9(2): e960, 2020 02.
Article in English | MEDLINE | ID: mdl-31680488

ABSTRACT

The amino acid serine plays diverse metabolic roles, yet bacteria actively degrade exogenously provided serine via deamination to pyruvate. Serine deamination is thought to be a detoxification mechanism due to the ability of serine to inhibit several biosynthetic reactions, but this pathway remains highly active even in nutrient-replete conditions. While investigating the physiological roles of serine deamination in different growth conditions, we discovered that Escherichia coli cells lacking the sdaCB operon, which encodes the serine transporter SdaC and the serine deaminase SdaB, lyse upon glucose depletion in a medium containing no exogenous serine but all other amino acids and nucleobases. Unexpectedly, this lysis phenotype can be recapitulated by deleting sdaC alone and can be rescued by heterologous expression of SdaC. Lysis of ΔsdaC cells can be prevented by omitting glycine from the medium, inhibiting the glycine cleavage system, or by increasing alanine availability. Together, our results reveal that the serine transporter SdaC plays a critical role in maintaining amino acid homeostasis during shifts in nutrient availability in E. coli.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Glucose/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amino Acids/metabolism , Biological Transport , Energy Metabolism , Gene Expression Regulation, Bacterial , L-Serine Dehydratase/genetics , Microbial Viability/genetics , Operon , Serine/metabolism
11.
Elife ; 62017 05 12.
Article in English | MEDLINE | ID: mdl-28498106

ABSTRACT

Ribosome stalling on mRNAs can decrease protein expression. To decipher ribosome kinetics at stall sites, we induced ribosome stalling at specific codons by starving the bacterium Escherichia coli for the cognate amino acid. We measured protein synthesis rates from a reporter library of over 100 variants that encoded systematic perturbations of translation initiation rate, the number of stall sites, and the distance between stall sites. Our measurements are quantitatively inconsistent with two widely-used kinetic models for stalled ribosomes: ribosome traffic jams that block initiation, and abortive (premature) termination of stalled ribosomes. Rather, our measurements support a model in which collision with a trailing ribosome causes abortive termination of the stalled ribosome. In our computational analysis, ribosome collisions selectively stimulate abortive termination without fine-tuning of kinetic rate parameters at ribosome stall sites. We propose that ribosome collisions serve as a robust timer for translational quality control pathways to recognize stalled ribosomes.


Subject(s)
Escherichia coli/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Computer Simulation , Kinetics
12.
Elife ; 2: e01501, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24347549

ABSTRACT

We report the discovery of a simple environmental sensing mechanism for biofilm formation in the bacterium Bacillus subtilis that operates without the involvement of a dedicated RNA or protein. Certain serine codons, the four TCN codons, in the gene for the biofilm repressor SinR caused a lowering of SinR levels under biofilm-inducing conditions. Synonymous substitutions of these TCN codons with AGC or AGT impaired biofilm formation and gene expression. Conversely, switching AGC or AGT to TCN codons upregulated biofilm formation. Genome-wide ribosome profiling showed that ribosome density was higher at UCN codons than at AGC or AGU during biofilm formation. Serine starvation recapitulated the effect of biofilm-inducing conditions on ribosome occupancy and SinR production. As serine is one of the first amino acids to be exhausted at the end of exponential phase growth, reduced translation speed at serine codons may be exploited by other microbes in adapting to stationary phase. DOI: http://dx.doi.org/10.7554/eLife.01501.001.


Subject(s)
Bacillus subtilis/metabolism , Biosensing Techniques , Serine/metabolism , Bacillus subtilis/genetics , Biofilms , Codon
SELECTION OF CITATIONS
SEARCH DETAIL