Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS One ; 18(5): e0285856, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37192202

RESUMEN

Pseudomonas aeruginosa causes a wide range of severe infections. Ceftazidime, a cephalosporin, is a key antibiotic for treating infections but a significant proportion of isolates are ceftazidime-resistant. The aim of this research was to identify mutations that contribute to resistance, and to quantify the impacts of individual mutations and mutation combinations. Thirty-five mutants with reduced susceptibility to ceftazidime were evolved from two antibiotic-sensitive P. aeruginosa reference strains PAO1 and PA14. Mutations were identified by whole genome sequencing. The evolved mutants tolerated ceftazidime at concentrations between 4 and 1000 times that of the parental bacteria, with most mutants being ceftazidime resistant (minimum inhibitory concentration [MIC] ≥ 32 mg/L). Many mutants were also resistant to meropenem, a carbapenem antibiotic. Twenty-eight genes were mutated in multiple mutants, with dacB and mpl being the most frequently mutated. Mutations in six key genes were engineered into the genome of strain PAO1 individually and in combinations. A dacB mutation by itself increased the ceftazidime MIC by 16-fold although the mutant bacteria remained ceftazidime sensitive (MIC < 32 mg/L). Mutations in ampC, mexR, nalC or nalD increased the MIC by 2- to 4-fold. The MIC of a dacB mutant was increased when combined with a mutation in ampC, rendering the bacteria resistant, whereas other mutation combinations did not increase the MIC above those of single mutants. To determine the clinical relevance of mutations identified through experimental evolution, 173 ceftazidime-resistant and 166 sensitive clinical isolates were analysed for the presence of sequence variants that likely alter function of resistance-associated genes. dacB and ampC sequence variants occur most frequently in both resistant and sensitive clinical isolates. Our findings quantify the individual and combinatorial effects of mutations in different genes on ceftazidime susceptibility and demonstrate that the genetic basis of ceftazidime resistance is complex and multifactorial.


Asunto(s)
Ceftazidima , Infecciones por Pseudomonas , Humanos , Ceftazidima/farmacología , Pseudomonas aeruginosa , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Antibacterianos/farmacología , Cefalosporinas/farmacología , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Combinación de Medicamentos , Compuestos de Azabiciclo/farmacología
3.
Biochem J ; 479(24): 2511-2527, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36504127

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa is one of leading causes of disability and mortality worldwide and the world health organisation has listed it with the highest priority for the need of new antimicrobial therapies. P. aeruginosa strains responsible for the poorest clinical outcomes express either ExoS or ExoU, which are injected into target host cells via the type III secretion system (T3SS). ExoS is a bifunctional cytotoxin that promotes intracellular survival of invasive P. aeruginosa by preventing targeting of the bacteria to acidified intracellular compartments. ExoU is a phospholipase which causes destruction of host cell plasma membranes, leading to acute tissue damage and bacterial dissemination. Fluoroquinolones are usually employed as a first line of therapy as they have been shown to be more active against P. aeruginosa in vitrothan other antimicrobial classes. Their overuse over the past decade, however, has resulted in the emergence of antibiotic resistance. In certain clinical situations, aminoglycosides have been shown to be more effective then fluoroquinolones, despite their reduced potency towards P. aeruginosa in vitro. In this study, we evaluated the effects of fluoroquinolones (moxifloxacin and ciprofloxacin) and aminoglycosides (tobramycin and gentamycin) on T3SS expression and toxicity, in corneal epithelial cell infection models. We discovered that tobramycin disrupted T3SS expression and reduced both ExoS and ExoU mediated cytotoxicity, protecting infected HCE-t cells at concentrations below the minimal inhibitory concentration (MIC). The fluoroquinolones moxifloxacin and ciprofloxacin, however, up-regulated the T3SS and did not inhibit and may have increased the cytotoxic effects of ExoS and ExoU.


Asunto(s)
Antiinfecciosos , Infecciones por Pseudomonas , Humanos , Fluoroquinolonas/farmacología , Fluoroquinolonas/metabolismo , Fluoroquinolonas/uso terapéutico , Aminoglicósidos/farmacología , Pseudomonas aeruginosa , Factores de Virulencia/metabolismo , Moxifloxacino/farmacología , Genotipo , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , ADP Ribosa Transferasas/genética , Antibacterianos/metabolismo , Tobramicina/metabolismo , Tobramicina/farmacología , Ciprofloxacina/metabolismo , Ciprofloxacina/farmacología , Antiinfecciosos/farmacología , Proteínas Bacterianas/metabolismo
4.
JCI Insight ; 7(22)2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36194492

RESUMEN

Pseudomonas aeruginosa undergoes diversification during infection of the cystic fibrosis (CF) lung. Understanding these changes requires model systems that capture the complexity of the CF lung environment. We previously identified loss-of-function mutations in the 2-component regulatory system sensor kinase gene pmrB in P. aeruginosa from CF lung infections and from experimental infection of mice. Here, we demonstrate that, while such mutations lowered in vitro minimum inhibitory concentrations for multiple antimicrobial classes, this was not reflected in increased antibiotic susceptibility in vivo. Loss of PmrB impaired aminoarabinose modification of LPS, increasing the negative charge of the outer membrane and promoting uptake of cationic antimicrobials. However, in vivo, this could be offset by increased membrane binding of other positively charged molecules present in lungs. The polyamine spermidine readily coated the surface of PmrB-deficient P. aeruginosa, reducing susceptibility to antibiotics that rely on charge differences to bind the outer membrane and increasing biofilm formation. Spermidine was elevated in lungs during P. aeruginosa infection in mice and during episodes of antimicrobial treatment in people with CF. These findings highlight the need to study antimicrobial resistance under clinically relevant environmental conditions. Microbial mutations carrying fitness costs in vitro may be advantageous during infection, where host resources can be utilized.


Asunto(s)
Antiinfecciosos , Fibrosis Quística , Ratones , Animales , Pseudomonas aeruginosa/genética , Poliaminas/metabolismo , Espermidina/metabolismo , Pruebas de Sensibilidad Microbiana , Fibrosis Quística/tratamiento farmacológico , Antiinfecciosos/metabolismo
5.
ERJ Open Res ; 7(2)2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33898620

RESUMEN

Bronchiectasis has been a largely overlooked disease area in respiratory medicine. This is reflected by a shortage of large-scale studies and lack of approved therapies, in turn leading to a variation of treatment across centres. BronchUK (Bronchiectasis Observational Cohort and Biobank UK) is a multicentre, prospective, observational cohort study working collaboratively with the European Multicentre Bronchiectasis Audit and Research Collaboration project. The inclusion criteria for patients entering the study are a clinical history consistent with bronchiectasis and computed tomography demonstrating bronchiectasis. Main exclusion criteria are 1) patients unable to provide informed consent, 2) bronchiectasis due to known cystic fibrosis or where bronchiectasis is not the main or co-dominant respiratory disease, 3) age <18 years, and 4) prior lung transplantation for bronchiectasis. The study is aligned to standard UK National Health Service (NHS) practice with an aim to recruit a minimum of 1500 patients from across at least nine secondary care centres. Patient data collected at baseline includes demographics, aetiology testing, comorbidities, lung function, radiology, treatments, microbiology and quality of life. Patients are followed up annually for a maximum of 5 years and, where able, blood and/or sputa samples are collected and stored in a central biobank. BronchUK aims to collect robust longitudinal data that can be used for analysis into current NHS practice and patient outcomes, and to become an integral resource to better inform future interventional studies in bronchiectasis.

6.
Microb Genom ; 7(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33720817

RESUMEN

The Liverpool epidemic strain (LES) is an important transmissible clonal lineage of Pseudomonas aeruginosa that chronically infects the lungs of people with cystic fibrosis (CF). Previous studies have focused on the genomics of the LES in a limited number of isolates, mostly from one CF centre in the UK, and from studies highlighting identification of the LES in Canada. Here we significantly extend the current LES genome database by genome sequencing 91 isolates from multiple CF centres across the UK, and we describe the comparative genomics of this large collection of LES isolates from the UK and Canada. Phylogenetic analysis revealed that the 145 LES genomes analysed formed a distinct clonal lineage when compared with the wider P. aeruginosa population. Notably, the isolates formed two clades: one associated with isolates from Canada, and the other associated with UK isolates. Further analysis of the UK LES isolates revealed clustering by clinic geography. Where isolates clustered closely together, the association was often supported by clinical data linking isolates or patients. When compared with the earliest known isolate, LESB58 (from 1988), many UK LES isolates shared common loss-of-function mutations, such as in genes gltR and fleR. Other loss-of-function mutations identified in previous studies as common adaptations during CF chronic lung infections were also identified in multiple LES isolates. Analysis of the LES accessory genome (including genomic islands and prophages) revealed variations in the carriage of large genomic regions, with some evidence for shared genomic island/prophage complement according to clinic location. Our study reveals divergence and adaptation during the spread of the LES, within the UK and between continents.


Asunto(s)
Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/transmisión , Pseudomonas aeruginosa/aislamiento & purificación , Adaptación Fisiológica , Canadá , Fibrosis Quística/complicaciones , Epidemias , Genoma Bacteriano , Humanos , Pulmón/microbiología , Infecciones Oportunistas/microbiología , Infecciones Oportunistas/transmisión , Filogenia , Infecciones por Pseudomonas/etiología , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/fisiología , Reino Unido/epidemiología
7.
Biochem J ; 478(3): 647-668, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33459338

RESUMEN

Pseudomonas aeruginosa has recently been highlighted by the World Health Organisation (WHO) as a major threat with high priority for the development of new therapies. In severe P. aeruginosa infections, the phospholipase activity of the type 3 secretion system toxin, ExoU, induces lysis of target host cells and results in the poorest clinical outcomes. We have developed an integrated pipeline to evaluate small molecule inhibitors of ExoU in vitro and in cultured cell models, including a disease-relevant corneal epithelial (HCE-T) scratch and infection model using florescence microscopy and cell viability assays. Compounds Pseudolipasin A, compound A and compound B were effective in vitro inhibitors of ExoU and mitigated P. aeruginosa ExoU-dependent cytotoxicity after infection of HCE-T cells at concentrations as low as 0.5 µM. Addition of the antimicrobial moxifloxacin controlled bacterial load, allowing these assays to be extended from 6 h to 24 h. P. aeruginosa remained cytotoxic to HCE-T cells with moxifloxacin, present at the minimal inhibitory concentration for 24 h, but, when used in combination with either Pseudolipasin A, compound A or compound B, a greater amount of viable cells and scratch healing were observed. Thus, our pipeline provides evidence that ExoU inhibitors could be used in combination with certain antimicrobials as a novel means to treat infections due to ExoU producing P. aeruginosa, as well as the means to identify more potent ExoU inhibitors for future therapeutics.


Asunto(s)
Antibacterianos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Pseudomonas aeruginosa/efectos de los fármacos , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Células Cultivadas , Sinergismo Farmacológico , Células Epiteliales , Epitelio Corneal/citología , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Moxifloxacino/farmacología , Conformación Proteica , Proteínas Recombinantes/efectos de los fármacos , Transfección
8.
J Cyst Fibros ; 20(6): 994-1002, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33358119

RESUMEN

BACKGROUND: The objective of this study was to explore the clinical and microbiological outcomes associated with substituting inhaled aztreonam lysine for an intravenous antibiotic in the treatment of acute pulmonary exacerbations of CF. METHODS: An open-label randomised crossover pilot trial was conducted at a UK CF centre among 16 adults with CF and P. aeruginosa infection. Median [IQR] age was 29.5 [24.5-32.5], mean ± SD forced expiratory volume in 1 second (FEV1) was 52.4 ± 14.7 % predicted. Over the course of two exacerbations, participants were randomised to sequentially receive 14 days of inhaled aztreonam lysine plus IV colistimethate (AZLI+IV), or dual IV antibiotics (IV+IV). Primary outcome was absolute change in % predicted FEV1. Other outcomes evaluated changes in quality of life, bacterial load and the lung microbiota. RESULTS: The difference between mean change in lung function at day 14 between AZLI+IV and IV+IV was +4.6% (95% CI 2.1-7.2, p=0.002). The minimum clinically important difference of the Cystic Fibrosis Revised Questionnaire (CFQ-R) was achieved more frequently with AZLI+IV (10/12, 83.3%) than IV+IV (7/16, 43.8%), p=0.05. No differences were observed for modulation of serum white cell count, C-reactive protein or sputum bacterial load. Microbiome compositional changes were observed with IV+IV (Bray-Curtis r2=0.14, p=0.02), but not AZLI+IV (r2=0.03, p=0.64). CONCLUSION: In adults with CF and P. aeruginosa infection experiencing an acute pulmonary exacerbation, AZLI+IV improved lung function and quality of life compared to the current standard treatment. These findings support the need for larger definitive trials of inhaled antibiotics in the acute setting. CLINICAL TRIAL REGISTRATION: EudraCT 2016-002832-34 ClinicalTrials.org NCT02894684.


Asunto(s)
Antibacterianos/administración & dosificación , Aztreonam/administración & dosificación , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/microbiología , Infecciones por Pseudomonas/tratamiento farmacológico , Administración por Inhalación , Adulto , Estudios Cruzados , Femenino , Humanos , Masculino , Proyectos Piloto , Brote de los Síntomas , Reino Unido
10.
FEMS Microbiol Lett ; 367(14)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32592387

RESUMEN

This study investigated the genetic basis of multidrug resistance in two strains of Achromobacter xylosoxidans isolated from patients attending a hospital in Thailand in 2012. These isolates were highly resistant to cephalosporins, aminoglycosides, fluoroquinolones, co-trimoxazole and carbapenems. Whole genome sequencing revealed that the two isolates were not clonally related and identified a carbapenem resistance gene-habouring integron (In687), residing in a novel genomic island, AcGI1. This In687 shares 100% identical nucleotide sequence with ones found in Acinetobacter baumannii Aci 16, isolated from the same hospital in 2007. We report the first analysis of multidrug-resistant A. xylosoxidans isolated in Thailand, and the first example of this island in A. xylosoxidans. Our data support the idea that resistance has spread in Thailand via horizontal gene transfer between species and suggest the possibility of A. xylosoxidans may serve as a reservoir of antibiotic resistance, especially in hospital setting.


Asunto(s)
Achromobacter denitrificans/genética , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Islas Genómicas , Infecciones por Bacterias Gramnegativas/microbiología , Integrones , Achromobacter denitrificans/clasificación , Achromobacter denitrificans/efectos de los fármacos , Achromobacter denitrificans/aislamiento & purificación , Aminoglicósidos/farmacología , Fluoroquinolonas/farmacología , Hospitales de Enseñanza , Humanos , Pruebas de Sensibilidad Microbiana , Filogenia , Análisis de Secuencia de ADN , Tailandia
11.
Nat Commun ; 11(1): 1370, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32170080

RESUMEN

Multidrug resistance (MDR) represents a global threat to health. Here, we used whole genome sequencing to characterise Pseudomonas aeruginosa MDR clinical isolates from a hospital in Thailand. Using long-read sequence data we obtained complete sequences of two closely related megaplasmids (>420 kb) carrying large arrays of antibiotic resistance genes located in discrete, complex and dynamic resistance regions, and revealing evidence of extensive duplication and recombination events. A comprehensive pangenomic and phylogenomic analysis indicates that: 1) these large plasmids comprise an emerging family present in different members of the Pseudomonas genus, and associated with multiple sources (geographical, clinical or environmental); 2) the megaplasmids encode diverse niche-adaptive accessory traits, including multidrug resistance; 3) the accessory genome of the megaplasmid family is highly flexible and diverse. The history of the megaplasmid family, inferred from our analysis of the available database, suggests that members carrying multiple resistance genes date back to at least the 1970s.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/genética , Genes Bacterianos/genética , Plásmidos/genética , Pseudomonas/genética , Antibacterianos/farmacología , ADN Bacteriano/genética , Evolución Molecular , Genómica , Humanos , Pruebas de Sensibilidad Microbiana , Filogenia , Plásmidos/clasificación , Plásmidos/aislamiento & purificación , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Tailandia , Secuenciación Completa del Genoma
12.
PLoS One ; 15(1): e0227535, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31999701

RESUMEN

BACKGROUND: Campylobacter jejuni is the most common bacterial cause of human infectious intestinal disease. METHODS: We genome sequenced 601 human C. jejuni isolates, obtained from two large prospective studies of infectious intestinal disease (IID1 [isolates from 1993-1996; n = 293] and IID2 [isolates from 2008-2009; n = 93]), the INTEGRATE project [isolates from 2016-2017; n = 52] and the ENIGMA project [isolates from 2017; n = 163]. RESULTS: There was a significant increase in the prevalence of the T86I mutation conferring resistance to fluoroquinolone between each of the three later studies (IID2, INTEGRATE and ENIGMA) and IID1. Although the distribution of major multilocus sequence types (STs) was similar between the studies, there were changes in both the abundance of minority STs associated with the T86I mutation, and the abundance of clones within single STs associated with the T86I mutation. DISCUSSION: Four population-based studies of community diarrhoea over a 25 year period revealed an increase over time in the prevalence of the T86I amongst isolates of C. jejuni associated with human gastrointestinal disease in the UK. Although associated with many STs, much of the increase is due to the expansion of clones associated with the resistance mutation.


Asunto(s)
Campylobacter jejuni/efectos de los fármacos , Campylobacter jejuni/genética , Farmacorresistencia Bacteriana/genética , Fluoroquinolonas/farmacología , Enfermedades Intestinales/microbiología , Mutación , Campylobacter jejuni/aislamiento & purificación , Campylobacter jejuni/fisiología , Niño , Genoma Bacteriano/genética , Humanos , Filogenia , Polimorfismo de Nucleótido Simple , Prevalencia , Reino Unido
13.
J Med Microbiol ; 69(1): 3-13, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31750813

RESUMEN

Pseudomonas aeruginosa, a versatile Gram-negative pathogen that can cause a wide range of infections, is the most common causative agent in cases of bacterial keratitis associated with contact-lens use. Corneal infections with P. aeruginosa often have poor clinical outcomes and can result in long and costly treatments. During the infection process, the pathogen exploits its large genome, encoding complex regulatory networks and a wide range of virulence factors, including motility and the secretion of various proteases and toxins. Although antibiotic resistance levels in the UK are low, higher levels have been seen in some other countries. In the face of increasing antibiotic resistance, alternative therapeutic approaches such as antivirulence strategies and phage therapy are being developed. There is increasing evidence to suggest that keratitis infections are associated with a phylogenetic subgroup of P. aeruginosa isolates carrying the gene encoding the potent cytotoxin exotoxin U, one of two mutually exclusive exotoxins secreted via the type III secretion system. The mechanisms behind this association are unclear, but understanding the genetic differences that predispose P. aeruginosa to cause corneal infections may allow for the development of targeted and more effective future treatments to reduce the morbidity of P. aeruginosa keratitis. In order to minimize the risk of severe P. aeruginosa eye infections, a wide range of contact-lens disinfection solutions are available. Constant exposure to biocides at a range of concentrations, from sub-inhibitory to inhibitory, could contribute to the development of resistance to both antibiotics and disinfectants.


Asunto(s)
Lentes de Contacto/efectos adversos , Queratitis/epidemiología , Queratitis/microbiología , Infecciones por Pseudomonas/epidemiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/aislamiento & purificación , Farmacorresistencia Bacteriana , Exotoxinas/metabolismo , Genotipo , Salud Global , Humanos , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/genética , Factores de Virulencia/metabolismo
14.
Antimicrob Agents Chemother ; 63(12)2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31570397

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen that causes a wide range of acute and chronic infections. An increasing number of isolates have mutations that make them antibiotic resistant, making treatment difficult. To identify resistance-associated mutations we experimentally evolved the antibiotic sensitive strain P. aeruginosa PAO1 to become resistant to three widely used anti-pseudomonal antibiotics, ciprofloxacin, meropenem and tobramycin. Mutants could tolerate up to 2048-fold higher concentrations of antibiotic than strain PAO1. Genome sequences were determined for thirteen mutants for each antibiotic. Each mutant had between 2 and 8 mutations. For each antibiotic at least 8 genes were mutated in multiple mutants, demonstrating the genetic complexity of resistance. For all three antibiotics mutations arose in genes known to be associated with resistance, but also in genes not previously associated with resistance. To determine the clinical relevance of mutations uncovered in this study we analysed the corresponding genes in 558 isolates of P. aeruginosa from patients with chronic lung disease and in 172 isolates from the general environment. Many genes identified through experimental evolution had predicted function-altering changes in clinical isolates but not in environmental isolates, showing that mutated genes in experimentally evolved bacteria can predict those that undergo mutation during infection. Additionally, large deletions of up to 479kb arose in experimentally evolved meropenem resistant mutants and large deletions were present in 87 of the clinical isolates. These findings significantly advance understanding of antibiotic resistance in P. aeruginosa and demonstrate the validity of experimental evolution in identifying clinically-relevant resistance-associated mutations.

15.
JMIR Res Protoc ; 8(9): e13941, 2019 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-31573952

RESUMEN

BACKGROUND: Diarrheal disease, which affects 1 in 4 people in the United Kingdom annually, is the most common cause of outbreaks in community and health care settings. Traditional surveillance methods tend to detect point-source outbreaks of diarrhea and vomiting; they are less effective at identifying low-level and intermittent food supply contamination. Furthermore, it can take up to 9 weeks for infections to be confirmed, reducing slow-burn outbreak recognition, potentially impacting hundreds or thousands of people over wide geographical areas. There is a need to address fundamental problems in traditional diarrheal disease surveillance because of underreporting and subsequent unconfirmed infection by patients and general practitioners (GPs); varying submission practices and selective testing of samples in laboratories; limitations in traditional microbiological diagnostics, meaning that the timeliness of sample testing and etiology of most cases remains unknown; and poorly integrated human and animal surveillance systems, meaning that identification of zoonoses is delayed or missed. OBJECTIVE: This study aims to detect anomalous patterns in the incidence of gastrointestinal disease in the (human) community; to target sampling; to test traditional diagnostic methods against rapid, modern, and sensitive molecular and genomic microbiology methods that identify and characterize responsible pathogens rapidly and more completely; and to determine the cost-effectiveness of rapid, modern, sensitive molecular and genomic microbiology methods. METHODS: Syndromic surveillance will be used to aid identification of anomalous patterns in microbiological events based on temporal associations, demographic similarities among patients and animals, and changes in trends in acute gastroenteritis cases using a point process statistical model. Stool samples will be obtained from patients' consulting GPs, to improve the timeliness of cluster detection and characterize the pathogens responsible, allowing health protection professionals to investigate and control outbreaks quickly, limiting their size and impact. The cost-effectiveness of the proposed system will be examined using formal cost-utility analysis to inform decisions on national implementation. RESULTS: The project commenced on April 1, 2013. Favorable approval was obtained from the Research Ethics Committee on June 15, 2015, and the first patient was recruited on October 13, 2015, with 1407 patients recruited and samples processed using traditional laboratory techniques as of March 2017. CONCLUSIONS: The overall aim of this study is to create a new One Health paradigm for detecting and investigating diarrhea and vomiting in the community in near-real time, shifting from passive human surveillance and management of laboratory-confirmed infection toward an integrated, interdisciplinary enhanced surveillance system including management of people with symptoms. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/13941.

16.
Proc Biol Sci ; 286(1912): 20191794, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31594506

RESUMEN

Transposable temperate phages randomly insert into bacterial genomes, providing increased supply and altered spectra of mutations available to selection, thus opening alternative evolutionary trajectories. Transposable phages accelerate bacterial adaptation to new environments, but their effect on adaptation to the social environment is unclear. Using experimental evolution of Pseudomonas aeruginosa in iron-limited and iron-rich environments, where the cost of producing cooperative iron-chelating siderophores is high and low, respectively, we show that transposable phages promote divergence into extreme siderophore production phenotypes. Iron-limited populations with transposable phages evolved siderophore overproducing clones alongside siderophore non-producing cheats. Low siderophore production was associated with parallel mutations in pvd genes, encoding pyoverdine biosynthesis, and pqs genes, encoding quinolone signalling, while high siderophore production was associated with parallel mutations in phenazine-associated gene clusters. Notably, some of these parallel mutations were caused by phage insertional inactivation. These data suggest that transposable phages, which are widespread in microbial communities, can mediate the evolutionary divergence of social strategies.


Asunto(s)
Pseudomonas aeruginosa/fisiología , Adaptación Fisiológica , Bacteriófagos , Evolución Biológica , Mutación , Fenazinas , Sideróforos
17.
J Med Microbiol ; 68(11): 1591-1595, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31553303

RESUMEN

Patient-derived isolates of the opportunistic pathogen Pseudomonas aeruginosa are frequently resistant to antibiotics due to the presence of sequence variants in resistance-associated genes. However, the frequency of antibiotic resistance and of resistance-associated sequence variants in environmental isolates of P. aeruginosa has not been well studied. Antimicrobial susceptibility testing (ciprofloxacin, ceftazidime, meropenem, tobramycin) of environmental (n=50) and cystic fibrosis (n=42) P. aeruginosa isolates was carried out. Following whole genome sequencing of all isolates, 25 resistance-associated genes were analysed for the presence of likely function-altering sequence variants. Environmental isolates were susceptible to all antibiotics with one exception, whereas patient-derived isolates had significant frequencies of resistance to each antibiotic and a greater number of likely resistance-associated genetic variants. These findings indicate that the natural environment does not act as a reservoir of antibiotic-resistant P. aeruginosa, supporting a model in which antibiotic susceptible environmental bacteria infect patients and develop resistance during infection.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Microbiología Ambiental , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Ceftazidima/farmacología , Ciprofloxacina/farmacología , Fibrosis Quística/microbiología , Genómica , Humanos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , Tobramicina/farmacología
18.
Ann Am Thorac Soc ; 16(11): 1375-1382, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31319678

RESUMEN

Rationale: Ivacaftor can greatly improve clinical outcomes in people with cystic fibrosis (CF) and has been shown to have in vitro antibacterial properties, yet the long-term microbiological outcomes of treatment are unknown.Objectives: To investigate changes in respiratory microbiology associated with long-term ivacaftor use.Methods: This was a retrospective cohort study using data from the UK CF Registry 2011-2016. Primary outcome was the annual prevalence ratios for key CF pathogens between ivacaftor users and their contemporaneous comparators. Multivariable log-binomial regression models were designed to adjust for confounders. Changes in Pseudomonas aeruginosa status were compared between groups using nonparametric maximum likelihood estimate for the purposes of Kaplan-Meier approximation.Results: Ivacaftor use was associated with early and sustained reduction in P. aeruginosa rates (2016 adjusted prevalence ratio, 0.68; 95% confidence interval, 0.58-0.79; P < 0.001) via a combination of increased clearance in those with infection (ivacaftor: 33/87 [37.9%] vs. nonivacaftor: 432/1,872 [22.8%]; P < 0.001) and reduced acquisition in those without infection (49/134 [36.6%] vs. 1,157/2,382 [48.6%]; P = 0.01). The improved prevalence of P. aeruginosa infection was independent of reduced sampling in the ivacaftor cohort. Ivacaftor was also associated with reduced prevalence of Staphylococcus aureus and Aspergillus spp. but not Burkholderia cepacia complex.Conclusions: In this study, long-term ivacaftor use was associated with reduced infection with important CF pathogens including P. aeruginosa. These findings have implications for antibiotic stewardship and the need for ongoing chronic antimicrobial therapy in this cohort.


Asunto(s)
Aminofenoles/uso terapéutico , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Activadores de Enzimas/uso terapéutico , Infecciones por Pseudomonas/epidemiología , Quinolonas/uso terapéutico , Adolescente , Adulto , Aspergillus/aislamiento & purificación , Burkholderia cepacia/aislamiento & purificación , Niño , Femenino , Humanos , Estimación de Kaplan-Meier , Funciones de Verosimilitud , Masculino , Análisis Multivariante , Pseudomonas aeruginosa/aislamiento & purificación , Sistema de Registros , Análisis de Regresión , Estudios Retrospectivos , Staphylococcus aureus/aislamiento & purificación , Resultado del Tratamiento , Reino Unido/epidemiología , Adulto Joven
19.
Microb Genom ; 5(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31170060

RESUMEN

Pseudomonas aeruginosa is a highly versatile, antibiotic-resistant Gram-negative bacterium known for causing opportunistic infections and contamination of industrial products. Despite extensive genomic analysis of clinical P. aeruginosa strains, no genomes exist for preservative-tolerant industrial strains. A unique collection of 69 industrial isolates was assembled and compared to clinical and environmental strains; 16 genetically distinct industrial strains were subjected to array tube genotyping, multilocus sequence typing and whole-genome sequencing. The industrial strains possessed high preservative tolerance and were dispersed widely across P. aeruginosa as a species, but recurrence of strains from the same lineage within specific industrial products and locations was identified. The industrial P. aeruginosa genomes (mean=7.0 Mb) were significantly larger than those of previously sequenced environmental (mean=6.5 Mb; n=19) and clinical (mean=6.6 Mb; n=66) strains. Complete sequencing of the P. aeruginosa industrial strain RW109, which encoded the largest genome (7.75 Mb), revealed a multireplicon structure including a megaplasmid (555 265 bp) and large plasmid (151 612 bp). The RW109 megaplasmid represented an emerging plasmid family conserved in seven industrial and two clinical P. aeruginosa strains, and associated with extremely stress-resilient phenotypes, including antimicrobial resistance and solvent tolerance. Here, by defining the detailed phylogenomics of P. aeruginosa industrial strains, we show that they uniquely possess multireplicon, megaplasmid-bearing genomes, and significantly greater genomic content worthy of further study.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Genoma Bacteriano/genética , Plásmidos/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , ADN Bacteriano/genética , Humanos , Microbiología Industrial , Filogenia , Replicón , Secuenciación Completa del Genoma
20.
Microorganisms ; 7(12)2019 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-31888268

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa employs the type III secretion system (T3SS) and four effector proteins, ExoS, ExoT, ExoU, and ExoY, to disrupt cellular physiology and subvert the host's innate immune response. Of the effector proteins delivered by the T3SS, ExoU is the most toxic. In P. aeruginosa infections, where the ExoU gene is expressed, disease severity is increased with poorer prognoses. This is considered to be due to the rapid and irreversible damage exerted by the phospholipase activity of ExoU, which cannot be halted before conventional antibiotics can successfully eliminate the pathogen. This review will discuss what is currently known about ExoU and explore its potential as a therapeutic target, highlighting some of the small molecule ExoU inhibitors that have been discovered from screening approaches.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA