Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Infect Immun ; 70(5): p.2681-9, 2002.
Artículo en Inglés | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib9595
2.
Cell Microbiol ; 3(12): 865-71, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11736997

RESUMEN

Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system (TTSS) to inject effector proteins into the plasma membrane and cytosol of infected cells. To translocate proteins, EPEC, like Salmonella and Shigella, is believed to assemble a macromolecular complex (type III secreton) that spans both bacterial membranes and has a short needle-like projection. However, there is a special interest in studying the EPEC TTSS owing to the fact that one of the secreted proteins, EspA, is assembled into a unique filamentous structure also required for protein translocation. In this report we present electron micrographs of EspA filaments which reveal a regular segmented substructure. Recently we have shown that deletion of the putative structural needle protein, EscF, abolished protein secretion and formation of EspA filaments. Moreover, we demonstrated that EspA can bind directly to EscF, suggesting that EspA filaments are physically linked to the EPEC needle complex. In this paper we provide direct evidence for the association between an EPEC bacterial membrane needle complex and EspA filaments, defining a new class of filamentous TTSS.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Proteínas Portadoras/ultraestructura , Proteínas de Escherichia coli , Escherichia coli/ultraestructura , Membrana Celular/ultraestructura , Escherichia coli/metabolismo
3.
Cell Microbiol ; 3(11): 753-62, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11696035

RESUMEN

Type III secretion systems, designed to deliver effector proteins across the bacterial cell envelope and the plasma membrane of the target eukaryotic cell, are involved in subversion of eukaryotic cell functions in a variety of human, animal and plant pathogens. In enteropathogenic Escherichia coli (EPEC), several protein substrates for the secretion apparatus were identified, including EspA, EspB and EspD. EspA is a structural protein and the major component of a large transiently expressed filamentous surface organelle that forms a direct link between the bacterium and the host cell, whereas EspD and EspB seem to form the mature translocation pore. Recent studies of the type III secretion systems of Shigella and Salmonella pathogenicity island (SPI)-1 revealed the existence of a macromolecular complex that spans both bacterial membranes and consists of a basal structure with two upper and two lower rings and a needle-like projection that extends outwards from the bacterial surface. MxiH (Shigella) and PrgI (Salmonella) are the main components of the needle of the type III secretion complex. A needle-like complex has not yet been reported in EPEC. In this study, we investigated EscF, a protein sharing sequence similarity with MxiH and PrgI. We report that EscF is required for type III protein secretion and EspA filament assembly. Moreover, we show that EscF binds EspA, suggesting that EspA filaments are an extension of the type III secretion needle complexes in EPEC.


Asunto(s)
Proteínas del Citoesqueleto , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Transporte de Proteínas/fisiología , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Línea Celular , Clonación Molecular , Eritrocitos/microbiología , Escherichia coli/fisiología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Prueba de Complementación Genética , Humanos , Ratones , Microscopía Electrónica , Mutación , Conejos , Análisis de Secuencia de ADN
4.
FEMS Microbiol Lett ; 203(2): 199-205, 2001 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-11583848

RESUMEN

The pathogenic mechanisms of enteroaggregative Escherichia coli (EAEC) are not well defined. We investigated the interaction of EAEC strain 236 (serotype O111:H12) with polarised Caco-2 and T84 human intestinal epithelial cells lines, and with human jejunal and colonic mucosa. Strain 236 adhered to both polarised cell lines and to both intestinal tissue types, but caused severe damage and was invasive only in T84 cells and colonic mucosa. In contrast, prototype EAEC strain 042, which also adhered to the cultured intestinal cell lines, did not adhere to or invade jejunal or colonic tissue. These observations suggest a heterogeneity of virulence properties within the EAEC category of diarrhoea-causing E. coli.


Asunto(s)
Colon/microbiología , Escherichia coli/patogenicidad , Mucosa Intestinal/microbiología , Adhesión Bacteriana , Células CACO-2 , Línea Celular , Niño , Escherichia coli/clasificación , Humanos , Yeyuno/microbiología , Microscopía Electrónica , Serotipificación , Virulencia
5.
J Pediatr Gastroenterol Nutr ; 33(1): 32-40, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11479405

RESUMEN

BACKGROUND: A hospital-based study was performed to (1) compare phenotypic and genotypic diagnostic tests for enteropathogenic Escherichia coli, enteroaggregative E. coli, and diffuse-adhering E. coli (collectively termed adherent E. coli) and (2) to assess the importance of these different classes of adherent E. coli as causes of infant diarrhea in the United Kingdom in comparison with other enteropathogens. METHODS: E. coli isolated from 1,496 infants with diarrheal disease and from 546 age-related controls were screened for enteropathogenic E. coli, enteroaggregative E. coli, and diffuse-adhering E. coli using HEp-2 cell adherence assays and DNA probes. RESULTS: Marked discrepancies between the phenotype and genotype of isolates indicate significant heterogeneity among enteroaggregative E. coli and diffuse-adhering E. coli strains. Depending on the assay used, adherent E. coli were isolated as the only putative pathogen in 23% to 27% of diarrhea cases, a significantly higher incidence than in the control group. Individually, enteroaggregative E. coli (8.5-8.6% of cases) and diffuse-adhering E. coli (10.4-11.3% of cases), but not enteropathogenic E. coli (4.5-7.5% of cases), were significantly associated with diarrhea. CONCLUSIONS: These studies indicate that adherent E. coli may be an important cause of diarrhea in infants in the United Kingdom; they also emphasize the need for more specific virulence-based tests for these putative classes of "diarrheagenic" (diarrhea causing) E. coli.


Asunto(s)
Adhesión Bacteriana/fisiología , Diarrea/microbiología , Infecciones por Escherichia coli/diagnóstico , Escherichia coli/fisiología , Escherichia coli/patogenicidad , Adhesión Bacteriana/genética , Estudios de Casos y Controles , Línea Celular , Sondas de ADN , Diarrea/diagnóstico , Diarrea/genética , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/genética , Femenino , Genotipo , Humanos , Lactante , Masculino , Microscopía Electrónica de Rastreo , Microscopía de Contraste de Fase , Fenotipo , Estudios Prospectivos , Reino Unido
6.
Infect Immun ; 69(6): 4055-64, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11349076

RESUMEN

Many animal and plant pathogens use type III secretion systems to secrete key virulence factors, some directly into the host cell cytosol. However, the basis for such protein translocation has yet to be fully elucidated for any type III secretion system. We have previously shown that in enteropathogenic and enterohemorrhagic Escherichia coli the type III secreted protein EspA is assembled into a filamentous organelle that attaches the bacterium to the plasma membrane of the host cell. Formation of EspA filaments is dependent on expression of another type III secreted protein, EspD. The carboxy terminus of EspD, a protein involved in formation of the translocation pore in the host cell membrane, is predicted to adopt a coiled-coil conformation with 99% probability. Here, we demonstrate EspD-EspD protein interaction using the yeast two-hybrid system and column overlays. Nonconservative triple amino acid substitutions of specific EspD carboxy-terminal residues generated an enteropathogenic E. coli mutant that was attenuated in its ability to induce attaching and effacing lesions on HEp-2 cells. Although the mutation had no effect on EspA filament biosynthesis, it also resulted in reduced binding to and reduced hemolysis of red blood cells. These results segregate, for the first time, functional domains of EspD that control EspA filament length from EspD-mediated cell attachment and pore formation.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli , Escherichia coli/patogenicidad , Hemólisis , Proteínas de la Membrana/química , Secuencia de Aminoácidos , Western Blotting , Línea Celular , Escherichia coli/fisiología , Infecciones por Escherichia coli/microbiología , Regulación Bacteriana de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Electrónica , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia de ADN , Técnicas del Sistema de Dos Híbridos , Virulencia
7.
Trends Microbiol ; 9(5): 214-8, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11336837

RESUMEN

Intimate bacterial adhesion to the intestinal epithelium is a pathogenic mechanism shared by several human and animal enteric pathogens, including enteropathogenic and enterohaemorrhagic Escherichia coli. Two bacterial protein partners involved in this intimate association have been identified, intimin and Tir. Some key remaining questions include whether intimin specifically interacts with one or more host-cell-encoded molecules and whether these contacts are a prerequisite for the subsequent intimate intimin-Tir association. Recent data support the hypothesis that the formation of a stable intimin-Tir relationship is the consequence of intimin protein interactions involving both host and bacterial components.


Asunto(s)
Adhesinas Bacterianas , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Portadoras , Proteínas de Escherichia coli , Escherichia coli/fisiología , Receptores de Superficie Celular/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/química , Células Cultivadas , Epitelio/metabolismo , Epitelio/microbiología , Humanos , Microscopía Inmunoelectrónica , Unión Proteica , Receptores de Superficie Celular/química
8.
Cell Microbiol ; 3(4): 213-22, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11298645

RESUMEN

Type III secretion allows bacteria to inject effector proteins into host cells. In enteropathogenic Escherichia coli (EPEC), three type III secreted proteins, EspA, EspB and EspD, have been shown to be required for translocation of the Tir effector protein into host cells. EspB and EspD have been proposed to form a pore in the host cell membrane, whereas EspA, which forms a large filamentous structure bridging bacterial and host cell surfaces, is thought to provide a conduit for translocation of effector proteins between pores in the bacterial and host cell membranes. Type III secretion has been correlated with an ability to cause contact-dependent haemolysis of red blood cells (RBCs) in vitro. As EspA filaments link bacteria and the host cell, we predicted that intimate bacteria-RBC contact would not be required for EPEC-induced haemolysis and, therefore, in this study we investigated the interaction of EPEC with monolayers of RBCs attached to polylysine-coated cell culture dishes. EPEC caused total RBC haemolysis in the absence of centrifugation and osmoprotection studies were consistent with the insertion of a hydrophilic pore into the RBC membrane. Cell attachment and haemolysis involved interaction between EspA filaments and the RBC membrane and was dependent upon a functional type III secretion system and on EspD, whereas EPEC lacking EspB still caused some haemolysis. Following haemolysis, only EspD was consistently detected in the RBC membrane. This study shows that intimate bacteria-RBC membrane contact is not a requirement for EPEC-induced haemolysis; it also provides further evidence that EspA filaments are a conduit for protein translocation and that EspD may be the major component of a translocation pore in the host cell membrane.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Adhesión Bacteriana/fisiología , Proteínas de la Membrana Bacteriana Externa , Proteínas Bacterianas/metabolismo , Transporte Biológico , Línea Celular , Membrana Eritrocítica/microbiología , Membrana Eritrocítica/ultraestructura , Eritrocitos/microbiología , Eritrocitos/ultraestructura , Escherichia coli/patogenicidad , Escherichia coli/ultraestructura , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/fisiología , Fimbrias Bacterianas/ultraestructura , Hemólisis , Humanos
9.
Curr Opin Infect Dis ; 14(5): 559-65, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11964876

RESUMEN

Unlike many gastrointestinal pathogens, enteropathogenic Escherichia coli orchestrates the modulation of host cellular and immune responses from the exterior of the infected cell, chiefly via the secreted and translocated components of a type III secretion system. Close inspection of these enteropathogenic Escherichia coli proteins and the interactions they mediate provides an increasingly coherent picture of the pathogenic mechanisms that enteropathogenic Escherichia coli uses to exploit its host.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli/patogenicidad , Intestinos/microbiología , Adhesinas Bacterianas/fisiología , Evolución Biológica , Proteínas Portadoras/fisiología , Mapeo Cromosómico , Enterocitos/microbiología , Escherichia coli/genética , Escherichia coli/inmunología , Operón , Receptores de Superficie Celular/fisiología , Virulencia
10.
Trends Microbiol ; 9(5): p.214-8, 2001.
Artículo en Inglés | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib11453
12.
Mol Microbiol ; 35(6): 1483-92, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10760148

RESUMEN

Enteropathogenic Escherichia coli (EPEC), like many bacterial pathogens, use a type III secretion system to deliver effector proteins across the bacterial cell wall. In EPEC, four proteins, EspA, EspB, EspD and Tir are known to be exported by a type III secretion system and to be essential for 'attaching and effacing' (A/E) lesion formation, the hallmark of EPEC pathogenicity. EspA was recently shown to be a structural protein and a major component of a large, transiently expressed, filamentous surface organelle which forms a direct link between the bacterium and the host cell. In contrast, EspB is translocated into the host cell where it is localized to both membrane and cytosolic cell fractions. EspA and EspB are required for translocation of Tir to the host cell membrane suggesting that they may both be components of the translocation apparatus. In this study, we show that EspB co-immunoprecipitates with the EspA filaments and that, during EPEC infection of HEp-2 cells, EspB localizes closely with EspA. Using a number of binding assays, we also show that EspB can bind and be copurified with EspA. Nevertheless, binding of EspA filaments to the host cell membranes occurred even in the absence of EspB. These results suggest that following initial attachment of the EspA filaments to the target cells, EspB is delivered into the host cell membrane and that the interaction between EspA and EspB may be important for protein translocation.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/aislamiento & purificación , Transporte Biológico , Línea Celular/microbiología , Células Epiteliales/microbiología , Glutatión Transferasa/genética , Glutatión Transferasa/aislamiento & purificación , Glutatión Transferasa/metabolismo , Humanos , Ratones , Mutación , Pruebas de Precipitina/métodos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
J Biol Chem ; 274(50): 35969-74, 1999 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-10585486

RESUMEN

Enteropathogenic E. coli (EPEC) utilize a type III secretion system to deliver virulence-associated effector proteins to the host cell. Four proteins, EspA, EspB, EspD, and Tir, which are integral to the formation of characteristic "attaching and effacing" (A/E) intestinal lesions, are known to be exported via the EPEC type III secretion system. Recent work demonstrated that EspA is a major component of a filamentous structure, elaborated on the surface of EPEC, which is required for translocation of EspB and Tir. The carboxyl terminus of EspA is predicted to comprise an alpha-helical region, which demonstrates heptad periodicity whereby positions a and d in the heptad repeat unit abcdefg are occupied by hydrophobic residues, indicating a propensity for coiled-coil interactions. Here we demonstrate multimeric EspA isoforms in EPEC culture supernatants and EspA:EspA interaction on solid phase. Non-conservative amino acid substitution of specific EspA heptad residues generated EPEC mutants defective in filament assembly but which retained the ability to induce A/E lesions; additional mutation totally abolished EspA filament assembly and A/E lesion formation. These results demonstrate a similarity to flagellar biosynthesis and indicate that the coiled-coil domain of EspA is required for assembly of the EspA filament-associated type III secretion translocon.


Asunto(s)
Proteínas Bacterianas/química , Membrana Celular/fisiología , Proteínas de Escherichia coli , Escherichia coli/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/ultraestructura , Clonación Molecular , Escherichia coli/patogenicidad , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Transducción de Señal , Programas Informáticos , Virulencia
14.
J Clin Microbiol ; 37(12): 3822-7, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10565891

RESUMEN

Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) constitute a significant risk to human health worldwide. A hallmark of both pathogens is their ability to produce characteristic attaching-and-effacing (A/E) lesions in intestinal epithelial cells. Genes encoding A/E lesion formation map to a chromosomal pathogenicity island termed the locus of enterocyte effacement (LEE). Intimin, an LEE-encoded bacterial adhesion molecule, mediates the intimate bacterium-host cell interaction characteristic of A/E lesions. On the basis of characterization of the C-terminal 280-amino-acid cell binding domain of intimin (Int280(661-939)), four distinct Int280 types (types alpha, beta, gamma, and delta) have been identified. Importantly, Int280alpha and Int280beta antisera specifically recognized their respective intimin types. Using a conserved region of the intimin molecule (Int(388-667)) and primers synthesized to generate the recombinant Int(388-667), we have now generated universal intimin antiserum and PCR primers that are reactive with the different intimin types expressed by both human and animal A/E lesion-forming strains. Use of immunogold electron microscopy to visualize intimin on the surfaces of EPEC and EHEC strains revealed, in general, a uniform distribution on the bacterial cell surface. However, a filamentous staining pattern was observed with a few strains expressing intimin gamma. Cloning of the intimin eae gene from one such strain (strain ICC57) into strain CVD206, an EPEC strain which harbors a null deletion in eae, produced a uniform intimin staining pattern indicating that, if the filamentous staining pattern defines a filamentous form of intimin gamma, it is dependent upon the genetic background of the strain and is not a feature of the intimin molecule.


Asunto(s)
Adhesinas Bacterianas , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras , Cartilla de ADN , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli , Escherichia coli/química , Escherichia coli/aislamiento & purificación , Animales , Anticuerpos Antibacterianos/biosíntesis , Proteínas de la Membrana Bacteriana Externa/análisis , Proteínas de la Membrana Bacteriana Externa/genética , Western Blotting , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/patogenicidad , Humanos , Sueros Inmunes/inmunología , Inmunohistoquímica , Reacción en Cadena de la Polimerasa
15.
Mol Microbiol ; 33(6): 1176-89, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10510232

RESUMEN

The locus of enterocyte effacement of enteropathogenic Escherichia coli encodes a type III secretion system, an outer membrane protein adhesin (intimin, the product of eae ) and Tir, a translocated protein that becomes a host cell receptor for intimin. Many type III secreted proteins require chaperones, which function to stabilize proteins, prevent inappropriate protein-protein interactions and aid in secretion. An open reading frame located between tir and eae, previously named orfU, was predicted to encode a protein with partial similarity to the Yersinia SycH chaperone. We examined the potential of the orfU gene product to serve as a chaperone for Tir. The orfU gene encoded a 15 kDa cytoplasmic protein that specifically interacted with Tir as demonstrated by the yeast two-hybrid assay, column binding and coimmunoprecipitation experiments. An orfU mutant was defective in attaching-effacing lesion formation and Tir secretion, but was unaffected in expression of other virulence factors. OrfU appeared to stabilize Tir levels in the cytoplasm, but was not absolutely necessary for secretion of Tir. Based upon the physical similarities, phenotypic characteristics and the demonstrated interaction with Tir, orfU is redesignated as cesT for the chaperone for E. coli secretion of T ir.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas de Escherichia coli , Escherichia coli/fisiología , Escherichia coli/patogenicidad , Chaperonas Moleculares/fisiología , Receptores de Superficie Celular/fisiología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Secuencia de Bases , Clonación Molecular , Secuencia Conservada , Cartilla de ADN/genética , Escherichia coli/genética , Genes Bacterianos , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Mutación , Fenotipo , Unión Proteica , Receptores de Superficie Celular/genética , Homología de Secuencia de Aminoácido , Shigella/genética , Shigella/fisiología , Transformación Genética , Virulencia/genética , Virulencia/fisiología
16.
Mol Microbiol ; 33(3): 499-509, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10417641

RESUMEN

BFP, a plasmid-encoded type IV bundle-forming pilus produced by enteropathogenic Escherichia coli (EPEC), has recently been shown to be associated with the aggregation of bacteria and dispersal of bacteria from bacterial microcolonies. In standard 3 h HEp-2 cell assays, EPEC adhere in localized microcolonies; after 6 h, bacterial microcolonies are no longer present, indicating that bacterial aggregation and dispersal occurs in vitro during EPEC adhesion to cultured epithelial cells. To examine the role of BFP in EPEC aggregation and dispersal, we examined HEp-2 cell adhesion of strain E2348/69 and defined E2348/69 mutants by immunofluorescence and immunoelectron microscopy. BFP was expressed initially as approximately 40 nm diameter pilus bundles that promoted bacteria-bacteria interaction and microcolony formation. BFP subsequently underwent a striking alteration in structural organization with the formation of much longer and thicker ( approximately 100 nm diameter) pilus bundles, which frequently aggregated laterally to form even thicker bundles often arranged in a loose three-dimensional network; EPEC dispersal from bacterial microcolonies was associated with this transformation of BFP from thin to thick bundles. Bacterial dispersal and transformation of BFP from thin to thick bundles did not occur with a bfpF mutant of strain E2348/69. It is concluded that BFP promotes both the formation and the dispersal of EPEC microcolonies, that the dispersal phase requires BfpF and that dispersal is associated with dramatic alterations in the structure of BFP bundles.


Asunto(s)
Adhesión Bacteriana , Escherichia coli/patogenicidad , Fimbrias Bacterianas/química , Proteínas de la Membrana/química , Escherichia coli/ultraestructura , Proteínas Fimbrias , Fimbrias Bacterianas/ultraestructura , Humanos , Inmunohistoquímica , Microscopía Electrónica de Rastreo , Microscopía de Contraste de Fase , Mutación , Plásmidos , Células Tumorales Cultivadas
17.
Mol Microbiol ; 32(1): 151-8, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10216868

RESUMEN

Enteropathogenic Escherichia coli (EPEC) induce characteristic attaching and effacing (A/E) lesions on epithelial cells. This event is mediated, in part, by binding of the bacterial outer membrane protein, intimin, to a second EPEC protein, Tir (translocated intimin receptor), which is exported by the bacteria and integrated into the host cell plasma membrane. In this study, we have localized the intimin-binding domain of Tir to a central 107-amino-acid region, designated Tir-M. We provide evidence that both the amino- and carboxy-termini of Tir are located within the host cell. In addition, using immunogold labelling electron microscopy, we have confirmed that intimin can bind independently to host cells even in the absence of Tir. This Tir-independent interaction and the ability of EPEC to induce A/E lesions requires an intact lectin-like module residing at the carboxy-terminus of the intimin polypeptide. Using the yeast two-hybrid system and gel overlays, we show that intimin can bind both Tir and Tir-M even when the lectin-like domain is disrupted. These data provide strong evidence that intimin interacts not only with Tir but also in a lectin-like manner with a host cell intimin receptor.


Asunto(s)
Adhesinas Bacterianas , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Receptores de Superficie Celular/metabolismo , Línea Celular , Clonación Molecular , Escherichia coli/patogenicidad , Células HeLa , Humanos , Inmunohistoquímica , Microscopía Electrónica , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Modelos Genéticos , Modelos Moleculares , Plásmidos , Unión Proteica , Levaduras/genética , beta-Galactosidasa/metabolismo
18.
FEMS Microbiol Lett ; 169(1): 73-80, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9851036

RESUMEN

Enteropathogenic Escherichia coli (EPEC) encode a type III secretion system located on a pathogenicity island known as the locus for enterocyte effacement. Four proteins are known to be exported by this type III secretion system--EspA, EspB and EspD required for subversion of host cell signal transduction pathways and a translocated intimin receptor protein (Tir) required for intimin-mediated intimate attachment and attaching and effacing lesion formation. The espA gene is located within the locus for enterocyte effacement and the EspA polypeptide from the prototype EPEC strain E2348/69 (O127:H6) has recently been shown to be a component of a filamentous structure involved in bacteria-host cell interaction and locus for enterocyte effacement-encoded protein translocation involved in attaching and effacing lesion formation. In this study we have extended our investigation of EspA to strains belonging to other classical EPEC serotypes. DNA sequencing demonstrated that the espA gene from the different EPEC strains share at least 65% DNA identity. In addition, we detected morphologically and antigenically similar EspA filaments in all but one of the bacterial strains examined including recombinant, non-pathogenic E. coli expressing espA from a cloned locus for enterocyte effacement region (HB101(pCVD462)).


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Escherichia coli , Escherichia coli/genética , Escherichia coli/patogenicidad , Secuencia de Aminoácidos , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Células Cultivadas , ADN Bacteriano/genética , Escherichia coli/clasificación , Escherichia coli/ultraestructura , Expresión Génica , Mucosa Intestinal/microbiología , Datos de Secuencia Molecular , Plásmidos , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
19.
Infect Immun ; 66(12): 6049-53, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9826396

RESUMEN

The pathophysiology of enteropathogenic Escherichia coli (EPEC) diarrhea remains uncertain. In vitro, EPEC stimulates a rapid increase in short-circuit current (Isc) across Caco-2 cell monolayers coincident with intimate attaching and effacing (A/E) bacterial adhesion. This study has examined the roles of specific EPEC virulence proteins in this Isc response. EPEC genes encoding EspA, EspB, and EspD, essential for signal transduction in host cells and A/E activity, were also required for modulation of Caco-2 electrolyte transport.


Asunto(s)
Adhesinas Bacterianas , Proteínas Bacterianas/metabolismo , Proteínas Portadoras , Colon/fisiología , Proteínas de Escherichia coli , Escherichia coli/patogenicidad , Genes Bacterianos , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/biosíntesis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Transporte Biológico , Células CACO-2 , Colon/citología , Colon/microbiología , Conductividad Eléctrica , Impedancia Eléctrica , Electrólitos/metabolismo , Escherichia coli/genética , Humanos , Transducción de Señal
20.
Infect Immun ; 66(8): 3900-8, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9673278

RESUMEN

Elevated concentrations of intracellular calcium ([Ca]i) have been implicated as an important signalling event during attaching and effacing (A/E) lesion formation by enteropathogenic Escherichia coli (EPEC). The highly localized nature of the cytoskeletal and cell surface alterations occurring during A/E lesion formation suggests that there should be equally localized EPEC-induced signalling events. To analyze further the calcium responses to infection of HEp-2 cells by EPEC, we employed calcium-imaging fluorescence microscopy, which allows both temporal and spatial measurements of [Ca]i in live cells. Using this imaging technique, not only were we unable to detect any significant elevation in [Ca]i at sites of A/E EPEC adhesion, but, with several different classical EPEC and enterohemorrhagic E. coli (EHEC) strains and three different infection procedures, each of which resulted in extensive A/E bacterial adhesion, we were unable to detect any significant alterations in [Ca]i in infected cells compared to uninfected cells. In addition, chelation of intracellular free calcium with bis-(aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid (BAPTA) did not, as previously reported, prevent A/E lesion formation. We conclude that increased [Ca]i are not required for A/E lesion formation by EPEC and EHEC.


Asunto(s)
Calcio/metabolismo , Escherichia coli/fisiología , Humanos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...