Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38759643

RESUMO

Urinary tract infection (UTI), mainly caused by Escherichia coli, are frequent and have a recurrent nature even after antibiotic treatment. Potential bacterial escape mechanisms include growth defects, but probing bacterial division in vivo and establishing its relation to the antibiotic response remain challenging. Using a synthetic reporter of cell division, we follow the temporal dynamics of cell division for different E. coli clinical strains in a UTI mouse model with and without antibiotics. We show that more bacteria are actively dividing in the kidneys and urine compared with the bladder. Bacteria that survive antibiotic treatment are consistently non-dividing in three sites of infection. Additionally, we demonstrate how both the strain in vitro persistence profile and the microenvironment impact infection and treatment dynamics. Understanding the relative contribution of the host environment, growth heterogeneity, non-dividing bacteria, and antibiotic persistence is crucial to improve therapies for recurrent infections.

2.
Sci Adv ; 10(6): eadj0767, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38335294

RESUMO

Protein carboxymethyltransferase (Pcm) is a highly evolutionarily conserved enzyme that initiates the conversion of abnormal isoaspartate to aspartate residues. While it is commonly believed that Pcm facilitates the repair of damaged proteins, a number of observations suggest that it may have another role in cell functioning. We investigated whether Pcm provides a means for Escherichia coli to recycle aspartate, which is essential for protein synthesis and other cellular processes. We showed that Pcm is required for the energy production, the maintenance of cellular redox potential and of S-adenosylmethionine synthesis, which are critical for the proper functioning of many metabolic pathways. Pcm contributes to the full growth capacity both under aerobic and anaerobic conditions. Last, we showed that Pcm enhances the robustness of bacteria when exposed to sublethal antibiotic treatments and improves their fitness in the mammalian urinary tract. We propose that Pcm plays a crucial role in E. coli metabolism by ensuring a steady supply of aspartate.


Assuntos
Ácido Aspártico , Escherichia coli , Animais , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Antibacterianos , Redes e Vias Metabólicas , Mamíferos/metabolismo
3.
Nat Commun ; 14(1): 3667, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37339949

RESUMO

The intrinsic virulence of extra-intestinal pathogenic Escherichia coli is associated with numerous chromosomal and/or plasmid-borne genes, encoding diverse functions such as adhesins, toxins, and iron capture systems. However, the respective contribution to virulence of those genes seems to depend on the genetic background and is poorly understood. Here, we analyze genomes of 232 strains of sequence type complex STc58 and show that virulence (quantified in a mouse model of sepsis) emerged in a sub-group of STc58 due to the presence of the siderophore-encoding high-pathogenicity island (HPI). When extending our genome-wide association study to 370 Escherichia strains, we show that full virulence is associated with the presence of the aer or sit operons, in addition to the HPI. The prevalence of these operons, their co-occurrence and their genomic location depend on strain phylogeny. Thus, selection of lineage-dependent specific associations of virulence-associated genes argues for strong epistatic interactions shaping the emergence of virulence in E. coli.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Animais , Camundongos , Virulência/genética , Ferro , Infecções por Escherichia coli/patologia , Ilhas Genômicas/genética , Estudo de Associação Genômica Ampla , Filogenia
4.
mBio ; 13(2): e0038522, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35377167

RESUMO

Toxin-antitoxin systems are genetic elements that are widespread in prokaryotes. Although molecular mode of action of many of these toxins has been identified, their biological functions are mostly unknown. We investigated the functional integration of the TisB/IstR toxin-antitoxin system in the Escherichia coli SOS genotoxic stress response network. We showed that the tisB gene is induced in cells exposed to high doses of the genotoxic antibiotic trimethoprim. However, we also found that TisB contributes to trimethoprim-induced lethality. This is a consequence of the TisB-induced drop in the proton motive force (PMF), which results in blocking the thymine import and therefore the functioning of the pyrimidine salvage pathway. Conversely, a TisB-induced PMF drop protects cells by preventing the import of some other toxic compounds, like the aminoglycoside antibiotic gentamicin and colicin M, in the SOS-induced cells. Colicins are cytotoxic molecules produced by Enterobacterales when they are exposed to strong genotoxic stresses in order to compete with other microbiota members. We indeed found that TisB contributes to E. coli's fitness during mouse gut colonization. Based on the results obtained here, we propose that the primary biological role of the TisB toxin is to increase the probability of survival and maintenance in the mammalian gut of their bacterial hosts when they have to simultaneously deal with massive DNA damages and a fierce chemical warfare with other microbiota members. IMPORTANCE The contribution of toxin-antitoxin systems to the persistence of bacteria to antibiotics has been intensively studied. This is also the case with the E. coli TisB/IstR toxin-antitoxin system, but the contribution of TisB to the persistence to antibiotics turned out to be not as straightforward as anticipated. In this study, we show that TisB can decrease, but also increase, cytotoxicity of different antibiotics. This inconsistency has a common origin, i.e., TisB-induced collapse of the PMF, which impacts the import and the action of different antibiotics. By taking into account the natural habitat of TisB bacterial hosts, the facts that this toxin-antitoxin system is integrated into the genotoxic stress response regulon SOS and that both SOS regulon and TisB are required for E. coli to colonize the host intestine, and the phenotypic consequences of the collapse of the PMF, we propose that TisB protects its hosts from cytotoxic molecules produced by competing intestinal bacteria.


Assuntos
Colicinas , Infecções por Escherichia coli , Animais , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Colicinas/genética , Dano ao DNA , Escherichia coli/metabolismo , Mamíferos , Camundongos , Trimetoprima
5.
Environ Microbiol ; 23(11): 7139-7151, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34431197

RESUMO

To get a global picture of the population structure of the Escherichia coli phylogroup E, encompassing the O157:H7 EHEC lineage, we analysed the whole genome of 144 strains isolated from various continents, hosts and lifestyles and representative of the phylogroup diversity. The strains possess 4331 to 5440 genes with a core genome of 2771 genes and a pangenome of 33 722 genes. The distribution of these genes among the strains shows an asymmetric U-shaped distribution. E phylogenetic strains have the largest genomes of the species, partly explained by the presence of mobile genetic elements. Sixty-eight lineages were delineated, some of them exhibiting extra-intestinal virulence genes and being virulent in the mouse sepsis model. Except for the EHEC lineages and the reference EPEC, EIEC and ETEC strains, very few strains possess intestinal virulence genes. Most of the strains were devoid of acquired resistance genes, but eight strains possessed extended-spectrum beta-lactamase genes. Human strains belong to specific lineages, some of them being virulent and antibiotic-resistant [sequence type complexes (STcs) 350 and 2064]. The E phylogroup mimics all the features of the species as a whole, a phenomenon already observed at the STc level, arguing for a fractal population structure of E. coli.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Animais , Escherichia coli , Proteínas de Escherichia coli/genética , Camundongos , Filogenia , Virulência/genética , Fatores de Virulência/genética
6.
mSphere ; 6(1)2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33408235

RESUMO

Adaptive processes in chronic bacterial infections are well described, but much less is known about the processes at play during acute infections. Here, by sequencing seven randomly selected isolates per patient, we analyzed Escherichia coli populations from three acute extraintestinal infections in adults (meningitis, pyelonephritis, and peritonitis), in which a high-mutation-rate isolate or mutator isolate was found. The isolates of single patients displayed between a few dozen and more than 200 independent mutations, with up to half being specific to the mutator isolate. Multiple signs of positive selection were evidenced: a high ratio of nonsynonymous to synonymous mutations (Ka /Ks ratio) and strong mutational convergence within and between patients, some of them at loci well known for their adaptive potential, such as rpoS, rbsR, fimH, and fliC For all patients, the mutator isolate was likely due to a large deletion of a methyl-directed mismatch repair gene, and in two instances, the deletion extended to genes involved in some genetic convergence, suggesting potential coselection. Intrinsic extraintestinal virulence assessed in a mouse model of sepsis showed variable patterns of virulence ranging from non-mouse killer to mouse killer for the isolates from single patients. However, genomic signature and gene inactivation experiments did not establish a link between a single gene and the capacity to kill mice, highlighting the complex and multifactorial nature of the virulence. Altogether, these data indicate that E. coli isolates are adapting under strong selective pressure when colonizing an extraintestinal site.IMPORTANCE Little is known about the dynamics of adaptation in acute bacterial infections. By sequencing multiple isolates from monoclonal extraintestinal Escherichia coli infections in several patients, we were able to uncover traces of selection taking place at short time scales compared to chronic infection. High genomic diversity was observed in the patient isolates, with an excess of nonsynonymous mutations, and the comparison within and between different infections showed patterns of convergence at the gene level, both constituting strong signs of adaptation. The genes targeted were coding mostly for proteins involved in global regulation, metabolism, and adhesion/motility. Moreover, virulence assessed in a mouse model of sepsis was variable among the isolates of single patients, but this difference was left unexplained at the molecular level. This work gives us clues about the E. coli lifestyle transition between commensalism and pathogenicity.


Assuntos
Adaptação Fisiológica/genética , Infecções por Escherichia coli/microbiologia , Evolução Molecular , Escherichia coli Extraintestinal Patogênica/genética , Escherichia coli Extraintestinal Patogênica/patogenicidade , Genoma Bacteriano , Doença Aguda , Animais , Infecções por Escherichia coli/classificação , Escherichia coli Extraintestinal Patogênica/isolamento & purificação , Feminino , Genótipo , Humanos , Meningite/microbiologia , Camundongos , Mutação , Peritonite/microbiologia , Pielonefrite/microbiologia , Fatores de Virulência/genética
7.
PLoS Genet ; 16(10): e1009065, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33112851

RESUMO

The genus Escherichia is composed of several species and cryptic clades, including E. coli, which behaves as a vertebrate gut commensal, but also as an opportunistic pathogen involved in both diarrheic and extra-intestinal diseases. To characterize the genetic determinants of extra-intestinal virulence within the genus, we carried out an unbiased genome-wide association study (GWAS) on 370 commensal, pathogenic and environmental strains representative of the Escherichia genus phylogenetic diversity and including E. albertii (n = 7), E. fergusonii (n = 5), Escherichia clades (n = 32) and E. coli (n = 326), tested in a mouse model of sepsis. We found that the presence of the high-pathogenicity island (HPI), a ~35 kbp gene island encoding the yersiniabactin siderophore, is highly associated with death in mice, surpassing other associated genetic factors also related to iron uptake, such as the aerobactin and the sitABCD operons. We confirmed the association in vivo by deleting key genes of the HPI in E. coli strains in two phylogenetic backgrounds. We then searched for correlations between virulence, iron capture systems and in vitro growth in a subset of E. coli strains (N = 186) previously phenotyped across growth conditions, including antibiotics and other chemical and physical stressors. We found that virulence and iron capture systems are positively correlated with growth in the presence of numerous antibiotics, probably due to co-selection of virulence and resistance. We also found negative correlations between virulence, iron uptake systems and growth in the presence of specific antibiotics (i.e. cefsulodin and tobramycin), which hints at potential "collateral sensitivities" associated with intrinsic virulence. This study points to the major role of iron capture systems in the extra-intestinal virulence of the genus Escherichia.


Assuntos
Infecções por Escherichia coli/genética , Escherichia coli/genética , Ferro/metabolismo , Sepse/genética , Sideróforos/genética , Animais , Modelos Animais de Doenças , Escherichia coli/classificação , Escherichia coli/patogenicidade , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Variação Genética/genética , Estudo de Associação Genômica Ampla , Ilhas Genômicas/genética , Humanos , Camundongos , Fenóis/metabolismo , Filogenia , Sepse/microbiologia , Sepse/patologia , Sideróforos/metabolismo , Tiazóis/metabolismo , Virulência/genética
8.
Infect Immun ; 88(12)2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-32989036

RESUMO

Escherichia coli O25b:H4 sequence type 131 (ST131), which is resistant to fluoroquinolones and which is a producer of CTX-M-15, is globally one of the major extraintestinal pathogenic E. coli (ExPEC) lineages. Phylogenetic analyses showed that multidrug-resistant ST131 strains belong to clade C, which recently emerged from clade B by stepwise evolution. It has been hypothesized that features other than multidrug resistance could contribute to this dissemination since other major global ExPEC lineages (ST73 and ST95) are mostly antibiotic susceptible. To test this hypothesis, we compared early biofilm production, presence of ExPEC virulence factors (VFs), and in vivo virulence in a mouse sepsis model in 19 and 20 epidemiologically relevant strains of clades B and C, respectively. Clade B strains were significantly earlier biofilm producers (P < 0.001), carriers of more VFs (P = 4e-07), and faster killers of mice (P = 2e-10) than clade C strains. Gene inactivation experiments showed that the H30-fimB and ibeART genes were associated with in vivo virulence. Competition assays in sepsis, gut colonization, and urinary tract infection models between the most anciently diverged strain (B1 subclade), one C1 subclade strain, and a B4 subclade recombining strain harboring some clade C-specific genetic events showed that the B1 strain always outcompeted the C1 strain, whereas the B4 strain outcompeted the C1 strain, depending on the mouse niches. All these findings strongly suggest that clade C evolution includes a progressive loss of virulence involving multiple genes, possibly enhancing overall strain fitness by avoiding severe infections, even if it comes at the cost of a lower colonization ability.


Assuntos
Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Escherichia coli Extraintestinal Patogênica/genética , Sepse/microbiologia , Fatores de Virulência/genética , Virulência/genética , Animais , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/metabolismo , Evolução Molecular , Escherichia coli Extraintestinal Patogênica/efeitos dos fármacos , Escherichia coli Extraintestinal Patogênica/patogenicidade , Genótipo , Integrases/genética , Integrases/metabolismo , Estimativa de Kaplan-Meier , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Fenótipo , Infecções Urinárias/microbiologia , Sequenciamento Completo do Genoma
9.
Mol Ecol ; 28(19): 4470-4485, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31482587

RESUMO

In vitro experimental evolution has taught us many lessons on the molecular bases of adaptation. To move towards more natural settings, evolution in the mice gut has been successfully performed. Yet, these experiments suffered from the use of laboratory strains as well as the use of axenic or streptomycin-treated mice to maintain the inoculated strains. To circumvent these limitations, we conducted a one-year experimental evolution in vivo using a natural isolate of E. coli, strain 536, in conditions mimicking as much as possible natural environment with mother-to-offspring microbiota transmission. Mice were then distributed in 24 independent cages and separated into two different diets: a regular one (chow diet, CD) and high-fat and high-sugar one (Western Diet, WD). Genome sequences revealed an early and rapid selection during the breastfeeding period that selected the constitutive expression of the well-characterized lactose operon. E. coli was lost significantly more in CD than WD; however, we could not detect any genomic signature of selection, nor any diet specificities during the later part of the experiments. The apparently neutral evolution presumably due to low population size maintained nevertheless at high frequency the early selected mutations affecting lactose regulation. The rapid loss of lactose operon regulation challenges the idea that plastic gene expression is both optimal and stable in the wild.


Assuntos
Adaptação Fisiológica/genética , Escherichia coli/genética , Deriva Genética , Genoma Bacteriano/genética , Transmissão Vertical de Doenças Infecciosas , Óperon Lac/genética , Animais , Evolução Biológica , Dieta Hiperlipídica , Escherichia coli/fisiologia , Feminino , Trato Gastrointestinal/microbiologia , Camundongos , Mutação
10.
Sci Rep ; 9(1): 11331, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31383878

RESUMO

The microbiota of the human gut is a complex and rich community where bacteria and their viruses, the bacteriophages, are dominant. There are few studies on the phage community and no clear standard for isolating them, sequencing and analysing their genomes. Since this makes comparisons between studies difficult, we aimed at defining an easy, low-cost, and reproducible methodology. We analysed five different techniques to isolate phages from human adult faeces and developed an approach to analyse their genomes in order to quantify contamination and classify phage contigs in terms of taxonomy and lifestyle. We chose the polyethylene glycol concentration method to isolate phages because of its simplicity, low cost, reproducibility, and of the high number and diversity of phage sequences that we obtained. We also tested the reproducibility of this method with multiple displacement amplification (MDA) and showed that MDA severely decreases the phage genetic diversity of the samples and the reproducibility of the method. Lastly, we studied the influence of sequencing depth on the analysis of phage diversity and observed the beginning of a plateau for phage contigs at 20,000,000 reads. This work contributes to the development of methods for the isolation of phages in faeces and for their comparative analysis.


Assuntos
Bacteriófagos/genética , Intestinos/virologia , Metagenoma/genética , Filogenia , Bacteriófagos/isolamento & purificação , Biologia Computacional , Análise Custo-Benefício , Fezes , Genoma Viral , Humanos , Metagenômica , Microbiota/genética
11.
Environ Microbiol ; 21(8): 3107-3117, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31188527

RESUMO

The phylogeny of the Escherichia coli species, with the identification of seven phylogroups (A, B1, B2, C, D, E and F), is linked to the lifestyle of the strains. With the accumulation of whole genome sequence data, it became clear that some strains belong to a group intermediate between the F and B2 phylogroups, designated as phylogroup G. Here, we studied the complete sequences of 112 strains representative of the G phylogroup diversity and showed that it is composed of one main sequence type complex (STc)117 and four other STcs (STc657, STc454, STc738 and STc174). STc117, which phylogeny is characterized by very short internal branches, exhibits extensive O diversity, but little H-type and fimH allele diversity, whereas the other STcs are characterized by a main O, H and fimH type. STc117 strains possess many traits associated with extra-intestinal virulence, are virulent in a mouse sepsis model and exhibit multi-drug resistance such as CTX-M production. Epidemiologic data on 4,524 Australian and French strains suggest that STc117 is a poultry-associated lineage that can also establish in humans and cause extra-intestinal diseases. We propose an easy identification method that will help to trace this potentially virulent and resistant phylogroup in epidemiologic studies.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Infecções por Escherichia coli/microbiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Animais , Austrália , Resistência Microbiana a Medicamentos , Escherichia coli/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Filogenia , Virulência , Fatores de Virulência/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-31138573

RESUMO

We previously identified an operon involved in an arginine deiminase (ADI) pathway (arc operon) on a CTX-M-producing plasmid from an O102-ST405 strain of Escherichia coli As the ADI pathway was shown to be involved in the virulence of various Gram-positive bacteria, we tested whether the ADI pathway could be involved in the epidemiological success of extended-spectrum-ß-lactamase (ESBL)-producing E. coli strains. We studied two collections of human E. coli isolated in France (n = 493) and England (n = 1,509) and show that the prevalence of the arc operon (i) is higher in ESBL-producing strains (12.1%) than in nonproducers (2.5%), (ii) is higher in CTX-M-producing strains (16%) than in other ESBL producers (3.5%), and (iii) increased over time in ESBL-producing strains from 0% before 2000 to 43.3% in 2011 to 2012. The arc operon, found in strains from various phylogenetic backgrounds, is carried by IncF plasmids (85%) or chromosomes (15%) in regions framed by numerous insertion sequences, indicating multiple arrivals. Competition experiments showed that the arc operon enhances fitness of the strain in vitro in lysogeny broth with arginine. In vivo competition experiments showed that the arc operon is advantageous for the strain in a mouse model of urinary tract infection (UTI), whereas it is a burden in a mouse model of intestinal colonization. In summary, we have identified a trait linked to CTX-M-producing strains that is responsible for a trade-off between two main E. coli lifestyles, UTI and gut commensalism. This trait alone cannot explain the wide spread of ESBLs in E. coli but merits epidemiological surveillance.


Assuntos
Escherichia coli/genética , Hidrolases/genética , Óperon/genética , beta-Lactamases/genética , Animais , Inglaterra , Infecções por Escherichia coli/microbiologia , França , Humanos , Camundongos , Testes de Sensibilidade Microbiana/métodos , Filogenia , Plasmídeos/genética , Infecções Urinárias/microbiologia
13.
Transplantation ; 101(1): 74-82, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27681266

RESUMO

BACKGROUND: Transplant recipients are treated with immunosuppressive (IS) therapies, which impact host-microbial interactions. We examined the impact of IS drugs on gut microbiota and on the expression of ileal antimicrobial peptides. METHODS: Mice were treated for 14 days with prednisolone, mycophenolate mofetil, tacrolimus, a combination of these 3 drugs, everolimus, or water. Feces were collected before and after treatment initiation. Ileal samples were collected after sacrifice. Fecal and ileal microbiota were analyzed by pyrosequencing of 16S rRNA genes and enumeration of selected bacteria by culture, and C-type lectins were assessed in ileal tissues by reverse transcriptase-quantitative polymerase chain reaction. RESULTS: Prednisolone disrupted fecal microbiota community structure, decreased Bacteroidetes, and increased Firmicutes in the feces. Prednisolone, tacrolimus, and mycophenolate mofetil modified fecal microbiota at the family level in each experimental replicate, but changes were not consistent between the replicates. In ileal samples, the genus Clostridium sensu stricto was dramatically reduced in the prednisolone and combined IS drug groups. These modifications corresponded to an altered ileal expression of C-type lectins Reg3γ and Reg3ß, and of interleukin 22. Interestingly, the combined IS treatment enabled a commensal Escherichia coli to flourish, and dramatically increased colonization by uropathogenic E. coli strain 536. CONCLUSIONS: IS treatment alters innate antimicrobial defenses and disrupts the gut microbiota, which leads to overgrowth of indigenous E. coli and facilitates colonization by opportunistic pathogens.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Íleo/efeitos dos fármacos , Imunossupressores/toxicidade , Ácido Micofenólico/toxicidade , Prednisolona/toxicidade , Tacrolimo/toxicidade , Escherichia coli Uropatogênica/crescimento & desenvolvimento , Animais , Quimioterapia Combinada , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Fezes/microbiologia , Interações Hospedeiro-Patógeno , Íleo/imunologia , Íleo/metabolismo , Íleo/microbiologia , Hospedeiro Imunocomprometido , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Camundongos Endogâmicos C57BL , Modelos Animais , Infecções Oportunistas/imunologia , Infecções Oportunistas/microbiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribotipagem , Fatores de Tempo , Infecções Urinárias/imunologia , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/imunologia , Escherichia coli Uropatogênica/patogenicidade
14.
Mol Ecol ; 26(7): 1802-1817, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27661780

RESUMO

Although microbial ecology of the gut is now a major focus of interest, little is known about the molecular determinants of microbial adaptation in the gut. Experimental evolution coupled with whole-genome sequencing can provide insights of the adaptive process. In vitro experiments have revealed some conserved patterns: intermediate convergence, and epistatic interactions between beneficial mutations and mutations in global regulators. To test the relevance of these patterns and to identify the selective pressures acting in vivo, we have performed a long-term adaptation of an E. coli natural isolate, the streptomycin-resistant strain 536, in the digestive tract of streptomycin-treated mice. After a year of evolution, a clone from 15 replicates was sequenced. Consistently with in vitro observations, the identified mutations revealed a strong pattern of convergence at the mutation, gene, operon and functional levels. Yet, the rate of molecular evolution was lower than in in vitro, and no mutations in global regulators were recovered. More specific targets were observed: the dgo operon, involved in the galactonate pathway that improved growth on D-galactonate, and rluD and gidB, implicated in the maturation of the ribosomes, which mutations improved growth only in the presence of streptomycin. As in vitro, the nonrandom associations of mutations within the same pathways suggested a role of epistasis in shaping the adaptive landscape. Overall, we show that 'evolve and sequence' approach coupled with an analysis of convergence, when applied to a natural isolate, can be used to study adaptation in vivo and uncover the specific selective pressures of that environment.


Assuntos
Adaptação Fisiológica , Escherichia coli/genética , Evolução Molecular , Trato Gastrointestinal/microbiologia , Estreptomicina/farmacologia , Animais , Farmacorresistência Bacteriana , Escherichia coli/efeitos dos fármacos , Feminino , Genes Bacterianos , Camundongos , Mutação , Óperon
15.
J Antimicrob Chemother ; 71(11): 3072-3080, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27387322

RESUMO

OBJECTIVES: Amongst the highly diverse Escherichia coli population, the ST131-O25b:H4 clonal complex is particularly worrisome as it is associated with a high level of antibiotic resistance. The lack of new antibiotics, the worldwide continuous increase of infections caused by MDR bacteria and the need for narrow-spectrum antimicrobial agents have revived interest in phage therapy. In this article, we describe a virulent bacteriophage, LM33_P1, which specifically infects O25b strains, and provide data related to its therapeutic potential. METHODS: A large panel of E. coli strains (n = 283) was used to assess both the specificity and the activity of bacteriophage LM33_P1. Immunology, biochemistry and genetics-based methods confirmed this specificity. Virology methods and sequencing were used to characterize this bacteriophage in vitro, while three relevant mouse models were employed to show its in vivo efficacy. RESULTS: Bacteriophage LM33_P1 exclusively infects O25b E. coli strains with a 70% coverage on sequence types associated with high antibiotic resistance (ST131 and ST69). This specificity is due to an interaction with the LPS mediated by an original tail fibre. LM33_P1 also has exceptional intrinsic properties with a high adsorption constant and produces over 300 virions per cell in <10 min. Using animal pneumonia, septicaemia and urinary tract infection models, we showed the in vivo efficacy of LM33_P1 to reduce the bacterial load in several organs. CONCLUSIONS: Bacteriophage LM33_P1 represents the first weapon that specifically and quickly kills O25b E. coli strains. Therapeutic approaches derived from this bacteriophage could be developed to stop or slow down the spread of the ST131-O25b:H4 drug-resistant clonal complex in humans.


Assuntos
Colífagos/crescimento & desenvolvimento , Escherichia coli/fisiologia , Escherichia coli/virologia , Viabilidade Microbiana , Animais , Colífagos/isolamento & purificação , Modelos Animais de Doenças , Escherichia coli/classificação , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/terapia , Genoma Viral , Genótipo , Camundongos , Terapia por Fagos/métodos , Análise de Sequência de DNA
16.
PLoS One ; 11(4): e0153034, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27096607

RESUMO

Uropathogenic Escherichia coli (UPEC) strains live as commensals in the digestive tract of the host, but they can also initiate urinary tract infections. The aim of this work was to determine how a host detects the presence of a new UPEC strain in the digestive tract. Mice were orally challenged with UPEC strains 536 and CFT073, non-pathogenic strain K12 MG1655, and ΔPAI-536, an isogenic mutant of strain 536 lacking all 7 pathogenicity islands whose virulence is drastically attenuated. Intestinal colonization was measured, and cytokine expression was determined in various organs recovered from mice after oral challenge. UPEC strain 536 efficiently colonized the mouse digestive tract, and prior Enterobacteriaceae colonization was found to impact strain 536 colonization efficiency. An innate immune response, detected as the production of TNFα, IL-6 and IL-10 cytokines, was activated in the ileum 48 hours after oral challenge with strain 536, and returned to baseline within 8 days, without a drop in fecal pathogen load. Although inflammation was detected in the ileum, histology was normal at the time of cytokine peak. Comparison of cytokine secretion 48h after oral gavage with E. coli strain 536, CFT073, MG1655 or ΔPAI-536 showed that inflammation was more pronounced with UPECs than with non-pathogenic or attenuated strains. Pathogenicity islands also seemed to be involved in host detection, as IL-6 intestinal secretion was increased after administration of E. coli strain 536, but not after administration of ΔPAI-536. In conclusion, UPEC colonization of the mouse digestive tract activates acute phase inflammatory cytokine secretion but does not trigger any pathological changes, illustrating the opportunistic nature of UPECs. This digestive tract colonization model will be useful for studying the factors controlling the switch from commensalism to pathogenicity.


Assuntos
Infecções por Escherichia coli/imunologia , Imunidade Inata , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Infecções Urinárias/imunologia , Escherichia coli Uropatogênica/imunologia , Animais , Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Escherichia coli/microbiologia , Humanos , Interleucina-10/imunologia , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/imunologia , Infecções Urinárias/microbiologia
17.
Mol Biol Evol ; 33(4): 898-914, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26613786

RESUMO

In the context of the great concern about the impact of human activities on the environment, we studied 403 commensal Escherichia coli/Escherichia clade strains isolated from several animal and human populations that have variable contacts to one another. Multilocus sequence typing (MLST) showed a decrease of diversity 1) in strains isolated from animals that had an increasing contact with humans and 2) in all strains that had increased antimicrobial resistance. A specific B1 phylogroup clonal complex (CC87, Institut Pasteur schema nomenclature) of animal origin was identified and characterized as being responsible for the increased antimicrobial resistance prevalence observed in strains from the environments with a high human-mediated antimicrobial pressure. CC87 strains have a high capacity of acquiring and disseminating resistance genes with specific metabolic and genetic determinants as demonstrated by high-throughput sequencing and phenotyping. They are good mouse gut colonizers but are not virulent. Our data confirm the predominant role of human activities in the emergence of antimicrobial resistance in the environmental bacterial strains and unveil a particular E. coli clonal complex of animal origin capable of spreading antimicrobial resistance to other members of microbial communities.


Assuntos
Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/genética , Variação Genética , Animais , Anti-Infecciosos/efeitos adversos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Genótipo , Humanos , Camundongos , Tipagem de Sequências Multilocus , Filogenia
18.
J Insect Physiol ; 76: 56-66, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25747870

RESUMO

Insect molting and metamorphosis are controlled by the molt stimulating hormone ecdysone. A recent study suggests that reduced tissue oxygenation correlates with the size-sensing mechanism responsible for triggering molting. When reared in hypoxia, larvae of Manduca sexta and Drosophila melanogaster initiate molting at lower weights than do larvae reared in normoxia. Furthermore, in Drosophila, the signaling gas nitric oxide (NO) appears to be required for normal developmental timing. As observed in Drosophila, NO signaling targets the nuclear hormone receptor beta fushi tarazu transcription factor 1 (ßFTZ-F1) through activation of Drosophila hormone receptor 3 (DHR3), two key regulators of ecdysone production and metamorphic tissue progression. We set out to directly examine the effects of hypoxia and NO on ecdysone secretion using prothoracic glands from feeding fifth (last) larval stage M. sexta. Our results indicate that in vitro treatment of prothoracic glands with hypoxia (2% oxygen) or the NO donor DETA-NONOate significantly inhibit ecdysone secretion. Protein markers of glandular activity were also in keeping with an initial inhibition, measured a decrease in phosphorylated ERK (extracellular signal regulated kinase) and an increase in non-phosphorylated 4EBP (eukaryotic initiation factor 4E binding protein). Additionally, gene expression levels of Manduca hormone receptor 3 (mhr3), ßftz-f1, nitric oxide synthase (nos), and the PTTH receptor torso, were quantified using real-time PCR. NO treatment increased mhr3 expression and decreased nos expression. Hypoxia increased mhr3 transcription after 2 hr, but decreased transcription after 12 hr, with no effect on nos expression. Both NO and hypoxia had small effects on ßftz-f1 expression, yet strongly increased torso transcription. Our results demonstrate that, in isolated prothoracic glands, hypoxia and NO signaling directly inhibit ecdysteroid secretion, but at the same time alter aspects of prothoracic gland function that may enhance secretory response.


Assuntos
Ecdisona/metabolismo , Manduca/crescimento & desenvolvimento , Manduca/metabolismo , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Anaerobiose , Animais , Glândulas Endócrinas/metabolismo , Proteínas de Insetos/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Metamorfose Biológica/fisiologia , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais
19.
PLoS One ; 9(9): e108738, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25268639

RESUMO

The Escherichia coli species is divided in phylogenetic groups that differ in their virulence and commensal distribution. Strains belonging to the B2 group are involved in extra-intestinal pathologies but also appear to be more prevalent as commensals among human occidental populations. To investigate the genetic specificities of B2 sub-group, we used 128 sequenced genomes and identified genes of the core genome that showed marked difference between B2 and non-B2 genomes. We focused on the gene and its surrounding region with the strongest divergence between B2 and non-B2, the antiporter gene nhaA. This gene is part of the nhaAR operon, which is in the core genome but flanked by mobile regions, and is involved in growth at high pH and high sodium concentrations. Consistently, we found that a panel of non-B2 strains grew faster than B2 at high pH and high sodium concentrations. However, we could not identify differences in expression of the nhaAR operon using fluorescence reporter plasmids. Furthermore, the operon deletion had no differential impact between B2 and non-B2 strains, and did not result in a fitness modification in a murine model of gut colonization. Nevertheless, sequence analysis and experiments in a murine model of septicemia revealed that recombination in nhaA among B2 strains was observed in strains with low virulence. Finally, nhaA and nhaAR operon deletions drastically decreased virulence in one B2 strain. This effect of nhaAR deletion appeared to be stronger than deletion of all pathogenicity islands. Thus, a population genetic approach allowed us to identify an operon in the core genome without strong effect in commensalism but with an important role in extra-intestinal virulence, a landmark of the B2 strains.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Escherichia coli/genética , Escherichia coli/patogenicidade , Trocadores de Sódio-Hidrogênio/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/classificação , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/classificação , Proteínas de Escherichia coli/metabolismo , Feminino , Genoma Bacteriano , Concentração de Íons de Hidrogênio , Camundongos , Óperon , Concentração Osmolar , Fenótipo , Filogenia , Sepse/microbiologia , Sepse/mortalidade , Sepse/patologia , Trocadores de Sódio-Hidrogênio/classificação , Trocadores de Sódio-Hidrogênio/metabolismo , Taxa de Sobrevida , Fatores de Transcrição/classificação , Fatores de Transcrição/metabolismo , Virulência
20.
PLoS One ; 9(1): e87025, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498015

RESUMO

Escherichia coli sequence type (ST)131 is an emerging disseminated public health threat implicated in multidrug-resistant extraintestinal infections worldwide. Although the majority of ST131 isolates belong to O25b:H4 serotype, new variants with different serotypes, STs using the discriminative multilocus sequence typing scheme of Pasteur Institute, and virulence-gene profiles (virotypes) have been reported with unknown implications on the pattern of spread, persistence and virulence. The aim of the present study was to compare virulence in a mouse subcutaneous sepsis model of representative ST131 clinical isolates belonging to 2 serotypes (O25b:H4, O16:H5) and nine virotypes and subtypes (A, B, C, D1, D2, D3, D4, D5 and E). Fourteen out of the 23 ST131 isolates tested (61%) killed 90 to 100% of mice challenged, and 18 of 23 (78%) at least 50%. Interestingly, different virulence patterns in association with virotypes were observed, from highly rapid lethality (death in less than 24 h) to low final lethality (death at 7 days) but with presence of an acute inflammation. This is the first study to assess virulence of ST131 isolates belonging to serotype O16:H5, which exhibited virotype C. In spite of their low virulence-gene score, O16:H5 isolates did not show significant differences in final lethality compared with highly virulent O25b:H4 isolates of virotypes A, B and C, but killed mice less rapidly. Significant differences were found, however, between virotypes A, B, C (final lethality ≥80% of mice challenged) and virotypes D, E. Particularly unexpected was the low lethality of the newly assigned virotype E taking into account that it exhibited high virulence-gene score, and the same clonotype H30 as highly virulent O25b:H4 isolates of virotypes A, B and C. In vivo virulence diversity reported in this study would reflect the genetic variability within ST131 clonal group evidenced by molecular typing.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/patogenicidade , Sepse/microbiologia , Adesinas de Escherichia coli/genética , Animais , Animais não Endogâmicos , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Feminino , Proteínas de Fímbrias/genética , Variação Genética , Humanos , Camundongos , Tipagem de Sequências Multilocus , Filogenia , Sorogrupo , Virulência , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...