Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
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.
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.

3.
PLoS Pathog ; 19(9): e1011630, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37669280

RESUMEN

Streptococcus pneumoniae is a leading cause of community-acquired pneumonia and bacteraemia and is capable of remarkable phenotypic plasticity, responding rapidly to environmental change. Pneumococcus is a nasopharyngeal commensal, but is responsible for severe, acute infections following dissemination within-host. Pneumococcus is adept at utilising host resources, but the airways are compartmentalised and those resources are not evenly distributed. Challenges and opportunities in metabolite acquisition within different airway niches may contribute to the commensal-pathogen switch when pneumococcus moves from nasopharynx into lungs. We used NMR to characterise the metabolic landscape of the mouse airways, in health and during infection. Using paired nasopharynx and lung samples from naïve animals, we identified fundamental differences in metabolite bioavailability between airway niches. Pneumococcal pneumonia was associated with rapid and dramatic shifts in the lung metabolic environment, whilst nasopharyngeal carriage led to only modest change in upper airway metabolite profiles. NMR spectra derived from the nasopharynx of mice infected with closely-related pneumococcal strains that differ in their colonisation potential could be distinguished from one another using multivariate dimensionality reduction methods. The resulting models highlighted that increased branched-chain amino acid (BCAA) bioavailability in nasopharynx is a feature of infection with the high colonisation potential strain. Subsequent analysis revealed increased expression of BCAA transport genes and increased intracellular concentrations of BCAA in that same strain. Movement from upper to lower airway environments is associated with shifting challenges in metabolic resource allocation for pneumococci. Efficient biosynthesis, liberation or acquisition of BCAA is a feature of adaptation to nasopharyngeal colonisation.


Asunto(s)
Nariz , Infecciones Neumocócicas , Animales , Ratones , Metabolómica , Streptococcus pneumoniae , Aminoácidos de Cadena Ramificada
4.
Microbiology (Reading) ; 169(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37526960

RESUMEN

Burkholderia multivorans is the dominant Burkholderia pathogen recovered from lung infection in people with cystic fibrosis. However, as an understudied pathogen there are knowledge gaps in relation to its population biology, phenotypic traits and useful model strains. A phylogenomic study of B. multivorans was undertaken using a total of 283 genomes, of which 73 were sequenced and 49 phenotypically characterized as part of this study. Average nucleotide identity analysis (ANI) and phylogenetic alignment of core genes demonstrated that the B. multivorans population separated into two distinct evolutionary clades, defined as lineage 1 (n=58 genomes) and lineage 2 (n=221 genomes). To examine the population biology of B. multivorans, a representative subgroup of 77 B. multivorans genomes (28 from the reference databases and the 49 novel short-read genome sequences) were selected based on multilocus sequence typing (MLST), isolation source and phylogenetic placement criteria. Comparative genomics was used to identify B. multivorans lineage-specific genes - ghrB_1 in lineage 1 and glnM_2 in lineage 2 - and diagnostic PCRs targeting them were successfully developed. Phenotypic analysis of 49 representative B. multivorans strains showed considerable inter-strain variance, but the majority of the isolates tested were motile and capable of biofilm formation. A striking absence of B. multivorans protease activity in vitro was observed, but no lineage-specific phenotypic differences were demonstrated. Using phylogenomic and phenotypic criteria, three model B. multivorans CF strains were identified, BCC0084 (lineage 1), BCC1272 (lineage 2a) and BCC0033 lineage 2b, and their complete genome sequences determined. B. multivorans CF strains BCC0033 and BCC0084, and the environmental reference strain, ATCC 17616, were all capable of short-term survival within a murine lung infection model. By mapping the population biology, identifying lineage-specific PCRs and model strains, we provide much needed baseline resources for future studies of B. multivorans.


Asunto(s)
Infecciones por Burkholderia , Burkholderia , Fibrosis Quística , Filogenia , Animales , Ratones , Burkholderia/clasificación , Burkholderia/genética , Infecciones por Burkholderia/complicaciones , Infecciones por Burkholderia/microbiología , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Tipificación de Secuencias Multilocus , Genoma Bacteriano/genética , Ratones Endogámicos BALB C , Femenino
5.
ACS Appl Mater Interfaces ; 15(17): 20822-20832, 2023 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-37074085

RESUMEN

High sulfur content polymers prepared by inverse vulcanization have many reported potential applications, including as novel antimicrobial materials. High sulfur content polymers usually have limited water-solubility and dispersibility due to their hydrophobic nature, which could limit the development of their applications. Herein, we report the formulation of high sulfur content polymeric nanoparticles by a nanoprecipitation and emulsion-based method. High sulfur content polymeric nanoparticles were found to have an inhibitory effect against important bacterial pathogens, including Gram-positive methicillin-resistant Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa. Salt-stable particles were formulated with the addition of a surfactant, which did not inhibit the antibacterial activity of the polymeric particles. Furthermore, the polymeric nanoparticles were found to inhibit S. aureus biofilm formation and exhibited low cytotoxicity against mammalian liver cells. Interaction of the polymeric particles with cellular thiols could be a potential mechanism of action against bacterial cells, as demonstrated by reaction with cysteine as a model thiol. The findings presented demonstrate methods of preparing aqueous dispersions of high sulfur content polymeric nanoparticles that could have useful biological applications.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Nanopartículas , Animales , Staphylococcus aureus , Polímeros/farmacología , Polímeros/química , Antibacterianos/farmacología , Antibacterianos/química , Azufre/química , Nanopartículas/química , Pruebas de Sensibilidad Microbiana , Mamíferos
6.
Sci Adv ; 9(12): eade1851, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36947610

RESUMEN

Sensing of pathogens by ubiquitination is a critical arm of cellular immunity. However, universal ubiquitination targets on microbes remain unidentified. Here, using in vitro, ex vivo, and in vivo studies, we identify the first protein-based ubiquitination substrates on phylogenetically diverse bacteria by unveiling a strategy that uses recognition of degron-like motifs. Such motifs form a new class of intra-cytosolic pathogen-associated molecular patterns (PAMPs). Their incorporation enabled recognition of nonubiquitin targets by host ubiquitin ligases. We find that SCFFBW7 E3 ligase, supported by the regulatory kinase, glycogen synthase kinase 3ß, is crucial for effective pathogen detection and clearance. This provides a mechanistic explanation for enhanced risk of infections in patients with chronic lymphocytic leukemia bearing mutations in F-box and WD repeat domain containing 7 protein. We conclude that exploitation of this generic pathogen sensing strategy allows conservation of host resources and boosts antimicrobial immunity.


Asunto(s)
Proteínas F-Box , Humanos , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Fosforilación , Ubiquitinación , Bacterias/metabolismo
7.
Cell Rep ; 42(2): 112054, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36724074

RESUMEN

Streptococcus pneumoniae is a pathogen of global morbidity and mortality. Pneumococcal pneumonia can lead to systemic infections associated with high rates of mortality. We find that, upon pneumococcal infection, pulmonary Treg cells are activated and have upregulated TNFR2 expression. TNFR2-deficient mice have compromised Treg cell responses and highly activated IL-17A-producing γδ T cell (γδT17) responses, resulting in significantly enhanced neutrophil infiltration, tissue damage, and rapid development of bacteremia, mirroring responses in Treg cell-depleted mice. Deletion of total Treg cells predominantly activate IFNγ-T cell responses, whereas adoptive transfer of TNFR2+ Treg cells specifically suppress the γδT17 response, suggesting a targeted control of γδT17 activation by TNFR2+ Treg cells. Blocking IL-17A at early stage of infection significantly reduces bacterial blood dissemination and improves survival in TNFR2-deficient mice. Our results demonstrate that TNFR2 is critical for Treg cell-mediated regulation of pulmonary γδT17-neutrophil axis, with impaired TNFR2+ Treg cell responses increasing susceptibility to disease.


Asunto(s)
Bacteriemia , Neumonía Neumocócica , Ratones , Animales , Neumonía Neumocócica/metabolismo , Linfocitos T Reguladores/metabolismo , Interleucina-17/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral , Pulmón/metabolismo , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo
8.
Microbiol Spectr ; 11(1): e0310322, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36507681

RESUMEN

The short generation time of many bacterial pathogens allows the accumulation of de novo mutations during routine culture procedures used for the preparation and propagation of bacterial stocks. Taking the major human pathogen Streptococcus pneumoniae as an example, we sought to determine the influence of standard laboratory handling of microbes on within-strain genetic diversity and explore how these changes influence virulence characteristics and experimental outcomes. A single culture of S. pneumoniae D39 grown overnight resulted in the enrichment of previously rare genotypes present in bacterial freezer stocks and the introduction of new variation to the bacterial population through the acquisition of mutations. A comparison of D39 stocks from different laboratories demonstrated how changes in bacterial population structure taking place during individual culture events can cumulatively lead to fixed, divergent change that profoundly alters virulence characteristics. The passage of D39 through mouse models of infection, a process used to standardize virulence, resulted in the enrichment of high-fitness genotypes that were originally rare (<2% frequency) in D39 culture collection stocks and the loss of previously dominant genotypes. In the most striking example, the selection of a <2%-frequency genotype carrying a mutation in sdhB, a gene thought to be essential for the establishment of lung infection, was associated with enhanced systemic virulence. Three separately passaged D39 cultures originating from the same frozen stocks showed considerable genetic divergence despite comparable virulence. IMPORTANCE Laboratory bacteriology involves the use of high-density cultures that we often assume to be clonal but that in reality are populations consisting of multiple genotypes at various abundances. We have demonstrated that the genetic structure of a single population of a widely used Streptococcus pneumoniae strain can be substantially altered by even short-term laboratory handling and culture and that, over time, this can lead to changes in virulence characteristics. Our findings suggest that caution should be applied when comparing data generated in different laboratories using the same strain but also when comparing data within laboratories over time. Given the dramatic reductions in the cost of next-generation sequencing technology in recent years, we advocate for the frequent sampling and sequencing of bacterial isolate collections.


Asunto(s)
Proteínas Bacterianas , Streptococcus pneumoniae , Animales , Ratones , Proteínas Bacterianas/genética , Mutación , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Virulencia/genética
9.
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
10.
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
11.
Am J Respir Crit Care Med ; 206(9): 1070-1080, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35649181

RESUMEN

Streptococcus pneumoniae (the pneumococcus) is the leading cause of pneumonia and bacterial meningitis. A number of recent studies indicate an association between the incidence of pneumococcal disease and exposure to air pollution. Although the epidemiological evidence is substantial, the underlying mechanisms by which the various components of air pollution (particulate matter and gases such as NO2 and SO2) can increase susceptibility to pneumococcal infection are less well understood. In this review, we summarize the various effects air pollution components have on pneumococcal pathogenesis and transmission; exposure to air pollution can enhance host susceptibility to pneumococcal colonization by impairing the mucociliary activity of the airway mucosa, reducing the function and production of key antimicrobial peptides, and upregulating an important pneumococcal adherence factor on respiratory epithelial cells. Air pollutant exposure can also impair the phagocytic killing ability of macrophages, permitting increased replication of S. pneumoniae. In addition, particulate matter has been shown to activate various extra- and intracellular receptors of airway epithelial cells, which may lead to increased proinflammatory cytokine production. This increases recruitment of innate immune cells, including macrophages and neutrophils. The inflammatory response that ensues may result in significant tissue damage, thereby increasing susceptibility to invasive disease, because it allows S. pneumoniae access to the underlying tissues and blood. This review provides an in-depth understanding of the interaction between air pollution and the pneumococcus, which has the potential to aid the development of novel treatments or alternative strategies to prevent disease, especially in areas with high concentrations of air pollution.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Infecciones Neumocócicas , Neumonía , Humanos , Streptococcus pneumoniae , Contaminación del Aire/análisis , Contaminantes Atmosféricos/análisis , Material Particulado/análisis , Neumonía/epidemiología , Infecciones Neumocócicas/complicaciones
12.
J Mater Chem B ; 10(22): 4153-4162, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35438120

RESUMEN

Elemental sulfur (S8), a by-product of the petroleum refining industries, possesses many favourable properties including photocatalytic activity and antibacterial activity, in addition to being intrinsically hydrophobic. Despite this, there is a relative lack of research employing elemental sulfur and/or sulfur copolymers within superhydrophobic materials design. In this work, we present the use of sulfur copolymers to produce superhydrophobic materials with advanced functionalities. Using inverse vulcanization and the use of a natural organic crosslinker, perillyl alcohol (PER), stable S8-PER copolymers were synthesised and later combined with silica (SiO2) nanoparticles, to achieve highly water repellent composites that displayed both antimicrobial and photocatalytic properties, in the absence of carcinogenic and/or expensive materials. Here, we investigated the antibacterial performance of coatings against the Staphylococcus aureus bacterial strain, where coatings displayed great promise for use in antifouling applications, as they were found to limit surface adhesion by more than 99%, when compared to uncoated glass samples. Furthermore, UV dye degradation tests were performed, utilizing the commercially available dye resazurin, and it was shown that coatings had the potential to simultaneously exhibit surface hydrophobicity and photoactivity, demonstrating a great advancement in the field of superhydrophobic materials.


Asunto(s)
Antiinfecciosos , Polímeros , Antibacterianos/farmacología , Polímeros/química , Polímeros/farmacología , Dióxido de Silicio/química , Azufre/química , Agua/química
13.
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
14.
Biomacromolecules ; 22(12): 5223-5233, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34784205

RESUMEN

Inverse vulcanization is a bulk polymerization method for synthesizing high sulfur content polymers from elemental sulfur, a byproduct of the petrochemical industry, with vinylic comonomers. There is growing interest in polysulfides as novel antimicrobial agents due to the antimicrobial activity of natural polysulfides found in garlic and onions (Tsao et al. J. Antimicrob. Chemother. 2001, 47, 665-670). Herein, we report the antibacterial properties of several inverse vulcanized polymers against Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa, two common causes of nosocomial infection and pathogens identified by the World Health Organization as priorities for antimicrobial development. High sulfur content polymers were synthesized with different divinyl comonomers and at different sulfur/comonomer ratios, to determine the effect of such variables on the antibacterial properties of the resulting materials. Furthermore, polymers were tested for their potential as antibacterial materials at different temperatures. It was found that the test temperature influenced the antibacterial efficacy of the polymers and could be related to the glass transition temperature of the polymer. These findings provide further understanding of the antibacterial properties of inverse vulcanized polymers and show that such polymers have the potential to be used as antibacterial surfaces.


Asunto(s)
Antibacterianos , Polímeros , Antibacterianos/química , Antibacterianos/farmacología , Polimerizacion , Polímeros/química , Polímeros/farmacología , Staphylococcus aureus , Azufre/química
15.
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
16.
Sci Rep ; 11(1): 6984, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772094

RESUMEN

Infection with Streptococcus pneumoniae is the leading cause of death in children and burden of disease is greatest where helminth infections are also common. We investigated the impact of intestinal helminth co-infection on pneumococcal carriage; a risk factor for invasive disease. We used a mouse co-infection model and clinical data to assess the impact of co-infection on carriage density. Co-infection in mice was associated with increased pneumococcal carriage density and dissemination into lungs. Helminth-infected children also exhibited increased carriage density as compared to uninfected children. Anthelmintic treatment may be a cost-effective method of reducing pneumococcal disease burden in lower-income countries.


Asunto(s)
Coinfección/microbiología , Helmintiasis/microbiología , Parasitosis Intestinales/microbiología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/aislamiento & purificación , Animales , Niño , Preescolar , Coinfección/epidemiología , Ecuador/epidemiología , Femenino , Helmintiasis/epidemiología , Humanos , Parasitosis Intestinales/epidemiología , Masculino , Ratones , Infecciones Neumocócicas/epidemiología , Factores de Riesgo
17.
Mol Biol Evol ; 38(6): 2209-2226, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33502519

RESUMEN

Streptococcus pneumoniae is a commensal of the human nasopharynx and a major cause of respiratory and invasive disease. We examined adaptation and evolution of pneumococcus, within nasopharynx and lungs, in an experimental system where the selective pressures associated with transmission were removed. This was achieved by serial passage of pneumococci, separately, in mouse models of nasopharyngeal carriage or pneumonia. Passaged pneumococci became more effective colonizers of the respiratory tract and we observed several examples of potential parallel evolution. The cell wall-modifying glycosyltransferase LafA was under strong selection during lung passage, whereas the surface expressed pneumococcal vaccine antigen gene pvaA and the glycerol-3-phosphate dehydrogenase gene gpsA were frequent targets of mutation in nasopharynx-passaged pneumococci. These mutations were not identified in pneumococci that were separately evolved by serial passage on laboratory agar. We focused on gpsA, in which the same single nucleotide polymorphism arose in two independently evolved nasopharynx-passaged lineages. We describe a new role for this gene in nasopharyngeal carriage and show that the identified single nucleotide change confers resistance to oxidative stress and enhanced nasopharyngeal colonization potential. We demonstrate that polymorphisms in gpsA arise and are retained during human colonization. These findings highlight how within-host environmental conditions can determine trajectories of bacterial evolution. Relative invasiveness or attack rate of pneumococcal lineages may be defined by genes that make niche-specific contributions to bacterial fitness. Experimental evolution in animal infection models is a powerful tool to investigate the relative roles played by pathogen virulence and colonization factors within different host niches.


Asunto(s)
Adaptación Biológica/genética , Evolución Biológica , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Animales , Femenino , Genoma Bacteriano , Humanos , Pulmón/microbiología , Ratones , Nasofaringe/microbiología , Distribución Aleatoria , Streptococcus pneumoniae/genética , Factores de Virulencia
19.
mBio ; 11(6)2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293378

RESUMEN

Streptococcus pneumoniae is a frequent colonizer of the human nasopharynx and a major cause of life-threating invasive infections such as pneumonia, meningitis and sepsis. Over 1 million people die every year due to invasive pneumococcal disease (IPD), mainly in developing countries. Serotype 1 is a common cause of IPD; however, unlike other serotypes, it is rarely found in the carrier state in the nasopharynx, which is often considered a prerequisite for disease. The aim of this study was to understand this dichotomy. We used murine models of carriage and IPD to characterize the pathogenesis of African serotype 1 (sequence type 217) pneumococcal strains obtained from the Queen Elizabeth Central Hospital in Blantyre, Malawi. We found that ST217 pneumococcal strains were highly virulent in a mouse model of invasive pneumonia, but in contrast to the generally accepted assumption, can also successfully establish nasopharyngeal carriage. Interestingly, we found that cocolonizing serotypes may proliferate in the presence of serotype 1, suggesting that acquisition of serotype 1 carriage could increase the risk of developing IPD by other serotypes. RNA sequencing analysis confirmed that key virulence genes associated with inflammation and tissue invasiveness were upregulated in serotype 1. These data reveal important new insights into serotype 1 pathogenesis, with implications for carriage potential and risk of invasive disease through interactions with other cocolonizing serotypes, an often-overlooked factor in transmission and disease progression.IMPORTANCE The pneumococcus causes serious diseases such as pneumonia, sepsis, and meningitis and is a major cause of morbidity and mortality worldwide. Serotype 1 accounts for the majority of invasive pneumococcal disease cases in sub-Saharan Africa but is rarely found during nasopharyngeal carriage. Understanding the mechanisms leading to nasopharyngeal carriage and invasive disease by this serotype can help reduce its burden on health care systems worldwide. In this study, we also uncovered the potential impact of serotype 1 on disease progression of other coinfecting serotypes, which can have important implications for vaccine efficacy. Understanding the interactions between different serotypes during nasopharyngeal carriage may lead to improved intervention methods and therapies to reduce pneumococcal invasive disease levels.


Asunto(s)
Portador Sano/microbiología , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Viabilidad Microbiana , Infecciones Neumocócicas/metabolismo , Serogrupo , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Factores de Tiempo , Virulencia
20.
PLoS Pathog ; 16(11): e1009016, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33216805

RESUMEN

The opportunistic pathogen Streptococcus pneumoniae has dual lifestyles: one of an asymptomatic colonizer in the human nasopharynx and the other of a deadly pathogen invading sterile host compartments. The latter triggers an overwhelming inflammatory response, partly driven via pore forming activity of the cholesterol dependent cytolysin (CDC), pneumolysin. Although pneumolysin-induced inflammation drives person-to-person transmission from nasopharynx, the primary reservoir for pneumococcus, it also contributes to high mortality rates, creating a bottleneck that hampers widespread bacterial dissemination, thus acting as a double-edged sword. Serotype 1 ST306, a widespread pneumococcal clone, harbours a non-hemolytic variant of pneumolysin (Ply-NH). Performing crystal structure analysis of Ply-NH, we identified Y150H and T172I as key substitutions responsible for loss of its pore forming activity. We uncovered a novel inter-molecular cation-π interaction, governing formation of the transmembrane ß-hairpins (TMH) in the pore state of Ply, which can be extended to other CDCs. H150 in Ply-NH disrupts this interaction, while I172 provides structural rigidity to domain-3, through hydrophobic interactions, inhibiting TMH formation. Loss of pore forming activity enabled improved cellular invasion and autophagy evasion, promoting an atypical intracellular lifestyle for pneumococcus, a finding that was corroborated in in vivo infection models. Attenuation of inflammatory responses and tissue damage promoted tolerance of Ply-NH-expressing pneumococcus in the lower respiratory tract. Adoption of this altered lifestyle may be necessary for ST306 due to its limited nasopharyngeal carriage, with Ply-NH, aided partly by loss of its pore forming ability, facilitating a benign association of SPN in an alternative, intracellular host niche.


Asunto(s)
Adaptación Fisiológica , Inflamación/microbiología , Mutación con Pérdida de Función , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Estreptolisinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Membrana Celular/microbiología , Colesterol/metabolismo , Citoplasma/microbiología , Femenino , Humanos , Ratones , Modelos Estructurales , Perforina/genética , Perforina/metabolismo , Alineación de Secuencia , Streptococcus pneumoniae/genética , Estreptolisinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA