Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cell ; 159(5): 1086-1095, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416947

ABSTRACT

Fighting viral infections is hampered by the scarcity of viral targets and their variability, resulting in development of resistance. Viruses depend on cellular molecules-which are attractive alternative targets-for their life cycle, provided that they are dispensable for normal cell functions. Using the model organism Drosophila melanogaster, we identify the ribosomal protein RACK1 as a cellular factor required for infection by internal ribosome entry site (IRES)-containing viruses. We further show that RACK1 is an essential determinant for hepatitis C virus translation and infection, indicating that its function is conserved for distantly related human and fly viruses. Inhibition of RACK1 does not affect Drosophila or human cell viability and proliferation, and RACK1-silenced adult flies are viable, indicating that this protein is not essential for general translation. Our findings demonstrate a specific function for RACK1 in selective mRNA translation and uncover a target for the development of broad antiviral intervention.


Subject(s)
Dicistroviridae/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/virology , GTP-Binding Proteins/metabolism , Hepatocytes/virology , Insect Viruses/metabolism , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Line, Tumor , Drosophila melanogaster/metabolism , Hepacivirus/metabolism , Hepatocytes/metabolism , Humans , Models, Molecular , Peptide Initiation Factors/metabolism , Protein Biosynthesis , Receptors for Activated C Kinase , Regulatory Sequences, Ribonucleic Acid , Virus Replication
2.
RNA ; 30(3): 200-212, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38164596

ABSTRACT

rRNA modifications play crucial roles in fine-tuning the delicate balance between translation speed and accuracy, yet the underlying mechanisms remain elusive. Comparative analyses of the rRNA modifications in taxonomically distant bacteria could help define their general, as well as species-specific, roles. In this study, we identified a new methyltransferase, RlmQ, in Staphylococcus aureus responsible for the Gram-positive specific m7G2601, which is not modified in Escherichia coli (G2574). We also demonstrate the absence of methylation on C1989, equivalent to E. coli C1962, which is methylated at position 5 by the Gram-negative specific RlmI methyltransferase, a paralog of RlmQ. Both modifications (S. aureus m7G2601 and E. coli m5C1962) are situated within the same tRNA accommodation corridor, hinting at a potential shared function in translation. Inactivation of S. aureus rlmQ causes the loss of methylation at G2601 and significantly impacts growth, cytotoxicity, and biofilm formation. These findings unravel the intricate connections between rRNA modifications, translation, and virulence in pathogenic Gram-positive bacteria.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Staphylococcus aureus/genetics , Escherichia coli Proteins/genetics , RNA , Virulence/genetics , RNA, Ribosomal, 23S/genetics , Methyltransferases/genetics
3.
Mol Microbiol ; 117(1): 193-214, 2022 01.
Article in English | MEDLINE | ID: mdl-34783400

ABSTRACT

Staphylococcus aureus RsaG is a 3'-untranslated region (3'UTR) derived sRNA from the conserved uhpT gene encoding a glucose-6-phosphate (G6P) transporter expressed in response to extracellular G6P. The transcript uhpT-RsaG undergoes degradation from 5'- to 3'-end by the action of the exoribonucleases J1/J2, which are blocked by a stable hairpin structure at the 5'-end of RsaG, leading to its accumulation. RsaG together with uhpT is induced when bacteria are internalized into host cells or in the presence of mucus-secreting cells. Using MS2-affinity purification coupled with RNA sequencing, several RNAs were identified as targets including mRNAs encoding the transcriptional factors Rex, CcpA, SarA, and the sRNA RsaI. Our data suggested that RsaG contributes to the control of redox homeostasis and adjusts metabolism to changing environmental conditions. RsaG uses different molecular mechanisms to stabilize, degrade, or repress the translation of its mRNA targets. Although RsaG is conserved only in closely related species, the uhpT 3'UTR of the ape pathogen S. simiae harbors an sRNA, whose sequence is highly different, and which does not respond to G6P levels. Our results hypothesized that the 3'UTRs from UhpT transporter encoding mRNAs could have rapidly evolved to enable adaptation to host niches.


Subject(s)
Antiporters/metabolism , Monosaccharide Transport Proteins/metabolism , RNA, Small Untranslated/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Transcription Factors/metabolism , Untranslated Regions/genetics , Adaptation, Physiological , Antiporters/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Gene Expression Regulation, Bacterial , Glucose-6-Phosphate/metabolism , Homeostasis , Monosaccharide Transport Proteins/genetics , Oxidation-Reduction , RNA Stability , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Transcription Factors/genetics
4.
EMBO J ; 38(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30760492

ABSTRACT

Pathogenic bacteria must rapidly adapt to ever-changing environmental signals resulting in metabolism remodeling. The carbon catabolite repression, mediated by the catabolite control protein A (CcpA), is used to express genes involved in utilization and metabolism of the preferred carbon source. Here, we have identified RsaI as a CcpA-repressed small non-coding RNA that is inhibited by high glucose concentrations. When glucose is consumed, RsaI represses translation initiation of mRNAs encoding a permease of glucose uptake and the FN3K enzyme that protects proteins against damage caused by high glucose concentrations. RsaI also binds to the 3' untranslated region of icaR mRNA encoding the transcriptional repressor of exopolysaccharide production and to sRNAs induced by the uptake of glucose-6 phosphate or nitric oxide. Furthermore, RsaI expression is accompanied by a decreased transcription of genes involved in carbon catabolism pathway and an activation of genes involved in energy production, fermentation, and nitric oxide detoxification. This multifaceted RNA can be considered as a metabolic signature when glucose becomes scarce and growth is arrested.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Glucose/deficiency , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Repressor Proteins/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Binding Sites , Biofilms/drug effects , Gene Expression Regulation, Bacterial , Glucose/administration & dosage , Metabolic Networks and Pathways , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Ribosomes/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Sweetening Agents/administration & dosage , Transcriptome
5.
Nat Chem Biol ; 16(1): 69-76, 2020 01.
Article in English | MEDLINE | ID: mdl-31636432

ABSTRACT

Live-cell imaging of RNA has remained a challenge because of the lack of naturally fluorescent RNAs. Recently developed RNA aptamers that can light-up small fluorogenic dyes could overcome this limitation, but they still suffer from poor brightness and photostability. Here, we propose the concept of a cell-permeable fluorogenic dimer of self-quenched sulforhodamine B dyes (Gemini-561) and the corresponding dimerized aptamer (o-Coral) that can drastically enhance performance of the current RNA imaging method. The improved brightness and photostability, together with high affinity of this complex, allowed direct fluorescence imaging in live mammalian cells of RNA polymerase III transcription products as well as messenger RNAs labeled with a single copy of the aptamer; that is, without tag multimerization. The developed fluorogenic module enables fast and sensitive detection of RNA inside live cells, while the proposed design concept opens the route to new generation of ultrabright RNA probes.


Subject(s)
Fluorescent Dyes/chemistry , RNA/chemistry , Spectrometry, Fluorescence/methods , Aptamers, Nucleotide/genetics , Dimerization , Fluorescence , Gene Library , HEK293 Cells , HeLa Cells , Humans , Microfluidics/methods , RNA/analysis , Rhodamines/chemistry , Spectrophotometry
6.
Mol Microbiol ; 113(3): 603-612, 2020 03.
Article in English | MEDLINE | ID: mdl-31705780

ABSTRACT

Trans-acting small regulatory RNAs (sRNAs) are key players in the regulation of gene expression in bacteria. There are hundreds of different sRNAs in a typical bacterium, which in contrast to eukaryotic microRNAs are more heterogeneous in length, sequence composition, and secondary structure. The vast majority of sRNAs function post-transcriptionally by binding to other RNAs (mRNAs, sRNAs) through rather short regions of imperfect sequence complementarity. Besides, every single sRNA may interact with dozens of different target RNAs and impact gene expression either negatively or positively. These facts contributed to the view that the entirety of the regulatory targets of a given sRNA, its targetome, is challenging to identify. However, recent developments show that a more comprehensive sRNAs targetome can be achieved through the combination of experimental and computational approaches. Here, we give a short introduction into these methods followed by a description of two sRNAs, RyhB, and RsaA, to illustrate the particular strengths and weaknesses of these approaches in more details. RyhB is an sRNA involved in iron homeostasis in Enterobacteriaceae, while RsaA is a modulator of virulence in Staphylococcus aureus. Using such a combined strategy, a better appreciation of the sRNA-dependent regulatory networks is now attainable.


Subject(s)
Computational Biology/methods , Gene Expression Regulation, Bacterial/genetics , RNA, Small Untranslated/genetics , Bacteria/genetics , Enterobacteriaceae/genetics , Gene Expression/genetics , Genes, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Small Untranslated/metabolism , Staphylococcus aureus/genetics
7.
Annu Rev Microbiol ; 70: 299-316, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27482744

ABSTRACT

Staphylococcus aureus RNAIII is one of the main intracellular effectors of the quorum-sensing system. It is a multifunctional RNA that encodes a small peptide, and its noncoding parts act as antisense RNAs to regulate the translation and/or the stability of mRNAs encoding transcriptional regulators, major virulence factors, and cell wall metabolism enzymes. In this review, we explain how regulatory proteins and RNAIII are embedded in complex regulatory circuits to express virulence factors in a dynamic and timely manner in response to stress and environmental and metabolic changes.


Subject(s)
Quorum Sensing , RNA, Bacterial/metabolism , Regulon , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Virulence Factors/genetics , Animals , Bacterial Proteins , Gene Expression Regulation, Bacterial , Humans , RNA, Bacterial/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology , Virulence , Virulence Factors/metabolism
8.
Nucleic Acids Res ; 47(18): 9871-9887, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31504767

ABSTRACT

The human opportunistic pathogen Staphylococcus aureus produces numerous small regulatory RNAs (sRNAs) for which functions are still poorly understood. Here, we focused on an atypical and large sRNA called RsaC. Its length varies between different isolates due to the presence of repeated sequences at the 5' end while its 3' part is structurally independent and highly conserved. Using MS2-affinity purification coupled with RNA sequencing (MAPS) and quantitative differential proteomics, sodA mRNA was identified as a primary target of RsaC sRNA. SodA is a Mn-dependent superoxide dismutase involved in oxidative stress response. Remarkably, rsaC gene is co-transcribed with the major manganese ABC transporter MntABC and, consequently, RsaC is mainly produced in response to Mn starvation. This 3'UTR-derived sRNA is released from mntABC-RsaC precursor after cleavage by RNase III. The mature and stable form of RsaC inhibits the synthesis of the Mn-containing enzyme SodA synthesis and favors the oxidative stress response mediated by SodM, an alternative SOD enzyme using either Mn or Fe as co-factor. In addition, other putative targets of RsaC are involved in oxidative stress (ROS and NOS) and metal homeostasis (Fe and Zn). Consequently, RsaC may balance two interconnected defensive responses, i.e. oxidative stress and metal-dependent nutritional immunity.


Subject(s)
Bacterial Proteins/genetics , Oxidative Stress/genetics , Staphylococcal Infections/genetics , Staphylococcus aureus/genetics , Bacterial Proteins/chemistry , Gene Expression Regulation, Bacterial/genetics , Homeostasis/genetics , Humans , Manganese/chemistry , Oxidation-Reduction , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Starvation , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics
9.
Nucleic Acids Res ; 45(11): 6746-6760, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28379505

ABSTRACT

The virulon of Staphyloccocus aureus is controlled by intricate connections between transcriptional and post-transcriptional regulators including proteins and small non-coding RNAs (sRNAs). Many of the sRNAs regulate gene expression through base-pairings with mRNAs. However, characterization of the direct sRNA targets in Gram-positive bacteria remained a difficult challenge. Here, we have applied and adapted the MS2-affinity purification approach coupled to RNA sequencing (MAPS) to determine the targetome of RsaA sRNA of S. aureus, known to repress the synthesis of the transcriptional regulator MgrA. Several mRNAs were enriched with RsaA expanding its regulatory network. Besides mgrA, several of these mRNAs encode a family of SsaA-like enzymes involved in peptidoglycan metabolism and the secreted anti-inflammatory FLIPr protein. Using a combination of in vivo and in vitro approaches, these mRNAs were validated as direct RsaA targets. Quantitative differential proteomics of wild-type and mutant strains corroborated the MAPS results. Additionally, it revealed that RsaA indirectly activated the synthesis of surface proteins supporting previous data that RsaA stimulated biofilm formation and favoured chronic infections. All together, this study shows that MAPS could also be easily applied in Gram-positive bacteria for identification of sRNA targetome.


Subject(s)
Membrane Proteins/genetics , RNA, Untranslated/physiology , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Gene Expression Regulation, Bacterial , Membrane Proteins/metabolism , Proteome/genetics , Proteome/metabolism , RNA Interference , RNA, Bacterial , RNA, Messenger , Staphylococcus aureus/metabolism , Transcriptome
10.
Methods ; 117: 48-58, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28027957

ABSTRACT

RNA-protein interactions are at the bases of many biological processes, forming either tight and stable functional ribonucleoprotein (RNP) complexes (i.e. the ribosome) or transitory ones, such as the complexes involving RNA chaperone proteins. To localize the sites where a protein interacts on an RNA molecule, a common simple and inexpensive biochemical method is the footprinting technique. The protein leaves its footprint on the RNA acting as a shield to protect the regions of interaction from chemical modification or cleavages obtained with chemical or enzymatic nucleases. This method has proven its efficiency to study in vitro the organization of stable RNA-protein complexes. Nevertheless, when the protein binds the RNA very dynamically, with high off-rates, protections are very often difficult to observe. For the analysis of these transient complexes, we describe an alternative strategy adapted from the Site Directed Chemical Probing (SDCP) approach and we compare it with classical footprinting. SDCP relies on the modification of the RNA binding protein to tether an RNA probe (usually Fe-EDTA) to specific protein positions. Local cleavages on the regions of interaction can be used to localize the protein and position its domains on the RNA molecule. This method has been used in the past to monitor stable complexes; we provide here a detailed protocol and a practical example of its application to the study of Escherichia coli RNA chaperone protein S1 and its transitory complexes with mRNAs.


Subject(s)
Molecular Chaperones/chemistry , Molecular Imprinting/methods , RNA-Binding Proteins/chemistry , RNA/chemistry , Staining and Labeling/methods , Base Sequence , Biphenyl Compounds/chemistry , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydroxyl Radical/metabolism , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Probes/chemistry , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Protein Conformation , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Mol Cell ; 37(1): 21-33, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20129052

ABSTRACT

Cold induction of cspA, the paradigm Escherichia coli cold-shock gene, is mainly subject to posttranscriptional control, partly promoted by cis-acting elements of its transcript, whose secondary structure at 37 degrees C and at cold-shock temperature has been elucidated here by enzymatic and chemical probing. The structures, which were also validated by mutagenesis, demonstrate that cspA mRNA undergoes a temperature-dependent structural rearrangement, likely resulting from stabilization in the cold of an otherwise thermodynamically unstable folding intermediate. At low temperature, the "cold-shock" structure is more efficiently translated and somewhat less susceptible to degradation than the 37 degrees C structure. Overall, our data shed light on a molecular mechanism at the basis of the cold-shock response, indicating that cspA mRNA is able to sense temperature downshifts, adopting functionally distinct structures at different temperatures, even without the aid of trans-acting factors. Unlike with other previously studied RNA thermometers, these structural rearrangements do not result from melting of hairpin structures.


Subject(s)
Cold Temperature , Escherichia coli Proteins/physiology , Escherichia coli/genetics , Heat-Shock Proteins/physiology , Protein Biosynthesis , RNA, Messenger/physiology , 5' Untranslated Regions , Acclimatization , Cold Shock Proteins and Peptides , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/genetics , Models, Genetic , Nucleic Acid Conformation , RNA, Messenger/chemistry
12.
Nucleic Acids Res ; 44(21): 10491-10504, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906650

ABSTRACT

Comparative structural studies of ribosomes from various organisms keep offering exciting insights on how species-specific or environment-related structural features of ribosomes may impact translation specificity and its regulation. Although the importance of such features may be less obvious within more closely related organisms, their existence could account for vital yet species-specific mechanisms of translation regulation that would involve stalling, cell survival and antibiotic resistance. Here, we present the first full 70S ribosome structure from Staphylococcus aureus, a Gram-positive pathogenic bacterium, solved by cryo-electron microscopy. Comparative analysis with other known bacterial ribosomes pinpoints several unique features specific to S. aureus around a conserved core, at both the protein and the RNA levels. Our work provides the structural basis for the many studies aiming at understanding translation regulation in S. aureus and for designing drugs against this often multi-resistant pathogen.


Subject(s)
Bacterial Proteins/chemistry , Protein Biosynthesis , RNA, Bacterial/chemistry , Ribosomal Proteins/chemistry , Ribosomes/ultrastructure , Staphylococcus aureus/chemistry , Amino Acid Sequence , Bacillus subtilis/chemistry , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cryoelectron Microscopy , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Molecular Dynamics Simulation , Nucleic Acid Conformation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Bacterial/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Thermus thermophilus/chemistry , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
13.
PLoS Biol ; 11(12): e1001731, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24339747

ABSTRACT

Regulation of translation initiation is well appropriate to adapt cell growth in response to stress and environmental changes. Many bacterial mRNAs adopt structures in their 5' untranslated regions that modulate the accessibility of the 30S ribosomal subunit. Structured mRNAs interact with the 30S in a two-step process where the docking of a folded mRNA precedes an accommodation step. Here, we used a combination of experimental approaches in vitro (kinetic of mRNA unfolding and binding experiments to analyze mRNA-protein or mRNA-ribosome complexes, toeprinting assays to follow the formation of ribosomal initiation complexes) and in vivo (genetic) to monitor the action of ribosomal protein S1 on the initiation of structured and regulated mRNAs. We demonstrate that r-protein S1 endows the 30S with an RNA chaperone activity that is essential for the docking and the unfolding of structured mRNAs, and for the correct positioning of the initiation codon inside the decoding channel. The first three OB-fold domains of S1 retain all its activities (mRNA and 30S binding, RNA melting activity) on the 30S subunit. S1 is not required for all mRNAs and acts differently on mRNAs according to the signals present at their 5' ends. This work shows that S1 confers to the ribosome dynamic properties to initiate translation of a large set of mRNAs with diverse structural features.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/physiology , Protein Biosynthesis/physiology , RNA Folding/physiology , RNA, Messenger/physiology , Ribosomal Proteins/physiology , Ribosomes/physiology , Gene Expression Regulation, Bacterial/physiology
14.
Proc Natl Acad Sci U S A ; 110(39): 15656-61, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24029017

ABSTRACT

Translation initiation factor 2 (IF2) promotes 30S initiation complex (IC) formation and 50S subunit joining, which produces the 70S IC. The architecture of full-length IF2, determined by small angle X-ray diffraction and cryo electron microscopy, reveals a more extended conformation of IF2 in solution and on the ribosome than in the crystal. The N-terminal domain is only partially visible in the 30S IC, but in the 70S IC, it stabilizes interactions between IF2 and the L7/L12 stalk of the 50S, and on its deletion, proper N-formyl-methionyl(fMet)-tRNA(fMet) positioning and efficient transpeptidation are affected. Accordingly, fast kinetics and single-molecule fluorescence data indicate that the N terminus promotes 70S IC formation by stabilizing the productive sampling of the 50S subunit during 30S IC joining. Together, our data highlight the dynamics of IF2-dependent ribosomal subunit joining and the role played by the N terminus of IF2 in this process.


Subject(s)
Prokaryotic Initiation Factor-2/chemistry , Prokaryotic Initiation Factor-2/metabolism , Ribosome Subunits/metabolism , Thermus thermophilus/metabolism , Cryoelectron Microscopy , Models, Molecular , Mutant Proteins/metabolism , Peptide Chain Initiation, Translational , Prokaryotic Initiation Factor-2/ultrastructure , Protein Binding , Protein Structure, Tertiary , Ribosome Subunits, Large, Bacterial , Ribosome Subunits, Small, Bacterial , Scattering, Small Angle , Structure-Activity Relationship , X-Ray Diffraction
15.
Nature ; 455(7211): 416-20, 2008 Sep 18.
Article in English | MEDLINE | ID: mdl-18758445

ABSTRACT

Translation initiation, the rate-limiting step of the universal process of protein synthesis, proceeds through sequential, tightly regulated steps. In bacteria, the correct messenger RNA start site and the reading frame are selected when, with the help of initiation factors IF1, IF2 and IF3, the initiation codon is decoded in the peptidyl site of the 30S ribosomal subunit by the fMet-tRNA(fMet) anticodon. This yields a 30S initiation complex (30SIC) that is an intermediate in the formation of the 70S initiation complex (70SIC) that occurs on joining of the 50S ribosomal subunit to the 30SIC and release of the initiation factors. The localization of IF2 in the 30SIC has proved to be difficult so far using biochemical approaches, but could now be addressed using cryo-electron microscopy and advanced particle separation techniques on the basis of three-dimensional statistical analysis. Here we report the direct visualization of a 30SIC containing mRNA, fMet-tRNA(fMet) and initiation factors IF1 and GTP-bound IF2. We demonstrate that the fMet-tRNA(fMet) is held in a characteristic and precise position and conformation by two interactions that contribute to the formation of a stable complex: one involves the transfer RNA decoding stem which is buried in the 30S peptidyl site, and the other occurs between the carboxy-terminal domain of IF2 and the tRNA acceptor end. The structure provides insights into the mechanism of 70SIC assembly and rationalizes the rapid activation of GTP hydrolysis triggered on 30SIC-50S joining by showing that the GTP-binding domain of IF2 would directly face the GTPase-activated centre of the 50S subunit.


Subject(s)
Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Peptide Chain Initiation, Translational , Ribosomes/metabolism , Ribosomes/ultrastructure , Thermus thermophilus/enzymology , Thermus thermophilus/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Prokaryotic Initiation Factor-1/chemistry , Prokaryotic Initiation Factor-1/genetics , Prokaryotic Initiation Factor-1/metabolism , Prokaryotic Initiation Factor-1/ultrastructure , Prokaryotic Initiation Factor-2/chemistry , Prokaryotic Initiation Factor-2/genetics , Prokaryotic Initiation Factor-2/metabolism , Prokaryotic Initiation Factor-2/ultrastructure , Protein Conformation , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer, Met/chemistry , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism , RNA, Transfer, Met/ultrastructure , Ribosome Subunits/chemistry , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Ribosomes/chemistry , Thermus thermophilus/genetics
17.
Methods Mol Biol ; 2741: 73-100, 2024.
Article in English | MEDLINE | ID: mdl-38217649

ABSTRACT

Noncoding RNAs, including regulatory RNAs (sRNAs), are instrumental in regulating gene expression in pathogenic bacteria, allowing them to adapt to various stresses encountered in their host environments. Staphylococcus aureus is a well-studied model for RNA-mediated regulation of virulence and pathogenicity, with sRNAs playing significant roles in shaping S. aureus interactions with human and animal hosts. By modulating the translation and/or stability of target mRNAs, sRNAs regulate the synthesis of virulence factors and regulatory proteins required for pathogenesis. Moreover, perturbation of the levels of RNA modifications in two other classes of noncoding RNAs, rRNAs, and tRNAs, has been proposed to contribute to stress adaptation. However, the study of how these various factors affect translation regulation has often been restricted to specific genes, using in vivo reporters and/or in vitro translation systems. Genome-wide sequencing approaches offer novel perspectives for studying RNA-dependent regulation. In particular, ribosome profiling methods provide a powerful resource for characterizing the overall landscape of translational regulation, contributing to a better understanding of S. aureus physiopathology. Here, we describe protocols that we have adapted to perform ribosome profiling in S. aureus.


Subject(s)
Ribosome Profiling , Staphylococcus aureus , Animals , Humans , Staphylococcus aureus/metabolism , Gene Expression Regulation , RNA, Ribosomal/genetics , RNA, Messenger/genetics , Gene Expression Regulation, Bacterial
18.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 6): 925-33, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23695237

ABSTRACT

Translation initiation factor 2 (IF2) is involved in the early steps of bacterial protein synthesis. It promotes the stabilization of the initiator tRNA on the 30S initiation complex (IC) and triggers GTP hydrolysis upon ribosomal subunit joining. While the structure of an archaeal homologue (a/eIF5B) is known, there are significant sequence and functional differences in eubacterial IF2, while the trimeric eukaryotic IF2 is completely unrelated. Here, the crystal structure of the apo IF2 protein core from Thermus thermophilus has been determined by MAD phasing and the structures of GTP and GDP complexes were also obtained. The IF2-GTP complex was trapped by soaking with GTP in the cryoprotectant. The structures revealed conformational changes of the protein upon nucleotide binding, in particular in the P-loop region, which extend to the functionally relevant switch II region. The latter carries a catalytically important and conserved histidine residue which is observed in different conformations in the GTP and GDP complexes. Overall, this work provides the first crystal structure of a eubacterial IF2 and suggests that activation of GTP hydrolysis may occur by a conformational repositioning of the histidine residue.


Subject(s)
Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Prokaryotic Initiation Factor-2/chemistry , Thermus thermophilus/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Molecular Conformation , Prokaryotic Initiation Factor-2/metabolism , Thermus thermophilus/metabolism , X-Ray Diffraction
19.
Front Microbiol ; 14: 1118329, 2023.
Article in English | MEDLINE | ID: mdl-36846801

ABSTRACT

Escherichia coli CspA is an RNA binding protein that accumulates during cold-shock and stimulates translation of several mRNAs-including its own. Translation in the cold of cspA mRNA involves a cis-acting thermosensor element, which enhances ribosome binding, and the trans-acting action of CspA. Using reconstituted translation systems and probing experiments we show that, at low temperature, CspA specifically promotes the translation of the cspA mRNA folded in the conformation less accessible to the ribosome, which is formed at 37°C but is retained upon cold shock. CspA interacts with its mRNA without inducing large structural rearrangements, but allowing the progression of the ribosomes during the transition from translation initiation to translation elongation. A similar structure-dependent mechanism may be responsible for the CspA-dependent translation stimulation observed with other probed mRNAs, for which the transition to the elongation phase is progressively facilitated during cold acclimation with the accumulation of CspA.

20.
RNA Biol ; 9(12): 1461-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23134978

ABSTRACT

The endoribonuclease III (RNase III) belongs to the enzyme family known to process double-stranded RNAs. Staphylococcus aureus RNase III was shown to regulate, in concert with the quorum sensing induced RNAIII, the degradation of several mRNAs encoding virulence factors and the transcriptional repressor of toxins Rot. Two of the mRNA-RNAIII complexes involve fully base paired loop-loop interactions with similar sequences that are cleaved by RNase III at a unique position. We show here that the sequence of the base pairs within the loop-loop interaction is not critical for RNase III cleavage, but that the co-axial stacking of three consecutive helices provides an ideal topology for RNase III recognition. In contrast, RNase III induces several strong cleavages in a regular helix, which carries a sequence similar to the loop-loop interaction. The introduction of a bulged loop that interrupts the regular helix restrains the number of cleavages. This work shows that S. aureus RNase III is able to bind and cleave a variety of RNA-mRNA substrates, and that specific structure elements direct the action of RNase III.


Subject(s)
Gene Expression Regulation, Bacterial , RNA, Bacterial/metabolism , Ribonuclease III/metabolism , Staphylococcus aureus/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enzyme Activation , Models, Molecular , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Porins/genetics , Porins/metabolism , Protein Binding , Protein Biosynthesis , Protein Interaction Mapping , Quorum Sensing , RNA Stability , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III/genetics , Staphylococcus aureus/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL