Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 256
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732126

ABSTRACT

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.


Subject(s)
Enterohemorrhagic Escherichia coli , Ileum , Organoids , Rectum , Animals , Cattle , Ileum/microbiology , Ileum/metabolism , Ileum/ultrastructure , Rectum/microbiology , Enterohemorrhagic Escherichia coli/pathogenicity , Organoids/metabolism , Organoids/microbiology , Mucus/metabolism , Escherichia coli Infections/microbiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure
2.
Nat Commun ; 15(1): 4462, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796512

ABSTRACT

Virulence and metabolism are often interlinked to control the expression of essential colonisation factors in response to host-associated signals. Here, we identified an uncharacterised transporter of the dietary monosaccharide ʟ-arabinose that is widely encoded by the zoonotic pathogen enterohaemorrhagic Escherichia coli (EHEC), required for full competitive fitness in the mouse gut and highly expressed during human infection. Discovery of this transporter suggested that EHEC strains have an enhanced ability to scavenge ʟ-arabinose and therefore prompted us to investigate the impact of this nutrient on pathogenesis. Accordingly, we discovered that ʟ-arabinose enhances expression of the EHEC type 3 secretion system, increasing its ability to colonise host cells, and that the underlying mechanism is dependent on products of its catabolism rather than the sensing of ʟ-arabinose as a signal. Furthermore, using the murine pathogen Citrobacter rodentium, we show that ʟ-arabinose metabolism provides a fitness benefit during infection via virulence factor regulation, as opposed to supporting pathogen growth. Finally, we show that this mechanism is not restricted to ʟ-arabinose and extends to other pentose sugars with a similar metabolic fate. This work highlights the importance integrating central metabolism with virulence regulation in order to maximise competitive fitness of enteric pathogens within the host-niche.


Subject(s)
Arabinose , Citrobacter rodentium , Enterohemorrhagic Escherichia coli , Arabinose/metabolism , Animals , Mice , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/metabolism , Citrobacter rodentium/genetics , Humans , Virulence , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Virulence Factors/metabolism , Virulence Factors/genetics , Enterobacteriaceae Infections/microbiology , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Escherichia coli Infections/microbiology , Female
3.
Georgian Med News ; (348): 78-80, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38807396

ABSTRACT

Escherichia coli is a gram-negative bacillus and considered to be the normal pathogen of intestinal and extraintestinal manifestations depending upon the strain. A variety of strains exist that are responsible for causing myriads of clinical presentation. E.coli O157: H7 being the most common and severe bacterial pathogen is the leading cause of bloody diarrhea. EHEC (Enterohemorrhagic E.coli) is responsible for causing severe complications like HC (Hemorrhagic colitis). Herein, we present the case of a young girl with E.coli O157:H7 infection and review the related literature. A previously healthy 37-year-old female presented with bloody diarrhea, fever, headache, and lower abdominal pain. As per history she had eaten a hamburger, denied any recent travel and absence of inflammatory bowel disease or bloody stools in family history. Physical examination revealed normal vital signs and the physical findings were unremarkable except for severe abdominal pain. Her stool was hem-occult positive. The complete blood count was within normal limits except neutrophilia and leukocytosis. An abdominal ultrasound showed thickened bowel loops consistent with colitis. First week of her hospital course, she continued to have bloody diarrhea and severe abdominal pain. Her final stool submitted to the laboratory on day 7 was consistent with a blood clot, following her developed low urine output and hematuria, with a serum creatinine of 2.1 mg/dl on day 5. Her renal symptoms were treated with fluids. She was given supportive treatment, and her platelet count and hemoglobin were stabilized. In early stages of bloody diarrhea, parental hydration plays a major role in accelerating volume expansion. Rapid stool analysis for these bacteria can alert specialists to deal with severe complications like HUS.


Subject(s)
Escherichia coli Infections , Hemolytic-Uremic Syndrome , Humans , Female , Adult , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/diagnosis , Escherichia coli Infections/microbiology , Escherichia coli Infections/diagnosis , Escherichia coli Infections/complications , Diarrhea/microbiology , Escherichia coli O157/isolation & purification , Abdominal Pain/microbiology , Abdominal Pain/etiology , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/isolation & purification
4.
PLoS One ; 19(5): e0298746, 2024.
Article in English | MEDLINE | ID: mdl-38787890

ABSTRACT

Enterohemorrhagic E. coli (EHEC) is considered to be the most dangerous pathotype of E. coli, as it causes severe conditions such as hemorrhagic colitis (HC) and hemolytic uremic syndrome (HUS). Antibiotic treatment of EHEC infections is generally not recommended since it may promote the production of the Shiga toxin (Stx) and lead to worsened symptoms. This study explores how exposure to the fluoroquinolone ciprofloxacin reorganizes the transcriptome and proteome of EHEC O157:H7 strain EDL933, with special emphasis on virulence-associated factors. As expected, exposure to ciprofloxacin caused an extensive upregulation of SOS-response- and Stx-phage proteins, including Stx. A range of other virulence-associated factors were also upregulated, including many genes encoded by the LEE-pathogenicity island, the enterohemolysin gene (ehxA), as well as several genes and proteins involved in LPS production. However, a large proportion of the genes and proteins (17 and 8%, respectively) whose expression was upregulated upon ciprofloxacin exposure (17 and 8%, respectively) are not functionally assigned. This indicates a knowledge gap in our understanding of mechanisms involved in EHECs response to antibiotic-induced stress. Altogether, the results contribute to better understanding of how exposure to ciprofloxacin influences the virulome of EHEC and generates a knowledge base for further studies on how EHEC responds to antibiotic-induced stress. A deeper understanding on how EHEC responds to antibiotics will facilitate development of novel and safer treatments for EHEC infections.


Subject(s)
Ciprofloxacin , Proteomics , Transcriptome , Ciprofloxacin/pharmacology , Proteomics/methods , Virulence/drug effects , Transcriptome/drug effects , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Virulence Factors/genetics , Virulence Factors/metabolism , Proteome/metabolism , Gene Expression Profiling , Humans
5.
J Biosci Bioeng ; 137(6): 445-452, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38553372

ABSTRACT

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.


Subject(s)
Quorum Sensing , Salmonella typhimurium , Quorum Sensing/drug effects , Salmonella typhimurium/drug effects , Virulence , Bacillus/metabolism , Anti-Bacterial Agents/pharmacology , Lactones/pharmacology , Lactones/metabolism , Virulence Factors/metabolism , Virulence Factors/genetics , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/metabolism , Biofilms/drug effects , Biofilms/growth & development , Homoserine/analogs & derivatives
6.
Cell Rep ; 43(4): 114004, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38522070

ABSTRACT

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.


Subject(s)
Caspase 3 , Gasdermins , Intracellular Signaling Peptides and Proteins , Macrophages , Phosphate-Binding Proteins , Pyroptosis , Pyroptosis/drug effects , Phosphate-Binding Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Intracellular Signaling Peptides and Proteins/metabolism , Caspase 3/metabolism , Humans , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/metabolism , Caspases/metabolism , Lipopolysaccharides/pharmacology , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Caspases, Initiator/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Interleukin-1beta/metabolism
7.
J Med Microbiol ; 71(1)2022 Jan.
Article in English | MEDLINE | ID: mdl-35037615

ABSTRACT

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.


Subject(s)
Campylobacter Infections , Campylobacter , Dysentery , Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Campylobacter/genetics , Campylobacter/pathogenicity , Campylobacter Infections/epidemiology , Child, Preschool , Diarrhea/epidemiology , Diarrhea/microbiology , Dysentery/epidemiology , Dysentery/microbiology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/epidemiology , Feces , Humans , India , Multiplex Polymerase Chain Reaction , Prevalence , Real-Time Polymerase Chain Reaction , Shigella/genetics
8.
Mol Microbiol ; 117(1): 86-101, 2022 01.
Article in English | MEDLINE | ID: mdl-34411346

ABSTRACT

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.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Host Factor 1 Protein/metabolism , RNA, Small Untranslated/genetics , Trans-Activators/metabolism , Enterohemorrhagic Escherichia coli/growth & development , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , Humans , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Biosynthesis , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Activators/genetics , Type III Secretion Systems , Virulence
9.
Toxins (Basel) ; 13(10)2021 09 26.
Article in English | MEDLINE | ID: mdl-34678979

ABSTRACT

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.


Subject(s)
Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/pathogenicity , Virulence Factors/genetics , Animals , Enterohemorrhagic Escherichia coli/classification , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Europe , Humans , Phylogeny , Serogroup , Shiga-Toxigenic Escherichia coli/classification , Virulence , Whole Genome Sequencing
10.
Nucleic Acids Res ; 49(19): 10988-11004, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34591974

ABSTRACT

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.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Small Untranslated/genetics , Transcription Factors/genetics , Virulence Factors/genetics , Animals , Base Pairing , Base Sequence , Colon/metabolism , Colon/microbiology , Endoribonucleases/chemistry , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/pathology , Escherichia coli Proteins/metabolism , Ethanolamine/metabolism , Female , Gene Expression Regulation, Bacterial , Genetic Fitness , HeLa Cells , Host Microbial Interactions/genetics , Humans , Mice , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Small Untranslated/metabolism , Sequence Analysis, RNA , Transcription Factors/metabolism , Virulence , Virulence Factors/metabolism
11.
Genetics ; 217(1): 1-17, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33683370

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Escherichia coli Infections/genetics , Immunity, Innate , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Intestines/microbiology , Muscle Proteins/genetics , Muscle Proteins/metabolism
12.
Ren Fail ; 43(1): 382-387, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33641616

ABSTRACT

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.


Subject(s)
Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/microbiology , Insulin-Like Growth Factor Binding Protein 2/blood , Adolescent , Adult , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Escherichia coli Infections/complications , Female , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , Male , ROC Curve , Severity of Illness Index , Young Adult
13.
PLoS One ; 16(2): e0245470, 2021.
Article in English | MEDLINE | ID: mdl-33539359

ABSTRACT

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.


Subject(s)
Diarrhea/epidemiology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/epidemiology , Acute Disease , Adolescent , Adult , Aged , Child , Child, Preschool , Cross-Sectional Studies , Diarrhea/microbiology , Enterohemorrhagic Escherichia coli/isolation & purification , Enteropathogenic Escherichia coli/isolation & purification , Escherichia coli Infections/microbiology , Feces/microbiology , Female , Genes, Bacterial , Humans , Infant , Infant, Newborn , Iran/epidemiology , Male , Middle Aged , Polymerase Chain Reaction , Prevalence , Seasons , Virulence/genetics , Young Adult
14.
Mol Microbiol ; 116(1): 168-183, 2021 07.
Article in English | MEDLINE | ID: mdl-33567149

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Caenorhabditis elegans/microbiology , Enterohemorrhagic Escherichia coli/pathogenicity , Shiga Toxin 1/biosynthesis , Trans-Activators/genetics , Virulence Factors/biosynthesis , Animals , Bacterial Proteins/metabolism , Digestive System/microbiology , Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Promoter Regions, Genetic/genetics , Shiga Toxin 1/genetics , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Type III Secretion Systems/biosynthesis , Type III Secretion Systems/genetics , Virulence Factors/genetics
15.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468701

ABSTRACT

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.


Subject(s)
Arachidonic Acid/metabolism , Enterohemorrhagic Escherichia coli/physiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Fatty Acids/metabolism , Host-Pathogen Interactions , Arachidonic Acid/pharmacology , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Models, Biological , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence/genetics , Virulence Factors/genetics
16.
Nat Commun ; 12(1): 90, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397943

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , Enterohemorrhagic Escherichia coli/physiology , Host-Pathogen Interactions , Microvilli/microbiology , Microvilli/pathology , Actins/metabolism , Animals , Caco-2 Cells , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/ultrastructure , Carbohydrate Epimerases/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Formins , Humans , Intestines/microbiology , Microvilli/metabolism , Phosphorylation , Phosphothreonine/metabolism , Virulence
17.
J Biol Chem ; 296: 100299, 2021.
Article in English | MEDLINE | ID: mdl-33460651

ABSTRACT

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.


Subject(s)
Galactosyltransferases/chemistry , Globosides/chemistry , Shiga Toxin 1/chemistry , Trihexosylceramides/chemistry , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Animals , Binding Sites , CHO Cells , Carbohydrate Sequence , Cricetulus , Enterohemorrhagic Escherichia coli/chemistry , Enterohemorrhagic Escherichia coli/pathogenicity , Galactose/chemistry , Galactose/metabolism , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gene Expression , Globosides/biosynthesis , Globosides/metabolism , Glucose/chemistry , Glucose/metabolism , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga Toxin 1/metabolism , Shiga Toxin 2/chemistry , Shiga Toxin 2/metabolism , Trihexosylceramides/biosynthesis
18.
Curr Genet ; 67(2): 221-224, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33219834

ABSTRACT

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.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Infections/genetics , Genetic Heterogeneity , Serine/genetics , Adaptation, Physiological/genetics , Anti-Bacterial Agents/pharmacology , Drug Tolerance/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Humans , Mutation/genetics
19.
Infect Immun ; 89(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33229368

ABSTRACT

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.


Subject(s)
Antimicrobial Cationic Peptides/drug effects , Bile Acids and Salts/pharmacology , Bile Acids and Salts/therapeutic use , Disease Resistance/drug effects , Enterohemorrhagic Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Virulence/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/pathology , Humans
20.
Front Immunol ; 11: 561337, 2020.
Article in English | MEDLINE | ID: mdl-33329523

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Host Microbial Interactions/immunology , Membrane Proteins/immunology , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Escherichia coli Infections/microbiology , Gene Knockdown Techniques , Immunity, Innate , Membrane Proteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...