Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Microbiol Mol Biol Rev ; 87(4): e0003622, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38047635

ABSTRACT

SUMMARYAminoglycosides (AGs) are long-known molecules successfully used against Gram-negative pathogens. While their use declined with the discovery of new antibiotics, they are now classified as critically important molecules because of their effectiveness against multidrug-resistant bacteria. While they can efficiently cross the Gram-negative envelope, the mechanism of AG entry is still incompletely understood, although this comprehension is essential for the development of new therapies in the face of the alarming increase in antibiotic resistance. Increasing antibiotic uptake in bacteria is one strategy to enhance effective treatments. This review aims, first, to consolidate old and recent knowledge about AG uptake; second, to explore the connection between AG-dependent bacterial stress and drug uptake; and finally, to present new strategies of potentiation of AG uptake for more efficient antibiotic therapies. In particular, we emphasize on the connection between sugar transport and AG potentiation.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Aminoglycosides/pharmacology , Aminoglycosides/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Gram-Negative Bacteria , Bacteria , Drug Resistance, Multiple, Bacterial
2.
Microbiol Spectr ; 11(6): e0173023, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37861314

ABSTRACT

IMPORTANCE: The RavA-ViaA complex was previously found to sensitize Escherichia coli to aminoglycosides (AGs) in anaerobic conditions, but the mechanism is unknown. AGs are antibiotics known for their high efficiency against Gram-negative bacteria. In order to elucidate how the expression of the ravA-viaA genes increases bacterial susceptibility to aminoglycosides, we aimed at identifying partner functions necessary for increased tolerance in the absence of RavA-ViaA, in Vibrio cholerae. We show that membrane stress response systems Cpx and Zra2 are required in the absence of RavA-ViaA, for the tolerance to AGs and for outer membrane integrity. In the absence of these systems, the ∆ravvia strain's membrane becomes permeable to external agents such as the antibiotic vancomycin.


Subject(s)
Escherichia coli Proteins , Vibrio cholerae , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Aminoglycosides/pharmacology , Aminoglycosides/metabolism , Adenosine Triphosphatases/metabolism
3.
bioRxiv ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37502966

ABSTRACT

The possible active entry of aminoglycosides in bacterial cells has been debated since the development of this antibiotic family. Here we report the identification of their active transport mechanism in Vibrio species. We combined genome-wide transcriptional analysis and fitness screens to identify alterations driven by treatment of V. cholerae with sub-minimum inhibitory concentrations (sub-MIC) of the aminoglycoside tobramycin. RNA-seq data showed downregulation of the small non-coding RNA ncRNA586 during such treatment, while Tn-seq revealed that inactivation of this sRNA was associated with improved fitness in the presence of tobramycin. This sRNA is located near sugar transport genes and previous work on a homologous region in Vibrio tasmaniensis suggested that this sRNA stabilizes gene transcripts for carbohydrate transport and utilization, as well as phage receptors. The role for ncRNA586, hereafter named ctrR, in the transport of both carbohydrates and aminoglycosides, was further investigated. Flow cytometry on cells treated with a fluorescent aminoglycoside confirmed the role of ctrR and of carbohydrate transporters in differential aminoglycoside entry. Despite sequence diversity, ctrR showed functional conservation across the Vibrionales. This system in directly modulated by carbon sources, suggesting regulation by carbon catabolite repression, a widely conserved mechanism in Gram-negative bacteria, priming future research on aminoglycoside uptake by sugar transporters in other bacterial species.

4.
Res Microbiol ; 174(1-2): 103997, 2023.
Article in English | MEDLINE | ID: mdl-36347445

ABSTRACT

Vibrio cholerae N16961 genome encodes 18 type II Toxin/Antitoxin (TA) systems, all but one located inside gene cassettes of its chromosomal superintegron (SI). This study aims to investigate additional TA systems in this genome. We screened for all two-genes operons of uncharacterized function by analyzing previous RNAseq data. Assays on nine candidates, revealed one additional functional type II TA encoded by the VCA0497-0498 operon, carried inside a SI cassette. We showed that VCA0498 antitoxin alone and in complex with VCA0497 represses its own operon promoter. VCA0497-0498 is the second element of the recently identified dhiT/dhiA superfamily uncharacterized type II TA system. RNAseq analysis revealed that another SI cassette encodes a novel type I TA system: VCA0495 gene and its two associated antisense non-coding RNAs, ncRNA495 and ncRNA496. Silencing of both antisense ncRNAs lead to cell death, demonstrating the type I TA function. Both VCA0497 and VCA0495 toxins do not show any homology to functionally characterized toxins, however our preliminary data suggest that their activity may end up in mRNA degradation, directly or indirectly. Our findings increase the TA systems number carried in this SI to 19, preferentially located in its distal end, confirming their importance in this large cassette array.


Subject(s)
Antitoxins , Bacterial Toxins , Vibrio cholerae , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Escherichia coli/metabolism , Antitoxins/genetics , Antitoxins/metabolism , Promoter Regions, Genetic
5.
Elife ; 112022 01 17.
Article in English | MEDLINE | ID: mdl-35037621

ABSTRACT

The plasmid-mediated quinolone resistance (PMQR) genes have been shown to promote high-level bacterial resistance to fluoroquinolone antibiotics, potentially leading to clinical treatment failures. In Escherichia coli, sub-minimum inhibitory concentrations (sub-MICs) of the widely used fluoroquinolones are known to induce the SOS response. Interestingly, the expression of several PMQR qnr genes is controlled by the SOS master regulator, LexA. During the characterization of a small qnrD-plasmid carried in E. coli, we observed that the aminoglycosides become able to induce the SOS response in this species, thus leading to the elevated transcription of qnrD. Our findings show that the induction of the SOS response is due to nitric oxide (NO) accumulation in the presence of sub-MIC of aminoglycosides. We demonstrated that the NO accumulation is driven by two plasmid genes, ORF3 and ORF4, whose products act at two levels. ORF3 encodes a putative flavin adenine dinucleotide (FAD)-binding oxidoreductase which helps NO synthesis, while ORF4 codes for a putative fumarate and nitrate reductase (FNR)-type transcription factor, related to an O2-responsive regulator of hmp expression, able to repress the Hmp-mediated NO detoxification pathway of E. coli. Thus, this discovery, that other major classes of antibiotics may induce the SOS response could have worthwhile implications for antibiotic stewardship efforts in preventing the emergence of resistance.


Subject(s)
Aminoglycosides/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli , Plasmids/genetics , SOS Response, Genetics/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/physiology , Nitric Oxide/metabolism , Quinolones
6.
Microlife ; 3: uqac019, 2022.
Article in English | MEDLINE | ID: mdl-37223353

ABSTRACT

Antimicrobial resistance develops as a major problem in infectious diseases treatment. While antibiotic resistance mechanisms are usually studied using lethal antibiotic doses, lower doses allowing bacterial growth are now considered as factors influencing the development and selection of resistance. Starting with a high-density Tn insertion library in Vibrio cholerae and following its evolution by TN-seq in the presence of subinhibitory concentrations of antibiotics, we discovered that RNA modification genes can have opposite fates, being selected or counter-selected. We, thus have undertaken the phenotypic characterization of 23 transfer RNA (tRNA) and ribosomal RNA (rRNA) modifications deletion mutants, for which growth is globally not affected in the absence of stress. We uncover a specific involvement of different RNA modification genes in the response to aminoglycosides (tobramycin and gentamicin), fluoroquinolones (ciprofloxacin), ß-lactams (carbenicillin), chloramphenicol, and trimethoprim. Our results identify t/rRNA modification genes, not previously associated to any antibiotic resistance phenotype, as important factors affecting the bacterial response to low doses of antibiotics from different families. This suggests differential translation and codon decoding as critical factors involved in the bacterial response to stress.

7.
Cells ; 10(11)2021 11 18.
Article in English | MEDLINE | ID: mdl-34831448

ABSTRACT

Antibiotics are well known drugs which, when present above certain concentrations, are able to inhibit the growth of certain bacteria. However, a growing body of evidence shows that even when present at lower doses (subMIC, for sub-minimal inhibitory concentration), unable to inhibit or affect microbial growth, antibiotics work as signaling molecules, affect gene expression and trigger important bacterial stress responses. However, how subMIC antibiotic signaling interplays with other well-known signaling networks in bacteria (and the consequences of such interplay) is not well understood. In this work, through transcriptomic and genetic approaches, we have explored how quorum-sensing (QS) proficiency of V. cholerae affects this pathogen's response to subMIC doses of the aminoglycoside tobramycin (TOB). We show that the transcriptomic signature of V. cholerae in response to subMIC TOB depends highly on the presence of QS master regulator HapR. In parallel, we show that subMIC doses of TOB are able to negatively interfere with the AI-2/LuxS QS network of V. cholerae, which seems critical for survival to aminoglycoside treatment and TOB-mediated induction of SOS response in this species. This interplay between QS and aminoglycosides suggests that targeting QS signaling may be a strategy to enhance aminoglycoside efficacy in V. cholerae.


Subject(s)
Aminoglycosides/pharmacology , Microbial Viability , Quorum Sensing/drug effects , Vibrio cholerae/physiology , Gene Expression Regulation, Bacterial/drug effects , Microbial Viability/drug effects , Mutation/genetics , Quorum Sensing/genetics , SOS Response, Genetics/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Tobramycin/pharmacology , Transcriptome/genetics , Vibrio cholerae/drug effects , Vibrio cholerae/genetics , Vibrio cholerae/growth & development
8.
PLoS Genet ; 17(10): e1009748, 2021 10.
Article in English | MEDLINE | ID: mdl-34669693

ABSTRACT

Antibiotic resistance has become a major global issue. Understanding the molecular mechanisms underlying microbial adaptation to antibiotics is of keen importance to fight Antimicrobial Resistance (AMR). Aminoglycosides are a class of antibiotics that target the small subunit of the bacterial ribosome, disrupting translational fidelity and increasing the levels of misfolded proteins in the cell. In this work, we investigated the role of VchM, a DNA methyltransferase, in the response of the human pathogen Vibrio cholerae to aminoglycosides. VchM is a V. cholerae specific orphan m5C DNA methyltransferase that generates cytosine methylation at 5'-RCCGGY-3' motifs. We show that deletion of vchM, although causing a growth defect in absence of stress, allows V. cholerae cells to cope with aminoglycoside stress at both sub-lethal and lethal concentrations of these antibiotics. Through transcriptomic and genetic approaches, we show that groESL-2 (a specific set of chaperonin-encoding genes located on the second chromosome of V. cholerae), are upregulated in cells lacking vchM and are needed for the tolerance of vchM mutant to lethal aminoglycoside treatment, likely by fighting aminoglycoside-induced misfolded proteins. Interestingly, preventing VchM methylation of the four RCCGGY sites located in groESL-2 region, leads to a higher expression of these genes in WT cells, showing that the expression of these chaperonins is modulated in V. cholerae by DNA methylation.


Subject(s)
Aminoglycosides/genetics , Chaperonins/genetics , Cytosine/metabolism , DNA Methylation/genetics , DNA/genetics , Vibrio cholerae/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Methyltransferases/genetics
9.
iScience ; 24(10): 103128, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34611612

ABSTRACT

Indole is a molecule proposed to be involved in bacterial signaling. We find that indole secretion is induced by sublethal tobramycin concentrations and increases persistence to aminoglycosides in V. cholerae. Indole transcriptomics showed increased expression of raiA, a ribosome associated factor. Deletion of raiA abolishes the appearance of indole dependent persisters to aminoglycosides, although its overexpression leads to 100-fold increase of persisters, and a reduction in lag phase, evocative of increased active 70S ribosome content, confirmed by sucrose gradient analysis. We propose that, under stress conditions, RaiA-bound inactive 70S ribosomes are stored as "sleeping ribosomes", and are rapidly reactivated upon stress relief. Our results point to an active process of persister formation through ribosome protection during translational stress (e.g., aminoglycoside treatment) and reactivation upon antibiotic removal. Translation is a universal process, and these results could help elucidate a mechanism of persistence formation in a controlled, thus inducible way.

10.
mBio ; 10(4)2019 07 02.
Article in English | MEDLINE | ID: mdl-31266870

ABSTRACT

We have previously identified Vibrio cholerae mutants in which the stress response to subinhibitory concentrations of aminoglycoside is altered. One gene identified, VC1636, encodes a putative DNA/RNA helicase, recently named RadD in Escherichia coli Here we combined extensive genetic characterization and high-throughput approaches in order to identify partners and molecular mechanisms involving RadD. We show that double-strand DNA breaks (DSBs) are formed upon subinhibitory tobramycin treatment in the absence of radD and recBCD and that formation of these DSBs can be overcome by RNase H1 overexpression. Loss of RNase H1, or of the transcription-translation coupling factor EF-P, is lethal in the radD deletion mutant. We propose that R-loops are formed upon sublethal aminoglycoside treatment, leading to the formation of DSBs that can be repaired by the RecBCD homologous recombination pathway, and that RadD counteracts such R-loop accumulation. We discuss how R-loops that can occur upon translation-transcription uncoupling could be the link between tobramycin treatment and DNA break formation.IMPORTANCE Bacteria frequently encounter low concentrations of antibiotics. Active antibiotics are commonly detected in soil and water at concentrations much below lethal concentration. Although sub-MICs of antibiotics do not kill bacteria, they can have a major impact on bacterial populations by contributing to the development of antibiotic resistance through mutations in originally sensitive bacteria or acquisition of DNA from resistant bacteria. It was shown that concentrations as low as 100-fold below the MIC can actually lead to the selection of antibiotic-resistant cells. We seek to understand how bacterial cells react to such antibiotic concentrations using E. coli, the Gram-negative bacterial paradigm, and V. cholerae, the causative agent of cholera. Our findings shed light on the processes triggered at the DNA level by antibiotics targeting translation, how damage occurs, and what the bacterial strategies are to respond to such DNA damage.


Subject(s)
Adenosine Triphosphatases/metabolism , Anti-Bacterial Agents/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , R-Loop Structures , Tobramycin/metabolism , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Anti-Bacterial Agents/pharmacology , DNA, Bacterial/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Microbial Sensitivity Tests , Microbial Viability , Tobramycin/pharmacology , Vibrio cholerae/drug effects , Vibrio cholerae/enzymology
11.
Res Microbiol ; 165(6): 476-80, 2014.
Article in English | MEDLINE | ID: mdl-24946128

ABSTRACT

Low concentrations of aminoglycosides induce the SOS response in Vibrio cholerae but not in Escherichia coli. In order to determine whether a specific factor present in E. coli prevents this induction, we developed a genetic screen where only SOS inducing mutants are viable. We identified the vsr gene coding for the Vsr protein of the very short patch mismatch repair (VSPR) pathway. The effect of mismatch repair (MMR) mutants was also studied. We propose that lesions formed upon aminoglycoside treatment are preferentially repaired by VSPR without SOS induction in E. coli and by MMR when VSPR is impaired.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , DNA Mismatch Repair , DNA Repair Enzymes/metabolism , Endodeoxyribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/physiology , SOS Response, Genetics , DNA Repair Enzymes/genetics , Endodeoxyribonucleases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics
12.
FEMS Microbiol Rev ; 38(6): 1126-45, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24923554

ABSTRACT

The presence of an abnormal amount of single-stranded DNA in the bacterial cell constitutes a genotoxic alarm signal that induces the SOS response, a broad regulatory network found in most bacterial species to address DNA damage. The aim of this review was to point out that beyond being a repair process, SOS induction leads to a very strong but transient response to genotoxic stress, during which bacteria can rearrange and mutate their genome, induce several phenotypic changes through differential regulation of genes, and sometimes acquire characteristics that potentiate bacterial survival and adaptation to changing environments. We review here the causes and consequences of SOS induction, but also how this response can be modulated under various circumstances and how it is connected to the network of other important stress responses. In the first section, we review articles describing the induction of the SOS response at the molecular level. The second section discusses consequences of this induction in terms of DNA repair, changes in the genome and gene expression, and sharing of genomic information, with their effects on the bacteria's life and evolution. The third section is about the fine tuning of this response to fit with the bacteria's 'needs'. Finally, we discuss recent findings linking the SOS response to other stress responses. Under these perspectives, SOS can be perceived as a powerful bacterial strategy against aggressions.


Subject(s)
Bacteria/metabolism , SOS Response, Genetics , Stress, Physiological/physiology , Bacteria/genetics , DNA Repair/physiology , Gene Expression Regulation, Bacterial , Stress, Physiological/genetics
13.
Nucleic Acids Res ; 42(4): 2366-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24319148

ABSTRACT

Sub-inhibitory concentrations (sub-MIC) of antibiotics play a very important role in selection and development of resistances. Unlike Escherichia coli, Vibrio cholerae induces its SOS response in presence of sub-MIC aminoglycosides. A role for oxidized guanine residues was observed, but the mechanisms of this induction remained unclear. To select for V. cholerae mutants that do not induce low aminoglycoside-mediated SOS induction, we developed a genetic screen that renders induction of SOS lethal. We identified genes involved in this pathway using two strategies, inactivation by transposition and gene overexpression. Interestingly, we obtained mutants inactivated for the expression of proteins known to destabilize the RNA polymerase complex. Reconstruction of the corresponding mutants confirmed their specific involvement in induction of SOS by low aminoglycoside concentrations. We propose that DNA lesions formed on aminoglycoside treatment are repaired through the formation of single-stranded DNA intermediates, inducing SOS. Inactivation of functions that dislodge RNA polymerase leads to prolonged stalling on these lesions, which hampers SOS induction and repair and reduces viability under antibiotic stress. The importance of these mechanisms is illustrated by a reduction of aminoglycoside sub-MIC. Our results point to a central role for transcription blocking at DNA lesions in SOS induction, so far underestimated.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/physiology , DNA Helicases/physiology , SOS Response, Genetics , Transcription Factors/physiology , Transcription, Genetic , Vibrio cholerae/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/radiation effects , Gene Deletion , Genes, Bacterial , MutS DNA Mismatch-Binding Protein/genetics , Mutation , Ribonuclease H/metabolism , Tobramycin/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Ultraviolet Rays , Vibrio cholerae/drug effects , Vibrio cholerae/enzymology
14.
PLoS Genet ; 9(4): e1003421, 2013.
Article in English | MEDLINE | ID: mdl-23613664

ABSTRACT

Bacteria encounter sub-inhibitory concentrations of antibiotics in various niches, where these low doses play a key role for antibiotic resistance selection. However, the physiological effects of these sub-lethal concentrations and their observed connection to the cellular mechanisms generating genetic diversification are still poorly understood. It is known that, unlike for the model bacterium Escherichia coli, sub-minimal inhibitory concentrations (sub-MIC) of aminoglycosides (AGs) induce the SOS response in Vibrio cholerae. SOS is induced upon DNA damage, and since AGs do not directly target DNA, we addressed two issues in this study: how sub-MIC AGs induce SOS in V. cholerae and why they do not do so in E. coli. We found that when bacteria are grown with tobramycin at a concentration 100-fold below the MIC, intracellular reactive oxygen species strongly increase in V. cholerae but not in E. coli. Using flow cytometry and gfp fusions with the SOS regulated promoter of intIA, we followed AG-dependent SOS induction. Testing the different mutation repair pathways, we found that over-expression of the base excision repair (BER) pathway protein MutY relieved this SOS induction in V. cholerae, suggesting a role for oxidized guanine in AG-mediated indirect DNA damage. As a corollary, we established that a BER pathway deficient E. coli strain induces SOS in response to sub-MIC AGs. We finally demonstrate that the RpoS general stress regulator prevents oxidative stress-mediated DNA damage formation in E. coli. We further show that AG-mediated SOS induction is conserved among the distantly related Gram negative pathogens Klebsiella pneumoniae and Photorhabdus luminescens, suggesting that E. coli is more of an exception than a paradigm for the physiological response to antibiotics sub-MIC.


Subject(s)
Bacterial Proteins/genetics , DNA Damage/genetics , Escherichia coli/genetics , SOS Response, Genetics , Sigma Factor/genetics , Vibrio cholerae/genetics , Aminoglycosides/pharmacology , DNA Repair/genetics , Escherichia coli/growth & development , Gene Expression Regulation, Bacterial , Oxidative Stress , Reactive Oxygen Species , Tobramycin/pharmacology , Vibrio cholerae/growth & development
15.
Antibiotics (Basel) ; 2(2): 288-315, 2013 May 30.
Article in English | MEDLINE | ID: mdl-27029305

ABSTRACT

The emergence of multi-resistant bacterial strains is a major source of concern and has been correlated with the widespread use of antibiotics. The origins of resistance are intensively studied and many mechanisms involved in resistance have been identified, such as exogenous gene acquisition by horizontal gene transfer (HGT), mutations in the targeted functions, and more recently, antibiotic tolerance through persistence. In this review, we focus on factors leading to integron rearrangements and gene capture facilitating antibiotic resistance acquisition, maintenance and spread. The role of stress responses, such as the SOS response, is discussed.

16.
J Bacteriol ; 194(7): 1659-67, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22287520

ABSTRACT

The human pathogen Vibrio cholerae carries a chromosomal superintegron (SI). The SI contains an array of hundreds of gene cassettes organized in tandem which are stable under conditions when no particular stress is applied to bacteria (such as during laboratory growth). Rearrangements of these cassettes are catalyzed by the activity of the associated integron integrase. Understanding the regulation of integrase expression is pivotal to fully comprehending the role played by this genetic reservoir for bacterial adaptation and its connection with the development of antibiotic resistance. Our previous work established that the integrase is regulated by the bacterial SOS response and that it is induced during bacterial conjugation. Here, we show that transformation, another horizontal gene transfer (HGT) mechanism, also triggers integrase expression through SOS induction, underlining the importance of HGT in genome plasticity. Moreover, we report a new cyclic AMP (cAMP)-cAMP receptor protein (CRP)-dependent regulation mechanism of the integrase, highlighting the influence of the extracellular environment on chromosomal gene content. Altogether, our data suggest an interplay between different stress responses and regulatory pathways for the modulation of the recombinase expression, thus showing how the SI remodeling mechanism is merged into bacterial physiology.


Subject(s)
Bacterial Proteins/genetics , Carbon/metabolism , Genome, Bacterial , Integrases/genetics , SOS Response, Genetics , Transformation, Bacterial , Vibrio cholerae/enzymology , Vibrio cholerae/genetics , Bacterial Proteins/metabolism , Base Sequence , Cyclic AMP Receptor Protein/genetics , Cyclic AMP Receptor Protein/metabolism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Integrases/metabolism , Integrons , Molecular Sequence Data , Vibrio cholerae/metabolism
17.
J Biol Chem ; 286(25): 22372-83, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21531731

ABSTRACT

In bacteria, RuvABC is required for the resolution of Holliday junctions (HJ) made during homologous recombination. The RuvAB complex catalyzes HJ branch migration and replication fork reversal (RFR). During RFR, a stalled fork is reversed to form a HJ adjacent to a DNA double strand end, a reaction that requires RuvAB in certain Escherichia coli replication mutants. The exact structure of active RuvAB complexes remains elusive as it is still unknown whether one or two tetramers of RuvA support RuvB during branch migration and during RFR. We designed an E. coli RuvA mutant, RuvA2(KaP), specifically impaired for RuvA tetramer-tetramer interactions. As expected, the mutant protein is impaired for complex II (two tetramers) formation on HJs, although the binding efficiency of complex I (a single tetramer) is as wild type. We show that although RuvA complex II formation is required for efficient HJ branch migration in vitro, RuvA2(KaP) is fully active for homologous recombination in vivo. RuvA2(KaP) is also deficient at forming complex II on synthetic replication forks, and the binding affinity of RuvA2(KaP) for forks is decreased compared with wild type. Accordingly, RuvA2(KaP) is inefficient at processing forks in vitro and in vivo. These data indicate that RuvA2(KaP) is a separation-of-function mutant, capable of homologous recombination but impaired for RFR. RuvA2(KaP) is defective for stimulation of RuvB activity and stability of HJ·RuvA·RuvB tripartite complexes. This work demonstrates that the need for RuvA tetramer-tetramer interactions for full RuvAB activity in vitro causes specifically an RFR defect in vivo.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Replication , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Movement , Protein Multimerization , Adenosine Triphosphatases/metabolism , DNA Helicases/genetics , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Mutagenesis , Mutation , Protein Stability , Protein Structure, Quaternary
18.
Antimicrob Agents Chemother ; 55(5): 2438-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21300836

ABSTRACT

Antibiotic resistance development has been linked to the bacterial SOS stress response. In Escherichia coli, fluoroquinolones are known to induce SOS, whereas other antibiotics, such as aminoglycosides, tetracycline, and chloramphenicol, do not. Here we address whether various antibiotics induce SOS in Vibrio cholerae. Reporter green fluorescent protein (GFP) fusions were used to measure the response of SOS-regulated promoters to subinhibitory concentrations of antibiotics. We show that unlike the situation with E. coli, all these antibiotics induce SOS in V. cholerae.


Subject(s)
Anti-Bacterial Agents/pharmacology , SOS Response, Genetics/drug effects , Vibrio cholerae/drug effects , Aminoglycosides/pharmacology , Chloramphenicol/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Fluoroquinolones/pharmacology , Mutagenesis , Tetracycline/pharmacology
19.
Microbiol Mol Biol Rev ; 74(4): 570-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21119018

ABSTRACT

Structured forms of DNA with intrastrand pairing are generated in several cellular processes and are involved in biological functions. These structures may arise on single-stranded DNA (ssDNA) produced during replication, bacterial conjugation, natural transformation, or viral infections. Furthermore, negatively supercoiled DNA can extrude inverted repeats as hairpins in structures called cruciforms. Whether they are on ssDNA or as cruciforms, hairpins can modify the access of proteins to DNA, and in some cases, they can be directly recognized by proteins. Folded DNAs have been found to play an important role in replication, transcription regulation, and recognition of the origins of transfer in conjugative elements. More recently, they were shown to be used as recombination sites. Many of these functions are found on mobile genetic elements likely to be single stranded, including viruses, plasmids, transposons, and integrons, thus giving some clues as to the manner in which they might have evolved. We review here, with special focus on prokaryotes, the functions in which DNA secondary structures play a role and the cellular processes giving rise to them. Finally, we attempt to shed light on the selective pressures leading to the acquisition of functions for DNA secondary structures.


Subject(s)
DNA, Bacterial/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA, Viral/genetics , Inverted Repeat Sequences/genetics , DNA Replication , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , DNA, Single-Stranded/chemistry , DNA, Viral/chemistry , DNA, Viral/metabolism , Evolution, Molecular , Nucleic Acid Conformation
20.
PLoS Genet ; 6(10): e1001165, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975940

ABSTRACT

Conjugation is one mechanism for intra- and inter-species horizontal gene transfer among bacteria. Conjugative elements have been instrumental in many bacterial species to face the threat of antibiotics, by allowing them to evolve and adapt to these hostile conditions. Conjugative plasmids are transferred to plasmidless recipient cells as single-stranded DNA. We used lacZ and gfp fusions to address whether conjugation induces the SOS response and the integron integrase. The SOS response controls a series of genes responsible for DNA damage repair, which can lead to recombination and mutagenesis. In this manuscript, we show that conjugative transfer of ssDNA induces the bacterial SOS stress response, unless an anti-SOS factor is present to alleviate this response. We also show that integron integrases are up-regulated during this process, resulting in increased cassette rearrangements. Moreover, the data we obtained using broad and narrow host range plasmids strongly suggests that plasmid transfer, even abortive, can trigger chromosomal gene rearrangements and transcriptional switches in the recipient cell. Our results highlight the importance of environments concentrating disparate bacterial communities as reactors for extensive genetic adaptation of bacteria.


Subject(s)
Bacteria/genetics , DNA, Single-Stranded/genetics , Integrons/genetics , SOS Response, Genetics/genetics , Adaptation, Physiological/genetics , Bacteria/metabolism , Conjugation, Genetic , Drug Resistance, Microbial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Gene Transfer, Horizontal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Lac Operon/genetics , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Time Factors , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...