Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Intervalo de año de publicación
1.
Microb Cell Fact ; 21(1): 133, 2022 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-35780105

RESUMEN

BACKGROUND: Bacterial type III secretion systems (T3SSs) assemble a multiprotein complex termed the injectisome, which acts as a molecular syringe for translocation of specific effector proteins into the cytoplasm of host cells. The use of injectisomes for delivery of therapeutic proteins into mammalian cells is attractive for biomedical applications. With that aim, we previously generated a non-pathogenic Escherichia coli strain, called Synthetic Injector E. coli (SIEC), which assembles functional injectisomes from enteropathogenic E. coli (EPEC). The assembly of injectisomes in EPEC is assisted by the lytic transglycosylase EtgA, which degrades the peptidoglycan layer. As SIEC lacks EtgA, we investigated whether expression of this transglycosylase enhances the protein translocation capacity of the engineered bacterium. RESULTS: The etgA gene from EPEC was integrated into the SIEC chromosome under the control of the inducible tac promoter, generating the strain SIEC-eEtgA. The controlled expression of EtgA had no effect on the growth or viability of bacteria. Upon induction, injectisome assembly was ~ 30% greater in SIEC-eEtgA than in the parental strain, as determined by the level of T3SS translocon proteins, the hemolytic activity of the bacterial strain, and the impairment in flagellar motility. The functionality of SIEC-eEtgA injectisomes was evaluated in a derivative strain carrying a synthetic operon (eLEE5), which was capable of delivering Tir effector protein into the cytoplasm of HeLa cells triggering F-actin polymerization beneath the attached bacterium. Lastly, using ß-lactamase as a reporter of T3SS-protein injection, we determined that the protein translocation capacity was ~ 65% higher in the SIEC-EtgA strain than in the parental SIEC strain. CONCLUSIONS: We demonstrate that EtgA enhances the assembly of functional injectisomes in a synthetic injector E. coli strain, enabling the translocation of greater amounts of proteins into the cytoplasm of mammalian cells. Accordingly, EtgA expression may boost the protein translocation of SIEC strains programmed as living biotherapeutics.


Asunto(s)
Ingeniería Celular , Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Glicosiltransferasas , Ingeniería Celular/métodos , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicosiltransferasas/metabolismo , Células HeLa , Humanos , Transporte de Proteínas
2.
Gut Microbes ; 14(1): 2013763, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34965187

RESUMEN

Many bacterial pathogens employ a protein complex, termed the type III secretion system (T3SS), to inject bacterial effectors into host cells. These effectors manipulate various cellular processes to promote bacterial growth and survival. The T3SS complex adopts a nano-syringe shape that is assembled across the bacterial membranes, with an extracellular needle extending toward the host cell membrane. The assembly of the T3SS is initiated by the association of three proteins, known as SctR, SctS, and SctT, which create an entry portal to the translocation channel within the bacterial inner membrane. Using the T3SS of enteropathogenic Escherichia coli, we investigated, by mutational and functional analyses, the role of two structural construction sites formed within the SctRST complex and revealed that they are mutation-resistant components that are likely to act as seals preventing leakage of ions and metabolites rather than as substrate gates. In addition, we identified two residues in the SctS protein, Pro23, and Lys54, that are critical for the proper activity of the T3SS. We propose that Pro23 is critical for the physical orientation of the SctS transmembrane domains that create the tip of the SctRST complex and for their positioning with regard to other T3SS substructures. Surprisingly, we found that SctS Lys54, which was previously suggested to mediate the SctS self-oligomerization, is critical for T3SS activity due to its essential role in SctS-SctT hetero-interactions.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Secuencias de Aminoácidos , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Unión Proteica , Dominios Proteicos , Sistemas de Secreción Tipo III/química , Sistemas de Secreción Tipo III/genética
3.
Mikrochim Acta ; 188(11): 396, 2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34714421

RESUMEN

Rapid and sensitive detection of enteropathogenic Escherichia coli (EPEC) in fluids with complex background is an important task for safety quality control in the field of medicine, environment, and food. In this study, a gold foil paper-based aptasensor was developed for the detection of enteropathogenic EPEC O26:K60 with surface-enhanced Raman spectroscopy (SERS) and magnetic separation technology mediated by Fe3O4@Au composite. The gold foil paper was firstly modified with thiolated capture probe and SERS tag. The thiolated aptamer probe for EPEC was immobilized onto a Fe3O4@Au composite. In the presence of EPEC, highly specific recognition between the aptamer probe and EPEC made the Fe3O4@Au composite partially dissociated from the gold foil paper. This led to a decreased Raman intensity response, which showed an obvious negative linear correlation with increasing concentration of EPEC over a wide concentration range from 10 to 107 CFU/mL under an excitation wavelength of 633 nm. The detection limit was about 2.86 CFU/mL in a buffer solution and a licorice extractum and the detection time was only 2.5 h. The results demonstrate that the gold foil paper-based aptasensor can be an excellent biosensing platform that offers a reliable, rapid, and sensitive alternative for EPEC detection.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Escherichia coli Enteropatógena/aislamiento & purificación , Oro/química , Papel , Ácidos Borónicos/química , Escherichia coli Enteropatógena/química , Glycyrrhiza/microbiología , Ácidos Nucleicos Inmovilizados/química , Límite de Detección , Fenómenos Magnéticos , Nanopartículas de Magnetita/química , Nanocompuestos/química , Espectrometría Raman , Compuestos de Sulfhidrilo/química
4.
Sci Rep ; 9(1): 138, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30644424

RESUMEN

The carbon storage regulator A (CsrA) is a conserved global regulatory system known to control central carbon pathways, biofilm formation, motility, and pathogenicity. The aim of this study was to characterize changes in major metabolic pathways induced by CsrA in human enteropathogenic Escherichia coli (EPEC) grown under virulence factor-inducing conditions. For this purpose, the metabolomes and transcriptomes of EPEC and an isogenic ∆csrA mutant derivative were analyzed by untargeted mass spectrometry and RNA sequencing, respectively. Of the 159 metabolites identified from untargeted GC/MS and LC/MS data, 97 were significantly (fold change ≥ 1.5; corrected p-value ≤ 0.05) regulated between the knockout and the wildtype strain. A lack of csrA led to an accumulation of fructose-6-phosphate (F6P) and glycogen synthesis pathway products, whereas metabolites in lower glycolysis and the citric acid cycle were downregulated. Associated pathways from the citric acid cycle like aromatic amino acid and siderophore biosynthesis were also negatively influenced. The nucleoside salvage pathways were featured by an accumulation of nucleosides and nucleobases, and a downregulation of nucleotides. In addition, a pronounced downregulation of lyso-lipid metabolites was observed. A drastic change in the morphology in the form of vesicle-like structures of the ∆csrA knockout strain was visible by electron microscopy. Colanic acid synthesis genes were strongly (up to 50 fold) upregulated, and the abundance of colanic acid was 3 fold increased according to a colorimetric assay. The findings expand the scope of pathways affected by the csrA regulon and emphasize its importance as a global regulator.


Asunto(s)
Escherichia coli Enteropatógena/química , Proteínas de Escherichia coli/farmacología , Metaboloma/efectos de los fármacos , Proteínas de Unión al ARN/farmacología , Proteínas Represoras/farmacología , Transcriptoma/efectos de los fármacos , Secuencia de Bases , Cromatografía Liquida , Ciclo del Ácido Cítrico/efectos de los fármacos , Escherichia coli , Cromatografía de Gases y Espectrometría de Masas , Regulación Bacteriana de la Expresión Génica , Glucógeno/biosíntesis , Humanos , Regulón/genética
5.
Anat Rec (Hoboken) ; 301(12): 2103-2111, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30312538

RESUMEN

The ingestion of enteropathogenic Escherichia coli (EPEC), Listeria monocytogenes, or Salmonella enterica serovar Typhimurium leads to their colonization of the intestinal lumen, which ultimately causes an array of ailments ranging from diarrhea to bacteremia. Once in the intestines, these microbes generate various actin-rich structures to attach, invade, or move within the host intestinal epithelial cells. Although an assortment of actin-associated proteins has been identified to varying degrees at these structures, the localization of many actin stabilizing proteins have yet to be analyzed. Here, we examined the recruitment of the actin-associated proteins, calponin 1 and 2 at EPEC pedestals, L. monocytogenes actin clouds, comet tails and listeriopods, and S. Typhimurium membrane ruffles. In other systems, calponins are known to bind to and stabilize actin filaments. In EPEC pedestals, calponin 1 was recruited uniformly throughout the structures while calponin 2 was enriched at the apical tip. During L. monocytogenes infections, calponin 1 was found through all the actin-rich structures generated by the bacteria, while calponin 2 was only present within actin-rich structures formed by L. monocytogenes near the host cell membrane. Finally, both calponins were found within S. Typhimurium-generated membrane ruffles. Taken together, we have shown that although calponin 1 is recruited to actin-rich structures formed by the three bacteria, calponin 2 is specifically recruited to only membrane-bound actin-rich structures formed by the bacteria. Thus, our findings suggest that calponin 2 is a novel marker for membrane-bound actin structures formed by pathogenic bacteria. Anat Rec, 301:2103-2111, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Actinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Escherichia coli Enteropatógena/metabolismo , Listeria monocytogenes/metabolismo , Proteínas de Microfilamentos/metabolismo , Salmonella enterica/metabolismo , Actinas/análisis , Células CACO-2 , Proteínas de Unión al Calcio/análisis , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Escherichia coli Enteropatógena/química , Humanos , Listeria monocytogenes/química , Proteínas de Microfilamentos/análisis , Salmonella enterica/química , Calponinas
6.
J Bacteriol ; 200(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29084860

RESUMEN

The ß-barrel assembly machinery (BAM) complex is the core machinery for the assembly of ß-barrel membrane proteins, and inhibition of BAM complex activity is lethal to bacteria. Discovery of integral membrane proteins that are key to pathogenesis and yet do not require assistance from the BAM complex raises the question of how these proteins assemble into bacterial outer membranes. Here, we address this question through a structural analysis of the type 2 secretion system (T2SS) secretin from enteropathogenic Escherichia coli O127:H6 strain E2348/69. Long ß-strands assemble into a barrel extending 17 Å through and beyond the outer membrane, adding insight to how these extensive ß-strands are assembled into the E. coli outer membrane. The substrate docking chamber of this secretin is shown to be sufficient to accommodate the substrate mucinase SteC.IMPORTANCE In order to cause disease, bacterial pathogens inhibit immune responses and induce pathology that will favor their replication and dissemination. In Gram-negative bacteria, these key attributes of pathogenesis depend on structures assembled into or onto the outer membrane. One of these is the T2SS. The Vibrio-type T2SS mediates cholera toxin secretion in Vibrio cholerae, and in Escherichia coli O127:H6 strain E2348/69, the same machinery mediates secretion of the mucinases that enable the pathogen to penetrate intestinal mucus and thereby establish deadly infections.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Escherichia coli Enteropatógena/química , Secretina/química , Sistemas de Secreción Tipo II/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli Enteropatógena/metabolismo , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Lipoproteínas/química , Microscopía Electrónica/métodos , Modelos Moleculares , Polisacárido Liasas/metabolismo , Unión Proteica , Conformación Proteica , Sistemas de Translocación de Proteínas/química , Sistemas de Translocación de Proteínas/metabolismo , Transporte de Proteínas , Secretina/genética , Secretina/aislamiento & purificación , Sistemas de Secreción Tipo II/metabolismo , Vibrio cholerae/química , Vibrio cholerae/metabolismo
7.
Biomedica ; 36(0): 118-27, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-27622632

RESUMEN

INTRODUCTION: Diarrheagenic Escherichia coli is an important causative agent of acute diarrheic syndrome.  OBJECTIVE: To identify clonal groups of enteropathogenic E. coli (EPEC), in 485 children with acute diarrhea aged 0 to 10 years attending health care centers in Arismendi, Benítez and Sucre municipalities, Sucre state, Venezuela, from March to December, 2011.  MATERIALS AND METHODS: After obtaining the informed consent, stool samples were collected. Escherichia coli was identified using standard coproculture methods and serology with polyvalent and monovalent antisera. DNA was isolated, and eae (intimin) and bfpA (bundlin) genes were amplified through two multiplex polymerase chain reactions (PCR).  RESULTS: The presence of bacterial infection was determined in 39.6% of coprocultures. The prevalence of E. coli was 54.7%; 82.9% of these isolates were positive by serology for the evaluated serogroups and serotypes, which were mostly identified in children between 0 and 2 years (37.9%); 48.6% of E. coli strains amplified the eae gene; of these, 58.8% were classified as typical EPEC (eae+ y bfp+). EPEC II was the most common serogroup (38.7%), with predominance of typical EPEC (60%). In positive strains for eae gene, the ß intimin allele was the most frequently identified (74.5%). Only four strains with O157:H7 serotype were identified, which showed no PCR amplification of the eae and bfpA genes.  CONCLUSION: This study showed the importance of molecular tests to identify diarrheagenic E. coli strains causing clinical conditions of varying severity.


Asunto(s)
Diarrea/epidemiología , Escherichia coli Enteropatógena/aislamiento & purificación , Infecciones por Escherichia coli/epidemiología , Escherichia coli/aislamiento & purificación , Reacción en Cadena de la Polimerasa/métodos , Niño , Escherichia coli Enteropatógena/química , Escherichia coli/química , Humanos , Venezuela
8.
J Microbiol ; 54(8): 559-64, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27480636

RESUMEN

Enteropathogenic E. coli causes attaching and effacing (A/E) intestinal lesions. The genes involved in the formation of A/E lesions are encoded within a chromosomal island comprising of five major operons, LEE1-5. The global regulator H-NS represses the expression of these operons. Ler, a H-NS homologue, counteracts the H-NS-mediated repression. Using a novel genetic approach, we identified the amino acid residues in Ler that are involved in the interaction with H-NS: I20 and L23 in the C-terminal portion of α-helix 3, and I42 in the following unstructured linker region.


Asunto(s)
Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Operón , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Transactivadores/química , Transactivadores/metabolismo , Secuencias de Aminoácidos , Regulación hacia Abajo , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/metabolismo , Fosfoproteínas/metabolismo , Transactivadores/genética
9.
J Biol Chem ; 291(11): 5803-5816, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26786100

RESUMEN

Attaching and effacing Escherichia coli cause diarrhea and typically produce lymphostatin (LifA), an inhibitor of mitogen-activated proliferation of lymphocytes and pro-inflammatory cytokine synthesis. A near-identical factor (Efa1) has been reported to mediate adherence of E. coli to epithelial cells. An amino-terminal region of LifA shares homology with the catalytic domain of the large clostridial toxins, which are retaining glycosyltransferases with a DXD motif involved in binding of a metal ion. Understanding the mode(s) of action of lymphostatin has been constrained by difficulties obtaining a stably transformed plasmid expression clone. We constructed a tightly inducible clone of enteropathogenic E. coli O127:H6 lifA for affinity purification of lymphostatin. The purified protein inhibited mitogen-activated proliferation of bovine T lymphocytes in the femtomolar range. It is a monomer in solution and the molecular envelope was determined using both transmission electron microscopy and small-angle x-ray scattering. Domain architecture was further studied by limited proteolysis. The largest proteolytic fragment containing the putative glycosyltransferase domain was tested in isolation for activity against T cells, and was not sufficient for activity. Tryptophan fluorescence studies indicated thatlymphostatin binds uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) but not UDP-glucose (UDP-Glc). Substitution of the predicted DXD glycosyltransferase motif with alanine residues abolished UDP-GlcNAc binding and lymphostatin activity, although other biophysical properties were unchanged. The data indicate that lymphostatin has UDP-sugar binding potential that is critical for activity, and is a major leap toward identifying the nature and consequences of modifications of host cell factors.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Enfermedades de los Bovinos/inmunología , Escherichia coli Enteropatógena/inmunología , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/inmunología , Linfocitos T/microbiología , Secuencia de Aminoácidos , Animales , Bovinos , Enfermedades de los Bovinos/microbiología , Escherichia coli Enteropatógena/química , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Glicosiltransferasas/química , Glicosiltransferasas/inmunología , Humanos , Activación de Linfocitos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Linfocitos T/inmunología , Factores de Virulencia/inmunología , Difracción de Rayos X
10.
Glycobiology ; 25(8): 845-54, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25941008

RESUMEN

The affinities of the most abundant oligosaccharides found in human milk for four bacterial exotoxins (from Vibrio cholerae and pathogenic Escherichia coli) were quantified for the first time. Association constants (Ka) for a library of 20 human milk oligosaccharides (HMOs) binding to Shiga toxin type 2 holotoxin (Stx2) and the B subunit homopentamers of cholera toxin, heat-labile toxin and Shiga toxin type 1 (CTB5, HLTB5 and Stx1B5) were measured at 25°C and pH 7 using the direct electrospray ionization mass spectrometry assay. Notably, all four bacterial toxins bind to a majority of the HMOs tested and five of the HMOs (2'-fucosyllactose, lacto-N-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II and lacto-N-fucopentaose III) are ligands for all four toxins. These five HMOs are also reported to bind to other bacterial toxins (e.g. toxin A and toxin B of Clostridium difficile). In all cases, the HMO affinities (apparent Ka) are relatively modest (≤15,000 M(-1)). However, at the high concentrations of HMOs typically ingested by infants, a significant fraction of these toxins, if present, is expected to be bound to HMOs. Binding measurements carried out with 2'-fucosyllactose or lacto-N-fucopentaose I, together with a high-affinity ligand based on the native carbohydrate receptor, revealed that all four toxins possess HMO-binding sites that are distinct from those of the native receptors, although evidence of competitive binding was found for lacto-N-fucopentaose I with Stx2 and 2'-fucosyllactose and lacto-N-fucopentaose I with HLTB5. Taken together, the results of this study suggest that, while HMOs are expected to bind extensively to these bacterial toxins, it is unlikely that HMO binding will effectively inhibit their interactions with their cellular receptors.


Asunto(s)
Clostridioides difficile/química , Escherichia coli Enteropatógena/química , Leche Humana/química , Vibrio cholerae/química , Amino Azúcares/química , Amino Azúcares/aislamiento & purificación , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Toxinas Bacterianas/química , Toxinas Bacterianas/aislamiento & purificación , Sitios de Unión , Secuencia de Carbohidratos , Toxina del Cólera/química , Toxina del Cólera/aislamiento & purificación , Enterotoxinas/química , Enterotoxinas/aislamiento & purificación , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Oligosacáridos/química , Oligosacáridos/aislamiento & purificación , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Unión Proteica , Toxina Shiga I/química , Toxina Shiga I/aislamiento & purificación , Toxina Shiga II/química , Toxina Shiga II/aislamiento & purificación , Espectrometría de Masa por Ionización de Electrospray , Trisacáridos/química , Trisacáridos/aislamiento & purificación
11.
mBio ; 6(2)2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25805731

RESUMEN

UNLABELLED: Type IV pili (T4Ps) are surface appendages used by Gram-negative and Gram-positive pathogens for motility and attachment to epithelial surfaces. In Gram-negative bacteria, such as the important pediatric pathogen enteropathogenic Escherichia coli (EPEC), during extension and retraction, the pilus passes through an outer membrane (OM) pore formed by the multimeric secretin complex. The secretin is common to Gram-negative assemblies, including the related type 2 secretion (T2S) system and the type 3 secretion (T3S) system. The N termini of the secretin monomers are periplasmic and in some systems have been shown to mediate substrate specificity. In this study, we mapped the topology of BfpB, the T4P secretin from EPEC, using a combination of biochemical and biophysical techniques that allowed selective identification of periplasmic and extracellular residues. We applied rules based on solved atomic structures of outer membrane proteins (OMPs) to generate our topology model, combining the experimental results with secondary structure prediction algorithms and direct inspection of the primary sequence. Surprisingly, the C terminus of BfpB is extracellular, a result confirmed by flow cytometry for BfpB and a distantly related T4P secretin, PilQ, from Pseudomonas aeruginosa. Keeping with prior evidence, the C termini of two T2S secretins and one T3S secretin were not detected on the extracellular surface. On the basis of our data and structural constraints, we propose that BfpB forms a beta barrel with 16 transmembrane beta strands. We propose that the T4P secretins have a C-terminal segment that passes through the center of each monomer. IMPORTANCE: Secretins are multimeric proteins that allow the passage of secreted toxins and surface structures through the outer membranes (OMs) of Gram-negative bacteria. To date, there have been no atomic structures of the C-terminal region of a secretin, although electron microscopy (EM) structures of the complex are available. This work provides a detailed topology prediction of the membrane-spanning domain of a type IV pilus (T4P) secretin. Our study used innovative techniques to provide new and comprehensive information on secretin topology, highlighting similarities and differences among secretin subfamilies. Additionally, the techniques used in this study may prove useful for the study of other OM proteins.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Escherichia coli Enteropatógena/química , Proteínas de Escherichia coli/química , Lipoproteínas/química , Sistemas de Secreción Tipo IV/química , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/genética , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Citometría de Flujo , Lipoproteínas/genética , Modelos Moleculares , Conformación Proteica , Sistemas de Secreción Tipo IV/genética
12.
Mol Microbiol ; 95(1): 80-100, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25353290

RESUMEN

Intimin and Invasin are prototypical inverse (Type Ve) autotransporters and important virulence factors of enteropathogenic Escherichia coli and Yersinia spp. respectively. In addition to a C-terminal extracellular domain and a ß-barrel transmembrane domain, both proteins also contain a short N-terminal periplasmic domain that, in Intimin, includes a lysin motif (LysM), which is thought to mediate binding to peptidoglycan. We show that the periplasmic domain of Intimin does bind to peptidoglycan both in vitro and in vivo, but only under acidic conditions. We were able to determine a dissociation constant of 0.8 µM for this interaction, whereas the Invasin periplasmic domain, which lacks a LysM, bound only weakly in vitro and failed to bind peptidoglycan in vivo. We present the solution structure of the Intimin LysM, which has an additional α-helix conserved within inverse autotransporter LysMs but lacking in others. In contrast to previous reports, we demonstrate that the periplasmic domain of Intimin mediates dimerisation. We further show that dimerisation and peptidoglycan binding are general features of LysM-containing inverse autotransporters. Peptidoglycan binding by the periplasmic domain in the infection process may aid in resisting mechanical and chemical stress during transit through the gastrointestinal tract.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Escherichia coli Enteropatógena/metabolismo , Peptidoglicano/metabolismo , Yersinia/metabolismo , Adhesinas Bacterianas/genética , Sitios de Unión , Biología Computacional/métodos , Dimerización , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Concentración de Iones de Hidrógeno , Modelos Moleculares , Multimerización de Proteína , Estructura Secundaria de Proteína , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Yersinia/química , Yersinia/genética
13.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 40-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24419377

RESUMEN

NleC is one of the virulence factors that is injected into infected host cells by enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC) via a needle-like protein complex called the type III secretion system (T3SS). The cytosolic NleC specifically cleaves the p65 subunit of NF-κB in the p65-p50 heterodimeric complex just after the Cys38 site in its N-terminal domain. The degradation of the remainder of the p65 C-terminal domain by the proteasome disrupts the NF-κB signalling pathway, thus dampening the host inflammatory response. Here, the crystal structure of NleC is reported at 1.55 Šresolution. In conjunction with biochemical analyses, the structure reveals that NleC is a member of the zincin zinc protease family and that the configuration of the NleC active site resembles that of the metzincin clan of metallopeptidases but without the canonical Met turn of astacin. The extended zinc-binding motif of NleC (HEXXHXXTXXXD) includes three metal ligands. The fifth zinc ligand, a conserved tyrosine (a bound water molecule is the fourth ligand), lies 45 residues downstream of the zincin motif. Furthermore, the electrostatic potential complementarity between NleC and p65 also contributes to the cleavage activity of the protease. These results not only provide important insights into the mechanism of how NleC recognizes its substrates, but also shed light on the design of new antibiotics for the food-borne diseases arising from EPEC and EHEC.


Asunto(s)
Escherichia coli Enteropatógena/química , Proteínas de Escherichia coli/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Conformación Proteica , Zinc/metabolismo
14.
J Bacteriol ; 195(11): 2481-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23524615

RESUMEN

Type III secretion systems (T3SSs) are central virulence mechanisms used by a variety of Gram-negative bacteria to inject effector proteins into host cells. The needle polymer is an essential part of the T3SS that provides the effector proteins a continuous channel into the host cytoplasm. It has been shown for a few T3SSs that two chaperones stabilize the needle protein within the bacterial cytosol to prevent its premature polymerization. In this study, we characterized the chaperones of the enteropathogenic Escherichia coli (EPEC) needle protein EscF. We found that Orf2 and Orf29, two poorly characterized proteins encoded within the EPEC locus of enterocyte effacement (LEE), function as the needle protein cochaperones. Our finding demonstrated that both Orf2 and Orf29 are essential for type III secretion (T3S). In addition, we found that Orf2 and Orf29 associate with the bacterial membrane and form a complex with EscF. Orf2 and Orf29 were also shown to disrupt the polymerization of EscF in vitro. Prediction of the tertiary structures of Orf2 and Orf29 showed high structural homology to chaperones of other T3SS needle proteins. Overall, our data suggest that Orf2 and Orf29 function as the chaperones of the needle protein, and therefore, they have been renamed EscE and EscG.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/aislamiento & purificación , Proteínas del Citoesqueleto/metabolismo , Enterocitos/metabolismo , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/aislamiento & purificación , Expresión Génica , Regulación Bacteriana de la Expresión Génica , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/aislamiento & purificación , Datos de Secuencia Molecular , Complejos Multiproteicos , Mutación , Fosfoproteínas/genética , Multimerización de Proteína , Transporte de Proteínas , Proteínas Recombinantes , Alineación de Secuencia
15.
J Bacteriol ; 194(11): 2819-28, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22447907

RESUMEN

The virulence of many Gram-negative pathogens is associated with type III secretion systems (T3SSs), which deliver virulence effector proteins into the cytoplasm of host cells. Components of enteropathogenic Escherichia coli (EPEC) T3SS are encoded within the locus of enterocyte effacement (LEE). While most LEE-encoded T3SS proteins in EPEC have assigned names and functions, a few of them remain poorly characterized. Here, we studied a small LEE-encoded protein, Orf15, that shows no homology to other T3SS/flagellar proteins and is only present in attaching and effacing pathogens, including enterohemorrhagic E. coli and Citrobacter rodentium. Our findings demonstrated that it is essential for type III secretion (T3S) and that it is localized to the periplasm and associated with the inner membrane. Membrane association was driven by the N-terminal 19 amino acid residues, which were also shown to be essential for T3S. Consistent with its localization, Orf15 was found to interact with the EPEC T3SS outer membrane ring component, EscC, which was previously shown to be embedded within the outer membrane and protruding into the periplasmic space. Interestingly, we found that the predicted coiled-coil structure of Orf15 is critical for the protein's function. Overall, our findings suggest that Orf15 is a structural protein that contributes to the structural integrity of the T3S complex, and therefore we propose to rename it EscA.


Asunto(s)
Sistemas de Secreción Bacterianos , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Secuencia de Aminoácidos , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia
16.
Biochem J ; 442(1): 119-25, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22087554

RESUMEN

The T3SS (type III secretion system) is a multi-protein complex that plays a central role in the virulence of many gram-negative bacterial pathogens. This apparatus spans both bacterial membranes and transports virulence factors from the bacterial cytoplasm into eukaryotic host cells. The T3SS exports substrates in a hierarchical and temporal manner. The first secreted substrates are the rod/needle proteins which are incorporated into the T3SS apparatus and are required for the secretion of later substrates, the translocators and effectors. In the present study, we provide evidence that rOrf8/EscI, a poorly characterized locus of enterocyte effacement-encoded protein, functions as the inner rod protein of the T3SS of EPEC (enteropathogenic Escherichia coli). We demonstrate that EscI is essential for type III secretion and is also secreted as an early substrate of the T3SS. We found that EscI interacts with EscU, the integral membrane protein that is linked to substrate specificity switching, implicating EscI in the substrate-switching event. Furthermore, we showed that EscI self-associates and interacts with the outer membrane secretin EscC, further supporting its function as an inner rod protein. Overall, the results of the present study suggest that EscI is the YscI/PrgJ/MxiI homologue in the T3SS of attaching and effacing pathogens.


Asunto(s)
Escherichia coli Enteropatógena/química , Proteínas de Escherichia coli/metabolismo , Factores de Virulencia/metabolismo , Proteínas de Escherichia coli/química
17.
Appl Environ Microbiol ; 77(14): 4949-58, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21642413

RESUMEN

A national survey of Escherichia coli O26 in Norwegian sheep flocks was conducted, using fecal samples to determine the prevalence. In total, 491 flocks were tested, and E. coli O26 was detected in 17.9% of the flocks. One hundred forty-two E. coli O26 isolates were examined for flagellar antigens (H typing) and four virulence genes, including stx and eae, to identify possible Shiga toxin-producing E. coli (STEC) and enteropathogenic E. coli (EPEC). Most isolates (129 out of 142) were identified as E. coli O26:H11. They possessed eae and may have potential as human pathogens, although only a small fraction were identified as STEC O26:H11, giving a prevalence in sheep flocks of only 0.8%. Correspondingly, the sheep flock prevalence of atypical EPEC (aEPEC) O26:H11 was surprisingly high (15.9%). The genetic relationship between the E. coli O26:H11 isolates was investigated by pulsed-field gel electrophoresis (PFGE) and multilocus variable number tandem repeat analysis (MLVA), identifying 63 distinct PFGE profiles and 22 MLVA profiles. Although the MLVA protocol was less discriminatory than PFGE and a few cases of disagreement were observed, comparison by partition mapping showed an overall good accordance between the two methods. A close relationship between a few isolates of aEPEC O26:H11 and STEC O26:H11 was identified, but all the E. coli O26:H11 isolates should be considered potentially pathogenic to humans. The present study consisted of a representative sampling of sheep flocks from all parts of Norway. This is the first large survey of sheep flocks focusing on E. coli O26 in general, including results of STEC, aEPEC, and nonpathogenic isolates.


Asunto(s)
Antígenos Bacterianos/análisis , Escherichia coli Enteropatógena/aislamiento & purificación , Infecciones por Escherichia coli/veterinaria , Antígenos O/análisis , Enfermedades de las Ovejas/epidemiología , Ovinos/microbiología , Animales , Antígenos Bacterianos/inmunología , Técnicas de Tipificación Bacteriana , Electroforesis en Gel de Campo Pulsado , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/clasificación , Infecciones por Escherichia coli/epidemiología , Proteínas de Escherichia coli , Heces/microbiología , Tipificación de Secuencias Multilocus , Noruega/epidemiología , Antígenos O/inmunología , Serotipificación , Toxina Shiga/biosíntesis , Factores de Virulencia
18.
Biochemistry ; 50(24): 5465-76, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21449614

RESUMEN

We report the 1.9 Å resolution crystal structure of enteropathogenic Escherichia coli GfcC, a periplasmic protein encoded by the gfc operon, which is essential for assembly of group 4 polysaccharide capsule (O-antigen capsule). Presumed gene orthologs of gfcC are present in capsule-encoding regions of at least 29 genera of Gram-negative bacteria. GfcC, a member of the DUF1017 family, is comprised of tandem ß-grasp (ubiquitin-like) domains (D2 and D3) and a carboxyl-terminal amphipathic helix, a domain arrangement reminiscent of that of Wza that forms an exit pore for group 1 capsule export. Unlike the membrane-spanning C-terminal helix from Wza, the GfcC C-terminal helix packs against D3. Previously unobserved in a ß-grasp domain structure is a 48-residue helical hairpin insert in D2 that binds to D3, constraining its position and sequestering the carboxyl-terminal amphipathic helix. A centrally located and invariant Arg115 not only is essential for proper localization but also forms one of two mostly conserved pockets. Finally, we draw analogies between a GfcC protein fused to an outer membrane ß-barrel pore in some species and fusion proteins necessary for secreting biofilm-forming exopolysaccharides.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Secuencia de Aminoácidos , Cápsulas Bacterianas/química , Proteínas de la Membrana Bacteriana Externa/genética , Secuencia de Bases , Secuencia Conservada , Cristalografía por Rayos X , ADN Bacteriano/genética , Dimerización , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos , Modelos Moleculares , Datos de Secuencia Molecular , Operón , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido , Electricidad Estática
19.
J Biol Chem ; 285(42): 32336-42, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20688909

RESUMEN

We present a body of ultrastructural, biochemical, and genetic evidence that demonstrates the oligomerization of virulence-associated autotransporter proteins EspC or EspP produced by deadly human pathogens enterohemorrhagic and enteropathogenic Escherichia coli into novel macroscopic rope-like structures (>1 cm long). The rope-like structures showed high aggregation and insolubility, stability to anionic detergents and high temperature, and binding to Congo Red and thioflavin T dyes. These are properties also exhibited by human amyloidogenic proteins. These macroscopic ropes were not observed in cultures of nonpathogenic Escherichia coli or isogenic espP or espC deletion mutants of enterohemorrhagic or enteropathogenic Escherichia coli but were produced by an Escherichia coli K-12 strain carrying a plasmid expressing espP. Purified recombinant EspP monomers were able to self-assemble into macroscopic ropes upon incubation, suggesting that no other protein was required for assembly. The ropes bound to and showed cytopathic effects on cultured epithelial cells, served as a substratum for bacterial adherence and biofilm formation, and protected bacteria from antimicrobial compounds. We hypothesize that these ropes play a biologically significant role in the survival and pathogenic scheme of these organisms.


Asunto(s)
Adhesión Bacteriana , Escherichia coli Enterohemorrágica , Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Serina Endopeptidasas , Animales , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli Enterohemorrágica/química , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestructura , Células HeLa , Humanos , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Serina Endopeptidasas/ultraestructura
20.
Can J Microbiol ; 56(5): 366-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20555398

RESUMEN

The structure of the antigenic O-polysaccharide isolated from the lipopolysaccharide produced by enterohemorrhagic Escherichia coli O103:H2 was determined and shown to be composed of d-glucose (1 part), 2-acetamido-2-deoxy-d-glucose (2 parts), 2-acetamido-2-deoxy-d-galactose (1 part), and 3-deoxy-3-(R)-3-hydroxybutyramido-d-fucose (1 part). From the results of methylation analysis, Smith-type periodate oxidation degradation studies, and the use of one- and two-dimensional (1)H and (13)C NMR spectroscopy, the O-polysaccharide antigen was found to be an unbranched polymer of a repeating pentasaccharide unit having the following structure: -->2)-Beta-d-Glcp-(1-->2)-Beta-d-Fucp3NBu-(1-->6)-alpha-d-GlcpNAc-(1-->4)-alpha-d-GalpNAc-(1-->3)-Beta-d-GlcpNAc-(1-->,where Bu is (R)-3-hydroxybutyramido.


Asunto(s)
Escherichia coli Enteropatógena/química , Antígenos O/química , Secuencia de Carbohidratos , Lipopolisacáridos/química , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...