Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 21(1): e3001942, 2023 01.
Article in English | MEDLINE | ID: mdl-36603027

ABSTRACT

RNA processing and degradation shape the transcriptome by generating stable molecules that are necessary for translation (rRNA and tRNA) and by facilitating the turnover of mRNA, which is necessary for the posttranscriptional control of gene expression. In bacteria and the plant chloroplast, RNA degradosomes are multienzyme complexes that process and degrade RNA. In many bacterial species, the endoribonuclease RNase E is the central component of the RNA degradosome. RNase E-based RNA degradosomes are inner membrane proteins in a large family of gram-negative bacteria (ß- and γ-Proteobacteria). Until now, the reason for membrane localization was not understood. Here, we show that a mutant strain of Escherichia coli, in which the RNA degradosome is localized to the interior of the cell, has high levels of 20S and 40S particles that are defective intermediates in ribosome assembly. These particles have aberrant protein composition and contain rRNA precursors that have been cleaved by RNase E. After RNase E cleavage, rRNA fragments are degraded to nucleotides by exoribonucleases. In vitro, rRNA in intact ribosomes is resistant to RNase E cleavage, whereas protein-free rRNA is readily degraded. We conclude that RNA degradosomes in the nucleoid of the mutant strain interfere with cotranscriptional ribosome assembly. We propose that membrane-attached RNA degradosomes in wild-type cells control the quality of ribosome assembly after intermediates are released from the nucleoid. That is, the compact structure of mature ribosomes protects rRNA against cleavage by RNase E. Turnover of a proportion of intermediates in ribosome assembly explains slow growth of the mutant strain. Competition between mRNA and rRNA degradation could be the cause of slower mRNA degradation in the mutant strain. We conclude that attachment of the RNA degradosome to the bacterial inner cytoplasmic membrane prevents wasteful degradation of rRNA precursors, thus explaining the reason for conservation of membrane-attached RNA degradosomes throughout the ß- and γ-Proteobacteria.


Subject(s)
Escherichia coli Proteins , RNA, Ribosomal , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Ribosomes/metabolism , Multienzyme Complexes/metabolism , RNA/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Cell Membrane/metabolism , Bacteria/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Bacterial/genetics
2.
Annu Rev Microbiol ; 76: 533-552, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35671533

ABSTRACT

RNA degradosomes are multienzyme complexes composed of ribonucleases, RNA helicases, and metabolic enzymes. RNase E-based degradosomes are widespread in Proteobacteria. The Escherichia coli RNA degradosome is sequestered from transcription in the nucleoid and translation in the cytoplasm by localization to the inner cytoplasmic membrane, where it forms short-lived clusters that are proposed to be sites of mRNA degradation. In Caulobacter crescentus, RNA degradosomes localize to ribonucleoprotein condensates in the interior of the cell [bacterial ribonucleoprotein-bodies (BR-bodies)], which have been proposed to drive the concerted degradation of mRNA to nucleotides. The turnover of mRNA in growing cells is important for maintaining pools of nucleotides for transcription and DNA replication.Membrane attachment of the E. coli RNA degradosome is necessary to avoid wasteful degradation of intermediates in ribosome assembly. Sequestering RNA degradosomes to C. crescentus BR-bodies, which exclude structured RNA, could have a similar role in protecting intermediates in ribosome assembly from degradation.


Subject(s)
Caulobacter crescentus , Endoribonucleases , Escherichia coli , Multienzyme Complexes , Nucleotides , Polyribonucleotide Nucleotidyltransferase , RNA Helicases , RNA Stability , RNA, Messenger , Caulobacter crescentus/enzymology , Caulobacter crescentus/genetics , Endoribonucleases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nucleotides/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism
3.
Mol Microbiol ; 117(1): 54-66, 2022 01.
Article in English | MEDLINE | ID: mdl-34219284

ABSTRACT

Under conditions of nutrient adversity, bacteria adjust metabolism to minimize cellular energy usage. This is often achieved by controlling the synthesis and degradation of RNA. In Escherichia coli, RNase E is the central enzyme involved in RNA degradation and serves as a scaffold for the assembly of the multiprotein complex known as the RNA degradosome. The activity of RNase E against specific mRNAs can also be regulated by the action of small RNAs (sRNA). In this case, the ubiquitous bacterial chaperone Hfq bound to sRNAs can interact with the RNA degradosome for the sRNA guided degradation of target RNAs. The RNA degradosome and Hfq have never been visualized together in live bacteria. We now show that in long-term nitrogen starved E. coli, both RNase E and Hfq co-localize in a single, large focus. This subcellular assembly, which we refer to as the H-body, forms by a liquid-liquid phase separation type mechanism and includes components of the RNA degradosome, namely, the helicase RhlB and the exoribonuclease polynucleotide phosphorylase. The results support the existence of a hitherto unreported subcellular compartmentalization of a process(s) associated with RNA management in stressed bacteria.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Host Factor 1 Protein/metabolism , Multienzyme Complexes , Nitrogen/deficiency , Polyribonucleotide Nucleotidyltransferase , RNA Helicases , Cell Compartmentation , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Escherichia coli/enzymology , Escherichia coli/physiology , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , RNA Stability , RNA, Bacterial/genetics , Stress, Physiological
4.
mBio ; 12(5): e0193221, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34488454

ABSTRACT

The essential endoribonuclease RNase E, which is a component of the Escherichia coli multienzyme RNA degradosome, has a global role in RNA processing and degradation. RNase E localizes to the inner cytoplasmic membrane in small, short-lived clusters (puncta). Rifampin, which arrests transcription, inhibits RNase E clustering and increases its rate of diffusion. Here, we show that inhibition of clustering is due to the arrest of transcription using a rifampin-resistant control strain. Two components of the RNA degradosome, the 3' exoribonuclease polynucleotide phosphorylase (PNPase) and the DEAD box RNA helicase RhlB, colocalize with RNase E in puncta. Clustering of PNPase and RhlB is inhibited by rifampin, and their diffusion rates increase, as evidenced by in vivo photobleaching measurements. Results with rifampin treatment reported here show that RNA degradosome diffusion is constrained by interaction with RNA substrate. Kasugamycin, which arrests translation initiation, inhibits formation of puncta and increases RNA degradosome diffusion rates. Since kasugamycin treatment results in continued synthesis and turnover of ribosome-free mRNA but inhibits polyribosome formation, RNA degradosome clustering is therefore polyribosome dependent. Chloramphenicol, which arrests translation elongation, results in formation of large clusters (foci) of RNA degradosomes that are distinct from puncta. Since chloramphenicol-treated ribosomes are stable, the formation of RNA degradosome foci could be part of a stress response that protects inactive polyribosomes from degradation. Our results strongly suggest that puncta are sites where translationally active polyribosomes are captured by membrane-associated RNA degradosomes. These sites could be part of a scanning process that is an initial step in mRNA degradation. IMPORTANCE Here, we show that RNase E, RhlB, and PNPase act together as components of the multienzyme RNA degradosome in polyribosome-dependent clustering to form puncta on the inner cytoplasmic membrane. Our results support the hypothesis that RNA degradosome puncta are sites of mRNA degradation. We propose that clustering of RNA degradosomes is a pre-RNase E cleavage step in which polyribosomes are scanned in a search for ribosome-free mRNA. This work is part of an emerging view that posttranscriptional events such as tRNA maturation, late steps in ribosome assembly, and mRNA degradation are membrane associated and partitioned from translation in the cytoplasm and transcription in the nucleoid. This separation could protect newly synthesized transcripts from premature destructive interactions with the RNA degradosome. The scanning of ribosomes and polyribosomes could be part of a general mechanism in which defective stable RNA or ribosome-free mRNA is targeted for destruction by the RNA degradosome.


Subject(s)
Escherichia coli/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , Polyribosomes/metabolism , RNA Stability/genetics , Cluster Analysis , Endoribonucleases/metabolism , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Exoribonucleases , Multienzyme Complexes , RNA Helicases , RNA Processing, Post-Transcriptional , RNA, Bacterial , RNA, Messenger/metabolism , Rifampin/pharmacology
5.
RNA ; 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34099575

ABSTRACT

Rifampicin, a broad-spectrum antibiotic, inhibits bacterial RNA polymerase. Here we show that rifampicin treatment of Escherichia coli results in a 50% decrease in cell size due to a terminal cell division. This decrease is a consequence of inhibition of transcription as evidenced by an isogenic rifampicin-resistant strain. There is also a 50% decrease in total RNA due mostly to a 90% decrease in 23S and 16S rRNA levels. Control experiments showed this decrease is not an artifact of our RNA purification protocol and therefore due to degradation in vivo. Since chromosome replication continues after rifampicin treatment, ribonucleotides from rRNA degradation could be recycled for DNA synthesis. Rifampicin-induced rRNA degradation occurs under different growth conditions and in different strain backgrounds. However, rRNA degradation is never complete thus permitting the re-initiation of growth after removal of rifampicin. The orderly shutdown of growth under conditions where the induction of stress genes is blocked by rifampicin is noteworthy. Inhibition of protein synthesis by chloramphenicol resulted in a partial decrease in 23S and 16S rRNA levels whereas kasugamycin treatment had no effect. Analysis of temperature-sensitive mutant strains implicate RNase E, PNPase and RNase R in rifampicin-induced rRNA degradation. We cannot distinguish between a direct role for RNase E in rRNA degradation versus an indirect role involving a slowdown of mRNA degradation. Since mRNA and rRNA appear to be degraded by the same ribonucleases, competition by rRNA is likely to result in slower mRNA degradation rates in the presence of rifampicin than under normal growth conditions.

6.
Mol Microbiol ; 111(6): 1715-1731, 2019 06.
Article in English | MEDLINE | ID: mdl-30903628

ABSTRACT

The reason for RNase E attachment to the inner membrane is largely unknown. To understand the cell biology of RNA degradation, we have characterized a strain expressing RNase E lacking the membrane attachment site (cytoplasmic RNase E). Genome-wide data show a global slowdown in mRNA degradation. There is no correlation between mRNA stabilization and the function or cellular location of encoded proteins. The activity of cRNase E is comparable to the wild-type enzyme in vitro, but the mutant protein is unstable in vivo. Autoregulation of cRNase E synthesis compensates for protein instability. cRNase E associates with other proteins to assemble a cytoplasmic RNA degradosome. CsrB/C sRNAs, whose stability is regulated by membrane-associated CsrD, are stabilized. Membrane attachment of RNase E is thus necessary for CsrB/C turnover. In contrast to mRNA stability, ribosome-free transcripts are sensitive to inactivation by cRNase E. Our results show that effects on RNA degradation are not due to the differences in the activity or level of cRNase E, or failure to assemble the RNA degradosome. We propose that membrane attachment is necessary for RNase E stability, functional interactions with membrane-associated regulatory factors and protection of ribosome-free transcripts from premature interactions with RNase E in the nucleoid.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli/genetics , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , RNA Stability , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Proteolysis , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribosomes/genetics
7.
Methods Enzymol ; 612: 47-66, 2018.
Article in English | MEDLINE | ID: mdl-30502954

ABSTRACT

In this study, we compared different computational methods used for genome-wide determination of mRNA half-lives in Escherichia coli with a special focus on the impact on considering a delay before the onset of mRNA decay after transcription arrest. A wide variety of datasets were analyzed coming from different technical methods for mRNA quantification (microarrays, RNA-seq, and RT-qPCR) and different bacterial growth conditions. The exponential decay of mRNA levels was fitted using both linear and exponential models and with or without a delay. We showed that for all the models, independently of mRNA quantification methods and growth conditions, ignoring the delay resulted in only a modest overestimation of the half-life. For approximately 80% of the mRNAs, differences in mRNA half-life values were less than 34s. The correlation between half-lives estimated with and without a delay was extremely high. However, the slope of the linear regression between the half-lives with and without a delay tended to decrease with the delay. For the few mRNAs for which taking into account the delay influenced the estimated half-life, the impact was dependent on the model and the growth condition. The smallest impact was obtained for the linear model.


Subject(s)
Escherichia coli/genetics , RNA Stability/physiology , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA Stability/genetics , Transcription, Genetic/genetics
9.
Sci Rep ; 6: 25057, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27112822

ABSTRACT

Bacterial adaptation requires large-scale regulation of gene expression. We have performed a genome-wide analysis of the Csr system, which regulates many important cellular functions. The Csr system is involved in post-transcriptional regulation, but a role in transcriptional regulation has also been suggested. Two proteins, an RNA-binding protein CsrA and an atypical signaling protein CsrD, participate in the Csr system. Genome-wide transcript stabilities and levels were compared in wildtype E. coli (MG1655) and isogenic mutant strains deficient in CsrA or CsrD activity demonstrating for the first time that CsrA and CsrD are global negative and positive regulators of transcription, respectively. The role of CsrA in transcription regulation may be indirect due to the 4.6-fold increase in csrD mRNA concentration in the CsrA deficient strain. Transcriptional action of CsrA and CsrD on a few genes was validated by transcriptional fusions. In addition to an effect on transcription, CsrA stabilizes thousands of mRNAs. This is the first demonstration that CsrA is a global positive regulator of mRNA stability. For one hundred genes, we predict that direct control of mRNA stability by CsrA might contribute to metabolic adaptation by regulating expression of genes involved in carbon metabolism and transport independently of transcriptional regulation.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Membrane Proteins/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Carbon/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial , Membrane Proteins/genetics , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic
10.
Mol Microbiol ; 97(6): 1021-135, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096689

ABSTRACT

Ribonuclease E (RNase E) of Escherichia coli, which is the founding member of a widespread family of proteins in bacteria and chloroplasts, is a fascinating enzyme that still has not revealed all its secrets. RNase E is an essential single-strand specific endoribonuclease that is involved in the processing and degradation of nearly every transcript in E. coli. A striking enzymatic property is a preference for substrates with a 5' monophosphate end although recent work explains how RNase E can overcome the protection afforded by the 5' triphosphate end of a primary transcript. Other features of E. coli RNase E include its interaction with enzymes involved in RNA degradation to form the multienzyme RNA degradosome and its localization to the inner cytoplasmic membrane. The N-terminal catalytic core of the RNase E protomer associates to form a tetrameric holoenzyme. Each RNase E protomer has a large C-terminal intrinsically disordered (ID) noncatalytic region that contains sites for interactions with protein components of the RNA degradosome as well as RNA and phospholipid bilayers. In this review, RNase E homologs have been classified into five types based on their primary structure. A recent analysis has shown that type I RNase E in the γ-proteobacteria forms an orthologous group of proteins that has been inherited vertically. The RNase E catalytic core and a large ID noncatalytic region containing an RNA binding motif and a membrane targeting sequence are universally conserved features of these orthologs. Although the ID noncatalytic region has low composition and sequence complexity, it is possible to map microdomains, which are short linear motifs that are sites of interaction with protein and other ligands. Throughout bacteria, the composition of the multienzyme RNA degradosome varies with species, but interactions with exoribonucleases (PNPase, RNase R), glycolytic enzymes (enolase, aconitase) and RNA helicases (DEAD-box proteins, Rho) are common. Plasticity in RNA degradosome composition is due to rapid evolution of RNase E microdomains. Characterization of the RNase E-PNPase interaction in α-proteobacteria, γ-proteobacteria and cyanobacteria suggests that it arose independently several times during evolution, thus conferring an advantage in control and coordination of RNA processing and degradation.


Subject(s)
Bacteria/enzymology , Chloroplasts/enzymology , Endoribonucleases/chemistry , Endoribonucleases/genetics , Evolution, Molecular , Multienzyme Complexes/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/genetics , Animals , Bacteria/genetics , Endoribonucleases/classification , Endoribonucleases/metabolism , Escherichia coli/metabolism , Humans , Multienzyme Complexes/metabolism , Plants/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , Protein Interaction Domains and Motifs , RNA Helicases/metabolism
11.
PLoS Genet ; 11(2): e1004961, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25647427

ABSTRACT

RNase E, which is the central component of the multienzyme RNA degradosome, serves as a scaffold for interaction with other enzymes involved in mRNA degradation including the DEAD-box RNA helicase RhlB. Epifluorescence microscopy under live cell conditions shows that RNase E and RhlB are membrane associated, but neither protein forms cytoskeletal-like structures as reported earlier by Taghbalout and Rothfield. We show that association of RhlB with the membrane depends on a direct protein interaction with RNase E, which is anchored to the inner cytoplasmic membrane through an MTS (Membrane Targeting Sequence). Molecular dynamics simulations show that the MTS interacts with the phospholipid bilayer by forming a stabilized amphipathic α-helix with the helical axis oriented parallel to the plane of the bilayer and hydrophobic side chains buried deep in the acyl core of the membrane. Based on the molecular dynamics simulations, we propose that the MTS freely diffuses in the membrane by a novel mechanism in which a large number of weak contacts are rapidly broken and reformed. TIRFm (Total Internal Reflection microscopy) shows that RNase E in live cells rapidly diffuses over the entire inner membrane forming short-lived foci. Diffusion could be part of a scanning mechanism facilitating substrate recognition and cooperativity. Remarkably, RNase E foci disappear and the rate of RNase E diffusion increases with rifampicin treatment. Control experiments show that the effect of rifampicin is specific to RNase E and that the effect is not a secondary consequence of the shut off of E. coli transcription. We therefore interpret the effect of rifampicin as being due to the depletion of RNA substrates for degradation. We propose a model in which formation of foci and constraints on diffusion arise from the transient clustering of RNase E into cooperative degradation bodies.


Subject(s)
DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Escherichia coli Proteins/genetics , Multienzyme Complexes/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/genetics , RNA Stability/genetics , Cell Membrane Structures/chemistry , Cell Membrane Structures/genetics , DEAD-box RNA Helicases/chemistry , Endoribonucleases/chemistry , Escherichia coli/genetics , Molecular Dynamics Simulation , Multienzyme Complexes/chemistry , Nucleic Acid Conformation , Phospholipids/chemistry , Phospholipids/genetics , Polyribonucleotide Nucleotidyltransferase/chemistry , Protein Interaction Maps/genetics , RNA Helicases/chemistry , RNA, Messenger/genetics
12.
Mol Genet Genomics ; 290(3): 847-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25432321

ABSTRACT

RNase E of Escherichia coli is a membrane-associated endoribonuclease that has a major role in mRNA degradation. The enzyme has a large C-terminal noncatalytic region that is mostly intrinsically disordered (ID). Under standard growth conditions, RhlB, enolase and PNPase associate with the noncatalytic region to form the multienzyme RNA degradosome. To elucidate the origin and evolution of the RNA degradosome, we have identified and characterized orthologs of RNase E in the γ-Proteobacteria, a phylum of bacteria with diverse ecological niches and metabolic phenotypes and an ancient origin contemporary with the radiation of animals, plants and fungi. Intrinsic disorder, composition bias and tandem sequence repeats are conserved features of the noncatalytic region. Composition bias is bipartite with a catalytic domain proximal ANR-rich region and distal AEPV-rich region. Embedded in the noncatalytic region are microdomains (also known as MoRFs, MoREs or SLiMs), which are motifs that interact with protein and other ligands. Our results suggest that tandem repeat sequences are the progenitors of microdomains. We have identified 24 microdomains with phylogenetic signals that were acquired once with few losses. Microdomains involved in membrane association and RNA binding are universally conserved suggesting that they were present in ancestral RNase E. The RNA degradosome of E. coli arose in two steps with RhlB and PNPase acquisition early in a major subtree of the γ-Proteobacteria and enolase acquisition later. We propose a mechanism of microdomain acquisition and evolution and discuss implications of these results for the structure and function of the multienzyme RNA degradosome.


Subject(s)
Endoribonucleases/genetics , Evolution, Molecular , Gammaproteobacteria/genetics , Multienzyme Complexes/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/genetics , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/genetics , Conserved Sequence , Gammaproteobacteria/enzymology , Phylogeny , Protein Interaction Mapping , Protein Structure, Tertiary , RNA Stability , RNA, Bacterial/genetics , RNA, Messenger/genetics , Sequence Alignment
13.
Nucleic Acids Res ; 42(4): 2460-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24243845

ABSTRACT

Microorganisms extensively reorganize gene expression to adjust growth rate to changes in growth conditions. At the genomic scale, we measured the contribution of both transcription and transcript stability to regulating messenger RNA (mRNA) concentration in Escherichia coli. Transcriptional control was the dominant regulatory process. Between growth rates of 0.10 and 0.63 h(-1), there was a generic increase in the bulk mRNA transcription. However, many transcripts became less stable and the median mRNA half-life decreased from 4.2 to 2.8 min. This is the first evidence that mRNA turnover is slower at extremely low-growth rates. The destabilization of many, but not all, transcripts at high-growth rate correlated with transcriptional upregulation of genes encoding the mRNA degradation machinery. We identified five classes of growth-rate regulation ranging from mainly transcriptional to mainly degradational. In general, differential stability within polycistronic messages encoded by operons does not appear to be affected by growth rate. We show here that the substantial reorganization of gene expression involving downregulation of tricarboxylic acid cycle genes and acetyl-CoA synthetase at high-growth rates is controlled mainly by transcript stability. Overall, our results demonstrate that the control of transcript stability has an important role in fine-tuning mRNA concentration during changes in growth rate.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , RNA Stability , RNA, Messenger/metabolism , Transcription, Genetic , Escherichia coli/growth & development , Escherichia coli/metabolism , Glucose/metabolism
14.
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
15.
Biochim Biophys Acta ; 1829(6-7): 532-51, 2013.
Article in English | MEDLINE | ID: mdl-23403287

ABSTRACT

The ß-CASP ribonucleases, which are found in the three domains of life, have in common a core of 460 residues containing seven conserved sequence motifs involved in the tight binding of two catalytic zinc ions. A hallmark of these enzymes is their ability to catalyze both endo- and exo-ribonucleolytic degradation. Exo-ribonucleolytic degradation proceeds in the 5' to 3' direction and is sensitive to the phosphorylation state of the 5' end of a transcript. Recent phylogenomic analyses have shown that the ß-CASP ribonucleases can be partitioned into two major subdivisions that correspond to orthologs of eukaryal CPSF73 and bacterial RNase J. We discuss the known functions of the CPSF73 and RNase J orthologs, their association into complexes, and their structure as it relates to mechanism of action. Eukaryal CPSF73 is part of a large multiprotein complex that is involved in the maturation of the 3' end of RNA Polymerase II transcripts and the polyadenylation of messenger RNA. RNase J1 and J2 are paralogs in Bacillus subtilis that are involved in the degradation of messenger RNA and the maturation of non-coding RNA. RNase J1 and J2 co-purify as a heteromeric complex and there is recent evidence that they interact with other enzymes to form a bacterial RNA degradosome. Finally, we speculate on the evolutionary origin of ß-CASP ribonucleases and on their functions in Archaea. Orthologs of CPSF73 with endo- and exo-ribonuclease activity are strictly conserved throughout the archaea suggesting a role for these enzymes in the maturation and/or degradation of messenger RNA. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Subject(s)
Endoribonucleases/genetics , Multienzyme Complexes/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/genetics , RNA Stability/genetics , Archaea/enzymology , Archaea/genetics , Conserved Sequence/genetics , Endoribonucleases/chemistry , Evolution, Molecular , Humans , Multienzyme Complexes/chemistry , Polyribonucleotide Nucleotidyltransferase/chemistry , RNA Helicases/chemistry , mRNA Cleavage and Polyadenylation Factors/genetics
16.
Nucleic Acids Res ; 41(2): 1091-103, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23222134

ABSTRACT

Bacterial RNase J and eukaryal cleavage and polyadenylation specificity factor (CPSF-73) are members of the ß-CASP family of ribonucleases involved in mRNA processing and degradation. Here we report an in-depth phylogenomic analysis that delineates aRNase J and archaeal CPSF (aCPSF) as distinct orthologous groups and establishes their repartition in 110 archaeal genomes. The aCPSF1 subgroup, which has been inherited vertically and is strictly conserved, is characterized by an N-terminal extension with two K homology (KH) domains and a C-terminal motif involved in dimerization of the holoenzyme. Pab-aCPSF1 (Pyrococcus abyssi homolog) has an endoribonucleolytic activity that preferentially cleaves at single-stranded CA dinucleotides and a 5'-3' exoribonucleolytic activity that acts on 5' monophosphate substrates. These activities are the same as described for the eukaryotic cleavage and polyadenylation factor, CPSF-73, when engaged in the CPSF complex. The N-terminal KH domains are important for endoribonucleolytic cleavage at certain specific sites and the formation of stable high molecular weight ribonucleoprotein complexes. Dimerization of Pab-aCPSF is important for exoribonucleolytic activity and RNA binding. Altogether, our results suggest that aCPSF1 performs an essential function and that an enzyme with similar activities was present in the last common ancestor of Archaea and Eukarya.


Subject(s)
Archaeal Proteins/classification , Ribonucleases/classification , Amino Acid Sequence , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Cleavage And Polyadenylation Specificity Factor/classification , Conserved Sequence , Endoribonucleases/metabolism , Exoribonucleases/metabolism , Molecular Sequence Data , Phylogeny , Protein Multimerization , Protein Structure, Tertiary , Pyrococcus abyssi/enzymology , Ribonucleases/chemistry , Ribonucleases/metabolism
17.
Q Rev Biophys ; 45(2): 105-45, 2012 May.
Article in English | MEDLINE | ID: mdl-22169164

ABSTRACT

The RNA degradosome is a massive multi-enzyme assembly that occupies a nexus in RNA metabolism and post-transcriptional control of gene expression in Escherichia coli and many other bacteria. Powering RNA turnover and quality control, the degradosome serves also as a machine for processing structured RNA precursors during their maturation. The capacity to switch between destructive and processing modes involves cooperation between degradosome components and is analogous to the process of RNA surveillance in other domains of life. Recruitment of components and cellular compartmentalisation of the degradosome are mediated through small recognition domains that punctuate a natively unstructured segment within a scaffolding core. Dynamic in conformation, variable in composition and non-essential under certain laboratory conditions, the degradosome has nonetheless been maintained throughout the evolution of many bacterial species, due most likely to its diverse contributions in global cellular regulation. We describe the role of the degradosome and its components in RNA decay pathways in E. coli, and we broadly compare these pathways in other bacteria as well as archaea and eukaryotes. We discuss the modular architecture and molecular evolution of the degradosome, its roles in RNA degradation, processing and quality control surveillance, and how its activity is regulated by non-coding RNA. Parallels are drawn with analogous machinery in organisms from all life domains. Finally, we conjecture on roles of the degradosome as a regulatory hub for complex cellular processes.


Subject(s)
Endoribonucleases/chemistry , Endoribonucleases/metabolism , Escherichia coli/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/chemistry , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/chemistry , RNA Helicases/metabolism , Protein Conformation , RNA Processing, Post-Transcriptional , RNA Stability , Ribonucleases/chemistry , Ribonucleases/metabolism , Structure-Activity Relationship
18.
Mol Microbiol ; 82(6): 1305-10, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22074454

ABSTRACT

RNase E is an essential endoribonuclease with a preference for RNA substrates with 5'-monophosphate ends. Primary transcripts, which have 5' triphosphate ends, are thus protected from RNase E. Their conversion to 5'-monophosphate transcripts by RppH is a prerequisite for RNase E-mediated processing and degradation. 5'-monophosphate recognition involves binding to a subdomain in the catalytic core of RNase E known as the 5' sensor. There are, however, transcripts that can be attacked directly by RNase E in a 5'-end-independent pathway. Direct entry involves elements outside of the catalytic domain that are located in the carboxyl terminal half (CTH) of RNase E. Strains harbouring rne alleles that express variants of RNase E in which 5' sensing (rneR169Q) or direct entry (rneΔCTH) are inactivated, are viable. However, the rneR169Q/rneΔCTH and ΔrppH/rneΔCTH combinations are synthetic lethal suggesting that the essential function(s) of RNase E requires at least one of these pathways to be active. A striking result is the demonstration that mutations affecting Rho-dependent transcription termination can overcome synthetic lethality by a pathway that requires RNase H. It is hypothesized that R-loop formation and RNase H cleavage substitute for RNase E-dependent RNA processing and mRNA degradation.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Peptide Elongation Factors/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Endoribonucleases/chemistry , Endoribonucleases/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Peptide Elongation Factors/metabolism , Protein Structure, Tertiary , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rho Factor/genetics , Rho Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic
19.
BMC Genomics ; 12: 312, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21668986

ABSTRACT

BACKGROUND: Noncoding RNA (ncRNA) has been recognized as an important regulator of gene expression networks in Bacteria and Eucaryota. Little is known about ncRNA in thermococcal archaea except for the eukaryotic-like C/D and H/ACA modification guide RNAs. RESULTS: Using a combination of in silico and experimental approaches, we identified and characterized novel P. abyssi ncRNAs transcribed from 12 intergenic regions, ten of which are conserved throughout the Thermococcales. Several of them accumulate in the late-exponential phase of growth. Analysis of the genomic context and sequence conservation amongst related thermococcal species revealed two novel P. abyssi ncRNA families. The CRISPR family is comprised of crRNAs expressed from two of the four P. abyssi CRISPR cassettes. The 5'UTR derived family includes four conserved ncRNAs, two of which have features similar to known bacterial riboswitches. Several of the novel ncRNAs have sequence similarities to orphan OrfB transposase elements. Based on RNA secondary structure predictions and experimental results, we show that three of the twelve ncRNAs include Kink-turn RNA motifs, arguing for a biological role of these ncRNAs in the cell. Furthermore, our results show that several of the ncRNAs are subjected to processing events by enzymes that remain to be identified and characterized. CONCLUSIONS: This work proposes a revised annotation of CRISPR loci in P. abyssi and expands our knowledge of ncRNAs in the Thermococcales, thus providing a starting point for studies needed to elucidate their biological function.


Subject(s)
Genetic Loci/genetics , Pyrococcus abyssi/genetics , RNA, Archaeal/genetics , Riboswitch/genetics , 5' Untranslated Regions/genetics , Base Sequence , Conserved Sequence , DNA, Intergenic/genetics , Genome, Archaeal/genetics , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
20.
J Bacteriol ; 192(20): 5413-23, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20729366

ABSTRACT

The degradosome is a multienzyme complex involved in mRNA degradation in Escherichia coli. The essential endoribonuclease RNase E contains a large noncatalytic region necessary for protein-protein interactions with other components of the RNA degradosome. Interacting proteins include the DEAD-box RNA helicase RhlB, the glycolytic enzyme enolase, and the exoribonuclease PNPase. Pseudoalteromonas haloplanktis, a psychrotolerant gammaproteobacterium distantly related to E. coli, encodes homologs of each component of the RNA degradosome. In P. haloplanktis, RNase E associates with RhlB and PNPase but not enolase. Plasmids expressing P. haloplanktis RNase E (Ph-RNase E) can complement E. coli strains lacking E. coli RNase E (Ec-RNase E). Ph-RNase E, however, does not confer a growth advantage to E. coli at low temperature. Ph-RNase E has a heterologous protein-protein interaction with Ec-RhlB but not with Ec-enolase or Ec-PNPase. The Ph-RNase E binding sites for RhlB and PNPase were mapped by deletion analysis. The PNPase binding site is located at the C-terminal end of Ph-RNase E at the same position as that in Ec-RNase E, but the sequence of the site is not conserved. The sequence of the RhlB binding site in Ph-RNase E is related to the sequence in Ec-RNase E. Together with the heterologous interaction between Ph-RNase E and Ec-RhlB, our results suggest that the underlying structural motif for the RNase E-RhlB interaction is conserved. Since the activity of Ec-RhlB requires its physical interaction with Ec-RNase E, conservation of the underlying structural motif over a large evolutionary distance could be due to constraints involved in the control of RhlB activity.


Subject(s)
Endoribonucleases/metabolism , Gammaproteobacteria/genetics , Gene Expression Regulation, Bacterial/physiology , RNA Stability/genetics , RNA, Bacterial/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endoribonucleases/genetics , Escherichia coli , Genetic Complementation Test , Molecular Sequence Data , RNA Stability/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...