Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
ISME J ; 18(1)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38647527

RESUMEN

Pseudomonas aeruginosa is a cause of chronic respiratory tract infections in people with cystic fibrosis (CF), non-CF bronchiectasis, and chronic obstructive pulmonary disease. Prolonged infection allows the accumulation of mutations and horizontal gene transfer, increasing the likelihood of adaptive phenotypic traits. Adaptation is proposed to arise first in bacterial populations colonizing upper airway environments. Here, we model this process using an experimental evolution approach. Pseudomonas aeruginosa PAO1, which is not airway adapted, was serially passaged, separately, in media chemically reflective of upper or lower airway environments. To explore whether the CF environment selects for unique traits, we separately passaged PAO1 in airway-mimicking media with or without CF-specific factors. Our findings demonstrated that all airway environments-sinus and lungs, under CF and non-CF conditions-selected for loss of twitching motility, increased resistance to multiple antibiotic classes, and a hyper-biofilm phenotype. These traits conferred increased airway colonization potential in an in vivo model. CF-like conditions exerted stronger selective pressures, leading to emergence of more pronounced phenotypes. Loss of twitching was associated with mutations in type IV pili genes. Type IV pili mediate surface attachment, twitching, and induction of cAMP signalling. We additionally identified multiple evolutionary routes to increased biofilm formation involving regulation of cyclic-di-GMP signalling. These included the loss of function mutations in bifA and dipA phosphodiesterase genes and activating mutations in the siaA phosphatase. These data highlight that airway environments select for traits associated with sessile lifestyles and suggest upper airway niches support emergence of phenotypes that promote establishment of lung infection.


Asunto(s)
Adaptación Fisiológica , Biopelículas , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiología , Pseudomonas aeruginosa/metabolismo , Infecciones por Pseudomonas/microbiología , Biopelículas/crecimiento & desarrollo , Animales , Pulmón/microbiología , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Sistemas de Mensajero Secundario , Fibrosis Quística/microbiología , Ratones , Humanos , Antibacterianos/farmacología , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Mutación , Fenotipo
2.
Nat Rev Microbiol ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689039

RESUMEN

The ecological and evolutionary mechanisms of antimicrobial resistance (AMR) emergence within patients and how these vary across bacterial infections are poorly understood. Increasingly widespread use of pathogen genome sequencing in the clinic enables a deeper understanding of these processes. In this Review, we explore the clinical evidence to support four major mechanisms of within-patient AMR emergence in bacteria: spontaneous resistance mutations; in situ horizontal gene transfer of resistance genes; selection of pre-existing resistance; and immigration of resistant lineages. Within-patient AMR emergence occurs across a wide range of host niches and bacterial species, but the importance of each mechanism varies between bacterial species and infection sites within the body. We identify potential drivers of such differences and discuss how ecological and evolutionary analysis could be embedded within clinical trials of antimicrobials, which are powerful but underused tools for understanding why these mechanisms vary between pathogens, infections and individuals. Ultimately, improving understanding of how host niche, bacterial species and antibiotic mode of action combine to govern the ecological and evolutionary mechanism of AMR emergence in patients will enable more predictive and personalized diagnosis and antimicrobial therapies.

3.
Curr Opin Microbiol ; 78: 102436, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38368839

RESUMEN

Bacteria have evolved a variety of defence mechanisms to protect against mobile genetic elements, including restriction-modification systems and CRISPR-Cas. In recent years, dozens of previously unknown defence systems (DSs) have been discovered. Notably, diverse DSs often coexist within the same genome, and some co-occur at frequencies significantly higher than would be expected by chance, implying potential synergistic interactions. Recent studies have provided evidence of defence mechanisms that enhance or complement one another. Here, we review the interactions between DSs at the mechanistic, regulatory, ecological and evolutionary levels.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Bacterias/genética , Evolución Biológica , Bacteriófagos/genética
4.
Nat Commun ; 15(1): 1547, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378698

RESUMEN

Pseudomonas aeruginosa is a major nosocomial pathogen that causes severe disease including sepsis. Carbapenem-resistant P. aeruginosa is recognised by the World Health Organisation as a priority 1 pathogen, with urgent need for new therapeutics. As such, there is renewed interest in using bacteriophages as a therapeutic. However, the dynamics of treating pan-resistant P. aeruginosa with phage in vivo are poorly understood. Using a pan-resistant P. aeruginosa in vivo infection model, phage therapy displays strong therapeutic potential, clearing infection from the blood, kidneys, and spleen. Remaining bacteria in the lungs and liver displays phage resistance due to limiting phage adsorption. Yet, resistance to phage results in re-sensitisation to a wide range of antibiotics. In this work, we use phage steering in vivo, pre-exposing a pan resistant P. aeruginosa infection with a phage cocktail to re-sensitise bacteria to antibiotics, clearing the infection from all organs.


Asunto(s)
Bacteriófagos , Terapia de Fagos , Infecciones por Pseudomonas , Humanos , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología , Pulmón/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Terapia de Fagos/métodos , Pseudomonas aeruginosa
5.
J Vis Exp ; (203)2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38251745

RESUMEN

Temperate phages are found integrated as prophages in the majority of bacterial genomes. Some prophages are cryptic and fixed in the bacterial chromosome, but others are active and can be triggered into a replicative form either spontaneously or by exposure to inducing factors. Prophages are commonly associated with the ability to confer toxin production or other virulence-associated traits on their host cell. More recent studies have shown they can play a much bigger role in altering the physiology of their hosts. The technique described here has enabled us to investigate how prophages affect gene expression in the opportunistic bacterium Pseudomonas aeruginosa. In this work, the growth of the wild-type P. aeruginosa strain PAO1 was compared with that of isogenic lysogens carrying different combinations of prophages from the Liverpool Epidemic Strain (LES) LESB58. In a lysogen culture, a proportion of bacterial cells will be supporting lytic bacteriophage replication (spontaneous induction) with a high level of expression per cell of late phage genes, such as those associated with the assembly of phage particles, thus masking the low-level gene expression associated with lysogen-restricted gene expression. The impact of spontaneous induction can thus obscure prophage gene expression across a lysogen population. Growth profiling experiments were used to identify spontaneous induction, which was minimal during the early exponential growth phase. This study reports how to prepare sample cultures during the early exponential growth phase and how to set up adequate controls despite low cell numbers. These protocols ensure the reliable and reproducible comparison of wild-type and lysogenic bacteria under various conditions, thus improving the transcriptomic profiling of prophage genomes and aiding in the identification of previously unrecognized prophage functions.


Asunto(s)
Bacteriófagos , Bacteriófagos/genética , Perfilación de la Expresión Génica , Técnicas de Tipificación Bacteriana , Recuento de Células , Cromosomas Bacterianos
6.
Crit Rev Microbiol ; : 1-16, 2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-37999716

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen that is responsible for infections in people living with chronic respiratory conditions, such as cystic fibrosis (CF) and non-CF bronchiectasis (NCFB). Traditionally, in people with chronic respiratory disorders, P. aeruginosa infection has been managed with a combination of inhaled and intravenous antibiotic therapies. However, due in part to the prolonged use of antibiotics in these people, the emergence of multi-drug resistant P. aeruginosa strains is a growing concern. The development of anti-virulence therapeutics may provide a new means of treating P. aeruginosa lung infections whilst also combatting the AMR crisis, as these agents are presumed to exert reduced pressure for the emergence of drug resistance as compared to antibiotics. However, the pipeline for developing anti-virulence therapeutics is poorly defined, and it is currently unclear as to whether in vivo and in vitro models effectively replicate the complex pulmonary environment sufficiently to enable development and testing of such therapies for future clinical use. Here, we discuss potential targets for P. aeruginosa anti-virulence therapeutics and the effectiveness of the current models used to study them. Focus is given to the difficulty of replicating the virulence gene expression patterns of P. aeruginosa in the CF and NCFB lung under laboratory conditions and to the challenges this poses for anti-virulence therapeutic development.

7.
Biology (Basel) ; 12(2)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36829557

RESUMEN

Probiotics may have the potential to protect against breast cancer, partly through systemic immunomodulatory action and active impact upon intestinal microbiota. Given a few clinical studies on their curative role, we conducted a systematic review of the potential effects of probiotics in breast cancer patients and survivors of breast cancer, aiming to support further clinical studies. A literature search was performed using PubMed, Embase, and the CENTRAL databases from inception through to March 2022. A total of eight randomized clinical trials were identified from thirteen articles published between 2004 and 2022. We evaluated quality-of-life measures, observed bacterial species and diversity indices, probiotic-related metabolites, inflammatory biomarkers, and other responses in breast cancer patients and survivors. Results were synthesized qualitatively and quantitatively using random-effects meta-analysis. Different probiotics supplements utilized included Lactobacillus species alone (Lacto), with or without estriol; probiotic combinations of Lactobacillus with Bifidobacterium (ProLB), with or without prebiotic fructooligosaccharides (FOS); ProLB plus Streptococcus and FOS (ProLBS + FOS); and ProLB plus Enterococcus (ProLBE). We found that use of ProLBS with FOS in breast cancer patients and use of ProLBE in survivors of breast cancer show potential benefits in countering obesity and dyslipidemia. ProLBS with FOS use decreases pro-inflammatory TNF-α in breast cancer survivors and improves quality of life in those with breast-cancer-associated lymphedema. Supplementing probiotics capsules (109 CFU) with a prebiotic and using an intake duration of 10 weeks could provide a better approach than probiotics alone.

8.
F1000Res ; 11: 1007, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36519007

RESUMEN

The respiratory tract is a compartmentalised and heterogenous environment. The nasopharynx and sinuses of the upper airways have distinct properties from the lungs and these differences may shape bacterial adaptation and evolution. Upper airway niches act as early colonisation sites for respiratory bacterial pathogens, including those, such as Pseudomonas aeruginosa, that can go on to establish chronic infection of the lungs in people with cystic fibrosis (CF). Despite the importance of upper airway environments in facilitating early adaptation to host environments, currently available in vitro models for study of respiratory infection in CF focus exclusively on the lungs. Furthermore, animal models, widely used to bridge the gap between in vitro systems and the clinical scenario, do not allow the upper and lower airways to be studied in isolation. We have developed a suite of culture media reproducing key features of the upper and lower airways, for the study of bacterial adaptation and evolution in different respiratory environments. For both upper and lower airway-mimicking media, we have developed formulations that reflect airway conditions in health and those that reflect the altered environment of the CF respiratory tract. Here, we describe the development and validation of these media and their use for study of genetic and phenotypic adaptations in P. aeruginosa during growth under upper or lower airway conditions in health and in CF.


Asunto(s)
Fibrosis Quística , Infecciones por Pseudomonas , Animales , Medios de Cultivo , Pseudomonas aeruginosa/genética , Pulmón
9.
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
10.
Prog Retin Eye Res ; 89: 101031, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34915112

RESUMEN

Bacterial keratitis is a common corneal infection that is treated with topical antimicrobials. By the time of presentation there may already be severe visual loss from corneal ulceration and opacity, which may persist despite treatment. There are significant differences in the associated risk factors and the bacterial isolates between high income and low- or middle-income countries, so that general management guidelines may not be appropriate. Although the diagnosis of bacterial keratitis may seem intuitive there are multiple uncertainties about the criteria that are used, which impacts the interpretation of investigations and recruitment to clinical studies. Importantly, the concept that bacterial keratitis can only be confirmed by culture ignores the approximately 50% of cases clinically consistent with bacterial keratitis in which investigations are negative. The aetiology of these culture-negative cases is unknown. Currently, the estimation of bacterial susceptibility to antimicrobials is based on data from systemic administration and achievable serum or tissue concentrations, rather than relevant corneal concentrations and biological activity in the cornea. The provision to the clinician of minimum inhibitory concentrations of the antimicrobials for the isolated bacteria would be an important step forward. An increase in the prevalence of antimicrobial resistance is a concern, but the effect this has on disease outcomes is yet unclear. Virulence factors are not routinely assessed although they may affect the pathogenicity of bacteria within species and affect outcomes. New technologies have been developed to detect and kill bacteria, and their application to bacterial keratitis is discussed. In this review we present the multiple areas of clinical uncertainty that hamper research and the clinical management of bacterial keratitis, and we address some of the assumptions and dogma that have become established in the literature.


Asunto(s)
Infecciones Bacterianas del Ojo , Queratitis , Antibacterianos/uso terapéutico , Bacterias , Toma de Decisiones Clínicas , Córnea/microbiología , Infecciones Bacterianas del Ojo/tratamiento farmacológico , Infecciones Bacterianas del Ojo/microbiología , Humanos , Queratitis/tratamiento farmacológico , Queratitis/microbiología , Incertidumbre
11.
J Med Microbiol ; 71(12)2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36748497

RESUMEN

Chronic respiratory infection is the primary driver of mortality in individuals with cystic fibrosis (CF). Existing drug screening models utilised in preclinical antimicrobial development are unable to mimic the complex CF respiratory environment. Consequently, antimicrobials showing promising activity in preclinical models often fail to translate through to clinical efficacy in people with CF. Model systems used in CF anti-infective drug discovery and development range from antimicrobial susceptibility testing in nutrient broth, through to 2D and 3D in vitro tissue culture systems and in vivo models. No single model fully recapitulates every key aspect of the CF lung. To improve the outcomes of people with CF (PwCF) it is necessary to develop a set of preclinical models that collectively recapitulate the CF respiratory environment to a high degree of accuracy. Models must be validated for their ability to mimic aspects of the CF lung and associated lung infection, through evaluation of biomarkers that can also be assessed following treatment in the clinic. This will give preclinical models greater predictive power for identification of antimicrobials with clinical efficacy. The landscape of CF is changing, with the advent of modulator therapies that correct the function of the CFTR protein, while antivirulence drugs and phage therapy are emerging alternative treatments to chronic infection. This review discusses the challenges faced in current antimicrobial development pipelines, including the advantages and disadvantages of current preclinical models and the impact of emerging treatments.


Asunto(s)
Antiinfecciosos , Fibrosis Quística , Infecciones por Pseudomonas , Humanos , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Antibacterianos/uso terapéutico , Antiinfecciosos/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/uso terapéutico , Modelos Biológicos , Infección Persistente , Pseudomonas aeruginosa , Infecciones por Pseudomonas/tratamiento farmacológico
12.
Front Cell Infect Microbiol ; 11: 665759, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33937104

RESUMEN

P. aeruginosa is classified as a priority one pathogen by the World Health Organisation, and new drugs are urgently needed, due to the emergence of multidrug-resistant (MDR) strains. Antimicrobial-resistant nosocomial pathogens such as P. aeruginosa pose unwavering and increasing threats. Antimicrobial stewardship has been a challenge during the COVID-19 pandemic, with a majority of those hospitalized with SARS-CoV2 infection given antibiotics as a safeguard against secondary bacterial infection. This increased usage, along with increased handling of sanitizers and disinfectants globally, may further accelerate the development and spread of cross-resistance to antibiotics. In addition, P. aeruginosa is the primary causative agent of morbidity and mortality in people with the life-shortening genetic disease cystic fibrosis (CF). Prolonged periods of selective pressure, associated with extended antibiotic treatment and the actions of host immune effectors, results in widespread adaptive and acquired resistance in P. aeruginosa found colonizing the lungs of people with CF. This review discusses the arsenal of resistance mechanisms utilized by P. aeruginosa, how these operate under high-stress environments such as the CF lung and how their interconnectedness can result in resistance to multiple antibiotic classes. Intrinsic, adaptive and acquired resistance mechanisms will be described, with a focus on how each layer of resistance can serve as a building block, contributing to multi-tiered resistance to antimicrobial activity. Recent progress in the development of anti-resistance adjuvant therapies, targeting one or more of these building blocks, should lead to novel strategies for combatting multidrug resistant P. aeruginosa. Anti-resistance adjuvant therapy holds great promise, not least because resistance against such therapeutics is predicted to be rare. The non-bactericidal nature of anti-resistance adjuvants reduce the selective pressures that drive resistance. Anti-resistance adjuvant therapy may also be advantageous in facilitating efficacious use of traditional antimicrobials, through enhanced penetration of the antibiotic into the bacterial cell. Promising anti-resistance adjuvant therapeutics and targets will be described, and key remaining challenges highlighted. As antimicrobial stewardship becomes more challenging in an era of emerging and re-emerging infectious diseases and global conflict, innovation in antibiotic adjuvant therapy can play an important role in extending the shelf-life of our existing antimicrobial therapeutic agents.


Asunto(s)
COVID-19 , Infecciones por Pseudomonas , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Humanos , Pandemias , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa , ARN Viral , SARS-CoV-2
13.
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
14.
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
15.
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
16.
Polymers (Basel) ; 11(12)2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31766551

RESUMEN

As the current global threat of antimicrobial resistance (AMR) persists, developing alternatives to antibiotics that are less susceptible to resistance is becoming an urgent necessity. Recent advances in biomaterials have allowed for the development and fabrication of materials with discrete surface nanotopographies that can deter bacteria from adhering to their surface. Using binary polymer blends of polystyrene (PS), poly(methyl methacrylate) (PMMA) and polycaprolactone (PCL) and varying their relative concentrations, PS/PCL, PS/PMMA and PCL/PMMA polymer demixed thin films were developed with nanoisland, nanoribbon and nanopit topographies. In the PS/PCL system, PS segregates to the air-polymer interface, with the lower solubility PCL preferring the substrate-polymer interface. In the PS/PMMA and PCL/PMMA systems, PMMA prefers the air-polymer interface due to its greater solubility and lower surface energy. The anti-adhesion efficacy of the demixed films were tested against Pseudomonas aeruginosa (PA14). PS/PCL and PCL/PMMA demixed films showed a significant reduction in cell counts adhered on their surfaces compared to pure polymer control films, while no reduction was observed in the counts adhered on PS/PMMA demixed films. While the specific morphology did not affect the adhesion, a relationship between bacterial cell and topographical surface feature size was apparent. If the surface feature was smaller than the cell, then an anti-adhesion effect was observed; if the surface feature was larger than the cell, then the bacteria preferred to adhere.

17.
Vet Rec ; 185(7): 206, 2019 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-31239295

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is an opportunistic pathogen and a major cause of infections. Widespread resistance in human infections are increasing the use of last resort antimicrobials such as polymyxins. However, these have been used for decades in veterinary medicine. Companion animals are an understudied source of antimicrobial resistant P. aeruginosa isolates. This study evaluated the susceptibility of P. aeruginosa veterinary isolates to polymyxins to determine whether the veterinary niche represents a potential reservoir of resistance genes for pathogenic bacteria in both animals and humans. METHODS AND RESULTS: Clinical P. aeruginosa isolates (n=24) from UK companion animals were compared for antimicrobial susceptibility to a panel of human-associated isolates (n=37). Minimum inhibitory concentration (MIC) values for polymyxin B and colistin in the companion animals was significantly higher than in human isolates (P=0.033 and P=0.013, respectively). Genotyping revealed that the veterinary isolates were spread throughout the P. aeruginosa population, with shared array types from human infections such as keratitis and respiratory infections, suggesting the potential for zoonotic transmission. Whole genome sequencing revealed mutations in genes associated with polymyxin resistance and other antimicrobial resistance-related genes. CONCLUSION: The high levels of resistance to polymyxin shown here, along with genetic similarities between some human and animal isolates, together suggest a need for sustained surveillance of this veterinary niche as a potential reservoir for resistant, clinically relevant bacteria in both animals and humans.


Asunto(s)
Farmacorresistencia Bacteriana , Mascotas/microbiología , Polimixinas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Humanos , Pseudomonas aeruginosa/aislamiento & purificación , Reino Unido , Medicina Veterinaria
18.
Nat Commun ; 9(1): 2635, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29980663

RESUMEN

Pseudomonas aeruginosa colonises the upper airway of cystic fibrosis (CF) patients, providing a reservoir of host-adapted genotypes that subsequently establish chronic lung infection. We previously experimentally-evolved P. aeruginosa in a murine model of respiratory tract infection and observed early-acquired mutations in pmrB, encoding the sensor kinase of a two-component system that promoted establishment and persistence of infection. Here, using proteomics, we show downregulation of proteins involved in LPS biosynthesis, antimicrobial resistance and phenazine production in pmrB mutants, and upregulation of proteins involved in adherence, lysozyme resistance and inhibition of the chloride ion channel CFTR, relative to wild-type strain LESB65. Accordingly, pmrB mutants are susceptible to antibiotic treatment but show enhanced adherence to airway epithelial cells, resistance to lysozyme treatment, and downregulate host CFTR expression. We propose that P. aeruginosa pmrB mutations in CF patients are subject to an evolutionary trade-off, leading to enhanced colonisation potential, CFTR inhibition, and resistance to host defences, but also to increased susceptibility to antibiotics.


Asunto(s)
Adaptación Fisiológica , Proteínas Bacterianas/metabolismo , Evolución Biológica , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/metabolismo , Factores de Transcripción/metabolismo , Células A549 , Adaptación Fisiológica/efectos de los fármacos , Animales , Antiinfecciosos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Recuento de Colonia Microbiana , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulación hacia Abajo , Células Epiteliales/metabolismo , Fimbrias Bacterianas/efectos de los fármacos , Fimbrias Bacterianas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Pulmón/microbiología , Pulmón/patología , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Movimiento , Muramidasa/metabolismo , Mutación/genética , Análisis de Componente Principal , Proteómica , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación
19.
FEMS Microbiol Lett ; 365(14)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29897457

RESUMEN

Pseudomonas aeruginosa is an important opportunistic pathogen, especially in the context of infections of cystic fibrosis (CF). In order to facilitate coordinated study of this pathogen, an international reference panel of P. aeruginosa isolates was assembled. Here we report the genome sequencing and analysis of 33 of these isolates and 7 reference genomes to further characterise this panel. Core genome single nucleotide variant phylogeny demonstrated that the panel strains are widely distributed amongst the P. aeruginosa population. Common loss-of-function mutations reported as adaptive during CF (such as in mucA and mexA) were identified amongst isolates from chronic respiratory infections. From the 40 strains analysed, 37 unique resistomes were predicted, based on the Resistance Gene Identifier method using the Comprehensive Antibiotic Resistance Database. Notably, hierarchical clustering and phylogenetic reconstructions based on the presence/absence of genomic islands (GIs), prophages and other regions of genome plasticity (RGPs) supported the subdivision of P. aeruginosa into two main groups. This is the largest, most diverse analysis of GIs and associated RGPs to date, and the results suggest that, at least at the largest clade grouping level (group 1 vs group 2), each group may be drawing upon distinct mobile gene pools.


Asunto(s)
Genoma Bacteriano/genética , Pseudomonas aeruginosa/genética , Adaptación Fisiológica/genética , Fibrosis Quística/microbiología , Farmacorresistencia Microbiana/genética , Genes Bacterianos/genética , Islas Genómicas/genética , Genómica , Humanos , Mutación , Filogenia , Polimorfismo de Nucleótido Simple , Profagos/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/fisiología , Análisis de Secuencia de ADN
20.
Microb Genom ; 4(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29547097

RESUMEN

Pseudomonas aeruginosa chronic infections of cystic fibrosis (CF) airways are a paradigm for within-host evolution with abundant evidence for rapid evolutionary adaptation and diversification. Recently emerged transmissible strains have spread globally, with the Liverpool Epidemic Strain (LES) the most common strain infecting the UK CF population. Previously we have shown that highly divergent lineages of LES can be found within a single infection, consistent with super-infection among a cross-sectional cohort of patients. However, despite its clinical importance, little is known about the impact of transmission on the genetic structure of these infections over time. To characterize this, we longitudinally sampled a meta-population of 15 genetic lineages within the LES over 13 months among seven chronically infected CF patients by genome sequencing. Comparative genome analyses of P. aeruginosa populations revealed that the presence of coexisting lineages contributed more to genetic diversity within an infection than diversification in situ. We observed rapid and substantial shifts in the relative abundance of lineages and replacement of dominant lineages, likely to represent super-infection by repeated transmissions. Lineage dynamics within patients led to rapid changes in the frequencies of mutations across suites of linked loci carried by each lineage. Many loci were associated with important infection phenotypes such as antibiotic resistance, mucoidy and quorum sensing, and were repeatedly mutated in different lineages. These findings suggest that transmission leads to rapid shifts in the genetic structure of CF infections, including in clinically important phenotypes such as antimicrobial resistance, and is likely to impede accurate diagnosis and treatment.


Asunto(s)
Fibrosis Quística/epidemiología , Fibrosis Quística/microbiología , Epidemias , Metagenómica , Infecciones por Pseudomonas/epidemiología , Pseudomonas aeruginosa/genética , Estudios Transversales , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Sitios Genéticos , Humanos , Estudios Longitudinales , Pseudomonas aeruginosa/aislamiento & purificación , Percepción de Quorum , Sistema Respiratorio/microbiología , Análisis de Secuencia de ADN , Esputo/microbiología , Reino Unido/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...