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










Publication year range
1.
BMC Microbiol ; 24(1): 266, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026145

ABSTRACT

BACKGROUND: Legionella pneumophila is a Gram-negative intracellular bacillus and is the causative agent of a severe form of pneumonia called Legionnaires' disease which accounts for 2-9% of cases of community acquired pneumonia. It produces an extremely large protein belonging to the RTX (Repeats in ToXin) family, called RtxA, and we previously reported that RtxA is transported by a dedicated type 1 secretion system (T1SS) to the cell surface. RTX proteins have been shown to participate in the virulence or biofilm formation of various bacteria, the most studied models being the pore forming hemolysin A (HlyA) of Escherichia coli and the biofilm associated protein LapA of P. fluorescens. LapA localization depends on the enzymatic release by LapD/LapG complex activity. This study aimed to elucidate the dual localization (cell surface associated or released state) of L. pneumophila RTX protein (RtxA) and whether this released versus sequestered state of RtxA plays a role in L. pneumophila virulence. RESULTS: The hereby work reveals that, in vitro, LapG periplasmic protease cleaves RtxA N-terminus in the middle of a di-alanine motif (position 108-109). Consistently, a strain lacking LapG protease maintains RtxA on the cell surface, whereas a strain lacking the c-di-GMP receptor LapD does not exhibit cell surface RtxA because of its continuous cleavage and release, as in the LapA-D-G model of Pseudomonas fluorescens. Interestingly, our data point out a key role of RtxA in enhancing the infection process of amoeba cells, regardless of its location (embedded or released); therefore, this may be the result of a secondary role of this surface protein. CONCLUSIONS: This is the first experimental identification of the cleavage site within the RTX protein family. The primary role of RtxA in Legionella is still questionable as in many other bacterial species, hence it sounds reasonable to propose a major function in biofilm formation, promoting cell aggregation when RtxA is embedded in the outer membrane and facilitating biofilm dispersion in case of RtxA release. The role of RtxA in enhancing the infection process may be a result of its action on host cells (i.e., PDI interaction or pore-formation), and independently of its status (embedded or released).


Subject(s)
Bacterial Proteins , Legionella pneumophila , Legionella pneumophila/pathogenicity , Legionella pneumophila/metabolism , Legionella pneumophila/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Virulence , Bacterial Toxins/metabolism , Biofilms/growth & development , Legionnaires' Disease/microbiology , Type I Secretion Systems/metabolism , Type I Secretion Systems/genetics , Cell Membrane/metabolism
2.
Front Cell Infect Microbiol ; 13: 1219233, 2023.
Article in English | MEDLINE | ID: mdl-37600942

ABSTRACT

Bacterial persisters are a transient subpopulation of non-growing, antibiotic-tolerant cells. There is increasing evidence that bacterial persisters play an important role in treatment failure leading to recurring infections and promoting the development of antibiotic resistance. Current research reveals that recurring legionellosis is often the result of relapse rather than reinfection and suggests that the mechanism of bacterial persistence may play a role. The development of single-cell techniques such as the Timerbac system allows us to identify potential persister cells and investigate their physiology. Here, we tested the persister forming capacity of 7 pairs of Legionella pneumophila (Lp) clinical isolates, with isolate pairs corresponding to two episodes of legionellosis in the same patient. We distinguished non-growing subpopulations from their replicating counterparts during infection in an amoeba model. Imaging flow cytometry allowed us to identify single non-growing bacteria within amoeba cells 17 h post-infection, thus corresponding to this subpopulation of potential persister cells. Interestingly the magnitude of this subpopulation varies between the 7 pairs of Lp clinical isolates. Biphasic killing kinetics using ofloxacin stress confirmed the persister development capacity of ST1 clinical isolates, highlighting enhanced persister formation during the host cell infection. Thus, persister formation appears to be strain or ST (sequence type) dependent. Genome sequence analysis was carried out between ST1 clinical isolates and ST1 Paris. No genetic microevolution (SNP) linked to possible increase of persistence capacity was revealed among all the clones tested, even in clones issued from two persistence cycle experiments, confirming the transient reversible phenotypic status of persistence. Treatment failure in legionellosis is a serious issue as infections have a 5-10% mortality rate, and investigations into persistence in a clinical context and the mechanisms involved may allow us to combat this issue.


Subject(s)
Legionella pneumophila , Legionellosis , Humans , Legionella pneumophila/genetics , Reinfection , Anti-Bacterial Agents/pharmacology , Clone Cells
3.
Virulence ; 13(1): 160-173, 2022 12.
Article in English | MEDLINE | ID: mdl-35030980

ABSTRACT

Legionnaires' Disease (LD) is a severe pneumonia mainly caused in Europe by Legionella pneumophila serogroup 1 (Lp1). Sequence-based typing methods reveal that some sequence types (ST) are overrepresented in clinical samples such as ST1 and ST47, suggesting that some strains are more fit for infection than others. In the present study, a collection of 108 Lp1 clinical isolates were used to evaluate the strain-dependent immune responses from human macrophages. Clinical Lp1 isolates induced differential TNFα secretion from macrophages. ST1 isolates induced a significantly higher TNF-α secretion than non-ST1, whereas ST47 isolates induced a significantly lower TNF-α secretion than non-ST47 isolates. ST1 isolates induced a significantly higher cell death than ST47 isolates evaluated by lactate dehydrogenase activity (cytotoxicity) and caspase-3 activity (apoptosis). Treatment of macrophages with anti-TNF-α antibodies significantly reduced the cell death in macrophages infected with ST1 or ST47 strains. The TNF-α secretion was neither explained by a differential bacterial replication nor by the number or type (bystander or infected) of TNF-α producing cells following infection but by a differential response from macrophages. The Paris ST1 reference strain elicited a significantly higher TNF-α gene transcription and a higher induction of NF-κB signaling pathway than the Lorraine ST47 reference strain.Clinical Lp1 isolates induce a diverse immune response and cell death, which could be related to the genotype. The two predominant sequence-types ST1 and ST47 trigger opposite inflammatory response that could be related to the host susceptibility.


Subject(s)
Legionella pneumophila , Legionnaires' Disease , Genotype , Humans , Legionella pneumophila/genetics , Legionnaires' Disease/microbiology , Macrophages , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factor-alpha/genetics
4.
Methods Mol Biol ; 1921: 93-105, 2019.
Article in English | MEDLINE | ID: mdl-30694487

ABSTRACT

Studying bacterial physiology and pathogenesis often requires isolation of targeted mutants. From the early days of bacterial genetics, many genetic tools have been developed to achieve this goal in a lot of bacteria species, and a major key is to be able to manipulate the targeted genome region with a minimum impact on the rest of the genome. Here, we described a two-step protocol relevant in Legionella pneumophila. This efficient two-step protocol uses the natural transformability of L. pneumophila and linear DNA fragments as substrates for recombination without the necessity of intermediate hosts to amplify targeted DNA. Based on a suicide cassette strategy, this genetic toolbox enables to generate clean scar-free deletions, single-nucleotide mutation, transcriptional or translational fusions, as well as insertion at any chosen place in L. pneumophila chromosome, therefore enabling multiple mutations with no need of multiple selection markers.


Subject(s)
Gene Editing , Legionella pneumophila/physiology , Legionnaires' Disease/microbiology , Gene Editing/methods , Mutagenesis, Insertional , Recombination, Genetic , Sequence Deletion , Transformation, Bacterial
5.
Int J Antimicrob Agents ; 50(5): 684-689, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28782709

ABSTRACT

Legionnaires' disease is a severe pneumonia mainly caused by Legionella pneumophila that is treated by antibiotics. The purpose of this study was to describe the susceptibility of clinical strains of L. pneumophila to eight antibiotics used for treatment of legionellosis. The minimum inhibitory concentrations (MICs) of 109 well-characterised clinical strains of L. pneumophila serogroup 1 were determined by the broth microdilution method without charcoal and were compared with antibiotic-resistant strains selected in vitro. All strains were inhibited by low concentrations of fluoroquinolones, macrolides and rifampicin. The epidemiological cut-off values (ECOFFs) were 0.064 mg/L for ciprofloxacin, 0.064 mg/L for moxifloxacin, 0.032 mg/L for levofloxacin, 1 mg/L for erythromycin, 2 mg/L for azithromycin, 0.064 mg/L for clarithromycin, 2 mg/L for doxycycline and 0.001 mg/L for rifampicin. However, MIC distributions revealed a subpopulation of strains displaying reduced susceptibility to some macrolides (especially azithromycin), which correlated with the presence of the lpeAB genes encoding a macrolide efflux pump found specifically in sequence type (ST) ST1, ST701 and closely related STs. Thus, all isolates could be considered susceptible to the tested antibiotics, although macrolides were less active against some strains harbouring a specific efflux system.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Genes, Bacterial , Legionella pneumophila/drug effects , Legionella pneumophila/genetics , Macrolides/pharmacology , Fluoroquinolones/pharmacology , Humans , Legionella pneumophila/isolation & purification , Legionnaires' Disease/microbiology , Microbial Sensitivity Tests , Rifampin/pharmacology
6.
BMC Microbiol ; 17(1): 126, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545445

ABSTRACT

BACKGROUND: Nitroreductases, NAD(P)H dependent flavoenzymes, are found in most of bacterial species. Even if Enterococcus faecalis strains seems to present such activity because of their sensitivity to nitrofurans, no enzyme has been described. Nitroreductases were separated of others reductases due to their capacity to reduce nitro compounds. They are further classified based on their preference in cofactor: NADH and/or NADPH. However, recently, azoreductases have been studied for their strong activity on nitro compounds, especially nitro pro-drugs. This result suggests a crossing in azo and nitro reductase activities. For the moment, no nitroreductase was demonstrated to possess azoreductase activity. But due to sequence divergence and activity specificity linked to substrates, activity prediction is not evident and biochemical characterisation remains necessary. Identifying enzymes active on these two classes of compounds: azo and nitro is of interest to consider a common physiological role. RESULTS: Four putative nitroreductases, EF0404, EF0648, EF0655 and EF1181 from Enterococcus faecalis V583 were overexpressed as his-tagged recombinant proteins in Escherichia coli and purified following a native or a denaturing/renaturing protocol. EF0648, EF0655 and EF1181 showed nitroreductase activity and their cofactor preferences were in agreement with their protein sequence phylogeny. EF0404 showed both nitroreductase and azoreductase activity. Interestingly, the biochemical characteristics (substrate and cofactor specificity) of EF0404 resembled the properties of the known azoreductase AzoA. But its sequence matched within nitroreductase group, the same as EF0648. CONCLUSIONS: We here demonstrate nitroreductase activity of the putative reductases identified in the Enterococcus faecalis V583 genome. We identified the first nitroreductase able to reduce directly an azo compound, while its protein sequence is close to others nitroreductases. Consequently, it highlights the difficulty in classifying these enzymes solely on the basis of protein sequence alignment and hereby the necessity to experimentally demonstrate the activity. The results provide additional data to consider a broader functionality of these reductases.


Subject(s)
Enterococcus faecalis/enzymology , NADH, NADPH Oxidoreductases/isolation & purification , NADH, NADPH Oxidoreductases/metabolism , Nitroreductases/isolation & purification , Nitroreductases/metabolism , Amino Acid Sequence , Azo Compounds/metabolism , DNA, Bacterial/genetics , Enterococcus faecalis/genetics , Enzyme Assays , Escherichia coli/genetics , Genetic Vectors , Genome, Bacterial , NAD/metabolism , NADH, NADPH Oxidoreductases/classification , NADH, NADPH Oxidoreductases/genetics , NADP/metabolism , Nitroreductases/classification , Nitroreductases/genetics , Oxidation-Reduction , Oxidoreductases/metabolism , Phylogeny , Sequence Alignment , Substrate Specificity
7.
J Antimicrob Chemother ; 72(5): 1327-1333, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28137939

ABSTRACT

Objectives: A previous study on 12 in vitro -selected azithromycin-resistant Legionella pneumophila lineages showed that ribosomal mutations were major macrolide resistance determinants. In addition to these mechanisms that have been well described in many species, mutations upstream of lpeAB operon, homologous to acrAB in Escherichia coli , were identified in two lineages. In this study, we investigated the role of LpeAB and of these mutations in macrolide resistance of L. pneumophila . Methods: The role of LpeAB was studied by testing the antibiotic susceptibility of WT, deleted and complemented L. pneumophila Paris strains. Translational fusion experiments using GFP as a reporter were conducted to investigate the consequences of the mutations observed in the upstream sequence of lpeAB operon. Results: We demonstrated the involvement of LpeAB in an efflux pump responsible for a macrolide-specific reduced susceptibility of L. pneumophila Paris strain. Mutations in the upstream sequence of lpeAB operon were associated with an increased protein expression. Increased expression was also observed under sub-inhibitory macrolide concentrations in strains with both mutated and WT promoting regions. Conclusions: LpeAB are components of an efflux pump, which is a macrolide resistance determinant in L. pneumophila Paris strain. Mutations observed in the upstream sequence of lpeAB operon in resistant lineages led to an overexpression of this efflux pump. Sub-inhibitory concentrations of macrolides themselves participated in upregulating this efflux and could constitute a first step in the acquisition of a high macrolide resistance level.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Legionella pneumophila/drug effects , Macrolides/pharmacology , Membrane Transport Proteins/genetics , Azithromycin , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Erythromycin/pharmacology , Genes, Bacterial , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Mutation , Operon , RNA, Ribosomal, 23S
9.
mBio ; 6(3): e00354-15, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25944859

ABSTRACT

UNLABELLED: Legionella pneumophila, the etiological agent of legionellosis, replicates within phagocytic cells. Crucial to biogenesis of the replicative vacuole is the Dot/Icm type 4 secretion system, which translocates a large number of effectors into the host cell cytosol. Among them is LegK2, a protein kinase that plays a key role in Legionella infection. Here, we identified the actin nucleator ARP2/3 complex as a target of LegK2. LegK2 phosphorylates the ARPC1B and ARP3 subunits of the ARP2/3 complex. LegK2-dependent ARP2/3 phosphorylation triggers global actin cytoskeleton remodeling in cells, and it impairs actin tail formation by Listeria monocytogenes, a well-known ARP2/3-dependent process. During infection, LegK2 is addressed to the Legionella-containing vacuole surface and inhibits actin polymerization on the phagosome, as revealed by legK2 gene inactivation. Consequently, LegK2 prevents late endosome/lysosome association with the phagosome and finally contributes to remodeling of the bacterium-containing phagosome into a replicative niche. The inhibition of actin polymerization by LegK2 and its effect on endosome trafficking are ARP2/3 dependent since it can be phenocopied by a specific chemical inhibitor of the ARP2/3 complex. Thus, LegK2-ARP2/3 interplay highlights an original mechanism of bacterial virulence with an unexpected role in local actin remodeling that allows bacteria to control vesicle trafficking in order to escape host defenses. IMPORTANCE: Deciphering the individual contribution of each Dot/Icm type 4 secretion system substrate to the intracellular life-style of L. pneumophila remains the principal challenge in understanding the molecular basis of Legionella virulence. Our finding that LegK2 is a Dot/Icm effector that inhibits actin polymerization on the Legionella-containing vacuole importantly contributes to the deciphering of the molecular mechanisms evolved by Legionella to counteract the endocytic pathway. Indeed, our results highlight the essential role of LegK2 in preventing late endosomes from fusing with the phagosome. More generally, this work is the first demonstration of local actin remodeling as a mechanism used by bacteria to control organelle trafficking. Further, by characterizing the role of the bacterial protein kinase LegK2, we reinforce the concept that posttranslational modifications are key strategies used by pathogens to evade host cell defenses.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Phagosomes/metabolism , Phagosomes/microbiology , Protein Kinases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endosomes/metabolism , Legionella pneumophila/genetics , Lysosomes/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Transport , Vacuoles/microbiology
10.
BMC Microbiol ; 15: 40, 2015 Feb 21.
Article in English | MEDLINE | ID: mdl-25885688

ABSTRACT

BACKGROUND: One proposed mechanism for assembly of secreted heteropolysaccharides by many Gram positive bacteria relies on the coordinated action of a polymerization complex through reversible phosphorylation events. The role of the tyrosine protein kinase transmembrane modulator is, however, not well understood. RESULTS: The protein sequences deduced from the wzb, wzd and wze genes from Lactobacillus rhamnosus ATCC 9595 and RW-9595 M contain motifs also found in corresponding proteins CpsB, CpsC and CpsD from Streptococcus pneumoniae D39 (serotype 2). Use of an anti-phosphotyrosine antibody demonstrated that both Wzd and Wze can be found in tyrosine phosphorylated form. When tyrosine 266 was mutated to phenylalanine, WzdY266F showed slightly less phosphorylated protein than those produced by using eight other tyrosine mutated Wzd genes, when expressed along with Wze and Wzb in Lactococcus lactis subsp. cremoris MG1363. In order to demonstrate the importance of ATP for the interactions among these proteins, native and fusion Wzb, Wzd and Wze proteins were expressed and purified from Escherichia coli cultures. The modulator protein, Wzd, binds with the phosphotyrosine kinase Wze, irrespective of its phosphorylation status. However, Wze attained a higher phosphorylation level after interacting with phosphorylated Wzd in the presence of 10 mM ATP. This highly phosphorylated Wze did not remain in close association with phosphorylated Wzd. CONCLUSION: The Wze tyrosine kinase protein of Lactobacillus rhamnosus thus carries out tyrosine phosphorylation of Wzd in addition to auto- and trans- phosphorylation of the kinase itself.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Lacticaseibacillus rhamnosus/physiology , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Lacticaseibacillus rhamnosus/genetics , Lacticaseibacillus rhamnosus/metabolism , Phosphorylation , Protein Binding , Protein Interaction Mapping
11.
J Bacteriol ; 197(3): 563-71, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422301

ABSTRACT

Legionella pneumophila is a Gram-negative pathogen found mainly in water, either in a free-living form or within infected protozoans, where it replicates. This bacterium can also infect humans by inhalation of contaminated aerosols, causing a severe form of pneumonia called legionellosis or Legionnaires' disease. The involvement of type II and IV secretion systems in the virulence of L. pneumophila is now well documented. Despite bioinformatic studies showing that a type I secretion system (T1SS) could be present in this pathogen, the functionality of this system based on the LssB, LssD, and TolC proteins has never been established. Here, we report the demonstration of the functionality of the T1SS, as well as its role in the infectious cycle of L. pneumophila. Using deletion mutants and fusion proteins, we demonstrated that the repeats-in-toxin protein RtxA is secreted through an LssB-LssD-TolC-dependent mechanism. Moreover, fluorescence monitoring and confocal microscopy showed that this T1SS is required for entry into the host cell, although it seems dispensable to the intracellular cycle. Together, these results underline the active participation of L. pneumophila, via its T1SS, in its internalization into host cells.


Subject(s)
Bacterial Secretion Systems , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Membrane Proteins/metabolism , Virulence Factors/metabolism , Endocytosis , Gene Knockout Techniques , Humans , Legionella pneumophila/genetics , Membrane Proteins/genetics , Monocytes/microbiology , U937 Cells , Virulence
12.
Infect Immun ; 82(3): 1222-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379287

ABSTRACT

Legionella pneumophila is an intracellular pathogen which replicates within protozoan cells and can accidently infect alveolar macrophages, causing an acute pneumonia in humans. The second messenger cyclic di-GMP (c-di-GMP) has been shown to play key roles in the regulation of various bacterial processes, including virulence. While investigating the function of the 22 potential c-di-GMP-metabolizing enzymes of the L. pneumophila Lens strain, we found three that directly contribute to its ability to infect both protozoan and mammalian cells. These three enzymes display diguanylate cyclase (Lpl0780), phosphodiesterase (Lpl1118), and bifunctional diguanylate cyclase/phosphodiesterase (Lpl0922) activities, which are all required for the survival and intracellular replication of L. pneumophila. Mutants with deletions of the corresponding genes are efficiently taken up by phagocytic cells but are partially defective for the escape of the Legionella-containing vacuole (LCV) from the host degradative endocytic pathway and result in lower survival. In addition, Lpl1118 is required for efficient endoplasmic reticulum recruitment to the LCV. Trafficking and biogenesis of the LCV are dependent upon the orchestrated actions of several type 4 secretion system Dot/Icm effectors proteins, which exhibit differentially altered translocation in the three mutants. While translocation of some effectors remained unchanged, others appeared over- and undertranslocated. A general translocation offset of the large repertoire of Dot/Icm effectors may be responsible for the observed defects in the trafficking and biogenesis of the LCV. Our results suggest that L. pneumophila uses cyclic di-GMP signaling to fine-tune effector delivery and ensure effective evasion of the host degradative pathways and establishment of a replicative vacuole.


Subject(s)
Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Legionella pneumophila/metabolism , Legionnaires' Disease/metabolism , Cell Line, Tumor , Cyclic GMP/metabolism , Endocytosis/physiology , Endoplasmic Reticulum/metabolism , Escherichia coli Proteins/metabolism , Humans , Macrophages/metabolism , Phagocytes/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/metabolism , Protein Transport/physiology , Signal Transduction/physiology , U937 Cells , Virulence/physiology
13.
BMC Microbiol ; 13: 17, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23350929

ABSTRACT

BACKGROUND: Several cases of legionellosis have been diagnosed in the same French thermal spa in 1986, 1994 and 1997. L. pneumophila serogroup 1 (Lp1) strains have been isolated from several patients, but the source of contamination was not identified despite the presence of different Lp1 in water samples of the three natural springs feeding the spa at this period. RESULTS: Our strategy was to investigate L. pneumophila (Lp) strains from natural biofilms developed in a sulphur-rich warm spring of this contaminated site. Biofilm analysis revealed the presence of three Lp serogroups (Lp1, Lp10 and Lp12). Surprisingly, Lp10 and Lp12 were not reported in the previous described studies from water samples. Besides, the new seven Lp1 we isolated exhibit a high molecular diversity and have been differentiated in five classes according to their DNA genome patterns obtained by PFGE and mip sequences. It must be noted that these DNA patterns are original and unknown in databases. Interestingly, the 27 Lp environmental strains we isolated display a higher cytotoxicity and virulence towards the amoeba Acanthamoeba castellanii than those of known Lp1 epidemic strains. CONCLUSION: The characteristics of Legionella pneumophila Lp1 strains isolated from the warm spring are in agreement with their presence in biofilms and their probable long-term persistence in this ecosystem.


Subject(s)
Biofilms , Genetic Variation , Hot Springs/microbiology , Legionella pneumophila/classification , Legionella pneumophila/physiology , Acanthamoeba castellanii/drug effects , Acanthamoeba castellanii/microbiology , Bacterial Toxins/toxicity , DNA, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , France , Humans , Legionella pneumophila/genetics , Legionella pneumophila/isolation & purification , Molecular Typing
14.
J Biol Chem ; 286(36): 31136-44, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21757706

ABSTRACT

A significant part of bacterial two-component system response regulators contains effector domains predicted to be involved in metabolism of bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP), a second messenger that plays a key role in many physiological processes. The intracellular level of c-di-GMP is controlled by diguanylate cyclase and phosphodiesterases activities associated with GGDEF and EAL domains, respectively. The Legionella pneumophila Lens genome displays 22 GGDEF/EAL domain-encoding genes. One of them, lpl0329, encodes a protein containing a two-component system receiver domain and both GGDEF and EAL domains. Here, we demonstrated that the GGDEF and EAL domains of Lpl0329 are both functional and lead to simultaneous synthesis and hydrolysis of c-di-GMP. Moreover, these two opposite activities are finely regulated by Lpl0329 phosphorylation due to the atypical histidine kinase Lpl0330. Indeed, Lpl0330 was found to autophosphorylate on a histidine residue in an atypical H box, which is conserved in various bacteria species and thus defines a new histidine kinase subfamily. Lpl0330 also catalyzes the phosphotransferase to Lpl0329, which results in a diguanylate cyclase activity decrease whereas phosphodiesterase activity remains efficient. Altogether, these data present (i) a new histidine kinase subfamily based on the conservation of an original H box that we named HGN H box, and (ii) the first example of a bifunctional enzyme that modulates synthesis and turnover of c-di-GMP in response to phosphorylation of its receiver domain.


Subject(s)
Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/metabolism , Legionella pneumophila/enzymology , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/metabolism , Protein Kinases/physiology , Bacterial Proteins , Cyclic GMP/biosynthesis , Genes, Bacterial , Phosphorylation
15.
Microbiology (Reading) ; 157(Pt 6): 1612-1619, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21393368

ABSTRACT

Lactococcus lactis is known to take up extracellular peptides via at least three distinct peptide transporters. The well-described oligopeptide transporter Opp alone is able to ensure the growth of L. lactis in milk, while the di- and tripeptide transporter DtpT is involved in a peptide-dependent signalling mechanism. The oligopeptide Opt transporter displays two peptide-binding proteins, OptA and OptS. We previously demonstrated that OptA-dependent transport is dedicated to nutritional peptides, as an optABCDF mutant (of a strain devoid of Opp) has an impaired capacity to grow in milk. Using isogenic peptide transport mutants, this study shows that biosynthesis of the Opt transporter is much less sensitive to downregulation that is dependent on extracellular peptides taken up by DtpT than is Opp biosynthesis; this peptide-dependent regulation relies on the transcriptional repressor CodY. We demonstrate the dual function of the Opt system; while OptA contributes to the bacterial nutrition during growth in milk, OptS is involved in the transport of signalling peptides derived from milk and controlling opp expression. So, these results shed new light on the peptide-dependent regulation relying on two peptide transporters with different specificities: DtpT and Opt (via OptS).


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Oligopeptides/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Lactococcus lactis/genetics , Lactococcus lactis/growth & development , Lactococcus lactis/metabolism , Lactococcus lactis/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Milk/microbiology , Mutation , Signal Transduction , Substrate Specificity
16.
Infect Immun ; 79(5): 1936-50, 2011 May.
Article in English | MEDLINE | ID: mdl-21321072

ABSTRACT

Legionella pneumophila is the etiological agent of Legionnaires' disease. Crucial to the pathogenesis of this intracellular pathogen is its ability to subvert host cell defenses, permitting intracellular replication in specialized vacuoles within host cells. The Dot/Icm type IV secretion system (T4SS), which translocates a large number of bacterial effectors into host cell, is absolutely required for rerouting the Legionella phagosome. Many Legionella effectors display distinctive eukaryotic domains, among which are protein kinase domains. In silico analysis and in vitro phosphorylation assays identified five functional protein kinases, LegK1 to LegK5, encoded by the epidemic L. pneumophila Lens strain. Except for LegK5, the Legionella protein kinases are all T4SS effectors. LegK2 plays a key role in bacterial virulence, as demonstrated by gene inactivation. The legK2 mutant containing vacuoles displays less-efficient recruitment of endoplasmic reticulum markers, which results in delayed intracellular replication. Considering that a kinase-dead substitution mutant of legK2 exhibits the same virulence defects, we highlight here a new molecular mechanism, namely, protein phosphorylation, developed by L. pneumophila to establish a replicative niche and evade host cell defenses.


Subject(s)
Bacterial Secretion Systems/genetics , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Protein Kinases/genetics , Amino Acid Sequence , Animals , Endoplasmic Reticulum/enzymology , Intracellular Space/enzymology , Legionella pneumophila/enzymology , Mice , Molecular Sequence Data , Protein Kinases/metabolism , Sequence Alignment , Virulence
17.
PLoS One ; 4(11): e7732, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19888467

ABSTRACT

Pneumonia associated with Iegionnaires's disease is initiated in humans after inhalation of contaminated aerosols. In the environment, Legionella pneumophila is thought to survive and multiply as an intracellular parasite within free-living amoeba. In the genome of L. pneumophila Lens, we identified a unique gene, tolC, encoding a protein that is highly homologous to the outer membrane protein TolC of Escherichia coli. Deletion of tolC by allelic exchange in L. pneumophila caused increased sensitivity to various drugs. The complementation of the tolC mutation in trans restored drug resistance, indicating that TolC is involved in multi-drug efflux machinery. In addition, deletion of tolC caused a significant attenuation of virulence towards both amoebae and macrophages. Thus, the TolC protein appears to play a crucial role in virulence which could be mediated by its involvement in efflux pump mechanisms. These findings will be helpful in unraveling the pathogenic mechanisms of L. pneumophila as well as in developing new therapeutic agents affecting the efflux of toxic compounds.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/physiology , Drug Resistance, Multiple , Gene Expression Regulation, Bacterial , Legionella pneumophila/metabolism , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/physiology , Acanthamoeba castellanii/microbiology , Alleles , Gene Deletion , Genetic Complementation Test , Humans , Legionella pneumophila/pathogenicity , Microbial Sensitivity Tests , Models, Genetic , Plasmids/metabolism , Reactive Oxygen Species , U937 Cells
18.
BMC Biochem ; 9: 10, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-18387182

ABSTRACT

BACKGROUND: Reversible phosphorylation events within a polymerisation complex have been proposed to modulate capsular polysaccharide synthesis in Streptococcus pneumoniae. Similar phosphatase and kinase genes are present in the exopolysaccharide (EPS) biosynthesis loci of numerous lactic acid bacteria genomes. RESULTS: The protein sequence deduced from the wzb gene in Lactobacillus rhamnosus ATCC 9595 reveals four motifs of the polymerase and histidinol phosphatase (PHP) superfamily of prokaryotic O-phosphatases. Native and modified His-tag fusion Wzb proteins were purified from Escherichia coli cultures. Extracts showed phosphatase activity towards tyrosine-containing peptides. The purified fusion protein Wzb was active on p-nitrophenyl-phosphate (pNPP), with an optimal activity in presence of bovine serum albumin (BSA 1%) at pH 7.3 and a temperature of 75 degrees C. At 50 degrees C, residual activity decreased to 10 %. Copper ions were essential for phosphatase activity, which was significantly increased by addition of cobalt. Mutated fusion Wzb proteins exhibited reduced phosphatase activity on p-nitrophenyl-phosphate. However, one variant (C6S) showed close to 20% increase in phosphatase activity. CONCLUSION: These characteristics reveal significant differences with the manganese-dependent CpsB protein tyrosine phosphatase described for Streptococcus pneumoniae as well as with the polysaccharide-related phosphatases of Gram negative bacteria.


Subject(s)
Bacterial Proteins/metabolism , Lactobacillus/enzymology , Mutagenesis, Site-Directed/methods , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Catalysis/drug effects , Cobalt/pharmacology , Electrophoresis, Polyacrylamide Gel , Histidinol-Phosphatase/genetics , Histidinol-Phosphatase/metabolism , Hydrogen-Ion Concentration , Lactobacillus/genetics , Molecular Sequence Data , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Temperature , Vanadates/pharmacology
19.
Appl Environ Microbiol ; 69(6): 3377-84, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12788739

ABSTRACT

Belonging to the subtilase family, the cell surface proteinase (CSP) PrtB of Lactobacillus delbrueckii subsp. bulgaricus differs from other CSPs synthesized by lactic acid bacteria. Expression of the prtB gene under its own promoter was shown to complement the proteinase-deficient strain MG1363 (PrtP(-) PrtM(-)) of Lactococcus lactis subsp. cremoris. Surprisingly, the maturation process of PrtB, unlike that of lactococcal CSP PrtPs, does not require a specific PrtM-like chaperone. The carboxy end of PrtB was previously shown to be different from the consensus anchoring region of other CSPs and exhibits an imperfect duplication of 59 amino acids with a high lysine content. By using a deletion strategy, the removal of the last 99 amino acids, including the degenerated anchoring signal (LPKKT), was found to be sufficient to release a part of the truncated PrtB into the culture medium and led to an increase in PrtB activity. This truncated PrtB is still active and enables L. lactis MG1363 to grow in milk supplemented with glucose. By contrast, deletion of the last 806 amino acids of PrtB led to the secretion of an inactive proteinase. Thus, the utmost carboxy end of PrtB is involved in attachment to the bacterial cell wall. Proteinase PrtB constitutes a powerful tool for cell surface display of heterologous proteins like antigens.


Subject(s)
Bacterial Proteins , Cell Wall/metabolism , Endopeptidases/chemistry , Lactobacillus/enzymology , Lactococcus lactis/enzymology , Amino Acid Sequence , Animals , Caseins/metabolism , Culture Media , Endopeptidases/genetics , Endopeptidases/metabolism , Gene Deletion , Lactobacillus/genetics , Lactococcus lactis/genetics , Lactococcus lactis/growth & development , Milk/microbiology , Molecular Sequence Data , Sequence Analysis, DNA , Substrate Specificity
20.
J Biol Chem ; 278(17): 14832-40, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12590143

ABSTRACT

The specific oligopeptide transport system Opp is essential for growth of Lactococcus lactis in milk. We examined the biodiversity of oligopeptide transport specificity in the L. lactis species. Six strains were tested for (i) consumption of peptides during growth in a chemically defined medium and (ii) their ability to transport these peptides. Each strain demonstrated some specific preferences for peptide utilization, which matched the specificity of peptide transport. Sequencing of the binding protein OppA in some strains revealed minor differences at the amino acid level. The differences in specificity were used as a tool to unravel the role of the binding protein in transport specificity. The genes encoding OppA in four strains were cloned and expressed in L. lactis MG1363 deleted for its oppA gene. The substrate specificity of these engineered strains was found to be similar to that of the L. lactis MG1363 parental strain, whichever oppA gene was expressed. In situ binding experiments demonstrated the ability of OppA to interact with non-transported peptides. Taken together, these results provide evidence for a new concept. Despite that fact that OppA is essential for peptide transport, it is not the (main) determinant of peptide transport specificity in L. lactis.


Subject(s)
Carrier Proteins/genetics , Lactococcus lactis/metabolism , Oligopeptides/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Amino Acid Sequence , Bacterial Proteins , Carrier Proteins/physiology , Lactococcus lactis/chemistry , Lactococcus lactis/classification , Lipoproteins/genetics , Lipoproteins/physiology , Molecular Sequence Data , Protein Engineering , Protein Transport/genetics , Sequence Alignment , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...