Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 87(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31209149

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) has at least three enzymes, NorV, Hmp, and Hcp, that act independently to lower the toxicity of nitric oxide (NO), a potent antimicrobial molecule. This study aimed to reveal the cooperative roles of these defensive enzymes in EHEC against nitrosative stress. Under anaerobic conditions, combined deletion of all three enzymes significantly increased the NO sensitivity of EHEC determined by the growth at late stationary phase; however, the expression of norV restored the NO resistance of EHEC. On the other hand, the growth of Δhmp mutant EHEC was inhibited after early stationary phase, indicating that NorV and Hmp play a cooperative role in anaerobic growth. Under microaerobic conditions, the growth of Δhmp mutant EHEC was inhibited by NO, indicating that Hmp is the enzyme that protects cells from NO stress under microaerobic conditions. When EHEC cells were exposed to a lower concentration of NO, the NO level in bacterial cells of Δhcp mutant EHEC was higher than those of the other EHEC mutants, suggesting that Hcp is effective at regulating NO levels only at a low concentration. These findings of a low level of NO in bacterial cells with hcp indicate that the NO consumption activity of Hcp was suppressed by Hmp at a low range of NO concentrations. Taken together, these results show that the cooperative effects of NO-metabolizing enzymes are regulated by the range of NO concentrations to which the EHEC cells are exposed.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Óxido Nítrico/metabolismo , Estrés Nitrosativo/fisiología , Anaerobiosis , Escherichia coli Enterohemorrágica/metabolismo , Regulación Bacteriana de la Expresión Génica , Oxidorreductasas/metabolismo
2.
J Med Microbiol ; 67(5): 724-732, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29620505

RESUMEN

PURPOSE: In enterohaemorrhagic Escherichia coli (EHEC), stx1 or stx2 genes encode Shiga toxin (Stx1 or Stx2, respectively) and are carried by prophages. The production and release of both stx phages and toxin occur upon initiation of the phage lytic cycle. Phages can further disseminate stx genes by infecting naïve bacteria in the intestine. Here, the effect of RNase E deficiency on these two virulence traits was investigated. METHODOLOGY: Cultures of the EHEC strains TEA028-rne containing low versus normal RNase E levels or the parental strain (TEA028) were treated with mitomycin C (MMC) to induce the phage lytic cycle. Phages and Stx2 titres were quantified by the double-agar assay and the receptor ELISA technique, respectively. RESULTS: RNase E deficiency in MMC-treated cells significantly reduced the yield of infectious stx2 phages. Delayed cell lysis and the appearance of encapsidated phage DNA copies suggest a slow onset of the lytic cycle. However, these observations do not entirely explain the decrease of phage yields. stx1 phages were not detected under normal or deficient RNase E levels. After an initial delay, high levels of toxin were finally produced in MMC-treated cultures. CONCLUSION: RNase E scarcity reduces stx2 phage production but not toxin. Normal concentrations of RNase E are likely required for correct phage morphogenesis. Our future work will address the mechanism of RNase E action on phage morphogenesis.


Asunto(s)
Colifagos/crecimiento & desarrollo , Endorribonucleasas/metabolismo , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/virología , Profagos/crecimiento & desarrollo , Toxina Shiga II/biosíntesis , Bacteriólisis , Colifagos/genética , Endorribonucleasas/deficiencia , Ensayo de Inmunoadsorción Enzimática , Humanos , Profagos/genética , Toxina Shiga II/análisis , Ensayo de Placa Viral
3.
SLAS Discov ; 23(8): 815-822, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29630847

RESUMEN

Acetaldehyde dehydrogenase (AdhE) is a bifunctional acetaldehyde-coenzyme A (CoA) dehydrogenase and alcohol dehydrogenase involved in anaerobic metabolism in gram-negative bacteria. This enzyme was recently found to be a key regulator of the type three secretion (T3S) system in Escherichia coli. AdhE inhibitors can be used as tools to study bacterial virulence and a starting point for discovery of novel antibacterial agents. We developed a robust enzymatic assay, based on the acetaldehyde-CoA dehydrogenase activity of AdhE using both absorption and fluorescence detection models (Z' > 0.7). This assay was used to screen ~11,000 small molecules in 384-well format that resulted in three hits that were confirmed by resynthesis and validation. All three compounds are noncompetitive with respect to acetaldehyde and display a clear dose-response effect with hill slopes of 1-2. These new inhibitors will be used as chemical tools to study the interplay between metabolism and virulence and the role of AdhE in T3S regulation in gram-negative bacteria, and as starting points for the development of novel antibacterial agents.


Asunto(s)
Alcohol Deshidrogenasa/antagonistas & inhibidores , Aldehído Oxidorreductasas/antagonistas & inhibidores , Antibacterianos/farmacología , Evaluación Preclínica de Medicamentos , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/enzimología , Inhibidores Enzimáticos/farmacología , Proteínas de Escherichia coli/antagonistas & inhibidores , Alcohol Deshidrogenasa/genética , Alcohol Deshidrogenasa/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Antibacterianos/química , Línea Celular , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Escherichia coli Enterohemorrágica/genética , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Ratones , Flujo de Trabajo
4.
J Biol Chem ; 292(27): 11423-11430, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28522607

RESUMEN

Many Gram-negative bacterial pathogens use a syringe-like apparatus called a type III secretion system to inject virulence factors into host cells. Some of these effectors are enzymes that modify host proteins to subvert their normal functions. NleB is a glycosyltransferase that modifies host proteins with N-acetyl-d-glucosamine to inhibit antibacterial and inflammatory host responses. NleB is conserved among the attaching/effacing pathogens enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), and Citrobacter rodentium Moreover, Salmonella enterica strains encode up to three NleB orthologs named SseK1, SseK2, and SseK3. However, there are conflicting reports regarding the activities and host protein targets among the NleB/SseK orthologs. Therefore, here we performed in vitro glycosylation assays and cell culture experiments to compare the activities and substrate specificities of these effectors. SseK1, SseK3, EHEC NleB1, EPEC NleB1, and Crodentium NleB blocked TNF-mediated NF-κB pathway activation, whereas SseK2 and NleB2 did not. C. rodentium NleB, EHEC NleB1, and SseK1 glycosylated host GAPDH. C. rodentium NleB, EHEC NleB1, EPEC NleB1, and SseK2 glycosylated the FADD (Fas-associated death domain protein). SseK3 and NleB2 were not active against either substrate. We also found that EHEC NleB1 glycosylated two GAPDH arginine residues, Arg197 and Arg200, and that these two residues were essential for GAPDH-mediated activation of TNF receptor-associated factor 2 ubiquitination. These results provide evidence that members of this highly conserved family of bacterial virulence effectors target different host protein substrates and exhibit distinct cellular modes of action to suppress host responses.


Asunto(s)
Proteínas Bacterianas/metabolismo , Citrobacter rodentium/enzimología , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enteropatógena/enzimología , Proteínas de Escherichia coli/metabolismo , Salmonella enterica/enzimología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Glicosilación , Ratones , Células RAW 264.7 , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Salmonella enterica/genética , Salmonella enterica/patogenicidad , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitinación , Factores de Virulencia/genética
5.
J Bacteriol ; 198(24): 3318-3328, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27698085

RESUMEN

We recently discovered that exposure of enterohemorrhagic Escherichia coli (EHEC) to d-serine resulted in accumulation of this unusual amino acid, induction of the SOS regulon, and downregulation of the type III secretion system that is essential for efficient colonization of the host. Here, we have investigated the physiological relevance of this elevated SOS response, which is of particular interest given the presence of Stx toxin-carrying lysogenic prophages on the EHEC chromosome that are activated during the SOS response. We found that RecA elevation in response to d-serine, while being significant, was heterogeneous and not capable of activating stx expression or stx phage transduction to a nonlysogenic recipient. This "SOS-like response" was, however, capable of increasing the mutation frequency associated with low-level RecA activity, thus promoting genetic diversity. Furthermore, this response was entirely dependent on RecA and enhanced in the presence of a DNA-damaging agent, indicating a functional SOS response, but did not result in observable cleavage of the LexA repressor alone, indicating a controlled mechanism of induction. This work demonstrates that environmental factors not usually associated with DNA damage are capable of promoting an SOS-like response. We propose that this modulated induction of RecA allows EHEC to adapt to environmental insults such as d-serine while avoiding unwanted phage-induced lysis. IMPORTANCE: The SOS response is a global stress network that is triggered by the presence of DNA damage due to breakage or stalled replication forks. Activation of the SOS response can trigger the replication of lytic bacteriophages and promote genetic diversification through error-prone polymerases. We have demonstrated that the host-associated metabolite d-serine contributes to Escherichia coli niche specification and accumulates inside cells that cannot catabolize it. This results in a modulated activation of the SOS antirepressor RecA that is insufficient to trigger lytic bacteriophage but capable of increasing the SOS-associated mutation frequency. These findings describe how relevant signals not normally associated with DNA damage can hijack the SOS response, promoting diversity as E. coli strains adapt while avoiding unwanted phage lysis.


Asunto(s)
Escherichia coli Enterohemorrágica/metabolismo , Proteínas de Escherichia coli/metabolismo , Variación Genética , Rec A Recombinasas/metabolismo , Serina/metabolismo , Bacteriófagos/fisiología , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/virología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Rec A Recombinasas/genética , Respuesta SOS en Genética
6.
Protein Expr Purif ; 126: 122-126, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371359

RESUMEN

In enterohemorrhagic Escherichia coli (EHEC), the QseEF two-component system causes attaching and effacing (AE) lesion on epithelial cells. QseE histidine kinase senses the host hormone epinephrine, sulfate, and phosphate; it also regulates QseF response regulator, which activates LEE gene that encodes AE lesion. In order to understand the recognition of ligand molecules and signal transfer mechanism in pathogenic bacteria, structural studies of the sensor domain of QseE of Escherichia coli should be conducted. In this study, we describe the overexpression, purification, and structural and biophysical properties of the sensor domain of QseE. The fusion protein had a 6×His tag at its N-terminus; this protein was overexpressed as inclusion bodies in E. coli BL21 (DE3). The protein was denatured in 7M guanidine hydrochloride and refolded by dialysis. The purification of the refolded protein was carried out using Ni-NTA affinity column and size-exclusion chromatography. Thereafter, the characteristics of the refolded protein were determined from NMR, CD, and MALS spectroscopies. In a pH range of 7.4-5.0, the folded protein existed in a monomeric form with a predominantly helical structure. (1)H-(15)N HSQC NMR spectra shows that approximately 93% backbone amide peaks are detected at pH 5.0, suggesting that the number of backbone signals is sufficient for NMR studies. These data might provide an opportunity for structural and functional studies of the sensor domain of QseE.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli , Expresión Génica , Replegamiento Proteico , Receptores Adrenérgicos , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/aislamiento & purificación , Concentración de Iones de Hidrógeno , Resonancia Magnética Nuclear Biomolecular , Dominios Proteicos , Estructura Secundaria de Proteína , Receptores Adrenérgicos/biosíntesis , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación
7.
J Bacteriol ; 196(17): 3122-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24957618

RESUMEN

Escherichia coli displays O antigens on the outer membrane that play an important role in bacterial interactions with the environment. The O antigens of enterohemorrhagic E. coli O104 and O5 contain a Galß1-3GalNAc disaccharide at the reducing end of the repeating unit. Several other O antigens contain this disaccharide, which is identical to the mammalian O-glycan core 1 or the cancer-associated Thomsen-Friedenreich (TF) antigen. We identified the wbwC genes responsible for the synthesis of the disaccharide in E. coli serotypes O104 and O5. To functionally characterize WbwC, an acceptor substrate analog, GalNAcα-diphosphate-phenylundecyl, was synthesized. WbwC reaction products were isolated by high-pressure liquid chromatography and analyzed by mass spectrometry, nuclear magnetic resonance, galactosidase and O-glycanase digestion, and anti-TF antibody. The results clearly showed that the Galß1-3GalNAcα linkage was synthesized, confirming WbwCECO104 and WbwCECO5 as UDP-Gal:GalNAcα-diphosphate-lipid ß1,3-Gal-transferases. Sequence analysis revealed a conserved DxDD motif, and mutagenesis showed the importance of these Asp residues in catalysis. The purified enzymes require divalent cations (Mn(2+)) for activity and are specific for UDP-Gal and GalNAc-diphosphate lipid substrates. WbwC was inhibited by bis-imidazolium salts having aliphatic chains of 18 to 22 carbons. This work will help to elucidate mechanisms of polysaccharide synthesis in pathogenic bacteria and provide technology for vaccine synthesis.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Galactosiltransferasas/metabolismo , Secuencia de Aminoácidos , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/metabolismo , Proteínas de Escherichia coli/genética , Galactosiltransferasas/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica , Serotipificación
9.
PLoS One ; 8(7): e69196, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874912

RESUMEN

Life-threatening intestinal and systemic effects of the Shiga toxins produced by enterohemorrhagic Escherichia coli (EHEC) require toxin uptake and transcytosis across intestinal epithelial cells. We have recently demonstrated that EHEC infection of intestinal epithelial cells stimulates toxin macropinocytosis, an actin-dependent endocytic pathway. Host actin rearrangement necessary for EHEC attachment to enterocytes is mediated by the type 3 secretion system which functions as a molecular syringe to translocate bacterial effector proteins directly into host cells. Actin-dependent EHEC attachment also requires the outer membrane protein intimin, a major EHEC adhesin. Here, we investigate the role of type 3 secretion in actin turnover occurring during toxin macropinocytosis. Toxin macropinocytosis is independent of EHEC type 3 secretion and intimin attachment. EHEC soluble factors are sufficient to stimulate macropinocytosis and deliver toxin into enterocytes in vitro and in vivo; intact bacteria are not required. Intimin-negative enteroaggregative Escherichia coli (EAEC) O104:H4 robustly stimulate Shiga toxin macropinocytosis into intestinal epithelial cells. The apical macropinosomes formed in intestinal epithelial cells move through the cells and release their cargo at these cells' basolateral sides. Further analysis of EHEC secreted proteins shows that a serine protease EspP alone is able to stimulate host actin remodeling and toxin macropinocytosis. The observation that soluble factors, possibly serine proteases including EspP, from each of two genetically distinct toxin-producing strains, can stimulate Shiga toxin macropinocytosis and transcellular transcytosis alters current ideas concerning mechanisms whereby Shiga toxin interacts with human enterocytes. Mechanisms important for this macropinocytic pathway could suggest new potential therapeutic targets for Shiga toxin-induced disease.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Pinocitosis/fisiología , Serina Endopeptidasas/metabolismo , Toxina Shiga/metabolismo , Actinas/metabolismo , Animales , Sistemas de Secreción Bacterianos/fisiología , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Íleon/citología , Íleon/metabolismo , Íleon/ultraestructura , Mucosa Intestinal/ultraestructura , Ratones , Microscopía Electrónica de Transmisión
10.
Carbohydr Res ; 366: 17-24, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23261778

RESUMEN

The assembly of the repeating units of O-antigens in Gram negative bacteria is catalyzed by specific glycosyltransferases. Previously we used GlcNAc/GalNAcα-diphosphate-phenoxyundecyl as natural acceptor substrate analogs in assays of the transfer of radioactive sugars by bacterial glycosyltransferases. In order to develop new, fluorescence based assays we have synthesized a fluorescent acceptor P¹-[11-(anthracen-9-ylmethoxy)undecyl]-P²-(2-acetamido-2-deoxy-α-D-galactopyranosyl) diphosphate and have shown that the compound was an excellent acceptor for glucosyltransferase WbdN from Escherichia coli (E. coli) O157 and for galactosyltransferase WbwC from E. coli O5. This is the first report of the Gal-transferase activity of the wbwC gene product of E.coli O5. The presence of the fluorescent label in the acceptor molecule allows the detection of glycosyltransferase reaction products with high sensitivity, eliminating the need for radioactive nucleotide sugars.


Asunto(s)
Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli O157/metabolismo , Colorantes Fluorescentes/metabolismo , Glicosiltransferasas/metabolismo , Antígenos O/biosíntesis , Antígenos O/química , Biocatálisis , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli O157/enzimología , Colorantes Fluorescentes/química , Estructura Molecular , Estereoisomerismo
11.
Berl Munch Tierarztl Wochenschr ; 125(11-12): 469-75, 2012.
Artículo en Alemán | MEDLINE | ID: mdl-23227764

RESUMEN

In contrast to infections with enterohaemorrhagic E. coli (EHEC), which are thought to be classical zoonosis, the zoonotic potential of extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae is still widely unknown. The aim of our study was to determine the frequency of EHEC and ESBL-producing Enterobacteriaceae in domestic animals (dogs and cats) in the Tyrol. Among 228 fecal samples of dogs (n = 92) and cats (n = 136) three samples (1.3%) were positive in the EHEC-ELISA. In two of the three cases isolation of the organism was not possible, the third sample of a two-year-old crossbreed bitch yielded EHEC O103:H2. In twelve of 228 (5.3%) fecal samples 13 ESBL-producing Enterobacteriaceae (in ten cats and two dogs) were found.These animals mainly derived from homes for animals (ten animals, 83%). 75% of the isolates belonged to the CTX-M-1-group, 8% to the CTX-M-2-group and 17% to the CTX-M-9-group. One isolate was positive for CTX-M-1 and CTX-M-9. Typing of the 13 ESBL-producing isolates by multilocus sequence typing (MLST) showed ten different sequence types, which points out the importance of the horizontal transfer of mainly plasmid-coded ESBL genes. Transmission of EHEC and ESBL-producing Enterobacteriaceae from domestic animals to humans is possible, corroborated by the fact that the EHEC serotype found in one dog and the sequence types detected by MLST in several dogs and cats were previously reported to occur in severe human infection.


Asunto(s)
Enfermedades de los Gatos/microbiología , Enfermedades de los Perros/microbiología , Escherichia coli Enterohemorrágica/aislamiento & purificación , Infecciones por Escherichia coli/veterinaria , Zoonosis/microbiología , Animales , Antibacterianos/farmacología , Austria , Gatos , Perros , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Femenino , Humanos , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus
12.
Infect Immun ; 80(2): 483-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22144482

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC) are food-borne pathogens that cause serious diarrheal diseases. To colonize the human intestine, these pathogens must overcome innate immune defenses such as antimicrobial peptides (AMPs). Bacterial pathogens have evolved various mechanisms to resist killing by AMPs, including proteolytic degradation of AMPs. To examine the ability of the EHEC and EPEC OmpT outer membrane (OM) proteases to degrade α-helical AMPs, ompT deletion mutants were generated. Determination of MICs of various AMPs revealed that both mutant strains are more susceptible than their wild-type counterparts to α-helical AMPs, although to different extents. Time course assays monitoring the degradation of LL-37 and C18G showed that EHEC cells degraded both AMPs faster than EPEC cells in an OmpT-dependent manner. Mass spectrometry analyses of proteolytic fragments showed that EHEC OmpT cleaves LL-37 at dibasic sites. The superior protection provided by EHEC OmpT compared to EPEC OmpT against α-helical AMPs was due to higher expression of the ompT gene and, in turn, higher levels of the OmpT protein in EHEC. Fusion of the EPEC ompT promoter to the EHEC ompT open reading frame resulted in decreased OmpT expression, indicating that transcriptional regulation of ompT is different in EHEC and EPEC. We hypothesize that the different contributions of EHEC and EPEC OmpT to the degradation and inactivation of LL-37 may be due to their adaptation to their respective niches within the host, the colon and small intestine, respectively, where the environmental cues and abundance of AMPs are different.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enteropatógena/enzimología , Serina Endopeptidasas/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Farmacorresistencia Bacteriana , Transferencia Resonante de Energía de Fluorescencia , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Pruebas de Sensibilidad Microbiana , Regiones Promotoras Genéticas , Serina Endopeptidasas/genética , Catelicidinas
13.
Toxins (Basel) ; 5(1): 25-48, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23274272

RESUMEN

Enterohemorrhagic E. coli (EHEC) causes severe diseases in humans worldwide. One of its virulence factors is EspP, which belongs to the serine protease autotransporters of Enterobacteriaceae (SPATE) family. In this review we recapitulate the current data on prevalence, biogenesis, structural properties and functionality. EspP has been used to investigate mechanistic details of autotransport, and recent studies indicate that this transport mechanism is not autonomous but rather dependent on additional factors. Currently, five subtypes have been identified (EspPα-EspPε), with EspPα being associated with highly virulent EHEC serotypes and isolates from patients with severe disease. EspPα has been shown to degrade major proteins of the complement cascade, namely C3 and C5 and probably interferes with hemostasis by cleavage of coagulation factor V. Furthermore, EspPα is believed to contribute to biofilm formation perhaps by polymerization to rope-like structures. Together with the proteolytic activity, EspPα might ameliorate host colonization and interfere with host response.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Infecciones por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Serina Endopeptidasas/metabolismo , Células Cultivadas , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/química , Interacciones Huésped-Patógeno , Humanos , Estructura Terciaria de Proteína , Serina Endopeptidasas/química , Especificidad por Sustrato , Factores de Virulencia
14.
Microbiology (Reading) ; 157(Pt 11): 3094-3103, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21903756

RESUMEN

In enterobacteria such as Escherichia coli and Salmonella species, flagellar biogenesis is strictly dependent upon the master regulator flhDC. Here, we demonstrate that in enterohaemorrhagic E. coli (EHEC), the flagellar regulon is controlled by ClpXP, a member of the ATP-dependent protease family, through two pathways: (i) post-translational control of the FlhD/FlhC master regulator and (ii) transcriptional control of the flhDC operon. Both FlhD and FlhC proteins accumulated markedly following ClpXP depletion, and their half-lives were significantly longer in the mutant cells, suggesting that ClpXP is responsible for degrading FlhD and FlhC proteins, leading to downregulation of flagellar expression. ClpXP was involved in regulating the transcription of the flhD promoter only when the cells had entered stationary phase in a culture medium that markedly induced expression of the locus of enterocyte effacement (LEE). Comparative analyses of transcription from the flhD promoter in EHEC cells with different genetic backgrounds suggested that the downregulation of flhDC expression by ClpXP is dependent on the LEE-encoded GrlR-GrlA system. We have also shown that the degradation of FlhD and FlhC by ClpXP is responsible for downregulating flagellar expression even when LEE expression is induced.


Asunto(s)
Endopeptidasa Clp/metabolismo , Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Redes Reguladoras de Genes , Endopeptidasa Clp/genética , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/genética , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica , Mutación , Regiones Promotoras Genéticas , Regulón , Transactivadores/metabolismo
15.
Foodborne Pathog Dis ; 8(5): 643-6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21214372

RESUMEN

The antibacterial activity of an herbal combination composed of Mume Fructus, Coptidis Rhizoma, and Schizandrae Fructus extracts on enterohemorrhagic Escherichia coli (EHEC) was evaluated in the present study. The combination demonstrated antibacterial activity against all EHEC strains tested in this study, including those resistant to multiple antibiotics; minimum inhibitory concentration values ranged from 0.49 to 31.25 mg/mL. In in vivo antibacterial activity assay, the herbal combination was administered to mice after initial E. coli O157 infection and had significant effects on mouse mortality. The effects of the herbal combination on Shiga toxin release from EHEC O26, EHEC O111, and EHEC O157 strains containing the stx1 and stx2 genes were assessed by the reversed passive latex agglutination method, and there was no increased Shiga toxin release in the strain cultures containing the herbal combination. These results suggested that the herbal combination may be a safe and effective remedy for EHEC inhibition.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Escherichia coli Enterohemorrágica/efectos de los fármacos , Fitoterapia , Extractos Vegetales/farmacología , Prunus/química , Schisandra/química , Animales , Coptis chinensis , Combinación de Medicamentos , Farmacorresistencia Bacteriana Múltiple , Medicamentos Herbarios Chinos/uso terapéutico , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/mortalidad , Proteínas de Escherichia coli/metabolismo , Femenino , Pruebas de Fijación de Látex , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/uso terapéutico , Plantas Medicinales/química , Reacción en Cadena de la Polimerasa , Inhibidores de la Síntesis de la Proteína/metabolismo , Toxinas Shiga/metabolismo , Resultado del Tratamiento
16.
Infect Immun ; 78(10): 4294-301, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20643852

RESUMEN

Hemolytic-uremic syndrome (HUS) is a life-threatening disorder characterized by hemolytic anemia, thrombocytopenia, and renal insufficiency. It is caused mainly by infections with enterohemorrhagic Escherichia coli (EHEC). Recently, Shiga toxin 2, the best-studied virulence factor of EHEC, was reported to interact with complement, implying that complement may be involved in the pathogenesis of EHEC-induced HUS. The aim of the present study was to investigate whether or not the serine protease EspP, an important virulence factor of EHEC, interacts with complement proteins. EspP did not have any effect on the integrity of factor H or factor I. However, EspP was shown to cleave purified C3/C3b and C5. Cleavage of the respective complement proteins also occurred in normal human serum (NHS) as a source of C3/C3b or C5 or when purified complement proteins were added to the supernatant of an EspP-producing wild-type strain. Edman degradation allowed unequivocal mapping of all three main C3b fragments but not of the three main C5 fragments. Complement activation was significantly downregulated in all three pathways for C5-depleted serum to which C5, preincubated with EspP, was added (whereas C5 preincubated with an EspP mutant was able to fully reconstitute complement activation). This indicates that EspP markedly destroyed the functional activity, as measured by a commercial total complement enzyme-linked immunosorbent assay (Wieslab). Downregulation of complement by EspP in vivo may influence the colonization of EHEC bacteria in the gut or the disease severity of HUS.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C3/metabolismo , Complemento C3b/metabolismo , Complemento C5/metabolismo , Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Serina Endopeptidasas/metabolismo , Complemento C3/química , Complemento C3b/química , Complemento C5/química , Proteínas de Escherichia coli/genética , Regulación Enzimológica de la Expresión Génica , Síndrome Hemolítico-Urémico/microbiología , Humanos , Serina Endopeptidasas/genética
17.
PLoS Pathog ; 6(6): e1000960, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20585566

RESUMEN

NleG homologues constitute the largest family of Type 3 effectors delivered by pathogenic E. coli, with fourteen members in the enterohaemorrhagic (EHEC) O157:H7 strain alone. Identified recently as part of the non-LEE-encoded (Nle) effector set, this family remained uncharacterised and shared no sequence homology to other proteins including those of known function. The C-terminal domain of NleG2-3 (residues 90 to 191) is the most conserved region in NleG proteins and was solved by NMR. Structural analysis of this structure revealed the presence of a RING finger/U-box motif. Functional assays demonstrated that NleG2-3 as well as NleG5-1, NleG6-2 and NleG9' family members exhibited a strong autoubiquitination activity in vitro; a characteristic usually expressed by eukaryotic ubiquitin E3 ligases. When screened for activity against a panel of 30 human E2 enzymes, the NleG2-3 and NleG5-1 homologues showed an identical profile with only UBE2E2, UBE2E3 and UBE2D2 enzymes supporting NleG activity. Fluorescence polarization analysis yielded a binding affinity constant of 56+/-2 microM for the UBE2D2/NleG5-1 interaction, a value comparable with previous studies on E2/E3 affinities. The UBE2D2 interaction interface on NleG2-3 defined by NMR chemical shift perturbation and mutagenesis was shown to be generally similar to that characterised for human RING finger ubiquitin ligases. The alanine substitutions of UBE2D2 residues Arg5 and Lys63, critical for activation of eukaryotic E3 ligases, also significantly decreased both NleG binding and autoubiquitination activity. These results demonstrate that bacteria-encoded NleG effectors are E3 ubiquitin ligases analogous to RING finger and U-box enzymes in eukaryotes.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/crecimiento & desarrollo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Dominios RING Finger , Homología de Secuencia de Aminoácido , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
18.
Int Microbiol ; 12(3): 187-92, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19784925

RESUMEN

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a multifunctional housekeeping protein reported to be a target of several covalent modifications in many organisms. In a previous study, enterohemorrhagic (EHEC) and enteropathogenic (EPEC) Escherichia coli strains were shown to secrete GAPDH and the protein to bind human plasminogen and fibrinogen. Here we report that GAPDH of these pathogens is ADP-ribosylated either in the cytoplasm or in the extracellular medium. GAPDH catalyzes its own modification, which involves Cys-149 at the active site. ADP-ribosylation of extracellular GAPDH may play an important role in the host-pathogen interaction, as also proposed in other pathogens.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enteropatógena/enzimología , Escherichia coli Enteropatógena/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Procesamiento Proteico-Postraduccional , Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular
19.
Nat Chem Biol ; 5(4): 251-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19270684

RESUMEN

5'-Methylthioadenosine/S-adenosylhomocysteine nucleosidase (MTAN) is a bacterial enzyme involved in S-adenosylmethionine-related quorum sensing pathways that induce bacterial pathogenesis factors. Transition state analogs MT-DADMe-Immucillin-A, EtT-DADMe-Immucillin-A and BuT-DADMe-Immucillin-A are slow-onset, tight-binding inhibitors of Vibrio cholerae MTAN (VcMTAN), with equilibrium dissociation constants of 73, 70 and 208 pM, respectively. Structural analysis of VcMTAN with BuT-DADMe-Immucillin-A revealed interactions contributing to the high affinity. We found that in V. cholerae cells, these compounds are potent MTAN inhibitors with IC(50) values of 27, 31 and 6 nM for MT-, EtT- and BuT-DADMe-Immucillin-A, respectively; the compounds disrupt autoinducer production in a dose-dependent manner without affecting growth. MT- and BuT-DADMe-Immucillin-A also inhibited autoinducer-2 production in enterohemorrhagic Escherichia coli O157:H7 with IC(50) values of 600 and 125 nM, respectively. BuT-DADMe-Immucillin-A inhibition of autoinducer-2 production in both strains persisted for several generations and caused reduction in biofilm formation. These results support MTAN's role in quorum sensing and its potential as a target for bacterial anti-infective drug design.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/antagonistas & inhibidores , N-Glicosil Hidrolasas/antagonistas & inhibidores , Percepción de Quorum/efectos de los fármacos , Vibrio cholerae/enzimología , Adenina/análogos & derivados , Adenina/farmacología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Modelos Moleculares , Estructura Molecular , N-Glicosil Hidrolasas/química , N-Glicosil Hidrolasas/metabolismo , Pirrolidinas/farmacología , Especificidad por Sustrato , Vibrio cholerae/efectos de los fármacos , Vibrio cholerae/genética , Vibrio cholerae/metabolismo
20.
J Bacteriol ; 190(1): 438-41, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17981979

RESUMEN

Shiga toxin 2 (Stx2), one of the principal virulence factors of enterohemorrhagic Escherichia coli, is encoded by 933W, a lambda-like prophage. 933W prophage induction contributes to Stx2 production, and here, we provide evidence that Dam methyltransferase is essential for maintenance of 933W lysogeny. Our findings are consistent with the idea that the 933W prophage has a relatively low threshold for induction, which may promote Stx2 production during infection.


Asunto(s)
Colifagos/metabolismo , Escherichia coli Enterohemorrágica/enzimología , Escherichia coli Enterohemorrágica/virología , Toxina Shiga II/genética , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo , Colifagos/enzimología , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/genética , Kanamicina/farmacología , Lisogenia , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA