Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
RNA ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839110

ABSTRACT

ArcZ is a small regulatory RNA conserved in Enterobacterales It is a Hfq-dependent RNA that is cleaved by RNase E in a processed form of 55 to 60 nucleotides. This processed form is highly conserved for controlling the expression of target mRNAs. ArcZ expression is induced by abundant oxygen levels and reaches its peak during the stationary growth phase. This control is mediated by the oxygen-responsive two-component system ArcAB, leading to the repression of arcZ transcription under low-oxygen conditions in most bacteria in which it has been studied. ArcZ displays multiple targets, and it can control up to 10% of a genome and interact directly with more than 300 mRNAs in Escherichia coli and Salmonella enterica ArcZ displays a multi-faceted ability to regulate its targets through diverse mechanisms such as RNase recruitment, modulation of ribosome accessibility on the mRNA and interaction with translational enhancing regions. By influencing stress response, motility and virulence through the regulation of master regulators such as FlhDC or RpoS, ArcZ emerges as a major orchestrator of cell physiology within Enterobacterales.

2.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119746, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719030

ABSTRACT

Iron­sulfur (Fe-S) clusters are one of the most ancient and versatile inorganic cofactors present in the three domains of life. Fe-S clusters are essential cofactors for the activity of a large variety of metalloproteins that play crucial physiological roles. Fe-S protein biogenesis is a complex process that starts with the acquisition of the elements (iron and sulfur atoms) and their assembly into an Fe-S cluster that is subsequently inserted into the target proteins. The Fe-S protein biogenesis is ensured by multiproteic systems conserved across all domains of life. Here, we provide an overview on how bacterial genetics approaches have permitted to reveal and dissect the Fe-S protein biogenesis process in vivo.


Subject(s)
Bacterial Proteins , Iron-Sulfur Proteins , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron/metabolism , Sulfur/metabolism , Bacteria/genetics , Bacteria/metabolism
3.
Microlife ; 4: uqad003, 2023.
Article in English | MEDLINE | ID: mdl-37223744

ABSTRACT

Iron-sulfur (Fe-S) clusters are important cofactors conserved in all domains of life, yet their synthesis and stability are compromised in stressful conditions such as iron deprivation or oxidative stress. Two conserved machineries, Isc and Suf, assemble and transfer Fe-S clusters to client proteins. The model bacterium Escherichia coli possesses both Isc and Suf, and in this bacterium utilization of these machineries is under the control of a complex regulatory network. To better understand the dynamics behind Fe-S cluster biogenesis in E. coli, we here built a logical model describing its regulatory network. This model comprises three biological processes: 1) Fe-S cluster biogenesis, containing Isc and Suf, the carriers NfuA and ErpA, and the transcription factor IscR, the main regulator of Fe-S clusters homeostasis; 2) iron homeostasis, containing the free intracellular iron regulated by the iron sensing regulator Fur and the non-coding regulatory RNA RyhB involved in iron sparing; 3) oxidative stress, representing intracellular H2O2 accumulation, which activates OxyR, the regulator of catalases and peroxidases that decompose H2O2 and limit the rate of the Fenton reaction. Analysis of this comprehensive model reveals a modular structure that displays five different types of system behaviors depending on environmental conditions, and provides a better understanding on how oxidative stress and iron homeostasis combine and control Fe-S cluster biogenesis. Using the model, we were able to predict that an iscR mutant would present growth defects in iron starvation due to partial inability to build Fe-S clusters, and we validated this prediction experimentally.

4.
Adv Microb Physiol ; 76: 1-39, 2020.
Article in English | MEDLINE | ID: mdl-32408945

ABSTRACT

Iron sulfur (Fe-S) clusters rank among the most ancient and conserved prosthetic groups. Fe-S clusters containing proteins are present in most, if not all, organisms. Fe-S clusters containing proteins are involved in a wide range of cellular processes, from gene regulation to central metabolism, via gene expression, RNA modification or bioenergetics. Fe-S clusters are built by biogenesis machineries conserved throughout both prokaryotes and eukaryotes. We focus mostly on bacterial ISC machinery, but not exclusively, as we refer to eukaryotic ISC system when it brings significant complementary information. Besides covering the structural and regulatory aspects of Fe-S biogenesis, this review aims to highlight Fe-S biogenesis facets remaining matters of discussion, such as the role of frataxin, or the link between fatty acid metabolism and Fe-S homeostasis. Last, we discuss recent advances on strategies used by different species to make and use Fe-S clusters in changing redox environmental conditions.


Subject(s)
Escherichia coli/physiology , Iron-Sulfur Proteins/physiology , Iron/metabolism , Sulfur/metabolism , Acyl Carrier Protein/physiology , Fatty Acids/metabolism , Gene Expression Regulation, Bacterial , Homeostasis , Humans , Iron-Binding Proteins , Iron-Sulfur Proteins/chemistry , Oxidation-Reduction , Saccharomyces cerevisiae/physiology , Frataxin
5.
Mol Microbiol ; 113(3): 588-592, 2020 03.
Article in English | MEDLINE | ID: mdl-31971637

ABSTRACT

An outstanding question regards the ability of organisms to sense their environments and respond in a suitable way. Pathogenic bacteria in particular exploit host-temperature sensing as a cue for triggering virulence gene expression. This micro-review does not attempt to fully cover the field of bacterial thermosensors and in detail describe each identified case. Instead, the review focus on the time-period at the end of the 1990's and beginning of the 2000's when several key discoveries were made, identifying protein, DNA and RNA as potential thermosensors controlling gene expression in several different bacterial pathogens in general and on the prfA thermosensor of Listeria monocytogenes in particular.


Subject(s)
Bacteria/metabolism , Host Microbial Interactions/physiology , Thermoreceptors/physiology , Bacteria/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Hot Temperature , Listeria monocytogenes/metabolism , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , RNA/genetics , RNA/metabolism , Thermoreceptors/metabolism , Thermosensing/genetics , Thermosensing/physiology , Trans-Activators/metabolism , Virulence/genetics , Virulence Factors/genetics
6.
PLoS Genet ; 15(4): e1008078, 2019 04.
Article in English | MEDLINE | ID: mdl-31009454

ABSTRACT

Phenotypic resistance describes a bacterial population that becomes transiently resistant to an antibiotic without requiring a genetic change. We here investigated the role of the small regulatory RNA (sRNA) RyhB, a key contributor to iron homeostasis, in the phenotypic resistance of Escherichia coli to various classes of antibiotics. We found that RyhB induces phenotypic resistance to gentamicin, an aminoglycoside that targets the ribosome, when iron is scarce. RyhB induced resistance is due to the inhibition of respiratory complexes Nuo and Sdh activities. These complexes, which contain numerous Fe-S clusters, are crucial for generating a proton motive force (pmf) that allows gentamicin uptake. RyhB regulates negatively the expression of nuo and sdh, presumably by binding to their mRNAs and, as a consequence, inhibiting their translation. We further show that Isc Fe-S biogenesis machinery is essential for the maturation of Nuo. As RyhB also limits levels of the Isc machinery, we propose that RyhB may also indirectly impact the maturation of Nuo and Sdh. Notably, our study shows that respiratory complexes activity levels are predictive of the bacterial sensitivity to gentamicin. Altogether, these results unveil a new role for RyhB in the adaptation to antibiotic stress, an unprecedented consequence of its role in iron starvation stress response.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacteria/metabolism , Bacterial Physiological Phenomena/drug effects , Gentamicins/pharmacology , Iron/metabolism , RNA, Small Untranslated , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Models, Biological , Mutation , RNA, Bacterial , RNA, Messenger/chemistry , RNA, Messenger/genetics
7.
Microbiol Spectr ; 6(2)2018 03.
Article in English | MEDLINE | ID: mdl-29573257

ABSTRACT

While iron is essential to sustain growth, its excess can be detrimental to the cell by generating highly toxic reactive oxygen species. Regulation of iron homeostasis thus plays a vital role in almost all living organisms. During the last 15 years, the small RNA (sRNA) RyhB has been shown to be a key actor of iron homeostasis regulation in bacteria. Through multiple molecular mechanisms, RyhB represses expendable iron-utilizing proteins, promotes siderophore production, and coordinates Fe-S cluster cofactor biogenesis, thereby establishing a so-called iron-sparing response. In this review, we will summarize knowledge on how sRNAs control iron homeostasis mainly through studies on RyhB in Escherichia coli. The parallel roles and modes of action of other sRNAs in different bacteria will also be described. Finally, we will discuss what questions remain to be answered concerning this important stress response regulation by sRNAs.


Subject(s)
Bacteria/metabolism , Homeostasis , Iron/metabolism , Iron/pharmacology , Ribosome Subunits, Small, Bacterial/metabolism , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Iron-Sulfur Proteins/metabolism , RNA , RNA, Bacterial/metabolism , Reactive Oxygen Species , Siderophores , Transcription Factors/metabolism , Virulence Factors
8.
mBio ; 7(5)2016 09 20.
Article in English | MEDLINE | ID: mdl-27651365

ABSTRACT

UNLABELLED: Fe-S clusters are cofactors conserved through all domains of life. Once assembled by dedicated ISC and/or SUF scaffolds, Fe-S clusters are conveyed to their apo-targets via A-type carrier proteins (ATCs). Escherichia coli possesses four such ATCs. ErpA is the only ATC essential under aerobiosis. Recent studies reported a possible regulation of the erpA mRNA by the small RNA (sRNA) RyhB, which controls the expression of many genes under iron starvation. Surprisingly, erpA has not been identified in recent transcriptomic analysis of the iron starvation response, thus bringing into question the actual physiological significance of the putative regulation of erpA by RyhB. Using an sRNA library, we show that among 26 sRNAs, only RyhB represses the expression of an erpA-lacZ translational fusion. We further demonstrate that this repression occurs during iron starvation. Using mutational analysis, we show that RyhB base pairs to the erpA mRNA, inducing its disappearance. In addition, IscR, the master regulator of Fe-S homeostasis, represses expression of erpA at the transcriptional level when iron is abundant, but depleting iron from the medium alleviates this repression. The conjunction of transcriptional derepression by IscR and posttranscriptional repression by RyhB under Fe-limiting conditions is best described as an incoherent regulatory circuit. This double regulation allows full expression of erpA at iron concentrations for which Fe-S biogenesis switches from the ISC to the SUF system. We further provide evidence that this regulatory circuit coordinates ATC usage to iron availability. IMPORTANCE: Regulatory small RNAs (sRNAs) have emerged as major actors in the control of gene expression in the last few decades. Relatively little is known about how these regulators interact with classical transcription factors to coordinate genetic responses. We show here how an sRNA, RyhB, and a transcription factor, IscR, regulate expression of an essential gene, erpA, in the bacterium E. coli ErpA is involved in the biogenesis of Fe-S clusters, an important class of cofactors involved in a plethora of cellular reactions. Interestingly, we show that RyhB and IscR repress expression of erpA under opposite conditions in regard to iron concentration, forming a regulatory circuit called an "incoherent network." This incoherent network serves to maximize expression of erpA at iron concentrations where it is most needed. Altogether, our study paves the way for a better understanding of mixed regulatory networks composed of RNAs and transcription factors.


Subject(s)
Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Iron-Sulfur Proteins/metabolism , RNA, Bacterial/genetics , Transcription Factors/metabolism , Aerobiosis , Anaerobiosis , Escherichia coli Proteins/genetics , Iron/metabolism , Iron-Sulfur Proteins/genetics , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Transcription Factors/genetics
9.
Biochim Biophys Acta ; 1827(8-9): 923-37, 2013.
Article in English | MEDLINE | ID: mdl-23660107

ABSTRACT

Iron/sulfur centers are key cofactors of proteins intervening in multiple conserved cellular processes, such as gene expression, DNA repair, RNA modification, central metabolism and respiration. Mechanisms allowing Fe/S centers to be assembled, and inserted into polypeptides have attracted much attention in the last decade, both in eukaryotes and prokaryotes. Basic principles and recent advances in our understanding of the prokaryotic Fe/S biogenesis ISC and SUF systems are reviewed in the present communication. Most studies covered stem from investigations in Escherichia coli and Azotobacter vinelandii. Remarkable insights were brought about by complementary structural, spectroscopic, biochemical and genetic studies. Highlights of the recent years include scaffold mediated assembly of Fe/S cluster, A-type carriers mediated delivery of clusters and regulatory control of Fe/S homeostasis via a set of interconnected genetic regulatory circuits. Also, the importance of Fe/S biosynthesis systems in mediating soft metal toxicity was documented. A brief account of the Fe/S biosynthesis systems diversity as present in current databases is given here. Moreover, Fe/S biosynthesis factors have themselves been the object of molecular tailoring during evolution and some examples are discussed here. An effort was made to provide, based on the E. coli system, a general classification associating a given domain with a given function such as to help next search and annotation of genomes. This article is part of a Special Issue entitled: Metals in Bioenergetics and Biomimetics Systems.

10.
Curr Opin Microbiol ; 16(2): 125-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23415757

ABSTRACT

Small regulatory RNAs (sRNAs) are now considered as major post-transcriptional regulators of gene expression in bacteria. Their importance is related to their variety in probably all bacterial species as well as to the extreme diversity of physiological functions of their target genes. An increasing amount of data point to an intimate connection between sRNAs and transcriptional regulatory networks to control multiple functions as important as motility or group behavior. The resulting mixed circuits unravel novel regulatory links and their properties are just starting to be characterized.


Subject(s)
Bacteria/genetics , Gene Expression Regulation, Bacterial , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic , Models, Biological
11.
Biochim Biophys Acta ; 1827(3): 455-69, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23298813

ABSTRACT

Iron/sulfur centers are key cofactors of proteins intervening in multiple conserved cellular processes, such as gene expression, DNA repair, RNA modification, central metabolism and respiration. Mechanisms allowing Fe/S centers to be assembled, and inserted into polypeptides have attracted much attention in the last decade, both in eukaryotes and prokaryotes. Basic principles and recent advances in our understanding of the prokaryotic Fe/S biogenesis ISC and SUF systems are reviewed in the present communication. Most studies covered stem from investigations in Escherichia coli and Azotobacter vinelandii. Remarkable insights were brought about by complementary structural, spectroscopic, biochemical and genetic studies. Highlights of the recent years include scaffold mediated assembly of Fe/S cluster, A-type carriers mediated delivery of clusters and regulatory control of Fe/S homeostasis via a set of interconnected genetic regulatory circuits. Also, the importance of Fe/S biosynthesis systems in mediating soft metal toxicity was documented. A brief account of the Fe/S biosynthesis systems diversity as present in current databases is given here. Moreover, Fe/S biosynthesis factors have themselves been the object of molecular tailoring during evolution and some examples are discussed here. An effort was made to provide, based on the E. coli system, a general classification associating a given domain with a given function such as to help next search and annotation of genomes. This article is part of a Special Issue entitled: Metals in Bioenergetics and Biomimetics Systems.


Subject(s)
Iron-Sulfur Proteins/metabolism , Prokaryotic Cells/metabolism , Carrier Proteins/physiology , Escherichia coli/metabolism , Escherichia coli Proteins/physiology , Homeostasis , Oxidative Stress
12.
Methods Mol Biol ; 905: 41-60, 2012.
Article in English | MEDLINE | ID: mdl-22735997

ABSTRACT

Small regulatory RNAs (sRNAs) are versatile regulators that have been shown to be involved in the gene regulation of a growing number of biological pathways in bacteria. While finding the targets of a given sRNA has been the focus of many studies, fewer methods have been described to uncover which, if any, sRNAs regulate a given gene. Here I present two genetic screens that are designed to search for sRNAs regulating a gene of interest. Before the screens are performed, a translational fusion is made between the gene of interest and lacZ, designed so that mostly post-transcriptional effects on the gene's expression can be analyzed. I describe here a simple and rapid way to obtain this fusion, even when the transcriptional start site is unknown, by combining PCR or 5'RACE with recombination in the chromosome of a special strain of Escherichia coli. The first genetic screen uses a genomic multicopy library to find regulator genes that, when overexpressed, affect the expression of the fusion. While this technique is a classical genetic screen, particular attention is paid to how it can be used to specifically find sRNAs. A second screen is described that takes advantage of a specific library of sRNAs of E. coli that provides an easier and more rapid way to look for sRNA regulation. The library is transformed into the fusion containing strain using a serial transformation protocol developed in microtiter plates. The transformants can then be directly assayed for effects on the beta-galactosidase activity of the fusion in liquid, providing a precise and rapid way to evaluate sRNA regulation. Use of one or both of these screens should help uncover new pathways of regulation by sRNAs.


Subject(s)
Genetic Techniques , RNA, Bacterial/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/genetics , Genes, Reporter/genetics , Lac Operon/genetics , Protein Biosynthesis
13.
EMBO J ; 29(18): 3094-107, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20683441

ABSTRACT

The alternative sigma factor RpoS responds to multiple stresses and activates a large number of genes that allow bacteria to adapt to changing environments. The accumulation of RpoS is regulated at multiple levels, including the regulation of its translation by small regulatory RNAs (sRNAs). A library of plasmids expressing each of 26 Escherichia coli sRNAs that bind Hfq was created to globally and rapidly analyse regulation of an rpoS-lacZ translational fusion. The approach can be easily applied to any gene of interest. When overexpressed, four sRNAs, including OxyS, previously shown to repress rpoS, were observed to repress the expression of the rpoS-lacZ fusion. Along with DsrA and RprA, two previously defined activators of rpoS translation, a third new sRNA activator, ArcZ, was identified. The expression of arcZ is repressed by the aerobic/anaerobic-sensing ArcA-ArcB two-component system under anaerobic conditions and adds translational regulation to the ArcA-ArcB regulon. ArcZ directly represses, and is repressed by, arcB transcription, providing a negative feedback loop that may affect functioning of the ArcA-ArcB regulon.


Subject(s)
Oxygen/metabolism , RNA, Bacterial/physiology , RNA, Untranslated/physiology , Aerobiosis , Anaerobiosis , Base Sequence , Blotting, Northern , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Sigma Factor/genetics , Sigma Factor/metabolism
14.
Proc Natl Acad Sci U S A ; 107(21): 9602-7, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20457943

ABSTRACT

Bacterial small noncoding RNAs carry out both positive and negative regulation of gene expression by pairing with mRNAs; in Escherichia coli, this regulation often requires the RNA chaperone Hfq. Three small regulatory RNAs (sRNAs), DsrA, RprA, and ArcZ, positively regulate translation of the sigma factor RpoS, each pairing with the 5' leader to open up an inhibitory hairpin. In vitro, rpoS interaction with sRNAs depends upon an (AAN)(4) Hfq-binding site upstream of the pairing region. Here we show that both Hfq and this Hfq binding site are required for RprA or ArcZ to act in vivo and to form a stable complex with rpoS mRNA in vitro; both were partially dispensable for DsrA at 37 degrees C. ArcZ sRNA is processed from 121 nt to a stable 56 nt species that contains the pairing region; only the 56 nt ArcZ makes a strong Hfq-dependent complex with rpoS. For each of these sRNAs, the stability of the sRNA*mRNA complexes, rather than their rate of formation, best predicted in vivo activity. These studies demonstrate that binding of Hfq to the rpoS mRNA is critical for sRNA regulation under normal conditions, but if the stability of the sRNA*mRNA complex is sufficiently high, the requirement for Hfq can be bypassed.


Subject(s)
Escherichia coli/metabolism , Host Factor 1 Protein/metabolism , RNA, Bacterial/metabolism , RNA, Untranslated/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/chemistry , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Messenger/metabolism , RNA, Untranslated/chemistry , Sigma Factor/genetics , Sigma Factor/metabolism
15.
Mol Microbiol ; 76(2): 467-79, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20345657

ABSTRACT

Numerous small RNAs regulators of gene expression exist in bacteria. A large class of them binds to the RNA chaperone Hfq and act by base pairing interactions with their target mRNA, thereby affecting their translation and/or stability. They often have multiple direct targets, some of which may be regulators themselves, and production of a single sRNA can therefore affect the expression of dozens of genes. We show in this study that the synthesis of the Escherichia coli pleiotropic PhoPQ two-component system is repressed by MicA, a sigma(E)-dependent sRNA regulator of porin biogenesis. MicA directly pairs with phoPQ mRNA in the translation initiation region of phoP and presumably inhibits translation by competing with ribosome binding. Consequently, MicA downregulates several members of the PhoPQ regulon. By linking PhoPQ to sigma(E), our findings suggest that major cellular processes such as Mg(2+) transport, virulence, LPS modification or resistance to antimicrobial peptides are modulated in response to envelope stress. In addition, we found that Hfq strongly affects the expression of phoP independently of MicA, raising the possibility that even more sRNAs, which remain to be identified, could regulate PhoPQ synthesis.


Subject(s)
Bacterial Proteins/biosynthesis , Cell Membrane/metabolism , Cell Wall/metabolism , Escherichia coli Proteins/biosynthesis , Gene Expression Regulation, Bacterial , MicroRNAs/metabolism , Stress, Physiological , Bacterial Proteins/antagonists & inhibitors , Escherichia coli/physiology , Escherichia coli Proteins/antagonists & inhibitors , Protein Biosynthesis , RNA, Messenger/metabolism , Regulon
16.
Cell ; 139(4): 770-9, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914169

ABSTRACT

Riboswitches are RNA elements acting in cis, controlling expression of their downstream genes through a metabolite-induced alteration of their secondary structure. Here, we demonstrate that two S-adenosylmethionine (SAM) riboswitches, SreA and SreB, can also function in trans and act as noncoding RNAs in Listeria monocytogenes. SreA and SreB control expression of the virulence regulator PrfA by binding to the 5'-untranslated region of its mRNA. Absence of the SAM riboswitches SreA and SreB increases the level of PrfA and virulence gene expression in L. monocytogenes. Thus, the impact of the SAM riboswitches on PrfA expression highlights a link between bacterial virulence and nutrient availability. Together, our results uncover an unexpected role for riboswitches and a distinct class of regulatory noncoding RNAs in bacteria.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/genetics , Peptide Termination Factors/genetics , Regulatory Sequences, Ribonucleic Acid , 5' Untranslated Regions , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Molecular Sequence Data , Temperature , Virulence
17.
Genes Dev ; 23(17): 1981-5, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19723760

ABSTRACT

Many bacterial small regulatory RNAs (sRNAs) pair with mRNA targets, stimulating or inhibiting mRNA stability and/or translation. Regulation of these sRNAs is usually due to tight transcriptional regulation of synthesis.In this issue of Genes & Development and a related paper in Molecular Microbiology, Figueroa-Bossi and colleagues (pp. 2004-2015) and Overgaard and colleagues report a novel regulatory mechanism in which induction of a competing mRNA acts to titrate away the sRNA, allowing expression of an otherwise strongly inhibited target gene.


Subject(s)
Gene Expression Regulation, Bacterial , MicroRNAs/metabolism , RNA, Bacterial/metabolism , Chitin/metabolism , Eukaryotic Cells/metabolism , MicroRNAs/genetics , RNA, Bacterial/genetics , RNA, Messenger/metabolism
18.
Mol Microbiol ; 72(3): 551-65, 2009 May.
Article in English | MEDLINE | ID: mdl-19426207

ABSTRACT

In Escherichia coli, the largest class of small regulatory RNAs binds to the RNA chaperone Hfq and regulates the stability and/or translation of specific mRNAs. While recent studies have shown that some mRNAs could be subject to post-transcriptional regulation by sRNAs (e.g. mRNAs found by co-immunoprecipitation with Hfq), no method has yet been described to identify small RNAs that regulate them. We developed a method to easily make translational fusions of genes of interest to the lacZ reporter gene, under the control of a P(BAD)-inducible promoter. A multicopy plasmid library of the E. coli genome can then be used to screen for small RNAs that affect the activity of the fusion. This screening method was first applied to the dpiB gene from the dpiBA operon, which encodes a two-component signal transduction system involved in the SOS response to beta-lactams. One small RNA, RybC, was found to negatively regulate the expression of dpiB. Using mutants in the dpiB-lacZ fusion and compensatory mutations in the RybC sRNA, we demonstrate that RybC directly base pairs with the dpiBA mRNA.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Protein Kinases/genetics , RNA Processing, Post-Transcriptional , RNA, Bacterial/genetics , Base Sequence , Gene Expression Regulation, Bacterial , Gene Library , Molecular Sequence Data , Operon , Plasmids , Promoter Regions, Genetic , RNA, Messenger/genetics , SOS Response, Genetics , Sequence Alignment , Signal Transduction
19.
Nucleic Acids Res ; 35(3): 962-74, 2007.
Article in English | MEDLINE | ID: mdl-17259222

ABSTRACT

To identify noncoding RNAs (ncRNAs) in the pathogenic bacterium Listeria monocytogenes, we analyzed the intergenic regions (IGRs) of strain EGD-e by in silico-based approaches. Among the twelve ncRNAs found, nine are novel and specific to the Listeria genus, and two of these ncRNAs are expressed in a growth-dependent manner. Three of the ncRNAs are transcribed in opposite direction to overlapping open reading frames (ORFs), suggesting that they act as antisense on the corresponding mRNAs. The other ncRNA genes appear as single transcription units. One of them displays five repeats of 29 nucleotides. Five of these new ncRNAs are absent from the non-pathogenic species L. innocua, raising the possibility that they might be involved in virulence. To predict mRNA targets of the ncRNAs, we developed a computational method based on thermodynamic pairing energies and known ncRNA-mRNA hybrids. Three ncRNAs, including one of the putative antisense ncRNAs, were predicted to have more than one mRNA targets. Several of them were shown to bind efficiently to the ncRNAs suggesting that our in silico approach could be used as a general tool to search for mRNA targets of ncRNAs.


Subject(s)
Listeria monocytogenes/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , 5' Flanking Region , Base Sequence , Computational Biology , DNA, Intergenic/chemistry , Electrophoretic Mobility Shift Assay , Genes, Bacterial , Genomics , Listeria monocytogenes/metabolism , Molecular Sequence Data , RNA, Messenger/chemistry , RNA, Untranslated/analysis , RNA, Untranslated/metabolism
20.
Mol Microbiol ; 57(5): 1367-80, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16102006

ABSTRACT

Signature-tagged mutagenesis (STM) was used to identify new genes involved in the virulence of the Gram-positive intracellular pathogen Listeria monocytogenes. One of the mutants isolated by this technique had the transposon inserted in virR, a gene encoding a putative response regulator of a two-component system. Deletion of virR severely decreased virulence in mice as well as invasion in cell-culture experiments. Using a transcriptomic approach, we identified 12 genes regulated by VirR, including the dlt-operon, previously reported to be important for L. monocytogenes virulence. However, a strain lacking dltA, was not as impaired in virulence as the DeltavirR strain, suggesting a role in virulence for other members of the vir regulon. Another VirR-regulated gene is homologous to mprF, which encodes a protein that modifies membrane phosphatidyl glycerol with l-lysine and that is involved in resistance to human defensins in Staphylococcus aureus. VirR thus appears to control virulence by a global regulation of surface components modifications. These modifications may affect interactions with host cells, including components of the innate immune system. Surprisingly, although controlling the same set of genes as VirR, the putative cognate histidine kinase of VirR, VirS, encoded by a gene located three genes downstream of virR, was shown not to be essential for virulence. By monitoring the activity of VirR with a GFP reporter construct, we showed that VirR can be activated independently of VirS, for example through a mechanism involving variations in the level of intracellular acetyl phosphate. In silico analysis of the VirR-regulated promoters revealed a VirR DNA-binding consensus site and specific interaction between purified VirR protein and this consensus sequence was demonstrated by gel mobility shift assays. This study identifies a second key virulence regulon in L. monocytogenes, after the prfA regulon.


Subject(s)
Bacterial Proteins/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Bacterial , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Animals , Bacterial Proteins/genetics , Base Sequence , Caco-2 Cells , Consensus Sequence , DNA-Binding Proteins/genetics , Female , Genes, Bacterial , Histidine Kinase , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Operon/genetics , Promoter Regions, Genetic , Protein Kinases/metabolism , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL