Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
J Biol Chem ; 289(7): 4334-45, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24362261

RESUMEN

Type IV pili are produced by many pathogenic Gram-negative bacteria and are important for processes as diverse as twitching motility, cellular adhesion, and colonization. Recently, there has been an increased appreciation of the ability of Gram-positive species, including Clostridium difficile, to produce Type IV pili. Here we report the first three-dimensional structure of a Gram-positive Type IV pilin, PilJ, demonstrate its incorporation into Type IV pili, and offer insights into how the Type IV pili of C. difficile may assemble and function. PilJ has several unique structural features, including a dual-pilin fold and the incorporation of a structural zinc ion. We show that PilJ is incorporated into Type IV pili in C. difficile and present a model in which the incorporation of PilJ into pili exposes the C-terminal domain of PilJ to create a novel interaction surface.


Asunto(s)
Clostridioides difficile/química , Proteínas Fimbrias/química , Pliegue de Proteína , Clostridioides difficile/metabolismo , Clostridioides difficile/ultraestructura , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/química , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/ultraestructura , Estructura Terciaria de Proteína
3.
Cell Physiol Biochem ; 32(2): 417-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23988581

RESUMEN

BACKGROUND/AIMS: Stabilization of the hypoxia-inducible factor (HIF-1α) is proposed to provide a protective host-response to C. difficile intoxication. Here, we aimed to elucidate whether nitric oxide and/or reactive oxygen species produced during C. difficile toxin exposure could influence HIF-1α stability and initiate protection against epithelial cell damage. METHODS/RESULTS: HIF-1α and inducible nitric oxide synthase (iNOS) proteins were up-regulated whereas factor-inhibiting HIF-1 (FIH-1) protein was down-regulated in Caco-2 epithelial cell monolayers with in vitro toxin exposure. We demonstrate using the biotin-switch assay that the stabilization of HIF-1α protein occurred via iNOS-dependent nitrosylation. Inhibition of iNOS activity by selective inhibitor (1400W) attenuated HIF-1α stabilization and exacerbated toxin-dependent disruptions in Caco-2 monolayer morphology and tight junctional integrity in vitro. Treatment of Caco-2 cell monolayers with N-actylcysteine (NAC), a scavenger of reactive oxygen species (ROS), attenuated toxin-dependent increases in iNOS and HIF-1α protein levels but had no effect on FIH-1 responses. In addition, mice that were exposed to C. difficile toxin in vivo also demonstrated a significant increase in HIF-1α protein and nitrosylation levels. CONCLUSION: Taken together, these data suggest that important synergistic actions exist between nitric oxide and ROS to stabilize HIF-1α and its innate, protective actions in the context of C. difficile toxin-mediated epithelial injury.


Asunto(s)
Toxinas Bacterianas/toxicidad , Clostridioides difficile , Células Epiteliales/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Óxido Nítrico/farmacología , Especies Reactivas de Oxígeno/farmacología , Animales , Células CACO-2 , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Estabilidad Proteica/efectos de los fármacos
4.
Infect Immun ; 80(12): 4474-84, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23045481

RESUMEN

Clostridium difficile, a major cause of hospital-acquired diarrhea, triggers disease through the release of two toxins, toxin A (TcdA) and toxin B (TcdB). These toxins disrupt the cytoskeleton of the intestinal epithelial cell, increasing intestinal permeability and triggering the release of inflammatory mediators resulting in intestinal injury and inflammation. The most prevalent animal model to study TcdA/TcdB-induced intestinal injury involves injecting toxin into the lumen of a surgically generated "ileal loop." This model is time-consuming and exhibits variability depending on the expertise of the surgeon. Furthermore, the target organ of C. difficile infection (CDI) in humans is the colon, not the ileum. In the current study, we describe a new model of CDI that involves intrarectal instillation of TcdA/TcdB into the mouse colon. The administration of TcdA/TcdB triggered colonic inflammation and neutrophil and macrophage infiltration as well as increased epithelial barrier permeability and intestinal epithelial cell death. The damage and inflammation triggered by TcdA/TcdB isolates from the VPI and 630 strains correlated with the concentration of TcdA and TcdB produced. TcdA/TcdB exposure increased the expression of a number of inflammatory mediators associated with human CDI, including interleukin-6 (IL-6), gamma interferon (IFN-γ), and IL-1ß. Finally, we were able to demonstrate that TcdA was much more potent at inducing colonic injury than was TcdB but TcdB could act synergistically with TcdA to exacerbate injury. Taken together, our data indicate that the intrarectal murine model provides a robust and efficient system to examine the effects of TcdA/TcdB on the induction of inflammation and colonic tissue damage in the context of human CDI.


Asunto(s)
Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Clostridioides difficile/patogenicidad , Modelos Animales de Enfermedad , Enterocolitis Seudomembranosa/patología , Enterotoxinas/toxicidad , Inflamación/patología , Administración Rectal , Animales , Proteínas Bacterianas/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Clostridioides difficile/metabolismo , Colon/patología , Relación Dosis-Respuesta a Droga , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/mortalidad , Enterotoxinas/administración & dosificación , Femenino , Humanos , Inflamación/inmunología , Inflamación/mortalidad , Ratones , Ratones Endogámicos C57BL
5.
Infect Immun ; 79(10): 4061-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21788384

RESUMEN

Although toxins A and B are known to be important contributors to the acute phase of Clostridium difficile infection, the role of colonization and adherence to host tissues in the overall pathogenesis of these organisms remains unclear. Consequently, we used the recently introduced intron-based ClosTron gene interruption system to eliminate the expression of two reported C. difficile colonization factors, the major flagellar structural subunit (FliC) and the flagellar cap protein (FliD), to gain greater insight into how flagella and motility contribute to C. difficile's pathogenic strategy. The results demonstrate that interrupting either the fliC or the fliD gene results in a complete loss of flagella, as well as motility, in C. difficile. However, both the fliC and fliD mutant strains adhered better than the wild-type 630Δerm strain to human intestine-derived Caco-2 cells, suggesting that flagella and motility do not contribute to, or may even interfere with, C. difficile adherence to epithelial cell surfaces in vitro. Moreover, we found that the mutant strains were more virulent in hamsters, indicating either that flagella are unnecessary for virulence or that repression of motility may be a pathogenic strategy employed by C. difficile in hamsters.


Asunto(s)
Proteínas Bacterianas/genética , Clostridioides difficile/patogenicidad , Flagelos/metabolismo , Mutación , Animales , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Células CACO-2 , Clostridioides difficile/genética , Clostridioides difficile/metabolismo , Clostridioides difficile/fisiología , Infecciones por Clostridium/microbiología , Cricetinae , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Flagelos/genética , Humanos , Intestinos/microbiología , Virulencia
6.
Glycobiology ; 21(9): 1217-27, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21610194

RESUMEN

The binding of recombinant fragments of the C-terminal cell-binding domains of the two large exotoxins, toxin A (TcdA) and toxin B (TcdB), expressed by Clostridium difficile and a library consisting of the most abundant neutral and acidic human milk oligosaccharides (HMOs) was examined quantitatively at 25°C and pH 7 using the direct electrospray ionization mass spectrometry (ES-MS) assay. The results of the ES-MS measurements indicate that both toxin fragments investigated, TcdB-B1 and TcdA-A2, which possess one and two carbohydrate binding sites, respectively, bind specifically to HMOs ranging in size from tri- to heptasaccharides. Notably, five of the HMOs tested bind to both toxins: Fuc(α1-2)Gal(ß1-4)Glc, Gal(ß1-3)GlcNAc(ß1-3)Gal(ß1-4)Glc, Fuc(α1-2)Gal(ß1-3)GlcNAc(ß1-3)Gal(ß1-4)Glc, Gal(ß1-3)[Fuc(α1-4)]GlcNAc(ß1-3)Gal(ß1-4)Glc and Gal(ß1-4)[Fuc(α1-3)]GlcNAc(ß1-3)Gal(ß1-4)Glc. However, the binding of the HMOs is uniformly weak, with apparent affinities ≤10(3 )M(-1). The results of molecular docking simulations, taken together with the experimental binding data, suggest that a disaccharide moiety (lactose or lactosamine) represents the core HMO recognition element for both toxin fragments. The results of a Verocytotoxicity neutralization assay reveal that HMOs do not significantly inhibit the cytotoxic effects of TcdA or TcdB. The absence of protection is attributed to the very weak intrinsic affinities that the toxins exhibit towards the HMOs.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/química , Enterotoxinas/metabolismo , Leche Humana/química , Oligosacáridos , Fragmentos de Péptidos/metabolismo , Amino Azúcares/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacología , Toxinas Bacterianas/química , Toxinas Bacterianas/farmacología , Sitios de Unión , Secuencia de Carbohidratos , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Enterotoxinas/química , Enterotoxinas/farmacología , Humanos , Cinética , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Oligosacáridos/análisis , Oligosacáridos/química , Oligosacáridos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Unión Proteica , Espectrometría de Masa por Ionización de Electrospray , Células Vero
7.
Mol Microbiol ; 76(5): 1095-110, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20444097

RESUMEN

The Cpx envelope stress response is induced by the misfolding of periplasmic proteins and restores envelope homeostasis by upregulating several periplasmic protein folding and degrading factors. The Cpx response also regulates the expression of a variety of envelope-spanning protein complexes, including flagella, secretion systems and pili, which play an important role in pathogenesis. In a previous study, we inactivated the Cpx response in enteropathogenic Escherichia coli (EPEC), a causative agent of infant diarrhoea, and observed decreased expression of its major adhesin, the bundle-forming pilus (BFP). Here, we examined the mechanism underlying this BFP expression defect, and found that this phenotype can be attributed to insufficient expression of periplasmic folding factors, such as DsbA, DegP and CpxP. Hence, a low level of Cpx pathway activity promotes BFP synthesis by upregulating factors important for folding of BFP component proteins. Conversely, we found that full induction of the Cpx response inhibits BFP expression, mainly by repressing transcription of the bfp gene cluster. In combination with a previous report examining EPEC type III secretion, our results demonstrate that the Cpx response co-ordinates the repression of cell-surface structures during periods of envelope stress.


Asunto(s)
Escherichia coli Enteropatógena/citología , Escherichia coli Enteropatógena/fisiología , Proteínas de Escherichia coli/metabolismo , Fimbrias Bacterianas/metabolismo , Proteínas Quinasas/metabolismo , Estrés Fisiológico , Adhesión Bacteriana , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Humanos , Lactante , Pliegue de Proteína , Proteínas Quinasas/genética , Transcripción Genética
8.
Mol Microbiol ; 76(5): 1111-26, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20487271

RESUMEN

Enteropathogenic Escherichia coli (EPEC) are a major cause of infant morbidity and mortality due to diarrhoea in developing countries. The pathogenesis of EPEC is dependent on a coordinated multi-step process culminating in the intimate adherence of the organisms to the host's intestinal mucosa. During the initial stages of the EPEC colonization process, the fimbrial adhesin, bundle-forming pili (BFP), plays an integral role. We previously reported that the major BFP structural subunit, bundlin, displays lectin-like properties, which enables BFP to initially tether EPEC to N-acetyllactosamine (LacNAc) glycan receptors on host cell surfaces. We also reported that incubating EPEC with synthetic LacNAc-bearing neoglycoconjugates not only inhibits their adherence to host cells, but also induces BFP retraction and subsequent degradation of the bundlin subunits. Herein, we demonstrate that the periplasmic serine protease, DegP, is required for degrading bundlin during this process. We also show that DegP appears to act as a bundlin chaperone during BFP assembly and that LacNAc-BSA-induced BFP retraction is followed by transcriptional upregulation of the BFP operon and downregulation of the locus of enterocyte effacement operons in EPEC.


Asunto(s)
Amino Azúcares/farmacología , Escherichia coli Enteropatógena , Fimbrias Bacterianas/efectos de los fármacos , Fimbrias Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Escherichia coli Enteropatógena/citología , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Lactante , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Estructura Molecular , Operón , Proteínas Periplasmáticas/genética , Proteínas Periplasmáticas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transcripción Genética
9.
Gastroenterology ; 139(2): 542-52, 552.e1-3, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20398664

RESUMEN

BACKGROUND & AIMS: Clostridium difficile-associated disease (CDAD) is the leading cause of nosocomial diarrhea in the United States. C difficile toxins TcdA and TcdB breach the intestinal barrier and trigger mucosal inflammation and intestinal damage. The inflammasome is an intracellular danger sensor of the innate immune system. In the present study, we hypothesize that TcdA and TcdB trigger inflammasome-dependent interleukin (IL)-1beta production, which contributes to the pathogenesis of CDAD. METHODS: Macrophages exposed to TcdA and TcdB were assessed for IL-1beta production, an indication of inflammasome activation. Macrophages deficient in components of the inflammasome were also assessed. Truncated/mutated forms of TcdB were assessed for their ability to activate the inflammasome. The role of inflammasome signaling in vivo was assessed in ASC-deficient and IL-1 receptor antagonist-treated mice. RESULTS: TcdA and TcdB triggered inflammasome activation and IL-1beta secretion in macrophages and human mucosal biopsy specimens. Deletion of Nlrp3 decreased, whereas deletion of ASC completely abolished, toxin-induced IL-1beta release. TcdB-induced IL-1beta release required recognition of the full-length toxin but not its enzymatic function. In vivo, deletion of ASC significantly reduced toxin-induced inflammation and damage, an effect that was mimicked by pretreatment with the IL-1 receptor antagonist anakinra. CONCLUSIONS: TcdA and TcdB trigger IL-1beta release by activating an ASC-containing inflammasome, a response that contributes to toxin-induced inflammation and damage in vivo. Pretreating mice with the IL-1 receptor antagonist anakinra afforded the same level of protection that was observed in ASC-/- mice. These data suggest that targeting inflammasome or IL-1beta signaling may represent new therapeutic targets in the treatment of CDAD.


Asunto(s)
Clostridioides difficile/patogenicidad , Ileítis/inmunología , Íleon/inmunología , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Biopsia , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Línea Celular , Clostridioides difficile/genética , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Endocitosis , Endosomas/inmunología , Endosomas/microbiología , Enterotoxinas/genética , Humanos , Ileítis/microbiología , Ileítis/patología , Ileítis/prevención & control , Íleon/efectos de los fármacos , Íleon/microbiología , Íleon/patología , Inmunidad Innata/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR
10.
Proc Natl Acad Sci U S A ; 105(44): 16837-42, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18955695

RESUMEN

We demonstrate that interactions between multimeric receptors and multivalent ligands are dramatically enhanced by recruiting a complementary templating receptor such as an endogenous multimeric protein but only when individual ligands are attached to a polymer as preorganized, covalent, heterobifunctional pairs. This effect cannot be replicated by a multivalent ligand if the same recognition elements are independently arrayed on the scaffold. Application of this principle offers an approach to create high-avidity inhibitors for multimeric receptors. Judicious selection of the ligand that engages the templating protein allows appropriate effector function to be incorporated in the polymeric construct, thereby providing an opportunity for therapeutic applications. The power of this approach is exemplified by the design of exceptionally potent Escherichia coli Shiga toxin antagonists that protect transgenic mice that constitutively express a human pentraxin, serum amyloid P component.


Asunto(s)
Antibacterianos/química , Escherichia coli O157/efectos de los fármacos , Toxina Shiga I/antagonistas & inhibidores , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacología , Escherichia coli O157/metabolismo , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Polímeros/síntesis química , Polímeros/química , Polímeros/farmacología , Componente Amiloide P Sérico/metabolismo , Toxina Shiga I/química
11.
Mol Microbiol ; 72(4): 859-68, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19400799

RESUMEN

Bundle-forming pili (BFP) promote the adherence of typical enteropathogenic Escherichia coli (EPEC) to human intestinal epithelial cells. BFP are polymers of bundlin and nine bundlin alleles have been identified in EPEC isolated from diverse sources. These alleles are divided into two main groups, alpha and beta, based on their amino acid sequences. Alpha bundlins are also N-acetyllactosamine- (LacNAc) specific lectins and bind to HEp-2 cells, whereas beta bundlins do not display these characteristics. The four surface-exposed regions of amino acid sequence heterogeneity between alpha and beta bundlin were therefore investigated as potential LacNAc-specific carbohydrate-binding domains in a bundlin. Mutation of one of these domains, 137-GENNI-141, in alpha(1) bundlin to that of beta bundlin (136-SPDST-140) resulted in BFP that no longer bound to LacNAc or HEp-2 cells. Conversely, mutating the beta3 bundlin gene to encode the alpha bundlin sequence at this domain resulted in the gain of HEp-2 cell adherence. The importance of this domain in carbohydrate binding is supported by the finding that introducing the mutation GENNI-->GENNT altered the alpha1 bundlin carbohydrate-binding specificity from LacNAc to the Lewis X glycan sequence.


Asunto(s)
Amino Azúcares/metabolismo , Escherichia coli Enteropatógena/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Adhesión Bacteriana , Línea Celular , Proteínas de Escherichia coli/genética , Proteínas Fimbrias/genética , Humanos , Lectinas/metabolismo , Mutación , Estructura Terciaria de Proteína , Especificidad por Sustrato
12.
Biochemistry ; 48(23): 5365-74, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19400587

RESUMEN

A systematic investigation into the assembly and stability of native and modified subunits of the Shiga toxins (Stx) in vitro is described. Analysis of the assembly of native and modified B subunits of Stx1 and Stx2 in solution, carried out using electrospray ionization mass spectrometry (ES-MS), suggests that the lower thermodynamic stability of the B subunit homopentamer of Stx2, compared to that of Stx1, is due to the presence of a repulsive interaction involving Asp70 of the Stx2 B subunit. In Stx1 B, the corresponding (spatially) residue is Arg. Using temperature-controlled ES-MS, it is shown that the Stx1 and Stx2 holotoxins exhibit differences in their resistance to temperature- and acid-induced dissociation. However, both Stx1 and Stx2 are fully assembled at pH >3.5 and 37 degrees C. This finding has several important biological implications. First, it argues against the likelihood that the difference in Stx1 and Stx2 toxicity arises from differential dissociation of the toxins during the intracellular trafficking steps of the cellular intoxication process. Second, it implies that the activation of the A subunits of Stx1 and Stx2 by enzymatic cleavage must occur while the A subunit is assembled with the B subunit homopentamer. It is, therefore, proposed that the differential toxicities of Stx1 and Stx2 reflect the relative efficiencies of intracellular activation of the A subunits.


Asunto(s)
Toxinas Shiga/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Escherichia coli , Subunidades de Proteína/química , Toxina Shiga I/química , Toxina Shiga II/química , Temperatura
13.
Infect Immun ; 77(10): 4463-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19635829

RESUMEN

Enteropathogenic Escherichia coli (EPEC) strains cause watery diarrhea almost exclusively in young children. The basis for this age discrimination has never been determined, but it may be related to host cell receptors. During infection, EPEC strains express type IV bundle-forming pili composed of repeating subunits of the protein called bundlin. The very first interaction between EPEC and in vitro-cultured epithelial cells is mediated by the binding of alpha-bundlin to a carbohydrate receptor that contains, at a minimum, the N-acetyllactosamine (LacNAc) glycan sequence. However, bundlins expressed from the beta-bundlin allele do not bind LacNAc glycan sequences. Herein, we investigated whether EPEC strains use alpha-bundlin to mediate early adherence to human intestinal biopsy specimens cultured in vitro by assessing the ability of isogenic EPEC mutants expressing either the alpha(1)- or beta(1)-bundlin allele or a bundlin-deficient EPEC strain to bind to these specimens. Furthermore, we directly compared the abilities of a wild-type EPEC strain to bind to the epithelial surfaces of both human adult and pediatric biopsy specimens. Our results demonstrate that beta-bundlin does not act as an adhesin during early EPEC adherence to adult duodenal biopsy specimens. The results also indicate that EPEC binds equally well to adult and pediatric biopsy specimens in an early adherence assay. This result is supported by the finding that the early adherence of EPEC to both adult and pediatric biopsy specimens was inhibited by LacNAc neoglycoconjugates, suggesting that organisms expressing alpha-bundlin-type bundle-forming pili initially bind to related glycan receptors in both age groups.


Asunto(s)
Adhesinas Bacterianas/fisiología , Adhesión Bacteriana , Escherichia coli Enteropatógena/fisiología , Proteínas de Escherichia coli/fisiología , Proteínas Fimbrias/fisiología , Factores de Virulencia/fisiología , Adhesinas Bacterianas/genética , Adulto , Anciano , Preescolar , Proteínas de Escherichia coli/genética , Femenino , Proteínas Fimbrias/genética , Eliminación de Gen , Humanos , Lactante , Masculino , Persona de Mediana Edad , Factores de Virulencia/genética , Adulto Joven
14.
Cell Microbiol ; 10(1): 177-87, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17697132

RESUMEN

Synthetic N-acetyllactosamine (LacNAc) glycoside sequences coupled to BSA competitively inhibit enteropathogenic Escherichia coli (EPEC) localized adherence (LA) to human intestinal biopsy specimens and tissue culture cell monolayers. The LacNAc-specific adhesin appears to be associated with the bundle-forming pili (BFP) expressed by EPEC during the early stages of colonization. Herein, we report that recombinant bundlin inhibits EPEC LA to HEp-2 cells and binds to HEp-2 cells. Recombinant bundlin also binds, with millimolar association constants (K(assoc)), to synthetic LacNAc-Benzene and LacNAc-O(CH(2))(8)CONH(2) glycosides as assessed in the gas phase by nanoelectrospray ionization mass spectrometry. Furthermore, LacNAc-BSA inhibits LA only of EPEC strains that express alpha bundlin alleles, suggesting putative locations for the LacNAc-binding pocket in the alpha bundlin monomer. Collectively, these results suggest that alpha bundlin possesses lectin-like properties that are responsible for LacNAc-specific initial adherence of alpha bundlin-expressing EPEC strains to host intestinal epithelial cells.


Asunto(s)
Amino Azúcares/metabolismo , Adhesión Bacteriana/fisiología , Escherichia coli Enteropatógena/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Lectinas/metabolismo , Línea Celular , Proteínas Fimbrias/genética , Proteínas Fimbrias/aislamiento & purificación , Humanos , Cinética , Lectinas/genética , Lectinas/aislamiento & purificación , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray
17.
J Proteomics ; 198: 145-150, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30716422

RESUMEN

After we published our preliminary study on the use of liquid chromatography-tandem mass spectrometry (LC-MS/MS) and curated E. coli toxin databases on the identification of E. coli Shiga toxins (Stxs) in the Journal of Proteomics in year 2018, we were encouraged to further refine the method and test clinical isolates. In this study, different concentrations of mitomycin C (MMC) and ciprofloxacin (CF), two common antibiotic/chemotherapy agents capable of stimulating Stx production, were first tested and compared on three reference strains and eight clinical isolates to observe the toxin induction and subsequent identification. Notably, no differences were observed between the two agents other than the concentrations applied. Seventeen more clinical isolates were then tested using fixed MMC and CF concentrations and sample amount. This study confirms that the majority of stx2-positive E. coli strains can be stimulated to produce sufficient toxin for confident identification. This does not occur with stx1-positive E. coli isolates, however, despite the fact that both Stxs can be identified for several isolates without MMC or CF stimulation. BIOLOGICAL SIGNIFICANCE: Stxs, especially Stx2, are very important causes of severe food-borne disease, even death. This study confirms that receptor analogue-based affinity enrichment of Stxs, after MMC or CF treatment of E. coli, is useful for fast and accurate Stx2 identification through LC-MS/MS.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Proteómica , Toxina Shiga I , Toxina Shiga II , Escherichia coli Shiga-Toxigénica/metabolismo , Cromatografía Liquida , Humanos , Toxina Shiga I/análisis , Toxina Shiga I/metabolismo , Toxina Shiga II/análisis , Toxina Shiga II/metabolismo , Espectrometría de Masas en Tándem
18.
Glycobiology ; 18(9): 698-706, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18509107

RESUMEN

The biological and ligand-binding properties of recombinant C-terminal cell-binding domains (CBDs) and subdomains of the two large exotoxins, Toxin A (TcdA) and Toxin B (TcdB) expressed by Clostridium difficile were examined in the hemagglutination and Verocytotoxicity neutralization assays and by qualitative affinity chromatography using Sepharose-linked alpha Gal(1,3)betaGal(1,4)beta Glc as well as the direct electrospray ionization mass spectrometry (ES-MS) assay. These studies revealed that, whereas the full-length TcdA CBD agglutinated rabbit erythrocytes, neutralized TcdA-mediated Vero cell death and bound to alpha Gal(1,3)betaGal(1,4)beta Glc-derivatized Sepharose, the TcdB CBD was inactive in these functional assays. Moreover, retention by alpha Gal(1,3)betaGal(1,4)beta Glc-derivatized Sepharose corresponded to the number of available TcdA subdomain ligand-binding sites. By contrast, the ES-MS assays revealed that both the TcdA and TcdB CBD bind to 8-methoxycarbonyloctyl-alpha Gal(1,3)betaGal(1,4)beta Glc sequences with similar avidities. Additional ES-MS experiments using chemically altered alpha Gal(1,3)betaGal(1,4)beta Glc sequences also revealed that the TcdA and TcdB CBD will tolerate a fair amount of structural variation in their complementary glycan ligands. Although the studies are consistent with the known ligand-binding properties of the TcdA and TcdB holotoxins, they also revealed subtle heretofore unrecognized functional differences in their receptor recognition properties.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Toxinas Bacterianas/química , Enterotoxinas/química , Enterotoxinas/fisiología , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Chlorocebus aethiops , Clostridioides difficile/metabolismo , Enterotoxinas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Modelos Biológicos , Modelos Moleculares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/fisiología , Unión Proteica , Estructura Terciaria de Proteína/fisiología , Pruebas de Toxicidad , Células Vero
19.
J Proteomics ; 180: 36-40, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28602982

RESUMEN

Toxin expression is a key factor in Shiga toxin (Stx)-producing E. coli, a common pathogen involved in foodborne disease outbreaks. A liquid chromatography-tandem mass spectrometry (LC-MS/MS) based approach has been used in this study to identify commonly reported E. coli toxins, with a focus on Shiga toxins (Stxs). Different sample preparation methods using variable culture conditions and concentrations of mitomycin C (MMC), a common antibiotic/chemotherapy agent capable of stimulating Stx production, were first tested on reference strains EDL933 and 90-2380 by LC-MS/MS detection of tryptic digests of receptor-analogue affinity binding enriched Stx preparations from culture supernatants and lysates. A curated E. coli protein toxin database was also used for faster and more straightforward toxin identification. With eight more genetically confirmed E. coli strains examined to verify the method, this preliminary study indicates that receptor-analogue based affinity enrichment on cell lysate or supernatant is a sensitive and accurate method for Stx identification. BIOLOGICAL SIGNIFICANCE: The existence of Stx is very important for identifying Stx-producing E. coli and implementing a clinical treatment regime. This study demonstrates for the first time that using a curated E. coli toxin database, together with receptor-analogue-based affinity enrichment of Stxs after MMC treatment of E. coli, is an easy and appropriate approach for fast and accurate Stx identification through LC-MS/MS.


Asunto(s)
Bases de Datos de Proteínas , Proteínas de Escherichia coli/metabolismo , Toxina Shiga/metabolismo , Escherichia coli Shiga-Toxigénica/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem
20.
Cell Rep ; 25(6): 1525-1536.e7, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30404007

RESUMEN

The non-canonical caspase-4 and canonical NLRP3 inflammasomes are both activated by intracellular lipopolysaccharide (LPS), but the crosstalk between these two pathways remains unclear. Shiga toxin 2 (Stx2)/LPS complex, from pathogenic enterohemorrhagic Escherichia coli, activates caspase-4, gasdermin D (GSDMD), and the NLRP3 inflammasome in human THP-1 macrophages, but not mouse macrophages that lack the Stx receptor CD77. Stx2/LPS-mediated IL-1ß secretion and pyroptosis are dependent on mitochondrial reactive oxygen species (ROS) downstream of the non-canonical caspase-4 inflammasome and cleaved GSDMD, which is enriched at the mitochondria. Blockade of caspase-4 activation and ROS generation as well as GSDMD deficiency significantly reduces Stx2/LPS-induced IL-1ß production and pyroptosis. The NLRP3 inflammasome plays a significant role in amplifying Stx2/LPS-induced GSDMD cleavage and pyroptosis, with significant reduction of these responses in NLRP3-deficient THP-1 cells. Together, these data show that Stx2/LPS complex activates the non-canonical inflammasome and mitochondrial ROS upstream of the NLRP3 inflammasome to promote cytokine maturation and pyroptosis.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Lipopolisacáridos/farmacología , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Toxina Shiga/farmacología , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Proteínas de Unión a Fosfato , Piroptosis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA