Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Mol Biol Evol ; 41(4)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38415839

ABSTRACT

Siderophores are crucial for iron-scavenging in microorganisms. While many yeasts can uptake siderophores produced by other organisms, they are typically unable to synthesize siderophores themselves. In contrast, Wickerhamiella/Starmerella (W/S) clade yeasts gained the capacity to make the siderophore enterobactin following the remarkable horizontal acquisition of a bacterial operon enabling enterobactin synthesis. Yet, how these yeasts absorb the iron bound by enterobactin remains unresolved. Here, we demonstrate that Enb1 is the key enterobactin importer in the W/S-clade species Starmerella bombicola. Through phylogenomic analyses, we show that ENB1 is present in all W/S clade yeast species that retained the enterobactin biosynthetic genes. Conversely, it is absent in species that lost the ent genes, except for Starmerella stellata, making this species the only cheater in the W/S clade that can utilize enterobactin without producing it. Through phylogenetic analyses, we infer that ENB1 is a fungal gene that likely existed in the W/S clade prior to the acquisition of the ent genes and subsequently experienced multiple gene losses and duplications. Through phylogenetic topology tests, we show that ENB1 likely underwent horizontal gene transfer from an ancient W/S clade yeast to the order Saccharomycetales, which includes the model yeast Saccharomyces cerevisiae, followed by extensive secondary losses. Taken together, these results suggest that the fungal ENB1 and bacterial ent genes were cooperatively integrated into a functional unit within the W/S clade that enabled adaptation to iron-limited environments. This integrated fungal-bacterial circuit and its dynamic evolution determine the extant distribution of yeast enterobactin producers and cheaters.


Subject(s)
Enterobactin , Evolution, Molecular , Operon , Phylogeny , Enterobactin/metabolism , Enterobactin/genetics , Siderophores/metabolism , Siderophores/genetics , Genes, Fungal , Saccharomycetales/genetics , Saccharomycetales/metabolism , Gene Transfer, Horizontal
2.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34502502

ABSTRACT

Siderophores produced by microorganisms to scavenge iron from the environment have been shown to contribute to virulence and/or stress resistance of some plant pathogenic bacteria. Phytopathogenic bacteria of Pectobacterium genus possess genes for the synthesis of siderophore enterobactin, which role in plant-pathogen interactions has not been elucidated. In the present study we characterized the phenotype of the mutant strain of Pba deficient for the enterobactin-biosynthetic gene entA. We showed that enterobactin may be considered as a conditionally beneficial virulence factor of Pba. The entA knockout did not reduce Pba virulence on non-primed plants; however, salicylic acid-primed plants were more resistant to ΔentA mutant than to the wild type Pba. The reduced virulence of ΔentA mutant towards the primed plants is likely explained by its compromised resistance to oxidative stress.


Subject(s)
Enterobactin/genetics , Pectobacterium/genetics , Enterobactin/metabolism , Iron , Oxidative Stress , Pectobacterium/metabolism , Plants/metabolism , Siderophores/genetics , Siderophores/metabolism , Stress, Physiological/physiology , Virulence/genetics
4.
Virulence ; 12(1): 1323-1333, 2021 12.
Article in English | MEDLINE | ID: mdl-33970792

ABSTRACT

Virulence plasmids of hypervirulent Klebsiella pneumoniae (hvKp) have the potential to transfer to drug-resistant strains or integrate with other plasmids, facilitating the genome evolution of threatening pathogens. We conducted an in-depth analysis of the publicly available 156 complete genome sequences of hvKp together with a multi-region clinical cohort of 171 hvKp strains from China to provide evidence for the virulence plasmid evolution. Virulence plasmids were frequently detected in the ST23 and ST11 K. pneumoniae strains. Multidrug-resistant hvKp (MDR-hvKp) occupied a large proportion of hvKp, and the coexistence of virulence and resistance plasmids may be the major cause. Virulence plasmids commonly possessed multiple replicons, of which IncFIBK was the most prevalent (84.6%). We identified 49 IncFIBK alleles among 583 IncFIBK plasmids, and they could be divided into Clades I, II, and III. We further observed that conjugative and non-conjugative virulence plasmids could be distinguished by IncFIBK genetic diversity, and IncFIBK subtyping could also indirectly indicate a chimeric preference of conjugative virulence plasmids. On this basis, we developed an open-access web tool called KpVR for IncFIBK subtyping. In conclusion, the genetic diversity of IncFIBK virulence plasmids could be used for tracking the evolution of virulence plasmids, and further preventing the emergence of MDR-hvKp strains.


Subject(s)
Enterobactin/analogs & derivatives , Hydroxamic Acids , Klebsiella pneumoniae , Plasmids , Enterobactin/genetics , Genetic Variation , Humans , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Plasmids/genetics , Virulence/genetics
5.
Microbiology (Reading) ; 166(11): 1038-1046, 2020 11.
Article in English | MEDLINE | ID: mdl-33074088

ABSTRACT

Vibrio cholerae, the aetiological agent of cholera, possesses multiple iron acquisition systems, including those for the transport of siderophores. How these systems benefit V. cholerae in low-iron, polymicrobial communities in environmental settings or during infection remains poorly understood. Here, we demonstrate that in iron-limiting conditions, co-culture of V. cholerae with a number of individual siderophore-producing microbes significantly promoted V. cholerae growth in vitro. We further show that in the host environment with low iron, V. cholerae colonizes better in adult mice in the presence of the siderophore-producing commensal Escherichia coli. Taken together, our results suggest that in aquatic reservoirs or during infection, V. cholerae may overcome environmental and host iron restriction by hijacking siderophores from other microbes.


Subject(s)
Siderophores/metabolism , Vibrio cholerae/physiology , Vibrio cholerae/pathogenicity , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Coculture Techniques , Enterobactin/genetics , Enterobactin/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gastrointestinal Microbiome , Iron Deficiencies , Mice , Microbial Viability , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Seawater/microbiology , Siderophores/genetics
6.
Microbes Infect ; 22(3): 144-147, 2020 04.
Article in English | MEDLINE | ID: mdl-31954842

ABSTRACT

A functional synergy was previously demonstrated between microcin, salmochelin and colibactin islands in Escherichia coli strains from B2 phylogroup. We aimed to determine this association prevalence in uropathogenic E. coli, and whether it was predictive of the infection severity in a collection of 225 E. coli strains from urinary samples. The high prevalence of this triad, even if it wasn't correlated with infection severity, suggested that it might not be a virulence factor per se within the urinary tract, but would promote its colonization. This triad would enable the strain to dominate the rectal reservoir with a minimal genetic cost.


Subject(s)
Bacteriocins/genetics , Enterobactin/analogs & derivatives , Multigene Family , Peptides/genetics , Uropathogenic Escherichia coli/genetics , Enterobactin/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/urine , Humans , Polyketides , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/pathogenicity , Virulence/genetics
7.
Genomics ; 112(2): 1182-1191, 2020 03.
Article in English | MEDLINE | ID: mdl-31279858

ABSTRACT

Enterobacter bugandensis is a recently described species that has been largely associated with nosocomial infections. We report the genome of a non-clinical E. bugandensis strain, which was integrated with publicly available genomes to study the pangenome and general population structure of E. bugandensis. Core- and whole-genome multilocus sequence typing allowed the detection of five E. bugandensis phylogroups (PG-A to E), which contain important antimicrobial resistance and virulence determinants. We uncovered several extended-spectrum ß-lactamases, including blaCTX-M-55 and blaNDM-5, present in an IncX replicon type plasmid, described here for the first time in E. bugandensis. Genetic context analysis of blaNDM-5 revealed the resemblance of this plasmid with other IncX plasmids from other bacteria from the same country. Three distinctive siderophore producing operons were found in E. bugandensis: enterobactin (ent), aerobactin (iuc/iut), and salmochelin (iro). Our findings provide novel insights on the lifestyle, physiology, antimicrobial, and virulence profiles of E. bugandensis.


Subject(s)
Bacterial Proteins/genetics , Enterobacter/genetics , Genome, Bacterial , Iron/metabolism , beta-Lactamases/genetics , Bacterial Proteins/metabolism , Enterobacter/metabolism , Enterobactin/analogs & derivatives , Enterobactin/genetics , Enterobactin/metabolism , Hydroxamic Acids/metabolism , Operon , beta-Lactamases/metabolism
8.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Article in English | MEDLINE | ID: mdl-31545853

ABSTRACT

Although Escherichia coli Nissle 1917 (EcN) has been used therapeutically for over a century, the determinants of its probiotic properties remain elusive. EcN produces two siderophore-microcins (Mcc) responsible for an antagonistic activity against other Enterobacteriaceae. EcN also synthesizes the genotoxin colibactin encoded by the pks island. Colibactin is a virulence factor and a putative pro-carcinogenic compound. Therefore, we aimed to decouple the antagonistic activity of EcN from its genotoxic activity. We demonstrated that the pks-encoded ClbP, the peptidase that activates colibactin, is required for the antagonistic activity of EcN. The analysis of a series of ClbP mutants revealed that this activity is linked to the transmembrane helices of ClbP and not the periplasmic peptidase domain, indicating the transmembrane domain is involved in some aspect of Mcc biosynthesis or secretion. A single amino acid substitution in ClbP inactivates the genotoxic activity but maintains the antagonistic activity. In an in vivo salmonellosis model, this point mutant reduced the clinical signs and the fecal shedding of Salmonella similarly to the wild type strain, whereas the clbP deletion mutant could neither protect nor outcompete the pathogen. The ClbP-dependent antibacterial effect was also observed in vitro with other E. coli strains that carry both a truncated form of the Mcc gene cluster and the pks island. In such strains, siderophore-Mcc synthesis also required the glucosyltransferase IroB involved in salmochelin production. This interplay between colibactin, salmochelin, and siderophore-Mcc biosynthetic pathways suggests that these genomic islands were co-selected and played a role in the evolution of E. coli from phylogroup B2. This co-evolution observed in EcN illustrates the fine margin between pathogenicity and probiotic activity, and the need to address both the effectiveness and safety of probiotics. Decoupling the antagonistic from the genotoxic activity by specifically inactivating ClbP peptidase domain opens the way to the safe use of EcN.


Subject(s)
Escherichia coli/physiology , Mutagens/toxicity , Probiotics/therapeutic use , Animals , Antibiosis/genetics , Antibiosis/physiology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/toxicity , Biosynthetic Pathways/genetics , Enterobactin/analogs & derivatives , Enterobactin/genetics , Enterobactin/physiology , Enterobactin/toxicity , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Female , Genes, Bacterial , Genomic Islands , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Multigene Family , Mutation , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/physiology , Peptides/genetics , Peptides/physiology , Peptides/toxicity , Polyketides/toxicity , Probiotics/toxicity , Protein Domains , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/therapy , Salmonella typhimurium , Siderophores/genetics , Siderophores/physiology , Siderophores/toxicity , Virulence Factors/genetics , Virulence Factors/physiology , Virulence Factors/toxicity
9.
Gut Microbes ; 10(3): 412-423, 2019.
Article in English | MEDLINE | ID: mdl-30449241

ABSTRACT

Enterobactin (Ent), a prototypical bacterial siderophore known for its unparalleled affinity for iron, is widely conserved among members of the Enterobacteriaceae family of Gram-negative bacteria. In this study, we demonstrated that, aside from mediating iron acquisition, Ent also dampened the macrophages (MΦs) antimicrobial responses against intracellular infection by Salmonella enterica serovar Typhimurium. Accordingly, the loss of Ent expression (ΔentB) in Salmonella demoted their survivability against MΦs. Addition of exogenous Ent not only rescued the survival of ΔentB Salmonella, but also augmented WT Salmonella to better withstand the microbicidal activity of MΦs. The protection conferred to WT Salmonella was observed only when Ent was administered as iron-free, thus indicating the requirement of iron chelation in this context. In contrast, the exogenous iron-bound Ent retained its ability to promote the survival of ΔentB Salmonella, albeit modestly. Assessment on MΦs labile iron pool (LIP) revealed that iron-free Ent is able to permeate into MΦs, chelate the intracellular LIP, and regulate the expression of several key iron-regulatory proteins, i.e., divalent metal transporter 1, ferroportin, and hepcidin. Chelation of iron by Ent was also observed to promote the MΦs towards M2 polarization. Collectively, our findings demonstrated that Ent not only facilitates bacterial iron uptake but also disrupts MΦs iron homeostasis and M1/M2 polarization to safeguard intracellular bacteria against the anti-bacterial effects of their host.


Subject(s)
Enterobactin/metabolism , Host-Pathogen Interactions , Macrophages/microbiology , Salmonella typhimurium/physiology , Siderophores/metabolism , Animals , Bacterial Proteins/genetics , Cell Differentiation/immunology , Enterobactin/genetics , Enterobactin/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Homeostasis , Iron/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Microbial Viability , Mutation , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Siderophores/genetics , Siderophores/pharmacology
10.
Genome Med ; 10(1): 77, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30371343

ABSTRACT

BACKGROUND: Klebsiella pneumoniae is a recognised agent of multidrug-resistant (MDR) healthcare-associated infections; however, individual strains vary in their virulence potential due to the presence of mobile accessory genes. In particular, gene clusters encoding the biosynthesis of siderophores aerobactin (iuc) and salmochelin (iro) are associated with invasive disease and are common amongst hypervirulent K. pneumoniae clones that cause severe community-associated infections such as liver abscess and pneumonia. Concerningly, iuc has also been reported in MDR strains in the hospital setting, where it was associated with increased mortality, highlighting the need to understand, detect and track the mobility of these virulence loci in the K. pneumoniae population. METHODS: Here, we examined the genetic diversity, distribution and mobilisation of iuc and iro loci amongst 2503 K. pneumoniae genomes using comparative genomics approaches and developed tools for tracking them via genomic surveillance. RESULTS: Iro and iuc were detected at low prevalence (< 10%). Considerable genetic diversity was observed, resolving into five iro and six iuc lineages that show distinct patterns of mobilisation and dissemination in the K. pneumoniae population. The major burden of iuc and iro amongst the genomes analysed was due to two linked lineages (iuc1/iro1 74% and iuc2/iro2 14%), each carried by a distinct non-self-transmissible IncFIBK virulence plasmid type that we designate KpVP-1 and KpVP-2. These dominant types also carry hypermucoidy (rmpA) determinants and include all previously described virulence plasmids of K. pneumoniae. The other iuc and iro lineages were associated with diverse plasmids, including some carrying IncFII conjugative transfer regions and some imported from Escherichia coli; the exceptions were iro3 (mobilised by ICEKp1) and iuc4 (fixed in the chromosome of K. pneumoniae subspecies rhinoscleromatis). Iro/iuc mobile genetic elements (MGEs) appear to be stably maintained at high frequency within known hypervirulent strains (ST23, ST86, etc.) but were also detected at low prevalence in others such as MDR strain ST258. CONCLUSIONS: Iuc and iro are mobilised in K. pneumoniae via a limited number of MGEs. This study provides a framework for identifying and tracking these important virulence loci, which will be important for genomic surveillance efforts including monitoring for the emergence of hypervirulent MDR K. pneumoniae strains.


Subject(s)
Enterobactin/genetics , Genetic Loci , Hydroxamic Acids/metabolism , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Siderophores/genetics , DNA Transposable Elements/genetics , Escherichia coli/genetics , Genetic Variation , Phylogeny , Plasmids/genetics , Virulence
11.
Proc Natl Acad Sci U S A ; 115(29): 7581-7586, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29954861

ABSTRACT

The secretion of small Fe-binding molecules called siderophores is an important microbial strategy for survival in Fe-limited environments. Siderophore production is often regulated by quorum sensing (QS), a microbial counting technique that allows organisms to alter gene expression based on cell density. However, the identity and quantities of siderophores produced under QS regulation are rarely studied in the context of their roles in Fe uptake. We investigated the link between QS, siderophores, and Fe uptake in the model marine organism Vibrio harveyi where QS is thought to repress siderophore production. We find that V. harveyi uses a single QS- and Fe-repressed gene cluster to produce both cell-associated siderophores (amphiphilic enterobactins) as well as several related soluble siderophores, which we identify and quantify using liquid chromatography-coupled (LC)-MS as well as tandem high-resolution MS (LC-HR-MS/MS). Measurements of siderophore production show that soluble siderophores are present at ∼100× higher concentrations than amphi-enterobactin and that over the course of growth V. harveyi decreases amphi-enterobactin concentrations but accumulates soluble siderophores. 55Fe radio-tracer uptake experiments demonstrate that these soluble siderophores play a significant role in Fe uptake and that the QS-dictated concentrations of soluble siderophores in stationary phase are near the limit of cellular uptake capacities. We propose that cell-associated and soluble siderophores are beneficial to V. harveyi in different environmental and growth contexts and that QS allows V. harveyi to exploit "knowledge" of its population size to avoid unnecessary siderophore production.


Subject(s)
Enterobactin/biosynthesis , Iron/metabolism , Multigene Family/physiology , Quorum Sensing/physiology , Siderophores/biosynthesis , Vibrio/metabolism , Enterobactin/genetics , Siderophores/genetics , Vibrio/genetics
12.
Microb Genom ; 4(2)2018 02.
Article in English | MEDLINE | ID: mdl-29424684

ABSTRACT

The increasing prevalence of KPC-producing Klebsiella pneumoniae strains in clinical settings has been largely attributed to dissemination of organisms of specific multilocus sequence types, such as ST258 and ST11. Compared with the ST258 clone, which is prevalent in North America and Europe, ST11 is common in China but information regarding its genetic features remains scarce. In this study, we performed detailed genetic characterization of ST11 K. pneumoniae strains by analyzing whole-genome sequences of 58 clinical strains collected from diverse geographic locations in China. The ST11 genomes were found to be highly heterogeneous and clustered into at least three major lineages based on the patterns of single-nucleotide polymorphisms. Exhibiting five different capsular types, these ST11 strains were found to harbor multiple resistance and virulence determinants such as the blaKPC-2 gene, which encodes carbapenemase, and the yersiniabactin-associated virulence genes irp, ybt and fyu. Moreover, genes encoding the virulence factor aerobactin and the regulator of the mucoid phenotype (rmpA) were detectable in six genomes, whereas genes encoding salmochelin were found in three genomes. In conclusion, our data indicated that carriage of a wide range of resistance and virulence genes constitutes the underlying basis of the high level of prevalence of ST11 in clinical settings. Such findings provide insight into the development of novel strategies for prevention, diagnosis and treatment of K. pneumoniae infections.


Subject(s)
Genome, Bacterial/genetics , Klebsiella pneumoniae/classification , Klebsiella pneumoniae/genetics , Multilocus Sequence Typing/methods , Bacterial Proteins/genetics , Carbapenem-Resistant Enterobacteriaceae/genetics , China , DNA, Bacterial/isolation & purification , Drug Resistance, Multiple, Bacterial/genetics , Enterobactin/analogs & derivatives , Enterobactin/genetics , Genes, Bacterial/genetics , Humans , Hydroxamic Acids , Klebsiella Infections/microbiology , Microbial Sensitivity Tests , Phenols , Polymorphism, Single Nucleotide , Sequence Alignment , Sequence Analysis , Thiazoles , Virulence/genetics , Virulence Factors/genetics , beta-Lactamases/genetics
13.
IUBMB Life ; 69(6): 435-441, 2017 06.
Article in English | MEDLINE | ID: mdl-28295919

ABSTRACT

Highly pathogenic Escherichia coli strains that belong to the phylogenetic group B2 have developed a greater ability to acquire iron (heme receptor and numerous siderophores), to produce the genotoxin colibactin and to synthesize antimicrobial siderophore-microcins. There is an increased prevalence of these E. coli strains over the last 30 years in the intestinal microbiota in industrialized countries. Integrating the regulation of fitness/virulence factors, such as siderophores, colibactin and siderophore-microcins into networks that respond to specific environmental signals, such as the local iron concentration, could result in an accurate production of specific fitness/virulence factors, so that the E. coli can adapt to the competitive environment that is the gut and/or the blood. Iron deficiency is common in infancy, even in industrialized countries. Usual strategies for anemia correction are iron supplementation and iron fortification of foods. The long-term consequences and risks associated with high iron supply in the light of this iron-dependent network described in this review could explain at least in part the increased prevalence of E. coli B2 in the gut of people in industrialized countries. © 2017 IUBMB Life, 69(6):435-441, 2017.


Subject(s)
Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Iron/metabolism , Peptides/metabolism , Polyketides/metabolism , Siderophores/biosynthesis , Virulence Factors/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriocins/biosynthesis , Bacteriocins/genetics , Dietary Supplements , Enterobactin/biosynthesis , Enterobactin/genetics , Escherichia coli/classification , Escherichia coli/genetics , Gastrointestinal Microbiome/genetics , Homeostasis/genetics , Humans , Iron/administration & dosage , Peptides/genetics , Phylogeny , Repressor Proteins/genetics , Repressor Proteins/metabolism , Siderophores/genetics , Virulence Factors/metabolism
14.
J Bacteriol ; 199(1)2017 01 01.
Article in English | MEDLINE | ID: mdl-27795321

ABSTRACT

Iron is an essential micronutrient required for the viability of many organisms. Under oxidizing conditions, ferric iron is highly insoluble (∼10-9 to 10-18 M), yet bacteria typically require ∼10-6 M for survival. To overcome this disparity, many bacteria have adopted the use of extracellular iron-chelating siderophores coupled with specific iron-siderophore uptake systems. In the case of Bacillus subtilis, undomesticated strains produce the siderophore bacillibactin. However, many laboratory strains, e.g., JH642, have lost the ability to produce bacillibactin during the process of domestication. In this work, we identified a novel iron acquisition activity from strain JH642 that accumulates in the growth medium and coordinates the iron response with population density. The molecule(s) responsible for this activity was named elemental Fe(II/III) (Efe) acquisition factor because efeUOB (ywbLMN) is required for its activity. Unlike most iron uptake molecules, including siderophores and iron reductases, Efe acquisition factor is present under iron-replete conditions and is regulated independently of Fur repressor. Restoring bacillibactin production in strain JH642 inhibits the activity of Efe acquisition factor, presumably by sequestering available iron. A similar iron acquisition activity is produced from a mutant of Escherichia coli unable to synthesize the siderophore enterobactin. Given the conservation of efeUOB and its regulation by catecholic siderophores in B. subtilis and E. coli, we speculate that Efe acquisition factor is utilized by many bacteria, serves as an alternative to Fur-mediated iron acquisition systems, and provides cells with biologically available iron that would normally be inaccessible during aerobic growth under iron-replete conditions. IMPORTANCE: Iron is an essential micronutrient required for a variety of biological processes, yet ferric iron is highly insoluble during aerobic growth. In this work, we identified a novel iron acquisition activity that coordinates the iron response with population density in laboratory strains of Bacillus subtilis We named the molecule(s) responsible for this activity elemental Fe(II/III) (Efe) acquisition factor after the efeUOB (ywbLMN) operon required for its uptake into cells. Unlike most iron uptake systems, Efe acquisition factor is present under iron-replete conditions and is regulated independently of Fur, the master regulator of the iron response. We speculate that Efe acquisition factor is highly conserved among bacteria and serves as a backup to Fur-mediated iron acquisition systems.


Subject(s)
Bacillus subtilis/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Iron/metabolism , Bacillus subtilis/genetics , Biological Transport , Carrier Proteins/genetics , Enterobactin/genetics , Enterobactin/metabolism , Mutation , Oligopeptides/genetics , Oligopeptides/metabolism
15.
PLoS One ; 11(6): e0157799, 2016.
Article in English | MEDLINE | ID: mdl-27310257

ABSTRACT

Microorganisms produce siderophores to facilitate iron uptake and even though this trait has been extensively studied, there is growing evidence suggesting that siderophores may have other physiological roles aside from iron acquisition. In support of this notion, we previously linked the archetypal siderophore enterobactin with oxidative stress alleviation. To further characterize this association, we studied the sensitivity of Escherichia coli strains lacking different components of the enterobactin system to the classical oxidative stressors hydrogen peroxide and paraquat. We observed that strains impaired in enterobactin production, uptake and hydrolysis were more susceptible to the oxidative damage caused by both compounds than the wild-type strain. In addition, meanwhile iron supplementation had little impact on the sensitivity, the reducing agent ascorbic acid alleviated the oxidative stress and therefore significantly decreased the sensitivity to the stressors. This indicated that the enterobactin-mediated protection is independent of its ability to scavenge iron. Furthermore, enterobactin supplementation conferred resistance to the entE mutant but did not have any protective effect on the fepG and fes mutants. Thus, we inferred that only after enterobactin is hydrolysed by Fes in the cell cytoplasm and iron is released, the free hydroxyl groups are available for radical stabilization. This hypothesis was validated testing the ability of enterobactin to scavenge radicals in vitro. Given the strong connection between enterobactin and oxidative stress, we studied the transcription of the entE gene and the concomitant production of the siderophore in response to such kind of stress. Interestingly, we observed that meanwhile iron represses the expression and production of the siderophore, hydrogen peroxide and paraquat favour these events even if iron is present. Our results support the involvement of enterobactin as part of the oxidative stress response and highlight the existence of a novel regulation mechanism for enterobactin biosynthesis.


Subject(s)
Enterobactin/biosynthesis , Escherichia coli/genetics , Gene Expression Regulation , Siderophores/biosynthesis , Stress, Physiological/genetics , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Chlorides/pharmacology , Enterobactin/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ferric Compounds/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Hydrolysis , Iron/metabolism , Ligases/genetics , Ligases/metabolism , Mutation , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress , Paraquat/antagonists & inhibitors , Paraquat/pharmacology , Siderophores/genetics , Transcription, Genetic
16.
Nat Commun ; 6: 7113, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25964185

ABSTRACT

During an inflammatory response in the gut, some commensal bacteria such as E. coli can thrive and contribute to disease. Here we demonstrate that enterobactin (Ent), a catecholate siderophore released by E. coli, is a potent inhibitor of myeloperoxidase (MPO), a bactericidal enzyme of the host. Glycosylated Ent (salmochelin) and non-catecholate siderophores (yersiniabactin and ferrichrome) fail to inhibit MPO activity. An E. coli mutant (ΔfepA) that overproduces Ent, but not an Ent-deficient double mutant (ΔaroB/ΔfepA), inhibits MPO activity and exhibits enhanced survival in inflamed guts. This survival advantage is counter-regulated by lipocalin 2, a siderophore-binding host protein, which rescues MPO from Ent-mediated inhibition. Spectral analysis reveals that Ent interferes with compound I [oxoiron, Fe(IV)=O] and reverts the enzyme back to its native ferric [Fe(III)] state. These findings define a fundamental mechanism by which E. coli surpasses the host innate immune responses during inflammatory gut diseases and gains a distinct survival advantage.


Subject(s)
Acute-Phase Proteins/metabolism , Enterobactin/metabolism , Escherichia coli/physiology , Inflammation/chemically induced , Lipocalins/metabolism , Oncogene Proteins/metabolism , Peroxidase/metabolism , Proto-Oncogene Proteins/metabolism , Acute-Phase Proteins/genetics , Animals , Colitis/chemically induced , Dextran Sulfate/toxicity , Enterobactin/genetics , Female , Gene Expression Regulation , Inflammation/microbiology , Lipocalin-2 , Lipocalins/genetics , Mice, Inbred BALB C , Oncogene Proteins/genetics , Peroxidase/antagonists & inhibitors , Peroxidase/genetics , Proto-Oncogene Proteins/genetics
17.
PLoS One ; 10(3): e0117906, 2015.
Article in English | MEDLINE | ID: mdl-25756870

ABSTRACT

Iron is essential for Escherichia coli growth and survival in the host and the external environment, but its availability is generally low due to the poor solubility of its ferric form in aqueous environments and the presence of iron-withholding proteins in the host. Most E. coli can increase access to iron by excreting siderophores such as enterobactin, which have a very strong affinity for Fe3+. A smaller proportion of isolates can generate up to 3 additional siderophores linked with pathogenesis; aerobactin, salmochelin, and yersiniabactin. However, non-pathogenic E. coli are also able to synthesise these virulence-associated siderophores. This raises questions about their role in the ecology of E. coli, beyond virulence, and whether specific siderophores might be linked with persistence in the external environment. Under the assumption that selection favours phenotypes that confer a fitness advantage, we compared siderophore production and gene distribution in E. coli isolated either from agricultural plants or the faeces of healthy mammals. This population-level comparison has revealed that under iron limiting growth conditions plant-associated isolates produced lower amounts of siderophores than faecal isolates. Additionally, multiplex PCR showed that environmental isolates were less likely to contain loci associated with aerobactin and yersiniabactin synthesis. Although aerobactin was linked with strong siderophore excretion, a significant difference in production was still observed between plant and faecal isolates when the analysis was restricted to strains only able to synthesise enterobactin. This finding suggests that the regulatory response to iron limitation may be an important trait associated with adaptation to the non-host environment. Our findings are consistent with the hypothesis that the ability to produce multiple siderophores facilitates E. coli gut colonisation and plays an important role in E. coli commensalism.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/isolation & purification , Feces/microbiology , Plants/microbiology , Siderophores/metabolism , Adaptation, Biological , Animals , Enterobactin/analogs & derivatives , Enterobactin/genetics , Enterobactin/metabolism , Environmental Microbiology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Glucosides/genetics , Glucosides/metabolism , Humans , Hydroxamic Acids/metabolism , Iron/metabolism , Phenols/metabolism , Siderophores/genetics , Thiazoles/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
18.
J Bacteriol ; 197(13): 2122-2128, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25733620

ABSTRACT

UNLABELLED: Many bacteria produce secreted iron chelators called siderophores, which can be shared among cells with specific siderophore uptake systems regardless of whether the cell produces siderophores. Sharing secreted products allows freeloading, where individuals use resources without bearing the cost of production. Here we show that the Escherichia coli siderophore enterochelin is not evenly shared between producers and nonproducers. Wild-type Escherichia coli grows well in low-iron minimal medium, and an isogenic enterochelin synthesis mutant (ΔentF) grows very poorly. The enterochelin mutant grows well in low-iron medium supplemented with enterochelin. At high cell densities the ΔentF mutant can compete equally with the wild type in low-iron medium. At low cell densities the ΔentF mutant cannot compete. Furthermore, the growth rate of the wild type is unaffected by cell density. The wild type grows well in low-iron medium even at very low starting densities. Our experiments support a model where at least some enterochelin remains associated with the cells that produce it, and the cell-associated enterochelin enables iron acquisition even at very low cell density. Enterochelin that is not retained by producing cells at low density is lost to dilution. At high cell densities, cell-free enterochelin can accumulate and be shared by all cells in the group. Partial privatization is a solution to the problem of iron acquisition in low-iron, low-cell-density habitats. Cell-free enterochelin allows for iron scavenging at a distance at higher population densities. Our findings shed light on the conditions under which freeloaders might benefit from enterochelin uptake systems. IMPORTANCE: Sociality in microbes has become a topic of great interest. One facet of sociality is the sharing of secreted products, such as the iron-scavenging siderophores. We present evidence that the Escherichia coli siderophore enterochelin is relatively inexpensive to produce and is partially privatized such that it can be efficiently shared only at high producer cell densities. At low cell densities, cell-free enterochelin is scarce and only enterochelin producers are able to grow in low-iron medium. Because freely shared products can be exploited by freeloaders, this partial privatization may help explain how enterochelin production is stabilized in E. coli and may provide insight into when enterochelin is available for freeloaders.


Subject(s)
Enterobactin/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Siderophores/metabolism , Biological Transport , Enterobactin/genetics , Escherichia coli/genetics , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Iron/metabolism , Mutation , Peptide Synthases/genetics , Peptide Synthases/metabolism , Siderophores/genetics
19.
PLoS One ; 9(1): e84734, 2014.
Article in English | MEDLINE | ID: mdl-24392154

ABSTRACT

Numerous bacteria have evolved different iron uptake systems with the ability to make use of their own and heterologous siderophores. However, there is growing evidence attributing alternative roles for siderophores that might explain the potential adaptive advantages of microorganisms having multiple siderophore systems. In this work, we show the requirement of the siderophore enterobactin for Escherichia coli colony development in minimal media. We observed that a strain impaired in enterobactin production (entE mutant) was unable to form colonies on M9 agar medium meanwhile its growth was normal on LB agar medium. Given that, neither iron nor citrate supplementation restored colony growth, the role of enterobactin as an iron uptake-facilitator would not explain its requirement for colony development. The absence of colony development was reverted either by addition of enterobactin, the reducing agent ascorbic acid or by incubating in anaerobic culture conditions with no additives. Then, we associated the enterobactin requirement for colony development with its ability to reduce oxidative stress, which we found to be higher in media where the colony development was impaired (M9) compared with media where the strain was able to form colonies (LB). Since oxyR and soxS mutants (two major stress response regulators) formed colonies in M9 agar medium, we hypothesize that enterobactin could be an important piece in the oxidative stress response repertoire, particularly required in the context of colony formation. In addition, we show that enterobactin has to be hydrolyzed after reaching the cell cytoplasm in order to enable colony development. By favoring iron release, hydrolysis of the enterobactin-iron complex, not only would assure covering iron needs, but would also provide the cell with a molecule with exposed hydroxyl groups (hydrolyzed enterobactin). This molecule would be able to scavenge radicals and therefore reduce oxidative stress.


Subject(s)
Enterobactin/metabolism , Escherichia coli/growth & development , Escherichia coli/metabolism , Oxidative Stress , Bacterial Load , Culture Media , Enterobactin/genetics , Escherichia coli/genetics , Hydrolysis , Mutation , Reactive Oxygen Species
20.
mBio ; 3(6)2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23169997

ABSTRACT

UNLABELLED: Pathogenic bacteria require iron for replication within their host. Klebsiella pneumoniae and other Gram-negative pathogens produce the prototypical siderophore enterobactin (Ent) to scavenge iron in vivo. In response, mucosal surfaces secrete lipocalin 2 (Lcn2), an innate immune protein that binds Ent to disrupt bacterial iron acquisition and promote acute inflammation during colonization. A subset of K. pneumoniae isolates attempt to evade Lcn2 by producing glycosylated Ent (Gly-Ent, salmochelin) or the alternative siderophore yersiniabactin (Ybt). However, these siderophores are not functionally equivalent and differ in their abilities to promote growth in the upper respiratory tract, lungs, and serum. To understand how Lcn2 exploits functional differences between siderophores, isogenic mutants of an Ent(+) Gly-Ent(+) Ybt(+) K. pneumoniae strain were inoculated into Lcn2(+/+) and Lcn2(-/-) mice, and the pattern of pneumonia was examined. Lcn2 effectively protected against the iroA ybtS mutant (Ent(+) Gly-Ent(-) Ybt(-)). Lcn2(+/+) mice had small foci of pneumonia, whereas Lcn2(-/-) mice had many bacteria in the perivascular space. The entB mutant (Ent(-) Ybt(+) Gly-Ent(-)) caused moderate bronchopneumonia but did not invade the transferrin-containing perivascular space. Accordingly, transferrin blocked Ybt-dependent growth in vitro. The wild type and the iroA mutant, which both produce Ent and Ybt, had a mixed phenotype, causing a moderate bronchopneumonia in Lcn2(+/+) mice and perivascular overgrowth in Lcn2(-/-) mice. Together, these data indicate that Lcn2, in combination with transferrin, confines K. pneumoniae to the airways and prevents invasion into tissue containing the pulmonary vasculature. IMPORTANCE: Gram-negative bacteria are a common cause of severe hospital-acquired infections. To cause disease, they must obtain iron and secrete the small molecule enterobactin to do so. Animal models of pneumonia using Klebsiella pneumoniae indicate that enterobactin promotes severe disease. Accordingly, the host defense protein lipocalin 2 exploits this common target by binding enterobactin and disrupting its function. However, pathogenic bacteria often make additional siderophores that lipocalin 2 cannot bind, such as yersiniabactin, which could make this host defense ineffective. This work compares the pattern and severity of pneumonia caused by K. pneumoniae based on which siderophores it produces. The results indicate that enterobactin promotes growth around blood vessels that are rich in the iron-binding protein transferrin, but yersiniabactin does not. Together, transferrin and lipocalin 2 protect this space against all types of K. pneumoniae tested. Therefore, the ability to acquire iron determines where bacteria can grow in the lung.


Subject(s)
Acute-Phase Proteins/metabolism , Enterobactin/metabolism , Host-Pathogen Interactions , Klebsiella Infections/pathology , Klebsiella pneumoniae/pathogenicity , Lipocalins/metabolism , Oncogene Proteins/metabolism , Pneumonia, Bacterial/pathology , Transferrin/metabolism , Acute-Phase Proteins/deficiency , Animals , Enterobactin/antagonists & inhibitors , Enterobactin/genetics , Lipocalin-2 , Mice , Mice, Knockout , Oncogene Proteins/deficiency , Phenols/antagonists & inhibitors , Phenols/metabolism , Protein Binding , Thiazoles/antagonists & inhibitors , Thiazoles/metabolism , Virulence Factors/antagonists & inhibitors , Virulence Factors/genetics , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...