Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 253
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732126

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.


Asunto(s)
Escherichia coli Enterohemorrágica , Íleon , Organoides , Recto , Animales , Bovinos , Íleon/microbiología , Íleon/metabolismo , Íleon/ultraestructura , Recto/microbiología , Escherichia coli Enterohemorrágica/patogenicidad , Organoides/metabolismo , Organoides/microbiología , Moco/metabolismo , Infecciones por Escherichia coli/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura
2.
Cell Rep ; 43(4): 114004, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38522070

RESUMEN

During infections, host cells are exposed to pathogen-associated molecular patterns (PAMPs) and virulence factors that stimulate multiple signaling pathways that interact additively, synergistically, or antagonistically. The net effect of such higher-order interactions is a vital determinant of the outcome of host-pathogen interactions. Here, we demonstrate one such complex interplay between bacterial exotoxin- and PAMP-induced innate immune pathways. We show that two caspases activated during enterohemorrhagic Escherichia coli (EHEC) infection by lipopolysaccharide (LPS) and Shiga toxin (Stx) interact in a functionally antagonistic manner; cytosolic LPS-activated caspase-11 cleaves full-length gasdermin D (GSDMD), generating an active pore-forming N-terminal fragment (NT-GSDMD); subsequently, caspase-3 activated by EHEC Stx cleaves the caspase-11-generated NT-GSDMD to render it nonfunctional, thereby inhibiting pyroptosis and interleukin-1ß maturation. Bacteria typically subvert inflammasomes by targeting upstream components such as NLR sensors or full-length GSDMD but not active NT-GSDMD. Thus, our findings uncover a distinct immune evasion strategy where a bacterial toxin disables active NT-GSDMD by co-opting caspase-3.


Asunto(s)
Caspasa 3 , Gasderminas , Péptidos y Proteínas de Señalización Intracelular , Macrófagos , Proteínas de Unión a Fosfato , Piroptosis , Piroptosis/efectos de los fármacos , Proteínas de Unión a Fosfato/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Caspasa 3/metabolismo , Humanos , Animales , Ratones , Proteínas Reguladoras de la Apoptosis/metabolismo , Toxinas Bacterianas/metabolismo , Caspasas/metabolismo , Lipopolisacáridos/farmacología , Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/inmunología , Interleucina-1beta/metabolismo
3.
J Biosci Bioeng ; 137(6): 445-452, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38553372

RESUMEN

Bacteria produce and release small signal molecules, autoinducers, as an indicator of their cell density. The system, called a quorum-sensing (QS) system, is used to control not only virulence factors but also antibiotic production, sporulation, competence, and biofilm formation in bacteria. Different from antibiotics, QS inhibitors are expected to specifically repress the virulence factors in pathogenic bacteria without inhibiting growth or bactericidal effects. Therefore, since QS inhibitors have little risk of antibiotic-resistant bacteria emergence, they have been proposed as promising anti-bacterial agents. In the present study, we aimed to find new QS inhibitors that prohibit the signaling cascade of autoinducer 3 (AI-3) recognized by a QseCB two-component system that regulates some virulence factors of pathogens, such as enterohemorrhagic Escherichia coli (EHEC) and Salmonella enterica subsp. enterica serovar Typhimurium. We have established the method for QS-inhibitor screening using a newly constructed plasmid pLES-AQSA. E. coli DH5α transformed with the pLES-AQSA can produce ß-galactosidase that converts 5-bromo-4-chloro-3-indolyl ß-d-galactopyranoside (X-gal) into blue pigment (5-bromo-4-chloro-indoxyl) under the control of the QseCB system. By screening, Heyndrickxia coagulans (formerly Bacillus coagulans) 29-2E was found to produce an exopolysaccharide (EPS)-like water-soluble polymer that prohibits QseCB-mediated ß-galactosidase production without antibacterial activities. Further, the simultaneous injection of the 29-2E strain significantly improves the survival rate of Salmonella Typhimurium-infected silkworm larvae (from 0% to 83.3%), suggesting that the substance may be a promising inhibitor against the virulence of pathogens without risk of the emergence of antibiotic-resistant bacteria.


Asunto(s)
Percepción de Quorum , Salmonella typhimurium , Percepción de Quorum/efectos de los fármacos , Salmonella typhimurium/efectos de los fármacos , Virulencia , Bacillus/metabolismo , Antibacterianos/farmacología , Lactonas/farmacología , Lactonas/metabolismo , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enterohemorrágica/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Homoserina/análogos & derivados
4.
J Med Microbiol ; 71(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35037615

RESUMEN

Introduction. Bacterial dysentery is one of the greatest causes of morbidity and mortality worldwide. Campylobacter spp. and diarrhoeagenic Escherichia coli (DEC) are recognised as the most common causes of bacterial enteritis in developing countries including India.Hypothesis/Gap statement. Rapid and accurate identification of dysentery causing organisms using molecular methods is essential for better disease management, epidemiology and outbreak investigations.Aim. In view of the limited information available on the dysentery causing agents like Campylobacter spp., enterohemorrhagic E. coli (EHEC)/enteropathogenic E. coli (EPEC) and enteroinvasive E. coli (EIEC)/Shigella in India, this study was undertaken to investigate the presence of these pathogens in human and poultry stool samples by molecular methods.Methodology. In total, 400 human stool samples and 128 poultry samples were studied. Microaerophilic culture along with real-time multiplex PCR with the targets specific to the genus Campylobacter, Campylobacter jejuni, Campylobacter coli, EHEC, EPEC and EIEC/Shigella was performed. Further species confirmation was done using MALDI-TOF MS.Results. On microaerophilic culture, C. coli was isolated in one human sample and two C. jejuni and one C. fetus in poultry samples. On PCR analysis, among human stool samples, typical EPEC (42%) was predominantly seen followed by Campylobacter spp. (19%) and EIEC/Shigella (10%). In contrast, Campylobacter spp. (41%) was predominant in poultry samples, followed by typical EPEC (26%) and EIEC/Shigella (9%). Poly-infections with Campylobacter spp. and DEC were also observed among both sources.Conclusion. The present study documented the increased prevalence of Campylobacter spp. in humans compared with the results of previous studies from India. Typical EPEC was found to be predominant in children less than 5 years of age in this study. The high prevalence of coinfections in the current study indicates that a multiple aetiology of diarrhoea is common in our settings.


Asunto(s)
Infecciones por Campylobacter , Campylobacter , Disentería , Escherichia coli Enterohemorrágica , Infecciones por Escherichia coli , Campylobacter/genética , Campylobacter/patogenicidad , Infecciones por Campylobacter/epidemiología , Preescolar , Diarrea/epidemiología , Diarrea/microbiología , Disentería/epidemiología , Disentería/microbiología , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/epidemiología , Heces , Humanos , India , Reacción en Cadena de la Polimerasa Multiplex , Prevalencia , Reacción en Cadena en Tiempo Real de la Polimerasa , Shigella/genética
5.
Mol Microbiol ; 117(1): 86-101, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34411346

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) causes severe human diseases worldwide. The type 3 secretion system and effector proteins are essential for EHEC infection, and are encoded by the locus of enterocyte effacement (LEE). RNA-binding protein Hfq is essential for small regulatory RNA (sRNA)-mediated regulation at a posttranscriptional level and full virulence of many pathogenic bacteria. Although two early studies indicated that Hfq represses LEE expression by posttranscriptionally controlling the expression of genes grlRA and/or ler, both of which encode LEE regulators mediating a positive regulatory loop, the detailed molecular mechanism and biological significance remain unclear. Herein, we show that LEE overexpression was caused by defective RNA-binding activity of the Hfq distal face, which posttranscriptionally represses grlA and ler expression. In vitro analyses revealed that the Hfq distal face directly binds near the translational initiation site of grlA and ler mRNAs, and inhibits their translation. Taken together, we conclude that Hfq inhibits grlA and ler translation by binding their mRNAs through the distal face in an sRNA-independent manner. Additionally, we show that Hfq-mediated repression of LEE is critical for normal EHEC growth because all suppressor mutations that restored the growth defect in the hfq mutant abolished hfq deletion-induced overexpression of LEE.


Asunto(s)
Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Proteína de Factor 1 del Huésped/metabolismo , ARN Pequeño no Traducido/genética , Transactivadores/metabolismo , Escherichia coli Enterohemorrágica/crecimiento & desarrollo , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/genética , Proteína de Factor 1 del Huésped/genética , Humanos , Mutación , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas , ARN Bacteriano/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transactivadores/genética , Sistemas de Secreción Tipo III , Virulencia
6.
Toxins (Basel) ; 13(10)2021 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-34678979

RESUMEN

O80:H2 enterohemorrhagic Escherichia coli (EHEC) of sequence type ST301 is one of the main serotypes causing European hemolytic and uremic syndrome, but also invasive infections, due to extra-intestinal virulence factors (VFs). Here, we determined whether other such heteropathotypes exist among ST301. EnteroBase was screened for ST301 strains that were included in a general SNP-phylogeny. French strains belonging to a new heteropathotype clone were sequenced. ST, hierarchical clusters (HC), serotype, resistome, and virulome were determined using EnteroBase, the CGE website, and local BLAST. The ST301 general phylogeny shows two groups. Group A (n = 25) is mainly composed of enteropathogenic E. coli, whereas group B (n = 55) includes mostly EHEC. Three serotypes, O186:H2, O45:H2 and O55:H9, share the same virulome as one of the O80:H2 sub-clones from which they derive subsequent O-antigen switches. The O55:H9 clone, mainly present in France (n = 29), as well as in the UK (n = 5) and Germany (n = 1), has a low background of genetic diversity (four HC20), although it has three Stx subtypes, an H-antigen switch, and genes encoding the major extra-intestinal VF yersiniabactin, and extended-spectrum beta-lactamases. Diverse heteropathotype clones genetically close to the O80:H2 clone are present among the ST301, requiring close European monitoring, especially the virulent O55:H9 clone.


Asunto(s)
Escherichia coli Shiga-Toxigénica/genética , Escherichia coli Shiga-Toxigénica/patogenicidad , Factores de Virulencia/genética , Animales , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/microbiología , Europa (Continente) , Humanos , Filogenia , Serogrupo , Escherichia coli Shiga-Toxigénica/clasificación , Virulencia , Secuenciación Completa del Genoma
7.
Nucleic Acids Res ; 49(19): 10988-11004, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34591974

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 relies on sRNAs to coordinate expression of metabolic and virulence factors to colonize the host. Here, we focus on the sRNA, named MavR (metabolism and virulence regulator), that is conserved among pathogenic Enterobacteriaceae. MavR is constitutively expressed under in vitro conditions that promote EHEC virulence gene expression. Using MS2-affinity purification coupled with RNA sequencing, the eutR transcript was identified as a putative target of MavR. EutR is a transcription factor that promotes expression of genes required for ethanolamine metabolism as well as virulence factors important for host colonization. MavR binds to the eutR coding sequence to protect the eutR transcript from RNase E-mediated degradation. Ultimately, MavR promotes EutR expression and in turn ethanolamine utilization and ethanolamine-dependent growth. RNAseq analyses revealed that MavR also affected expression of genes important for other metabolic pathways, motility, oxidative stress and attaching and effacing lesion formation, which contribute to EHEC colonization of the gastrointestinal tract. In support of the idea that MavR-dependent gene expression affects fitness during infection, deletion of mavR resulted in significant (∼10- to 100-fold) attenuation in colonization of the mammalian intestine. Altogether, these studies reveal an important, extensive, and robust phenotype for a bacterial sRNA in host-pathogen interactions.


Asunto(s)
Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Pequeño no Traducido/genética , Factores de Transcripción/genética , Factores de Virulencia/genética , Animales , Emparejamiento Base , Secuencia de Bases , Colon/metabolismo , Colon/microbiología , Endorribonucleasas/química , Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/patología , Proteínas de Escherichia coli/metabolismo , Etanolamina/metabolismo , Femenino , Regulación Bacteriana de la Expresión Génica , Aptitud Genética , Células HeLa , Interacciones Microbiota-Huesped/genética , Humanos , Ratones , ARN Bacteriano/metabolismo , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo , Virulencia , Factores de Virulencia/metabolismo
8.
Ren Fail ; 43(1): 382-387, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33641616

RESUMEN

BACKGROUND: Insulin-like growth factor-binding protein (IGFBP) 2 plays an important role in the regulation of cell adhesion, migration, growth, and apoptosis. This study aimed to investigate the clinical significance of serum IGFBP2 as a biomarker for disease activity and severity in hemolytic uremic syndrome (HUS) induced by enterohemorrhagic Escherichia coli (EHEC). METHODS: IGFBP2 production by human renal glomerular endothelial cells (RGECs) after exposure to Shiga toxin 2 (Stx-2) was investigated in vitro. Serum IGFBP2 levels in blood samples obtained from 22 patients with HUS and 10 healthy controls (HCs) were quantified using an enzyme-linked immunosorbent assay. The results were compared to the clinical features of HUS and serum tau and cytokine levels. RESULTS: Stx-2 induced the production of IGFBP2 in RGECs in a dose-dependent manner. Serum IGFBP2 levels were significantly higher in patients with HUS than in HCs and correlated with disease severity. Additionally, serum IGFBP2 levels were significantly higher in patients with encephalopathy than in those without encephalopathy. A serum IGFBP2 level above 3585 pg/mL was associated with a high risk of encephalopathy. Furthermore, serum IGFBP2 levels significantly correlated with serum levels of tau and inflammatory cytokines associated with the development of HUS. CONCLUSIONS: Correlation of serum IGFBP2 level with disease activity in patients with HUS suggests that IGFBP2 may be considered as a possible indicator for disease activity and severity in HUS. Larger studies and additional experiments using various cells in central nervous system should elucidate the true value of IGFBP2 as a clinical diagnostic marker. ABBREVIATIONS: IGFBP: insulin-like growth factor-binding protein; HUS: hemolytic uremic syndrome; EHEC: enterohemorrhagic Escherichia coli; RGECs: renal glomerular endothelial cells; STx-2: Shiga toxin 2; HCs: healthy controls; LPS: lipopolysaccharide; ROC: receiver operating characteristic; sTNFR: soluble tumor necrosis factor receptor.


Asunto(s)
Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/microbiología , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/microbiología , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Adolescente , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Niño , Preescolar , Infecciones por Escherichia coli/complicaciones , Femenino , Síndrome Hemolítico-Urémico/patología , Humanos , Lactante , Masculino , Curva ROC , Índice de Severidad de la Enfermedad , Adulto Joven
9.
Genetics ; 217(1): 1-17, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33683370

RESUMEN

Infection with antibiotic-resistant bacteria is an emerging life-threatening issue worldwide. Enterohemorrhagic Escherichia coli O157: H7 (EHEC) causes hemorrhagic colitis and hemolytic uremic syndrome via contaminated food. Treatment of EHEC infection with antibiotics is contraindicated because of the risk of worsening the syndrome through the secreted toxins. Identifying the host factors involved in bacterial infection provides information about how to combat this pathogen. In our previous study, we showed that EHEC colonizes in the intestine of Caenorhabditis elegans. However, the host factors involved in EHEC colonization remain elusive. Thus, in this study, we aimed to identify the host factors involved in EHEC colonization. We conducted forward genetic screens to isolate mutants that enhanced EHEC colonization and named this phenotype enhanced intestinal colonization (Inc). Intriguingly, four mutants with the Inc phenotype showed significantly increased EHEC-resistant survival, which contrasts with our current knowledge. Genetic mapping and whole-genome sequencing (WGS) revealed that these mutants have loss-of-function mutations in unc-89. Furthermore, we showed that the tolerance of unc-89(wf132) to EHEC relied on HLH-30/TFEB activation. These findings suggest that hlh-30 plays a key role in pathogen tolerance in C. elegans.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Caenorhabditis elegans/genética , Infecciones por Escherichia coli/genética , Inmunidad Innata , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/inmunología , Intestinos/microbiología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
10.
PLoS One ; 16(2): e0245470, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539359

RESUMEN

BACKGROUND: Knowledge about the distribution of Escherichia Coli (E. coli) pathotypes in Iran is limited. This nation-wide survey aims to provide a comprehensive description of the distribution of five pathogenic E. coli in Iran. METHODS: Stool samples were collected from 1,306 acute diarrhea cases from 15 provinces (2013-2014). E. coli-positive cultures underwent PCR testing for the detection of STEC, ETEC, EPEC, EAEC, and EIEC pathotypes. Pathotype frequency by province, age-group, and season was estimated. RESULTS: 979 diarrhea samples (75.0%) were culture-positive for E. coli (95% CI: 72.6, 77.3%), and 659 (50.5%) were pathogenic E. coli (95% CI: 47.8, 53.2%). STEC was the most frequent pathotype (35.4%). ETEC (14.0%) and EPEC (13.1%) were the second and the third most frequent pathotypes, respectively. EAEC (4.3%) and EIEC (0.3%) were not highly prevalent. Fars (88.7%) and Khorasan-e-Razavi (34.8%) provinces had the highest and lowest frequencies, respectively. E. coli pathotypes were more frequent in warmer than cooler seasons, showed the highest frequency among children under five years of age (73%), and had no significant association with participants' gender. CONCLUSIONS: Diarrheagenic E. coli may be an important cause of acute diarrhea in adults and children in Iran. STEC and ETEC seem to be widespread in the country with a peak in warmer seasons, impacting the recommended use of seasonal STEC and ETEC vaccines, especially in high-risk groups. Monitoring the incidence of E. coli pathotypes, serotypes, and antibiotic resistance over time is highly recommended for evaluation of interventions.


Asunto(s)
Diarrea/epidemiología , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Infecciones por Escherichia coli/epidemiología , Enfermedad Aguda , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios Transversales , Diarrea/microbiología , Escherichia coli Enterohemorrágica/aislamiento & purificación , Escherichia coli Enteropatógena/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Heces/microbiología , Femenino , Genes Bacterianos , Humanos , Lactante , Recién Nacido , Irán/epidemiología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Prevalencia , Estaciones del Año , Virulencia/genética , Adulto Joven
11.
Mol Microbiol ; 116(1): 168-183, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33567149

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC), an enteropathogen that colonizes in the intestine, causes severe diarrhea and hemorrhagic colitis in humans by the expression of the type III secretion system (T3SS) and Shiga-like toxins (Stxs). However, how EHEC can sense and respond to the changes in the alimentary tract and coordinate the expression of these virulence genes remains elusive. The T3SS-related genes are known to be regulated by the locus of enterocyte effacement (LEE)-encoded regulators, such as Ler, as well as non-LEE-encoded regulators in response to different environmental cues. Herein, we report that OmpR, which participates in the adaptation of E. coli to osmolarity and pH alterations, is required for EHEC infection in Caenorhabditis elegans. OmpR protein was able to directly bind to the promoters of ler and stx1 (Shiga-like toxin 1) and regulate the expression of T3SS and Stx1, respectively, at the transcriptional level. Moreover, we demonstrated that the expression of ler in EHEC is in response to the intestinal environment and is regulated by OmpR in C. elegans. Taken together, we reveal that OmpR is an important regulator of EHEC which coordinates the expression of virulence factors during gastrointestinal infection in vivo.


Asunto(s)
Proteínas Bacterianas/genética , Caenorhabditis elegans/microbiología , Escherichia coli Enterohemorrágica/patogenicidad , Toxina Shiga I/biosíntesis , Transactivadores/genética , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/metabolismo , Sistema Digestivo/microbiología , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Toxina Shiga I/genética , Transactivadores/biosíntesis , Transactivadores/metabolismo , Transcripción Genética/genética , Activación Transcripcional/genética , Sistemas de Secreción Tipo III/biosíntesis , Sistemas de Secreción Tipo III/genética , Factores de Virulencia/genética
12.
Nat Commun ; 12(1): 90, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397943

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) induces changes to the intestinal cell cytoskeleton and formation of attaching and effacing lesions, characterized by the effacement of microvilli and then formation of actin pedestals to which the bacteria are tightly attached. Here, we use a Caenorhabditis elegans model of EHEC infection to show that microvillar effacement is mediated by a signalling pathway including mitotic cyclin-dependent kinase 1 (CDK1) and diaphanous-related formin 1 (CYK1). Similar observations are also made using EHEC-infected human intestinal cells in vitro. Our results support the use of C. elegans as a host model for studying attaching and effacing lesions in vivo, and reveal that the CDK1-formin signal axis is necessary for EHEC-induced microvillar effacement.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Escherichia coli Enterohemorrágica/fisiología , Interacciones Huésped-Patógeno , Microvellosidades/microbiología , Microvellosidades/patología , Actinas/metabolismo , Animales , Células CACO-2 , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/ultraestructura , Carbohidrato Epimerasas/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Forminas , Humanos , Intestinos/microbiología , Microvellosidades/metabolismo , Fosforilación , Fosfotreonina/metabolismo , Virulencia
13.
J Biol Chem ; 296: 100299, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460651

RESUMEN

The human Gb3/CD77 synthase, encoded by the A4GALT gene, is an unusually promiscuous glycosyltransferase. It synthesizes the Galα1→4Gal linkage on two different glycosphingolipids (GSLs), producing globotriaosylceramide (Gb3, CD77, Pk) and the P1 antigen. Gb3 is the major receptor for Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli. A single amino acid substitution (p.Q211E) ramps up the enzyme's promiscuity, rendering it able to attach Gal both to another Gal residue and to GalNAc, giving rise to NOR1 and NOR2 GSLs. Human Gb3/CD77 synthase was long believed to transfer Gal only to GSL acceptors, therefore its GSL products were, by default, considered the only human Stx receptors. Here, using soluble, recombinant human Gb3/CD77 synthase and p.Q211E mutein, we demonstrate that both enzymes can synthesize the P1 glycotope (terminal Galα1→4Galß1→4GlcNAc-R) on a complex type N-glycan and a synthetic N-glycoprotein (saposin D). Moreover, by transfection of CHO-Lec2 cells with vectors encoding human Gb3/CD77 synthase and its p.Q211E mutein, we demonstrate that both enzymes produce P1 glycotopes on N-glycoproteins, with the mutein exhibiting elevated activity. These P1-terminated N-glycoproteins are recognized by Stx1 but not Stx2 B subunits. Finally, cytotoxicity assays show that Stx1 can use P1 N-glycoproteins produced in CHO-Lec2 cells as functional receptors. We conclude that Stx1 can recognize and use P1 N-glycoproteins in addition to its canonical GSL receptors to enter and kill the cells, while Stx2 can use GSLs only. Collectively, these results may have important implications for our understanding of the Shiga toxin pathology.


Asunto(s)
Galactosiltransferasas/química , Globósidos/química , Toxina Shiga I/química , Trihexosilceramidas/química , Acetilgalactosamina/química , Acetilgalactosamina/metabolismo , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Animales , Sitios de Unión , Células CHO , Secuencia de Carbohidratos , Cricetulus , Escherichia coli Enterohemorrágica/química , Escherichia coli Enterohemorrágica/patogenicidad , Galactosa/química , Galactosa/metabolismo , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Expresión Génica , Globósidos/biosíntesis , Globósidos/metabolismo , Glucosa/química , Glucosa/metabolismo , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Trihexosilceramidas/biosíntesis
14.
mBio ; 12(1)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33468701

RESUMEN

The mammalian gastrointestinal tract is a complex biochemical organ that generates a diverse milieu of host- and microbe-derived metabolites. In this environment, bacterial pathogens sense and respond to specific stimuli, which are integrated into the regulation of their virulence programs. Previously, we identified the transcription factor FadR, a long-chain fatty acid (LCFA) acyl coenzyme A (acyl-CoA) sensor, as a novel virulence regulator in the human foodborne pathogen enterohemorrhagic Escherichia coli (EHEC). Here, we demonstrate that exogenous LCFAs directly inhibit the locus of enterocyte effacement (LEE) pathogenicity island in EHEC through sensing by FadR. Moreover, in addition to LCFAs that are 18 carbons in length or shorter, we introduce host-derived arachidonic acid (C20:4) as an additional LCFA that is recognized by the FadR system in EHEC. We show that arachidonic acid is processed by the acyl-CoA synthetase FadD, which permits binding to FadR and decreases FadR affinity for its target DNA sequences. This interaction enables the transcriptional regulation of FadR-responsive operons by arachidonic acid in EHEC, including the LEE. Finally, we show that arachidonic acid inhibits hallmarks of EHEC disease in a FadR-dependent manner, including EHEC attachment to epithelial cells and the formation of attaching and effacing lesions. Together, our findings delineate a molecular mechanism demonstrating how LCFAs can directly inhibit the virulence of an enteric bacterial pathogen. More broadly, our findings expand the repertoire of ligands sensed by the canonical LFCA sensing machinery in EHEC to include arachidonic acid, an important bioactive lipid that is ubiquitous within host environments.IMPORTANCE Polyunsaturated fatty acids (PUFAs) play important roles in host immunity. Manipulation of lipid content in host tissues through diet or pharmacological interventions is associated with altered severity of various inflammatory diseases. Our work introduces a defined host-pathogen interaction by which arachidonic acid, a host-derived and dietary PUFA, can impact the outcome of enteric infection with the human pathogen enterohemorrhagic Escherichia coli (EHEC). We show that long-chain fatty acids including arachidonic acid act as signaling molecules that directly suppress a key pathogenicity island in EHEC following recognition by the fatty acyl-CoA-responsive transcription factor FadR. Thus, in addition to its established effects on host immunity and its bactericidal activities against other pathogens, we demonstrate that arachidonic acid also acts as a signaling molecule that inhibits virulence in an enteric pathogen.


Asunto(s)
Ácido Araquidónico/metabolismo , Escherichia coli Enterohemorrágica/fisiología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Ácidos Grasos/metabolismo , Interacciones Huésped-Patógeno , Ácido Araquidónico/farmacología , Escherichia coli Enterohemorrágica/efectos de los fármacos , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Modelos Biológicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Virulencia/genética , Factores de Virulencia/genética
15.
Infect Immun ; 89(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33229368

RESUMEN

During passage through the human gastrointestinal tract, enterohemorrhagic Escherichia coli (EHEC) is exposed to membrane-damaging bile in the small intestine. We previously reported that EHEC treatment with a physiological bile salt mixture upregulates basRS, encoding a two-component system, and arnBCADTEF, encoding the aminoarabinose lipid A modification pathway (J. V. Kus, A. Gebremedhin, V. Dang, S. L. Tran, A. Serbanescu, and D. Barnett Foster, J Bacteriol 193: 4509-4515, 2011, https://doi.org/10.1128/JB.00200-11). The present study examined the effect of bile salt mix (BSM) treatment on EHEC resistance to three human gastrointestinal defense peptides-HD-5, HNP-1, and LL-37-as well as the role of basRS and arnT in the respective responses. After BSM treatment, EHEC resistance to HD-5 and HNP-1 was significantly increased in a BSM-, defensin dose-dependent manner. The resistance phenotype was dependent on both basRS and arnT However, the BSM treatment did not alter EHEC resistance to LL-37, even when the ompT gene, encoding an LL-37 cleavage protease, was disrupted. Interestingly, enteropathogenic E. coli, a related pathogen that infects the small intestine, showed a similar BSM-induced resistance phenotype. Using a model of EHEC infection in Galleria mellonella, we found significantly lower survival rates in wax moth larvae infected with BSM-treated wild-type EHEC than in those infected with a BSM-treated basS mutant, suggesting that treatment with a physiological BSM enhances virulence through a basS-mediated pathway. The results of this investigation provide persuasive evidence that bile salts typically encountered during transit through the small intestine can serve as an environmental cue for EHEC, enhancing resistance to several key host defense peptides.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/efectos de los fármacos , Ácidos y Sales Biliares/farmacología , Ácidos y Sales Biliares/uso terapéutico , Resistencia a la Enfermedad/efectos de los fármacos , Escherichia coli Enterohemorrágica/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Virulencia/efectos de los fármacos , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/patología , Humanos
16.
Curr Genet ; 67(2): 221-224, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33219834

RESUMEN

Phenotypic and genetic heterogeneities are conserved features of prokaryotic populations. During periods of stress, this programmed diversity increases the likelihood that variants within the population will survive the adverse conditions, allowing for proliferation. Phenotypic heterogeneity can have a mutational or indeed a non-mutational basis as observed in bet-hedging strategies adopted by antibiotic-tolerant persister cells. Genetic variants can arise by phase variation (slip-strand mispairing, promoter inversions etc.), nucleotide polymorphisms resulting from replication errors or larger rearrangements such as deletions and insertions. In the face of selective pressures, these alterations may be neutral, beneficial or deleterious.We recently described the genetic basis of tolerance to a normally toxic metabolite, D-serine (D-ser) in enterohaemorrhagic E. coli (EHEC). Here we summarize our work in the context of population dynamics, provide further discussion on the distinction between these tolerance mechanisms and the importance of heterogeneity for maximising adaptive potential.


Asunto(s)
Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/genética , Heterogeneidad Genética , Serina/genética , Adaptación Fisiológica/genética , Antibacterianos/farmacología , Tolerancia a Medicamentos/genética , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/microbiología , Humanos , Mutación/genética
17.
Front Immunol ; 11: 561337, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329523

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC), a human pathogen, also infects Caenorhabditis elegans. We demonstrated previously that C. elegans activates the p38 MAPK innate immune pathway to defend against EHEC infection. However, whether a C. elegans pattern recognition receptor (PRR) exists to regulate the immune pathway remains unknown. PRRs identified in other metazoans contain several conserved domains, including the leucine-rich repeat (LRR). By screening a focused RNAi library, we identified the IGLR-2, a transmembrane protein containing the LRR domain, as a potential immune regulator in C. elegans. Our data showed that iglr-2 regulates the host susceptibility to EHEC infection. Moreover, iglr-2 is required for pathogen avoidance to EHEC. The iglr-2 overexpressed strain, which was more resistant to EHEC originally, showed hypersusceptibility to EHEC upon knockdown of the p38 MAPK pathway. Together, our data suggested that iglr-2 plays an important role in C. elegans to defend EHEC by regulating pathogen-avoidance behavior and the p38 MAPK pathway.


Asunto(s)
Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/inmunología , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/inmunología , Interacciones Microbiota-Huesped/inmunología , Proteínas de la Membrana/inmunología , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Infecciones por Escherichia coli/microbiología , Técnicas de Silenciamiento del Gen , Inmunidad Innata , Proteínas de la Membrana/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Sci Immunol ; 5(53)2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33246946

RESUMEN

Inflammatory caspase-dependent cytosolic lipopolysaccharide (LPS) sensing is a critical arm of host defense against bacteria. How pathogens overcome this pathway to establish infections is largely unknown. Enterohemorrhagic Escherichia coli (EHEC) is a clinically important human pathogen causing hemorrhagic colitis and hemolytic uremic syndrome. We found that a bacteriophage-encoded virulence factor of EHEC, Shiga toxin (Stx), suppresses caspase-11-mediated activation of the cytosolic LPS sensing pathway. Stx was essential and sufficient to inhibit pyroptosis and interleukin-1 (IL-1) responses elicited specifically by cytosolic LPS. The catalytic activity of Stx was necessary for suppression of inflammasome responses. Stx impairment of inflammasome responses to cytosolic LPS occurs at the level of gasdermin D activation. Stx also suppresses inflammasome responses in vivo after LPS challenge and bacterial infection. Overall, this study assigns a previously undescribed inflammasome-subversive function to a well-known bacterial toxin, Stx, and reveals a new phage protein-based pathogen blockade of cytosolic immune surveillance.


Asunto(s)
Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/inmunología , Inflamasomas/inmunología , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Animales , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Caspasas Iniciadoras/genética , Caspasas Iniciadoras/metabolismo , Chlorocebus aethiops , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Modelos Animales de Enfermedad , Escherichia coli Enterohemorrágica/inmunología , Escherichia coli Enterohemorrágica/virología , Infecciones por Escherichia coli/microbiología , Femenino , Humanos , Vigilancia Inmunológica , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Proteínas de Unión a Fosfato/metabolismo , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Células Vero , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo
20.
Proc Natl Acad Sci U S A ; 117(40): 25055-25065, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32968018

RESUMEN

Enterohemorrhagic Escherichia coli is a significant human pathogen that causes disease ranging from hemorrhagic colitis to hemolytic uremic syndrome. The latter can lead to potentially fatal renal failure and is caused by the release of Shiga toxins that are encoded within lambdoid bacteriophages. The toxins are encoded within the late transcript of the phage and are regulated by antitermination of the PR' late promoter during lytic induction of the phage. During lysogeny, the late transcript is prematurely terminated at tR' immediately downstream of PR', generating a short RNA that is a byproduct of antitermination regulation. We demonstrate that this short transcript binds the small RNA chaperone Hfq, and is processed into a stable 74-nt regulatory small RNA that we have termed StxS. StxS represses expression of Shiga toxin 1 under lysogenic conditions through direct interactions with the stx1AB transcript. StxS acts in trans to activate expression of the general stress response sigma factor, RpoS, through direct interactions with an activating seed sequence within the 5' UTR. Activation of RpoS promotes high cell density growth under nutrient-limiting conditions. Many phages utilize antitermination to regulate the lytic/lysogenic switch and our results demonstrate that short RNAs generated as a byproduct of this regulation can acquire regulatory small RNA functions that modulate host fitness.


Asunto(s)
Escherichia coli Enterohemorrágica/genética , Síndrome Hemolítico-Urémico/genética , ARN Pequeño no Traducido/genética , Toxina Shiga/genética , Proteínas Bacterianas/genética , Bacteriófago lambda/genética , Bacteriófago lambda/patogenicidad , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Síndrome Hemolítico-Urémico/microbiología , Proteína de Factor 1 del Huésped/genética , Humanos , Lisogenia/genética , Regiones Promotoras Genéticas/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Factor sigma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...