Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 92
1.
ACS Infect Dis ; 10(3): 988-999, 2024 Mar 08.
Article En | MEDLINE | ID: mdl-38317607

Escherichia coli continues to be the predominant Gram-negative pathogen causing neonatal meningitis worldwide. Inflammatory mediators have been implicated in the pathogenesis of meningitis and are key therapeutic targets. The role of interleukin-22 (IL-22) in various diseases is diverse, with both protective and pathogenic effects. However, little is understood about the mechanisms underlying the damaging effects of IL-22 on the blood-brain barrier (BBB) in E. coli meningitis. We observed that meningitic E. coli infection induced IL-22 expression in the serum and brain of mice. The tight junction proteins (TJPs) components ZO-1, Occludin, and Claudin-5 were degraded in the mouse brain and human brain microvascular endothelial cells (hBMEC) following IL-22 administration. Moreover, the meningitic E. coli-caused increase in BBB permeability in wild-type mice was restored by knocking out IL-22. Mechanistically, IL-22 activated the STAT3-VEGFA signaling cascade in E. coli meningitis, thus eliciting the degradation of TJPs to induce BBB disruption. Our data indicated that IL-22 is an essential host accomplice during E. coli-caused BBB disruption and could be targeted for the therapy of bacterial meningitis.


Escherichia coli Infections , Meningitis, Bacterial , Meningitis, Escherichia coli , Humans , Mice , Animals , Blood-Brain Barrier , Meningitis, Escherichia coli/metabolism , Meningitis, Escherichia coli/microbiology , Meningitis, Escherichia coli/pathology , Escherichia coli/metabolism , Endothelial Cells , Interleukin-22 , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
2.
Int J Mol Sci ; 24(12)2023 Jun 08.
Article En | MEDLINE | ID: mdl-37373064

Bacterial meningitis is a devastating disease occurring worldwide, with up to half of survivors left with permanent neurological sequelae. Neonatal meningitis-causing Escherichia coli (NMEC) is the most common Gram-negative bacillary organism that causes meningitis, particularly during the neonatal period. Here, RNA-seq transcriptional profiles of microglia in response to NMEC infection show that microglia are activated to produce inflammatory factors. In addition, we found that the secretion of inflammatory factors is a double-edged sword that promotes polymorphonuclear neutrophil (PMN) recruitment to the brain to clear the pathogens but, at the same time, induces neuronal damage, which may be related to the neurological sequelae. New neuroprotective therapeutic strategies must be developed for the treatment of acute bacterial meningitis. We found that transforming growth factor-ß (TGF-ß) may be a strong candidate in the treatment of acute bacterial meningitis, as it shows a therapeutic effect on bacterial-meningitis-induced brain damage. Prevention of disease and early initiation of the appropriate treatment in patients with suspected or proven bacterial meningitis are the key factors in reducing morbidity and mortality. Novel antibiotic and adjuvant treatment strategies must be developed, and the main goal for new therapies will be dampening the inflammatory response. Based on this view, our findings may help develop novel strategies for bacterial meningitis treatment.


Escherichia coli Infections , Meningitis, Bacterial , Meningitis, Escherichia coli , Infant, Newborn , Humans , Microglia , Meningitis, Escherichia coli/microbiology , Meningitis, Bacterial/microbiology , Escherichia coli Infections/microbiology , Escherichia coli
3.
FASEB J ; 36(3): e22197, 2022 03.
Article En | MEDLINE | ID: mdl-35147989

Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.


Escherichia coli Proteins/toxicity , Hemolysin Proteins/toxicity , Inflammasomes/metabolism , Meningitis, Escherichia coli/metabolism , Neonatal Sepsis/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Cells, Cultured , Escherichia coli K12 , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Macrophages/metabolism , Meningitis, Escherichia coli/microbiology , Mice , Mice, Inbred C57BL , Neonatal Sepsis/microbiology , Receptors, Purinergic P2X/genetics
4.
Front Immunol ; 12: 745854, 2021.
Article En | MEDLINE | ID: mdl-34721415

Alpha 7 nicotinic acetylcholine receptor (α7 nAChR) is critical for the pathogenesis of Escherichia coli (E. coli) K1 meningitis, a severe central nervous system infection of the neonates. However, little is known about how E. coli K1 manipulates α7 nAChR signaling. Here, through employing immortalized cell lines, animal models, and human transcriptional analysis, we showed that E. coli K1 infection triggers releasing of secreted Ly6/Plaur domain containing 1 (SLURP1), an endogenous α7 nAChR ligand. Exogenous supplement of SLURP1, combined with SLURP1 knockdown or overexpression cell lines, showed that SLURP1 is required for E. coli K1 invasion and neutrophils migrating across the blood-brain barrier (BBB). Furthermore, we found that SLURP1 is required for E. coli K1-induced α7 nAChR activation. Finally, the promoting effects of SLURP1 on the pathogenesis of E. coli K1 meningitis was significantly abolished in the α7 nAChR knockout mice. These results reveal that E. coli K1 exploits SLURP1 to activate α7 nAChR and facilitate its pathogenesis, and blocking SLURP1-α7 nAChR interaction might represent a novel therapeutic strategy for E. coli K1 meningitis.


Antigens, Ly/physiology , Blood-Brain Barrier , Escherichia coli Infections/microbiology , Escherichia coli/physiology , Meningitis, Escherichia coli/physiopathology , Urokinase-Type Plasminogen Activator/physiology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Antigens, Ly/genetics , Cell Line , Cerebrospinal Fluid/microbiology , Endothelial Cells/microbiology , Escherichia coli/isolation & purification , Hippocampus/metabolism , Host-Pathogen Interactions , Humans , Infant, Newborn , Memantine/pharmacology , Meningitis, Escherichia coli/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/physiology , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Urokinase-Type Plasminogen Activator/genetics , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/deficiency
5.
Microbiology (Reading) ; 167(10)2021 10.
Article En | MEDLINE | ID: mdl-34623236

Appropriate interpretation of environmental signals facilitates niche specificity in pathogenic bacteria. However, the responses of niche-specific pathogens to common host signals are poorly understood. d-Serine (d-ser) is a toxic metabolite present in highly variable concentrations at different colonization sites within the human host that we previously found is capable of inducing changes in gene expression. In this study, we made the striking observation that the global transcriptional response of three Escherichia coli pathotypes - enterohaemorrhagic E. coli (EHEC), uropathogenic E. coli (UPEC) and neonatal meningitis-associated E. coli (NMEC) - to d-ser was highly distinct. In fact, we identified no single differentially expressed gene common to all three strains. We observed the induction of ribosome-associated genes in extraintestinal pathogens UPEC and NMEC only, and the induction of purine metabolism genes in gut-restricted EHEC, and UPEC indicating distinct transcriptional responses to a common signal. UPEC and NMEC encode dsdCXA - a genetic locus required for detoxification and hence normal growth in the presence of d-ser. Specific transcriptional responses were induced in strains accumulating d-ser (WT EHEC and UPEC/NMEC mutants lacking the d-ser-responsive transcriptional activator DsdC), corroborating the notion that d-ser is an unfavourable metabolite if not metabolized. Importantly, many of the UPEC-associated transcriptome alterations correlate with published data on the urinary transcriptome, supporting the hypothesis that d-ser sensing forms a key part of urinary niche adaptation in this pathotype. Collectively, our results demonstrate distinct pleiotropic responses to a common metabolite in diverse E. coli pathotypes, with important implications for niche selectivity.


Escherichia coli/genetics , Serine/metabolism , Transcriptome , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Meningitis, Escherichia coli/microbiology , Species Specificity , Urinary Tract Infections/microbiology
6.
Microb Pathog ; 160: 105199, 2021 Nov.
Article En | MEDLINE | ID: mdl-34560248

Neonatal bacterial meningitis is a life-threatening disease in newborns, and neonatal meningitis Escherichia coli (NMEC) is the second most frequent bacteria causing this disease worldwide. In order to further understand the characteristics of this pathogen, an E. coli isolate W224 N from newborns with meningitis was sequenced for detailed genetic characterization and the virulence was tested by a series of phenotypic experiments. W224 N has a circular chromosome and three plasmids. It belongs to ST95 and the serotype is O18:H7. Comparative genomic analysis showed that W224 N was closely related to E. coli neonatal meningitis isolates RS218 and NMEC O18. There are 11 genomic islands in W224 N and most of the GIs are specific to W224 N. W224 N has most of the virulence factors other neonatal meningitis isolates have. The virulence genes located both on the genome and plasmid. At the same time, we found a virulence factor cdiA only present in W224 N but absent in the other five genomes analyzed. In vitro experiment showed that W224 N has strong serum resistance ability, low biofilm formation ability and high flagellar motility. It also has a very strong toxicity to mice and amoeba. The whole genome as well as in vitro and in vivo experiments showed that W224 N is a high virulent strain. The results can help us better learn about the pathogenicity of neonatal meningitis E. coli.


Escherichia coli Proteins , Escherichia coli/genetics , Genome, Bacterial , Meningitis, Escherichia coli , Animals , Escherichia coli/pathogenicity , Membrane Proteins , Meningitis, Escherichia coli/microbiology , Mice , Virulence , Virulence Factors/genetics
7.
J Vis Exp ; (170)2021 04 20.
Article En | MEDLINE | ID: mdl-33970148

Escherichia coli (E. coli) is the most common Gram-negative bacteria causing neonatal meningitis. The occurrence of bacteremia and bacterial penetration through the blood-brain barrier are indispensable steps for the development of E. coli meningitis. Reactive oxygen species (ROS) represent the major bactericidal mechanisms of neutrophils to destroy the invaded pathogens. In this protocol, the time-dependent intracellular ROS production in neutrophils infected with meningitic E. coli was quantified using fluorescent ROS probes detected by a real-time fluorescence microplate reader. This method may also be applied to the assessment of ROS production in mammalian cells during pathogen-host interactions.


Meningitis, Escherichia coli/microbiology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Humans
8.
Cell Microbiol ; 22(10): e13231, 2020 10.
Article En | MEDLINE | ID: mdl-32447809

Escherichia coli is the most common Gram-negative bacillary organism causing neonatal meningitis. Escherichia coli meningitis remains an important cause of mortality and morbidity, but the pathogenesis of E. coli penetration of the blood-brain barrier remains incompletely understood. Escherichia coli entry into the brain occurs in the meningeal and cortex capillaries, not in the choroid plexus, and exploits epidermal growth factor receptor (EGFR) and cysteinyl leukotrienes (CysLTs) for invasion of the blood-brain barrier. The present study examined whether EGFR and CysLTs are inter-related in their contribution to E. coli invasion of the blood-brain barrier and whether counteracting EGFR and CysLTs is a beneficial adjunct to antibiotic therapy of E. coli meningitis. We showed that (a) meningitis isolates of E. coli exploit EGFR and CysLTs for invasion of the blood-brain barrier, (b) the contribution of EGFR is upstream of that of CysLTs, and (c) counteracting EGFR and CysLTs as an adjunctive therapy improved the outcome (survival, neuronal injury and memory impairment) of animals with E. coli meningitis. These findings suggest that investigation of host factors contributing to E. coli invasion of the blood-brain barrier will help in enhancing the pathogenesis and development of new therapeutic targets for E. coli meningitis in the era of increasing resistance to conventional antibiotics.


Acetates/therapeutic use , Blood-Brain Barrier/microbiology , Cyclopropanes/therapeutic use , Cysteine/metabolism , ErbB Receptors/metabolism , Escherichia coli/pathogenicity , Gefitinib/therapeutic use , Leukotrienes/metabolism , Meningitis, Escherichia coli/microbiology , Quinolines/therapeutic use , Sulfides/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Blood-Brain Barrier/physiopathology , Brain/blood supply , Ceftriaxone/therapeutic use , Cells, Cultured , Drug Therapy, Combination , Endothelial Cells , ErbB Receptors/antagonists & inhibitors , Female , Humans , Infant, Newborn , Leukotriene Antagonists/therapeutic use , Male , Meningitis, Escherichia coli/drug therapy , Mice , Permeability , Phospholipases A2, Cytosolic/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
9.
BMC Microbiol ; 19(1): 298, 2019 12 17.
Article En | MEDLINE | ID: mdl-31847813

BACKGROUND: The neonatal meningitis E. coli (NMEC) strain S88 carries a ColV plasmid named pS88 which is involved in meningeal virulence. Transcriptional analysis of pS88 in human serum revealed a strong upregulation of an ORF of unknown function: shiF, which is adjacent to the operon encoding the siderophore aerobactin. The aim of this work is to investigate the role of shiF in aerobactin production in strain S88. RESULTS: Study of the prevalence of shiF and aerobactin operon in a collection of 100 extra-intestinal pathogenic E. coli strains (ExPEC) and 50 whole genome-sequenced E. coli strains revealed the colocalization of these two genes for 98% of the aerobactin positive strains. We used Datsenko and Wanner's method to delete shiF in two S88 mutants. A cross-feeding assay showed that these mutants were able to excrete aerobactin meaning that shiF is dispensable for aerobactin excretion. Our growth assays revealed that the shiF-deleted mutants grew significantly slower than the wild-type strain S88 in iron-depleted medium with a decrease of maximum growth rates of 23 and 28% (p < 0.05). Using Liquid Chromatography-Mass Spectrometry, we identified and quantified siderophores in the supernatants of S88 and its shiF deleted mutants after growth in iron-depleted medium and found that these mutants secreted significantly less aerobactin than S88 (- 52% and - 49%, p < 0.001). CONCLUSIONS: ShiF is physically and functionally linked to aerobactin. It provides an advantage to E. coli S88 under iron-limiting conditions by increasing aerobactin secretion and may thus act as an auxiliary virulence factor.


Bacterial Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Hydroxamic Acids/metabolism , Iron/metabolism , Siderophores/metabolism , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Gene Expression Profiling , Humans , Meningitis, Escherichia coli/blood , Meningitis, Escherichia coli/microbiology , Operon , Plasmids/genetics , Virulence , Virulence Factors/genetics , Whole Genome Sequencing
10.
Sci Rep ; 9(1): 2600, 2019 02 22.
Article En | MEDLINE | ID: mdl-30796316

Pathoadaptive mutations linked to c-di-GMP signalling were investigated in neonatal meningitis-causing Escherichia coli (NMEC). The results indicated that NMEC strains deficient in RpoS (the global stress regulator) maintained remarkably low levels of c-di-GMP, a major bacterial sessility-motility switch. Deletion of ycgG2, shown here to encode a YcgG allozyme with c-di-GMP phosphodiesterase activity, and the restoration of RpoS led to a decrease in S-fimbriae, robustly produced in artificial urine, hinting that the urinary tract could serve as a habitat for NMEC. We showed that NMEC were skilled in aerobic citrate utilization in the presence of glucose, a property that normally does not exist in E. coli. Our data suggest that this metabolic novelty is a property of extraintestinal pathogenic E. coli since we reconstituted this ability in E. coli UTI89 (a cystitis isolate) via deactivation rpoS; additionally, a set of pyelonephritis E. coli isolates were shown here to aerobically use citrate in the presence of glucose. We found that the main reason for this metabolic capability is RpoS inactivation leading to the production of the citrate transporter CitT, exploited by NMEC for ferric citrate uptake dependent on YcgG2 (an allozyme with c-di-GMP phosphodiesterase activity).


Bacterial Proteins/genetics , Cyclic GMP/analogs & derivatives , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Meningitis, Escherichia coli/microbiology , Sigma Factor/genetics , Citric Acid/metabolism , Cyclic GMP/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Glucose/metabolism , Organic Anion Transporters/metabolism
11.
BMC Microbiol ; 19(1): 17, 2019 01 17.
Article En | MEDLINE | ID: mdl-30654756

BACKGROUND: To describe the temporal dynamics, molecular characterization, clinical and ex vivo virulence of emerging O1:K1 neonatal meningitis Escherichia coli (NMEC) strains of Sequence Type complex (STc) 95 in France. The national reference center collected NMEC strains and performed whole genome sequencing (WGS) of O1:K1 STc95 NMEC strains for phylogenetic and virulence genes content analysis. Data on the clinical and biological features of patients were also collected. Ex vivo virulence was assessed using the Dictyostelium discoideum amoeba model. RESULTS: Among 250 NMEC strains collected between 1998 and 2015, 38 belonged to O1:K1 STc95. This clonal complex was the most frequently collected after 2004, representing up to 25% of NMEC strains in France. Phylogenetic analysis demonstrated that most (74%) belonged to a cluster designated D-1, characterized by the adhesin FimH30. There is no clinical data to suggest that this cluster is more pathogenic than its counterparts, although it is highly predominant and harbors a large repertoire of extraintestinal virulence factors, including a pS88-like plasmid. Ex vivo virulence model showed that this cluster was generally less virulent than STc95 reference strains of O45S88:H7 and O18:H7 serotypes. However, the model showed differences between several subclones, although they harbor the same known virulence determinants. CONCLUSIONS: The emerging clonal group O1:K1 STc95 of NMEC strains is mainly composed of a cluster with many virulence factors but of only moderate virulence. Whether its emergence is due to its ability to colonize the gut thanks to FimH30 or pS88-like plasmid remains to be determined.


Escherichia coli/genetics , Genome, Bacterial/genetics , Infant, Newborn, Diseases/microbiology , Meningitis, Escherichia coli/microbiology , Whole Genome Sequencing , Escherichia coli/classification , Escherichia coli/pathogenicity , France , Humans , Infant, Newborn , Models, Genetic , Phylogeny , Virulence/genetics
13.
J Infect Dis ; 219(3): 470-479, 2019 01 09.
Article En | MEDLINE | ID: mdl-30202861

FimH-mediated bacterial invasion and polymorphonuclear neutrophil (PMN) transmigration across human brain microvascular endothelial cells (HBMECs) are required for the pathogenesis of Escherichia coli meningitis. However, the underlying mechanism remains unclear. This study demonstrated that the TnphoA mutant (22A33) and FimH-knockout mutant (ΔFimH) of E coli strain E44, which resulted in inactivation of FimH, were less invasive and less effective in promoting PMN transmigration than their wild-type strain. FimH protein induced PMN transmigration, whereas calmodulin inhibitor significantly blocked this effect. Moreover, immunofluorescence and co-immunoprecipitation analysis indicated that colocalized CD48 and α7 nAChR formed a complex on the surface of HBMECs that is associated with increased cofilin dephosphorylation, which could be remarkably enhanced by FimH+ E44. Our study concluded that FimH-induced E coli K1 invasion and PMN migration across HBMECs may be mediated by the CD48-α7nAChR complex in lipid rafts of HBMEC via Ca2+ signaling and cofilin dephosphorylation.


Adhesins, Escherichia coli/metabolism , Brain/microbiology , CD48 Antigen/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Neutrophils/microbiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Adhesins, Escherichia coli/genetics , Calcium/metabolism , Cell Movement , Escherichia coli/pathogenicity , Fimbriae Proteins/genetics , Gene Knockdown Techniques , Humans , Membrane Microdomains , Meningitis, Escherichia coli/microbiology , Meningitis, Escherichia coli/pathology , Signal Transduction , Virulence Factors/genetics , Virulence Factors/metabolism
14.
BMC Infect Dis ; 18(1): 544, 2018 Nov 01.
Article En | MEDLINE | ID: mdl-30497396

BACKGROUND: Classification of pathogenic Escherichia coli (E. coli) has traditionally relied on detecting specific virulence associated genes (VAGs) or combinations thereof. For E. coli isolated from faecal samples, the presence of specific genes associated with different intestinal pathogenic pathovars will determine their classification and further course of action. However, the E. coli genome is not a static entity, and hybrid strains are emerging that cross the pathovar definitions. Hybrid strains may show gene contents previously associated with several distinct pathovars making the correct diagnostic classification difficult. We extended the analysis of routinely submitted faecal isolates to include known virulence associated genes that are usually not examined in faecal isolates to detect the frequency of possible hybrid strains. METHODS: From September 2012 to February 2013, 168 faecal isolates of E. coli routinely submitted to the Norwegian Institute of Public Health (NIPH) from clinical microbiological laboratories throughout Norway were analysed for 33 VAGs using multiplex-PCR, including factors associated with extraintestinal pathogenic E. coli (ExPEC) strains. The strains were further typed by Multiple Locus Variable-Number Tandem-Repeat Analysis (MLVA), and the phylogenetic grouping was determined. One isolate from the study was selected for whole genome sequencing (WGS) with a combination of Oxford Nanopore's MinION and Illumina's MiSeq. RESULTS: The analysis showed a surprisingly high number of strains carrying ExPEC associated VAGs and strains carrying a combination of both intestinal pathogenic E. coli (IPEC) and ExPEC VAGs. In particular, 93.5% (101/108) of isolates classified as belonging to an IPEC pathovar additionally carried ExPEC VAGs. WGS analysis of a selected hybrid strain revealed that it could, with present classification criteria, be classified as belonging to all of the Enteropathogenic Escherichia coli (EPEC), Uropathogenic Escherichia coli (UPEC), Neonatal meningitis Escherichia coli (NMEC) and Avian pathogenic Escherichia coli (APEC) pathovars. CONCLUSION: Hybrid ExPEC/IPEC E. coli strains were found at a very high frequency in faecal samples and were in fact the predominant species present. A sequenced hybrid isolate was confirmed to be a cross-pathovar strain possessing recognised hallmarks of several pathovars, and a genome heavily influenced by horizontal gene transfer.


Enteropathogenic Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Extraintestinal Pathogenic Escherichia coli/isolation & purification , Feces/microbiology , Virulence Factors/analysis , Animals , Enteropathogenic Escherichia coli/genetics , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Extraintestinal Pathogenic Escherichia coli/genetics , Feces/chemistry , Humans , Incidence , Intestines/microbiology , Meningitis, Escherichia coli/epidemiology , Meningitis, Escherichia coli/microbiology , Norway/epidemiology , Phylogeny , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/isolation & purification , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/isolation & purification
15.
Intern Med ; 57(21): 3199-3204, 2018 Nov 01.
Article En | MEDLINE | ID: mdl-29877280

A 78-year-old man had a fever and exhibited disordered consciousness, which led to his transportation to our hospital. On arrival, he exhibited discharge from the ear. Because extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli was detected in the ear discharge and cerebrospinal fluid specimens, it was inferred to be the causal bacteria. Pulsed-field gel electrophoresis indicated the same ESBL-producing E. coli pattern in the patient's ear discharge, external auditory canal granulation, cerebrospinal fluid, and stool, indicating their common molecular epidemiological origin. Although ESBL-producing E. coli is an extremely rare cause of bacterial meningitis, it should be considered as a potential causal bacteria for community-acquired meningitis.


Cholesteatoma, Middle Ear/microbiology , Community-Acquired Infections/diagnosis , Escherichia coli Infections/microbiology , Escherichia coli/isolation & purification , Meningitis, Escherichia coli/diagnosis , Otitis Media/microbiology , Aged , Cholesteatoma, Middle Ear/complications , Community-Acquired Infections/microbiology , Electrophoresis, Gel, Pulsed-Field , Escherichia coli/enzymology , Humans , Male , Meningitis, Escherichia coli/microbiology , Otitis Media/complications , beta-Lactamases/biosynthesis
16.
BMJ Case Rep ; 20172017 Dec 20.
Article En | MEDLINE | ID: mdl-29269368

Intrathecal baclofen (ITB) delivery via an implanted pump is frequently used for the treatment of spasticity. This is an effective and safe neurosurgical and pharmacological intervention associated with an improvement in patient quality of life. There is, however, a risk of device-related infection. We present a patient with pump-site infection and Escherichia coli meningitis secondary to transcolonic perforation of an intrathecal baclofen pump catheter. While this is rare, we review the intraoperative precautions and best practices that should be taken to prevent and manage this unusual complication.


Anti-Bacterial Agents/therapeutic use , Baclofen/administration & dosage , Catheterization/adverse effects , Catheters, Indwelling/adverse effects , Infusion Pumps, Implantable/adverse effects , Intestinal Perforation/microbiology , Meningitis, Escherichia coli/microbiology , Multiple Sclerosis/drug therapy , Muscle Relaxants, Central/administration & dosage , Catheters, Indwelling/microbiology , Device Removal , Disabled Persons , Female , Humans , Iatrogenic Disease , Infusion Pumps, Implantable/microbiology , Infusions, Spinal/adverse effects , Intestinal Perforation/etiology , Meningitis, Escherichia coli/etiology , Microbial Sensitivity Tests , Middle Aged , Recurrence , Treatment Outcome
17.
Avian Dis ; 61(1): 135-138, 2017 Mar.
Article En | MEDLINE | ID: mdl-28301230

Three outbreaks of colibacillosis have occurred in chicks during the quarantine period after importation to Japan. All three were derived from three different countries without epidemiologic relevance. Some birds from each infected flock were examined pathologically and bacteriologically. The characteristic histologic finding common to all three cases was severe bacterial meningitis in the central nervous system. Pericarditis, perihepatitis, and omphalitis with bacterial colonies were also observed. The bacterial colonies observed histologically were immunohistochemically positive for Escherichia coli antigens. Escherichia coli was isolated from the organ samples from each outbreak. At least two E. coli isolates were serotyped as O18 and O161, which differed from the popular serotypes in Japan. These results suggest that avian pathogenic E. coli of uncommon serotypes can be imported from outside countries by infected chicks. Colibacillosis should be included in the differential diagnosis when meningitis is histologically observed in chicks.


Escherichia coli/isolation & purification , Meningitis, Escherichia coli/veterinary , Poultry Diseases/microbiology , Animals , Chickens/virology , Japan , Meningitis, Escherichia coli/economics , Meningitis, Escherichia coli/microbiology , Poultry Diseases/economics
18.
Toxins (Basel) ; 9(2)2017 01 26.
Article En | MEDLINE | ID: mdl-28134751

Enterohemorrhagic Escherichia coli (EHEC) is the most common cause of hemorrhagic colitis and hemolytic uremic syndrome in human patients, with brain damage and dysfunction the main cause of acute death. We evaluated the efficacy of urtoxazumab (TMA-15, Teijin Pharma Limited), a humanized monoclonal antibody against Shiga toxin (Stx) 2 for the prevention of brain damage, dysfunction, and death in a piglet EHEC infection model. Forty-five neonatal gnotobiotic piglets were inoculated orally with 3 × 108 colony-forming units of EHEC O157:H7 strain EDL933 (Stx1⁺, Stx2⁺) when 22-24 h old. At 24 h post-inoculation, piglets were intraperitoneally administered placebo or TMA-15 (0.3, 1.0 or 3.0 mg/kg body weight). Compared to placebo (n = 10), TMA-15 (n = 35) yielded a significantly greater probability of survival, length of survival, and weight gain (p <0.05). The efficacy of TMA-15 against brain lesions and death was 62.9% (p = 0.0004) and 71.4% (p = 0.0004), respectively. These results suggest that TMA-15 may potentially prevent or reduce vascular necrosis and infarction of the brain attributable to Stx2 in human patients acutely infected with EHEC. However, we do not infer that TMA-15 treatment will completely protect human patients infected with EHEC O157:H7 strains that produce both Stx1 and Stx2.


Antibodies, Monoclonal, Humanized/pharmacology , Brain Infarction/prevention & control , Brain/drug effects , Escherichia coli O157/drug effects , Hemolytic-Uremic Syndrome/prevention & control , Meningitis, Escherichia coli/prevention & control , Shiga Toxin 2/antagonists & inhibitors , Animals , Animals, Newborn , Brain/immunology , Brain/microbiology , Brain/pathology , Brain Infarction/immunology , Brain Infarction/microbiology , Diarrhea/drug therapy , Diarrhea/immunology , Diarrhea/microbiology , Disease Models, Animal , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Germ-Free Life , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Meningitis, Escherichia coli/immunology , Meningitis, Escherichia coli/microbiology , Necrosis , Severity of Illness Index , Shiga Toxin 2/immunology , Sus scrofa , Time Factors
19.
Infez Med ; 24(4): 293-298, 2016 12 01.
Article En | MEDLINE | ID: mdl-28011964

Acinetobacter baumannii and Enterobacteriaceae are two pathogens responsible for postneurosurgical meningitis. The aim of this retrospective study was to evaluate the factors that influenced the outcomes in patients with postneurosurgical meningitis caused by A. baumannii and Enterobacteriaceae. Patients with post-surgical meningitis were identified from infection control committee charts between 2007 and 2015. Subjects over 16 years old who had positive cerebral spinal fluid cultures for A. baumannii or Enterobacteriaceae were enrolled in the study. Clinical and laboratory data for 30 patients with A. baumannii meningitis were compared with those of 12 patients with Enterobacteriaceae meningitis. The mean age of patients was 51.9 years and 57.1% were male. Eleven patients had comorbidities, the most common being diabetes mellitus. Most patients were due to intracranial haemorrhage (78.6%). The rate of the patients who received an appropriate antimicrobial therapy was 35.7%, and the crude mortality rate was 64.3%. In univariate analysis, previous antibiotic use, an infection before meningitis and mechanical ventilation had an increased risk of A. baumannii meningitis. Moreover, intrathecal antimicrobial use, inappropriate empirical antimicrobial use, antimicrobial resistance and alanine aminotransferase elevation were significantly higher in patients with A. baumannii meningitis than in those with Enterobacteriaceae meningitis. Antimicrobial use before meningitis (8.84 times) and mechanical ventilation (7.28 times) resulted in an increased risk of A. baumannii meningitis. None of the results affected 30-day mortality. Avoidance of unnecessarily prolonged antimicrobial usage may help to prevent a selection of A. baumannii.


Acinetobacter Infections/complications , Acinetobacter baumannii/isolation & purification , Anti-Bacterial Agents/therapeutic use , Enterobacteriaceae Infections/complications , Enterobacteriaceae/isolation & purification , Meningitis, Bacterial/drug therapy , Meningitis, Bacterial/microbiology , Adolescent , Adult , Female , Humans , Male , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Escherichia coli/drug therapy , Meningitis, Escherichia coli/microbiology , Middle Aged , Retrospective Studies , Treatment Outcome
20.
EcoSal Plus ; 7(1)2016 05.
Article En | MEDLINE | ID: mdl-27223820

Escherichia coli is the most common Gram-negative bacillary organism causing meningitis, and E. coli meningitis continues to be an important cause of mortality and morbidity throughout the world. Our incomplete knowledge of its pathogenesis contributes to such mortality and morbidity. Recent reports of E. coli strains producing CTX-M-type or TEM-type extended-spectrum ß-lactamases create a challenge. Studies using in vitro and in vivo models of the blood-brain barrier have shown that E. coli meningitis follows a high degree of bacteremia and invasion of the blood-brain barrier. E. coli invasion of the blood-brain barrier, the essential step in the development of E. coli meningitis, requires specific microbial and host factors as well as microbe- and host-specific signaling molecules. Blockade of such microbial and host factors contributing to E. coli invasion of the blood-brain barrier is shown to be efficient in preventing E. coli penetration into the brain. The basis for requiring a high degree of bacteremia for E. coli penetration of the blood-brain barrier, however, remains unclear. Continued investigation on the microbial and host factors contributing to a high degree of bacteremia and E. coli invasion of the blood-brain barrier is likely to identify new targets for prevention and therapy of E. coli meningitis.


Blood-Brain Barrier/microbiology , Brain/microbiology , Escherichia coli/pathogenicity , Meningitis, Escherichia coli/microbiology , Animals , Bacteremia/microbiology , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Host-Pathogen Interactions , Humans , Meningitis, Escherichia coli/prevention & control , Meningitis, Escherichia coli/therapy , Mice , Models, Animal , Rats , beta-Lactamases/biosynthesis
...