Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-24025841

RESUMEN

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Tracto Gastrointestinal/microbiología , Transducción de Señal , Factores de Virulencia/metabolismo , Animales , Caspasa 8/metabolismo , Muerte Celular , Citrobacter rodentium/patogenicidad , Citrobacter rodentium/fisiología , Escherichia coli Enteropatógena/patogenicidad , Activación Enzimática , Infecciones por Escherichia coli/patología , Proteína Ligando Fas/antagonistas & inhibidores , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/química , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , N-Acetilglucosaminiltransferasas/metabolismo , Estructura Terciaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/química , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Receptor fas/deficiencia , Receptor fas/metabolismo
2.
Hum Mol Genet ; 23(15): 4064-76, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24643277

RESUMEN

iRHOM2 is a highly conserved, catalytically inactive member of the Rhomboid family, which has recently been shown to regulate the maturation of the multi-substrate ectodomain sheddase enzyme ADAM17 (TACE) in macrophages. Dominant iRHOM2 mutations are the cause of the inherited cutaneous and oesophageal cancer-susceptibility syndrome tylosis with oesophageal cancer (TOC), suggesting a role for this protein in epithelial cells. Here, using tissues derived from TOC patients, we demonstrate that TOC-associated mutations in iRHOM2 cause an increase in the maturation and activity of ADAM17 in epidermal keratinocytes, resulting in significantly upregulated shedding of ADAM17 substrates, including EGF-family growth factors and pro-inflammatory cytokines. This activity is accompanied by increased EGFR activity, increased desmosome processing and the presence of immature epidermal desmosomes, upregulated epidermal transglutaminase activity and heightened resistance to Staphylococcal infection in TOC keratinocytes. Many of these features are consistent with the presence of a constitutive wound-healing-like phenotype in TOC epidermis, which may shed light on a novel pathway in skin repair, regeneration and inflammation.


Asunto(s)
Proteínas ADAM/genética , Proteínas Portadoras/genética , Epidermis/metabolismo , Neoplasias Esofágicas/genética , Queratinocitos/metabolismo , Queratodermia Palmoplantar/genética , Infecciones Cutáneas Estafilocócicas/genética , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/metabolismo , Proteína ADAM17 , Proteínas Portadoras/metabolismo , Citocinas/biosíntesis , Desmosomas/metabolismo , Desmosomas/patología , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Epidermis/microbiología , Epidermis/patología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/microbiología , Neoplasias Esofágicas/patología , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/microbiología , Queratinocitos/patología , Queratodermia Palmoplantar/metabolismo , Queratodermia Palmoplantar/microbiología , Queratodermia Palmoplantar/patología , Masculino , Mutación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Infecciones Cutáneas Estafilocócicas/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus/fisiología , Transglutaminasas/genética , Transglutaminasas/metabolismo
3.
Vet Res ; 45: 81, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25175996

RESUMEN

Salmonella Gallinarum and Salmonella Enteritidis are genetically closely related however associated with different pathologies. Several studies have suggested that S. Gallinarum is less invasive in vitro than S. Enteritidis. In this study we confirm that the S. Gallinarum strains tested were much less invasive than the S. Enteritidis strains tested in cells of avian or human origin. In addition, the S. Gallinarum T3SS-1-dependent ability to invade host cells was delayed by two to three hours compared to S. Enteritidis, indicating that T3SS-1-dependent entry is less efficient in S. Gallinarum than S. Enteritidis. This was neither due to a decreased transcription of T3SS-1 related genes when bacteria come into contact with cells, as transcription of hilA, invF and sipA was similar to that observed for S. Enteritidis, nor to a lack of functionality of the S. Gallinarum T3SS-1 apparatus as this apparatus was able to secrete and translocate effector proteins into host cells. In contrast, genome comparison of four S. Gallinarum and two S. Enteritidis strains revealed that all S. Gallinarum genomes displayed the same point mutations in each of the main T3SS-1 effector genes sipA, sopE, sopE2, sopD and sopA.


Asunto(s)
Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/microbiología , Infecciones por Salmonella/microbiología , Salmonella enterica/fisiología , Salmonella enterica/patogenicidad , Salmonella enteritidis/fisiología , Salmonella enteritidis/patogenicidad , Animales , Adhesión Bacteriana , Línea Celular , Línea Celular Tumoral , Pollos , Humanos , Salmonella enterica/genética , Salmonella enteritidis/genética
4.
Cell Tissue Res ; 351(1): 107-16, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23138568

RESUMEN

Escherichia coli, including enteropathogenic E. coli (EPEC), represents the most common cause of diarrhoea worldwide and is therefore a serious public health burden. Treatment for gastrointestinal pathogens is hindered by the emergence of multiple antibiotic resistance, leading to the requirement for the development of new therapies. A variety of mechanisms act in combination to mediate gastrointestinal-bacterial-associated diarrhoea development. For example, EPEC infection of enterocytes induces attaching and effacing lesion formation and the disruption of tight junctions. An alternative enteric pathogen, Shigella flexneri, manipulates the expression of Connexin 26 (Cx26), a gap junction protein. S. flexneri can open Cx26 hemichannels allowing the release of ATP, whereas HeLa cells expressing mutant gap-junction-associated Cx26 are less susceptible to cellular invasion by S. flexneri than cells expressing wild-type (WT) Cx26. We have investigated further the link between Cx26 expression and gastrointestinal infection by using EPEC and S. flexneri as in vitro models of infection. In this study, a significant reduction in EPEC adherence was observed in cells expressing mutant Cx26 compared with WT Cx26. Furthermore, a significant reduction in both cellular invasion by S. flexneri and adherence by EPEC was demonstrated in human intestinal cell lines following treatment with Cx26 short interfering RNA. These in vitro results suggest that the loss of functional Cx26 expression provides improved protection against gastrointestinal bacterial pathogens. Thus, Cx26 represents a potential therapeutic target for gastrointestinal bacterial infection.


Asunto(s)
Conexinas/metabolismo , Disentería Bacilar/metabolismo , Escherichia coli Enteropatógena/fisiología , Infecciones por Escherichia coli/metabolismo , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/patología , Shigella flexneri/fisiología , Adhesión Bacteriana , Línea Celular Tumoral , Conexina 26 , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , ARN Interferente Pequeño/metabolismo , Transfección
5.
Proc Natl Acad Sci U S A ; 107(7): 3129-34, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133763

RESUMEN

The human pathogens enteropathogenic (EPEC) and enterohemorrhagic Escherichia coli and the related mouse pathogen Citrobacter rodentium subvert a variety of host cell signaling pathways via their plethora of type III secreted effectors, including triggering of an early apoptotic response. EPEC-infected cells do not develop late apoptotic symptoms, however. In this study we demonstrate that the NleH family effectors, homologs of the Shigella effector kinase OspG, blocks apoptosis. During EPEC infection, NleH effectors inhibit elevation of cytosolic Ca(2+) concentrations, nuclear condensation, caspase-3 activation, and membrane blebbing and promote cell survival. NleH1 alone is sufficient to prevent procaspase-3 cleavage induced by the proapoptotic compounds staurosporine, brefeldin A, and tunicamycin. Using C. rodentium, we found that NleH inhibits procaspase-3 cleavage at the bacterial attachment sites in vivo. A yeast two-hybrid screen identified the endoplasmic reticulum six-transmembrane protein Bax inhibitor-1 (BI-1) as an NleH-interacting partner. We mapped the NleH-binding site to the N-terminal 40 amino acids of BI-1. Knockdown of BI-1 resulted in the loss of NleH's antiapoptotic activity. These results indicate that NleH effectors are inhibitors of apoptosis that may act through BI-1 to carry out their cytoprotective function.


Asunto(s)
Apoptosis/efectos de los fármacos , Infecciones por Enterobacteriaceae/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Virulencia/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Citrobacter rodentium , Clonación Molecular , Citosol/metabolismo , Escherichia coli , Proteínas de Escherichia coli/farmacología , Femenino , Proteínas de la Membrana/genética , Ratones , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Organismos Libres de Patógenos Específicos , Técnicas del Sistema de Dos Híbridos , Factores de Virulencia/farmacología
6.
Mol Microbiol ; 80(1): 219-30, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21306441

RESUMEN

Many bacterial pathogens utilize a type III secretion system (T3SS) to inject virulence effector proteins into host cells during infection. Previously, we found that enteropathogenic Escherichia coli (EPEC) uses the type III effector, NleE, to block the inflammatory response by inhibiting IκB degradation and nuclear translocation of the p65 subunit of NF-κB. Here we screened further effectors with unknown function for their capacity to prevent p65 nuclear translocation. We observed that ectopic expression of GFP-NleC in HeLa cells led to the degradation of p65. Delivery of NleC by the T3SS of EPEC also induced degradation of p65 in infected cells as well as other NF-κB components, c-Rel and p50. Recombinant His(6) -NleC induced p65 and p50 cleavage in HeLa cell lysates and mutation of a consensus zinc metalloprotease motif, HEIIH, abrogated NleC proteolytic activity. NleC inhibited IL-8 production during prolonged EPEC infection of HeLa cells in a protease activity-dependent manner. A double nleE/nleC mutant was further impaired for its ability to inhibit IL-8 secretion than either a single nleE or a single nleC mutant. We conclude that NleC is a type III effector protease that degrades NF-κB thereby contributing the arsenal of bacterial effectors that inhibit innate immune activation.


Asunto(s)
Escherichia coli Enteropatógena/enzimología , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteínas de Escherichia coli/genética , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Immunoblotting , Interleucina-8/metabolismo , Microscopía Confocal , Factor de Transcripción ReIA/genética
7.
J Immunol ; 185(7): 4118-27, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20833837

RESUMEN

Intestinal dendritic cells (DCs) send processes between epithelial cells into the gut lumen to sample pathogens. Noninvasive enteropathogenic Escherichia coli (EPEC) colonize the gut using a type three secretion system (T3SS) to inject effector proteins into epithelial cells. We hypothesized that EPEC might also inject proteins into DC processes to dampen immune recognition. Using a T3SS-linked fluorescence resonance energy transfer-based system we show that EPEC injects effectors into in vitro grown human myeloid DCs. Injected cells emit a blue signal due to cleavage of the green fluorescence resonance energy transfer-based substrate CCF2/AM by ß-lactamase. When cultured with a mutant EPEC unable to translocate effector proteins, myeloid DCs show rapid activation of NF-κB, secrete large amounts of proinflammatory cytokines and increase expression of CD80, CD83, and CD86, whereas wild-type EPEC barely elicits cytokine production and shuts off nuclear translocation of NF-κB p65. By deleting effector protein genes, we identified NleE as being critical for this effect. Expression of NleE in HeLa cells completely prevented nuclear p65 accumulation in response to IL1-ß, and luciferase production in an NF-κB reporter cell line. DCs cocultured with wild-type EPEC or NleE-complemented strains were less potent at inducing MLR. EPEC was also able to inject effectors into DCs sending processes through model gut epithelium in a transwell system and into Peyer's patch myeloid DCs. Thus, EPEC translocate effectors into human DCs to dampen the inflammatory response elicited by its own pathogen-associated molecular patterns.


Asunto(s)
Células Dendríticas/microbiología , Escherichia coli Enteropatógena/patogenicidad , Infecciones por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/inmunología , Factores de Virulencia/metabolismo , Western Blotting , Separación Celular , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Escherichia coli Enteropatógena/inmunología , Escherichia coli Enteropatógena/metabolismo , Ensayo de Inmunoadsorción Enzimática , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Prueba de Cultivo Mixto de Linfocitos , Microscopía Confocal , FN-kappa B/inmunología , Factores de Virulencia/inmunología
8.
J Bacteriol ; 192(13): 3534-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20400543

RESUMEN

We report that the N terminus of the type III secretion system translocator proteins EspB, EspD, and EspA mediate protein secretion and translocation from wild-type enteropathogenic Escherichia coli and hypersecretion from sepL and sepD mutants. EspA containing the translocation signal of Map and Tir containing the secretion signal of EspA are biologically active.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
9.
Cell Rep ; 27(4): 1008-1017.e6, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018119

RESUMEN

Microbial infections can stimulate the assembly of inflammasomes, which activate caspase-1. The gastrointestinal pathogen enteropathogenic Escherichia coli (EPEC) causes localized actin polymerization in host cells. Actin polymerization requires the binding of the bacterial adhesin intimin to Tir, which is delivered to host cells via a type 3 secretion system (T3SS). We show that EPEC induces T3SS-dependent rapid non-canonical NLRP3 inflammasome activation in human macrophages. Notably, caspase-4 activation by EPEC triggers pyroptosis and cytokine processing through the NLRP3-caspase-1 inflammasome. Mechanistically, caspase-4 activation requires the detection of LPS and EPEC-induced actin polymerization, either via Tir tyrosine phosphorylation and the phosphotyrosine-binding adaptor NCK or Tir and the NCK-mimicking effector TccP. An engineered E. coli K12 could reconstitute Tir-intimin signaling, which is necessary and sufficient for inflammasome activation, ruling out the involvement of other virulence factors. Our studies reveal a crosstalk between caspase-4 and caspase-1 that is cooperatively stimulated by LPS and effector-driven actin polymerization.


Asunto(s)
Caspasas Iniciadoras/fisiología , Escherichia coli Enteropatógena/patogenicidad , Macrófagos/microbiología , Actinas/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasa 1/fisiología , Caspasas Iniciadoras/genética , Caspasas Iniciadoras/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inflamasomas/fisiología , Modelos Biológicos , Polimerizacion
10.
Infect Immun ; 76(1): 361-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17984209

RESUMEN

Typical enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) employ either Nck, TccP/TccP2, or Nck and TccP/TccP2 pathways to activate the neuronal Wiskott-Aldrich syndrome protein (N-WASP) and to trigger actin polymerization in cultured cells. This phenotype is used as a marker for the pathogenic potential of EPEC and EHEC strains. In this paper we report that EPEC O125:H6, which represents a large category of strains, lacks the ability to utilize either Nck or TccP/TccP2 and hence triggers actin polymerization in vitro only inefficiently. However, we show that infection of human intestinal biopsies with EPEC O125:H6 results in formation of typical attaching and effacing lesions. Expression of TccP in EPEC O125:H6, which harbors an EHEC O157-like Tir, resulted in efficient actin polymerization in vitro and enhanced colonization of human intestinal in vitro organ cultures with detectable N-WASP and electron-dense material at the site of bacterial adhesion. These results show the existence of a natural category of EPEC that colonizes the gut mucosa using Nck- and TccP-independent mechanisms. Importantly, the results highlight yet again the fact that conclusions made on the basis of in vitro cell culture models cannot be extrapolated wholesale to infection of mucosal surfaces and that the ability to induce actin polymerization on cultured cells should not be used as a definitive marker for EPEC and EHEC virulence.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Intestinos/patología , Proteínas Oncogénicas/metabolismo , Actinas/metabolismo , Adhesión Bacteriana , Biopsia , Proteínas de Escherichia coli/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Intestinos/microbiología , Datos de Secuencia Molecular , Receptores de Superficie Celular/genética
11.
Infect Immun ; 76(11): 4804-13, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18725419

RESUMEN

The human pathogen enterohemorrhagic Escherichia coli (EHEC) O157:H7 colonizes human and animal gut via formation of attaching and effacing lesions. EHEC strains use a type III secretion system to translocate a battery of effector proteins into the mammalian host cell, which subvert diverse signal transduction pathways implicated in actin dynamics, phagocytosis, and innate immunity. The genomes of sequenced EHEC O157:H7 strains contain two copies of the effector protein gene nleH, which share 49% sequence similarity with the gene for the Shigella effector OspG, recently implicated in inhibition of migration of the transcriptional regulator NF-kappaB to the nucleus. In this study we investigated the role of NleH during EHEC O157:H7 infection of calves and lambs. We found that while EHEC DeltanleH colonized the bovine gut more efficiently than the wild-type strain, in lambs the wild-type strain exhibited a competitive advantage over the mutant during mixed infection. Using the mouse pathogen Citrobacter rodentium, which shares many virulence factors with EHEC O157:H7, including NleH, we observed that the wild-type strain exhibited a competitive advantage over the mutant during mixed infection. We found no measurable differences in T-cell infiltration or hyperplasia in colons of mice inoculated with the wild-type or the nleH mutant strain. Using NF-kappaB reporter mice carrying a transgene containing a luciferase reporter driven by three NF-kappaB response elements, we found that NleH causes an increase in NF-kappaB activity in the colonic mucosa. Consistent with this, we found that the nleH mutant triggered a significantly lower tumor necrosis factor alpha response than the wild-type strain.


Asunto(s)
Citrobacter rodentium/patogenicidad , Escherichia coli O157/patogenicidad , Mucosa Intestinal/microbiología , Factores de Virulencia/metabolismo , Animales , Bovinos , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/patología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Técnica del Anticuerpo Fluorescente Indirecta , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Factores de Virulencia/genética
12.
FEMS Microbiol Lett ; 263(1): 32-40, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16958848

RESUMEN

Enterohaemorrhagic Escherichia coli (EHEC) causes bloody diarrhoea in humans and deploys a type III secretion system (T3SS) encoded by the locus of enterocyte effacement to elicit the formation of attaching and effacing (AE) lesions on intestinal epithelia. Here, we report the identification of a new secreted substrate of this system, z1829, which is encoded by cryptic prophage CP-933N. Elevated secretion of a beta-lactamase-z1829 fusion protein was detected upon mutation of sepD in EHEC O157:H7 and the fusion protein was translocated into infected epithelial cells in a T3SS-dependent manner; accordingly, we named the protein EspK. In common with the related Salmonella enterica type III secreted effector GogB, we observed that EspK localized to the cytoplasm when transiently expressed in COS-7 cells using EspK-specific antiserum. Inactivation of espK did not impair adherence or actin nucleation during infection of HeLa cells but affected persistence of EHEC O157:H7 in the intestines of orally inoculated calves. Inactivation of an orthologue of espK in the murine AE pathogen Citrobacter rodentium did not impair intestinal colonization in mice.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Factores de Virulencia , Animales , Células COS , Bovinos , Chlorocebus aethiops , Citrobacter rodentium/química , Citrobacter rodentium/genética , Citoplasma/química , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli O157/química , Escherichia coli O157/genética , Proteínas de Escherichia coli/aislamiento & purificación , Islas Genómicas , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Profagos/genética , Transfección
13.
Microbes Infect ; 5(10): 857-67, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12919854

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/genética , Escherichia coli/inmunología , Adhesinas de Escherichia coli/inmunología , Administración Oral , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Adhesión Bacteriana/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Peso Corporal , Diarrea/inmunología , Diarrea/microbiología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Eliminación de Gen , Células HeLa , Humanos , Intestinos/microbiología , Mutagénesis Insercional , Conejos , Receptores de Superficie Celular/genética , Virulencia/genética
14.
J Inflamm (Lond) ; 8: 11, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21539730

RESUMEN

The intestinal immune system and the epithelium are the first line of defense in the gut. Constantly exposed to microorganisms from the environment, the gut has complex defense mechanisms to prevent infections, as well as regulatory pathways to tolerate commensal bacteria and food antigens. Intestinal pathogens have developed strategies to regulate intestinal immunity and inflammation in order to establish or prolong infection. The organisms that employ a type III secretion system use a molecular syringe to deliver effector proteins into the cytoplasm of host cells. These effectors target the host cell cytoskeleton, cell organelles and signaling pathways. This review addresses the multiple mechanisms by which the type III secretion system targets the intestinal immune response, with a special focus on pathogenic E. coli.

15.
Proc Natl Acad Sci U S A ; 103(40): 14941-6, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-16990433

RESUMEN

Several pathogenic strains of Escherichia coli exploit type III secretion to inject "effector proteins" into human cells, which then subvert eukaryotic cell biology to the bacterium's advantage. We have exploited bioinformatics and experimental approaches to establish that the effector repertoire in the Sakai strain of enterohemorrhagic E. coli (EHEC) O157:H7 is much larger than previously thought. Homology searches led to the identification of >60 putative effector genes. Thirteen of these were judged to be likely pseudogenes, whereas 49 were judged to be potentially functional. In total, 39 proteins were confirmed experimentally as effectors: 31 through proteomics and 28 through translocation assays. At the protein level, the EHEC effector sequences fall into >20 families. The largest family, the NleG family, contains 14 members in the Sakai strain alone. EHEC also harbors functional homologs of effectors from plant pathogens (HopPtoH, HopW, AvrA) and from Shigella (OspD, OspE, OspG), and two additional members of the Map/IpgB family. Genes encoding proven or predicted effectors occur in >20 exchangeable effector loci scattered throughout the chromosome. Crucially, the majority of functional effector genes are encoded by nine exchangeable effector loci that lie within lambdoid prophages. Thus, type III secretion in E. coli is linked to a vast phage "metagenome," acting as a crucible for the evolution of pathogenicity.


Asunto(s)
Bacteriófago lambda/metabolismo , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Cromosomas Bacterianos/genética , Genoma Bacteriano/genética , Humanos , Profagos/genética , Homología de Secuencia , Shigella/metabolismo
16.
Infect Immun ; 74(4): 2328-37, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16552063

RESUMEN

Attaching and effacing (A/E) pathogens are a significant cause of gastrointestinal illness in humans and animals. All A/E pathogens carry a large pathogenicity island, termed the locus for enterocyte effacement (LEE), which encodes a type III secretion system that translocates several effector proteins into host cells. To identify novel virulence determinants in A/E pathogens, we performed a signature-tagged mutagenesis screen in C57BL/6 mice by using the mouse A/E pathogen Citrobacter rodentium. Five hundred seventy-six derivatives of C. rodentium were tested in pools of 12 mutants. One attenuated mutant carried a transposon insertion in nleB, which encodes a putative effector of the LEE-encoded type III secretion system (T3SS). nleB is present in a genomic pathogenicity island that also encodes another putative effector, NleE, immediately downstream. Using translational fusions with beta-lactamase (TEM-1), we showed that both NleB and NleE were translocated into host cells by the LEE-encoded T3SS of enteropathogenic Escherichia coli. In addition, deletion of the gene encoding NleB in C. rodentium resulted in reduced colonization of mice in single infections and reduced colonic hyperplasia. In contrast, the deletion of other non-LEE-encoded effector genes in C. rodentium, nleC, nleD, or nleE, had no effect on host colonization or disease. These results suggest that nleB encodes an important virulence determinant of A/E pathogens.


Asunto(s)
Proteínas Bacterianas/fisiología , Citrobacter rodentium/crecimiento & desarrollo , Citrobacter rodentium/patogenicidad , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Factores de Virulencia/fisiología , Animales , Secuencia de Bases , Citrobacter rodentium/genética , Colon/microbiología , Cartilla de ADN , Infecciones por Enterobacteriaceae/genética , Islas Genómicas/genética , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutagénesis , Transporte de Proteínas/genética , Factores de Virulencia/genética
17.
Infect Immun ; 73(12): 8411-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16299341

RESUMEN

Intestinal colonization by enteropathogenic and enterohemorrhagic Escherichia coli requires the locus of enterocyte effacement-encoded type III secretion system. We report that NleC and NleD are translocated into host cells via this system. Deletion mutants induced attaching and effacing lesions in vitro, while infection of calves or lambs showed that neither gene was required for colonization.


Asunto(s)
Enterocitos/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Enterocitos/ultraestructura , Escherichia coli O157/genética , Proteínas de Escherichia coli/análisis , Proteínas de Escherichia coli/genética , Eliminación de Gen , Datos de Secuencia Molecular , Mutación , Transporte de Proteínas , Análisis de Secuencia de ADN , Ovinos
18.
Infect Immun ; 71(7): 3995-4002, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12819087

RESUMEN

Fecal samples from healthy children under 2 years of age living in Berlin, Germany (205 infants), and Melbourne, Australia (184 infants), were investigated for the presence of attaching and effacing (AE) Escherichia coli (AEEC) strains by screening for eae (intimin) genes. Twenty-seven AEEC strains were isolated from 14 children (7.6%) from Melbourne and from 12 children (5.9%) from Berlin. The 27 AEEC strains were classified as enterohemorrhagic E. coli (one strain, producing Shiga toxin 1), typical enteropathogenic E. coli (EPEC) (one strain carrying an EPEC adherence factor [EAF] plasmid), and atypical EPEC (25 strains negative for Shiga toxins and EAF plasmids). The AEEC were divided into 18 different serotypes, O-nontypeable and O-rough strains. Typing of their intimin genes revealed the presence of intimin alpha in 6 strains, intimin beta in 11 strains, intimin gamma in 7 strains, intimin zeta in 2 strains, and intimin eta in one strain. Analysis of HEp-2 cell adherence showed diffuse adherence or localized adherence-like patterns in 26 AEEC strains; local adherence was found only with the EAF-positive strain. Ten AEEC strains showed an AE property with the fluorescent actin staining (FAS) test. The introduction of an EAF plasmid (pMAR7) converted 11 FAS-negative AEEC strains to FAS positive and increased the FAS reaction in six FAS-positive AEEC strains, indicating that the genes needed for the AE phenotype were functional in these strains. Our finding indicates that atypical EPEC strains could play a double role as strains that naturally immunize against intimin in humans and as reservoirs for new emerging human pathogenic EPEC strains.


Asunto(s)
Actinas/química , Adhesinas Bacterianas/genética , Adhesión Bacteriana , Proteínas Portadoras/genética , Proteínas de Escherichia coli , Escherichia coli/patogenicidad , Actinas/análisis , Adhesinas Bacterianas/clasificación , Proteínas Portadoras/clasificación , Línea Celular , Escherichia coli/genética , Genotipo , Humanos , Lactante , Recién Nacido , Plásmidos , Virulencia
19.
Cell Microbiol ; 6(12): 1167-83, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15527496

RESUMEN

Subversion of host cell actin microfilaments is the hallmark of enterohaemorrhagic (EHEC) and enteropathogenic (EPEC) Escherichia coli infections. Both pathogens translocate the trans-membrane receptor protein-translocated intimin receptor (Tir), which links the extracellular bacterium to the cell cytoskeleton. While both converge on neural Wiskott-Aldrich syndrome protein (N-WASP), Tir-mediated actin accretion by EPEC and EHEC differ in that Tir(EPEC) requires both tyrosine phosphorylation and the host adaptor protein Nck, whereas Tir(EHEC) is not phosphorylated and utilizes an unidentified linker. Here we report the identification of Tir-cytoskeleton coupling protein (TccP), a novel EHEC effector that displays an Nck-like coupling activity following translocation into host cells. A tccP mutant did not affect Tir translocation and focusing but failed to recruit alpha-actinin, Arp3, N-WASP and actin to the site of bacterial adhesion. When expressed in EPEC, bacterial-derived TccP restored actin polymerization activity following infection of an Nck-deficient cell line. TccP has a similar biological activity on infected human intestinal explants ex vivo. Purified TccP activates N-WASP stimulating, in the presence of Arp2/3, actin polymerization in vitro. These results show that EHEC translocates both its own receptor (Tir) and an Nck-like protein (TccP) to facilitate actin polymerization.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli/metabolismo , Proteínas Oncogénicas/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/genética , Células HeLa , Humanos , Íleon , Proteínas Oncogénicas/genética , Técnicas de Cultivo de Órganos
20.
Infect Immun ; 70(2): 945-52, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11796630

RESUMEN

Shiga toxin-producing Escherchia coli (STEC) comprises a group of attaching and effacing (A/E) enteric pathogens of animals and humans. Natural and experimental infection of calves with STEC may result in acute enteritis or subclinical infection, depending on serotype- and host-specific factors. To quantify intestinal secretory and inflammatory responses to STEC in the bovine intestine, serotypes that are associated with human disease (O103:H2 and O157:H7) were introduced into ligated mid-ileal loops in gnotobiotic and conventional calves, and fluid accumulation and recruitment of radiolabeled neutrophils were measured after 12 h. STEC serotype O103:H2, but not serotype O157:H7, elicited strong enteropathogenic responses. To determine if the inflammatory response to STEC O103:H2 in calves requires Shiga toxin 1 or intimate bacterial attachment to the intestinal epithelium, defined mutations were made in the stx1, eae, and tir genes. Our data indicate that some STEC induce intestinal inflammatory responses in calves by a mechanism that is independent of A/E-lesion formation, intimin, or Shiga toxin 1. This may have implications for strategies to reduce STEC carriage in cattle.


Asunto(s)
Adhesinas Bacterianas/fisiología , Proteínas Bacterianas/fisiología , Proteínas Portadoras/fisiología , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Proteínas de Escherichia coli , Receptores de Superficie Celular/fisiología , Toxina Shiga I/metabolismo , Adhesinas Bacterianas/genética , Animales , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Bovinos , Escherichia coli , Infecciones por Escherichia coli/patología , Escherichia coli O157/genética , Células HeLa , Humanos , Íleon/microbiología , Microscopía Confocal/métodos , Mutagénesis , Receptores de Superficie Celular/genética , Serotipificación , Toxina Shiga I/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA