Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbes Infect ; 22(3): 144-147, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31954842

RESUMO

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.


Assuntos
Bacteriocinas/genética , Enterobactina/análogos & derivados , Família Multigênica , Peptídeos/genética , Escherichia coli Uropatogênica/genética , Enterobactina/genética , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/urina , Humanos , Policetídeos , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/patogenicidade , Virulência/genética
2.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31545853

RESUMO

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.


Assuntos
Escherichia coli/fisiologia , Mutagênicos/toxicidade , Probióticos/uso terapêutico , Animais , Antibiose/genética , Antibiose/fisiologia , Bacteriocinas/genética , Bacteriocinas/metabolismo , Bacteriocinas/toxicidade , Vias Biossintéticas/genética , Enterobactina/análogos & derivados , Enterobactina/genética , Enterobactina/fisiologia , Enterobactina/toxicidade , Escherichia coli/genética , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Feminino , Genes Bacterianos , Ilhas Genômicas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Família Multigênica , Mutação , Peptídeo Hidrolases/química , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/fisiologia , Peptídeos/genética , Peptídeos/fisiologia , Peptídeos/toxicidade , Policetídeos/toxicidade , Probióticos/toxicidade , Domínios Proteicos , Salmonelose Animal/microbiologia , Salmonelose Animal/terapia , Salmonella typhimurium , Sideróforos/genética , Sideróforos/fisiologia , Sideróforos/toxicidade , Fatores de Virulência/genética , Fatores de Virulência/fisiologia , Fatores de Virulência/toxicidade
3.
Sci Rep ; 8(1): 11245, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30050168

RESUMO

Allergic asthma is characterized by a strong Th2 and Th17 response with inflammatory cell recruitment, airways hyperreactivity and structural changes in the lung. The protease allergen papain disrupts the airway epithelium triggering a rapid eosinophilic inflammation by innate lymphoid cell type 2 (ILC2) activation, leading to a Th2 immune response. Here we asked whether the daily oral administrations of the probiotic Escherichia coli strain Nissle 1917 (ECN) might affect the outcome of the papain protease induced allergic lung inflammation in BL6 mice. We find that ECN gavage significantly prevented the severe allergic response induced by repeated papain challenges and reduced lung inflammatory cell recruitment, Th2 and Th17 response and respiratory epithelial barrier disruption with emphysema and airway hyperreactivity. In conclusion, ECN administration attenuated severe protease induced allergic inflammation, which may be beneficial to prevent allergic asthma.


Assuntos
Alérgenos/administração & dosagem , Asma/prevenção & controle , Escherichia coli/crescimento & desenvolvimento , Fatores Imunológicos/administração & dosagem , Papaína/administração & dosagem , Probióticos/administração & dosagem , Administração Oral , Animais , Asma/induzido quimicamente , Asma/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Respiratória/patologia , Células Th17/imunologia , Células Th2/imunologia , Resultado do Tratamento
4.
Front Immunol ; 8: 1096, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28959254

RESUMO

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with an increasing incidence in developed countries. Recent reports suggest that modulation of the gut microbiota might be one promising therapy for MS. Here, we investigated whether the probiotic Escherichia coli strain Nissle 1917 (ECN) could modulate the outcome of experimental autoimmune encephalomyelitis (EAE), a murine model of MS. We evidenced that daily oral treatment with ECN, but not with the archetypal K12 E. coli strain MG1655, reduced the severity of EAE induced by immunization with the MOG35-55 peptide. This beneficial effect was associated with a decreased secretion of inflammatory cytokines and an increased production of the anti-inflammatory cytokine IL-10 by autoreactive CD4 T cells, both in peripheral lymph nodes and CNS. Interestingly, ECN-treated mice exhibited increased numbers of MOG-specific CD4+ T cells in the periphery contrasting with severely reduced numbers in the CNS, suggesting that ECN might affect T cell migration from the periphery to the CNS through a modulation of their activation and/or differentiation. In addition, we demonstrated that EAE is associated with a profound defect in the intestinal barrier function and that treatment with ECN, but not with MG1655, repaired intestinal permeability dysfunction. Collectively, our data reveal that EAE induces a disruption of the intestinal homeostasis and that ECN protects from disease and restores the intestinal barrier function.

5.
mBio ; 8(2)2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28292979

RESUMO

An increasing number of human beings from developed countries are colonized by Escherichia coli strains producing colibactin, a genotoxin suspected to be associated with the development of colorectal cancers. Deoxynivalenol (DON) is the most prevalent mycotoxin that contaminates staple food-especially cereal products-in Europe and North America. This study investigates the effect of the food contaminant DON on the genotoxicity of the E. coli strains producing colibactin. In vitro, intestinal epithelial cells were coexposed to DON and E. coli producing colibactin. In vivo, newborn rats colonized at birth with E. coli producing colibactin were fed a DON-contaminated diet. Intestinal DNA damage was estimated by the phosphorylation of histone H2AX. DON exacerbates the genotoxicity of the E. coli producing colibactin in a time- and dose-dependent manner in vitro Although DON had no effect on the composition of the gut microbiota, and especially on the number of E. coli, a significant increase in DNA damage was observed in intestinal epithelial cells of animals colonized by E. coli strains producing colibactin and coexposed to DON compared to animals colonized with E. coli strains unable to produce colibactin or animals exposed only to DON. In conclusion, our data demonstrate that the genotoxicity of E. coli strains producing colibactin, increasingly present in the microbiota of asymptomatic human beings, is modulated by the presence of DON in the diet. This raises questions about the synergism between food contaminants and gut microbiota with regard to intestinal carcinogenesis.IMPORTANCE An increasing number of human beings from developed countries are colonized by Escherichia coli strains producing colibactin, a genotoxin suspected to be associated with the development of colorectal cancers. Deoxynivalenol (DON) is the most prevalent mycotoxin that contaminates staple food-especially cereal products-in Europe and North America. Our in vitro and in vivo results demonstrate that the intestinal DNA damage induced by colibactin-producing E. coli strains was exacerbated by the presence of DON in the diet. This raises questions about the synergism between food contaminants and gut microbiota with regard to intestinal carcinogenesis.


Assuntos
Dano ao DNA/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Trato Gastrointestinal/microbiologia , Mutagênicos/toxicidade , Peptídeos/toxicidade , Policetídeos/toxicidade , Tricotecenos/metabolismo , Animais , Técnicas de Cocultura , Células Epiteliais/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Histonas/análise , Ratos
6.
Bio Protoc ; 7(16): e2520, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34541180

RESUMO

Strains of Escherichia coli bearing the pks genomic island synthesize the genotoxin colibactin. Exposure of eukaryotic cells to E. coli producing colibactin induces DNA damages, ultimately leading to cell cycle arrest, senescence and death. Here we describe a simple method to demonstrate the genotoxicity of bacteria producing colibactin following a short infection of cultured mammalian cells with pks + E. coli.

7.
Infect Immun ; 83(6): 2420-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25824839

RESUMO

The intestinal barrier controls the balance between tolerance and immunity to luminal antigens. When this finely tuned equilibrium is deregulated, inflammatory disorders can occur. There is a concomitant increase, in urban populations of developed countries, of immune-mediated diseases along with a shift in Escherichia coli population from the declining phylogenetic group A to the newly dominant group B2, including commensal strains producing a genotoxin called colibactin that massively colonized the gut of neonates. Here, we showed that mother-to-offspring early gut colonization by colibactin-producing E. coli impairs intestinal permeability and enhances the transepithelial passage of luminal antigen, leading to an increased immune activation. Functionally, this was accompanied by a dramatic increase in local and systemic immune responses against a fed antigen, decreased regulatory T cell population, tolerogenic dendritic cells, and enhanced mucosal delayed-type hypersensitivity response. Conversely, the abolition of colibactin expression by mutagenesis abrogates the alteration of oral tolerance induced by neonatal colonization by E. coli. In conclusion, the vertical colonization by E. coli producing the genotoxin colibactin enhances intestinal translocation and subsequently alters oral tolerance. Thus, early colonization by E. coli from the newly dominant phylogenetic group B2, which produces colibactin, may represent a risk factor for the development of immune-mediated diseases.


Assuntos
Escherichia coli/metabolismo , Trato Gastrointestinal/microbiologia , Peptídeos/metabolismo , Policetídeos/metabolismo , Animais , Animais Recém-Nascidos , Portador Sadio , Feminino , Gravidez , Ratos , Ratos Wistar
8.
Vet Res ; 45: 76, 2014 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-25106491

RESUMO

F17 fimbriae are produced by pathogenic Escherichia coli involved in diarrhea and septicemia outbreaks in calves and lambs. These proteins result from the expression of four different clustered genes, namely f17A, f17D, f17C and f17G, encoding a pilin protein, a periplasmic protein, an anchor protein and an adhesin protein, respectively. Several variants of f17A and f17G genes have been reported and found genetically associated with typical virulence factors of bovine pathogenic E. coli strains. In this study, a new F17e-A variant, closely related to F17b-A, was identified from a collection of 58 E. coli isolates from diarrheic calves in Iran. While highly prevalent in Iranian F17-producing clinical isolates from calves, this variant was rare among E. coli from a French healthy adult bovine population, suggesting a possible association with virulence. The f17Ae gene was also found in the genome of the Shiga-like toxin variant Stx1d-producing bovine E. coli strain MHI813, and belonged to a gene cluster also encoding a new F17-G3 variant, which greatly differed from F17-G1 and F17-G2. This gene cluster was located on a pathogenicity island integrated in the tRNA pheV gene. The gene coding for a third new F17f-A variant corresponding to a combination of F17c-A and F17d-A was also identified on the pVir68 plasmid in the bovine pathogenic E. coli strain 6.0900. In conclusion, we identified three new F17-A and F17-G variants in cattle E. coli, which may also have significant impact on the development of new diagnostics and vaccination tools.


Assuntos
Doenças dos Bovinos/genética , Diarreia/veterinária , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Fímbrias Bacterianas/genética , Adesinas de Escherichia coli/genética , Adesinas de Escherichia coli/metabolismo , Animais , Sequência de Bases , Bovinos , Doenças dos Bovinos/microbiologia , Diarreia/genética , Diarreia/microbiologia , Escherichia coli/metabolismo , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Fímbrias Bacterianas/metabolismo , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase/veterinária
9.
Gut Microbes ; 5(3): 313-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24971581

RESUMO

The neonatal gut is rapidly colonized by a newly dominant group of commensal Escherichia coli strains among which a large proportion produces a genotoxin called colibactin. In order to analyze the short- and long-term effects resulting from such evolution, we developed a rat model mimicking the natural transmission of E. coli from mothers to neonates. Genotoxic and non-genotoxic E. coli strains were equally transmitted to the offspring and stably colonized the gut across generations. DNA damage was only detected in neonates colonized with genotoxic E. coli strains. Signs of genotoxic stress such as anaphase bridges, higher occurrence of crypt fission and accelerated renewal of the mature epithelium were detected at adulthood. In addition, we observed alterations of secretory cell populations and gut epithelial barrier. Our findings illustrate how critical is the genotype of E. coli strains acquired at birth for gut homeostasis at adulthood.


Assuntos
Dano ao DNA/efeitos dos fármacos , Escherichia coli/metabolismo , Trato Gastrointestinal/microbiologia , Peptídeos/metabolismo , Policetídeos/metabolismo , Animais , Feminino , Humanos , Recém-Nascido , Masculino , Modelos Animais , Gravidez , Ratos Wistar
10.
J Infect Dis ; 210(2): 285-94, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24489107

RESUMO

Sepsis is a life-threatening infection. Escherichia coli is the first known cause of bacteremia leading to sepsis. Lymphopenia was shown to predict bacteremia better than conventional markers of infection. The pks genomic island, which is harbored by extraintestinal pathogenic E. coli (ExPEC) and encodes the genotoxin colibactin, is epidemiologically associated with bacteremia. To investigate a possible relationship between colibactin and lymphopenia, we examined the effects of transient infection of lymphocytes with bacteria that were and those that were not producing the genotoxin. A mouse model of sepsis was used to compare the virulence of a clinical ExPEC isolate with its isogenic mutant impaired for the production of colibactin. We observed that colibactin induced double-strand breaks in the DNA of infected lymphocytes, leading to cell cycle arrest and to cell death by apoptosis. E. coli producing colibactin induced a more profound lymphopenia in septicemic mice, compared with the isogenic mutant unable to produce colibactin. In a sepsis model in which the mice were treated by rehydration and antibiotics, the production of colibactin by the bacteria was associated with a significantly lower survival rate. In conclusion, we demonstrate that production of colibactin by E. coli exacerbates lymphopenia associated with septicemia and could impair the chances to survive sepsis.


Assuntos
Infecções por Escherichia coli/patologia , Escherichia coli/patogenicidade , Linfopenia/induzido quimicamente , Mutagênicos/toxicidade , Peptídeos/toxicidade , Policetídeos/toxicidade , Sepse/patologia , Fatores de Virulência/toxicidade , Animais , Morte Celular , Quebras de DNA de Cadeia Dupla , Infecções por Escherichia coli/complicações , Feminino , Deleção de Genes , Linfócitos/microbiologia , Linfócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/genética , Sepse/complicações , Análise de Sobrevida , Virulência , Fatores de Virulência/genética
11.
PLoS Pathog ; 9(7): e1003437, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23853582

RESUMO

In Escherichia coli, the biosynthetic pathways of several small iron-scavenging molecules known as siderophores (enterobactin, salmochelins and yersiniabactin) and of a genotoxin (colibactin) are known to require a 4'-phosphopantetheinyl transferase (PPTase). Only two PPTases have been clearly identified: EntD and ClbA. The gene coding for EntD is part of the core genome of E. coli, whereas ClbA is encoded on the pks pathogenicity island which codes for colibactin. Interestingly, the pks island is physically associated with the high pathogenicity island (HPI) in a subset of highly virulent E. coli strains. The HPI carries the gene cluster required for yersiniabactin synthesis except for a gene coding its cognate PPTase. Here we investigated a potential interplay between the synthesis pathways leading to the production of siderophores and colibactin, through a functional interchangeability between EntD and ClbA. We demonstrated that ClbA could contribute to siderophores synthesis. Inactivation of both entD and clbA abolished the virulence of extra-intestinal pathogenic E. coli (ExPEC) in a mouse sepsis model, and the presence of either functional EntD or ClbA was required for the survival of ExPEC in vivo. This is the first report demonstrating a connection between multiple phosphopantetheinyl-requiring pathways leading to the biosynthesis of functionally distinct secondary metabolites in a given microorganism. Therefore, we hypothesize that the strict association of the pks island with HPI has been selected in highly virulent E. coli because ClbA is a promiscuous PPTase that can contribute to the synthesis of both the genotoxin and siderophores. The data highlight the complex regulatory interaction of various virulence features with different functions. The identification of key points of these networks is not only essential to the understanding of ExPEC virulence but also an attractive and promising target for the development of anti-virulence therapy strategies.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Mutagênicos/metabolismo , Peptídeos/metabolismo , Policetídeos/metabolismo , Sideróforos/biossíntese , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Animais , Proteínas de Bactérias/genética , Enterobactina/análogos & derivados , Enterobactina/biossíntese , Escherichia coli/enzimologia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Deleção de Genes , Ilhas Genômicas , Glicopeptídeos/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Fenóis/metabolismo , Sepse/metabolismo , Sepse/microbiologia , Tiazóis/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Virulência
12.
Gut Microbes ; 3(6): 501-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22895085

RESUMO

Oral administration of the probiotic bacterium Escherichia coli Nissle 1917 improves chronic inflammatory bowel diseases, but the molecular basis for this therapeutic efficacy is unknown. E. coli Nissle 1917 harbors a cluster of genes coding for the biosynthesis of hybrid nonribosomal peptide-polyketide(s). This biosynthetic pathway confers the ability for bacteria to induce DNA double strand breaks in eukaryotic cells. Here we reveal that inactivation of the clbA gene within this genomic island abrogated the ability for the strain to induce DNA damage and chromosomal abnormalities in non-transformed cultured rat intestinal epithelial cells but is required for the probiotic activity of E. coli Nissle 1917. Thus, evaluation of colitis severity induced in rodent fed with E. coli Nissle 1917 or an isogenic non-genotoxic mutant demonstrated the need for a functional biosynthetic pathway both in the amelioration of the disease and in the modulation of cytokine expression. Feeding rodents with a complemented strain for which genotoxicity was restored confirmed that this biosynthetic pathway contributes to the health benefits of the probiotic by modulating its immunomodulatory properties. Our data provide additional evidence for the benefit of this currently used probiotic in colitis but remind us that an efficient probiotic may also have side effects as any other medication.


Assuntos
Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Doenças Inflamatórias Intestinais/terapia , Mutagênicos/metabolismo , Probióticos/administração & dosagem , Animais , Linhagem Celular , Colite/microbiologia , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Escherichia coli/genética , Técnicas de Inativação de Genes , Masculino , Policetídeos/metabolismo , Ratos , Ratos Wistar
13.
J Biol Chem ; 286(41): 35562-35570, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21795676

RESUMO

The pks genomic island of Escherichia coli encodes polyketide (PK) and nonribosomal peptide (NRP) synthases that allow assembly of a putative hybrid PK-NRP compound named colibactin that induces DNA double-strand breaks in eukaryotic cells. The pks-encoded machinery harbors an atypical essential protein, ClbP. ClbP crystal structure and mutagenesis experiments revealed a serine-active site and original structural features compatible with peptidase activity, which was detected by biochemical assays. Ten ClbP homologs were identified in silico in NRP genomic islands of closely and distantly related bacterial species. All tested ClbP homologs were able to complement a clbP-deficient E. coli mutant. ClbP is therefore a prototype of a new subfamily of extracytoplasmic peptidases probably involved in the maturation of NRP compounds. Such peptidases will be powerful tools for the manipulation of NRP biosynthetic pathways.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Peptídeo Hidrolases/química , Cristalografia por Raios X , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Técnicas de Silenciamento de Genes , Teste de Complementação Genética , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Policetídeo Sintases/química , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(25): 11537-42, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20534522

RESUMO

Escherichia coli is a normal inhabitant of the human gut. However, E. coli strains of phylogenetic group B2 harbor a genomic island called "pks" that codes for the production of a polyketide-peptide genotoxin, Colibactin. Here we report that in vivo infection with E. coli harboring the pks island, but not with a pks isogenic mutant, induced the formation of phosphorylated H2AX foci in mouse enterocytes. We show that a single, short exposure of cultured mammalian epithelial cells to live pks(+) E. coli at low infectious doses induced a transient DNA damage response followed by cell division with signs of incomplete DNA repair, leading to anaphase bridges and chromosome aberrations. Micronuclei, aneuploidy, ring chromosomes, and anaphase bridges persisted in dividing cells up to 21 d after infection, indicating occurrence of breakage-fusion-bridge cycles and chromosomal instability. Exposed cells exhibited a significant increase in gene mutation frequency and anchorage-independent colony formation, demonstrating the infection mutagenic and transforming potential. Therefore, colon colonization with these E. coli strains harboring the pks island could contribute to the development of sporadic colorectal cancer.


Assuntos
Dano ao DNA , Escherichia coli/metabolismo , Instabilidade Genômica , Intestinos/microbiologia , Animais , Células CHO , Adesão Celular , Aberrações Cromossômicas , Cricetinae , Cricetulus , Reparo do DNA , Escherichia coli/genética , Camundongos , Camundongos Endogâmicos BALB C , Mutagênicos/metabolismo , Mutação , Neoplasias/metabolismo
15.
Infect Immun ; 77(1): 492-500, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18981247

RESUMO

Five types of cytolethal distending toxin (CDT-I to CDT-V) have been identified in Escherichia coli. In the present study we cloned and sequenced the cdt-IV operon and flanking region from a porcine extraintestinal pathogenic E. coli (ExPEC) strain belonging to serogroup O75. We confirmed that similar to other CDTs, CDT-IV induced phosphorylation of host histone H2AX, a sensitive marker of DNA double-strand breaks, and blocked the HeLa cell cycle at the G(2)-M transition. The cdt-IV genes were framed by lambdoid prophage genes. We cloned and sequenced the cdt-I operon and flanking regions from a human ExPEC O18:K1:H7 strain and observed that cdt-I genes were also flanked by lambdoid prophage genes. PCR studies indicated that a gene coding for a putative protease was always associated with the cdtC-IV gene but was not associated with cdtC genes in strains producing CDT-I, CDT-III, and CDT-V. Our results suggest that the cdt-I and cdt-IV genes might have been acquired from a common ancestor by phage transduction and evolved in their bacterial hosts. The lysogenic bacteriophages have the potential to carry nonessential "cargo" genes or "morons" and therefore play a crucial role in the generation of genetic diversity within ExPEC.


Assuntos
Toxinas Bacterianas/genética , DNA Bacteriano/genética , Escherichia coli/genética , Escherichia coli/virologia , Prófagos/genética , Toxinas Bacterianas/toxicidade , Clonagem Molecular , DNA Bacteriano/química , Escherichia coli/isolamento & purificação , Evolução Molecular , Transferência Genética Horizontal , Células HeLa , Humanos , Dados de Sequência Molecular , Análise de Sequência de DNA
16.
Science ; 313(5788): 848-51, 2006 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16902142

RESUMO

Transient infection of eukaryotic cells with commensal and extraintestinal pathogenic Escherichia coli of phylogenetic group B2 blocks mitosis and induces megalocytosis. This trait is linked to a widely spread genomic island that encodes giant modular nonribosomal peptide and polyketide synthases. Contact with E. coli expressing this gene cluster causes DNA double-strand breaks and activation of the DNA damage checkpoint pathway, leading to cell cycle arrest and eventually to cell death. Discovery of hybrid peptide-polyketide genotoxins in E. coli will change our view on pathogenesis and commensalism and open new biotechnological applications.


Assuntos
Citotoxinas/metabolismo , Dano ao DNA , Escherichia coli/fisiologia , Escherichia coli/patogenicidade , Ilhas Genômicas , Mutagênicos/metabolismo , Peptídeos/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Linhagem Celular , Núcleo Celular/química , DNA/análise , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/genética , Fase G2 , Células HeLa , Histonas/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Dados de Sequência Molecular , Mutagênese , Fosforilação , Policetídeo Sintases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
17.
Mol Microbiol ; 50(5): 1553-67, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14651638

RESUMO

Enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC) are closely related pathogens. Both use a type III secretion system (TTSS) encoded by the 'locus of enterocyte effacement' (LEE) to subvert and attach to epithelial cells through the injection of a repertoire of effector molecules. Here, we report the identification of a new TTSS translocated effector molecule called Cif, which blocks cell cycle G2/M transition and induces the formation of stress fibres through the recruitment of focal adhesions. Cif is not encoded by the LEE but by a lambdoid prophage present in EPEC and EHEC. A cif mutant causes localized effacement of microvilli and intimately attaches to the host cell surface, but is defective in the ability to block mitosis. When expressed in TTSS competent LEE-positive pathogens, Cif is injected into the infected epithelial cells. These cells arrested at the G2/M phase displayed accumulation of inactive phosphorylated Cdk1. In conclusion, Cif is a new member of a growing family of bacterial cyclomodulins that subvert the host eukaryotic cell cycle.


Assuntos
Escherichia coli O157/patogenicidade , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Fase G2/efeitos dos fármacos , Mitose/efeitos dos fármacos , Actinas/efeitos dos fármacos , Actinas/metabolismo , Bacteriófago lambda , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/metabolismo , Ciclo Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/metabolismo , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/farmacologia , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Prófagos
18.
Microbes Infect ; 5(10): 857-67, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12919854

RESUMO

Enteropathogenic Escherichia coli (EPEC), a major cause of severe disease with diarrhea in infants, is also involved in weaned rabbit colibacillosis. EPEC O103 is frequent in rabbit-fattening units of Western Europe. It causes high mortality and growth retardation, leading to substantial economic losses. We report here the construction by allelic exchange of an EPEC O103 strain mutated in espB and tir, two essential virulence genes. Upon live oral administration to weaned rabbits, the E22DeltaTir/EspB mutant strain efficiently colonized the intestinal tract without any adverse consequences. The rabbits were challenged with the highly pathogenic parental strain E22. The mutant provided complete protection to rabbits and total resistance to intestinal colonization by E22. In addition, E22DeltaTir/EspB strain induced a specific humoral response against the bacterial adhesin AF/R2. These Abs prevent bacterial attachment to epithelial cells in vitro. These results open the way for the development of an efficient vaccine strategy against rabbit EPEC infections.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/genética , Escherichia coli/imunologia , Adesinas de Escherichia coli/imunologia , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Peso Corporal , Diarreia/imunologia , Diarreia/microbiologia , Ensaio de Imunoadsorção Enzimática , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Deleção de Genes , Células HeLa , Humanos , Intestinos/microbiologia , Mutagênese Insercional , Coelhos , Receptores de Superfície Celular/genética , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...