Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842944

ABSTRACT

The ompD transcript, encoding an outer membrane porin in Salmonella, harbors a controlling element in its coding region that base-pairs imperfectly with a 'seed' region of the small regulatory RNA (sRNA) MicC. When tagged with the sRNA, the ompD mRNA is cleaved downstream of the pairing site by the conserved endoribonuclease RNase E, leading to transcript destruction. We observe that the sRNA-induced cleavage site is accessible to RNase E in vitro upon recruitment of ompD into the 30S translation pre-initiation complex (PIC) in the presence of the degradosome components. Evaluation of substrate accessibility suggests that the paused 30S PIC presents the mRNA for targeted recognition and degradation. Ribonuclease activity on PIC-bound ompD is critically dependent on the recruitment of RNase E into the multi-enzyme RNA degradosome, and our data suggest a process of substrate capture and handover to catalytic sites within the degradosome, in which sequential steps of seed matching and duplex remodelling contribute to cleavage efficiency. Our findings support a putative mechanism of surveillance at translation that potentially terminates gene expression efficiently and rapidly in response to signals provided by regulatory RNA.

2.
RNA Biol ; 19(1): 419-436, 2022.
Article in English | MEDLINE | ID: mdl-35438047

ABSTRACT

In all domains of life, RNA chaperones safeguard and guide the fate of the cellular RNA pool. RNA chaperones comprise structurally diverse proteins that ensure proper folding, stability, and ribonuclease resistance of RNA, and they support regulatory activities mediated by RNA. RNA chaperones constitute a topologically diverse group of proteins that often present an unstructured region and bind RNA with limited nucleotide sequence preferences. In bacteria, three main proteins - Hfq, ProQ, and CsrA - have been shown to regulate numerous complex processes, including bacterial growth, stress response and virulence. Hfq and ProQ have well-studied activities as global chaperones with pleiotropic impact, while CsrA has a chaperone-like role with more defined riboregulatory function. Here, we describe relevant novel insights into their common features, including RNA binding properties, unstructured domains, and interplay with other proteins important to RNA metabolism.


Subject(s)
RNA, Bacterial , RNA, Small Untranslated , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , RNA-Binding Proteins/metabolism
3.
RNA ; 27(11): 1339-1352, 2021 11.
Article in English | MEDLINE | ID: mdl-34341070

ABSTRACT

The conserved endoribonuclease RNase E dominates the dynamic landscape of RNA metabolism and underpins control mediated by small regulatory RNAs in diverse bacterial species. We explored the enzyme's hydrolytic mechanism, allosteric activation, and interplay with partner proteins in the multicomponent RNA degradosome assembly of Escherichia coli. RNase E cleaves single-stranded RNA with preference to attack the phosphate located at the 5' nucleotide preceding uracil, and we corroborate key interactions that select that base. Unexpectedly, RNase E activity is impeded strongly when the recognized uracil is isomerized to 5-ribosyluracil (pseudouridine), from which we infer the detailed geometry of the hydrolytic attack process. Kinetics analyses support models for recognition of secondary structure in substrates by RNase E and for allosteric autoregulation. The catalytic power of the enzyme is boosted when it is assembled into the multienzyme RNA degradosome, most likely as a consequence of substrate capture and presentation. Our results rationalize the origins of substrate preferences of RNase E and illuminate its catalytic mechanism, supporting the roles of allosteric domain closure and cooperation with other components of the RNA degradosome complex.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , Pseudouridine/metabolism , RNA Helicases/metabolism , RNA, Bacterial/metabolism , Binding Sites , Endoribonucleases/chemistry , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Multienzyme Complexes/genetics , Nucleic Acid Conformation , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Conformation , RNA Helicases/genetics , RNA, Bacterial/genetics
4.
Mol Cell ; 81(14): 2901-2913.e5, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34157309

ABSTRACT

Polynucleotide phosphorylase (PNPase) is an ancient exoribonuclease conserved in the course of evolution and is found in species as diverse as bacteria and humans. Paradoxically, Escherichia coli PNPase can act not only as an RNA degrading enzyme but also by an unknown mechanism as a chaperone for small regulatory RNAs (sRNAs), with pleiotropic consequences for gene regulation. We present structures of the ternary assembly formed by PNPase, the RNA chaperone Hfq, and sRNA and show that this complex boosts sRNA stability in vitro. Comparison of structures for PNPase in RNA carrier and degradation modes reveals how the RNA is rerouted away from the active site through interactions with Hfq and the KH and S1 domains. Together, these data explain how PNPase is repurposed to protect sRNAs from cellular ribonucleases such as RNase E and could aid RNA presentation to facilitate regulatory actions on target genes.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Host Factor 1 Protein/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA, Bacterial/genetics , Catalytic Domain/genetics , Endoribonucleases/genetics , Exoribonucleases/genetics , Gene Expression Regulation, Bacterial/genetics , Molecular Chaperones/genetics , RNA Stability/genetics , RNA, Small Untranslated/genetics
5.
J Virol ; 95(14): e0066321, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33963053

ABSTRACT

RNA structural elements occur in numerous single-stranded positive-sense RNA viruses. The stem-loop 2 motif (s2m) is one such element with an unusually high degree of sequence conservation, being found in the 3' untranslated region (UTR) in the genomes of many astroviruses, some picornaviruses and noroviruses, and a variety of coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. The evolutionary conservation and its occurrence in all viral subgenomic transcripts imply a key role for s2m in the viral infection cycle. Our findings indicate that the element, while stably folded, can nonetheless be invaded and remodeled spontaneously by antisense oligonucleotides (ASOs) that initiate pairing in exposed loops and trigger efficient sequence-specific RNA cleavage in reporter assays. ASOs also act to inhibit replication in an astrovirus replicon model system in a sequence-specific, dose-dependent manner and inhibit SARS-CoV-2 replication in cell culture. Our results thus permit us to suggest that the s2m element is readily targeted by ASOs, which show promise as antiviral agents. IMPORTANCE The highly conserved stem-loop 2 motif (s2m) is found in the genomes of many RNA viruses, including SARS-CoV-2. Our findings indicate that the s2m element can be targeted by antisense oligonucleotides. The antiviral potential of this element represents a promising start for further research into targeting conserved elements in RNA viruses.


Subject(s)
COVID-19 , Genome, Viral , Nucleotide Motifs , RNA Folding , RNA, Viral , SARS-CoV-2/physiology , Virus Replication , Animals , COVID-19/genetics , COVID-19/metabolism , Chlorocebus aethiops , HEK293 Cells , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Vero Cells
6.
Curr Opin Struct Biol ; 61: 59-70, 2020 04.
Article in English | MEDLINE | ID: mdl-31869589

ABSTRACT

Through the activities of various multi-component assemblies, protein-coding transcripts can be chaperoned toward protein synthesis or nudged into a funnel of rapid destruction. The capacity of these machine-like assemblies to tune RNA lifetime underpins the harmony of gene expression in all cells. Some of the molecular machines that mediate transcript turnover also contribute to on-the-fly surveillance of aberrant mRNAs and non-coding RNAs. How these dynamic assemblies distinguish functional RNAs from those that must be degraded is an intriguing puzzle for understanding the regulation of gene expression and dysfunction associated with disease. Recent data illuminate what the machines look like, and how they find, recognise and operate on transcripts to sculpt the dynamic regulatory landscape. This review captures current structural and mechanistic insights into the key enzymes and their effector assemblies that contribute to the fate-determining decision points for RNA in post-transcriptional control of genetic information.


Subject(s)
Gene Expression Regulation , Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , Eukaryotic Cells/physiology , Gene Expression Regulation, Bacterial , Humans , Mitochondria/genetics , Mitochondria/metabolism , RNA/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Structure-Activity Relationship
7.
Nucleic Acids Res ; 47(3): 1482-1492, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30462307

ABSTRACT

Increasing numbers of 3'UTR-derived small, regulatory RNAs (sRNAs) are being discovered in bacteria, most generated by cleavage from longer transcripts. The enzyme required for these cleavages has been reported to be RNase E, the major endoribonuclease in enterica bacteria. Previous studies investigating RNase E have come to a range of different conclusions regarding the determinants for RNase E processing. To better understand the sequence and structure determinants for the precise processing of a 3' UTR-derived sRNA, we examined the cleavage of multiple mutant and chimeric derivatives of the 3' UTR-derived MicL sRNA in vivo and in vitro. Our results revealed that tandem stem-loops 3' to the cleavage site define optimal, correctly-positioned cleavage of MicL and probably other sRNAs. Moreover, our assays of MicL, ArcZ and CpxQ showed that sRNAs exhibit differential sensitivity to RNase E, likely a consequence of a hierarchy of sRNA features recognized by the endonuclease.


Subject(s)
Endoribonucleases/chemistry , Escherichia coli/genetics , RNA, Small Untranslated/genetics , 3' Untranslated Regions/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Small Untranslated/chemistry
8.
Mol Cell ; 72(2): 275-285.e4, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30270108

ABSTRACT

The endoribonuclease RNase E is a principal factor in RNA turnover and processing that helps to exercise fine control of gene expression in bacteria. While its catalytic activity can be strongly influenced by the chemical identity of the 5' end of RNA substrates, the enzyme can also cleave numerous substrates irrespective of the chemistry of their 5' ends through a mechanism that has remained largely unexplained. We report structural and functional data illuminating details of both operational modes. Our crystal structure of RNase E in complex with the sRNA RprA reveals a duplex recognition site that saddles an inter-protomer surface to help present substrates for cleavage. Our data also reveal an autoinhibitory pocket that modulates the overall activity of the ribonuclease. Taking these findings together, we propose how RNase E uses versatile modes of RNA recognition to achieve optimal activity and specificity.


Subject(s)
Endoribonucleases/genetics , Substrate Specificity/genetics , Amino Acid Sequence , Catalysis , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Protein Subunits/genetics , RNA/genetics , RNA, Bacterial/genetics , Sequence Alignment
9.
Microbiol Spectr ; 6(2)2018 04.
Article in English | MEDLINE | ID: mdl-29676248

ABSTRACT

The bacterial endoribonuclease RNase E occupies a pivotal position in the control of gene expression, as its actions either commit transcripts to an irreversible fate of rapid destruction or unveil their hidden functions through specific processing. Moreover, the enzyme contributes to quality control of rRNAs. The activity of RNase E can be directed and modulated by signals provided through regulatory RNAs that guide the enzyme to specific transcripts that are to be silenced. Early in its evolutionary history, RNase E acquired a natively unfolded appendage that recruits accessory proteins and RNA. These accessory factors facilitate the activity of RNase E and include helicases that remodel RNA and RNA-protein complexes, and polynucleotide phosphorylase, a relative of the archaeal and eukaryotic exosomes. RNase E also associates with enzymes from central metabolism, such as enolase and aconitase. RNase E-based complexes are diverse in composition, but generally bear mechanistic parallels with eukaryotic machinery involved in RNA-induced gene regulation and transcript quality control. That these similar processes arose independently underscores the universality of RNA-based regulation in life. Here we provide a synopsis and perspective of the contributions made by RNase E to sustain robust gene regulation with speed and accuracy.


Subject(s)
Bacteria/enzymology , Endoribonucleases/metabolism , Evolution, Molecular , Gene Expression Regulation, Bacterial , RNA, Bacterial/metabolism , Aconitate Hydratase/metabolism , Archaea/enzymology , Bacteria/genetics , Eukaryotic Cells/enzymology , Exosomes , Phosphopyruvate Hydratase/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , RNA Processing, Post-Transcriptional
10.
Nucleic Acids Res ; 46(1): 387-402, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29136196

ABSTRACT

The RNA degradosome is a multi-enzyme assembly that plays a central role in the RNA metabolism of Escherichia coli and numerous other bacterial species including pathogens. At the core of the assembly is the endoribonuclease RNase E, one of the largest E. coli proteins and also one that bears the greatest region predicted to be natively unstructured. This extensive unstructured region, situated in the C-terminal half of RNase E, is punctuated with conserved short linear motifs that recruit partner proteins, direct RNA interactions, and enable association with the cytoplasmic membrane. We have structurally characterized a subassembly of the degradosome-comprising a 248-residue segment of the natively unstructured part of RNase E, the DEAD-box helicase RhlB and the glycolytic enzyme enolase, and provide evidence that it serves as a flexible recognition centre that can co-recruit small regulatory RNA and the RNA chaperone Hfq. Our results support a model in which the degradosome captures substrates and regulatory RNAs through the recognition centre, facilitates pairing to cognate transcripts and presents the target to the ribonuclease active sites of the greater assembly for cooperative degradation or processing.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/metabolism , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , RNA, Bacterial/metabolism , Binding Sites/genetics , Crystallography, X-Ray , Endoribonucleases/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , Models, Molecular , Multienzyme Complexes/genetics , Nucleic Acid Conformation , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Binding , Protein Domains , RNA Helicases/genetics , RNA, Bacterial/chemistry , RNA, Bacterial/genetics
11.
Elife ; 52016 07 22.
Article in English | MEDLINE | ID: mdl-27447594

ABSTRACT

In all domains of life, the catalysed degradation of RNA facilitates rapid adaptation to changing environmental conditions, while destruction of foreign RNA is an important mechanism to prevent host infection. We have identified a virus-encoded protein termed gp37/Dip, which directly binds and inhibits the RNA degradation machinery of its bacterial host. Encoded by giant phage фKZ, this protein associates with two RNA binding sites of the RNase E component of the Pseudomonas aeruginosa RNA degradosome, occluding them from substrates and resulting in effective inhibition of RNA degradation and processing. The 2.2 Šcrystal structure reveals that this novel homo-dimeric protein has no identifiable structural homologues. Our biochemical data indicate that acidic patches on the convex outer surface bind RNase E. Through the activity of Dip, фKZ has evolved a unique mechanism to down regulate a key metabolic process of its host to allow accumulation of viral RNA in infected cells.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Host-Parasite Interactions , Multienzyme Complexes/antagonists & inhibitors , Polyribonucleotide Nucleotidyltransferase/antagonists & inhibitors , Pseudomonas Phages/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/virology , RNA Helicases/antagonists & inhibitors , Viral Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Viral Proteins/chemistry
12.
RNA ; 22(3): 360-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26759452

ABSTRACT

In all bacterial species examined thus far, small regulatory RNAs (sRNAs) contribute to intricate patterns of dynamic genetic regulation. Many of the actions of these nucleic acids are mediated by well-characterized chaperones such as the Hfq protein, but genetic screens have also recently identified the 3'-to-5' exoribonuclease polynucleotide phosphorylase (PNPase) as an unexpected stabilizer and facilitator of sRNAs in vivo. To understand how a ribonuclease might mediate these effects, we tested the interactions of PNPase with sRNAs and found that the enzyme can readily degrade these nucleic acids in vitro but, nonetheless, copurifies from cell extracts with the same sRNAs without discernible degradation or modification to their 3' ends, suggesting that the associated RNA is protected against the destructive activity of the ribonuclease. In vitro, PNPase, Hfq, and sRNA can form a ternary complex in which the ribonuclease plays a nondestructive, structural role. Such ternary complexes might be formed transiently in vivo, but could help to stabilize particular sRNAs and remodel their population on Hfq. Taken together, our results indicate that PNPase can be programmed to act on RNA in either destructive or stabilizing modes in vivo and may form complex, protective ribonucleoprotein assemblies that shape the landscape of sRNAs available for action.


Subject(s)
Polyribonucleotide Nucleotidyltransferase/metabolism , RNA/metabolism , Hydrolysis , Protein Binding
13.
Elife ; 32014 Dec 31.
Article in English | MEDLINE | ID: mdl-25551292

ABSTRACT

Bacterial small RNAs (sRNAs) are key elements of regulatory networks that modulate gene expression. The sRNA RydC of Salmonella sp. and Escherichia coli is an example of this class of riboregulators. Like many other sRNAs, RydC bears a 'seed' region that recognises specific transcripts through base-pairing, and its activities are facilitated by the RNA chaperone Hfq. The crystal structure of RydC in complex with E. coli Hfq at a 3.48 Å resolution illuminates how the protein interacts with and presents the sRNA for target recognition. Consolidating the protein-RNA complex is a host of distributed interactions mediated by the natively unstructured termini of Hfq. Based on the structure and other data, we propose a model for a dynamic effector complex comprising Hfq, small RNA, and the cognate mRNA target.


Subject(s)
Host Factor 1 Protein/metabolism , RNA, Bacterial/metabolism , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Bacterial/chemistry , Salmonella/genetics , Salmonella/metabolism
14.
RNA Biol ; 10(4): 627-35, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23580162

ABSTRACT

RNA enables the material interpretation of genetic information through time and in space. The creation, destruction and activity of RNA must be well controlled and tightly synchronized with numerous cellular processes. We discuss here the pathways and mechanism of bacterial RNA turnover, and describe how RNA itself modulates these processes as part of decision-making networks. The central roles of RNA decay and other aspects of RNA metabolism in cellular control are also suggested by their vulnerability to sabotage by phages; nonetheless, RNA can be used in defense against phage infection, and these processes are described here. Salient aspects of RNA turnover are drawn together to suggest how it could affect complex effects such as phenotypic diversity in populations and responses that persist for multiple generations.


Subject(s)
Bacillus subtilis/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Host Factor 1 Protein/chemistry , RNA Stability , RNA, Bacterial/metabolism , RNA-Binding Proteins/chemistry , Bacteriophages/metabolism , DEAD-box RNA Helicases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Evolution, Molecular , Exoribonucleases/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Polyadenylation , RNA, Small Untranslated/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
15.
Biochim Biophys Acta ; 1829(6-7): 514-22, 2013.
Article in English | MEDLINE | ID: mdl-23459248

ABSTRACT

Bacterial transcripts each have a characteristic half-life, suggesting that the processes of RNA degradation work in an active and selective manner. Moreover, the processes are well controlled, thereby ensuring that degradation is orderly and coordinated. Throughout much of the bacterial kingdom, RNA degradation processes originate through the actions of assemblies of key RNA enzymes, known as RNA degradosomes. Neither conserved in composition, nor unified by common evolutionary ancestry, RNA degradosomes nonetheless can be found in divergent bacterial lineages, implicating a common requirement for the co-localisation of RNA metabolic activities. We describe how the cooperation of components in the representative degradosome of Escherichia coli may enable controlled access to transcripts, so that they have defined and programmable lifetimes. We also discuss how this cooperation contributes to precursor processing and to the riboregulation of intricate post-transcriptional networks in the control of gene expression. The E. coli degradosome interacts with the cytoplasmic membrane, and we discuss how this interaction may spatially organise the assembly and contribute to subunit cooperation and substrate capture. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Subject(s)
Endoribonucleases/genetics , Multienzyme Complexes , Polyribonucleotide Nucleotidyltransferase , RNA Helicases , RNA Stability , RNA, Bacterial/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Endoribonucleases/chemistry , Escherichia coli/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Polyribonucleotide Nucleotidyltransferase/chemistry , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Conformation , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA, Bacterial/chemistry
16.
Mol Cell ; 47(6): 943-53, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22902561

ABSTRACT

Numerous small non-coding RNAs (sRNAs) in bacteria modulate rates of translation initiation and degradation of target mRNAs, which they recognize through base-pairing facilitated by the RNA chaperone Hfq. Recent evidence indicates that the ternary complex of Hfq, sRNA and mRNA guides endoribonuclease RNase E to initiate turnover of both the RNAs. We show that a sRNA not only guides RNase E to a defined site in a target RNA, but also allosterically activates the enzyme by presenting a monophosphate group at the 5'-end of the cognate-pairing "seed." Moreover, in the absence of the target the 5'-monophosphate makes the sRNA seed region vulnerable to an attack by RNase E against which Hfq confers no protection. These results suggest that the chemical signature and pairing status of the sRNA seed region may help to both 'proofread' recognition and activate mRNA cleavage, as part of a dynamic process involving cooperation of RNA, Hfq and RNase E.


Subject(s)
Endoribonucleases/metabolism , Porins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Endoribonucleases/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , RNA, Small Untranslated/metabolism , Salmonella/genetics , Salmonella/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...