1
|
Zhong Q, Nasreen M, Yang R, Struwe M, Kobe B, Kappler U. Beyond anaerobic respiration-new physiological roles for DmsABC and other S-/N-oxide reductases in Escherichia coli. J Bacteriol 2025; 207:e0046324. [PMID: 40162811 PMCID: PMC12096835 DOI: 10.1128/jb.00463-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Sulfoxide reductases in pathogenic bacteria have recently received increasing attention for their association with virulence and survival within the host. Here, we have re-investigated the physiological role of the molybdenum-containing DmsABC dimethyl sulfoxide (DMSO) reductase from Escherichia coli, which has a proposed role in anaerobic respiration with DMSO. Our investigation into potential physiological substrates revealed that DmsABC efficiently reduces pyrimidine N-oxide, nicotinamide N-oxide, and methionine sulfoxide, and exposure to host cell-produced stressors such as hypochlorite or hydrogen peroxide specifically increased expression of the E. coli dmsA gene. E. coli strains lacking dmsA showed increased lag times in the presence of hypochlorite, and these strains also showed up to a 90% reduction in adherence to human bladder cells. Interestingly, in the presence of hypochlorite, expression of multiple alternative S-/N-oxide reductases present in E. coli was elevated by 2- to 4-fold in a ∆dmsA strain compared to the wild-type strain, suggesting functional redundancy. The phenotypes of the E. coli ∆dmsA strains were strikingly similar to ∆dmsA strains of the respiratory pathogen Haemophilus influenzae, which confirms the role of both enzymes in supporting host-pathogen interactions. We propose that this function is conserved in enzymes closely related to E. coli DmsABC. Our study also uncovered that the expression of many E. coli Mo enzymes was induced by oxidative stressors, including metals such as copper, and further work should be directed at determining the connection of these enzymes to host-pathogen interactions.IMPORTANCEBacterial urinary tract infections are debilitating and frequently recurring in human populations worldwide, and Escherichia coli strains are a major cause of these infections. In this study, we have uncovered a new mechanism by which E. coli can avoid being killed by the human immune system. The bacteria use a set of seven related enzymes that can reverse damage to essential cell components such as amino acids, vitamins, and DNA building blocks. Antibacterial compounds produced by the human immune system specifically induced the production of these enzymes, confirming that they play a role in helping E. coli survive during infection and making these enzymes potential future drug targets.
Collapse
Affiliation(s)
- Qifeng Zhong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Marufa Nasreen
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ruizhe Yang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Michel Struwe
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
2
|
Tsai YY, Ienes Lima J, Alvarez Narvaez S, Logue CM. Whole-genome analysis of five Escherichia coli strains isolated from focal duodenal necrosis in laying hens reveals genetic similarities to the E. coli O25:H4 ST131 strain. Microbiol Spectr 2025; 13:e0211024. [PMID: 40162772 PMCID: PMC12054123 DOI: 10.1128/spectrum.02110-24] [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: 08/23/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Focal duodenal necrosis (FDN) is an intestinal disease causing significant economic losses in the table-egg industry due to reduced egg production in laying hens. Its etiology and pathogenesis remain poorly understood. Between 2021 and 2023, 111 Escherichia coli isolates were collected from FDN lesions and screened for the presence of virulence genes using PCR panels. Five strains-FDN-4, FDN-9, FDN-11, FDN-24, and FDN-50-were selected for whole-genome sequencing due to their high virulence gene content. Core-genome analyses found that the five FDN E. coli belong to different phylogroups and strain types (ST), but they all share multiple complete operons involved in key pathogenic functions, including host cell adhesion and invasion, iron acquisition, motility, biofilm formation, and acid resistance. Comparative genomic analyses identified FDN-4 as the most genetically distinct strain, closely resembling EC958, an O25b:H4 ST131 uropathogenic E. coli (UPEC) commonly associated with extended-spectrum beta-lactamase production. FDN-4 and EC958 share unique chromosomal virulence genes absent in the other FDN strains, all located within genomic islands. This study provides the first complete genomic characterization of E. coli isolated from FDN lesions and highlights FDN-4 as a genetically distinct strain with similarities to O25b:H4 ST131 UPEC.IMPORTANCEThis study presents the first complete genomic characterization of Escherichia coli isolated from focal duodenal necrosis (FDN) lesions. Notably, FDN-4 is the first E. coli strain from a poultry disease (FDN) to show significant similarity to O25b:H4 ST131 strains, commonly classified as uropathogenic E. coli and often associated with extended-spectrum beta-lactamase production. However, caution is warranted when attributing direct transmission routes between poultry and humans.
Collapse
Affiliation(s)
- Yu-Yang Tsai
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Julia Ienes Lima
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Sonsiray Alvarez Narvaez
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Catherine M. Logue
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
3
|
Obonyo NG, Raman S, Suen JY, Peters KM, Phan MD, Passmore MR, Bouquet M, Wilson ES, Hyslop K, Palmieri C, White N, Sato K, Farah SM, Gandini L, Liu K, Fior G, Heinsar S, Ijuin S, Kyun Ro S, Abbate G, Ainola C, Sato N, Lundon B, Portatadino S, Rachakonda RH, Schneider B, Harley A, See Hoe LE, Schembri MA, Li Bassi G, Fraser JF. An ovine septic shock model of live bacterial infusion. Intensive Care Med Exp 2024; 12:94. [PMID: 39467921 PMCID: PMC11519284 DOI: 10.1186/s40635-024-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Escherichia coli is the most common cause of human bloodstream infections and bacterial sepsis/septic shock. However, translation of preclinical septic shock resuscitative therapies remains limited mainly due to low-fidelity of available models in mimicking clinical illness. To overcome the translational barrier, we sought to replicate sepsis complexity by creating an acutely critically-ill preclinical bacterial septic shock model undergoing active 48-h intensive care management. AIM To develop a clinically relevant large-animal (ovine) live-bacterial infusion model for septic shock. METHODS Septic shock was induced by intravenous infusion of the live antibiotic resistant extra-intestinal pathogenic E. coli sequence type 131 strain EC958 in eight anesthetised and mechanically ventilated sheep. A bacterial dose range of 2 × 105-2 × 109 cfu/mL was used for the dose optimisation phase (n = 4) and upon dose confirmation the model was developed (n = 5). Post-shock the animals underwent an early-vasopressor and volume-restriction resuscitation strategy with active haemodynamic management and monitoring over 48 h. Serial blood samples were collected for testing of pro-inflammatory (IL-6, IL-8, VEGFA) and anti-inflammatory (IL-10) cytokines and hyaluronan assay to assess endothelial integrity. Tissue samples were collected for histopathology and transmission electron microscopy. RESULTS The 2 × 107 cfu/mL bacterial dose led to a reproducible distributive shock within a pre-determined 12-h period. Five sheep were used to demonstrate consistency of the model. Bacterial infusion led to development of septic shock in all animals. The baseline mean arterial blood pressure reduced from a median of 91 mmHg (71, 102) to 50 mmHg (48, 57) (p = 0.004) and lactate levels increased from a median of 0.5 mM (0.3, 0.8) to 2.1 mM (2.0, 2.3) (p = 0.02) post-shock. The baseline median hyaluronan levels increased significantly from 25 ng/mL (18, 86) to 168 ng/mL (86, 569), p = 0.05 but not the median vasopressor dependency index which increased within 1 h of resuscitation from zero to 0.39 mmHg-1 (0.06, 5.13), p = 0.065, and. Over the 48 h, there was a significant decrease in the systemic vascular resistance index (F = 7.46, p = 0.01) and increase in the pro-inflammatory cytokines [IL-6 (F = 8.90, p = 0.02), IL-8 (F = 5.28, p = 0.03), and VEGFA (F = 6.47, p = 0.02)]. CONCLUSIONS This critically ill large-animal model was consistent in reproducing septic shock and will be applied in investigating advanced resuscitation and therapeutic interventions.
Collapse
Affiliation(s)
- Nchafatso G Obonyo
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia.
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- KEMRI-Wellcome Trust Research Programme and Initiative to Develop African Research Leaders, Kilifi, Kenya.
- Wellcome Trust Centre for Global Health Research, Imperial College London, London, UK.
| | - Sainath Raman
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Children's Intensive Care Research Program, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD, Australia
- Queensland Paediatric Sepsis Program, Children's Health and Youth Network, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Kate M Peters
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Mahe Bouquet
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Chiara Palmieri
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, QLD, Australia
| | - Nicole White
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Samia M Farah
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Lucia Gandini
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Gabriele Fior
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Department of Intensive Care, North Estonia Medical Centre, Tallinn, Estonia
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia
| | - Shinichi Ijuin
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Sun Kyun Ro
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Brooke Lundon
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Sofia Portatadino
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Reema H Rachakonda
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Bailey Schneider
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda Harley
- Children's Intensive Care Research Program, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Paediatric Sepsis Program, Children's Health and Youth Network, Children's Health Queensland, Brisbane, Queensland, Australia
- Critical Care Nursing Management Team, Queensland Children's Hospital, Brisbane, QLD, Australia
- School of Nursing, Midwifery and Social Work, University of Queensland, Brisbane, QLD, Australia
| | - Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Mark A Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia.
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Chorro L, Ciolino T, Torres CL, Illenberger A, Aglione J, Corts P, Lypowy J, Ponce C, La Porte A, Burt D, Volberg GL, Ramaiah L, McGovern K, Hu J, Anderson AS, Silmon de Monerri NC, Kanevsky I, Donald RGK. A cynomolgus monkey E. coli urinary tract infection model confirms efficacy of new FimH vaccine candidates. Infect Immun 2024; 92:e0016924. [PMID: 39297649 PMCID: PMC11475676 DOI: 10.1128/iai.00169-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 10/16/2024] Open
Abstract
The increase in urinary tract infections (UTI) caused by antibiotic-resistant Escherichia coli requires the development of new therapeutic agents and prophylactic vaccines. To evaluate the efficacy of new lead candidates, we implemented a cynomolgus macaque UTI challenge model that mimics human uncomplicated cystitis in response to transurethral challenge with a multidrug-resistant (MDR) E. coli serotype O25b ST131 isolate. E. coli fimbrial adhesin FimH and O-antigens are separately under clinical evaluation by others as vaccine candidates to prevent UTI and invasive urosepsis disease, respectively. Accordingly, we assessed the protective efficacy of three 50-µg intramuscular doses of a novel recombinant FimH antigen adjuvanted with liposomal QS21/MPLA compared with saline placebo in groups of nine animals. A third group was vaccinated with this FimH formulation in combination with 1 µg each of a four-valent mixture of serotype O1a, O2, O6, and O25b O-antigen CRM197 lattice glycoconjugates. Both vaccines elicited high levels of serum FimH IgG and adhesin blocking antibodies at the time of bacterial challenge and, for the combination group, O-antigen-specific antibodies. Following bacterial challenge, both vaccinated groups showed >200- and >700-fold reduction in bacteriuria at day 2 and day 7 post-infection compared with placebo, respectively. In parallel, both vaccines significantly reduced levels of inflammatory biomarkers IL-8 and myeloperoxidase in the urine at day 2 post-infection relative to placebo. Results provide preclinical proof-of-concept for the prevention of an MDR UTI infection by these new vaccine formulations.
Collapse
Affiliation(s)
- Laurent Chorro
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - Tara Ciolino
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | | | | | - JohnPaul Aglione
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - Paula Corts
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | | | | | | | - Deborah Burt
- Pfizer Drug Safety Research and Development, Groton, Connecticut, USA
| | | | - Lila Ramaiah
- Pfizer Drug Safety Research and Development, Pearl River, New York, USA
| | - Kathryn McGovern
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | - Jianfang Hu
- Pfizer Research Biostatistics, Collegeville, Pennsylvania, USA
| | | | | | - Isis Kanevsky
- Pfizer Vaccine Research and Development, Pearl River, New York, USA
| | | |
Collapse
|
5
|
Nhu NTK, Forde BM, Ben Zakour NL, Phan MD, Roberts LW, Beatson SA, Schembri MA. Evolution of the pheV-tRNA integrated genomic island in Escherichia coli. PLoS Genet 2024; 20:e1011459. [PMID: 39446883 PMCID: PMC11537424 DOI: 10.1371/journal.pgen.1011459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/05/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Escherichia coli exhibit extensive genetic diversity at the genome level, particularly within their accessory genome. The tRNA integrated genomic islands (GIs), a part of the E. coli accessory genome, play an important role in pathogenicity. However, studies examining the evolution of GIs have been challenging due to their large size, considerable gene content variation and fragmented assembly in draft genomes. Here we examined the evolution of the GI integrated at pheV-tRNA (GI-pheV), with a primary focus on uropathogenic E. coli (UPEC) and the globally disseminated multidrug resistant ST131 clone. We show the gene content of GI-pheV is highly diverse and arranged in a modular configuration, with the P4 integrase encoding gene intP4 the only conserved gene. Despite this diversity, the GI-pheV gene content displayed conserved features among strains from the same pathotype. In ST131, GI-pheV corresponding to the reference strain EC958 (EC958_GI-pheV) was found in ~90% of strains. Phylogenetic analyses suggested that GI-pheV in ST131 has evolved together with the core genome, with the loss/gain of specific modules (or the entire GI) linked to strain specific events. Overall, we show GI-pheV exhibits a dynamic evolutionary pathway, in which modules and genes have evolved through multiple events including insertions, deletions and recombination.
Collapse
Affiliation(s)
- Nguyen Thi Khanh Nhu
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Brian M. Forde
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nouri L. Ben Zakour
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Leah W. Roberts
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Scott A. Beatson
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark A. Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Henry SA, Webster CM, Shaw LN, Torres NJ, Jobson ME, Totzke BC, Jackson JK, McGreig JE, Wass MN, Robinson GK, Shepherd M. Steroid Drugs Inhibit Bacterial Respiratory Oxidases and Are Lethal Toward Methicillin-Resistant Staphylococcus aureus. J Infect Dis 2024; 230:e149-e158. [PMID: 39052707 PMCID: PMC11272085 DOI: 10.1093/infdis/jiad540] [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: 07/21/2023] [Accepted: 11/28/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Cytochrome bd complexes are respiratory oxidases found exclusively in prokaryotes that are important during infection for numerous bacterial pathogens. METHODS In silico docking was employed to screen approved drugs for their ability to bind to the quinol site of Escherichia coli cytochrome bd-I. Respiratory inhibition was assessed with oxygen electrodes using membranes isolated from E. coli and methicillin-resistant Staphylococcus aureus strains expressing single respiratory oxidases (ie, cytochromes bd, bo', or aa3). Growth/viability assays were used to measure bacteriostatic and bactericidal effects. RESULTS The steroid drugs ethinylestradiol and quinestrol inhibited E. coli bd-I activity with median inhibitory concentration (IC50) values of 47 ± 28.9 µg/mL (158 ± 97.2 µM) and 0.2 ± 0.04 µg/mL (0.5 ± 0.1 µM), respectively. Quinestrol inhibited growth of an E. coli "bd-I only" strain with an IC50 of 0.06 ± 0.02 µg/mL (0.2 ± 0.07 µM). Growth of an S. aureus "bd only" strain was inhibited by quinestrol with an IC50 of 2.2 ± 0.43 µg/mL (6.0 ± 1.2 µM). Quinestrol exhibited potent bactericidal effects against S. aureus but not E. coli. CONCLUSIONS Quinestrol inhibits cytochrome bd in E. coli and S. aureus membranes and inhibits the growth of both species, yet is only bactericidal toward S. aureus.
Collapse
Affiliation(s)
- Samantha A Henry
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Calum M Webster
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Lindsey N Shaw
- Department of Molecular Biosciences, University of South Florida, Tampa
| | | | | | - Brendan C Totzke
- Department of Molecular Biosciences, University of South Florida, Tampa
| | - Jessica K Jackson
- Department of Molecular Biosciences, University of South Florida, Tampa
| | - Jake E McGreig
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Mark N Wass
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Gary K Robinson
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Mark Shepherd
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
7
|
Varney AM, Mannix-Fisher E, Thomas JC, McLean S. Evaluation of phenotypic and genotypic methods for the identification and characterization of bacterial isolates recovered from catheter-associated urinary tract infections. J Appl Microbiol 2024; 135:lxae155. [PMID: 38925648 DOI: 10.1093/jambio/lxae155] [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: 12/17/2023] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 06/28/2024]
Abstract
AIMS Urinary tract infections are the most common hospital-acquired infection, 80% of which are associated with catheterization. Diagnostic methods may influence the reported identities of these pathogens, and phenotypic testing under laboratory conditions may not reflect infection phenotypes. This study aimed to evaluate the efficacy of diagnostic methods and whether medium composition alters phenotypes by characterizing catheter-associated urinary tract infection isolates from a UK hospital. METHODS AND RESULTS We compared five bacterial identification methods, including biochemical testing, matrix-assisted laser desorption/ionization biotyping, and genome sequencing, finding differences in genus- or species-level identifications. Antibiotic susceptibility comparisons between phenotypic assays and genomic predictions showed high agreement only in multidrug-resistant strains. To determine whether growth rate and biofilm formation were affected by medium composition, strains were grown in both planktonic and biofilm states. Low planktonic growth and significant biofilm formation were observed in artificial urine compared to rich laboratory media, underscoring the importance of assay design. CONCLUSIONS This study highlights the risks of relying on a single diagnostic method for species identification, advocating for whole-genome sequencing for accuracy. It emphasizes the continued importance of phenotypic methods in understanding antibiotic resistance in clinical settings and the need for characterization conditions that mirror those encountered by pathogens in the body.
Collapse
Affiliation(s)
- Adam M Varney
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
- Medical Technologies Innovation Facility (MTIF), Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Eden Mannix-Fisher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Jonathan C Thomas
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Samantha McLean
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| |
Collapse
|
8
|
Huening KA, Groves JT, Wildenthal JA, Tabita FR, North JA. Escherichia coli possessing the dihydroxyacetone phosphate shunt utilize 5'-deoxynucleosides for growth. Microbiol Spectr 2024; 12:e0308623. [PMID: 38441472 PMCID: PMC10986504 DOI: 10.1128/spectrum.03086-23] [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: 08/12/2023] [Accepted: 02/17/2024] [Indexed: 03/08/2024] Open
Abstract
All organisms utilize S-adenosyl-l-methionine (SAM) as a key co-substrate for the methylation of biological molecules, the synthesis of polyamines, and radical SAM reactions. When these processes occur, 5'-deoxy-nucleosides are formed as byproducts such as S-adenosyl-l-homocysteine, 5'-methylthioadenosine (MTA), and 5'-deoxyadenosine (5dAdo). A prevalent pathway found in bacteria for the metabolism of MTA and 5dAdo is the dihydroxyacetone phosphate (DHAP) shunt, which converts these compounds into dihydroxyacetone phosphate and 2-methylthioacetaldehyde or acetaldehyde, respectively. Previous work in other organisms has shown that the DHAP shunt can enable methionine synthesis from MTA or serve as an MTA and 5dAdo detoxification pathway. Rather, the DHAP shunt in Escherichia coli ATCC 25922, when introduced into E. coli K-12, enables the use of 5dAdo and MTA as a carbon source for growth. When MTA is the substrate, the sulfur component is not significantly recycled back to methionine but rather accumulates as 2-methylthioethanol, which is slowly oxidized non-enzymatically under aerobic conditions. The DHAP shunt in ATCC 25922 is active under oxic and anoxic conditions. Growth using 5-deoxy-d-ribose was observed during aerobic respiration and anaerobic respiration with Trimethylamine N-oxide (TMAO), but not during fermentation or respiration with nitrate. This suggests the DHAP shunt may only be relevant for extraintestinal pathogenic E. coli lineages with the DHAP shunt that inhabit oxic or TMAO-rich extraintestinal environments. This reveals a heretofore overlooked role of the DHAP shunt in carbon and energy metabolism from ubiquitous SAM utilization byproducts and suggests a similar role may occur in other pathogenic and non-pathogenic bacteria with the DHAP shunt. IMPORTANCE The acquisition and utilization of organic compounds that serve as growth substrates are essential for Escherichia coli to grow and multiply. Ubiquitous enzymatic reactions involving S-adenosyl-l-methionine as a co-substrate by all organisms result in the formation of the 5'-deoxy-nucleoside byproducts, 5'-methylthioadenosine and 5'-deoxyadenosine. All E. coli possess a conserved nucleosidase that cleaves these 5'-deoxy-nucleosides into 5-deoxy-pentose sugars for adenine salvage. The DHAP shunt pathway is found in some extraintestinal pathogenic E. coli, but its function in E. coli possessing it has remained unknown. This study reveals that the DHAP shunt enables the utilization of 5'-deoxy-nucleosides and 5-deoxy-pentose sugars as growth substrates in E. coli strains with the pathway during aerobic respiration and anaerobic respiration with TMAO, but not fermentative growth. This provides an insight into the diversity of sugar compounds accessible by E. coli with the DHAP shunt and suggests that the DHAP shunt is primarily relevant in oxic or TMAO-rich extraintestinal environments.
Collapse
Affiliation(s)
| | - Joshua T. Groves
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - John A. Wildenthal
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - F. Robert Tabita
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Justin A. North
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Bernacchia L, Paris A, Gupta A, Charman RJ, McGreig J, Wass MN, Kad NM. Identification of a novel DNA repair inhibitor using an in silico driven approach shows effective combinatorial activity with genotoxic agents against multidrug-resistant Escherichia coli. Protein Sci 2024; 33:e4948. [PMID: 38501485 PMCID: PMC10949335 DOI: 10.1002/pro.4948] [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: 12/12/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 03/20/2024]
Abstract
Increasing antimicrobial drug resistance represents a global existential threat. Infection is a particular problem in immunocompromised individuals, such as patients undergoing cancer chemotherapy, due to the targeting of rapidly dividing cells by antineoplastic agents. We recently developed a strategy that targets bacterial nucleotide excision DNA repair (NER) to identify compounds that act as antimicrobial sensitizers specific for patients undergoing cancer chemotherapy. Building on this, we performed a virtual drug screening of a ~120,000 compound library against the key NER protein UvrA. From this, numerous target compounds were identified and of those a candidate compound, Bemcentinib (R428), showed a strong affinity toward UvrA. This NER protein possesses four ATPase sites in its dimeric state, and we found that Bemcentinib could inhibit UvrA's ATPase activity by ~90% and also impair its ability to bind DNA. As a result, Bemcentinib strongly diminishes NER's ability to repair DNA in vitro. To provide a measure of in vivo activity we discovered that the growth of Escherichia coli MG1655 was significantly inhibited when Bemcentinib was combined with the DNA damaging agent 4-NQO, which is analogous to UV. Using the clinically relevant DNA-damaging antineoplastic cisplatin in combination with Bemcentinib against the urological sepsis-causing E. coli strain EC958 caused complete growth inhibition. This study offers a novel approach for the potential development of new compounds for use as adjuvants in antineoplastic therapy.
Collapse
Affiliation(s)
| | - Antoine Paris
- School of Biological SciencesUniversity of KentCanterburyUK
| | - Arya Gupta
- School of Biological SciencesUniversity of KentCanterburyUK
| | | | - Jake McGreig
- School of Biological SciencesUniversity of KentCanterburyUK
| | - Mark N. Wass
- School of Biological SciencesUniversity of KentCanterburyUK
| | - Neil M. Kad
- School of Biological SciencesUniversity of KentCanterburyUK
| |
Collapse
|
10
|
Phan MD, Schirra HJ, Nhu NTK, Peters KM, Sarkar S, Allsopp LP, Achard MES, Kappler U, Schembri MA. Combined functional genomic and metabolomic approaches identify new genes required for growth in human urine by multidrug-resistant Escherichia coli ST131. mBio 2024; 15:e0338823. [PMID: 38353545 PMCID: PMC10936160 DOI: 10.1128/mbio.03388-23] [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: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 03/14/2024] Open
Abstract
Urinary tract infections (UTIs) are one of the most common bacterial infections in humans, with ~400 million cases across the globe each year. Uropathogenic Escherichia coli (UPEC) is the major cause of UTI and increasingly associated with antibiotic resistance. This scenario has been worsened by the emergence and spread of pandemic UPEC sequence type 131 (ST131), a multidrug-resistant clone associated with extraordinarily high rates of infection. Here, we employed transposon-directed insertion site sequencing in combination with metabolomic profiling to identify genes and biochemical pathways required for growth and survival of the UPEC ST131 reference strain EC958 in human urine (HU). We identified 24 genes required for growth in HU, which mapped to diverse pathways involving small peptide, amino acid and nucleotide metabolism, the stringent response pathway, and lipopolysaccharide biosynthesis. We also discovered a role for UPEC resistance to fluoride during growth in HU, most likely associated with fluoridation of drinking water. Complementary nuclear magnetic resonance (NMR)-based metabolomics identified changes in a range of HU metabolites following UPEC growth, the most pronounced being L-lactate, which was utilized as a carbon source via the L-lactate dehydrogenase LldD. Using a mouse UTI model with mixed competitive infection experiments, we demonstrated a role for nucleotide metabolism and the stringent response in UPEC colonization of the mouse bladder. Together, our application of two omics technologies combined with different infection-relevant settings has uncovered new factors required for UPEC growth in HU, thus enhancing our understanding of this pivotal step in the UPEC infection pathway. IMPORTANCE Uropathogenic Escherichia coli (UPEC) cause ~80% of all urinary tract infections (UTIs), with increasing rates of antibiotic resistance presenting an urgent threat to effective treatment. To cause infection, UPEC must grow efficiently in human urine (HU), necessitating a need to understand mechanisms that promote its adaptation and survival in this nutrient-limited environment. Here, we used a combination of functional genomic and metabolomic techniques and identified roles for the metabolism of small peptides, amino acids, nucleotides, and L-lactate, as well as the stringent response pathway, lipopolysaccharide biosynthesis, and fluoride resistance, for UPEC growth in HU. We further demonstrated that pathways involving nucleotide metabolism and the stringent response are required for UPEC colonization of the mouse bladder. The UPEC genes and metabolic pathways identified in this study represent targets for the development of innovative therapeutics to prevent UPEC growth during human UTI, an urgent need given the rapidly rising rates of global antibiotic resistance.
Collapse
Affiliation(s)
- Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Horst Joachim Schirra
- School of Environment and Science, Griffith University, Nathan, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Nguyen Thi Khanh Nhu
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Kate M. Peters
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Sohinee Sarkar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Luke P. Allsopp
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Maud E. S. Achard
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Ulrike Kappler
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark A. Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Das Gupta K, Curson JEB, Tarique AA, Kapetanovic R, Schembri MA, Fantino E, Sly PD, Sweet MJ. CFTR is required for zinc-mediated antibacterial defense in human macrophages. Proc Natl Acad Sci U S A 2024; 121:e2315190121. [PMID: 38363865 PMCID: PMC10895263 DOI: 10.1073/pnas.2315190121] [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: 09/19/2023] [Accepted: 12/22/2023] [Indexed: 02/18/2024] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is an anion transporter required for epithelial homeostasis in the lung and other organs, with CFTR mutations leading to the autosomal recessive genetic disease CF. Apart from excessive mucus accumulation and dysregulated inflammation in the airways, people with CF (pwCF) exhibit defective innate immune responses and are susceptible to bacterial respiratory pathogens such as Pseudomonas aeruginosa. Here, we investigated the role of CFTR in macrophage antimicrobial responses, including the zinc toxicity response that is used by these innate immune cells against intracellular bacteria. Using both pharmacological approaches, as well as cells derived from pwCF, we show that CFTR is required for uptake and clearance of pathogenic Escherichia coli by CSF-1-derived primary human macrophages. CFTR was also required for E. coli-induced zinc accumulation and zinc vesicle formation in these cells, and E. coli residing in macrophages exhibited reduced zinc stress in the absence of CFTR function. Accordingly, CFTR was essential for reducing the intramacrophage survival of a zinc-sensitive E. coli mutant compared to wild-type E. coli. Ectopic expression of the zinc transporter SLC30A1 or treatment with exogenous zinc was sufficient to restore antimicrobial responses against E. coli in human macrophages. Zinc supplementation also restored bacterial killing in GM-CSF-derived primary human macrophages responding to P. aeruginosa, used as an in vitro macrophage model relevant to CF. Thus, restoration of the zinc toxicity response could be pursued as a therapeutic strategy to restore innate immune function and effective host defense in pwCF.
Collapse
Affiliation(s)
- Kaustav Das Gupta
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| | - James E. B. Curson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| | - Abdullah A. Tarique
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Ronan Kapetanovic
- Friedrich Miescher Institute for Biomedical Research, Basel, BS4058, Switzerland
- Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly37380, France
| | - Mark A. Schembri
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD4072, Australia
| | - Emmanuelle Fantino
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Peter D. Sly
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| |
Collapse
|
12
|
White RT, Bull MJ, Barker CR, Arnott JM, Wootton M, Jones LS, Howe RA, Morgan M, Ashcroft MM, Forde BM, Connor TR, Beatson SA. Genomic epidemiology reveals geographical clustering of multidrug-resistant Escherichia coli ST131 associated with bacteraemia in Wales. Nat Commun 2024; 15:1371. [PMID: 38355632 PMCID: PMC10866875 DOI: 10.1038/s41467-024-45608-1] [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: 10/13/2021] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Antibiotic resistance is a significant global public health concern. Uropathogenic Escherichia coli sequence type (ST)131, a widely prevalent multidrug-resistant clone, is frequently associated with bacteraemia. This study investigates third-generation cephalosporin resistance in bloodstream infections caused by E. coli ST131. From 2013-2014 blood culture surveillance in Wales, 142 E. coli ST131 genomes were studied alongside global data. All three major ST131 clades were represented across Wales, with clade C/H30 predominant (n = 102/142, 71.8%). Consistent with global findings, Welsh strains of clade C/H30 contain β-lactamase genes from the blaCTX-M-1 group (n = 65/102, 63.7%), which confer resistance to third-generation cephalosporins. Most Welsh clade C/H30 genomes belonged to sub-clade C2/H30Rx (58.3%). A Wales-specific sub-lineage, named GB-WLS.C2, diverged around 1996-2000. An introduction to North Wales around 2002 led to a localised cluster by 2009, depicting limited genomic diversity within North Wales. This investigation emphasises the value of genomic epidemiology, allowing the detection of genetically similar strains in local areas, enabling targeted and timely public health interventions.
Collapse
Affiliation(s)
- Rhys T White
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, 4072, Australia
- Health Group, Institute of Environmental Science and Research, 5022, Porirua, New Zealand
| | - Matthew J Bull
- Microbiomes, Microbes and Informatics Group, Organisms and Environment Division, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, United Kingdom
- Public Health Wales Microbiology, University Hospital of Wales, Cardiff, Wales, CF14 4XW, United Kingdom
| | - Clare R Barker
- Microbiomes, Microbes and Informatics Group, Organisms and Environment Division, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, United Kingdom
| | - Julie M Arnott
- Healthcare Associated Infection, Antimicrobial Resistance & Prescribing Programme (HARP), Public Health Wales, 2 Capital Quarter, Tyndall Street, Cardiff, Wales, CF10 4BZ, United Kingdom
| | - Mandy Wootton
- Public Health Wales Microbiology, University Hospital of Wales, Cardiff, Wales, CF14 4XW, United Kingdom
| | - Lim S Jones
- Public Health Wales Microbiology, University Hospital of Wales, Cardiff, Wales, CF14 4XW, United Kingdom
| | - Robin A Howe
- Public Health Wales Microbiology, University Hospital of Wales, Cardiff, Wales, CF14 4XW, United Kingdom
| | - Mari Morgan
- Healthcare Associated Infection, Antimicrobial Resistance & Prescribing Programme (HARP), Public Health Wales, 2 Capital Quarter, Tyndall Street, Cardiff, Wales, CF10 4BZ, United Kingdom
| | - Melinda M Ashcroft
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Brian M Forde
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, 4072, Australia
- The University of Queensland, UQ Centre for Clinical Research (UQCCR), Royal Brisbane & Women's Hospital Campus, Brisbane, QLD, 4029, Australia
| | - Thomas R Connor
- Microbiomes, Microbes and Informatics Group, Organisms and Environment Division, School of Biosciences, Cardiff University, Cardiff, CF10 3AX, United Kingdom.
- Public Health Genomics Programme, Public Health Wales, 2 Capital Quarter, Tyndall Street, Cardiff, Wales, CF10 4BZ, United Kingdom.
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
13
|
Allegretti YH, Yamaji R, Adams-Sapper S, Riley LW. Genetic features of antimicrobial drug-susceptible extraintestinal pathogenic Escherichia coli pandemic sequence type 95. Microbiol Spectr 2024; 12:e0418922. [PMID: 38059630 PMCID: PMC10783064 DOI: 10.1128/spectrum.04189-22] [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: 10/16/2022] [Accepted: 11/13/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Despite the increasing prevalence of antibiotic-resistant Escherichia coli strains that cause urinary tract and bloodstream infections, a major pandemic lineage of extraintestinal pathogenic E. coli (ExPEC) ST95 has a comparatively low frequency of drug resistance. We compared the genomes of 1,749 ST95 isolates to identify genetic features that may explain why most strains of ST95 resist becoming drug-resistant. Identification of such genomic features could contribute to the development of novel strategies to prevent the spread of antibiotic-resistant genes and devise new measures to control antibiotic-resistant infections.
Collapse
Affiliation(s)
| | | | | | - Lee W. Riley
- University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
14
|
Bermudez TA, Brannon JR, Dudipala N, Reasoner S, Morales G, Wiebe M, Cecala M, DaCosta M, Beebout C, Amir O, Hadjifrangiskou M. Raising the alarm: fosfomycin resistance associated with non-susceptible inner colonies imparts no fitness cost to the primary bacterial uropathogen. Antimicrob Agents Chemother 2024; 68:e0080323. [PMID: 38078906 PMCID: PMC10777853 DOI: 10.1128/aac.00803-23] [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: 06/17/2023] [Accepted: 11/11/2023] [Indexed: 01/11/2024] Open
Abstract
IMPORTANCE While fosfomycin resistance is rare, the observation of non-susceptible subpopulations among clinical Escherichia coli isolates is a common phenomenon during antimicrobial susceptibility testing (AST) in American and European clinical labs. Previous evidence suggests that mutations eliciting this phenotype are of high biological cost to the pathogen during infection, leading to current recommendations of neglecting non-susceptible colonies during AST. Here, we report that the most common route to fosfomycin resistance, as well as novel routes described in this work, does not impair virulence in uropathogenic E. coli, the major cause of urinary tract infections, suggesting a re-evaluation of current susceptibility guidelines is warranted.
Collapse
Affiliation(s)
- Tomas A. Bermudez
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John R. Brannon
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Seth Reasoner
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Grace Morales
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michelle Wiebe
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mia Cecala
- Vanderbilt University, Nashville, Tennessee, USA
| | | | - Connor Beebout
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Omar Amir
- Vanderbilt University, Nashville, Tennessee, USA
| | - Maria Hadjifrangiskou
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Institute for Infection, Immunology & Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Bernacchia L, Gupta A, Paris A, Moores AA, Kad NM. Developing novel antimicrobials by combining cancer chemotherapeutics with bacterial DNA repair inhibitors. PLoS Pathog 2023; 19:e1011875. [PMID: 38060607 PMCID: PMC10729960 DOI: 10.1371/journal.ppat.1011875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/19/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
Cancer chemotherapeutics kill rapidly dividing cells, which includes cells of the immune system. The resulting neutropenia predisposes patients to infection, which delays treatment and is a major cause of morbidity and mortality. To tackle this problem, we have isolated several compounds that inhibit bacterial DNA repair, alone they are non-toxic, however in combination with DNA damaging anti-cancer drugs, they prevent bacterial growth. These compounds were identified through screening of an FDA-approved drug library in the presence of the anti-cancer compound cisplatin. Using a series of triage tests, the screen was reduced to a handful of drugs that were tested for specific activity against bacterial nucleotide excision DNA repair (NER). Five compounds emerged, of which three possess promising antimicrobial properties including cell penetrance, and the ability to block replication in a multi-drug resistant clinically relevant E. coli strain. This study suggests that targeting NER could offer a new therapeutic approach tailor-made for infections in cancer patients, by combining cancer chemotherapy with an adjuvant that targets DNA repair.
Collapse
Affiliation(s)
- Lorenzo Bernacchia
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Arya Gupta
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Antoine Paris
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | | | - Neil M. Kad
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| |
Collapse
|
16
|
Hawas S, Vagenas D, Haque A, Totsika M. Bladder-draining lymph nodes support germinal center B cell responses during urinary tract infection in mice. Infect Immun 2023; 91:e0031723. [PMID: 37882531 PMCID: PMC10652902 DOI: 10.1128/iai.00317-23] [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: 08/12/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
Bacterial urinary tract infections (UTIs) are both common and exhibit high recurrence rates in women. UTI healthcare costs are increasing due to the rise of multidrug-resistant (MDR) bacteria, necessitating alternative approaches for infection control. Here, we directly observed host adaptive immune responses in acute UTI. We employed a mouse model in which wild-type C57BL/6J mice were transurethrally inoculated with a clinically relevant MDR UTI strain of uropathogenic Escherichia coli (UPEC). Firstly, we noted that rag1-/- C57BL/6J mice harbored larger bacterial burdens than wild-type counterparts, consistent with a role for adaptive immunity in UTI control. Consistent with this, UTI triggered in the bladders of wild-type mice early increases of myeloid cells, including CD11chi conventional dendritic cells, suggesting possible involvement of these professional antigen-presenting cells. Importantly, germinal center B cell responses developed by 4 weeks post-infection in bladder-draining lymph nodes of wild-type mice and, although modest in magnitude and transient in nature, could not be boosted with a second UTI. Thus, our data reveal for the first time in a mouse model that UPEC UTI induces local B cell immune responses in bladder-draining lymph nodes, which could potentially serve to control infection.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Dimitrios Vagenas
- Research Methods Group, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ashraful Haque
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Atac N, Onbasli K, Koc I, Yagci Acar H, Can F. Fimbria targeting superparamagnetic iron oxide nanoparticles enhance the antimicrobial and antibiofilm activity of ciprofloxacin against quinolone-resistant E. coli. Microb Biotechnol 2023; 16:2072-2081. [PMID: 37602720 PMCID: PMC10616650 DOI: 10.1111/1751-7915.14327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
High quinolone resistance of Escherichia coli limits the therapy options for urinary tract infection (UTI). In response to the urgent need for efficient treatment of multidrug-resistant infections, we designed a fimbriae targeting superparamagnetic iron oxide nanoparticle (SPION) delivering ciprofloxacin to ciprofloxacin-resistant E. coli. Bovine serum albumin (BSA) conjugated poly(acrylic acid) (PAA) coated SPIONs (BSA@PAA@SPION) were developed for encapsulation of ciprofloxacin and the nanoparticles were tagged with 4-aminophenyl-α-D-mannopyrannoside (mannoside, Man) to target E. coli fimbriae. Ciprofloxacin-loaded mannoside tagged nanoparticles (Cip-Man-BSA@PAA@SPION) provided high antibacterial activity (97.1 and 97.5%, respectively) with a dose of 32 μg/mL ciprofloxacin against two ciprofloxacin-resistant E. coli isolates. Furthermore, a strong biofilm inhibition (86.9% and 98.5%, respectively) was achieved in the isolates at a dose 16 and 8 times lower than the minimum biofilm eradication concentration (MBEC) of ciprofloxacin. Weaker growth inhibition was observed with untargeted nanoparticles, Cip-BSA@PAA@SPIONs, confirming that targeting E. coli fimbria with mannoside-tagged nanoparticles increases the ciprofloxacin efficiency to treat ciprofloxacin-resistant E. coli. Enhanced killing activity against ciprofloxacin-resistant E. coli planktonic cells and strong growth inhibition of their biofilms suggest that Cip-Man-BSA@PAA@SPION system might be an alternative and/or complementary therapeutic option for the treatment of quinolone-resistant E. coli infections.
Collapse
Affiliation(s)
- Nazli Atac
- School of Medicine, Medical MicrobiologyKoç UniversityIstanbulTurkey
- Koç University‐İşbank Center for Infectious Diseases (KUISCID)IstanbulTurkey
| | - Kubra Onbasli
- Department of Metallurgical and Materials Engineeringİstanbul Technical UniversityIstanbulTurkey
| | - Irem Koc
- Graduate School of Materials Science and EngineeringKoç UniversityIstanbulTurkey
| | - Havva Yagci Acar
- Graduate School of Materials Science and EngineeringKoç UniversityIstanbulTurkey
- Department of ChemistryKoç UniversityIstanbulTurkey
| | - Fusun Can
- School of Medicine, Medical MicrobiologyKoç UniversityIstanbulTurkey
- Koç University‐İşbank Center for Infectious Diseases (KUISCID)IstanbulTurkey
| |
Collapse
|
18
|
Hawas S, Qin J, Wiedbrauk S, Fairfull-Smith K, Totsika M. Preclinical Evaluation of Nitroxide-Functionalised Ciprofloxacin as a Novel Antibiofilm Drug Hybrid for Urinary Tract Infections. Antibiotics (Basel) 2023; 12:1479. [PMID: 37887180 PMCID: PMC10604439 DOI: 10.3390/antibiotics12101479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Urinary tract infections (UTIs) are the second most common bacterial infection with high recurrence rates and can involve biofilm formation on patient catheters. Biofilms are inherently tolerant to antimicrobials, making them difficult to eradicate. Many antibiofilm agents alone do not have bactericidal activity; therefore, linking them to antibiotics is a promising antibiofilm strategy. However, many of these hybrid agents have not been tested in relevant preclinical settings, limiting their potential for clinical translation. Here, we evaluate a ciprofloxacin di-nitroxide hybrid (CDN11), previously reported to have antibiofilm activity against uropathogenic Escherichia coli (UPEC) strain UTI89 in vitro, as a potential UTI therapeutic using multiple preclinical models that reflect various aspects of UTI pathogenesis. We report improved in vitro activity over the parent drug ciprofloxacin against mature UTI89 biofilms formed inside polyethylene catheters. In bladder cell monolayers infected with UTI89, treatment with CDN11 afforded significant reduction in bacterial titers, including intracellular UPEC. Infected mouse bladders containing biofilm-like intracellular reservoirs of UPEC UTI89 showed decreased bacterial loads after ex vivo bladder treatment with CDN11. Activity for CDN11 was reported across different models of UTI, showcasing nitroxide-antibiotic hybridization as a promising antibiofilm approach. The pipeline we described here could be readily used in testing other new therapeutic compounds, fast-tracking the development of novel antibiofilm therapeutics.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jilong Qin
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
| | - Sandra Wiedbrauk
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD 4000, Australia; (S.W.); (K.F.-S.)
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Kathryn Fairfull-Smith
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD 4000, Australia; (S.W.); (K.F.-S.)
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
19
|
Huening KA, Groves JT, Wildenthal JA, Tabita FR, North JA. Utilization of 5'-deoxy-nucleosides as Growth Substrates by Extraintestinal Pathogenic E. coli via the Dihydroxyacetone Phosphate Shunt. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552779. [PMID: 37609188 PMCID: PMC10441430 DOI: 10.1101/2023.08.10.552779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
All organisms utilize S-adenosyl-l-methionine (SAM) as a key co-substrate for methylation of biological molecules, synthesis of polyamines, and radical SAM reactions. When these processes occur, 5'-deoxy-nucleosides are formed as byproducts such as S-adenosyl-l-homocysteine (SAH), 5'-methylthioadenosine (MTA), and 5'-deoxyadenosine (5dAdo). One of the most prevalent pathways found in bacteria for the metabolism of MTA and 5dAdo is the DHAP shunt, which converts these compounds into dihydroxyacetone phosphate (DHAP) and 2-methylthioacetaldehyde or acetaldehyde, respectively. Previous work has shown that the DHAP shunt can enable methionine synthesis from MTA or serve as an MTA and 5dAdo detoxification pathway. Here we show that in Extraintestinal Pathogenic E. coil (ExPEC), the DHAP shunt serves none of these roles in any significant capacity, but rather physiologically functions as an assimilation pathway for use of MTA and 5dAdo as growth substrates. This is further supported by the observation that when MTA is the substrate for the ExPEC DHAP shunt, the sulfur components is not significantly recycled back to methionine, but rather accumulates as 2-methylthioethanol, which is slowly oxidized non-enzymatically under aerobic conditions. While the pathway is active both aerobically and anaerobically, it only supports aerobic ExPEC growth, suggesting that it primarily functions in oxygenic extraintestinal environments like blood and urine versus the predominantly anoxic gut. This reveals a heretofore overlooked role of the DHAP shunt in carbon assimilation and energy metabolism from ubiquitous SAM utilization byproducts and suggests a similar role may occur in other pathogenic and non-pathogenic bacteria with the DHAP shunt.
Collapse
Affiliation(s)
| | - Joshua T. Groves
- The Ohio State University Department of Microbiology, Columbus, OH, 43210
| | - John A. Wildenthal
- The Ohio State University Department of Microbiology, Columbus, OH, 43210
| | - F. Robert Tabita
- The Ohio State University Department of Microbiology, Columbus, OH, 43210
| | - Justin A. North
- The Ohio State University Department of Microbiology, Columbus, OH, 43210
| |
Collapse
|
20
|
Forsyth JH, Barron NL, Scott L, Watson BNJ, Chisnall MAW, Meaden S, van Houte S, Raymond B. Decolonizing drug-resistant E. coli with phage and probiotics: breaking the frequency-dependent dominance of residents. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001352. [PMID: 37418300 PMCID: PMC10433417 DOI: 10.1099/mic.0.001352] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
Widespread antibiotic resistance in commensal bacteria creates a persistent challenge for human health. Resident drug-resistant microbes can prevent clinical interventions, colonize wounds post-surgery, pass resistance traits to pathogens or move to more harmful niches following routine interventions such as catheterization. Accelerating the removal of resistant bacteria or actively decolonizing particular lineages from hosts could therefore have a number of long-term benefits. However, removing resident bacteria via competition with probiotics, for example, poses a number of ecological challenges. Resident microbes are likely to have physiological and numerical advantages and competition based on bacteriocins or other secreted antagonists is expected to give advantages to the dominant partner, via positive frequency dependence. Since a narrow range of Escherichia coli genotypes (primarily those belonging to the clonal group ST131) cause a significant proportion of multidrug-resistant infections, this group presents a promising target for decolonization with bacteriophage, as narrow-host-range viral predation could lead to selective removal of particular genotypes. In this study we tested how a combination of an ST131-specific phage and competition from the well-known probiotic E. coli Nissle strain could displace E. coli ST131 under aerobic and anaerobic growth conditions in vitro. We showed that the addition of phage was able to break the frequency-dependent advantage of a numerically dominant ST131 isolate. Moreover, the addition of competing E. coli Nissle could improve the ability of phage to suppress ST131 by two orders of magnitude. Low-cost phage resistance evolved readily in these experiments and was not inhibited by the presence of a probiotic competitor. Nevertheless, combinations of phage and probiotic produced stable long-term suppression of ST131 over multiple transfers and under both aerobic and anaerobic growth conditions. Combinations of phage and probiotic therefore have real potential for accelerating the removal of drug-resistant commensal targets.
Collapse
Affiliation(s)
- Jessica H. Forsyth
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
- Present address: Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Natalie L. Barron
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | - Lucy Scott
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | | | | | - Sean Meaden
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
- Present address: Department of Biology, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Stineke van Houte
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| | - Ben Raymond
- Centre for Ecology and Conservation, University of Exeter, Penryn, TR10 9FE, UK
| |
Collapse
|
21
|
Mo Y, Oonsivilai M, Lim C, Niehus R, Cooper BS. Implications of reducing antibiotic treatment duration for antimicrobial resistance in hospital settings: A modelling study and meta-analysis. PLoS Med 2023; 20:e1004013. [PMID: 37319169 PMCID: PMC10270346 DOI: 10.1371/journal.pmed.1004013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Reducing antibiotic treatment duration is a key component of hospital antibiotic stewardship interventions. However, its effectiveness in reducing antimicrobial resistance is uncertain and a clear theoretical rationale for the approach is lacking. In this study, we sought to gain a mechanistic understanding of the relation between antibiotic treatment duration and the prevalence of colonisation with antibiotic-resistant bacteria in hospitalised patients. METHODS AND FINDINGS We constructed 3 stochastic mechanistic models that considered both between- and within-host dynamics of susceptible and resistant gram-negative bacteria, to identify circumstances under which shortening antibiotic duration would lead to reduced resistance carriage. In addition, we performed a meta-analysis of antibiotic treatment duration trials, which monitored resistant gram-negative bacteria carriage as an outcome. We searched MEDLINE and EMBASE for randomised controlled trials published from 1 January 2000 to 4 October 2022, which allocated participants to varying durations of systemic antibiotic treatments. Quality assessment was performed using the Cochrane risk-of-bias tool for randomised trials. The meta-analysis was performed using logistic regression. Duration of antibiotic treatment and time from administration of antibiotics to surveillance culture were included as independent variables. Both the mathematical modelling and meta-analysis suggested modest reductions in resistance carriage could be achieved by reducing antibiotic treatment duration. The models showed that shortening duration is most effective at reducing resistance carriage in high compared to low transmission settings. For treated individuals, shortening duration is most effective when resistant bacteria grow rapidly under antibiotic selection pressure and decline rapidly when stopping treatment. Importantly, under circumstances whereby administered antibiotics can suppress colonising bacteria, shortening antibiotic treatment may increase the carriage of a particular resistance phenotype. We identified 206 randomised trials, which investigated antibiotic duration. Of these, 5 reported resistant gram-negative bacteria carriage as an outcome and were included in the meta-analysis. The meta-analysis determined that a single additional antibiotic treatment day is associated with a 7% absolute increase in risk of resistance carriage (80% credible interval 3% to 11%). Interpretation of these estimates is limited by the low number of antibiotic duration trials that monitored carriage of resistant gram-negative bacteria, as an outcome, contributing to a large credible interval. CONCLUSIONS In this study, we found both theoretical and empirical evidence that reducing antibiotic treatment duration can reduce resistance carriage, though the mechanistic models also highlighted circumstances under which reducing treatment duration can, perversely, increase resistance. Future antibiotic duration trials should monitor antibiotic-resistant bacteria colonisation as an outcome to better inform antibiotic stewardship policies.
Collapse
Affiliation(s)
- Yin Mo
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Division of Infectious Diseases, University Medicine Cluster, National University Hospital, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Mathupanee Oonsivilai
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Cherry Lim
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rene Niehus
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States of America
| | - Ben S. Cooper
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
22
|
da Silva RA, Wong JJ, Antypas H, Choo PY, Goh K, Jolly S, Liang C, Tay Kwan Sing L, Veleba M, Hu G, Chen J, Kline KA. Mitoxantrone targets both host and bacteria to overcome vancomycin resistance in Enterococcus faecalis. SCIENCE ADVANCES 2023; 9:eadd9280. [PMID: 36812322 PMCID: PMC9946351 DOI: 10.1126/sciadv.add9280] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Antibiotic resistance critically limits treatment options for infection caused by opportunistic pathogens such as enterococci. Here, we investigate the antibiotic and immunological activity of the anticancer agent mitoxantrone (MTX) in vitro and in vivo against vancomycin-resistant Enterococcus faecalis (VRE). We show that, in vitro, MTX is a potent antibiotic against Gram-positive bacteria through induction of reactive oxygen species and DNA damage. MTX also synergizes with vancomycin against VRE, rendering the resistant strains more permeable to MTX. In a murine wound infection model, single-dose MTX treatment effectively reduces VRE numbers, with further reduction when combined with vancomycin. Multiple MTX treatments accelerate wound closure. MTX also promotes macrophage recruitment and proinflammatory cytokine induction at the wound site and augments intracellular bacterial killing in macrophages by up-regulating the expression of lysosomal enzymes. These results show that MTX represents a promising bacterium- and host-targeted therapeutic for overcoming vancomycin resistance.
Collapse
Affiliation(s)
- Ronni A. G. da Silva
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jun Jie Wong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
| | - Haris Antypas
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Pei Yi Choo
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Karlyn Goh
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Shreya Jolly
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Cui Liang
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
| | - Leona Tay Kwan Sing
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
| | - Mark Veleba
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Guangan Hu
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jianzhu Chen
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kimberly A. Kline
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Drug Resistance Interdisciplinary Research Group, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
23
|
Guan XL, Loh JYX, Lizwan M, Chan SCM, Kwan JMC, Lim TP, Koh TH, Hsu LY, Lee BTK. LipidA-IDER to Explore the Global Lipid A Repertoire of Drug-Resistant Gram-Negative Bacteria. Anal Chem 2023; 95:602-611. [PMID: 36599414 PMCID: PMC9850412 DOI: 10.1021/acs.analchem.1c03566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
With the global emergence of drug-resistant bacteria causing difficult-to-treat infections, there is an urgent need for a tool to facilitate studies on key virulence and antimicrobial resistant factors. Mass spectrometry (MS) has contributed substantially to the elucidation of the structure-function relationships of lipid A, the endotoxic component of lipopolysaccharide which also serves as an important protective barrier against antimicrobials. Here, we present LipidA-IDER, an automated structure annotation tool for system-level scale identification of lipid A from high-resolution tandem mass spectrometry (MS2) data. LipidA-IDER was validated against previously reported structures of lipid A in the reference bacteria, Escherichia coli and Pseudomonas aeruginosa. Using MS2 data of variable quality, we demonstrated LipidA-IDER annotated lipid A with a performance of 71.2% specificity and 70.9% sensitivity, offering greater accuracy than existing lipidomics software. The organism-independent workflow was further applied to a panel of six bacterial species: E. coli and Gram-negative members of ESKAPE pathogens. A comprehensive atlas comprising 188 distinct lipid A species, including remodeling intermediates, was generated and can be integrated with software including MS-DIAL and Metabokit for identification and semiquantitation. Systematic comparison of a pair of polymyxin-sensitive and polymyxin-resistant Acinetobacter baumannii isolated from a human patient unraveled multiple key lipid A structural features of polymyxin resistance within a single analysis. Probing the lipid A landscape of bacteria using LipidA-IDER thus holds immense potential for advancing our understanding of the vast diversity and structural complexity of a key lipid virulence and antimicrobial-resistant factor. LipidA-IDER is freely available at https://github.com/Systems-Biology-Of-Lipid-Metabolism-Lab/LipidA-IDER.
Collapse
Affiliation(s)
- Xue Li Guan
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore,. Tel: +65 6592 3957
| | - Johnathan Yi-Xiong Loh
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore
| | - Marco Lizwan
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore
| | - Sharon Cui Mun Chan
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore
| | - Jeric Mun Chung Kwan
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore
| | - Tze Peng Lim
- Department
of Pharmacy, Singapore General Hospital, Singapore 169608, Singapore
| | - Tse Hsien Koh
- Department
of Microbiology, Singapore General Hospital, Singapore 169608, Singapore
| | - Li-Yang Hsu
- Saw Swee
Hock School of Public Health, National University
of Singapore, Singapore 117549, Singapore
| | - Bernett Teck Kwong Lee
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921, Singapore,Centre
for Biomedical Informatics, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore,Singapore
Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| |
Collapse
|
24
|
Huang Y, Chen J, Jiang Q, Huang N, Ding X, Peng L, Deng X. The molybdate-binding protein ModA is required for Proteus mirabilis-induced UTI. Front Microbiol 2023; 14:1156273. [PMID: 37180242 PMCID: PMC10174112 DOI: 10.3389/fmicb.2023.1156273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 05/16/2023] Open
Abstract
Background Proteus mirabilis is one of the pathogens commonly causing urinary tract infections (UTIs). The molybdate-binding protein ModA encoded by modA binds molybdate with high affinity and transports it. Increasing evidence shows that ModA promotes the survival of bacteria in anaerobic environments and participates in bacterial virulence by obtaining molybdenum. However, the role of ModA in the pathogenesis of P. mirabilis remains unknown. Results In this study, a series of phenotypic assays and transcriptomic analyses were used to study the role of ModA in the UTIs induced by P. mirabilis. Our data showed that ModA absorbed molybdate with high affinity and incorporated it into molybdopterin, thus affecting the anaerobic growth of P. mirabilis. Loss of ModA enhanced bacterial swarming and swimming and up-regulated the expression of multiple genes in flagellar assembly pathway. The loss of ModA also resulted in decreased biofilm formation under anaerobic growth conditions. The modA mutant significantly inhibited bacterial adhesion and invasion to urinary tract epithelial cells and down-regulated the expression of multiple genes associated with pilus assembly. Those alterations were not due to anaerobic growth defects. In addition, the decreased bacteria in the bladder tissue, the weakened inflammatory damage, the low level of IL-6, and minor weight change was observed in the UTI mouse model infected with modA mutant. Conclusion Here, we reported that in P. mirabilis, ModA mediated the transport of molybdate, thereby affecting the activity of nitrate reductase and thus affecting the growth of bacteria under anaerobic conditions. Overall, this study clarified the indirect role of ModA in the anaerobic growth, motility, biofilm formation, and pathogenicity of P. mirabilis and its possible pathway, and emphasized the importance of the molybdate-binding protein ModA to P. mirabilis in mediating molybdate uptake, allowing the bacterium to adapt to complex environmental conditions and cause UTIs. Our results provided valuable information on the pathogenesis of ModA-induced P. mirabilis UTIs and may facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Yi Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinbin Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiao Jiang
- Guangdong 999 Brain Hospital, Guangzhou, Guangdong, China
| | - Nan Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Ding
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liang Peng
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Deng, ; Liang Peng,
| | - Xiaoyan Deng
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Deng, ; Liang Peng,
| |
Collapse
|
25
|
Liu L, Law COK, Nie Q, Pham HQ, Ma H, Zhang L, Ho PL, Lau TCK. Comparative analysis of outer membrane vesicles from uropathogenic Escherichia coli reveal the role of aromatic amino acids synthesis proteins in motility. Int J Med Microbiol 2023; 313:151573. [PMID: 36634604 DOI: 10.1016/j.ijmm.2023.151573] [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: 09/16/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) are causative agent that causes urinary tract infections (UTIs) and the recent emergence of multidrug resistance (MDR) of UPEC increases the burden on the community. Recent studies of bacterial outer membrane vesicles (OMV) identified various factors including proteins, nucleic acids, and small molecules which provided inter-cellular communication within the bacterial population. However, the components of UPEC-specific OMVs and their functional role remain unclear. Here, we systematically determined the proteomes of UPEC-OMVs and identified the specific components that provide functions to the recipient bacteria. Based on the functional network of OMVs' proteomes, a group of signaling peptides was found in all OMVs which provide communication among bacteria. Moreover, we demonstrated that treatment with UPEC-OMVs affected the motility and biofilm formation of the recipient bacteria, and further identified aromatic amino acid (AAA) biosynthesis proteins as the key factors to provide their movement.
Collapse
Affiliation(s)
- LiangZhe Liu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China; Center for Clinical Precision Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Carmen Oi Kwan Law
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China
| | - Qichang Nie
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China
| | - Hoa Quynh Pham
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China
| | - Haiying Ma
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region
| | - Liang Zhang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region
| | - Pak Leung Ho
- Department of Microbiology, Queen Mary Hospital, University of Hong Kong, Hong Kong Special Administrative Region
| | - Terrence Chi-Kong Lau
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China.
| |
Collapse
|
26
|
Downing T, Lee MJ, Archbold C, McDonnell A, Rahm A. Informing plasmid compatibility with bacterial hosts using protein-protein interaction data. Genomics 2022; 114:110509. [PMID: 36273742 DOI: 10.1016/j.ygeno.2022.110509] [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: 08/19/2022] [Revised: 09/23/2022] [Accepted: 10/19/2022] [Indexed: 01/15/2023]
Abstract
The compatibility of plasmids with new host cells is significant given their role in spreading antimicrobial resistance (AMR) and virulence factor genes. Evaluating this using in vitro screening is laborious and can be informed by computational analyses of plasmid-host compatibility through rates of protein-protein interactions (PPIs) between plasmid and host cell proteins. We identified large excesses of such PPIs in eight important plasmids, including pOXA-48, using most known bacteria (n = 4363). 23 species had high rates of interactions with four blaOXA-48-positive plasmids. We also identified 48 species with high interaction rates with plasmids common in Escherichia coli. We found a strong association between one plasmid and the fimbrial adhesin operon pil, which could enhance host cell adhesion in aqueous environments. An excess rate of PPIs could be a sign of host-plasmid compatibility, which is important for AMR control given that plasmids like pOXA-48 move between species with ease.
Collapse
Affiliation(s)
- Tim Downing
- School of Biotechnology, Dublin City University, Dublin, Ireland; The Pirbright Institute, UK.
| | - Min Jie Lee
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Conor Archbold
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Adam McDonnell
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Alexander Rahm
- GAATI Lab, University of French Polynesia, Tahiti, French Polynesia
| |
Collapse
|
27
|
Kremer A, Whitmer G, Diaz A, Sajwani A, Navarro A, Arshad M. ESBL Escherichia coli Isolates Have Enhanced Gut Colonization Capacity Compared to Non-ESBL Strains in Neonatal Mice. Microbiol Spectr 2022; 10:e0058222. [PMID: 36121240 PMCID: PMC9603109 DOI: 10.1128/spectrum.00582-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 08/23/2022] [Indexed: 12/30/2022] Open
Abstract
Extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli can cause invasive infections in infants and immunocompromised children with high associated morbidity and mortality. The gut is a major reservoir of these strains in the community. Current dogma dictates that antimicrobial resistance is associated with a fitness cost. However, recent data show that some contemporary ESBL E. coli strains may be more "fit" compared to nonresistant E. coli strains. Here, we use whole-genome sequencing to first characterize 15 ESBL E. coli strains isolated from infants in a Pakistani community, a clinical extraintestinal pathogenic ESBL E. coli ST131 strain, and a non-ESBL commensal E. coli strain, and then use a novel animal model of early life gut colonization to assess the ability of these strains to colonize the infant mouse gut. We determined that CTX-M-15 was present in all the ESBL strains, as well as additional beta-lactamases and genes conferring resistance to multiple antibiotic classes. In the animal model, 11/16 ESBL E. coli strains had significantly higher burden of colonization at week four of life compared to commensal strains, even in the absence of selective antibiotic pressure, suggesting that these strains may have enhanced fitness despite being highly antimicrobial resistant. IMPORTANCE Antimicrobial resistance is a global public health emergency. Infants, especially preterm infants and those in the neonatal intensive care unit, immunocompromised hosts, and those with chronic illnesses are at highest risk of adverse outcomes from invasive infections with antimicrobial-resistant strains. It has long been thought that resistance is associated with a fitness cost, i.e., antimicrobial-resistant strains are not able to colonize the gut as well as nonresistant strains, and that antibiotic exposure is a key risk factor for persistent colonization with resistant strains. Here, we use a novel infant mouse model to add to the growing body of literature that some highly-resistant contemporary Escherichia coli strains can persist in the gut with a significant burden of colonization despite absence of antibiotic exposure.
Collapse
Affiliation(s)
- Aspen Kremer
- Ann and Robert H. Lurie Children’s Hospital, Chicago, Illinois, USA
| | - Grant Whitmer
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alondra Diaz
- Ann and Robert H. Lurie Children’s Hospital, Chicago, Illinois, USA
| | - Alima Sajwani
- Ann and Robert H. Lurie Children’s Hospital, Chicago, Illinois, USA
| | - Alexis Navarro
- University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mehreen Arshad
- Ann and Robert H. Lurie Children’s Hospital, Chicago, Illinois, USA
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
28
|
Occurrence and dissemination of antibiotic resistance genes in mine soil ecosystems. Appl Microbiol Biotechnol 2022; 106:6289-6299. [PMID: 36002692 DOI: 10.1007/s00253-022-12129-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 11/02/2022]
Abstract
Metal(loid) selection contributes to selection pressure on antibiotic resistance, but to our knowledge, evidence of the dissemination of antibiotic resistance genes (ARGs) induced by metal(loid)s in mine soil ecosystems is rare. In the current study, using a high-throughput sequencing (HTS)-based metagenomic approach, 819 ARG subtypes were identified in a mine soil ecosystem, indicating that these environmental habitats are important reservoirs of ARGs. The results showed that metal(loid)-induced coselection has an important role in the distribution of soil ARGs. Furthermore, metal(loid) selection-induced ARGs were mainly associated with resistance-nodulation-division (RND) antibiotic efflux, which is distinct from what is observed in agricultural soil ecosystems. By using independent genome binning, metal(loid)s were shown impose coselection pressure on multiple ARGs residing on mobile genetic elements (MGEs), which promotes the dissemination of the antibiotic resistome. Interestingly, the current results showed that the density of several MGEs conferring ARGs was considerably higher in organisms most closely related to the priority pathogens Pseudomonas aeruginosa and Escherichia coli. Together, the results of this study indicate that mine soil ecosystems are important reservoirs of ARGs and that metal(loid)-induced coselection plays critical roles in the dissemination of ARGs in this type of soil habitat. KEY POINTS: • Mining soil ecosystem is a reservoir of antibiotic resistance genes (ARGs). • ARGs distribute via bacterial resistance-nodulation-division efflux systems. • Metal(loid)s coselected ARGs residing on mobile genetic elements in P. aeruginosa and E. coli.
Collapse
|
29
|
Khoshbayan A, Golmoradi Zadeh R, Taati Moghadam M, Mirkalantari S, Darbandi A. Molecular determination of O25b/ST131 clone type among extended spectrum β-lactamases production Escherichia coli recovering from urinary tract infection isolates. Ann Clin Microbiol Antimicrob 2022; 21:35. [PMID: 35927655 PMCID: PMC9351160 DOI: 10.1186/s12941-022-00526-2] [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: 02/20/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Escherichia coli (E. coli) O25b/ST131 clone causes urinary tract infection (UTI) and is associated with a broad spectrum of other infections, such as intra-abdominal and soft tissue infections, that can be affecting bloodstream infections. Therefore, since O25b/ST131 has been reported in several studies from Iran, in the current study, we have investigated the molecular characteristics, typing, and biofilm formation of O25b/ST131 clone type E. coli collected from UTI specimens. Methods A total of 173 E. coli isolates from UTI were collected. The susceptibility to all fourth generations of cephalosporins (cefazolin, cefuroxime, ceftriaxone, cefotaxime, ceftazidime, cefepime) and ampicillin, ampicillin-sulbactam and aztreonam was determined. Class A ESBLs, class D ESBL and the presence of pabB gene screenings to detect of O25b/ST131 clone type were performed by using of PCR. Biofilm formation was compared between O25b/ST131 isolates and non-O25b/ST131 isolates. Finally, ERIC-PCR was used for typing of ESBL positive isolates. Results Ninety-four ESBL positive were detected of which 79 of them were O25b/ST131. Antimicrobial susceptibility test data showed that most antibiotics had a higher rate of resistance in isolates of the O25b/ST131 clonal type. Biofilm formation showed that there was a weak association between O25b/ST131 clone type isolates and the level of the biofilm formation. ERIC-PCR results showed that E. coli isolates were genetically diverse and classified into 14 groups. Conclusion Our results demonstrated the importance and high prevalence of E. coli O25b/ST131 among UTI isolates with the ability to spread fast and disseminate antibiotic resistance genes.
Collapse
Affiliation(s)
- Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Golmoradi Zadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Taati Moghadam
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Shiva Mirkalantari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| | - Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Venturini C, Bowring B, Partridge SR, Ben Zakour NL, Fajardo-Lubian A, Lopez Ayala A, Qin J, Totsika M, van Galen G, Norris J, Iredell J. Co-Occurrence of Multidrug Resistant Klebsiella pneumoniae Pathogenic Clones of Human Relevance in an Equine Pneumonia Case. Microbiol Spectr 2022; 10:e0215821. [PMID: 35579468 PMCID: PMC9241755 DOI: 10.1128/spectrum.02158-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/13/2022] [Indexed: 11/20/2022] Open
Abstract
The global epidemiology of multidrug resistant Klebsiella pneumoniae, a serious threat to both animal and human health, is dominated by the spread of pathogenic clones, each separately evolving via acquisition of transferable antibiotic resistance or niche-specific virulence determinants. In horses, K. pneumoniae infection can lead to severe respiratory illness. Here, we characterized multiple isolates recovered from bronchial aspirates of a mare with pneumonia refractory to antibiotics. First, we used a combination of standard microbiology, bacteriophage cross-susceptibility and antibiotic resistance testing to profile the infecting K. pneumoniae population. The genomes of isolates with distinct fingerprints (pulsed-field gel electrophoresis) and unique combined bacteriophage/antibiotic profiles were then further analyzed using whole-genome sequencing. Adhesion to human epithelial cells and biofilm production were also measured as virulence indicators. Although it is commonly expected for one clone to dominate an infection episode, we identified five coexisting multidrug resistant K. pneumoniae sharing the same niche. One was a novel sequence type (ST4656), while the other four were all members of emerging human pathogenic clonal groups (ST307, ST628, ST893 and ST392). These isolates did not display significant differences from one another in terms of virulence or resistance and differed only in plasmid content from isolates implicated in severe human infections, with equal potential to prolong duration and severity of infection when sharing the same niche. This study highlights the importance of more precise surveillance and detection measures to uncover bacterial heterogeneity, reminding us that the "single clone" concept is not an absolute in invasive bacterial infections. IMPORTANCE Multidrug resistant Klebsiella pneumoniae are agents of life-threatening infections in animals and humans, with several multidrug resistant clones causing outbreaks of disease worldwide. It is generally accepted that only one clone will be dominant in an infection episode. In this study, we investigated K. pneumoniae isolates from a horse with severe pneumonia and demonstrated co-occurrence of multiple sequence types previously identified as emerging human pathogens. The equine isolates are not significantly different from one another in terms of virulence or resistance, with equal potential to prolong duration and severity of infection, and are indistinguishable from isolates recovered from humans, except for plasmid content. Our study highlights how the "one dominant clone" concept is not an absolute in severe infection, illustrating the need for improved diagnostics to track heterogeneity of infection, and reinforces the importance of cross-monitoring of environmental and human reservoirs of multidrug resistant pathogens.
Collapse
Affiliation(s)
- Carola Venturini
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| | - Bethany Bowring
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Sally R. Partridge
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Western Sydney Local Health District (WSLHD), Westmead, New South Wales, Australia
| | - Nouri L. Ben Zakour
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Alicia Fajardo-Lubian
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Ariana Lopez Ayala
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Jilong Qin
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Gaby van Galen
- Sydney School of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| | - Jacqueline Norris
- Sydney School of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| | - Jonathan Iredell
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Western Sydney Local Health District (WSLHD), Westmead, New South Wales, Australia
| |
Collapse
|
31
|
Tantoso E, Eisenhaber B, Kirsch M, Shitov V, Zhao Z, Eisenhaber F. To kill or to be killed: pangenome analysis of Escherichia coli strains reveals a tailocin specific for pandemic ST131. BMC Biol 2022; 20:146. [PMID: 35710371 PMCID: PMC9205054 DOI: 10.1186/s12915-022-01347-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Escherichia coli (E. coli) has been one of the most studied model organisms in the history of life sciences. Initially thought just to be commensal bacteria, E. coli has shown wide phenotypic diversity including pathogenic isolates with great relevance to public health. Though pangenome analysis has been attempted several times, there is no systematic functional characterization of the E. coli subgroups according to the gene profile. RESULTS Systematically scanning for optimal parametrization, we have built the E. coli pangenome from 1324 complete genomes. The pangenome size is estimated to be ~25,000 gene families (GFs). Whereas the core genome diminishes as more genomes are added, the softcore genome (≥95% of strains) is stable with ~3000 GFs regardless of the total number of genomes. Apparently, the softcore genome (with a 92% or 95% generation threshold) can define the genome of a bacterial species listing the critically relevant, evolutionarily most conserved or important classes of GFs. Unsupervised clustering of common E. coli sequence types using the presence/absence GF matrix reveals distinct characteristics of E. coli phylogroups B1, B2, and E. We highlight the bi-lineage nature of B1, the variation of the secretion and of the iron acquisition systems in ST11 (E), and the incorporation of a highly conserved prophage into the genome of ST131 (B2). The tail structure of the prophage is evolutionarily related to R2-pyocin (a tailocin) from Pseudomonas aeruginosa PAO1. We hypothesize that this molecular machinery is highly likely to play an important role in protecting its own colonies; thus, contributing towards the rapid rise of pandemic E. coli ST131. CONCLUSIONS This study has explored the optimized pangenome development in E. coli. We provide complete GF lists and the pangenome matrix as supplementary data for further studies. We identified biological characteristics of different E. coli subtypes, specifically for phylogroups B1, B2, and E. We found an operon-like genome region coding for a tailocin specific for ST131 strains. The latter is a potential killer weapon providing pandemic E. coli ST131 with an advantage in inter-bacterial competition and, suggestively, explains their dominance as human pathogen among E. coli strains.
Collapse
Affiliation(s)
- Erwin Tantoso
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Singapore, 138672, Republic of Singapore.,Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore
| | - Birgit Eisenhaber
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Singapore, 138672, Republic of Singapore.,Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore
| | - Miles Kirsch
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore.,Present address: Northeastern University, Boston, USA
| | - Vladimir Shitov
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore
| | - Zhiya Zhao
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore.,Present address: The University of Cambridge, Cambridge, UK
| | - Frank Eisenhaber
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Singapore, 138672, Republic of Singapore. .,Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street #07-01, Matrix Building, Singapore, 138671, Republic of Singapore. .,School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, 637551, Singapore, Republic of Singapore.
| |
Collapse
|
32
|
Plasmid-mediated quinolone resistance determinants in fluoroquinolone-nonsusceptible Escherichia coli isolated from patients with urinary tract infections in a university hospital, 2009-2010 and 2020: PMQR in UTI E. coli. J Glob Antimicrob Resist 2022; 30:241-248. [PMID: 35691573 DOI: 10.1016/j.jgar.2022.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES This study aimed to characterize the plasmid-mediated quinolone resistance (PMQR) in fluoroquinolone non-susceptible E. coli (FQNSEC) isolated from patients with urinary tract infections (UTIs) in 2019-2010 and 2020. METHODS A total of 844 E. coli isolates were collected from UTI patients at National Cheng Kung University Hospital (NCKUH). The antimicrobial susceptibility of E. coli isolates to 21 antibiotics was determined by disk diffusion tests. The distribution of phylogenetic groups, virulence factor, and PMQR genes, was determined by PCR. Conjugation assays were performed to investigate the transferrability of qnr genes from FQNSEC isolates to E. coli C600. RESULTS We found 211 (41.9%) and 152 (44.7%) E. coli isolates were FQNSEC in 2009-2010 and 2020, respectively. Phylogenetic group B2 was dominant in FQNSEC isolates (52.34%), followed by group F (10.47%), group B1 (9.64%), and group D (9.64%). FQNSEC isolates were more resistant to 17 of 19 tested antimicrobial agents, compared to the FQ susceptible E. coli. PMQR screening results showed that 34, 22, and 10 FQNSEC isolates containing aac(6')-Ib-cr, qnr genes, and efflux pump genes (qepA or oqxAB), respectively. PMQR E. coli isolates were more non-susceptible to gentamicin, amoxicillin, ampicillin/sulbactam, imipenem, cefazolin, cefuroxime, cefmetazole, ceftriaxone, ceftazidime, and cefepime, compared to non-PMQR FQNSEC. Moreover, 16 of 22 qnr-carrying plasmids were transferrable to the recipient C600. CONCLUSION Here, we reported the high prevalence of MDR- and XDR-E. coli in FQNSEC isolates. Moreover, qnr-carrying plasmids were highly transferable and lead to the resistance to other classes of antibiotics in the transconjugants.
Collapse
|
33
|
Loss of β-Ketoacyl Acyl Carrier Protein Synthase III Activity Restores Multidrug-Resistant Escherichia coli Sensitivity to Previously Ineffective Antibiotics. mSphere 2022; 7:e0011722. [PMID: 35574679 PMCID: PMC9241538 DOI: 10.1128/msphere.00117-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gram-negative pathogens are a major concern for global public health due to increasing rates of antibiotic resistance and the lack of new drugs. A major contributing factor toward antibiotic resistance in Gram-negative bacteria is their formidable outer membrane, which acts as a permeability barrier preventing many biologically active antimicrobials from reaching the intracellular targets and thus limiting their efficacy.
Collapse
|
34
|
Zhou Y, Fu H, Yang H, Wu J, Chen Z, Jiang H, Liu M, Liu Q, Huang L, Gao J, Chen C. Extensive metagenomic analysis of the porcine gut resistome to identify indicators reflecting antimicrobial resistance. MICROBIOME 2022; 10:39. [PMID: 35246246 PMCID: PMC8895625 DOI: 10.1186/s40168-022-01241-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 02/01/2022] [Indexed: 05/04/2023]
Abstract
BACKGROUND Antimicrobial resistance (AMR) has been regarded as a major threat to global health. Pigs are considered an important source of antimicrobial resistance genes (ARGs). However, there is still a lack of large-scale quantitative data on the distribution of ARGs in the pig production industry. The bacterial species integrated ARGs in the gut microbiome have not been clarified. RESULTS In the present study, we used deep metagenomic sequencing data of 451 samples from 425 pigs including wild boars, Tibetan pigs, and commercial or cross-bred experimental pigs under different rearing modes, to comprehensively survey the diversity and distribution of ARGs and detect the bacteria integrated in these ARGs. We identified a total of 1295 open reading frames (ORFs) recognized as antimicrobial resistance protein-coding genes. The ORFs were clustered into 349 unique types of ARGs, and these could be further classified into 69 drug resistance classes. Tetracycline resistance was most enriched in pig feces. Pigs raised on commercial farms had a significantly higher AMR level than pigs under semi-free ranging conditions or wild boars. We tracked the changes in the composition of ARGs at different growth stages and gut locations. There were 30 drug resistance classes showing significantly different abundances in pigs between 25 and 240 days of age. The richness of ARGs and 41 drug resistance classes were significantly different between cecum lumen and feces in pigs from commercial farms, but not in wild boars. We identified 24 bacterial species that existed in almost all tested samples (core bacteria) and were integrated 128 ARGs in their genomes. However, only nine ARGs of these 128 ARGs were core ARGs, suggesting that most of the ARGs in these bacterial species might be acquired rather than constitutive. We selected three subsets of ARGs as indicators for evaluating the pollution level of ARGs in samples with high accuracy (r = 0.73~0.89). CONCLUSIONS This study provides a primary overview of ARG profiles in various farms under different rearing modes, and the data serve as a reference for optimizing the use of antimicrobials and evaluating the risk of pollution by ARGs in pig farms. Video abstract.
Collapse
Affiliation(s)
- Yunyan Zhou
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Hao Fu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Hui Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Jinyuan Wu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Zhe Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Hui Jiang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Min Liu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Qin Liu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Lusheng Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Jun Gao
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| | - Congying Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045 China
| |
Collapse
|
35
|
Extraintestinal Pathogenic Escherichia coli: Beta-Lactam Antibiotic and Heavy Metal Resistance. Antibiotics (Basel) 2022; 11:antibiotics11030328. [PMID: 35326791 PMCID: PMC8944441 DOI: 10.3390/antibiotics11030328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/10/2022] Open
Abstract
Multiple-antibiotic-resistant (MAR) extra-intestinal pathogenic Escherichia coli (ExPEC) represents one of the most frequent causes of human nosocomial and community-acquired infections, whose eradication is of major concern for clinicians. ExPECs may inhabit indefinitely as commensal the gut of humans and other animals; from the intestine, they may move to colonize other tissues, where they are responsible for a number of diseases, including recurrent and uncomplicated UTIs, sepsis and neonatal meningitis. In the pre-antibiotic era, heavy metals were largely used as chemotherapeutics and/or as antimicrobials in human and animal healthcare. As with antibiotics, the global incidence of heavy metal tolerance in commensal, as well as in ExPEC, has increased following the ban in several countries of antibiotics as promoters of animal growth. Furthermore, it is believed that extensive bacterial exposure to heavy metals present in soil and water might have favored the increase in heavy-metal-tolerant microorganisms. The isolation of ExPEC strains with combined resistance to both antibiotics and heavy metals has become quite common and, remarkably, it has been recently shown that heavy metal resistance genes may co-select antibiotic-resistance genes. Despite their clinical relevance, the mechanisms underlining the development and spread of heavy metal tolerance have not been fully elucidated. The aim of this review is to present data regarding the development and spread of resistance to first-line antibiotics, such as beta-lactams, as well as tolerance to heavy metals in ExPEC strains.
Collapse
|
36
|
Kenneally C, Murphy CP, Sleator RD, Culligan EP. The Urinary Microbiome and Biological Therapeutics: Novel Therapies For Urinary Tract Infections. Microbiol Res 2022; 259:127010. [DOI: 10.1016/j.micres.2022.127010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
|
37
|
Abstract
Many antibiotic resistant uropathogenic Escherichia coli (UPEC) strains belong to clones defined by their multilocus sequence type (ST), with ST131 being the most dominant. Although we have a good understanding of resistance development to fluoroquinolones and third-generation cephalosporins by ST131, our understanding of the virulence repertoire that has contributed to its global dissemination is limited. Here we show that the genes encoding Afa/Dr fimbriae, a group of adhesins strongly associated with UPEC that cause gestational pyelonephritis and recurrent cystitis, are found in approximately one third of all ST131 strains. Sequence comparison of the AfaE adhesin protein revealed a unique allelic variant carried by 82.9% of afa-positive ST131 strains. We identify the afa regulatory region as a hotspot for the integration of insertion sequence (IS) elements, all but one of which alter afa transcription. Close investigation demonstrated that the integration of an IS1 element in the afa regulatory region leads to increased expression of Afa/Dr fimbriae, promoting enhanced adhesion to kidney epithelial cells and suggesting a mechanism for altered virulence. Finally, we provide evidence for a more widespread impact of IS1 on ST131 genome evolution, suggesting that IS dynamics contribute to strain level microevolution that impacts ST131 fitness. IMPORTANCE E. coli ST131 is the most common antibiotic resistant UPEC clone associated with human urinary tract and bloodstream infections. Understanding the features of ST131 that have driven its global dissemination remains a critical priority if we are to counter its increasing antibiotic resistance. Here, we utilized a large collection of ST131 isolates to investigate the prevalence, regulation, and function of Afa/Dr fimbriae, a well-characterized UPEC colonization and virulence factor. We show that the afa genes are found frequently in ST131 and demonstrate how the integration of IS elements in the afa regulatory region modulates Afa expression, presenting an example of altered virulence capacity. We also exploit a curated set of ST131 genomes to map the integration of the antibiotic resistance-associated IS1 element in the ST131 pangenome, providing evidence for its widespread impact on ST131 genome evolution.
Collapse
|
38
|
Proton motive force underpins respiration-mediated potentiation of aminoglycoside lethality in pathogenic Escherichia coli. Arch Microbiol 2022; 204:120. [PMID: 34989857 PMCID: PMC8739286 DOI: 10.1007/s00203-021-02710-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 11/24/2022]
Abstract
It is well known that loss of aerobic respiration in Gram-negative bacteria can diminish the efficacy of a variety of bactericidal antibiotics, which has lead to subsequent demonstrations that the formation of reactive oxygen species (ROS) and the proton motive force (PMF) can both play a role in antibiotic toxicity. The susceptibility of Gram-negative bacteria to aminoglycoside antibiotics, particularly gentamicin, has previously been linked to both the production of ROS and the rate of antibiotic uptake that is mediated by the PMF, although the relative contributions of ROS and PMF to aminoglycoside toxicity has remained poorly understood. Herein, gentamicin was shown to elicit a very modest increase in ROS levels in an aerobically grown Escherichia coli clinical isolate. The well-characterised uncoupler 2,4-dinitrophenol (DNP) was used to disrupt the PMF, which resulted in a significant decrease in gentamicin lethality towards E. coli. DNP did not significantly alter respiratory oxygen consumption, supporting the hypothesis that this uncoupler does not increase ROS production via elevated respiratory oxidase activity. These observations support the hypothesis that maintenance of PMF rather than induction of ROS production underpins the mechanism for how the respiratory chain potentiates the toxicity of aminoglycosides. This was further supported by the demonstration that the uncoupler DNP elicits a dramatic decrease in gentamicin lethality under anaerobic conditions. Together, these data strongly suggest that maintenance of the PMF is the dominant mechanism for the respiratory chain in potentiating the toxic effects of aminoglycosides.
Collapse
|
39
|
Qin J, Wilson KA, Sarkar S, Heras B, O'Mara ML, Totsika M. Conserved FimH mutations in the global Escherichia coli ST131 multi-drug resistant lineage weaken interdomain interactions and alter adhesin function. Comput Struct Biotechnol J 2022; 20:4532-4541. [PMID: 36090810 PMCID: PMC9428848 DOI: 10.1016/j.csbj.2022.08.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/27/2022] Open
Abstract
The binding of the type 1 fimbrial adhesin FimH to mannosylated receptors is allosterically regulated to enhance the fitness of uropathogenic Escherichia coli (UPEC) during urinary tract infection (UTI). Mutations in the two FimH domains (pilin and lectin) located outside the mannose binding pocket have been shown to influence mannose binding affinity, yet the details of the allostery mechanism are not fully elucidated. Here we characterised different FimH conformational states (termed low-affinity tense and high-affinity relaxed conformations) of natural FimH variants using molecular dynamics (MD) simulation techniques and report key structural dynamics differences between them. The clinically dominant FimH30 variant from the pandemic multidrug resistant E. coli ST131 lineage contains an R166H mutation that weakens FimH interdomain interactions and allows enhanced mannose interactions with pre-existing high-affinity relaxed conformations. When expressed in an isogenic ST131 strain background, FimH30 mediated high human cell adhesion and invasion, and enhanced biofilm formation over other variants. Collectively, our computational and experimental findings support a model of FimH protein allostery that is mediated by shifts in the pre-existing conformational equilibrium of FimH, additional to the sequential step-wise process of structural perturbations transmitted from one site to another within the protein. Importantly, it is the first study to shed light into how natural mutations in a clinically dominant FimH variant influence the protein’s conformational landscape optimising its function for ST131 fitness at intestinal and extraintestinal niches.
Collapse
|
40
|
Hapuarachchi IU, Hannaway RF, Roman T, Biswas A, Dyet K, Morgan X, Ussher JE. Genetic evaluation of ESBL-producing Escherichia coli urinary isolates in Otago, New Zealand. JAC Antimicrob Resist 2021; 3:dlab147. [PMID: 34778763 PMCID: PMC8578626 DOI: 10.1093/jacamr/dlab147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/01/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVES The incidence of infections with ESBL-producing Escherichia coli (ESBL-Ec) in New Zealand is increasing. ESBL-Ec most commonly cause urinary tract infections and are seen in both community and hospitalized patients. The reason for the increasing incidence of ESBL-Ec infections is unknown. METHODS In this study, 65 urinary ESBL-Ec isolates from the Otago region in 2015 were fully genetically characterized to understand the mechanisms of transmission. The ESBL gene, E. coli STs, plasmid types and genetic context (e.g. insertion sequences) of ESBL genes were determined by a combination of whole genome and plasmid sequencing. The phylogenetic relationships of the isolates were compared with ESBL-Ec isolates sequenced as part of the 2016 nationwide survey. RESULTS Significant diversity of E. coli strains, plasmids, and the genetic context of ESBL genes was seen. However, there was evidence of common mobile genetic elements in unrelated ESBL-Ec. CONCLUSIONS Multiple introductions of ESBL resistance genes or resistant bacterial strains with limited horizontal transmission of mobile genetic elements accounts for the increased incidence of ESBL-Ec in this low prevalence area. Future studies should investigate modes of transmission of ESBL-Ec in the Otago region.
Collapse
Affiliation(s)
- Isuri U Hapuarachchi
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Rachel F Hannaway
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tabatha Roman
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Ambarish Biswas
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- AgResearch, Palmerston North, New Zealand
| | | | - Xochitl Morgan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - James E Ussher
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Southern Community Laboratories, Dunedin, New Zealand
| |
Collapse
|
41
|
Plasmid-mediated ciprofloxacin resistance imparts a selective advantage on Escherichia coli ST131. Antimicrob Agents Chemother 2021; 66:e0214621. [PMID: 34780264 DOI: 10.1128/aac.02146-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli ST131 is a recently emerged antibiotic resistant clone responsible for high rates of urinary tract and bloodstream infections. Despite its global dominance, the precise mechanisms that have driven the rapid dissemination of ST131 remain unknown. Here, we show that the plasmid-associated resistance gene encoding the AAC(6')-Ib-cr enzyme that inactivates the fluoroquinolone antibiotic ciprofloxacin is present in >70% of strains from the most rapidly expanding subgroup of multidrug resistant ST131. Using a series of genome-edited and plasmid-cured isogenic strains, we demonstrate that the aac(6')-Ib-cr gene confers a selective advantage on ST131 in the presence of ciprofloxacin, even in strains containing chromosomal GyrA and ParC FQ-resistance mutations. Further, we identify a pattern of emerging carbapenem resistance in other common E. coli clones carrying both aac(6')-Ib-cr and chromosomal FQ-resistance mutations, suggesting this dual resistance combination may also impart a selective advantage on these non-ST131 antibiotic resistant lineages.
Collapse
|
42
|
Milano A, Sulejmani A, Intra J, Sala MR, Leoni V, Carcione D. Antimicrobial Resistance Trends of Escherichia coli Isolates from Outpatient and Inpatient Urinary Infections over a 20-Year Period. Microb Drug Resist 2021; 28:63-72. [PMID: 34520265 DOI: 10.1089/mdr.2021.0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial resistance is a worldwide problem, and resistance in Enterobacteriaceae, particularly Escherichia coli and Klebsiella pneumoniae, is a critical threat to human health. Inappropriate and unnecessary use of antibiotics in human health care is the most common cause for the development and spread of antimicrobial resistance. In this work, we retrospectively analyzed the antimicrobial data of E. coli strains isolated from midstream urinary samples over a 20-year period (2000-2019). The aim was to provide useful information to clinicians to prescribe a more appropriate empirical antibiotic therapy. A total of 30,955 unique E. coli isolates from positive midstream urine samples of inpatients (1,198) and outpatients (29,757) were collected. Except for carbapenems, over time all the antibiotics tested showed increasing resistance rates in both groups (p < 0.0001). On the other hand, fosfomycin and nitrofurantoin presented significant decreasing trends in resistance rate (p < 0.05). There were significant increases in extended-spectrum β-lactamases- and multidrug resistance positive isolates starting in 2000 (p < 0.0001), with similar results in both groups. Ciprofloxacin, gentamicin, trimethoprim/sulfamethoxazole, and third-generation cephalosporin resistances significantly increased with increasing age (p < 0.0001). Collectively, E. coli resistance rates severely increased during the study period, except for fosfomycin and nitrofurantoin. The need of monitoring studies about antibiotic nonsusceptibilities at local and regional levels are necessary to enhance the focus on antimicrobial stewardship, to reduce antimicrobial consumption and to detect alarming resistance mechanisms.
Collapse
Affiliation(s)
- Antonio Milano
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| | - Adela Sulejmani
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| | - Jari Intra
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| | - Maria Roberta Sala
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| | - Valerio Leoni
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| | - Davide Carcione
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale di Monza ASST-Monza, Desio Hospital, Desio, Italy
| |
Collapse
|
43
|
Host Colonization as a Major Evolutionary Force Favoring the Diversity and the Emergence of the Worldwide Multidrug-Resistant Escherichia coli ST131. mBio 2021; 12:e0145121. [PMID: 34425698 PMCID: PMC8406181 DOI: 10.1128/mbio.01451-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The emergence of multidrug-resistant Escherichia coli ST131 is a major worldwide public health problem in humans. According to the “one health” approach, this study investigated animal reservoirs of ST131, their relationships with human strains, and the genetic features associated with host colonization. High-quality genomes originating from human, avian, and canine hosts were classified on the basis of their accessory gene content using pangenomic. Pangenomic clusters and subclusters were specifically and significantly associated with hosts. The functions of clustering accessory genes were mainly enriched in functions involved in DNA acquisition, interactions, and virulence (e.g., pathogenesis, response to biotic stimulus and interaction between organisms). Accordingly, networks of cooccurrent host interaction factors were significantly associated with the pangenomic clusters and the originating hosts. The avian strains exhibited a specific content in virulence factors. Rarely found in humans, they corresponded to pathovars responsible for severe human infections. An emerging subcluster significantly associated with both human and canine hosts was evidenced. This ability to significantly colonize canine hosts in addition to humans was associated with a specific content in virulence factors (VFs) and metabolic functions encoded by a new pathogenicity island in ST131 and an improved fitness that is probably involved in its emergence. Overall, VF content, unlike the determinants of antimicrobial resistance, appeared as a key actor of bacterial host adaptation. The host dimension emerges as a major driver of genetic evolution that shapes ST131 genome, enhances its diversity, and favors its dissemination.
Collapse
|
44
|
Phan MD, Bottomley AL, Peters KM, Harry EJ, Schembri MA. Uncovering novel susceptibility targets to enhance the efficacy of third-generation cephalosporins against ESBL-producing uropathogenic Escherichia coli. J Antimicrob Chemother 2021; 75:1415-1423. [PMID: 32073605 DOI: 10.1093/jac/dkaa023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/17/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Uropathogenic Escherichia coli (UPEC) are a major cause of urinary tract infection (UTI), one of the most common infectious diseases in humans. UPEC are increasingly associated with resistance to multiple antibiotics. This includes resistance to third-generation cephalosporins, a common class of antibiotics frequently used to treat UTI. METHODS We employed a high-throughput genome-wide screen using saturated transposon mutagenesis and transposon directed insertion-site sequencing (TraDIS) together with phenotypic resistance assessment to identify key genes required for survival of the MDR UPEC ST131 strain EC958 in the presence of the third-generation cephalosporin cefotaxime. RESULTS We showed that blaCMY-23 is the major ESBL gene in EC958 responsible for mediating resistance to cefotaxime. Our screen also revealed that mutation of genes involved in cell division and the twin-arginine translocation pathway sensitized EC958 to cefotaxime. The role of these cell-division and protein-secretion genes in cefotaxime resistance was confirmed through the construction of mutants and phenotypic testing. Mutation of these genes also sensitized EC958 to other cephalosporins. CONCLUSIONS This work provides an exemplar for the application of TraDIS to define molecular mechanisms of resistance to antibiotics. The identification of mutants that sensitize UPEC to cefotaxime, despite the presence of a cephalosporinase, provides a framework for the development of new approaches to treat infections caused by MDR pathogens.
Collapse
Affiliation(s)
- Minh-Duy Phan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Amy L Bottomley
- The ithree institute, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kate M Peters
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth J Harry
- The ithree institute, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
45
|
Louka C, Ravensbergen SJ, Ott A, Zhou X, García-Cobos S, Friedrich AW, Pournaras S, Rosema S, Rossen JW, Stienstra Y, Bathoorn E. Predominance of CTX-M-15-producing ST131 strains among ESBL-producing Escherichia coli isolated from asylum seekers in the Netherlands. J Antimicrob Chemother 2021; 76:70-76. [PMID: 33009805 PMCID: PMC7729386 DOI: 10.1093/jac/dkaa395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/19/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives Numerous studies show increased prevalence of MDR bacteria amongst asylum seekers, but data on the molecular profiles of such strains are limited. We aimed to evaluate the molecular profiles of ESBL-producing Escherichia coli (ESBL-E. coli) strains isolated from asylum seekers and investigate their phylogenetic relatedness. Methods WGS data of ESBL-E. coli isolates from asylum seekers, retrieved from 1 January to 31 December 2016, were analysed to assess MLST STs, fim types, phylogroups and resistance genes. Fifty-two ESBL-E. coli isolates from the Dutch–German border region were used for genome comparison purposes as a control group. Results Among 112 ESBL-E. coli isolates from asylum seekers, originating mostly from Syria (n = 40) and Iraq (n = 15), the majority belonged to ST131 (21.4%) and ST10 (17.0%). The predominant gene for β-lactam resistance was blaCTX-M-15 (67.9%), followed by the often co-detected blaTEM-1B (39.3%). No mcr or carbapenemase genes were detected. The majority of the strains belonged to phylogroups B2 (38.4%) and A (32.1%), carrying fimH27 (25%) and fimH30 (19.6%). A core genome MLST minimum spanning tree did not reveal clusters containing strains from the asylum seekers and the control group. Five clusters were formed within the asylum seeker group, by strains isolated from people originating from different countries. Conclusions The most frequently isolated clones in this study were isolated on a regular basis within the Dutch population before the increase in the asylum seeker population. No mcr- or carbapenemase-producing clones were detected among the asylum seeker population. Minor clustering was observed amongst the asylum seeker strains.
Collapse
Affiliation(s)
- Christina Louka
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine/Infectious Diseases, Groningen, The Netherlands.,ESCMID Study Group for Infections in Travellers and Migrants, Basel, Switzerland
| | - Sofanne J Ravensbergen
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine/Infectious Diseases, Groningen, The Netherlands.,ESCMID Study Group for Infections in Travellers and Migrants, Basel, Switzerland
| | - Alewijn Ott
- Department of Medical Microbiology and Infection Prevention, Certe, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - Xuewei Zhou
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - Silvia García-Cobos
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - Alexander W Friedrich
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - Spyros Pournaras
- Department of Medical Microbiology, 'ATTIKON' University Hospital of Athens, Athens, Greece
| | - Sigrid Rosema
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - John W Rossen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| | - Ymkje Stienstra
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine/Infectious Diseases, Groningen, The Netherlands.,ESCMID Study Group for Infections in Travellers and Migrants, Basel, Switzerland
| | - Erik Bathoorn
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, The Netherlands
| |
Collapse
|
46
|
Multidrug Resistance Dissemination in Escherichia coli Isolated from Wild Animals: Bacterial Clones and Plasmid Complicity. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Objectives. Epidemiological data concerning third-generation cephalosporin (3GC) resistance in wild fauna are scarce. The aim of this study was to characterize the resistance genes, their genetic context, and clonal relatedness in 17 Escherichia coli resistant to 3GC isolated from wild animals. Methods. The isolates were characterized by short-read whole genome sequencing, and long-read sequencing was used for the hybrid assembly of plasmid sequences. Results. The 3GC resistance gene most identified in the isolates was the extended-spectrum β-lactamases (ESBL)-encoding gene blaCTX-M-1 (82.3%), followed by blaCTX-M-32 (5.9%), blaCTX-M-14 (5.9%), and blaSHV-12 (5.9%). E. coli isolates mainly belonged to the sequence types (STs) rarely reported from humans. The single nucleotide polymorphism (SNP)-based typing showed that most E. coli genomes from wild animals (wild boars, birds of prey, and buzzards) formed clonal clusters (<5 SNPs), showing a clonal dissemination crossing species boundaries. blaCTX-M-1-harboring IncI1-ST3 plasmid was the predominant ESBL-encoding plasmid (76.4%) in wild animal isolates. Plasmid comparison revealed a 110-kb self-transferable plasmid consisting of a conserved backbone and two variable regions involved in antimicrobial resistance and in interaction with recipient cells during conjugation. Conclusion. Our results highlighted the unexpected clonal dissemination of blaCTX-M-1-encoding clones and the complicity of IncI1-ST3 plasmid in the spread of blaCTX-M-1 within wild fauna.
Collapse
|
47
|
Varney AM, Smitten KL, Thomas JA, McLean S. Transcriptomic Analysis of the Activity and Mechanism of Action of a Ruthenium(II)-Based Antimicrobial That Induces Minimal Evolution of Pathogen Resistance. ACS Pharmacol Transl Sci 2021; 4:168-178. [PMID: 33615170 PMCID: PMC7887750 DOI: 10.1021/acsptsci.0c00159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Indexed: 01/30/2023]
Abstract
Increasing concern over rising levels of antibiotic resistance among pathogenic bacteria has prompted significant research into developing efficacious alternatives to antibiotic treatment. Previously, we have reported on the therapeutic activity of a dinuclear ruthenium(II) complex against pathogenic, multi-drug-resistant bacterial pathogens. Herein, we report that the solubility properties of this lead are comparable to those exhibited by orally available therapeutics that in comparison to clinically relevant antibiotics it induces very slow evolution of resistance in the uropathogenic, therapeutically resistant, E. coli strain EC958, and this resistance was lost when exposure to the compound was temporarily removed. With the aim of further investigating the mechanism of action of this compound, the regulation of nine target genes relating to the membrane, DNA damage, and other stress responses provoked by exposure to the compound was also studied. This analysis confirmed that the compound causes a significant transcriptional downregulation of genes involved in membrane transport and the tricarboxylic acid cycle. By contrast, expression of the chaperone protein-coding gene, spy, was significantly increased suggesting a requirement for repair of damaged proteins in the region of the outer membrane. The complex was also found to display activity comparable to that in E. coli in a range of other therapeutically relevant Gram-negative pathogens.
Collapse
Affiliation(s)
- Adam M. Varney
- School
of Science and Technology, Nottingham Trent
University, Nottingham NG11 8NS, United Kingdom
| | - Kirsty L. Smitten
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, United
Kingdom
| | - Jim A. Thomas
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, United
Kingdom
| | - Samantha McLean
- School
of Science and Technology, Nottingham Trent
University, Nottingham NG11 8NS, United Kingdom
| |
Collapse
|
48
|
Blackburn SA, Shepherd M, Robinson GK. Reciprocal Packaging of the Main Structural Proteins of Type 1 Fimbriae and Flagella in the Outer Membrane Vesicles of "Wild Type" Escherichia coli Strains. Front Microbiol 2021; 12:557455. [PMID: 33643229 PMCID: PMC7907004 DOI: 10.3389/fmicb.2021.557455] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/22/2021] [Indexed: 11/23/2022] Open
Abstract
Fundamental aspects of outer membrane vesicle (OMV) biogenesis and the engineering of producer strains have been major research foci for many in recent years. The focus of this study was OMV production in a variety of Escherichia coli strains including wild type (WT) (K12 and BW25113), mutants (from the Keio collection) and proprietary [BL21 and BL21 (DE3)] strains. The present study investigated the proteome and prospective mechanism that underpinned the key finding that the dominant protein present in E. coli K-12 WT OMVs was fimbrial protein monomer (FimA) (a polymerizable protein which is the key structural monomer from which Type 1 fimbriae are made). However, mutations in genes involved in fimbriae biosynthesis (ΔfimA, B, C, and F) resulted in the packaging of flagella protein monomer (FliC) (the major structural protein of flagella) into OMVs instead of FimA. Other mutations (ΔfimE, G, H, I, and ΔlrhA-a transcriptional regulator of fimbriation and flagella biosynthesis) lead to the packaging of both FimA and Flagellin into the OMVs. In the majority of instances shown within this research, the production of OMVs is considered in K-12 WT strains where structural appendages including fimbriae or flagella are temporally co-expressed throughout the growth curve as shown previously in the literature. The hypothesis, proposed and supported within the present paper, is that the vesicular packaging of the major FimA is reciprocally regulated with the major FliC in E. coli K-12 OMVs but this is abrogated in a range of mutated, non-WT E. coli strains. We also demonstrate, that a protein of interest (GFP) can be targeted to OMVs in an E. coli K-12 strain by protein fusion with FimA and that this causes normal packaging to be disrupted. The findings and underlying implications for host interactions and use in biotechnology are discussed.
Collapse
Affiliation(s)
| | | | - Gary K. Robinson
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
49
|
Nennig M, Llarena AK, Herold M, Mossong J, Penny C, Losch S, Tresse O, Ragimbeau C. Investigating Major Recurring Campylobacter jejuni Lineages in Luxembourg Using Four Core or Whole Genome Sequencing Typing Schemes. Front Cell Infect Microbiol 2021; 10:608020. [PMID: 33489938 PMCID: PMC7819963 DOI: 10.3389/fcimb.2020.608020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/23/2020] [Indexed: 12/31/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial gastroenteritis, which has motivated the monitoring of genetic profiles circulating in Luxembourg since 13 years. From our integrated surveillance using a genotyping strategy based on an extended MLST scheme including gyrA and porA markers, an unexpected endemic pattern was discovered in the temporal distribution of genotypes. We aimed to test the hypothesis of stable lineages occurrence by implementing whole genome sequencing (WGS) associated with comprehensive and internationally validated schemes. This pilot study assessed four WGS-based typing schemes to classify a panel of 108 strains previously identified as recurrent or sporadic profiles using this in-house typing system. The strain collection included four common lineages in human infection (N = 67) initially identified from recurrent combination of ST-gyrA-porA alleles also detected in non-human samples: veterinary (N = 19), food (N = 20), and environmental (N = 2) sources. An additional set of 19 strains belonging to sporadic profiles completed the tested panel. All the strains were processed by WGS by using Illumina technologies and by applying stringent criteria for filtering sequencing data; we ensure robustness in our genomic comparison. Four typing schemes were applied to classify the strains: (i) the cgMLST SeqSphere+ scheme of 637 loci, (ii) the cgMLST Oxford scheme of 1,343 loci, (iii) the cgMLST INNUENDO scheme of 678 loci, and (iv) the wgMLST INNUENDO scheme of 2,795 loci. A high concordance between the typing schemes was determined by comparing the calculated adjusted Wallace coefficients. After quality control and analyses with these four typing schemes, 60 strains were confirmed as members of the four recurrent lineages regardless of the method used (N = 32, 12, 7, and 9, respectively). Our results indicate that, regardless of the typing scheme used, epidemic or endemic signals were detected as reflected by lineage B (ST2254-gyrA9-porA1) in 2014 or lineage A (ST19-gyrA8-porA7), respectively. These findings support the clonal expansion of stable genomes in Campylobacter population exhibiting a multi-host profile and accounting for the majority of clinical strains isolated over a decade. Such recurring genotypes suggest persistence in reservoirs, sources or environment, emphasizing the need to investigate their survival strategy in greater depth.
Collapse
Affiliation(s)
- Morgane Nennig
- Epidemiology and Microbial Genomics, Laboratoire National de Santé, Dudelange, Luxembourg.,INRAE, Oniris, SECALIM, Nantes, France
| | - Ann-Katrin Llarena
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Malte Herold
- Epidemiology and Microbial Genomics, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Joël Mossong
- Epidemiology and Microbial Genomics, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Christian Penny
- Luxembourg Institute of Science and Technology, Environmental Research and Innovation Department, Belvaux, Luxembourg
| | - Serge Losch
- Laboratoire de Médecine Vétérinaire de l'Etat, Veterinary Services Administration, Dudelange, Luxembourg
| | | | - Catherine Ragimbeau
- Epidemiology and Microbial Genomics, Laboratoire National de Santé, Dudelange, Luxembourg
| |
Collapse
|
50
|
Chen WC, Hung CH, Chen YS, Cheng JS, Lee SSJ, Tseng FC, Cheng MF, Wang JL. Bloodstream Infections Caused by Extended-Spectrum Beta-Lactamase-Producing Escherichia coli in Patients with Liver Cirrhosis. Pathogens 2021; 10:pathogens10010037. [PMID: 33466521 PMCID: PMC7824895 DOI: 10.3390/pathogens10010037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 11/16/2022] Open
Abstract
Background: This study aimed to investigate the frequency of sequence type (ST) 131 strains and outcome of cirrhotic patients with bloodstream infections (BSIs) caused by extended-spectrum beta-lactamase-producing Escherichiacoli (ESBLEC) and non-extended-spectrum beta-lactamase-producing Escherichiacoli (NESBLEC). Methods: The incidence of ST 131 strains, hospital stay, and 30-day re-admission/mortality were compared between 51 ESBLEC and 51 NESBLEC bacteremic patients with cirrhosis. Results: ST 131 strains were found in 35.3% of the ESBLEC group and 0% of the NESBLEC group (p < 0.001). Mean hospital stay was 26.5 days in the ESBLEC group and 17.1 days in the NESBLEC group (p = 0.006). Thirty-day re-admission rates were 11.8% in the ESBLEC group and 5.9% in the NESBLEC group (p = 0.5). ST 131 strains were associated with 30-day re-admission (odds ratio: 4.5, 95% confidence interval: 1.1–18.9). Thirty-day mortality rate was 31.4% in the ESBLEC group and 23.5% in the NESBLEC group (p = 0.4). Conclusion: In patients with cirrhosis, the ESBLEC BSIs group had a higher frequency of ST 131 strains and longer hospital stay than the NESBLEC BSIs group with similar 30-day re-admission/mortality. ST 131 strains were associated with 30-day re-admission.
Collapse
Affiliation(s)
- Wen-Chi Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.C.); (J.-S.C.)
| | - Chih-Hsin Hung
- Department of Chemical Engineering and Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 840, Taiwan;
| | - Yao-Shen Chen
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-S.C.); (S.S.-J.L.)
| | - Jin-Shiung Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (W.-C.C.); (J.-S.C.)
| | - Susan Shin-Jung Lee
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (Y.-S.C.); (S.S.-J.L.)
| | - Fan-Chen Tseng
- Graduate Institute of Basic Medical Science, China Medical University and Department of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan;
| | - Ming-Fang Cheng
- Department of Chemical Engineering and Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 840, Taiwan;
- Department of Pediatrics, Kaohsiung Veterans General Hospital and School of Medicine, Kaohsiung 813, Taiwan
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Nursing, Fooyin University, Kaohsiung 831, Taiwan
- Correspondence: (M.-F.C.); (J.-L.W.); Tel.: +886-7-3468203 (M.-F.C.); +886-6-2757575 (J.-L.W.); Fax: +886-7-3468207 (M.-F.C.); +886-6-2766175 (J.-L.W.)
| | - Jiun-Ling Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan
- Department of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Correspondence: (M.-F.C.); (J.-L.W.); Tel.: +886-7-3468203 (M.-F.C.); +886-6-2757575 (J.-L.W.); Fax: +886-7-3468207 (M.-F.C.); +886-6-2766175 (J.-L.W.)
| |
Collapse
|