Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.743
Filter
1.
Nat Commun ; 15(1): 8173, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289353

ABSTRACT

Riboswitches are structured RNA elements that regulate gene expression upon binding to small molecule ligands. Understanding the mechanisms by which small molecules impact riboswitch activity is key to developing potent, selective ligands for these and other RNA targets. We report the structure-informed design of chemically diverse synthetic ligands for PreQ1 riboswitches. Multiple X-ray co-crystal structures of synthetic ligands with the Thermoanaerobacter tengcongensis (Tte)-PreQ1 riboswitch confirm a common binding site with the cognate ligand, despite considerable chemical differences among the ligands. Structure probing assays demonstrate that one ligand causes conformational changes similar to PreQ1 in six structurally and mechanistically diverse PreQ1 riboswitch aptamers. Single-molecule force spectroscopy is used to demonstrate differential modes of riboswitch stabilization by the ligands. Binding of the natural ligand brings about the formation of a persistent, folded pseudoknot structure, whereas a synthetic ligand decreases the rate of unfolding through a kinetic mechanism. Single round transcription termination assays show the biochemical activity of the ligands, while a GFP reporter system reveals compound activity in regulating gene expression in live cells without toxicity. Taken together, this study reveals that diverse small molecules can impact gene expression in live cells by altering conformational changes in RNA structures through distinct mechanisms.


Subject(s)
Nucleic Acid Conformation , Riboswitch , Thermoanaerobacter , Riboswitch/genetics , Ligands , Thermoanaerobacter/metabolism , Thermoanaerobacter/genetics , Binding Sites , Crystallography, X-Ray , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/chemistry , Gene Expression Regulation, Bacterial , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/chemistry , Kinetics , Models, Molecular
2.
Structure ; 32(9): 1298-1300, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241762

ABSTRACT

In this issue of Structure, Elghondakly et al.1 present the crystal structure of Thermoanaerobacter pseudethanolicus antiterminator LoaP, a member of a ubiquitous family of NusG transcription factors, bound to its target, a dfn RNA hairpin. LoaP uses RNA as a recognition determinant, which is unique among NusG paralogs and makes unusual contacts in the major groove of the RNA.


Subject(s)
Bacterial Proteins , DNA-Directed RNA Polymerases , Thermoanaerobacter , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Thermoanaerobacter/enzymology , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Models, Molecular , RNA/metabolism , RNA/chemistry
3.
Commun Biol ; 7(1): 1130, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271946

ABSTRACT

Pneumolysin (Ply) of Streptococcus pneumoniae (pneumococcus) at relatively high and low levels facilitates pneumococcal invasion into the lung and brain, respectively; however, the regulatory mechanisms of Ply expression are poorly understood. Here, we find that a small RNA plyT, processed from the 3'UTR of the ply operon, is expressed higher in anaerobically- than in statically-cultured pneumococcus D39. Using bioinformatic, biochemical and genetic approaches, we reveal that PlyT inhibits Ply synthesis and hemolytic activities by pairing with an RBS-embedded intergenic region of the ply operon. The RNA-binding protein SPD_1558 facilitates the pairing. Importantly, PlyT inhibition of Ply synthesis is stronger in anaerobic culture and leads to lower Ply abundance. Deletion of plyT decreases the number of pneumococci in the infected mouse brain and reduces the virulence, demonstrating that PlyT-regulated lower Ply in oxygen-void microenvironments, such as the blood, is important for pneumococcus to cross the blood-brain barrier and invade the brain. PlyT-mediated repression of Ply synthesis at anoxic niches is also verified in pneumococcal serotype 4 and 14 strains; moreover, the ply operon with a 3'UTR-embedded plyT, and the pairing sequences of IGR and plyT are highly conserved among pneumococcal strains, implying PlyT-regulated Ply synthesis might be widely employed by pneumococcus.


Subject(s)
3' Untranslated Regions , Bacterial Proteins , Brain , Pneumococcal Infections , Streptococcus pneumoniae , Streptolysins , Streptolysins/metabolism , Streptolysins/genetics , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Mice , Pneumococcal Infections/microbiology , Brain/metabolism , Brain/microbiology , Gene Expression Regulation, Bacterial , Virulence/genetics , Operon , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism
4.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273334

ABSTRACT

Listeria pathogenicity island 1 (LIPI-1) is a genetic region containing a cluster of genes essential for virulence of the bacterial pathogen Listeria monocytogenes. Main virulence factors in LIPI-1 include long 5' untranslated regions (5'UTRs), among which is Rli51, a small RNA (sRNA) in the 5'UTR of the Zn-metalloprotease-coding mpl. So far, Rli51 function and molecular mechanisms have remained obscure. Here, we show that Rli51 exhibits a dual mechanism of regulation, functioning as a cis- and as a trans-acting sRNA. Under nutrient-rich conditions, rli51-mpl transcription is prematurely terminated, releasing a short 121-nucleotide-long sRNA. Rli51 is predicted to function as a transcription attenuator that can fold into either a terminator or a thermodynamically more stable antiterminator. We show that the sRNA Rli21/RliI binds to a single-stranded RNA loop in Rli51, which is essential to mediate premature transcription termination, suggesting that sRNA binding could stabilize the terminator fold. During intracellular infection, rli51 transcription is increased, which generates a higher abundance of the short Rli51 sRNA and allows for transcriptional read-through into mpl. Comparative intracellular bacterial transcriptomics in rli51-null mutants and the wild-type reference strain EGD-e suggests that Rli51 upregulates iron-scavenging proteins and downregulates virulence factors from LIPI-1. MS2 affinity purification confirmed that Rli51 binds transcripts of the heme-binding protein Lmo2186 and Lmo0937 in vivo. These results prove that Rli51 functions as a trans-acting sRNA in intracellular bacteria. Our research shows a growth condition-dependent mechanism of regulation for Rli51, preventing unintended mpl transcription in extracellular bacteria and regulating genes important for virulence in intracellular bacteria.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Listeria monocytogenes , RNA, Bacterial , RNA, Small Untranslated , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Genomic Islands/genetics , Transcription, Genetic , 5' Untranslated Regions , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Listeriosis/microbiology
5.
PLoS Genet ; 20(8): e1011349, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39088561

ABSTRACT

Cellular processes require precise and specific gene regulation, in which continuous mRNA degradation is a major element. The mRNA degradation mechanisms should be able to degrade a wide range of different RNA substrates with high efficiency, but should at the same time be limited, to avoid killing the cell by elimination of all cellular RNA. RNase Y is a major endoribonuclease found in most Firmicutes, including Bacillus subtilis and Staphylococcus aureus. However, the molecular interactions that direct RNase Y to cleave the correct RNA molecules at the correct position remain unknown. In this work we have identified transcripts that are homologs in S. aureus and B. subtilis, and are RNase Y targets in both bacteria. Two such transcript pairs were used as models to show a functional overlap between the S. aureus and the B. subtilis RNase Y, which highlighted the importance of the nucleotide sequence of the RNA molecule itself in the RNase Y targeting process. Cleavage efficiency is driven by the primary nucleotide sequence immediately downstream of the cleavage site and base-pairing in a secondary structure a few nucleotides downstream. Cleavage positioning is roughly localised by the downstream secondary structure and fine-tuned by the nucleotide immediately upstream of the cleavage. The identified elements were sufficient for RNase Y-dependent cleavage, since the sequence elements from one of the model transcripts were able to convert an exogenous non-target transcript into a target for RNase Y.


Subject(s)
Bacillus subtilis , Gene Expression Regulation, Bacterial , RNA Cleavage , RNA Stability , RNA, Bacterial , Staphylococcus aureus , Staphylococcus aureus/genetics , Staphylococcus aureus/enzymology , Bacillus subtilis/genetics , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Nucleic Acid Conformation , Base Sequence
6.
Proc Natl Acad Sci U S A ; 121(36): e2405510121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190361

ABSTRACT

Synonymous codons were originally viewed as interchangeable, with no phenotypic consequences. However, substantial evidence has now demonstrated that synonymous substitutions can perturb a variety of gene expression and protein homeostasis mechanisms, including translational efficiency, translational fidelity, and cotranslational folding of the encoded protein. To date, most studies of synonymous codon-derived perturbations have focused on effects within a single gene. Here, we show that synonymous codon substitutions made far within the coding sequence of Escherichia coli plasmid-encoded chloramphenicol acetyltransferase (cat) can significantly increase expression of the divergent upstream tetracycline resistance gene, tetR. In four out of nine synonymously recoded cat sequences tested, expression of the upstream tetR gene was significantly elevated due to transcription of a long antisense RNA (asRNA) originating from a transcription start site within cat. Surprisingly, transcription of this asRNA readily bypassed the native tet transcriptional repression mechanism. Even more surprisingly, accumulation of the TetR protein correlated with the level of asRNA, rather than total tetR RNA. These effects of synonymous codon substitutions on transcription and translation of a neighboring gene suggest that synonymous codon usage in bacteria may be under selection to both preserve the amino acid sequence of the encoded gene and avoid DNA sequence elements that can significantly perturb expression of neighboring genes. Avoiding such sequences may be especially important in plasmids and prokaryotic genomes, where genes and regulatory elements are often densely packed. Similar considerations may apply to the design of genetic circuits for synthetic biology applications.


Subject(s)
Chloramphenicol O-Acetyltransferase , Codon , Escherichia coli , Protein Biosynthesis , RNA, Antisense , Transcription, Genetic , RNA, Antisense/genetics , RNA, Antisense/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Codon/genetics , Gene Expression Regulation, Bacterial , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Plasmids/genetics , Plasmids/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Silent Mutation
7.
Genes Dev ; 38(13-14): 597-613, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39111824

ABSTRACT

Small RNAs base pair with and regulate mRNA translation and stability. For both bacterial small regulatory RNAs and eukaryotic microRNAs, association with partner proteins is critical for the stability and function of the regulatory RNAs. We review the mechanisms for degradation of these RNAs: displacement of the regulatory RNA from its protein partner (in bacteria) or destruction of the protein and its associated microRNAs (in eukaryotes). These mechanisms can allow specific destruction of a regulatory RNA via pairing with a decay trigger RNA or function as global off switches by disrupting the stability or function of the protein partner.


Subject(s)
MicroRNAs , RNA Stability , MicroRNAs/metabolism , MicroRNAs/genetics , RNA Stability/genetics , Animals , Humans , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , Gene Expression Regulation
8.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201508

ABSTRACT

Transfer RNA (tRNA) modifications are essential for the temperature adaptation of thermophilic and psychrophilic organisms as they control the rigidity and flexibility of transcripts. To further understand how specific tRNA modifications are adjusted to maintain functionality in response to temperature fluctuations, we investigated whether tRNA modifications represent an adaptation of bacteria to different growth temperatures (minimal, optimal, and maximal), focusing on closely related psychrophilic (P. halocryophilus and E. sibiricum), mesophilic (B. subtilis), and thermophilic (G. stearothermophilus) Bacillales. Utilizing an RNA sequencing approach combined with chemical pre-treatment of tRNA samples, we systematically profiled dihydrouridine (D), 4-thiouridine (s4U), 7-methyl-guanosine (m7G), and pseudouridine (Ψ) modifications at single-nucleotide resolution. Despite their close relationship, each bacterium exhibited a unique tRNA modification profile. Our findings revealed increased tRNA modifications in the thermophilic bacterium at its optimal growth temperature, particularly showing elevated levels of s4U8 and Ψ55 modifications compared to non-thermophilic bacteria, indicating a temperature-dependent regulation that may contribute to thermotolerance. Furthermore, we observed higher levels of D modifications in psychrophilic and mesophilic bacteria, indicating an adaptive strategy for cold environments by enhancing local flexibility in tRNAs. Our method demonstrated high effectiveness in identifying tRNA modifications compared to an established tool, highlighting its potential for precise tRNA profiling studies.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Transfer , Temperature , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Pseudouridine/metabolism
9.
Nat Commun ; 15(1): 6592, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097611

ABSTRACT

T-box riboswitches are noncoding RNA elements involved in genetic regulation of most Gram-positive bacteria. They regulate amino acid metabolism by assessing the aminoacylation status of tRNA, subsequently affecting the transcription or translation of downstream amino acid metabolism-related genes. Here we present single-molecule FRET studies of the Mycobacterium tuberculosis IleS T-box riboswitch, a paradigmatic translational T-box. Results support a two-step binding model, where the tRNA anticodon is recognized first, followed by interactions with the NCCA sequence. Furthermore, after anticodon recognition, tRNA can transiently dock into the discriminator domain even in the absence of the tRNA NCCA-discriminator interactions. Establishment of the NCCA-discriminator interactions significantly stabilizes the fully bound state. Collectively, the data suggest high conformational flexibility in translational T-box riboswitches; and supports a conformational selection model for NCCA recognition. These findings provide a kinetic framework to understand how specific RNA elements underpin the binding affinity and specificity required for gene regulation.


Subject(s)
Anticodon , Mycobacterium tuberculosis , Nucleic Acid Conformation , RNA, Bacterial , RNA, Transfer , Riboswitch , Riboswitch/genetics , RNA, Transfer/metabolism , RNA, Transfer/genetics , RNA, Transfer/chemistry , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/genetics , Anticodon/metabolism , Anticodon/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/chemistry , Fluorescence Resonance Energy Transfer , Protein Biosynthesis , Gene Expression Regulation, Bacterial , Kinetics
10.
Nucleic Acids Res ; 52(16): 10017-10028, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39126322

ABSTRACT

Vital organismal processes, including development, differentiation and adaptation, involve altered gene expression. Although expression is frequently controlled at the transcriptional stage, various regulation mechanisms operate at downstream levels. Here, we leverage the photoreceptor NmPAL to optogenetically induce RNA refolding and the translation of bacterial mRNAs. Blue-light-triggered NmPAL binding disrupts a cis-repressed mRNA state, thereby relieves obstruction of translation initiation, and upregulates gene expression. Iterative probing and optimization of the circuit, dubbed riboptoregulator, enhanced induction to 30-fold. Given action at the mRNA level, the riboptoregulator can differentially regulate individual structural genes within polycistronic operons. Moreover, it is orthogonal to and can be wed with other gene-regulatory circuits for nuanced and more stringent gene-expression control. We thus advance the pAurora2 circuit that combines transcriptional and translational mechanisms to optogenetically increase bacterial gene expression by >1000-fold. The riboptoregulator strategy stands to upgrade numerous regulatory circuits and widely applies to expression control in microbial biotechnology, synthetic biology and materials science.


Subject(s)
Gene Expression Regulation, Bacterial , Light , RNA, Messenger , RNA, Messenger/genetics , RNA, Messenger/metabolism , Optogenetics/methods , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Operon/genetics , Protein Biosynthesis , Photoreceptors, Microbial/genetics , Photoreceptors, Microbial/metabolism , RNA Folding
11.
Nat Commun ; 15(1): 6946, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138169

ABSTRACT

Hypervirulent Klebsiella pneumoniae (HvKP) is an emerging bacterial pathogen causing invasive infection in immune-competent humans. The hypervirulence is strongly linked to the overproduction of hypermucoviscous capsule, but the underlying regulatory mechanisms of hypermucoviscosity (HMV) have been elusive, especially at the post-transcriptional level mediated by small noncoding RNAs (sRNAs). Using a recently developed RNA interactome profiling approach iRIL-seq, we interrogate the Hfq-associated sRNA regulatory network and establish an intracellular RNA-RNA interactome in HvKP. Our data reveal numerous interactions between sRNAs and HMV-related mRNAs, and identify a plethora of sRNAs that repress or promote HMV. One of the strongest HMV repressors is ArcZ, which is activated by the catabolite regulator CRP and targets many HMV-related genes including mlaA and fbp. We discover that MlaA and its function in phospholipid transport is crucial for capsule retention and HMV, inactivation of which abolishes Klebsiella virulence in mice. ArcZ overexpression drastically reduces bacterial burden in mice and reduces HMV in multiple hypervirulent and carbapenem-resistant clinical isolates, indicating ArcZ is a potent RNA inhibitor of bacterial pneumonia with therapeutic potential. Our work unravels a novel CRP-ArcZ-MlaA regulatory circuit of HMV and provides mechanistic insights into the posttranscriptional virulence control in a superbug of global concern.


Subject(s)
Bacterial Capsules , Bacterial Proteins , Gene Expression Regulation, Bacterial , Klebsiella Infections , Klebsiella pneumoniae , RNA, Bacterial , RNA, Small Untranslated , Klebsiella pneumoniae/pathogenicity , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Animals , Virulence/genetics , Mice , Klebsiella Infections/microbiology , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Humans , Female , Host Factor 1 Protein/metabolism , Host Factor 1 Protein/genetics
12.
Sci Rep ; 14(1): 19505, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174714

ABSTRACT

Surface-enhanced Raman spectroscopy (SERS) is widely utilized in bacterial analyses, with the dominant SERS peaks attributed to purine metabolites released during sample preparation. Although adenosine triphosphate (ATP) and nucleic acids are potential molecular origins of these metabolites, research on their exact contributions remains limited. This study explored purine metabolite release from E. coli and RNA integrity following various sample preparation methods. Standard water washing generated dominant SERS signals within 10 s, a duration shorter than the anticipated RNA half-lives under starvation. Evaluating RNA integrity indicated that the most abundant ribosomal RNA species remained intact for hours post-washing, whereas messenger RNA and transfer RNA species degraded gradually. This suggests that bacterial SERS signatures observed after the typical washing step could originate from only a small fraction of endogenous purine-containing molecules. In contrast, acid depurination led to degradation of most RNA species, releasing about 40 times more purine derivatives than water washing. Mild heating also instigated the RNA degradation and released more purine derivatives than water washing. Notably, differences were also evident in the dominant SERS signals following these treatments. This work provides insights into SERS-based studies of purine metabolites released by bacteria and future development of methodologies.


Subject(s)
Escherichia coli , RNA, Bacterial , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , Purines/metabolism , Adenosine Triphosphate/metabolism
13.
J Bacteriol ; 206(9): e0025124, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39171914

ABSTRACT

Escherichia coli possesses four initiator tRNA (i-tRNA) genes, three of which are present together as metZWV and the fourth one as metY. In E. coli B, all four genes (metZWV and metY) encode i-tRNAfMet1, in which the G at position 46 is modified to m7G46 by TrmB (m7G methyltransferase). However, in E. coli K, because of a single-nucleotide polymorphism, metY encodes a variant, i-tRNAfMet2, having an A in place of m7G46. We generated E. coli strains to explore the importance of this polymorphism in i-tRNAs. The strains were sustained either on metYA46 (metY of E. coli K origin encoding i-tRNAfMet2) or its derivative metYG46 (encoding i-tRNAfMet1) in single (chromosomal) or plasmid-borne copies. We show that the strains sustained on i-tRNAfMet1 have a growth fitness advantage over those sustained on i-tRNAfMet2. The growth fitness advantages are more pronounced for the strains sustained on i-tRNAfMet1 in nutrient-rich media than in nutrient-poor media. The growth fitness of the strains correlates well with the relative stabilities of the i-tRNAs in vivo. Furthermore, the atomistic molecular dynamics simulations support the higher stability of i-tRNAfMet1 than that of i-tRNAfMet2. The stability of i-tRNAfMet1 remains unaffected upon the deletion of TrmB. These studies highlight how metYG46 and metYA46 alleles might influence the growth fitness of E. coli under certain nutrient-limiting conditions. IMPORTANCE: Escherichia coli harbors four initiator tRNA (i-tRNA) genes: three of these at metZWV and the fourth one at metY loci. In E. coli B, all four genes encode i-tRNAfMet1. In E. coli K, because of a single-nucleotide polymorphism, metY encodes a variant, i-tRNAfMet2, having an A in place of G at position 46 of i-tRNA sequence in metY. We show that G46 confers stability to i-tRNAfMet1. The strains sustained on i-tRNAfMet1 have a growth fitness advantage over those sustained on i-tRNAfMet2. Strains harboring metYG46 (B mimic) or metYA46 (K mimic) show that while in the nutrient-rich media, the K mimic is outcompeted rapidly; in the nutrient-poor medium, the K mimic is outcompeted less rapidly.


Subject(s)
Escherichia coli , RNA, Transfer, Met , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Bacterial/metabolism
14.
J Bacteriol ; 206(9): e0000424, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39171913

ABSTRACT

Streptococcus pneumoniae (pneumococcus) causes a wide range of important human infectious diseases, including pneumonia, pneumonia-derived sepsis, otitis media, and meningitis. Pneumococcus produces numerous secreted proteins that are critical for normal physiology and pathogenesis. The membrane targeting and translocation of these secreted proteins are partly mediated by the signal recognition particle (SRP) complex, which consists of 4.5S small cytoplasmic RNA (ScRNA), and the Ffh, and FtsY proteins. Here, we report that pneumococcal ∆scRNA, ∆ffh, and ∆ftsY mutants were significantly impaired in competence induction, competence pili production, exogenous DNA uptake, and genetic transformation. Also, the ∆scRNA mutant was significantly attenuated in the mouse models of bacteremia and pneumonia. Interestingly, unlike the ∆scRNA, both ∆ffh and ∆ftsY mutants had growth defects on Todd-Hewitt Agar, which were alleviated by the provision of free amino acids or serum. Differences in nutritional requirements between ∆ffh and ∆ftsY vs ∆scRNA suggest that Ffh and FtsY may be partially functional in the absence of ScRNA. Finally, the insertase YidC2, which could functionally rescue some SRP mutations in other streptococcal species, was not essential for pneumococcal genetic transformation. Collectively, these results indicate that ScRNA is crucial for the successful development of genetic competence and virulence in pneumococcus. IMPORTANCE: Streptococcus pneumoniae (pneumococcus) causes multiple important infectious diseases in humans. The signal recognition particle (SRP) complex, which comprised 4.5S small cytoplasmic RNA (ScRNA), and the Ffh and FtsY proteins, mediates membrane targeting and translocation of secreted proteins in all organisms. However, the role of SRP and ScRNA has not been characterized during the induction of the competence system for genetic transformation and virulence in pneumococcus. By using a combination of genetic, biochemical, proteomic, and imaging approaches, we demonstrated that the SRP complex plays a significant role in membrane targeting of competence system-regulated effectors important for genetic transformation, virulence during bacteremia and pneumonia infections, and nutritional acquisition.


Subject(s)
Bacterial Proteins , Streptococcus pneumoniae , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Streptococcus pneumoniae/metabolism , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence , Animals , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Pneumococcal Infections/microbiology , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , DNA Transformation Competence , Bacteremia/microbiology
15.
Proc Natl Acad Sci U S A ; 121(32): e2401981121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39078675

ABSTRACT

Dihydrouridine (D), a prevalent and evolutionarily conserved base in the transcriptome, primarily resides in tRNAs and, to a lesser extent, in mRNAs. Notably, this modification is found at position 2449 in the Escherichia coli 23S rRNA, strategically positioned near the ribosome's peptidyl transferase site. Despite the prior identification, in E. coli genome, of three dihydrouridine synthases (DUS), a set of NADPH and FMN-dependent enzymes known for introducing D in tRNAs and mRNAs, characterization of the enzyme responsible for D2449 deposition has remained elusive. This study introduces a rapid method for detecting D in rRNA, involving reverse transcriptase-blockage at the rhodamine-labeled D2449 site, followed by PCR amplification (RhoRT-PCR). Through analysis of rRNA from diverse E. coli strains, harboring chromosomal or single-gene deletions, we pinpoint the yhiN gene as the ribosomal dihydrouridine synthase, now designated as RdsA. Biochemical characterizations uncovered RdsA as a unique class of flavoenzymes, dependent on FAD and NADH, with a complex structural topology. In vitro assays demonstrated that RdsA dihydrouridylates a short rRNA transcript mimicking the local structure of the peptidyl transferase site. This suggests an early introduction of this modification before ribosome assembly. Phylogenetic studies unveiled the widespread distribution of the yhiN gene in the bacterial kingdom, emphasizing the conservation of rRNA dihydrouridylation. In a broader context, these findings underscore nature's preference for utilizing reduced flavin in the reduction of uridines and their derivatives.


Subject(s)
Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Ribosomal, 23S/metabolism , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/chemistry , Uridine/analogs & derivatives , Uridine/metabolism , Uridine/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/chemistry
16.
RNA ; 30(10): 1328-1344, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38981655

ABSTRACT

T-box riboswitches are widespread bacterial regulatory noncoding RNAs that directly interact with tRNAs and switch conformations to regulate the transcription or translation of genes related to amino acid metabolism. Recent studies in Bacilli have revealed the core mechanisms of T-boxes that enable multivalent, specific recognition of both the identity and aminoacylation status of the tRNA substrates. However, in-depth knowledge on a vast number of T-boxes in other bacterial species remains scarce, although a remarkable structural diversity, particularly among pathogens, is apparent. In the present study, analysis of T-boxes that control the transcription of glycyl-tRNA synthetases from four prominent human pathogens revealed significant structural idiosyncrasies. Nonetheless, these diverse T-boxes maintain functional T-box:tRNAGly interactions both in vitro and in vivo. Probing analysis not only validated recent structural observations, but also expanded our knowledge on the substantial diversities among T-boxes and suggest interesting distinctions from the canonical Bacilli T-boxes. Surprisingly, some glycyl T-boxes seem to redirect the T-box trajectory in the absence of recognizable K-turns or contain Stem II modules that are generally absent in glycyl T-boxes. These results consolidate the notion of a lineage-specific diversification and elaboration of the T-box mechanism and corroborate the potential of T-boxes as promising species-specific RNA targets for next-generation antibacterial compounds.


Subject(s)
Nucleic Acid Conformation , RNA, Bacterial , Riboswitch , Riboswitch/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/chemistry , Gene Expression Regulation, Bacterial , Glycine-tRNA Ligase/genetics , Glycine-tRNA Ligase/metabolism , Glycine-tRNA Ligase/chemistry , RNA, Transfer, Gly/metabolism , RNA, Transfer, Gly/genetics , RNA, Transfer, Gly/chemistry , Base Sequence , Bacteria/genetics , Bacteria/metabolism , Humans , RNA, Transfer/metabolism , RNA, Transfer/genetics , RNA, Transfer/chemistry
17.
RNA ; 30(10): 1264-1276, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39043438

ABSTRACT

Ribosomal RNAs are transcribed as part of larger precursor molecules. In Escherichia coli, complementary RNA segments flank each rRNA and form long leader-trailer (LT) helices, which are crucial for subunit biogenesis in the cell. A previous study of 15 representative species suggested that most but not all prokaryotes contain LT helices. Here, we use a combination of in silico folding and covariation methods to identify and characterize LT helices in 4464 bacterial and 260 archaeal organisms. Our results suggest that LT helices are present in all phyla, including Deinococcota, which had previously been suspected to lack LT helices. In very few organisms, our pipeline failed to detect LT helices for both 16S and 23S rRNA. However, a closer case-by-case look revealed that LT helices are indeed present but escaped initial detection. Over 3600 secondary structure models, many well supported by nucleotide covariation, were generated. These structures show a high degree of diversity. Yet, all exhibit extensive base-pairing between the leader and trailer strands, in line with a common and essential function.


Subject(s)
Archaea , Nucleic Acid Conformation , RNA, Archaeal , RNA, Bacterial , RNA, Archaeal/genetics , RNA, Archaeal/chemistry , RNA, Archaeal/metabolism , Archaea/genetics , RNA, Bacterial/genetics , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Bacteria/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Precursors/chemistry , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/chemistry , RNA, Ribosomal, 23S/metabolism , Base Sequence , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/chemistry , Base Pairing
18.
Nucleic Acids Res ; 52(15): 8746-8762, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39011882

ABSTRACT

Bacterial gene expression is a complex process involving extensive regulatory mechanisms. Along with growing interests in this field, Nanopore Direct RNA Sequencing (DRS) provides a promising platform for rapid and comprehensive characterization of bacterial RNA biology. However, the DRS of bacterial RNA is currently deficient in the yield of mRNA-mapping reads and has yet to be exploited for transcriptome-wide RNA modification mapping. Here, we showed that pre-processing of bacterial total RNA (size selection followed by ribosomal RNA depletion and polyadenylation) guaranteed high throughputs of sequencing data and considerably increased the amount of mRNA reads. This way, complex transcriptome architectures were reconstructed for Escherichia coli and Staphylococcus aureus and extended the boundaries of 225 known E. coli operons and 89 defined S. aureus operons. Utilizing unmodified in vitro-transcribed (IVT) RNA libraries as a negative control, several Nanopore-based computational tools globally detected putative modification sites in the E. coli and S. aureus transcriptomes. Combined with Next-Generation Sequencing-based N6-methyladenosine (m6A) detection methods, 75 high-confidence m6A candidates were identified in the E. coli protein-coding transcripts, while none were detected in S. aureus. Altogether, we demonstrated the potential of Nanopore DRS in systematic and convenient transcriptome and epitranscriptome analysis.


Subject(s)
Escherichia coli , Nanopore Sequencing , RNA, Bacterial , Sequence Analysis, RNA , Staphylococcus aureus , Transcriptome , Escherichia coli/genetics , Escherichia coli/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Transcriptome/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Sequence Analysis, RNA/methods , Nanopore Sequencing/methods , High-Throughput Nucleotide Sequencing/methods , Operon/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/genetics , Nanopores , Gene Expression Regulation, Bacterial , Gene Expression Profiling/methods
19.
Nucleic Acids Res ; 52(15): 8947-8966, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39036964

ABSTRACT

Small RNAs (sRNAs) are major regulators of gene expression in bacteria, exerting their regulation primarily via base pairing with their target transcripts and modulating translation. Accumulating evidence suggest that sRNAs can also affect the stability of their target transcripts by altering their accessibility to endoribonucleases. Yet, the effects of sRNAs on transcript stability and the mechanisms underlying them have not been studied in wide scale. Here we employ large-scale RNA-seq-based methodologies in the model bacterium Escherichia coli to quantitatively study the functional interaction between a sRNA and an endoribonuclease in regulating gene expression, using the well-established sRNA, GcvB, and the major endoribonuclease, RNase E. Studying single and double mutants of gcvB and rne and analysing their RNA-seq results by the Double Mutant Cycle approach, we infer distinct modes of the interplay between GcvB and RNase E. Transcriptome-wide mapping of RNase E cleavage sites provides further support to the results of the RNA-seq analysis, identifying cleavage sites in targets in which the functional interaction between GcvB and RNase E is evident. Together, our results indicate that the most dominant mode of GcvB-RNase E functional interaction is GcvB enhancement of RNase E cleavage, which varies in its magnitude between different targets.


Subject(s)
Endoribonucleases , Escherichia coli Proteins , Escherichia coli , RNA, Bacterial , RNA, Small Untranslated , Endoribonucleases/metabolism , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Mutation , RNA Stability/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA, Small Untranslated/metabolism , RNA, Small Untranslated/genetics
20.
Curr Opin Struct Biol ; 88: 102893, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39067113

ABSTRACT

Riboswitches are specialized RNA structures that orchestrate gene expression in response to sensing specific metabolite or ion ligands, mostly in bacteria. Upon ligand binding, these conformationally dynamic RNA motifs undergo structural changes that control critical gene expression processes such as transcription termination and translation initiation, thereby enabling cellular homeostasis and adaptation. Because RNA folds rapidly and co-transcriptionally, riboswitches make use of the low complexity of RNA sequences to adopt alternative, transient conformations on the heels of the transcribing RNA polymerase (RNAP), resulting in kinetic partitioning that defines the regulatory outcome. This review summarizes single molecule microscopy evidence that has begun to unveil a sophisticated network of dynamic, kinetically balanced interactions between riboswitch architecture and the gene expression machinery that, together, integrate diverse cellular signals.


Subject(s)
Gene Expression Regulation, Bacterial , Riboswitch , Ligands , Single Molecule Imaging , Nucleic Acid Conformation , Bacteria/genetics , Bacteria/metabolism , Signal Transduction , RNA, Bacterial/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL