Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 433(21): 167188, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34454944

RESUMEN

Type III protein secretion is widespread in Gram-negative pathogens. It comprises the injectisome with a surface-exposed needle and an inner membrane translocase. The translocase contains the SctRSTU export channel enveloped by the export gate subunit SctV that binds chaperone/exported clients and forms a putative ante-chamber. We probed the assembly, function, structure and dynamics of SctV from enteropathogenic E. coli (EPEC). In both EPEC and E. coli lab strains, SctV forms peripheral oligomeric clusters that are detergent-extracted as homo-nonamers. Membrane-embedded SctV9 is necessary and sufficient to act as a receptor for different chaperone/exported protein pairs with distinct C-domain binding sites that are essential for secretion. Negative staining electron microscopy revealed that peptidisc-reconstituted His-SctV9 forms a tripartite particle of ∼22 nm with a N-terminal domain connected by a short linker to a C-domain ring structure with a ∼5 nm-wide inner opening. The isolated C-domain ring was resolved with cryo-EM at 3.1 Å and structurally compared to other SctV homologues. Its four sub-domains undergo a three-stage "pinching" motion. Hydrogen-deuterium exchange mass spectrometry revealed this to involve dynamic and rigid hinges and a hyper-flexible sub-domain that flips out of the ring periphery and binds chaperones on and between adjacent protomers. These motions are coincident with local conformational changes at the pore surface and ring entry mouth that may also be modulated by the ATPase inner stalk. We propose that the intrinsic dynamics of the SctV protomer are modulated by chaperones and the ATPase and could affect allosterically the other subunits of the nonameric ring during secretion.


Asunto(s)
Adenosina Trifosfatasas/química , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/química , Flagelos/ultraestructura , Canales de Translocación SEC/química , Sistemas de Secreción Tipo III/ultraestructura , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Regulación Alostérica , Sitios de Unión , Clonación Molecular , Microscopía por Crioelectrón , Medición de Intercambio de Deuterio , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelos/metabolismo , Expresión Génica , Regulación Bacteriana de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Cinética , Espectrometría de Masas , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo , Especificidad por Sustrato , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo
2.
EBioMedicine ; 43: 325-332, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31036531

RESUMEN

BACKGROUND: The predominance of specific bacteria such as adherent-invasive Escherichia coli (AIEC) within the Crohn's disease (CD) intestine remains poorly understood with little evidence uncovered to support a selective pressure underlying their presence. Intestinal ethanolamine is however readily accessible during periods of intestinal inflammation, and enables pathogens to outcompete the host microbiota under such circumstances. METHODS: Quantitative RT-PCR (qRT-PCR) to determine expression of genes central to ethanolamine metabolism; transmission electron microscopy to detect presence of bacterial microcompartments (MCPs); in vitro infections of both murine and human macrophage cell lines examining intracellular replication of the AIEC-type strain LF82 and clinical E. coli isolates in the presence of ethanolamine; determination of E. coli ethanolamine utilization (eut) operon transcription in faecal samples from healthy patients, patients with active CD and the same patients in remission following treatment. RESULTS: Growth on the intestinal short chain fatty acid propionic acid (PA) stimulates significantly increased transcription of the eut operon (fold change relative to glucose: >16.9; p-value <.01). Additionally ethanolamine was accessible to intra-macrophage AIEC and stimulated significant increases in growth intracellularly when it was added extracellularly at concentrations comparable to those in the human intestine. Finally, qRT-PCR indicated that expression of the E. coli eut operon was increased in children with active CD compared to healthy controls (fold change increase: >4.72; P < .02). After clinical remission post-exclusive enteral nutrition treatment, the same CD patients exhibited significantly reduced eut expression (Pre vs Post fold change decrease: >15.64; P < .01). INTERPRETATION: Our data indicates a role for ethanolamine metabolism in selecting for AIEC that are consistently overrepresented in the CD intestine. The increased E. coli metabolism of ethanolamine seen in the intestine during active CD, and its decrease during remission, indicates ethanolamine use may be a key factor in shaping the intestinal microbiome in CD patients, particularly during times of inflammation. FUND: This work was funded by Biotechnology and Biological Sciences Research Council (BBSRC) grants BB/K008005/1 & BB/P003281/1 to DMW; by a Tenovus Scotland grant to MJO; by Glasgow Children's Hospital Charity, Nestle Health Sciences, Engineering and Physical Sciences Research Council (EPSRC) and Catherine McEwan Foundation grants awarded to KG; and by a Natural Environment Research Council (NERC) fellowship (NE/L011956/1) to UZI. The IBD team at the Royal Hospital for Children, Glasgow are supported by the Catherine McEwan Foundation and Yorkhill IBD fund. RKR and RH are supported by NHS Research Scotland Senior fellowship awards.


Asunto(s)
Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/metabolismo , Escherichia coli Enteropatógena , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/microbiología , Etanolamina/metabolismo , Animales , Línea Celular , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Escherichia coli Enteropatógena/fisiología , Escherichia coli Enteropatógena/ultraestructura , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/patología , Ácidos Grasos/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Operón
3.
J Microbiol ; 54(11): 745-752, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27796929

RESUMEN

Using clonal phylogenetic methods, it has been demonstrated that O111:H25 atypical enteropathogenic E. coli (aEPEC) strains belong to distinct clones, suggesting the possibility that their ability to interact with different hosts and abiotic surfaces can vary from one clone to another. Accordingly, the ability of O111:H25 aEPEC strains derived from human, cat and dogs to adhere to epithelial cells has been investigated, along with their ability to interact with macrophages and to form biofilms on polystyrene, a polymer used to make biomedical devices. The results demonstrated that all the strains analyzed were able to adhere to, and to form pedestals on, epithelial cells, mechanisms used by E. coli to become strongly attached to the host. The strains also show a Localized-Adherence-Like (LAL) pattern of adhesion on HEp-2 cells, a behavior associated with acute infantile diarrhea. In addition, the O111:H25 aEPEC strains derived either from human or domestic animals were able to form long filaments, a phenomenon used by some bacteria to avoid phagocytosis. O111:H25 aEPEC strains were also encountered inside vacuoles, a characteristic described for several bacterial strains as a way of protecting themselves against the environment. They were also able to induce TNF-α release via two routes, one dependent on TLR-4 and the other dependent on binding of Type I fimbriae. These O111:H25 strains were also able to form biofilms on polystyrene. In summary the results suggest that, regardless of their source (i.e. linked to human origin or otherwise), O111:H25 aEPEC strains carry the potential to cause human disease.


Asunto(s)
Adhesión Bacteriana , Escherichia coli Enteropatógena/metabolismo , Escherichia coli Enteropatógena/patogenicidad , Infecciones por Escherichia coli/microbiología , Animales , Biopelículas/crecimiento & desarrollo , Gatos , Perros , Escherichia coli Enteropatógena/aislamiento & purificación , Escherichia coli Enteropatógena/ultraestructura , Células Epiteliales/microbiología , Proteínas de Escherichia coli , Fimbrias Bacterianas/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Filogenia , Poliestirenos , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Virulencia
4.
J Bacteriol ; 198(5): 846-56, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26712935

RESUMEN

UNLABELLED: Enteropathogenic Escherichia coli (EPEC) remains a significant cause of infant diarrheal illness and associated morbidity and mortality in developing countries. EPEC strains are characterized by their ability to colonize the small intestines of their hosts by a multistep program involving initial loose attachment to intestinal epithelial cells followed by an intimate adhesion phase. The initial loose interaction of typical EPEC with host intestinal cells is mediated by bundle-forming pili (BFP). BFP are type 4b pili (T4bP) based on structural and functional properties shared with T4bP expressed by other bacteria. The major structural subunit of BFP is called bundlin, a T4b pilin expressed from the bfpA gene in the BFP operon, which contains three additional genes that encode the pilin-like proteins BfpI, BfpJ, and BfpK. In this study, we show that, in the absence of the BFP retraction ATPase (BfpF), BfpI, BfpJ, and BfpK are dispensable for BFP biogenesis. We also demonstrate that these three minor pilins are incorporated along with bundlin into the BFP filament and contribute to its structural integrity and host cell adhesive properties. The results confirm that previous findings in T4aP systems can be extended to a model T4bP such as BFP. IMPORTANCE: Bundle-forming pili contribute to the host colonization strategy of enteropathogenic Escherichia coli. The studies described here investigate the role for three minor pilin subunits in the structure and function of BFP in EPEC. The studies also suggest that these subunits could be antigens for vaccine development.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/genética , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Inmunohistoquímica , Mutación
5.
Infect Immun ; 82(5): 1801-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24549324

RESUMEN

Enteropathogenic Escherichia coli (EPEC) is recognized as an important intestinal pathogen that frequently causes acute and persistent diarrhea in humans and animals. The use of probiotic bacteria to prevent diarrhea is gaining increasing interest. The probiotic E. coli strain Nissle 1917 (EcN) is known to be effective in the treatment of several gastrointestinal disorders. While both in vitro and in vivo studies have described strong inhibitory effects of EcN on enteropathogenic bacteria, including pathogenic E. coli, the underlying molecular mechanisms remain largely unknown. In this study, we examined the inhibitory effect of EcN on infections of porcine intestinal epithelial cells with atypical enteropathogenic E. coli (aEPEC) with respect to single infection steps, including adhesion, microcolony formation, and the attaching and effacing phenotype. We show that EcN drastically reduced the infection efficiencies of aEPEC by inhibiting bacterial adhesion and growth of microcolonies, but not the attaching and effacing of adherent bacteria. The inhibitory effect correlated with EcN adhesion capacities and was predominantly mediated by F1C fimbriae, but also by H1 flagella, which served as bridges between EcN cells. Furthermore, EcN seemed to interfere with the initial adhesion of aEPEC to host cells by secretion of inhibitory components. These components do not appear to be specific to EcN, but we propose that the strong adhesion capacities enable EcN to secrete sufficient local concentrations of the inhibitory factors. The results of this study are consistent with a mode of action whereby EcN inhibits secretion of virulence-associated proteins of EPEC, but not their expression.


Asunto(s)
Escherichia coli Enteropatógena/fisiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/clasificación , Proteínas Fimbrias/metabolismo , Flagelos/fisiología , Probióticos/farmacología , Animales , Adhesión Bacteriana , Línea Celular , Escherichia coli Enteropatógena/patogenicidad , Escherichia coli Enteropatógena/ultraestructura , Células Epiteliales/microbiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Mucosa Intestinal/citología , Porcinos , Virulencia
6.
PLoS One ; 8(1): e55284, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383137

RESUMEN

Enteropathogenic E. coli (EPEC) is a human pathogen that targets the small intestine, causing severe and often fatal diarrhoea in infants. A defining feature of EPEC disease is the loss (effacement) of absorptive microvilli (MV) from the surface of small intestinal enterocytes. Much of our understanding of EPEC pathogenesis is derived from studies using cell lines such as Caco-2 - the most extensively used small intestinal model. However, previous work has revealed fundamental differences between Caco-2 cells and in vivo differentiated enterocytes in relation to MV effacement. This, and the high heterogeneity and low transfection efficiency of the Caco-2 cell line prompted the isolation of several sub-clones (NCL-1-12) to identify a more tractable and improved in vivo-like cell model. Along with established Caco-2 clones (TC-7, BBE1), sub-clones were assessed for growth rate, apical surface morphology, epithelial barrier function and transfection efficiency. TC-7 cells provided the best all-round clone and exhibited highest levels of ectopic gene expression following cell polarisation. Novel alterations in EGFP-labelled mitochondria, that were not previously documented in non-polarised cell types, highlighted the potential of the TC-7 model for defining dynamic enterocyte-specific changes during infection. Crucially, the TC-7 cell line also mimicked ex vivo derived enterocytes with regard to MV effacement, enabling a better dissection of the process. Effacement activity caused by the EPEC protein Map in the Caco-2 but not ex vivo model, was linked to a defect in suppressing its Cdc42-dependent functionality. MV effacement activity of the EPEC protein EspF in the TC-7 model was dependent on its N-WASP binding motif, which is also shown to play an essential role in epithelial barrier dysfunction. Together, this study highlights the many advantages of using TC-7 cells as a small intestinal model to study host-pathogen interactions.


Asunto(s)
Enterocitos/microbiología , Escherichia coli Enteropatógena/patogenicidad , Intestino Delgado/citología , Modelos Animales , Análisis de Varianza , Células CACO-2 , Impedancia Eléctrica , Enterocitos/patología , Escherichia coli Enteropatógena/ultraestructura , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Microscopía Confocal , Microscopía Electrónica de Rastreo
7.
Comp Immunol Microbiol Infect Dis ; 35(6): 539-49, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22768807

RESUMEN

Atypical enteropathogenic Escherichia coli (aEPEC) are heterogeneous in terms of serotypes, adherence patterns and the presence of non-locus of enterocyte effacement virulence factors. In this study, the low-molecular mass proteomes of four representative aEPEC, comprising three different adhesion phenotypes (localized-like, aggregative and diffuse) and one non-adherent isolate, were analyzed and compared by 2D gel electrophoresis and LC-MS/MS. By mass spectrometry, a total of 59 proteins were identified according to their annotated function, with most of them being involved in metabolism, protection, and transport; some of them still classified as hypothetical proteins. Thus, in this comparative proteomic analysis of low-molecular mass extracted proteins from different aEPEC isolates, the proteins identified are mainly involved in key metabolic pathways. Also, the majority of the hypothetical and filamentous proteins identified in the isolates studied are products of genes originally identified in the genome of enterohemorrhagic E. coli.


Asunto(s)
Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Proteoma/genética , Factores de Virulencia/genética , Adhesión Bacteriana , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Escherichia coli Enteropatógena/clasificación , Escherichia coli Enteropatógena/aislamiento & purificación , Escherichia coli Enteropatógena/ultraestructura , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Humanos , Microscopía Electrónica de Transmisión , Peso Molecular , Proteoma/metabolismo , Proteómica , Espectrometría de Masas en Tándem , Factores de Virulencia/metabolismo
8.
J Bacteriol ; 194(7): 1646-58, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22247509

RESUMEN

Type IV pili (T4P) are filamentous surface appendages required for tissue adherence, motility, aggregation, and transformation in a wide array of bacteria and archaea. The bundle-forming pilus (BFP) of enteropathogenic Escherichia coli (EPEC) is a prototypical T4P and confirmed virulence factor. T4P fibers are assembled by a complex biogenesis machine that extrudes pili through an outer membrane (OM) pore formed by the secretin protein. Secretins constitute a superfamily of proteins that assemble into multimers and support the transport of macromolecules by four evolutionarily ancient secretion systems: T4P, type II secretion, type III secretion, and phage assembly. Here, we determine that the lipoprotein transport pathway is not required for targeting the BfpB secretin protein of the EPEC T4P to the OM and describe the ultrastructure of the single particle averaged structures of the assembled complex by transmission electron microscopy. Furthermore, we use photoactivated localization microscopy to determine the distribution of single BfpB molecules fused to photoactivated mCherry. In contrast to findings in other T4P systems, we found that BFP components predominantly have an uneven distribution through the cell envelope and are only found at one or both poles in a minority of cells. In addition, we report that concurrent mutation of both the T4bP secretin and the retraction ATPase can result in viable cells and found that these cells display paradoxically low levels of cell envelope stress response activity. These results imply that secretins can direct their own targeting, have complex distributions and provide feedback information on the state of pilus biogenesis.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Fimbrias Bacterianas/metabolismo , Lipoproteínas/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/ultraestructura , Lipoproteínas/genética , Transporte de Proteínas
9.
Anat Rec (Hoboken) ; 295(2): 201-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22190417

RESUMEN

Enteropathogenic Escherichia coli (EPEC) manipulate the cytoskeleton of host intestinal epithelial cells, producing membrane protrusions termed pedestals that the bacteria reside on throughout the course of their infections. By definition pedestals are actin-based structures, however recent work has identified the spectrin cytoskeleton as a necessary component of EPEC pedestals. Here, we investigated the detailed arrangement of the spectrin and actin cytoskeletons within these structures. Immunofluorescent imaging revealed that the spectrin network forms a peripheral cage around actin at the membranous regions of pedestals. Myosin S1 fragment decorated actin filaments examined by electron microscopy demonstrated that actin filaments orientate with their fast-growing barbed ends toward the lateral membranes of EPEC pedestals. These findings provide a detailed descriptive analysis, which further illustrate the spectrin cytoskeletal organization within these structures.


Asunto(s)
Extensiones de la Superficie Celular/ultraestructura , Citoesqueleto/ultraestructura , Escherichia coli Enteropatógena/ultraestructura , Células HeLa/ultraestructura , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Animales , Extensiones de la Superficie Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Escherichia coli Enteropatógena/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Células HeLa/metabolismo , Células HeLa/microbiología , Humanos , Masculino , Microscopía Electrónica de Transmisión , Subfragmentos de Miosina/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrina/metabolismo
10.
BMC Microbiol ; 10: 57, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20175929

RESUMEN

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) are enteropathogenic strains identified by the aggregative adhesion (AA) pattern that share the capability to form biofilms. Citrobacter freundii is classically considered as an indigenous intestinal species that is sporadically associated with diarrhea. RESULTS: During an epidemiologic study focusing on infantile diarrhea, aggregative C. freundii (EACF) and EAEC strains were concomitantly recovered from a severe case of mucous diarrhea. Thereby, the occurrence of synergic events involving these strains was investigated. Coinfection of HeLa cells with EACF and EAEC strains showed an 8-fold increase in the overall bacterial adhesion compared with single infections (P < 0.001). The synergic effect was mediated by physical interactions among the bacteria and primed in the absence of chemical signaling and without the participation of host cells. Thus, significant increases (2.7-fold on average) in bacterial adhesion were also observed during the formation of mixed biofilms on abiotic surfaces. Bacterial settling assays showed that EAEC strains harboring F-pili genes (traA) were capable of forming bacterial aggregates only in the presence of EACF. Scanning electronic microscopy analyses revealed that bacterial aggregates as well as enhanced biofilms formed by EACF and traA-positive EAEC were mediated by non-bundle forming, flexible pili. Moreover, mixed biofilms formed by EACF and traA-positive EAEC strains were significantly reduced using nonlethal concentration of zinc, a specific inhibitor of F pili. In addition, EAEC strains isolated from diarrheic children frequently produced single biofilms sensitive to zinc. CONCLUSIONS: Putative F pili expressed by EAEC strains boosted mixed biofilm formation when in the presence of aggregative C. freundii.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Citrobacter freundii/fisiología , Diarrea/microbiología , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enteropatógena/fisiología , Proteínas de Escherichia coli/fisiología , Proteínas Fimbrias/fisiología , Adhesinas de Escherichia coli/genética , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Adhesión Bacteriana/genética , Biopelículas/efectos de los fármacos , Citrobacter freundii/patogenicidad , Citrobacter freundii/ultraestructura , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Vidrio , Células HeLa , Humanos , Lactante , Microscopía Electrónica de Rastreo , Percepción de Quorum , Zinc/farmacología
11.
Phytother Res ; 23(9): 1229-36, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19441013

RESUMEN

Bacterial adhesion is the first step in the sequence of events leading to infection. Previous data are available on the effect of Holarrhena antidysenterica on antidiarrhoeal and antibacterial action, but there is little information on the mechanism of action of the various aspects of EPEC-induced diarrhoea, namely adherence and translocation of the effector molecule to intestinal epithelial cells. The aim of the present study was to investigate the effects of alkaloids of Holarrhena antidysenterica (AHA) on interference in the mechanism of enteropathogenic Escherichia coli (EPEC) adhesion on host epithelial cells (INT 407 and HEp2). To determine the impact of AHA on epithelial cells, cytotoxicity (LDH), adherence, apoptotic and ultrastructural studies were performed. To analyse the effect of AHA on EPEC secreted proteins, especially EspD, INT 407 monolayers were infected with EPEC and AHA-treated EPEC, followed by immunoblotting, probed with anti EspD antisera. The maximum percentage of LDH leakage was reduced in AHA-treated EPEC (400 microg/mL) in both cell lines. Reduced bacterial adherence was observed under light microscopy and altered apoptotic changes were visualized using propidium iodide staining in conjunction with fluorescence microscopy, in both cell lines infected with AHA-treated EPEC and these results were confirmed with transmission electron microscope images. The suppression of type III secretory proteins (TTSPs), EspD ( approximately 40 kDa), was detected in INT 407 cells infected with AHA-treated EPEC. In conclusion, AHA reduces initial bacterial adhesion to intact epithelial cells and it may exert an antiadherence effect against the pathogenesis of EPEC in host epithelial cells. Thus, the investigations provide a rational basis for the treatment of EPEC-mediated diarrhoea with AHA.


Asunto(s)
Alcaloides/farmacología , Adhesión Bacteriana/efectos de los fármacos , Escherichia coli Enteropatógena/efectos de los fármacos , Células Epiteliales/microbiología , Holarrhena/química , Alcaloides/aislamiento & purificación , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Línea Celular , Escherichia coli Enteropatógena/metabolismo , Escherichia coli Enteropatógena/ultraestructura , Células Epiteliales/ultraestructura , Humanos , Lactato Deshidrogenasas/metabolismo , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Extractos Vegetales/farmacología
12.
J Bacteriol ; 191(11): 3451-61, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19218393

RESUMEN

Although the bundle-forming pilus (BFP) of enteropathogenic Escherichia coli (EPEC) mediates microcolony formation on epithelial cells, the adherence of BFP-deficient mutants is significantly abrogated, but the mutants are still adherent due to the presence of intimin and possibly other adhesins. In this study we investigated the contribution of the recently described E. coli common pilus (ECP) to the overall adherence properties of EPEC. We found that ECP and BFP structures can be simultaneously observed in the course (between zero time and 7 h during infection) of formation of localized adherence on cultured epithelial cells. These two pilus types colocalized at different levels of the microcolony topology, tethering the adhering bacteria. No evidence of BFP disappearance was found after prolonged infection. When expressed from a plasmid present in nonadherent E. coli HB101, ECP rendered this organism highly adherent at levels comparable to those of HB101 expressing the BFP. Purified ECP bound in a dose-dependent manner to epithelial cells, and the binding was blocked with anti-ECP antibodies, confirming that the pili possess adhesin properties. An ECP mutant showed only a modest reduction in adherence to cultured cells due to background expression levels of BFP and intimin. However, isogenic mutants not expressing EspA or BFP were significantly less adherent when the ecpA gene was also deleted. Furthermore, a DeltaespA DeltaecpA double mutant (unable to translocate Tir and to establish intimate adhesion) was at least 10-fold less adherent than the DeltaespA and DeltaecpA single mutants, even in the presence of BFP. A Delta bfp DeltaespA DeltaecpA triple mutant showed the least adherence compared to the wild type and all the isogenic mutant strains tested, suggesting that ECP plays a synergistic role in adherence. Our data indicate that ECP is an accessory factor that, in association with BFP and other adhesins, contributes to the multifactorial complex interaction of EPEC with host epithelial cells.


Asunto(s)
Adhesión Bacteriana/fisiología , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/fisiología , Proteínas Fimbrias/fisiología , Fimbrias Bacterianas/metabolismo , Adhesión Bacteriana/genética , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/ultraestructura , Células Epiteliales/microbiología , Proteínas de Escherichia coli/genética , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Citometría de Flujo , Células HT29 , Células HeLa , Humanos , Microscopía Electrónica de Transmisión
13.
J Bacteriol ; 190(14): 5063-74, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18502854

RESUMEN

Enterohemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC, respectively) strains represent a major global health problem. Their virulence is mediated by the concerted activity of an array of virulence factors including toxins, a type III protein secretion system (TTSS), pili, and others. We previously showed that EPEC O127 forms a group 4 capsule (G4C), and in this report we show that EHEC O157 also produces a G4C, whose assembly is dependent on the etp, etk, and wzy genes. We further show that at early time points postinfection, these G4Cs appear to mask surface structures including intimin and the TTSS. This masking inhibited the attachment of EPEC and EHEC to tissue-cultured epithelial cells, diminished their capacity to induce the formation of actin pedestals, and attenuated TTSS-mediated protein translocation into host cells. Importantly, we found that Ler, a positive regulator of intimin and TTSS genes, represses the expression of the capsule-related genes, including etp and etk. Thus, the expression of TTSS and G4C is conversely regulated and capsule production is diminished upon TTSS expression. Indeed, at later time points postinfection, the diminishing capsule no longer interferes with the activities of intimin and the TTSS. Notably, by using the rabbit infant model, we found that the EHEC G4C is required for efficient colonization of the rabbit large intestine. Taken together, our results suggest that temporal expression of the capsule, which is coordinated with that of the TTSS, is required for optimal EHEC colonization of the host intestine.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Cápsulas Bacterianas/metabolismo , Escherichia coli Enteropatógena/patogenicidad , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Factores de Virulencia/metabolismo , Animales , Adhesión Bacteriana , Cápsulas Bacterianas/ultraestructura , Línea Celular , Escherichia coli Enteropatógena/metabolismo , Escherichia coli Enteropatógena/ultraestructura , Células Epiteliales/microbiología , Eritrocitos/microbiología , Infecciones por Escherichia coli , Escherichia coli O157/metabolismo , Escherichia coli O157/ultraestructura , Proteínas de Escherichia coli/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Humanos , Intestino Grueso/microbiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Mutagénesis Insercional , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Conejos , Transactivadores/metabolismo
14.
Cell Microbiol ; 10(2): 415-25, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17910741

RESUMEN

Enteropathogenic Escherichia coli (EPEC) forms attaching and effacing lesions in the intestinal mucosa characterized by intimate attachment to the epithelium by means of intimin (an outer membrane adhesin encoded by eae). EPEC is subgrouped into typical (tEPEC) and atypical (aEPEC); only tEPEC carries the EAF (EPEC adherence factor) plasmid that encodes the bundle-forming pilus (BFP). Characteristically, after 3 h of incubation, tEPEC produces localized adherence (LA) (with compact microcolonies) in HeLa/HEp-2 cells by means of BFP, whereas most aEPEC form looser microcolonies. We have previously identified nine aEPEC strains displaying LA in extended (6 h) assays (LA6). In this study, we analysed the kinetics of LA6 pattern development and the role of intimin in the process. Transmission electron microscopy and confocal laser microscopy showed that the invasive process of strain 1551-2 displays a LA phenotype. An eae-defective mutant of strain 1551-2 prevented the invasion although preserving intense diffused adherence. Sequencing of eae revealed that strain 1551-2 expresses the omicron subtype of intimin. We propose that the LA phenotype of aEPEC strain 1551-2 is mediated by intimin omicron and hypothesize that this strain expresses an additional novel adhesive structure. The present study is the first to report the association of compact microcolony formation and an intense invasive ability in aEPEC.


Asunto(s)
Adhesinas Bacterianas/fisiología , Adhesión Bacteriana/fisiología , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/fisiología , Actinas/metabolismo , Adhesinas Bacterianas/química , Secuencia de Aminoácidos , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Células HeLa , Humanos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Fenotipo , Fosforilación , Receptores de Superficie Celular/metabolismo , Alineación de Secuencia
15.
S Afr Med J ; 97(11 Pt 3): 1182-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18250933

RESUMEN

First described in 1885, Escherichia coli gradually achieved recognition as a cause of diarrhoea. Strains of E. coli which belonged to a limited number of O-serogroups and had been associated with outbreaks of diarrhoea in hospitalised children were designated 'enteropathogenic' E. coli (EPEC) to distinguish them from E. coli strains that cause other types of infection. The discovery that some strains of E. coli can produce toxins or invade epithelial cells in a similar fashion to established pathogens, such as Vibrio cholerae and Shigella species, shed new light on E. coli virulence. As EPEC do not exhibit either of these properties, however, their pathogenicity was brought into question. Further studies revealed that EPEC constitute a distinctive group of pathogenic bacteria that display characteristic adherence to cultured epithelial cells and produce distinctive histopathological changes, termed 'attaching-effacing lesions', in the intestinal epithelium. The ability to evoke these lesions, which are essential for virulence, is associated with a series of linked genes, known as the locus for enterocyte effacement, on the bacterial chromosome. In addition, the pathogenicity of some EPEC strains is associated with the presence of a plasmid-encoded, bundle-forming pilus. Naturally occurring strains of EPEC which lack this pilus are known as atypical EPEC and are an emerging cause of diarrhoea throughout the world.


Asunto(s)
Escherichia coli Enteropatógena/patogenicidad , Escherichia coli Enteropatógena/ultraestructura , Infecciones por Escherichia coli/microbiología , Humanos , Enfermedades Intestinales/economía , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...