Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
1.
Antimicrob Agents Chemother ; 66(1): e0155921, 2022 01 18.
Article de Anglais | MEDLINE | ID: mdl-34748383

RÉSUMÉ

Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infections are associated with significant morbidity and mortality. MRSA secretes a number of virulence factors and pore-forming toxins that enable tissue invasion. Prior studies have found associations between decreased toxin production and poor outcomes in invasive MRSA infection, particularly in pneumonia. In this retrospective observational cohort study of MRSA bacteremia in adult patients from 2007 to 2015, we examined whether cytotoxicity was associated with 30-day mortality. Isolates were obtained from 776 patients and screened for cytotoxicity in a human HL-60 cell model, antimicrobial susceptibility, and spa type, and clinical data were abstracted from charts. We did not find an association between low cytotoxic activity and 30-day mortality in univariate logistic regression analyses. There was a difference in distribution of the genotypes across cytotoxicity phenotypes, with spa-CC008 accounting for a larger proportion of isolates in the high cytotoxicity group. Isolates with a skin and soft tissue primary infective site had a higher median cytotoxicity. There was no association between cytotoxicity and host factors such as age or comorbidity burden. The isolates in our study came from heterogeneous primary sites of infection and were predominantly from spa-CC002 and spa-CC008 lineages, so it is possible that findings in prior studies reflect a different distribution in genotypes and clinical syndromes. Overall, in this large study of cytotoxicity of MRSA bloodstream isolates, we did not find the low cytotoxicity phenotype to be predictive of poor outcomes in MRSA bacteremia.


Sujet(s)
Bactériémie , Staphylococcus aureus résistant à la méticilline , Infections à staphylocoques , Antibactériens/pharmacologie , Bactériémie/traitement médicamenteux , Humains , Staphylococcus aureus résistant à la méticilline/génétique , Tests de sensibilité microbienne , Études rétrospectives , Infections à staphylocoques/traitement médicamenteux , Facteurs de virulence/génétique
2.
Proc Natl Acad Sci U S A ; 116(27): 13563-13572, 2019 07 02.
Article de Anglais | MEDLINE | ID: mdl-31217288

RÉSUMÉ

The pathogen Staphylococcus aureus colonizes and infects a variety of different sites within the human body. To adapt to these different environments, S. aureus relies on a complex and finely tuned regulatory network. While some of these networks have been well-elucidated, the functions of more than 50% of the transcriptional regulators in S. aureus remain unexplored. Here, we assess the contribution of the LacI family of metabolic regulators to staphylococcal virulence. We found that inactivating the purine biosynthesis regulator purR resulted in a strain that was acutely virulent in bloodstream infection models in mice and in ex vivo models using primary human neutrophils. Remarkably, these enhanced pathogenic traits are independent of purine biosynthesis, as the purR mutant was still highly virulent in the presence of mutations that disrupt PurR's canonical role. Through the use of transcriptomics coupled with proteomics, we revealed that a number of virulence factors are differentially regulated in the absence of purR Indeed, we demonstrate that PurR directly binds to the promoters of genes encoding virulence factors and to master regulators of virulence. These results guided us into further ex vivo and in vivo studies, where we discovered that S. aureus toxins drive the death of human phagocytes and mice, whereas the surface adhesin FnbA contributes to the increased bacterial burden observed in the purR mutant. Thus, S. aureus repurposes a metabolic regulator to directly control the expression of virulence factors, and by doing so, tempers its pathogenesis.


Sujet(s)
Protéines bactériennes/métabolisme , Régulation de l'expression des gènes bactériens , Purines/biosynthèse , Protéines de répression/métabolisme , Staphylococcus aureus/métabolisme , Facteurs de virulence/métabolisme , Animaux , Protéines bactériennes/physiologie , Régulation de l'expression des gènes bactériens/physiologie , Humains , Souris , Protéines de répression/physiologie , Infections à staphylocoques/microbiologie , Staphylococcus aureus/pathogénicité , Facteurs de transcription/métabolisme , Facteurs de transcription/physiologie , Facteurs de virulence/physiologie
3.
Proc Natl Acad Sci U S A ; 116(5): 1745-1754, 2019 01 29.
Article de Anglais | MEDLINE | ID: mdl-30635416

RÉSUMÉ

The past two decades have witnessed an alarming expansion of staphylococcal disease caused by community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA). The factors underlying the epidemic expansion of CA-MRSA lineages such as USA300, the predominant CA-MRSA clone in the United States, are largely unknown. Previously described virulence and antimicrobial resistance genes that promote the dissemination of CA-MRSA are carried by mobile genetic elements, including phages and plasmids. Here, we used high-resolution genomics and experimental infections to characterize the evolution of a USA300 variant plaguing a patient population at increased risk of infection to understand the mechanisms underlying the emergence of genetic elements that facilitate clonal spread of the pathogen. Genetic analyses provided conclusive evidence that fitness (manifest as emergence of a dominant clone) changed coincidently with the stepwise emergence of (i) a unique prophage and mutation of the regulator of the pyrimidine nucleotide biosynthetic operon that promoted abscess formation and colonization, respectively, thereby priming the clone for success; and (ii) a unique plasmid that conferred resistance to two topical microbiocides, mupirocin and chlorhexidine, frequently used for decolonization and infection prevention. The resistance plasmid evolved through successive incorporation of DNA elements from non-S. aureus spp. into an indigenous cryptic plasmid, suggesting a mechanism for interspecies genetic exchange that promotes antimicrobial resistance. Collectively, the data suggest that clonal spread in a vulnerable population resulted from extensive clinical intervention and intense selection pressure toward a pathogen lifestyle that involved the evolution of consequential mutations and mobile genetic elements.


Sujet(s)
Infections communautaires/microbiologie , Staphylococcus aureus résistant à la méticilline/génétique , Virulence/génétique , Animaux , Antibactériens/pharmacologie , Enfant , Chlorhexidine/pharmacologie , Infections communautaires/traitement médicamenteux , Génome bactérien/génétique , Humains , Staphylococcus aureus résistant à la méticilline/effets des médicaments et des substances chimiques , Souris , Tests de sensibilité microbienne/méthodes , Mupirocine/pharmacologie , Phylogenèse , Plasmides/génétique , Infections à staphylocoques/traitement médicamenteux , Infections à staphylocoques/microbiologie
4.
Infect Immun ; 86(10)2018 10.
Article de Anglais | MEDLINE | ID: mdl-30061376

RÉSUMÉ

Therapy for bacteremia caused by Staphylococcus aureus is often ineffective, even when treatment conditions are optimal according to experimental protocols. Adapted subclones, such as those bearing mutations that attenuate agr-mediated virulence activation, are associated with persistent infection and patient mortality. To identify additional alterations in agr-defective mutants, we sequenced and assembled the complete genomes of clone pairs from colonizing and infected sites of several patients in whom S. aureus demonstrated a within-host loss of agr function. We report that events associated with agr inactivation result in agr-defective blood and nares strain pairs that are enriched in mutations compared to pairs from wild-type controls. The random distribution of mutations between colonizing and infecting strains from the same patient, and between strains from different patients, suggests that much of the genetic complexity of agr-defective strains results from prolonged infection or therapy-induced stress. However, in one of the agr-defective infecting strains, multiple genetic changes resulted in increased virulence in a murine model of bloodstream infection, bypassing the mutation of agr and raising the possibility that some changes were selected. Expression profiling correlated the elevated virulence of this agr-defective mutant to restored expression of the agr-regulated ESAT6-like type VII secretion system, a known virulence factor. Thus, additional mutations outside the agr locus can contribute to diversification and adaptation during infection by S. aureus agr mutants associated with poor patient outcomes.


Sujet(s)
Protéines bactériennes/génétique , Génome bactérien , Infections à staphylocoques/microbiologie , Staphylococcus aureus/génétique , Staphylococcus aureus/métabolisme , Transactivateurs/génétique , Animaux , Bactériémie/microbiologie , Protéines bactériennes/métabolisme , Femelle , Régulation de l'expression des gènes bactériens , Humains , Souris , Mutation , Phylogenèse , Staphylococcus aureus/classification , Staphylococcus aureus/pathogénicité , Transactivateurs/métabolisme , Virulence
5.
mBio ; 9(1)2018 01 23.
Article de Anglais | MEDLINE | ID: mdl-29362239

RÉSUMÉ

Staphylococcus aureus is a versatile bacterial pathogen that can cause significant disease burden and mortality. Like other pathogens, S. aureus must adapt to its environment to produce virulence factors to survive the immune responses evoked by infection. Despite the importance of environmental signals for S. aureus pathogenicity, only a limited number of these signals have been investigated in detail for their ability to modulate virulence. Here we show that pyruvate, a central metabolite, causes alterations in the overall metabolic flux of S. aureus and enhances its pathogenicity. We demonstrate that pyruvate induces the production of virulence factors such as the pore-forming leucocidins and that this induction results in increased virulence of community-acquired methicillin-resistant S. aureus (CA-MRSA) clone USA300. Specifically, we show that an efficient "pyruvate response" requires the activation of S. aureus master regulators AgrAC and SaeRS as well as the ArlRS two-component system. Altogether, our report further establishes a strong relationship between metabolism and virulence and identifies pyruvate as a novel regulatory signal for the coordination of the S. aureus virulon through intricate regulatory networks.IMPORTANCE Delineation of the influence of host-derived small molecules on the makeup of human pathogens is a growing field in understanding host-pathogen interactions. S. aureus is a prominent pathogen that colonizes up to one-third of the human population and can cause serious infections that result in mortality in ~15% of cases. Here, we show that pyruvate, a key nutrient and central metabolite, causes global changes to the metabolic flux of S. aureus and activates regulatory networks that allow significant increases in the production of leucocidins. These and other virulence factors are critical for S. aureus to infect diverse host niches, initiate infections, and effectively subvert host immune responses. Understanding how environmental signals, particularly ones that are essential to and prominent in the human host, affect virulence will allow us to better understand pathogenicity and consider more-targeted approaches to tackling the current S. aureus epidemic.


Sujet(s)
Staphylococcus aureus résistant à la méticilline/métabolisme , Staphylococcus aureus résistant à la méticilline/pathogénicité , Acide pyruvique/métabolisme , Facteurs de virulence/biosynthèse , Régulation de l'expression des gènes bactériens/effets des médicaments et des substances chimiques , Humains , Métabolisme/effets des médicaments et des substances chimiques , Infections à staphylocoques , Virulence
6.
Mol Cell Proteomics ; 16(4 suppl 1): S15-S28, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28196877

RÉSUMÉ

Staphylococcus aureus (Sa) is the leading cause of a variety of bacterial infections ranging from superficial skin infections to invasive and life threatening diseases such as septic bacteremia, necrotizing pneumonia, and endocarditis. The success of Sa as a human pathogen is contributed to its ability to adapt to different environments by changing expression, production, or secretion of virulence factors. Although Sa immune evasion is well-studied, the regulation of virulence factors under different nutrient and growth conditions is still not well understood. Here, we used label-free quantitative mass spectrometry to quantify and compare the Sa exoproteins (i.e. exoproteomes) of master regulator mutants or established reference strains. Different environmental conditions were addressed by growing the bacteria in rich or minimal media at different phases of growth. We observed clear differences in the composition of the exoproteomes depending on the genetic background or growth conditions. The relative abundance of cytotoxins determined in our study correlated well with differences in cytotoxicity measured by lysis of human neutrophils. Our findings demonstrate that label-free quantitative mass spectrometry is a versatile tool for predicting the virulence of bacterial strains and highlights the importance of the experimental design for in vitro studies. Furthermore, the results indicate that label-free proteomics can be used to cluster isolates into groups with similar virulence properties, highlighting the power of label-free quantitative mass spectrometry to distinguish Sa strains.


Sujet(s)
Spectrométrie de masse/méthodes , Granulocytes neutrophiles/microbiologie , Staphylococcus aureus/croissance et développement , Facteurs de virulence/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Cytotoxines/génétique , Cytotoxines/métabolisme , Régulation de l'expression des gènes bactériens , Génotype , Humains , Protéomique/méthodes , Infections à staphylocoques/microbiologie , Staphylococcus aureus/génétique , Staphylococcus aureus/métabolisme , Staphylococcus aureus/pathogénicité , Facteurs de virulence/génétique
7.
Pathog Dis ; 75(1)2017 01 01.
Article de Anglais | MEDLINE | ID: mdl-28104617

RÉSUMÉ

Staphylococcus aureus is an eminent human pathogen that can colonize the human host and cause severe life-threatening illnesses. This bacterium can reside in and infect a wide range of host tissues, ranging from superficial surfaces like the skin to deeper tissues such as in the gastrointestinal tract, heart and bones. Due to its multifaceted lifestyle, S. aureus uses complex regulatory networks to sense diverse signals that enable it to adapt to different environments and modulate virulence. In this minireview, we explore well-characterized environmental and host cues that S. aureus responds to and describe how this pathogen modulates virulence in response to these signals. Lastly, we highlight therapeutic approaches undertaken by several groups to inhibit both signaling and the cognate regulators that sense and transmit these signals downstream.


Sujet(s)
Interactions hôte-pathogène , Infections à staphylocoques/microbiologie , Staphylococcus aureus/physiologie , Animaux , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Métabolisme énergétique/effets des médicaments et des substances chimiques , Humains , Oxydoréduction/effets des médicaments et des substances chimiques , Détection du quorum/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Infections à staphylocoques/traitement médicamenteux , Infections à staphylocoques/métabolisme , Staphylococcus aureus/effets des médicaments et des substances chimiques , Virulence
8.
mSystems ; 1(1)2016.
Article de Anglais | MEDLINE | ID: mdl-27822514

RÉSUMÉ

Hundreds of small RNAs (sRNAs) have been identified in diverse bacterial species, and while the functions of most remain unknown, some regulate key processes, particularly stress responses. The sRNA DicF was identified over 25 years ago as an inhibitor of cell division but since then has remained uncharacterized. DicF consists of 53 nucleotides and is encoded by a gene carried on a prophage (Qin) in the genomes of many Escherichia coli strains. We demonstrated that DicF inhibits cell division via direct base pairing with ftsZ mRNA to repress translation and prevent new synthesis of the bacterial tubulin homolog FtsZ. Systems analysis using computational and experimental methods identified additional mRNA targets of DicF: xylR and pykA mRNAs, encoding the xylose uptake and catabolism regulator and pyruvate kinase, respectively. Genetic analyses showed that DicF directly base pairs with and represses translation of these targets. Phenotypes of cells expressing DicF variants demonstrated that DicF-associated growth inhibition is not solely due to repression of ftsZ, indicating that the physiological consequences of DicF-mediated regulation extend beyond effects on cell division caused by reduced FtsZ synthesis. IMPORTANCE sRNAs are ubiquitous and versatile regulators of bacterial gene expression. A number of well-characterized examples in E. coli are highly conserved and present in the E. coli core genome. In contrast, the sRNA DicF (identified over 20 years ago but remaining poorly characterized) is encoded by a gene carried on a defective prophage element in many E. coli genomes. Here, we characterize DicF in order to better understand how horizontally acquired sRNA regulators impact bacterial gene expression and physiology. Our data confirm the long-hypothesized DicF-mediated regulation of ftsZ, encoding the bacterial tubulin homolog required for cell division. We further uncover DicF-mediated posttranscriptional control of metabolic gene expression. Ectopic production of DicF is highly toxic to E. coli cells, but the toxicity is not attributable to DicF regulation of ftsZ. Further work is needed to reveal the biological roles of and benefits for the host conferred by DicF and other products encoded by defective prophages.

9.
mBio ; 7(3)2016 06 21.
Article de Anglais | MEDLINE | ID: mdl-27329753

RÉSUMÉ

UNLABELLED: Staphylococcus aureus is a formidable human pathogen that uses secreted cytolytic factors to injure immune cells and promote infection of its host. Of these proteins, the bicomponent family of pore-forming leukocidins play critical roles in S. aureus pathogenesis. The regulatory mechanisms governing the expression of these toxins are incompletely defined. In this work, we performed a screen to identify transcriptional regulators involved in leukocidin expression in S. aureus strain USA300. We discovered that a metabolic sensor-regulator, RpiRc, is a potent and selective repressor of two leukocidins, LukED and LukSF-PV. Whole-genome transcriptomics, S. aureus exoprotein proteomics, and metabolomic analyses revealed that RpiRc influences the expression and production of disparate virulence factors. Additionally, RpiRc altered metabolic fluxes in the trichloroacetic acid cycle, glycolysis, and amino acid metabolism. Using mutational analyses, we confirmed and extended the observation that RpiRc signals through the accessory gene regulatory (Agr) quorum-sensing system in USA300. Specifically, RpiRc represses the rnaIII promoter, resulting in increased repressor of toxins (Rot) levels, which in turn negatively affect leukocidin expression. Inactivation of rpiRc phenocopied rot deletion and increased S. aureus killing of primary human polymorphonuclear leukocytes and the pathogenesis of bloodstream infection in vivo. Collectively, our results suggest that S. aureus senses metabolic shifts by RpiRc to differentially regulate the expression of leukocidins and to promote invasive disease. IMPORTANCE: The bicomponent pore-forming leukocidins play pivotal roles in the ability of S. aureus to kill multiple host immune cells, thus enabling this pathogen to have diverse tissue- and species-tropic effects. While the mechanisms of leukocidin-host receptor interactions have been studied in detail, the regulatory aspects of leukocidin expression are less well characterized. Moreover, the expression of the leukocidins is highly modular in vitro, suggesting the presence of regulators other than the known Agr, Rot, and S. aureus exoprotein pathways. Here, we describe how RpiRc, a metabolite-sensing transcription factor, mediates the repression of two specific leukocidin genes, lukED and pvl, which in turn has complex effects on the pathogenesis of S. aureus Our findings highlight the intricacies of leukocidin regulation by S. aureus and demonstrate the involvement of factors beyond traditional virulence factor regulators.


Sujet(s)
Régulation de l'expression des gènes bactériens , Leucocidine/biosynthèse , Protéines de répression/métabolisme , Staphylococcus aureus/génétique , Staphylococcus aureus/pathogénicité , Facteurs de virulence/biosynthèse , Acides aminés/métabolisme , Animaux , Protéines bactériennes/métabolisme , Survie cellulaire , Cellules cultivées , Cycle citrique , Analyse de mutations d'ADN , Modèles animaux de maladie humaine , Délétion de gène , Analyse de profil d'expression de gènes , Glycolyse , Humains , Analyse des flux métaboliques , Métabolome , Souris , Granulocytes neutrophiles/microbiologie , Granulocytes neutrophiles/physiologie , Protéome/analyse , Protéines de répression/génétique , Sepsie/microbiologie , Sepsie/anatomopathologie , Transduction du signal , Staphylococcus aureus/métabolisme , Transactivateurs/métabolisme , Virulence
10.
Science ; 347(6228): 1371-4, 2015 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-25792329

RÉSUMÉ

Base-pairing interactions between nucleic acids mediate target recognition in many biological processes. We developed a super-resolution imaging and modeling platform that enabled the in vivo determination of base pairing-mediated target recognition kinetics. We examined a stress-induced bacterial small RNA, SgrS, which induces the degradation of target messenger RNAs (mRNAs). SgrS binds to a primary target mRNA in a reversible and dynamic fashion, and formation of SgrS-mRNA complexes is rate-limiting, dictating the overall regulation efficiency in vivo. Examination of a secondary target indicated that differences in the target search kinetics contribute to setting the regulation priority among different target mRNAs. This super-resolution imaging and analysis approach provides a conceptual framework that can be generalized to other small RNA systems and other target search processes.


Sujet(s)
Appariement de bases , Imagerie moléculaire/méthodes , Stabilité de l'ARN , ARN messager/composition chimique , Petit ARN non traduit/composition chimique , Endoribonucleases/composition chimique , Endoribonucleases/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme , Cinétique , Mutation , Phosphoenolpyruvate-fructose phosphotransferase/génétique
11.
J Bacteriol ; 195(20): 4620-30, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23935052

RÉSUMÉ

Bacterial dual-function small RNAs regulate gene expression by RNA-RNA base pairing and also code for small proteins. SgrS is a dual-function small RNA in Escherichia coli and Salmonella that is expressed under stress conditions associated with accumulation of sugar-phosphates, and its activity is crucial for growth during stress. The base-pairing function of SgrS regulates a number of mRNA targets, resulting in reduced uptake and enhanced efflux of sugars. SgrS also encodes the SgrT protein, which reduces sugar uptake by a mechanism that is independent of base pairing. While SgrS base-pairing activity has been characterized in detail, little is known about how base pairing and translation of sgrT are coordinated. In the current study, we utilized a series of mutants to determine how translation of sgrT affected the efficiency of base pairing-dependent regulation and vice versa. Mutations that abrogated sgrT translation had minimal effects on base-pairing activity. Conversely, mutations that impaired base-pairing interactions resulted in increased SgrT production. Furthermore, while ectopic overexpression of sgrS mutant alleles lacking only one of the two functions rescued cell growth under stress conditions, the SgrS base-pairing function alone was indispensable for growth rescue when alleles were expressed from the native locus. Collectively, the results suggest that during stress, repression of sugar transporter synthesis via base pairing with sugar transporter mRNAs is the first priority of SgrS. Subsequently, SgrT is made and acts on preexisting transporters. The combined action of these two functions produces an effective stress response.


Sujet(s)
Protéines bactériennes/métabolisme , Régulation de l'expression des gènes bactériens/physiologie , ARN bactérien/métabolisme , Salmonella typhimurium/métabolisme , Séquence d'acides aminés , Protéines bactériennes/génétique , Séquence nucléotidique , Données de séquences moléculaires , Mutation , Salmonella typhimurium/génétique
12.
Nucleic Acids Res ; 41(14): e140, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23716638

RÉSUMÉ

Recent advances in high-throughput RNA sequencing (RNA-seq) have enabled tremendous leaps forward in our understanding of bacterial transcriptomes. However, computational methods for analysis of bacterial transcriptome data have not kept pace with the large and growing data sets generated by RNA-seq technology. Here, we present new algorithms, specific to bacterial gene structures and transcriptomes, for analysis of RNA-seq data. The algorithms are implemented in an open source software system called Rockhopper that supports various stages of bacterial RNA-seq data analysis, including aligning sequencing reads to a genome, constructing transcriptome maps, quantifying transcript abundance, testing for differential gene expression, determining operon structures and visualizing results. We demonstrate the performance of Rockhopper using 2.1 billion sequenced reads from 75 RNA-seq experiments conducted with Escherichia coli, Neisseria gonorrhoeae, Salmonella enterica, Streptococcus pyogenes and Xenorhabdus nematophila. We find that the transcriptome maps generated by our algorithms are highly accurate when compared with focused experimental data from E. coli and N. gonorrhoeae, and we validate our system's ability to identify novel small RNAs, operons and transcription start sites. Our results suggest that Rockhopper can be used for efficient and accurate analysis of bacterial RNA-seq data, and that it can aid with elucidation of bacterial transcriptomes.


Sujet(s)
Algorithmes , Analyse de profil d'expression de gènes , ARN bactérien/composition chimique , Analyse de séquence d'ARN , Régions 5' non traduites , Génome bactérien , Génomique/méthodes , Séquençage nucléotidique à haut débit , Opéron , ARN bactérien/métabolisme , Petit ARN non traduit/composition chimique , Petit ARN non traduit/métabolisme , Alignement de séquences , Logiciel , Transcription génétique
13.
RNA Biol ; 10(3): 337-41, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23392245

RÉSUMÉ

Small regulatory RNAs (sRNAs) are influential post-transcriptional modulators of gene expression in bacteria. They regulate gene expression by base pairing to target mRNAs, leading to inhibition of translation and/or alteration of mRNA stability. Recently, several sRNAs have been discovered to regulate genes encoded in operons. In some cases, these sRNAs regulate all the genes encoded by the polycistronic mRNA (coordinate regulation) while in other cases, only a select subset of cistrons is controlled by the sRNA (discoordinate regulation). In this point of view, mechanisms of regulation and characteristics of sRNA-mRNA interactions involving polycistronic mRNAs are described. The diversity in mechanisms represented by these few characterized examples suggests that we still have much to learn about sRNA regulation of long polycistronic messages.


Sujet(s)
Régulation de l'expression des gènes bactériens , Opéron , ARN bactérien/physiologie , ARN messager/métabolisme , Petit ARN non traduit/physiologie , Appariement de bases , Gènes , Gènes bactériens , Stabilité de l'ARN , ARN messager/génétique
14.
Proc Natl Acad Sci U S A ; 109(40): E2691-8, 2012 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-22988087

RÉSUMÉ

In animal systems, mRNAs subject to posttranscriptional regulation by small RNAs (sRNAs) often possess multiple binding sites with imperfect complementarity to a given sRNA. In contrast, small RNA-mRNA interactions in bacteria and plants typically involve a single binding site. In a previous study, we demonstrated that the Escherichia coli sRNA SgrS base pairs with a site in the coding region of the first gene of a polycistronic message, manXYZ. This interaction was shown to be responsible for translational repression of manX and to contribute to destabilization of the manXYZ mRNA. In the current study, we report that translational repression of the manY and manZ genes by SgrS requires a second binding site located in the manX-manY intergenic region. Pairing at this site can repress translation of manY and manZ even when mRNA degradation is blocked. Base pairing between SgrS and the manX site does not affect translation of manY or manZ. Pairing at both sites is required for optimal SgrS-mediated degradation of the full-length manXYZ mRNA and for a particular stress phenotype. These results suggest that bacterial sRNAs may use target-site multiplicity to enhance the efficiency and stringency of regulation. Moreover, use of multiple binding sites may be particularly important for coordinating regulation of multiple genes encoded in operons.


Sujet(s)
Protéines Escherichia coli/génétique , Escherichia coli/génétique , Régulation de l'expression des gènes/génétique , Biosynthèse des protéines/génétique , ARN messager/métabolisme , Séquences régulatrices de l'acide ribonucléique/génétique , Appariement de bases , Sites de fixation/génétique , ADN intergénique/génétique , Régulation de l'expression des gènes/physiologie , Mutagenèse , Oligonucléotides/génétique , Biosynthèse des protéines/physiologie , Stabilité de l'ARN/physiologie , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , beta-Galactosidase
15.
Biochimie ; 93(11): 1943-9, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21816203

RÉSUMÉ

The importance of small RNA (sRNA) regulators has been recognized across all domains of life. In bacteria, sRNAs typically control the expression of virulence and stress response genes via antisense base pairing with mRNA targets. Originally dubbed "non-coding RNAs," a number of bacterial antisense sRNAs have been found to encode functional proteins. Although very few of these dual-function sRNAs have been characterized, they have been found in both gram-negative and gram-positive organisms. Among the few known examples, the functions and mechanisms of regulation by dual-function sRNAs are variable. Some dual-function sRNAs depend on the RNA chaperone Hfq for base pairing-dependent regulation (riboregulation); this feature appears so far exclusive to gram-negative bacterial sRNAs. Other variations can be found in the spatial organization of the coding region with respect to the riboregulation determinants. How the functions of encoded proteins relate to riboregulation is for the most part not understood. However, in one case it appears that there is physiological redundancy between protein and riboregulation functions. This mini-review focuses on the two best-studied bacterial dual-function sRNAs: RNAIII from Staphylococcus aureus and SgrS from Escherichia coli and includes a discussion of what is known about the structure, function and physiological roles of these sRNAs as well as what questions remain outstanding.


Sujet(s)
Protéines Escherichia coli/métabolisme , Escherichia coli/métabolisme , Régulation de l'expression des gènes bactériens , ARN bactérien/métabolisme , ARN messager/génétique , Petit ARN non traduit/génétique , Staphylococcus aureus/métabolisme , Escherichia coli/génétique , Protéines Escherichia coli/génétique , Évolution moléculaire , Chaperons moléculaires/génétique , Cadres ouverts de lecture/génétique , ARN antisens/génétique , ARN bactérien/composition chimique , ARN bactérien/génétique , Staphylococcus aureus/génétique
16.
J Bacteriol ; 191(14): 4534-45, 2009 Jul.
Article de Anglais | MEDLINE | ID: mdl-19429624

RÉSUMÉ

Azotobacter vinelandii is a soil bacterium related to the Pseudomonas genus that fixes nitrogen under aerobic conditions while simultaneously protecting nitrogenase from oxygen damage. In response to carbon availability, this organism undergoes a simple differentiation process to form cysts that are resistant to drought and other physical and chemical agents. Here we report the complete genome sequence of A. vinelandii DJ, which has a single circular genome of 5,365,318 bp. In order to reconcile an obligate aerobic lifestyle with exquisitely oxygen-sensitive processes, A. vinelandii is specialized in terms of its complement of respiratory proteins. It is able to produce alginate, a polymer that further protects the organism from excess exogenous oxygen, and it has multiple duplications of alginate modification genes, which may alter alginate composition in response to oxygen availability. The genome analysis identified the chromosomal locations of the genes coding for the three known oxygen-sensitive nitrogenases, as well as genes coding for other oxygen-sensitive enzymes, such as carbon monoxide dehydrogenase and formate dehydrogenase. These findings offer new prospects for the wider application of A. vinelandii as a host for the production and characterization of oxygen-sensitive proteins.


Sujet(s)
Azotobacter vinelandii/génétique , ADN bactérien/composition chimique , ADN bactérien/génétique , Génome bactérien , Analyse de séquence d'ADN , Protéines bactériennes/génétique , Séquence nucléotidique , Métabolisme/génétique , Données de séquences moléculaires , Phylogenèse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...