Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 36(3): e22197, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35147989

RESUMO

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.


Assuntos
Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/toxicidade , Inflamassomos/metabolismo , Meningite devida a Escherichia coli/metabolismo , Sepse Neonatal/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animais , Células Cultivadas , Escherichia coli K12 , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Macrófagos/metabolismo , Meningite devida a Escherichia coli/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Sepse Neonatal/microbiologia , Receptores Purinérgicos P2X/genética
2.
Int J Mol Sci ; 24(12)2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37373064

RESUMO

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.


Assuntos
Infecções por Escherichia coli , Meningites Bacterianas , Meningite devida a Escherichia coli , Recém-Nascido , Humanos , Microglia , Meningite devida a Escherichia coli/microbiologia , Meningites Bacterianas/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli
3.
Microbiology (Reading) ; 167(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34623236

RESUMO

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.


Assuntos
Escherichia coli/genética , Serina/metabolismo , Transcriptoma , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/isolamento & purificação , Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Meningite devida a Escherichia coli/microbiologia , Especificidade da Espécie , Infecções Urinárias/microbiologia
4.
Microb Pathog ; 160: 105199, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34560248

RESUMO

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.


Assuntos
Proteínas de Escherichia coli , Escherichia coli/genética , Genoma Bacteriano , Meningite devida a Escherichia coli , Animais , Escherichia coli/patogenicidade , Proteínas de Membrana , Meningite devida a Escherichia coli/microbiologia , Camundongos , Virulência , Fatores de Virulência/genética
5.
Cell Microbiol ; 22(10): e13231, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32447809

RESUMO

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.


Assuntos
Acetatos/uso terapêutico , Barreira Hematoencefálica/microbiologia , Ciclopropanos/uso terapêutico , Cisteína/metabolismo , Receptores ErbB/metabolismo , Escherichia coli/patogenicidade , Gefitinibe/uso terapêutico , Leucotrienos/metabolismo , Meningite devida a Escherichia coli/microbiologia , Quinolinas/uso terapêutico , Sulfetos/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Barreira Hematoencefálica/fisiopatologia , Encéfalo/irrigação sanguínea , Ceftriaxona/uso terapêutico , Células Cultivadas , Quimioterapia Combinada , Células Endoteliais , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Recém-Nascido , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Meningite devida a Escherichia coli/tratamento farmacológico , Camundongos , Permeabilidade , Fosfolipases A2 Citosólicas/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
6.
J Infect Dis ; 219(3): 470-479, 2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30202861

RESUMO

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.


Assuntos
Adesinas de Escherichia coli/metabolismo , Encéfalo/microbiologia , Antígeno CD48/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Escherichia coli/metabolismo , Proteínas de Fímbrias/metabolismo , Neutrófilos/microbiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Adesinas de Escherichia coli/genética , Cálcio/metabolismo , Movimento Celular , Escherichia coli/patogenicidade , Proteínas de Fímbrias/genética , Técnicas de Silenciamento de Genes , Humanos , Microdomínios da Membrana , Meningite devida a Escherichia coli/microbiologia , Meningite devida a Escherichia coli/patologia , Transdução de Sinais , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
7.
BMC Microbiol ; 19(1): 298, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31847813

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Escherichia coli Extraintestinal Patogênica/genética , Ácidos Hidroxâmicos/metabolismo , Ferro/metabolismo , Sideróforos/metabolismo , Escherichia coli Extraintestinal Patogênica/patogenicidade , Perfilação da Expressão Gênica , Humanos , Meningite devida a Escherichia coli/sangue , Meningite devida a Escherichia coli/microbiologia , Óperon , Plasmídeos/genética , Virulência , Fatores de Virulência/genética , Sequenciamento Completo do Genoma
8.
BMC Microbiol ; 19(1): 17, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30654756

RESUMO

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.


Assuntos
Escherichia coli/genética , Genoma Bacteriano/genética , Doenças do Recém-Nascido/microbiologia , Meningite devida a Escherichia coli/microbiologia , Sequenciamento Completo do Genoma , Escherichia coli/classificação , Escherichia coli/patogenicidade , França , Humanos , Recém-Nascido , Modelos Genéticos , Filogenia , Virulência/genética
9.
BMC Infect Dis ; 18(1): 544, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30497396

RESUMO

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.


Assuntos
Escherichia coli Enteropatogênica/isolamento & purificação , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli Extraintestinal Patogênica/isolamento & purificação , Fezes/microbiologia , Fatores de Virulência/análise , Animais , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/isolamento & purificação , Escherichia coli Extraintestinal Patogênica/genética , Fezes/química , Humanos , Incidência , Intestinos/microbiologia , Meningite devida a Escherichia coli/epidemiologia , Meningite devida a Escherichia coli/microbiologia , Noruega/epidemiologia , Filogenia , Escherichia coli Uropatogênica/genética , Escherichia coli Uropatogênica/isolamento & purificação , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/isolamento & purificação
10.
J Biol Chem ; 289(45): 30937-49, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25231998

RESUMO

Neonatal meningitis, caused by Escherichia coli K1, is a serious central nervous system disease. We have established that macrophages serve as permissive niches for E. coli K1 to multiply in the host and for attaining a threshold level of bacterial load, which is a prerequisite for the onset of the disease. Here, we demonstrate experimentally that three N-glycans in FcγRIa interact with OmpA of E. coli K1 for binding to and entering the macrophages. Adoptive transfer of FcγRIa(-/-) bone marrow-derived macrophages transfected with FcγRIa into FcγRIa(-/-) newborn mice renders them susceptible to E. coli K1-induced meningitis. In contrast, mice that received bone marrow-derived macrophages transfected with FcγRIa in which N-glycosylation sites 1, 4, and 5 are mutated to alanines exhibit resistance to E. coli K1 infection. Our molecular dynamics and simulation studies predict that N-glycan 5 exhibits strong binding at the barrel site of OmpA formed by loops 3 and 4, whereas N-glycans 1 and 4 interact with loops 1, 3, and 4 of OmpA at tip regions. Molecular modeling data also suggest no role for the IgG binding site in the invasion process. In agreement, experimental mutations in IgG binding site had no effect on the E. coli K1 entry into macrophages in vitro or on the onset of meningitis in newborn mice. Together, this integration of experimental and computational studies reveals how the N-glycans in FcγRIa interact with the OmpA of E. coli K1 for inducing the disease pathogenesis.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Escherichia coli/metabolismo , Macrófagos/microbiologia , Meningite devida a Escherichia coli/microbiologia , Polissacarídeos/química , Receptores de IgG/química , Transferência Adotiva , Animais , Animais Recém-Nascidos , Proteínas da Membrana Bacteriana Externa/metabolismo , Sítios de Ligação , Linhagem Celular , Glicosilação , Macrófagos/metabolismo , Meningite devida a Escherichia coli/genética , Camundongos , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Transfecção
11.
Clin Infect Dis ; 61(5): 779-86, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25944342

RESUMO

BACKGROUND: We aimed to describe features of Escherichia coli meningitis in a large population of children and the molecular characteristics of the involved strains to determine factors associated with severe disease or death. METHODS: Between 2001 and 2013, a prospective national survey collected data for 325 children hospitalized with E. coli meningitis. The national reference center genetically characterized 141 isolates. RESULTS: Among the 325 cases, 65.2% were term, 22.4% late preterm, and 12.5% very/extremely preterm infants. Escherichia coli meningitis was 7-fold more frequent in preterm than term infants. Median age at diagnosis was 14 days; 71.1% of infants were neonates, with 2 peaks of infection at age 0-3 days (mostly preterm neonates) and 11-15 days (mostly term neonates); 8.9% were >89 days old. In total, 51.1% patients were considered to have severe disease, and 9.2% died. B2.1 phylogenetic subgroup (56%) and O1 serogroup (27.7%) were the most frequently identified. On multivariate analysis, death was associated with preterm birth (odds ratio [OR], 3.3 [95% confidence interval {CI}, 1.3-8.4], P = .015 for late preterm infants; OR, 7.3 [95% CI, 2.7-20.9], P < .001 for very/extremely preterm infants) and cerebrospinal fluid (CSF) to blood glucose ratio <0.10 (OR, 15.3 [95% CI, 1.8-128.3], P = .012). Death was associated with uncommon O serogroup strains (P = .014) and severe disease with O7 serogroup (P = .034) and PapGII adhesin (OR, 2.3 [95% CI, 1.2-4.5], P = .015). CONCLUSIONS: In this large study of 325 cases of E. coli meningitis, risk factors of severe disease or death were preterm birth, severe hypoglycorrhachia, CSF/blood glucose ratio <0.10, and molecular characteristics of strains, which should help optimize therapeutic management.


Assuntos
Doenças do Recém-Nascido/epidemiologia , Doenças do Recém-Nascido/microbiologia , Meningite devida a Escherichia coli/epidemiologia , Meningite devida a Escherichia coli/microbiologia , Glicemia , Escherichia coli/classificação , Escherichia coli/genética , Feminino , França/epidemiologia , Glucose/líquido cefalorraquidiano , Humanos , Lactente , Recém-Nascido , Doenças do Recém-Nascido/mortalidade , Doenças do Recém-Nascido/fisiopatologia , Masculino , Meningite devida a Escherichia coli/mortalidade , Meningite devida a Escherichia coli/fisiopatologia , Estudos Prospectivos , Fatores de Risco
12.
BMC Infect Dis ; 15: 567, 2015 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-26654107

RESUMO

BACKGROUND: Escherichia coli is a rare cause of community-acquired meningitis in adults unless predisposing factors are present (e.g., previous penetrating cranio-cerebral injury or neurosurgery, immunosuppression, chronic alcoholism, history of cancer, diabetes mellitus, advanced age). CASE PRESENTATION: We describe the case of a 53-year-old woman, resident in Germany, suffering from community-acquired bacterial meningitis caused by CTX-M-9 type extended spectrum ß-lactamase producing Escherichia coli. Because typical predisposing factors were not apparent, pathogen identification resulted in expanded diagnostics to exclude a distant or contiguous primary focus. By magnetic resonance tomography, a previously unrecognized large retropharyngeal abscess with cervical spondylodiscitis was detected. In retrospect, the patient had complained about neck pain for a few weeks prior to meningitis onset, but the symptoms were interpreted as being related to a herniated disk. Meningitis and osteomyelitis resolved completely under surgical treatment and meropenem therapy. CONCLUSION: In case of adult Escherichia coli meningitis, underlying diseases should always be carefully excluded, especially if predisposing factors are not apparent.


Assuntos
Discite/diagnóstico , Infecções por Escherichia coli/diagnóstico , Meningite devida a Escherichia coli/diagnóstico , Abscesso Retrofaríngeo/diagnóstico , Discite/microbiologia , Discite/cirurgia , Escherichia coli/enzimologia , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Feminino , Alemanha , Humanos , Imageamento por Ressonância Magnética , Meningite devida a Escherichia coli/microbiologia , Meningite devida a Escherichia coli/cirurgia , Meropeném , Pessoa de Meia-Idade , Osteomielite/diagnóstico , Osteomielite/cirurgia , Abscesso Retrofaríngeo/microbiologia , Abscesso Retrofaríngeo/cirurgia , Tienamicinas , beta-Lactamases/metabolismo
13.
J Bacteriol ; 196(7): 1343-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24443535

RESUMO

The ability to capture iron is a challenge for most bacteria. The neonatal meningitis Escherichia coli strain S88 possesses several iron uptake systems, notably including siderophores. Transcriptional analysis of the ColV plasmid pS88 has shown strong induction of a previously undescribed gene with low identity to three E. coli chromosomal genes encoding phospho-2-dehydro-3-deoxyheptonate aldolases involved in aromatic amino acid and catecholate/phenolate siderophore biosynthesis through the shikimate pathway. Here, we investigated the role of this gene, ssbLp (ssbL carried on the plasmid), in siderophore biosynthesis and, consequently, in S88 virulence. We constructed an S88 mutant designated S88 ΔssbLp, which exhibited reduced growth under low-iron conditions compared to the wild-type strain. Liquid chromatography-mass spectroscopy analysis of culture supernatants showed that the mutant secreted significantly smaller amounts of enterobactin, salmochelin SX, and yersiniabactin than the wild-type strain. The mutant was also less virulent in a neonatal rat sepsis model, with significantly lower bacteremia and mortality. Supplementation with chorismate, the final product of the shikimate pathway, restored the wild-type phenotype in vitro. In a collection of human extraintestinal E. coli isolates, we found that ssbL was present only in strains harboring the iro locus, encoding salmochelins, and was located either on the chromosome or on plasmids. Acquisition of the iro locus has been accompanied by acquisition of the auxiliary gene ssbL, which boosts the metabolic pathway essential for catecholate/phenolate siderophore biosynthesis and could represent potential therapeutic targets.


Assuntos
Aldeído Liases/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Meningite devida a Escherichia coli/microbiologia , Plasmídeos/genética , Ácido Chiquímico/metabolismo , Sideróforos/biossíntese , Fatores de Virulência/metabolismo , Aldeído Liases/genética , Animais , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Humanos , Ferro/metabolismo , Redes e Vias Metabólicas , Plasmídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Virulência/genética
14.
J Am Chem Soc ; 136(26): 9244-7, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24932672

RESUMO

The gene cluster responsible for synthesis of the unknown molecule "colibactin" has been identified in mutualistic and pathogenic Escherichia coli. The pathway endows its producer with a long-term persistence phenotype in the human bowel, a probiotic activity used in the treatment of ulcerative colitis, and a carcinogenic activity under host inflammatory conditions. To date, functional small molecules from this pathway have not been reported. Here we implemented a comparative metabolomics and targeted structural network analyses approach to identify a catalog of small molecules dependent on the colibactin pathway from the meningitis isolate E. coli IHE3034 and the probiotic E. coli Nissle 1917. The structures of 10 pathway-dependent small molecules are proposed based on structural characterizations and network relationships. The network will provide a roadmap for the structural and functional elucidation of a variety of other small molecules encoded by the pathway. From the characterized small molecule set, in vitro bacterial growth inhibitory and mammalian CNS receptor antagonist activities are presented.


Assuntos
Escherichia coli/metabolismo , Metabolômica/métodos , Peptídeos/metabolismo , Policetídeos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Algoritmos , Bacillus subtilis/efeitos dos fármacos , Antagonistas de Dopamina/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Escherichia coli/isolamento & purificação , Escherichia coli/patogenicidade , Células HeLa/efeitos dos fármacos , Humanos , Espectroscopia de Ressonância Magnética , Meningite devida a Escherichia coli/microbiologia , Redes e Vias Metabólicas , Estrutura Molecular , Peptídeos/genética , Probióticos , Bibliotecas de Moléculas Pequenas/farmacologia
15.
Curr Opin Infect Dis ; 27(5): 459-64, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25023740

RESUMO

PURPOSE OF REVIEW: Colonization of the host epithelia by pathogenic Escherichia coli is influenced by the ability of the bacteria to interact with host surfaces. Because the initial step of an E. coli infection is to adhere, invade, and persist within host cells, some strategies used by intestinal and extraintestinal E. coli to infect host cell are presented. RECENT FINDINGS: This review highlights recent progress understanding how extraintestinal pathogenic E. coli strains express specific adhesins or invasins that allow colonization of the urinary tract or the meninges, while intestinal E. coli strains are able to colonize different regions of the intestinal tract using other specialized adhesins or invasins. Finally, evaluation of different diets and environmental conditions regulating the colonization of these pathogens is discussed. SUMMARY: Discovery of new interactions between pathogenic E. coli and the host epithelial cells unravels the need for more mechanistic studies that can provide new clues regarding how to combat these infections.


Assuntos
Aderência Bacteriana/imunologia , Translocação Bacteriana/imunologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/isolamento & purificação , Trato Gastrointestinal/microbiologia , Meningite devida a Escherichia coli/microbiologia , Infecções Urinárias/microbiologia , Adesinas de Escherichia coli , Animais , Barreira Hematoencefálica/microbiologia , Modelos Animais de Doenças , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/imunologia , Proteínas da Matriz Extracelular , Trato Gastrointestinal/imunologia , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Meningite devida a Escherichia coli/imunologia , Camundongos , Infecções Urinárias/etiologia , Infecções Urinárias/imunologia
16.
BMC Microbiol ; 14: 203, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25164788

RESUMO

BACKGROUND: Escherichia coli is the most predominant Gram-negative bacterial pathogen associated with neonatal meningitis. Previous studies indicated that the prototypic neonatal meningitis E. coli (NMEC) strain RS218 (O18:K1:H7) harbors one large plasmid. Objectives of the present study were to analyze the complete nucleotide sequence of this large plasmid (pRS218) and its contribution to NMEC pathogenesis using in vitro and in vivo models of neonatal meningitis. RESULTS: The plasmid is 114,231 bp in size, belongs to the incompatibility group FIB/IIA (IncFIB/IIA), and contains a genetic load region that encodes several virulence and fitness traits such as enterotoxicity, iron acquisition and copper tolerance. The nucleotide sequence of pRS218 showed a 41- 46% similarity to other neonatal meningitis-causing E. coli (NMEC) plasmids and remarkable nucleotide sequence similarity (up to 100%) to large virulence plasmids of E. coli associated with acute cystitis. Some genes located on pRS218 were overly represented by NMEC strains compared to fecal E. coli isolated from healthy individuals. The plasmid-cured strain was significantly attenuated relative to the RS218 wild-type strain as determined in vitro by invasion potential to human cerebral microvascular endothelial cells and in vivo by mortalities, histopathological lesions in the brain tissue, and bacterial recovery from the cerebrospinal fluid of infected rat pups. CONCLUSIONS: The pRS218 is an IncFIB/IIA plasmid which shares a remarkable nucleotide sequence similarity to large plasmids of E. coli associated with cystitis. Both in vitro and in vivo experiments indicated that pRS218 plays an important role in NMEC pathogenesis.


Assuntos
Escherichia coli/genética , Meningite devida a Escherichia coli/microbiologia , Plasmídeos , Fatores de Virulência/genética , Animais , Líquido Cefalorraquidiano/microbiologia , DNA Bacteriano/química , DNA Bacteriano/genética , Escherichia coli/isolamento & purificação , Ordem dos Genes , Humanos , Recém-Nascido , Dados de Sequência Molecular , Ratos Sprague-Dawley , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico , Análise de Sobrevida , Virulência
17.
J Infect Dis ; 207(1): 61-71, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23100563

RESUMO

Elevated levels of pterins and nitric oxide (NO) are observed in patients with septic shock and bacterial meningitis. We demonstrate that Escherichia coli K1 infection of human brain microvascular endothelial cells (HBMECs) induces the expression of guanosine triphosphate cyclohydrolase (GCH1), the rate-limiting enzyme in pterin synthesis, thereby elevating levels of biopterin. DAHP (2,4-diamino hydroxyl pyrimidine), a specific inhibitor of GCH1, prevented biopterin and NO production and invasion of E. coli K1 in HBMECs. GCH1 interaction with Ecgp96, the receptor for outer membrane protein A of E. coli K1, also increases on infection, and suppression of Ecgp96 expression prevents GCH1 activation and biopterin synthesis. Pretreatment of newborn mice with DAHP prevented the production of biopterin and the development of meningitis. These results suggest a novel role for biopterin synthesis in the pathogenesis of E. coli K1 meningitis.


Assuntos
Biopterinas/metabolismo , Encéfalo/microbiologia , Células Endoteliais/microbiologia , Escherichia coli/patogenicidade , Meningite devida a Escherichia coli/prevenção & controle , Animais , Animais Recém-Nascidos , Proteínas da Membrana Bacteriana Externa/metabolismo , Biopterinas/biossíntese , Encéfalo/citologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , GTP Cicloidrolase/antagonistas & inibidores , Humanos , Hipoxantinas/farmacologia , Hipoxantinas/uso terapêutico , Meningite devida a Escherichia coli/metabolismo , Meningite devida a Escherichia coli/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Microvasos/metabolismo , Microvasos/microbiologia , Óxido Nítrico/análise , Óxido Nítrico/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Virulência
18.
ACS Infect Dis ; 10(3): 988-999, 2024 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-38317607

RESUMO

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.


Assuntos
Infecções por Escherichia coli , Meningites Bacterianas , Meningite devida a Escherichia coli , Humanos , Camundongos , Animais , Barreira Hematoencefálica , Meningite devida a Escherichia coli/metabolismo , Meningite devida a Escherichia coli/microbiologia , Meningite devida a Escherichia coli/patologia , Escherichia coli/metabolismo , Células Endoteliais , Interleucina 22 , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/farmacologia
19.
Antimicrob Agents Chemother ; 57(12): 5808-10, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24002097

RESUMO

In this study, the efficacy of ceftaroline fosamil was compared with that of cefepime in an experimental rabbit meningitis model against two Gram-negative strains (Escherichia coli QK-9 and Klebsiella pneumoniae 1173687). The penetration of ceftaroline into inflamed and uninflamed meninges was also investigated. Both regimens were bactericidal, but ceftaroline fosamil was significantly superior to cefepime against K. pneumoniae and E. coli in this experimental rabbit meningitis model (P < 0.0007 against K. pneumoniae and P < 0.0016 against E. coli). The penetration of ceftaroline was approximately 15% into inflamed meninges and approximately 3% into uninflamed meninges.


Assuntos
Antibacterianos/farmacologia , Cefalosporinas/farmacologia , Infecções por Klebsiella/tratamento farmacológico , Meningite devida a Escherichia coli/tratamento farmacológico , Animais , Antibacterianos/líquido cefalorraquidiano , Antibacterianos/farmacocinética , Cefepima , Cefalosporinas/líquido cefalorraquidiano , Cefalosporinas/farmacocinética , Modelos Animais de Doenças , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Infecções por Klebsiella/líquido cefalorraquidiano , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/fisiologia , Meninges/efeitos dos fármacos , Meninges/metabolismo , Meninges/microbiologia , Meningite devida a Escherichia coli/líquido cefalorraquidiano , Meningite devida a Escherichia coli/microbiologia , Permeabilidade , Coelhos , Resultado do Tratamento , Ceftarolina
20.
J Negat Results Biomed ; 12: 8, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23634997

RESUMO

BACKGROUND: Proteases are well-known virulence factors that promote survival, pathogenesis and immune evasion of many pathogens. Several lines of evidence suggest that the blood-brain barrier permeability is a prerequisite in microbial invasion of the central nervous system. Because proteases are frequently associated with vascular permeability by targeting junctional proteins, here it is hypothesized that neuropathogenic Escherichia coli K1 exhibit proteolytic activities to exert its pathogenicity. METHODS: Zymographic assays were performed using collagen and gelatin as substrates. The lysates of whole E. coli K1 strain E44, or E. coli K-12 strain HB101 were tested for proteolytic activities. The conditioned media were prepared by incubating bacteria in RPMI-1640 in the presence or absence of serum. The cell-free supernatants were collected and tested for proteases in zymography as mentioned above. Additionally, proteolytic degradation of host immune factors was determined by co-incubating conditioned media with albumin/immunoglobulins using protease assays. RESULTS: When collagen or gelatin were used as substrates in zymographic assays, neither whole bacteria nor conditioned media exhibited proteolytic activities. The conditioned media of neuropathogenic E. coli K1 strain E44, or E. coli K-12 strain HB101 did not affect degradation of albumin and immunoglobulins using protease assays. CONCLUSIONS: Neither zymographic assays nor protease assays detected proteolytic activities in either the whole bacteria or conditioned media of E. coli K1 strain E44 and E. coli K-12 strain HB101. These findings suggest that host cell monolayer disruptions and immune evasion strategies are likely independent of proteolytic activities of neuropathogenic E. coli K1.


Assuntos
Escherichia coli/patogenicidade , Meningite devida a Escherichia coli/microbiologia , Meningite devida a Escherichia coli/patologia , Proteólise , Animais , Bovinos , Meios de Cultivo Condicionados/química , Ensaios Enzimáticos , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/metabolismo , Meningite devida a Escherichia coli/metabolismo , Peptídeo Hidrolases/metabolismo , Coelhos , Soroalbumina Bovina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa