1
|
Roy A, Anbarasu A. Unveiling Berberine analogues as potential inhibitors of Escherichia coli FtsZ through machine learning molecular docking and molecular dynamics approach. Sci Rep 2025; 15:14668. [PMID: 40287515 PMCID: PMC12033256 DOI: 10.1038/s41598-025-98835-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
The bacterial cell division protein FtsZ, a crucial GTPase, plays a vital role in the formation of the contractile Z-ring, which is essential for bacterial cytokinesis. Consequently, inhibiting FtsZ could prevent the formation of proto-filaments and interfere with the cell division machinery. The remarkable conservation of FtsZ across diverse bacterial species makes it a promising drug target for combating drug resistance. In the present study, 1072 berberine analogues were screened for favorable pharmacokinetic properties. A total of 60 compounds that fulfilled the drug-likeliness criteria and were found to be non-toxic were selected for virtual screening against Escherichia coli FtsZ protein (PDB ID: 8GZY). Molecular docking revealed a strong binding affinity of ZINC000524729297 (- 8.73 kcal/mol) and ZINC000604405393 (and - 8.55 kcal/mol) with FtsZ by strong intermolecular hydrogen bonds and hydrophobic interactions. Subsequently, the docking profiles were validated through a 500 ns MD simulation and MMPBSA analysis of the FtsZ-ligand complexes. The analysis revealed the FtsZ- ZINC524729297 and FtsZ-ZINC000604405393 complexes had the lowest root-mean-square deviation with lowest binding energy and enhanced conformational stability in a dynamic environment. These findings suggest that ZINC524729297 and ZINC000604405393 are the potent lead compound that targets FtsZ and requires further experimental validation.
Collapse
Affiliation(s)
- Aditi Roy
- Medical and Biological Computing Laboratory, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
- Department of Biotechnology, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
2
|
Song Y, Baniakina LFT, Jiang L, Chai L. Metagenomic insights into the alterations of gut microbial community in Bufo gargarizans tadpoles following lead exposure. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101522. [PMID: 40288073 DOI: 10.1016/j.cbd.2025.101522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Lead (Pb), a prevalent heavy metal contaminant in aquatic environments, has complex effects on the gut microbiome function of aquatic animals. In this study, metagenomic analysis of Bufo gargarizans tadpoles was carried out following Pb exposure. Moreover, histological analysis was performed on the intestines. The results showed that Pb exposure induced histological damage to the intestinal epithelium. Significant differences in microbial abundance and function were detected in the 200 μg/L Pb group compared to the control group. Specifically, an increase in Bosea and Klebsiella was noted at 200 μg/L Pb, which potentially could induce inflammation in tadpoles. Notably, the decrease in the abundance of glycoside hydrolases subsequent to exposure to 200 μg/L Pb is likely to attenuate carbohydrate metabolism. Furthermore, increased fluoroquinolone-related antibiotic resistance genes (ARGs), phenolic-related ARGs, and iron uptake systems following 200 μg/L Pb exposure might heighten the disease risk for tadpoles. These discoveries augment our comprehension of the influences of Pb on the intestinal well-being of amphibians and offer valuable insights for further assessment of the ecological risks that Pb poses to amphibians.
Collapse
Affiliation(s)
- Yanjiao Song
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of the Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Lod Fabuleux Tresor Baniakina
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Eco-hydrology and Water Security in Arid and Semi-arid Regions of Ministry of Water Resources, Chang'an University, Xi'an 710054, China
| | - Ling Jiang
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of the Ministry of Education, Chang'an University, Xi'an 710054, China
| | - Lihong Chai
- School of Water and Environment, Chang'an University, Xi'an 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of the Ministry of Education, Chang'an University, Xi'an 710054, China.
| |
Collapse
|
3
|
Asgari B, Burke JR, Quigley BL, Bradford G, Hatje E, Kuballa A, Katouli M. Identification of Virulence Genes Associated with Pathogenicity of Translocating Escherichia coli with Special Reference to the Type 6 Secretion System. Microorganisms 2024; 12:1851. [PMID: 39338525 PMCID: PMC11433802 DOI: 10.3390/microorganisms12091851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Recent genomic characterisation of translocating Escherichia coli HMLN-1 isolated from mesenteric lymph nodes (MLNs) and blood of a patient with a fatal case of pancreatitis revealed the presence of a type 6 secretion system (T6SS) that was not present in non-translocating E. coli strains. This strain was also genomically similar to adherent-invasive E. coli (AIEC) LF82 pathotype. We aimed to identify the role of T6SS-1 in the pathogenesis of this strain and other pathogenic E. coli. The HMLN-1 strain was initially tested for the presence of six virulence genes (VGs) associated with AIEC strains and an iron sequestering system. Additionally, HMLN-1's interaction with a co-culture of Caco-2:HT29-MTX cells and its intra-macrophagic survival was evaluated. We subsequently screened a collection of 319 pathogenic E. coli strains isolated from patients with urinary tract infection (UTI), diarrhoea, inflammatory bowel disease (IBD) and septicaemia for the presence of T6SS-1 and its expression related to adhesion, invasion and translocation via the above co-culture of the intestinal cell lines. The results showed that HMLN-1 harboured four of the AIEC-associated VGs (dsbA, htrA, ompC and afaC). Screening of the pathogenic E. coli collection detected the presence of the T6SS-1 genes in septicaemic and UTI E. coli strains at a significantly higher level than diarrhoea and IBD strains (p < 0.0001). The high expression of T6SS-1 in E. coli HMLN-1 upon adhesion and invasion, as well as its high prevalence among extra-intestinal E. coli strains, suggests a role for T6SS-1 in the pathogenesis of translocating E. coli.
Collapse
Affiliation(s)
- Behnoush Asgari
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| | - Jarred R. Burke
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- Servatus Biopharmaceuticals, Coolum Beach, QLD 4573, Australia
| | - Bonnie L. Quigley
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- Thompson Institute, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia
| | - Georgia Bradford
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| | - Eva Hatje
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Anna Kuballa
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- School of Health, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia
| | - Mohammad Katouli
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| |
Collapse
|
4
|
Del Carpio AMG, Freire CA, Andrade FB, Piazza RMF, Silva RM, Carvalho E, Elias WP. Genomic Dissection of an Enteroaggregative Escherichia coli Strain Isolated from Bacteremia Reveals Insights into Its Hybrid Pathogenic Potential. Int J Mol Sci 2024; 25:9238. [PMID: 39273188 PMCID: PMC11394720 DOI: 10.3390/ijms25179238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 09/15/2024] Open
Abstract
Escherichia coli is a frequent pathogen isolated from bloodstream infections. This study aimed to characterize the genetic features of EC092, an E. coli strain isolated from bacteremia that harbors enteroaggregative E. coli (EAEC) genetic markers, indicating its hybrid pathogenic potential. Whole-genome sequencing showed that EC092 belongs to phylogroup B1, ST278, and serotype O165:H4. Genes encoding virulence factors such as fimbriae, toxins, iron-uptake systems, autotransporter proteins (Pet, Pic, Sat, and SepA), and secretion systems were detected, as well as EAEC virulence genes (aggR, aatA, aaiC, and aap). EC092 was found to be closely related to the other EAEC prototype strains and highly similar in terms of virulence to three EAEC strains isolated from diarrhea. The genomic neighborhood of pet, pic, sat, sepA, and the EAEC virulence genes of EC092 and its three genetically related fecal EAEC strains showed an identical genomic organization and nucleotide sequences. Also, EC092 produced and secreted Pet, Pic, Sat, and SepA in the culture supernatant and resisted the bactericidal activity of normal human serum. Our results demonstrate that the strain EC092, isolated from bacteremia, is a hybrid pathogenic extraintestinal E. coli (ExPEC)/EAEC with virulence features that could mediate both extraintestinal and intestinal infections.
Collapse
Affiliation(s)
| | - Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Fernanda B Andrade
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Roxane M F Piazza
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Rosa M Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Eneas Carvalho
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil
| |
Collapse
|
5
|
Poteete O, Cox P, Ruffin F, Sutton G, Brinkac L, Clarke TH, Fouts DE, Fowler VG, Thaden JT. Serum susceptibility of Escherichia coli and its association with patient clinical outcomes. PLoS One 2024; 19:e0307968. [PMID: 39074102 DOI: 10.1371/journal.pone.0307968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024] Open
Abstract
The innate immune system eliminates bloodstream pathogens such as Escherichia coli in part through complement protein deposition and subsequent bacterial death (i.e., "serum killing"). Some E. coli strains have developed mechanisms to resist serum killing, though the extent of variation in serum killing among bloodstream infection (BSI) isolates and the clinical impact of this variation is not well understood. To address this issue, we developed a novel assay that uses flow cytometry to perform high throughput serum bactericidal assays (SBAs) with E. coli BSI isolates (n = 183) to define the proportion of surviving bacteria after exposure to serum. We further determined whether E. coli resistance to serum killing is associated with clinical outcomes (e.g., in-hospital attributable mortality, in-hospital total mortality, septic shock) and bacterial genotype in the corresponding patients with E. coli BSI. Our novel flow cytometry-based SBA performed similarly to a traditional SBA, though with significantly decreased hands-on bench work. Among E. coli BSI isolates, the mean proportion that survived exposure to 25% serum was 0.68 (Standard deviation 0.02, range 0.57-0.93). We did not identify associations between E. coli resistance to serum killing and clinical outcomes in our adjusted models. Together, this study describes a novel flow cytometry-based approach to the bacterial SBA that allowed for high-throughput testing of E. coli BSI isolates and identified high variability in resistance to serum killing among a large set of BSI isolates.
Collapse
Affiliation(s)
- Orianna Poteete
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States of America
| | - Phillip Cox
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States of America
| | - Felicia Ruffin
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States of America
| | - Granger Sutton
- J. Craig Venter Institute, Rockville, MD, United States of America
| | - Lauren Brinkac
- J. Craig Venter Institute, Rockville, MD, United States of America
| | - Thomas H Clarke
- J. Craig Venter Institute, Rockville, MD, United States of America
| | - Derrick E Fouts
- J. Craig Venter Institute, Rockville, MD, United States of America
| | - Vance G Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States of America
- Duke Clinical Research Institute, Durham, NC, United States of America
| | - Joshua T Thaden
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States of America
| |
Collapse
|
6
|
Fierro CA, Sarnecki M, Spiessens B, Go O, Day TA, Davies TA, van den Dobbelsteen G, Poolman J, Abbanat D, Haazen W. A randomized phase 1/2a trial of ExPEC10V vaccine in adults with a history of UTI. NPJ Vaccines 2024; 9:106. [PMID: 38877036 PMCID: PMC11178786 DOI: 10.1038/s41541-024-00885-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/07/2024] [Indexed: 06/16/2024] Open
Abstract
The safety, reactogenicity, and immunogenicity of 3 doses of ExPEC10V (VAC52416), a vaccine candidate to prevent invasive Escherichia coli disease, were assessed in a phase 1/2a study (NCT03819049). In Cohort 1, ExPEC10V was well tolerated; the high dose was selected as optimal and further characterized in Cohort 2. Cohort 2 comprised a maximum 28-day screening, vaccination (Day 1), double-blind 181-day follow-up, and open-label long-term follow-up until Year 1. Healthy participants (≥60 years) with a history of urinary tract infection (UTI) within 5 years were randomized to receive ExPEC10V or placebo. The primary endpoint evaluated the safety and reactogenicity of ExPEC10V (solicited local and systemic AEs [until Day 15]; unsolicited AEs [until Day 30], SAEs [until Day 181], and immunogenicity [Day 30]) via multiplex electrochemiluminescent (ECL) and multiplex opsonophagocytic assay (MOPA). 416 participants (ExPEC10V, n = 278; placebo, n = 138) were included (mean age [SD], 68.8 [6.52] years; female, 79.6%; White, 96.1%). The incidence of solicited AEs was higher with ExPEC10V (local, 50.0% [n = 139]; systemic, 50.0% [n = 139]) than placebo (15.9% [n = 22]; 38.4% [n = 53]); rates of unsolicited AEs were comparable (ExPEC10V, 28.4% [n = 79]; placebo, 26.1% [n = 36]). No vaccine-related SAEs or deaths were reported. ExPEC10V elicited a robust antibody-mediated immunogenic response across all serotypes with ECL (Day 30 geometric mean fold increase, 2.33-8.18) and demonstrated functional opsonophagocytic killing activity across all measured serotypes (Day 30 geometric mean fold increase, 1.81-9.68). ExPEC10V exhibited an acceptable safety profile and a robust vaccine-induced functional immunogenic response in participants with a history of UTI. Clinical trial registration details: https://clinicaltrials.gov/study/NCT03819049 .
Collapse
Affiliation(s)
| | - Michal Sarnecki
- Janssen Research & Development, Infectious Diseases & Vaccines, Janssen Vaccines, Bern, Switzerland
| | - Bart Spiessens
- Janssen Research & Development, Infectious Diseases & Vaccines, Janssen Pharmaceutica, Beerse, Belgium
| | - Oscar Go
- Janssen Research & Development, Raritan, NJ, USA
| | - Tracey A Day
- Bacterial Vaccines Discovery and Early Development, Janssen Vaccines & Prevention B.V., Leiden, Netherlands
| | | | - Germie van den Dobbelsteen
- Bacterial Vaccines Discovery and Early Development, Janssen Vaccines & Prevention B.V., Leiden, Netherlands
| | - Jan Poolman
- Bacterial Vaccines Discovery and Early Development, Janssen Vaccines & Prevention B.V., Leiden, Netherlands.
| | | | - Wouter Haazen
- Janssen Research & Development, Infectious Diseases & Vaccines, Janssen Pharmaceutica, Beerse, Belgium
| |
Collapse
|
7
|
Maldonado N, López-Hernández I, García-Montaner A, López-Cortés LE, Pérez-Crespo PMM, Retamar-Gentil P, Sousa-Domínguez A, Goikoetxea J, Pulido-Navazo Á, Labayru-Echeverría C, Natera-Kindelán C, Jover-Sáenz A, Del Arco-Jiménez A, Armiñanzas-Castillo C, Aller AI, Fernández-Suárez J, Marrodán-Ciordia T, Boix-Palop L, Smithson-Amat A, Reguera-Iglesias JM, Galán-Sánchez F, Bahamonde A, Sánchez Calvo JM, Gea-Lázaro I, Pérez-Camacho I, Reyes-Bertos A, Becerril-Carral B, Rodríguez-Baño J, Pascual Á. Whole-genome characterisation of Escherichia coli isolates from patients with bacteraemia presenting with sepsis or septic shock in Spain: a multicentre cross-sectional study. THE LANCET. MICROBE 2024; 5:e390-e399. [PMID: 38547882 DOI: 10.1016/s2666-5247(23)00369-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 04/08/2024]
Abstract
BACKGROUND Escherichia coli is the most frequent cause of bloodstream infections (BSIs). About one-third of patients with BSIs due to E coli develop sepsis or shock. The objective of this study is to characterise the microbiological features of E coli blood isolates causing sepsis or septic shock to provide exploratory information for future diagnostic, preventive, or therapeutic interventions. METHODS E coli blood isolates from a multicentre cross-sectional study of patients older than 14 years presenting with sepsis or septic shock (according to the Third International Consensus Definitions for Sepsis and Septic Shock criteria) from hospitals in Spain between Oct 4, 2016, and Oct 15, 2017, were studied by whole-genome sequencing. Phylogroups, sequence types (STs), serotype, FimH types, antimicrobial resistance (AMR) genes, pathogenicity islands, and virulence factors were identified. Susceptibility testing was performed by broth microdilution. The main outcome of this study was the characterisation of the E coli blood isolates in terms of population structure by phylogroups, groups (group 1: phylogroups B2, F, and G; group 2: A, B1, and C; group 3: D), and STs and distribution by geographical location and bloodstream infection source. Other outcomes were virulence score and prevalence of virulence-associated genes, pathogenicity islands, AMR, and AMR-associated genes. Frequencies were compared using χ² or Fisher's exact tests, and continuous variables using the Mann-Whitney test, with Bonferroni correction for multiple comparisons. FINDINGS We analysed 224 isolates: 140 isolates (63%) were included in phylogenetic group 1, 52 (23%) in group 2, and 32 (14%) in group 3. 85 STs were identified, with four comprising 44% (n=98) of the isolates: ST131 (38 [17%]), ST73 (25 [11%]), ST69 (23 [10%]), and ST95 (12 [5%]). No significant differences in phylogroup or ST distribution were found according to geographical areas or source of bloodstream infection, except for ST95, which was more frequent in urinary tract infections than in other sources (11 [9%] of 116 vs 1 [1%] of 108, p=0·0045). Median virulence score was higher in group 1 (median 25·0 [IQR 20·5-29·0) than in group 2 (median 14·5 [9·0-20·0]; p<0·0001) and group 3 (median 21 [16·5-23·0]; p<0·0001); prevalence of several pathogenicity islands was higher in group 1. No significant differences were found between phylogenetic groups in proportions of resistance to antibiotics. ST73 had higher median virulence score (32 [IQR 29-35]) than the other predominant clones (median range 21-28). Some virulence genes and pathogenicity islands were significantly associated with each ST. ST131 isolates had higher prevalence of AMR and a higher proportion of AMR genes, notably blaCTX-M-15 and blaOXA-1. INTERPRETATION In this exploratory study, the population structure of E coli causing sepsis or shock was similar to previous studies that included all bacteraemic isolates. Virulence genes, pathogenicity islands, and AMR genes were not randomly distributed among phylogroups or STs. These results provide a comprehensive characterisation of invasive E coli isolates causing severe response syndrome. Future studies are required to determine the contribution of these microbiological factors to severe clinical presentation and worse outcomes in patients with E coli bloodstream infection. FUNDING Instituto de Salud Carlos III.
Collapse
Affiliation(s)
- Natalia Maldonado
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
| | - Inmaculada López-Hernández
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea García-Montaner
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain
| | - Luis Eduardo López-Cortés
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Pilar Retamar-Gentil
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | - Clara Natera-Kindelán
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | | | - Carlos Armiñanzas-Castillo
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Hospital Universitario de Marqués de Valdecilla, Santander, Spain
| | | | - Jonathan Fernández-Suárez
- Hospital Universitario Central de Asturias, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | | | | | - José Mª Reguera-Iglesias
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
| | | | | | - Juan Manuel Sánchez Calvo
- Hospital Universitario de Jerez, Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Jerez de la Frontera, Spain
| | | | | | | | | | - Jesús Rodríguez-Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Álvaro Pascual
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Departamentos de Medicina y Microbiología, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Distribution of Pathogenicity Island Markers and Virulence Factors Genes of Extraintestinal Pathogenic Escherichia coli Isolates. Jundishapur J Microbiol 2022. [DOI: 10.5812/jjm-121044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Extraintestinal pathogenic Escherichia coli (ExPEC) can colonize the urinary system with various virulence factors. Escaping from the host defense, can lead to serious infections, such as bacteremia and sepsis. Objectives: This study aimed to investigate the presence of phylogenetic groups, various virulence factor genes, and pathogenicity island (PAI) markers of extraintestinal and commensal E. coli isolates. Methods: Within January 2016 and December 2017, 50 E. coli isolates were isolated from simultaneous blood and urine samples of 25 patients hospitalized in Mersin University Faculty of Medicine, Turkey, and 50 commensal E. coli isolates were isolated from fecal 18 samples of healthy volunteers as the control group was included in the study. Phylogenetic groups, virulence factors genes, and PAI markers were determined by the multiplex polymerase chain reaction method. Results: The majority of ExPEC isolates were observed to be in phylogenetic group B2. All of the commensal E. coli isolates were detected in phylogenetic group A. The most predominant virulence factor gene was traT (80%). The most prevalent PAI marker was PAI IV536 (86%). However, the PAI I536, IJ96, and IIJ96 markers were not detected in any isolates. Only PAI IV536 was detected in commensal E. coli isolates. Conclusions: It was determined that E. coli isolates that cause uropathogenic and bloodstream infections contain a higher frequency of PAI than commensal isolates.
Collapse
|
9
|
Freire CA, Silva RM, Ruiz RC, Pimenta DC, Bryant JA, Henderson IR, Barbosa AS, Elias WP. Secreted Autotransporter Toxin (Sat) Mediates Innate Immune System Evasion. Front Immunol 2022; 13:844878. [PMID: 35251044 PMCID: PMC8891578 DOI: 10.3389/fimmu.2022.844878] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Several strategies are used by Escherichia coli to evade the host innate immune system in the blood, such as the cleavage of complement system proteins by secreted proteases. Members of the Serine Proteases Autotransporters of Enterobacteriaceae (SPATE) family have been described as presenting proteolytic effects against complement proteins. Among the SPATE-encoding genes sat (secreted autotransporter toxin) has been detected in high frequencies among strains of E. coli isolated from bacteremia. Sat has been characterized for its cytotoxic action, but the possible immunomodulatory effects of Sat have not been investigated. Therefore, this study aimed to evaluate the proteolytic effects of Sat on complement proteins and the role in pathogenesis of BSI caused by extraintestinal E. coli (ExPEC). E. coli EC071 was selected as a Sat-producing ExPEC strain. Whole-genome sequencing showed that sat sequences of EC071 and uropathogenic E. coli CFT073 present 99% identity. EC071 was shown to be resistant to the bactericidal activity of normal human serum (NHS). Purified native Sat was used in proteolytic assays with proteins of the complement system and, except for C1q, all tested substrates were cleaved by Sat in a dose and time-dependent manner. Moreover, E. coli DH5α survived in NHS pre-incubated with Sat. EC071-derivative strains harboring sat knockout and in trans complementations producing either active or non-active Sat were tested in a murine sepsis model. Lethality was reduced by 50% when mice were inoculated with the sat mutant strain. The complemented strain producing active Sat partially restored the effect caused by the wild-type strain. The results presented in this study show that Sat presents immunomodulatory effects by cleaving several proteins of the three complement system pathways. Therefore, Sat plays an important role in the establishment of bloodstream infections and sepsis.
Collapse
Affiliation(s)
- Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Rosa M Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rita C Ruiz
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Daniel C Pimenta
- Laboratório de Bioquímica, Instituto Butantan, São Paulo, Brazil
| | - Jack A Bryant
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom.,Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Angela S Barbosa
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
10
|
Genetic Characterization of Multidrug-Resistant E. coli Isolates from Bloodstream Infections in Lithuania. Microorganisms 2022; 10:microorganisms10020449. [PMID: 35208903 PMCID: PMC8880137 DOI: 10.3390/microorganisms10020449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) isolates are a main cause of bloodstream infections. The aim of this study was to characterize 256 β-lactam–resistant, bacteremia-causing E. coli isolates collected from 12 healthcare institutions in Lithuania in 2014 and 2018. All isolates were identified as E. coli via MALDI-TOF MS and VITEK ®2. In addition, the isolates were analyzed for the presence of 29 resistance genes and 13 virulence genes, divided into phylogenetic groups (A, B1, B2, C, D, E, and F), and characterized using rep-PCR genotyping methods (BOX-PCR and (GTG)5-PCR). Analyzing the results of this study showed tetA-strB-sul2-TEM-NDM-strA-fosA-AIM-sul3-aadA-CTX-M-9 to be the most common resistance gene combination (67.2% of all isolates). Additionally, the most common virulence genes established were fimH (98.4% of all isolates), fyuA (91.8%), and traT (81.3%) and the most common gene combination was fuyA-fimH-iroN (58.6% of all isolates). Next, the isolates were separated into four phylogenetic groups: A, B1, B2, and F, where group A isolates were detected at a significantly higher frequency (79.3% of all isolates). Finally, a total of 235 genotyping profiles were established using rep-PCR methods, and all profiles were separated into fourteen genotypic clusters, with each cluster containing profiles with a variety of virulence and resistance genes not restricted to any specific cluster. The results of this study elucidate E. coli antimicrobial resistance patterns by highlighting the variability and diversity of resistance and virulence genes and providing phylogenetic classification, genetic profiling, and clustering data. These results may improve clinical control of multidrug-resistant infections in healthcare institutions and contribute to the prevention of potential outbreaks.
Collapse
|
11
|
Genome Informatics and Machine Learning-Based Identification of Antimicrobial Resistance-Encoding Features and Virulence Attributes in Escherichia coli Genomes Representing Globally Prevalent Lineages, Including High-Risk Clonal Complexes. mBio 2021; 13:e0379621. [PMID: 35164570 PMCID: PMC8844930 DOI: 10.1128/mbio.03796-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Escherichia coli, a ubiquitous commensal/pathogenic member from the Enterobacteriaceae family, accounts for high infection burden, morbidity, and mortality throughout the world. With emerging multidrug resistance (MDR) on a massive scale, E. coli has been listed as one of the Global Antimicrobial Resistance and Use Surveillance System (GLASS) priority pathogens. Understanding the resistance mechanisms and underlying genomic features appears to be of utmost importance to tackle further spread of these multidrug-resistant superbugs. While a few of the globally prevalent sequence types (STs) of E. coli, such as ST131, ST69, ST405, and ST648, have been previously reported to be highly virulent and harboring MDR, there is no clarity if certain ST lineages have a greater propensity to acquire MDR. In this study, large-scale comparative genomics of a total of 5,653 E. coli genomes from 19 ST lineages revealed ST-wide prevalence patterns of genomic features, such as antimicrobial resistance (AMR)-encoding genes/mutations, virulence genes, integrons, and transposons. Interpretation of the importance of these features using a Random Forest Classifier trained with 11,988 genomic features from whole-genome sequence data identified ST-specific or phylogroup-specific signature proteins mostly belonging to different protein superfamilies, including the toxin-antitoxin systems. Our study provides a comprehensive understanding of a myriad of genomic features, ST-specific proteins, and resistance mechanisms entailing different lineages of E. coli at the level of genomes; this could be of significant downstream importance in understanding the mechanisms of AMR, in clinical discovery, in epidemiology, and in devising control strategies. IMPORTANCE With the leap in whole-genome data being generated, the application of relevant methods to mine biologically significant information from microbial genomes is of utmost importance to public health genomics. Machine-learning methods have been used not only to mine, curate, or classify the data but also to identify the relevant features that could be linked to a particular class/target. This is perhaps one of the pioneering studies that has attempted to classify a large repertoire of E. coli genome data sets (5,653 genomes) belonging to 19 different STs (including well-studied as well as understudied STs) using machine learning approaches. Important features identified by these approaches have revealed ST-specific signature proteins, which could be further studied to predict possible associations with the phenotypic profiles, thereby providing a better understanding of virulence and the resistance mechanisms among different clonal lineages of E. coli.
Collapse
|
12
|
Li D, Reid CJ, Kudinha T, Jarocki VM, Djordjevic SP. Genomic analysis of trimethoprim-resistant extraintestinal pathogenic Escherichia coli and recurrent urinary tract infections. Microb Genom 2020; 6:mgen000475. [PMID: 33206038 PMCID: PMC8116683 DOI: 10.1099/mgen.0.000475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Urinary tract infections (UTIs) are the most common bacterial infections requiring medical attention and a leading justification for antibiotic prescription. Trimethoprim is prescribed empirically for uncomplicated cases. UTIs are primarily caused by extraintestinal pathogenic Escherichia coli (ExPEC) and ExPEC strains play a central role in disseminating antimicrobial-resistance genes worldwide. Here, we describe the whole-genome sequences of trimethoprim-resistant ExPEC and/or ExPEC from recurrent UTIs (67 in total) from patients attending a regional Australian hospital from 2006 to 2008. Twenty-three sequence types (STs) were observed, with ST131 predominating (28 %), then ST69 and ST73 (both 7 %). Co-occurrence of trimethoprim-resistance genes with genes conferring resistance to extended-spectrum β-lactams, heavy metals and quaternary ammonium ions was a feature of the ExPEC described here. Seven trimethoprim-resistance genes were identified, most commonly dfrA17 (38 %) and dfrA12 (18 %). An uncommon dfrB4 variant was also observed. Two blaCTX-M variants were identified - blaCTX-M-15 (16 %) and blaCTX-M-14 (10 %). The former was always associated with dfrA12, the latter with dfrA17, and all blaCTX-M genes co-occurred with chromate-resistance gene chrA. Eighteen class 1 integron structures were characterized, and chrA featured in eight structures; dfrA genes featured in seventeen. ST131 H30Rx isolates possessed distinct antimicrobial gene profiles comprising aac(3)-IIa, aac(6)-Ib-cr, aph(3')-Ia, aadA2, blaCTX-M-15, blaOXA-1 and dfrA12. The most common virulence-associated genes (VAGs) were fimH, fyuA, irp2 and sitA (all 91 %). Virulence profile clustering showed ST131 H30 isolates carried similar VAGs to ST73, ST405, ST550 and ST1193 isolates. The sole ST131 H27 isolate carried molecular predictors of enteroaggregative E. coli/ExPEC hybrid strains (aatA, aggR, fyuA). Seven isolates (10 %) carried VAGs suggesting ColV plasmid carriage. Finally, SNP analysis of serial UTI patients experiencing worsening sequelae demonstrated a high proportion of point mutations in virulence factors.
Collapse
Affiliation(s)
- Dmitriy Li
- Ithree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Cameron J. Reid
- Ithree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Timothy Kudinha
- NSW Health Pathology, Microbiology, Orange Hospital, Orange, NSW 2800, Australia
- School of Biomedical Sciences, Charles Sturt University, Orange, NSW 2800, Australia
| | - Veronica M. Jarocki
- Ithree Institute, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | | |
Collapse
|
13
|
Bogema DR, McKinnon J, Liu M, Hitchick N, Miller N, Venturini C, Iredell J, Darling AE, Roy Chowdury P, Djordjevic SP. Whole-genome analysis of extraintestinal Escherichia coli sequence type 73 from a single hospital over a 2 year period identified different circulating clonal groups. Microb Genom 2020; 6. [PMID: 30810518 PMCID: PMC7067039 DOI: 10.1099/mgen.0.000255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sequence type (ST)73 has emerged as one of the most frequently isolated extraintestinal pathogenic Escherichia coli. To examine the localized diversity of ST73 clonal groups, including their mobile genetic element profile, we sequenced the genomes of 16 multiple-drug resistant ST73 isolates from patients with urinary tract infection from a single hospital in Sydney, Australia, between 2009 and 2011. Genome sequences were used to generate a SNP-based phylogenetic tree to determine the relationship of these isolates in a global context with ST73 sequences (n=210) from public databases. There was no evidence of a dominant outbreak strain of ST73 in patients from this hospital, rather we identified at least eight separate groups, several of which reoccurred, over a 2 year period. The inferred phylogeny of all ST73 strains (n=226) including the ST73 clone D i2 reference genome shows high bootstrap support and clusters into four major groups that correlate with serotype. The Sydney ST73 strains carry a wide variety of virulence-associated genes, but the presence of iss, pic and several iron-acquisition operons was notable.
Collapse
Affiliation(s)
- D R Bogema
- Elizabeth Macarthur Agricultural Institute, NSW Department of Primary Industries, Menangle, NSW 2568, Australia.,The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| | - J McKinnon
- The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| | - M Liu
- The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| | - N Hitchick
- San Pathology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - N Miller
- San Pathology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - C Venturini
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - J Iredell
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - A E Darling
- The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| | - P Roy Chowdury
- The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| | - S P Djordjevic
- The ithree Institute, University of Technology Sydney, NSW 2007, Australia
| |
Collapse
|
14
|
Virulence, phylogenetic background and antimicrobial resistance in Escherichia coli associated with extraintestinal infections. J Infect Public Health 2020; 13:1537-1543. [DOI: 10.1016/j.jiph.2020.06.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/05/2020] [Accepted: 06/25/2020] [Indexed: 01/20/2023] Open
|
15
|
Coolen JPM, den Drijver EPM, Kluytmans JAJW, Verweij JJ, Lamberts BA, Soer JACJ, Verhulst C, Wertheim HFL, Kolwijck E. Development of an algorithm to discriminate between plasmid- and chromosomal-mediated AmpC β-lactamase production in Escherichia coli by elaborate phenotypic and genotypic characterization. J Antimicrob Chemother 2020; 74:3481-3488. [PMID: 31504559 PMCID: PMC7183348 DOI: 10.1093/jac/dkz362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES AmpC-β-lactamase production is an under-recognized antibiotic resistance mechanism that renders Gram-negative bacteria resistant to common β-lactam antibiotics, similar to the well-known ESBLs. For infection control purposes, it is important to be able to discriminate between plasmid-mediated AmpC (pAmpC) production and chromosomal-mediated AmpC (cAmpC) hyperproduction in Gram-negative bacteria as pAmpC requires isolation precautions to minimize the risk of horizontal gene transmission. Detecting pAmpC in Escherichia coli is challenging, as both pAmpC production and cAmpC hyperproduction may lead to third-generation cephalosporin resistance. METHODS We tested a collection of E. coli strains suspected to produce AmpC. Elaborate susceptibility testing for third-generation cephalosporins, WGS and machine learning were used to develop an algorithm to determine ampC genotypes in E. coli. WGS was applied to detect pampC genes, cAmpC hyperproducers and STs. RESULTS In total, 172 E. coli strains (n=75 ST) were divided into a training set and two validation sets. Ninety strains were pampC positive, the predominant gene being blaCMY-2 (86.7%), followed by blaDHA-1 (7.8%), and 59 strains were cAmpC hyperproducers. The algorithm used a cefotaxime MIC value above 6 mg/L to identify pampC-positive E. coli and an MIC value of 0.5 mg/L to discriminate between cAmpC-hyperproducing and non-cAmpC-hyperproducing E. coli strains. Accuracy was 0.88 (95% CI=0.79-0.94) on the training set, 0.79 (95% CI=0.64-0.89) on validation set 1 and 0.85 (95% CI=0.71-0.94) on validation set 2. CONCLUSIONS This approach resulted in a pragmatic algorithm for differentiating ampC genotypes in E. coli based on phenotypic susceptibility testing.
Collapse
Affiliation(s)
- Jordy P M Coolen
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evert P M den Drijver
- Department of Infection Control, Amphia Ziekenhuis, Breda, The Netherlands.,Laboratory for Medical Microbiology and Immunology, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Jan A J W Kluytmans
- Department of Infection Control, Amphia Ziekenhuis, Breda, The Netherlands.,Laboratory for Microbiology, Microvida, Location Breda, The Netherlands.,Julius Center for Health Sciences and Primary Care, UMCU, Utrecht, The Netherlands
| | - Jaco J Verweij
- Laboratory for Medical Microbiology and Immunology, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Bram A Lamberts
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joke A C J Soer
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carlo Verhulst
- Department of Infection Control, Amphia Ziekenhuis, Breda, The Netherlands.,Laboratory for Microbiology, Microvida, Location Breda, The Netherlands
| | - Heiman F L Wertheim
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eva Kolwijck
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Rahmani HK, Tabar GH, Badouei MA, Khoramian B. Development of three multiplex-PCR assays for virulence profiling of different iron acquisition systems in Escherichia coli. IRANIAN JOURNAL OF MICROBIOLOGY 2020; 12:281-288. [PMID: 32994898 PMCID: PMC7502150 DOI: 10.18502/ijm.v12i4.3930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND OBJECTIVES Escherichia coli is responsible for various enteric and extraintestinal infections in animals and humans. Iron as an essential nutrient, has a proven role in pathogenicity of E. coli. Pathogenic E. coli benefits of having complicated systems for iron acquisition but our current knowledge is limited because of complexity of these systems. In the present study, three multiplex-PCR assays were developed to screen nine different virulence genes related to diverse iron acquisition systems in E. coli. MATERIALS AND METHODS The multiplex-PCR systems were designed and optimized in three panels. Each panel includes a triplex-PCR cocktail. The panels are as follow: panel 1: iroN, iutA and fecA; panel 2: fyuA, sitA and irp2; and panel 3: iucD, chuA and tonB. A total of 39 pathogenic E. coli was screened according to the designed multiplex-PCR. RESULTS In total, the top three frequent genes were tonB (100%), fecA (66.6%) and sitA (58.9%). With the exception of fecA and tonB, comparing the prevalence of genes among different origin of isolates (human, cattle, poultry and pigeon) showed significant associations (P < 0.05). Moreover, the iroN, sitA and iucD genes were significantly prevalent (P < 0.05) among members of extraintestinal pathogenic E. coli in comparison with the group of diarrheagenic E. coli. CONCLUSION The current multiplex-PCR assays could be a valuable, rapid and economic tool to investigate diverse iron acquisition systems in E. coli for more precise virulence typing of pathogenic or commensal strains.
Collapse
Affiliation(s)
- Hamideh Kalateh Rahmani
- Department of Pathobiology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Gholamreza Hashemi Tabar
- Department of Pathobiology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mahdi Askari Badouei
- Department of Pathobiology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Babak Khoramian
- Department of Clinical Sciences, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
17
|
Dutra IL, Araújo LG, Assunção RG, Lima YA, Nascimento JR, Vale AAM, Alves PCS, Trovão LO, Santos ACM, Silva RM, Silva LA, Maciel MCG, de Sousa EM, Elias WP, Nascimento FRF, Abreu AG. Pic-Producing Escherichia coli Induces High Production of Proinflammatory Mediators by the Host Leading to Death by Sepsis. Int J Mol Sci 2020; 21:ijms21062068. [PMID: 32197297 PMCID: PMC7139334 DOI: 10.3390/ijms21062068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 11/30/2022] Open
Abstract
Escherichia coli is an important pathogen responsible for a variety of diseases. We have recently shown that Pic, a serine protease secreted by E. coli, mediates immune evasion by the direct cleavage of complement molecules. The aim of this study was to investigate the action of a Pic-producing bacteria in a murine model of sepsis. Mice were infected with Pic-producing E. coli (F5) or F5∆pic mutant. Animal survival was monitored for five days, and a subset of mice was euthanized after 12 h for sample acquisition. The inoculation of Pic-producing bacteria induced 100% death within 24 h. The colony forming units count in the organs was significantly higher in F5. Hematological analysis showed a decrease of total leukocytes. Nitric oxide and cytokines were detected in serum, as well as on peritoneal lavage of the F5 group in higher levels than those detected in the other groups. In addition, immunophenotyping showed a decrease of activated lymphocytes and macrophages in the F5 group. Therefore, Pic represents an important virulence factor, allowing the survival of the bacterium in the bloodstream and several organs, as well as inducing a high production of proinflammatory mediators by the host, and concomitantly a cellular immunosuppression, leading to sepsis and death.
Collapse
Affiliation(s)
- Itaynara L. Dutra
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil; (I.L.D.); (L.G.A.); (R.G.A.)
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
| | - Lorena G. Araújo
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil; (I.L.D.); (L.G.A.); (R.G.A.)
- Programa de Pós-Graduação em Biologia Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
| | - Raissa G. Assunção
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil; (I.L.D.); (L.G.A.); (R.G.A.)
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
| | - Yago A. Lima
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Johnny R. Nascimento
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - André A. M. Vale
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Patrícia C. S. Alves
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Liana O. Trovão
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Ana Carolina M. Santos
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil; (A.C.M.S.); (R.M.S.)
| | - Rosa M. Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil; (A.C.M.S.); (R.M.S.)
| | - Lucilene A. Silva
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Márcia C. G. Maciel
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Departamento de Biologia Celular, Universidade de Brasília, Brasília 70878-040, Brazil
| | - Eduardo M. de Sousa
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Programa de Pós-Graduação em Biologia Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
| | - Waldir P. Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Flávia R. F. Nascimento
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão, São Luís 65080-805, Brazil;
| | - Afonso G. Abreu
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil; (I.L.D.); (L.G.A.); (R.G.A.)
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, Brazil; (J.R.N.); (A.A.M.V.); (P.C.S.A.); (L.O.T.); (L.A.S.); (M.C.G.M.); (E.M.d.S.); (F.R.F.N.)
- Programa de Pós-Graduação em Biologia Microbiana, Universidade Ceuma, São Luís 65075-120, Brazil
- Correspondence:
| |
Collapse
|
18
|
Vieira PCG, Espinoza-Culupú AO, Nepomuceno R, Alves MR, Lebrun I, Elias WP, Ruiz RC. Secreted autotransporter toxin (Sat) induces cell damage during enteroaggregative Escherichia coli infection. PLoS One 2020; 15:e0228959. [PMID: 32084148 PMCID: PMC7034920 DOI: 10.1371/journal.pone.0228959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
Secreted autotransporter toxin (Sat) is a 107-kDa serine protease autotransporter of Enterobacteriaceae (SPATE) presenting cytotoxic activity in renal and bladder cells. Further studies have detected the Sat-encoding gene (sat) in enteroaggregative Escherichia coli (EAEC) and in E. coli strains isolated from neonatal septicemia and meningitis. Here, we investigated the role of Sat as a cytotoxin of EAEC. Sat was purified from a strain of E. coli harboring sat (DEC/Sat+, O126:H2) and used to raise antibodies in rabbit. The presence of Sat was detected by ELISA in the supernatant of 93.7% of EAEC strains harboring sat and in none lacking the gene. The effect of Sat during infection was investigated in polarized Caco-2 cells infected with Sat-producing EAEC (CV323/77, O125ab:H21). This strain induced intense cell detachment, which was inhibited by PMSF or Sat antiserum. Also, sat transcription and Sat production were detected during infection. Here we demonstrate that Sat is internalized in polarized cells leading to F-actin disruption which preceded cell detachment. A comparative study of the toxin action in cell lines corresponding to the infection sites in which bacteria carrying the sat gene have been isolated was performed. Cells originating from the gastrointestinal tract (Caco-2), urinary (LLC-PK1) and endothelium (HUVEC) were incubated with purified Sat. The time required for observation of cell damage differed according to the cell line. HUVEC cells were more sensitive to Sat than cells derived from urinary and intestinal tracts. The intense activity of Sat on the endothelial cells suggests that Sat could also be a virulence factor for the bacteria in the bloodstream. In addition, this is the first work demonstrating that Sat induces cytotoxic effect during EAEC infection in vitro. The cell damage observed during infection indicates that Sat may be another toxin with cytotoxic role in the EAEC pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Marina R. Alves
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| | - Ivo Lebrun
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, SP, Brazil
| | - Waldir P. Elias
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| | - Rita C. Ruiz
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Freire CA, Santos ACM, Pignatari AC, Silva RM, Elias WP. Serine protease autotransporters of Enterobacteriaceae (SPATEs) are largely distributed among Escherichia coli isolated from the bloodstream. Braz J Microbiol 2020; 51:447-454. [PMID: 31965549 DOI: 10.1007/s42770-020-00224-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/07/2020] [Indexed: 12/29/2022] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is the major cause of Gram-negative-related sepsis. Bacterial survival in the bloodstream is mediated by a variety of virulence traits, including those mediating immune system evasion. Serine protease autotransporters of Enterobacteriaceae (SPATE) constitute a superfamily of virulence factors that can cause tissue damage and cleavage of molecules of the complement system, which is a key feature for the establishment of infection in the bloodstream. In this study, we analyzed 278 E. coli strains isolated from human bacteremia from inpatients of both genders, different ages, and clinical conditions. These strains were screened for the presence of SPATE-encoding genes as well as for phylogenetic classification and intrinsic virulence of ExPEC. SPATE-encoding genes were detected in 61.2% of the strains and most of these strains (44.6%) presented distinct SPATE-encoding gene profiles. sat was the most frequent gene among the entire collection, found in 34.2%, followed by vat (28.4%), pic (8.3%), and tsh (4.7%). Although in low frequencies, espC (0.7%), eatA (1.1%), and espI (1.1%) were detected and are being reported for the first time in extraintestinal isolates. The presence of SPATE-encoding genes was positively associated to phylogroup B2 and intrinsic virulent strains. These findings suggest that SPATEs are highly prevalent and involved in diverse steps of the pathogenesis of bacteremia caused by E. coli.
Collapse
Affiliation(s)
- Claudia A Freire
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brazil 1500, São Paulo, SP, 05503-900, Brazil
| | - Ana Carolina M Santos
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Antonio C Pignatari
- Laboratório Especial de Microbiologia Clínica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Rosa M Silva
- Disciplina de Microbiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Waldir P Elias
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brazil 1500, São Paulo, SP, 05503-900, Brazil.
| |
Collapse
|
20
|
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) strains are responsible for a majority of human extraintestinal infections globally, resulting in enormous direct medical and social costs. ExPEC strains are comprised of many lineages, but only a subset is responsible for the vast majority of infections. Few systematic surveillance systems exist for ExPEC. To address this gap, we systematically reviewed and meta-analyzed 217 studies (1995 to 2018) that performed multilocus sequence typing or whole-genome sequencing to genotype E. coli recovered from extraintestinal infections or the gut. Twenty major ExPEC sequence types (STs) accounted for 85% of E. coli isolates from the included studies. ST131 was the most common ST from 2000 onwards, covering all geographic regions. Antimicrobial resistance-based isolate study inclusion criteria likely led to an overestimation and underestimation of some lineages. European and North American studies showed similar distributions of ExPEC STs, but Asian and African studies diverged. Epidemiology and population dynamics of ExPEC are complex; summary proportion for some STs varied over time (e.g., ST95), while other STs were constant (e.g., ST10). Persistence, adaptation, and predominance in the intestinal reservoir may drive ExPEC success. Systematic, unbiased tracking of predominant ExPEC lineages will direct research toward better treatment and prevention strategies for extraintestinal infections.
Collapse
|
21
|
Daga AP, Koga VL, Soncini JGM, de Matos CM, Perugini MRE, Pelisson M, Kobayashi RKT, Vespero EC. Escherichia coli Bloodstream Infections in Patients at a University Hospital: Virulence Factors and Clinical Characteristics. Front Cell Infect Microbiol 2019; 9:191. [PMID: 31245301 PMCID: PMC6563721 DOI: 10.3389/fcimb.2019.00191] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/20/2019] [Indexed: 01/24/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) isolates are responsible for many bloodstream infections. The aim of this study was to characterize E. coli isolated from the bloodstreams of patients (n = 48) at the University Hospital in Brazil. Epidemiological data were obtained through the analysis of medical records and laboratory tests. By PCR analysis, we investigated the presence of virulence factors (VFs), pathogenicity islands (PAIs), extended-spectrum β-lactamase (ESBL), phylogenetic classifications (A, B1, B2, C, D, E, and F) and molecular genotype by enterobacterial repetitive intergenic consensus-polymerase chain reaction (ERIC-PCR). The mortality analysis showed that 33.3% of the deaths were associated with bacteraemia due to E. coli infections; in addition, an age between 60 and 75 years (p < 0.001; OR = 6.3[2.1–18.9]) and bacteraemia with an abdominal origin (p = 0.02; OR = 5[1.2–20.5]) were risk factors for the severity of the infection. Additionally, the presence of the afa gene was associated with mortality due to E. coli bacteraemia (p = 0.027; OR = 11.4[1.5–85.7]). Immunosuppression (27.1%), intestinal diseases (25.0%) and diabetes (18.8%), were prevalent among patients, and most of the bacteraemia cases were secondary to urinary tract infections (50.0%). The serum resistance gene traT was present in 77.1% of isolates, group capsular 2 (kpsMT II) was present in 45.8% and the K5 capsule was present in 20.8% of isolates. The isolates also showed a high prevalence for the siderophore yersiniabactina (fyuA) (70.8%) and PAI IV536 (77.1%). Phylogenetic analysis showed that group B2 (45.8%) was the most prevalent, and was the phylogroup that had a higher prevalence of VFs and PAIs. However, in this study, a considerable number of isolated bacteria were classified as group B1 (18.8%) and as group E (14.6%). Eight (16.7%) isolates were resistant to third and fourth generation cephalosporin and group CTX-M-1 (CTX-M-15) was the most prevalent ESBL type. The molecular genotyping showed two clonal lineages and several isolates that were not related to each other. This study provides additional information on the epidemiological and molecular characteristics of E. coli bloodstream infections in Brazil.
Collapse
Affiliation(s)
- Ana Paula Daga
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Vanessa Lumi Koga
- Laboratory of Microbiology, Department of Microbiology, State University of Londrina, Londrina, Brazil
| | - João Gabriel Material Soncini
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Caroline Martins de Matos
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Marcia Regina Eches Perugini
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Marsileni Pelisson
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | | | - Eliana Carolina Vespero
- Laboratory of Clinical Microbiology, Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, State University of Londrina, Londrina, Brazil
| |
Collapse
|
22
|
Bozcal E, Eldem V, Aydemir S, Skurnik M. The relationship between phylogenetic classification, virulence and antibiotic resistance of extraintestinal pathogenic Escherichia coli in İzmir province, Turkey. PeerJ 2018; 6:e5470. [PMID: 30155366 PMCID: PMC6110251 DOI: 10.7717/peerj.5470] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/21/2018] [Indexed: 12/25/2022] Open
Abstract
Background Extraintestinal pathogenic Escherichia coli (ExPEC) is an important bacterium and responsible for many bloodstream infections, including urinary tract infections and even fatal bacteremia. The aim of this research was to investigate whether ExPEC strains isolated from Turkish blood cultures have a relationship between 16S rRNA based phylogenetic clusters and antibiotic resistance profiles, virulence factors or clonal lineages. Methods Phenotypically identified ExPEC blood culture isolates (n = 104) were included in this study. The 16S rRNA partial sequence analysis was performed for genotypic identification of ExPEC isolates. Antibiotic susceptibility and Extended-Spectrum β-Lactamase testing of isolates were performed. Phylogenetic classification (A, B1, B2 and D), Multi Locus Sequence Typing analysis and virulence-associated genes were investigated. Results Based on 16S rRNA partial sequence analysis, 97 out of 104 (93.26%) ExPEC isolates were confirmed as E. coli. Ampicillin (74.22%) and cefuroxime axetil (65.97%) resistances had the highest frequencies among the ExPEC isolates. In terms of phylogenetic classification of ExPEC, D (38.14%, 37/97) was the most prevalent group after A (29.89%, 29/97), B2 (20.61%, 20/97), and B1 (11.34%, 11/97). The sequence types of the 20 ExPEC isolates belonging to the B2 phylogenetic group were analyzed by Multi Locus Sequence Typing. Ten isolates out of 20 (50.0%) were identified as ST131. The other STs were ST95 (n = 1), ST14 (n = 1), ST10 (n = 1), ST69 (n = 1), ST1722 (n = 2), ST141 (n = 1), ST88 (n = 1), ST80 (n = 1), and ST998 (n = 1). Of the ST131 strains, six (60%, 6/10) represented serogroup O25. The most common virulence factor genes were serum resistance factor gene, traT (55.7%) aerobactin siderophore receptor and yersiniabactin encoding genes iutA (45.3%) and fyuA (50.5%), respectively. In addition, PAI (41.2%), iroN (23.7%), hlyA (15.4%), kpsII (13.4%), ompT (13.4%), papG (12.4%), iss (9.3%), cnf1 (7.2%), ibeA (2.06%), and sfaS (2.06%) genes were present in the ExPEC isolates. Conclusion The 16S rRNA-based phylogenetic relationship tree analysis showed that a large cluster was present among 97 ExPEC isolates along with related reference strains. There were 21 main clusters with 32 closely related subclusters. Based on our findings, different clonal lineages of ExPEC can display different antibiotic susceptibilities and virulence properties. We also concluded that virulence factors were not distributed depending on phylogenetic groups (A, B1, B2, and D). The ExPEC isolates belonging to the same phylogenetic group and sequence type could display different resistance and virulence characteristics.
Collapse
Affiliation(s)
- Elif Bozcal
- Department of Biology, Basic and Industrial Microbiology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Vahap Eldem
- Department of Biology, Zoology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Sohret Aydemir
- Department of Medical Microbiology, Faculty of Medicine, Ege University, İzmir, Turkey
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, Medicum and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Kato K, Mizumoto H, Matsubara K, Hata A, Wachino JI, Arakawa Y, Hata D. Recurrence of Escherichia coli meningitis in a preterm infant and co-infection of echovirus 18. IDCases 2017; 10:135-137. [PMID: 29181300 PMCID: PMC5695537 DOI: 10.1016/j.idcr.2017.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 11/29/2022] Open
Abstract
Introduction Bacterial meningitis may relapse after adequate antibiotic treatment. In most cases, however, the pathophysiology cannot be identified. Presentation of case We describe a preterm infant with recurrent episodes of meningitis due to infection with an identical Escherichia coli strain both at birth and at 10 days after cessation of a 3 week course of appropriate antibiotic treatment. At the time of recurrence, the patient presented with fulminant severe cardiac failure due to acute myocarditis, coupled with a concurrent echovirus 18 infection (confirmed by stool culture and serological analysis). Conclusion Co-infection by echovirus may underlie recurrence of Escherichia coli meningitis in this case.
Collapse
Affiliation(s)
- Kentaro Kato
- Department of Pediatrics, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Hiroshi Mizumoto
- Department of Pediatrics, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | | | - Atsuko Hata
- Department of Pediatrics, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Jun-Ichi Wachino
- Department of Bacteriology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshichika Arakawa
- Department of Bacteriology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hata
- Department of Pediatrics, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| |
Collapse
|
24
|
Kallonen T, Brodrick HJ, Harris SR, Corander J, Brown NM, Martin V, Peacock SJ, Parkhill J. Systematic longitudinal survey of invasive Escherichia coli in England demonstrates a stable population structure only transiently disturbed by the emergence of ST131. Genome Res 2017; 27:1437-1449. [PMID: 28720578 PMCID: PMC5538559 DOI: 10.1101/gr.216606.116] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 06/07/2017] [Indexed: 12/19/2022]
Abstract
Escherichia coli associated with urinary tract infections and bacteremia has been intensively investigated, including recent work focusing on the virulent, globally disseminated, multidrug-resistant lineage ST131. To contextualize ST131 within the broader E. coli population associated with disease, we used genomics to analyze a systematic 11-yr hospital-based survey of E. coli associated with bacteremia using isolates collected from across England by the British Society for Antimicrobial Chemotherapy and from the Cambridge University Hospitals NHS Foundation Trust. Population dynamics analysis of the most successful lineages identified the emergence of ST131 and ST69 and their establishment as two of the five most common lineages along with ST73, ST95, and ST12. The most frequently identified lineage was ST73. Compared to ST131, ST73 was susceptible to most antibiotics, indicating that multidrug resistance was not the dominant reason for prevalence of E. coli lineages in this population. Temporal phylogenetic analysis of the emergence of ST69 and ST131 identified differences in the dynamics of emergence and showed that expansion of ST131 in this population was not driven by sequential emergence of increasingly resistant subclades. We showed that over time, the E. coli population was only transiently disturbed by the introduction of new lineages before a new equilibrium was rapidly achieved. Together, these findings suggest that the frequency of E. coli lineages in invasive disease is driven by negative frequency-dependent selection occurring outside of the hospital, most probably in the commensal niche, and that drug resistance is not a primary determinant of success in this niche.
Collapse
Affiliation(s)
- Teemu Kallonen
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Hayley J Brodrick
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Simon R Harris
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Jukka Corander
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
- Department of Mathematics and Statistics, University of Helsinki, 00014 Helsinki, Finland
- Department of Biostatistics, University of Oslo, 0372 Oslo, Norway
| | - Nicholas M Brown
- Public Health England, Clinical Microbiology and Public Health Laboratory, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Veronique Martin
- British Society of Antimicrobial Chemotherapy, Birmingham B1 3NJ, United Kingdom
| | - Sharon J Peacock
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Julian Parkhill
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
25
|
de Souza da-Silva AP, de Sousa VS, Martins N, da Silva Dias RC, Bonelli RR, Riley LW, Moreira BM. Escherichia coli sequence type 73 as a cause of community acquired urinary tract infection in men and women in Rio de Janeiro, Brazil. Diagn Microbiol Infect Dis 2017; 88:69-74. [PMID: 28214224 PMCID: PMC5392417 DOI: 10.1016/j.diagmicrobio.2017.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/25/2017] [Accepted: 01/31/2017] [Indexed: 12/22/2022]
Abstract
Escherichia coli clones ST131, ST69, ST95, and ST73 are frequent causes of urinary tract infections (UTI) and bloodstream infections. Specific clones and virulence profiles of E. coli causing UTI in men has been rarely described. The aim of this study was to characterize patient and clonal characteristics of community-acquired UTI caused by E. coli in men (n=12) and women (n=127) in Rio de Janeiro, Brazil, complementing a previous work. We characterized isolates in phylogenetic groups, ERIC2-PCR and PFGE types, MLST, genome similarity and virulence gene-profiles. UTI from men were more frequently caused by phylogenetic group B2 isolates (83% versus 42%, respectively, P = 0.01), a group with significantly higher virulence scores compared with women. ST73 was the predominant clone in men (50%) and the second most frequent in women (12%), with the highest virulence score (mean and median=9) among other clones. ST73 gnomes formed at least six clusters. E. coli from men carried significantly higher numbers of virulence genes, such as sfa/focDE (67% versus 27%), hlyA (58% versus 24%), cnf 1 (58% versus 16%), fyuA (100% versus 82%) and MalX (92% versus 44%), compared with isolates from women. These data suggest the predominance and spread of ST73 isolates likely relates to an abundance of virulence determinants.
Collapse
Affiliation(s)
| | - Viviane Santos de Sousa
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Rio de Janeiro, 21941-902, Brazil
| | - Natacha Martins
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Rio de Janeiro, 21941-902, Brazil
| | | | - Raquel Regina Bonelli
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Rio de Janeiro, 21941-902, Brazil
| | - Lee W Riley
- School of Public Health, University of California, Berkeley, CA
| | - Beatriz Meurer Moreira
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
26
|
Mac Aogáin M, Miajlovic H, Moloney G, Chotirmall SH, Rogers TR, Smith SGJ. Identification of a novel sequence type of Escherichia coli as the causative agent of pyelonephritis and bloodstream infection. JMM Case Rep 2017; 3:e005061. [PMID: 28348784 PMCID: PMC5343142 DOI: 10.1099/jmmcr.0.005061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/06/2016] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Globally, extra-intestinal pathogenic Escherichia coli are one of the predominant causative agents of bacteraemia. CASE PRESENTATION This case report outlines a presentation of community-acquired pyelonephritis and secondary bloodstream infection in an 81-year-old man. Laboratory investigations revealed that the causative isolate was a multi-drug-resistant E. coli of a novel multi-locus sequence type. This sequence type (ST) was designated ST-458 and was most closely related to the globally prevalent ST-131 lineage. CONCLUSION This is the first report of a novel E. coli ST, ST-458, which caused pyelonephritis and bacteraemia.
Collapse
Affiliation(s)
- Micheál Mac Aogáin
- Department of Clinical Microbiology, Sir Patrick Dun Research Laboratory, School of Medicine, Trinity College , Dublin , Ireland
| | - Helen Miajlovic
- Department of Clinical Microbiology, Sir Patrick Dun Research Laboratory, School of Medicine, Trinity College , Dublin , Ireland
| | - Geraldine Moloney
- Department of Clinical Microbiology, Sir Patrick Dun Research Laboratory, School of Medicine, Trinity College , Dublin , Ireland
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Thomas R Rogers
- Department of Clinical Microbiology, Sir Patrick Dun Research Laboratory, School of Medicine, Trinity College , Dublin , Ireland
| | - Stephen G J Smith
- Department of Clinical Microbiology, Sir Patrick Dun Research Laboratory, School of Medicine, Trinity College , Dublin , Ireland
| |
Collapse
|