Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Microbiology (Reading) ; 169(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37505800

RESUMEN

Antimicrobial resistance (AMR) in bacteria is a major public health problem. The main route for AMR acquisition in clinically important bacteria is the horizontal transfer of plasmids carrying resistance genes. AMR plasmids allow bacteria to survive antibiotics, but they also entail physiological alterations in the host cell. Multiple studies over the last few years have indicated that these alterations can translate into a fitness cost when antibiotics are absent. However, due to technical limitations, most of these studies are based on analysing new associations between plasmids and bacteria generated in vitro, and we know very little about the effects of plasmids in their native bacterial hosts. In this study, we used a CRISPR-Cas9-tool to selectively cure plasmids from clinical enterobacteria to overcome this limitation. Using this approach, we were able to study the fitness effects of the carbapenem resistance plasmid pOXA-48 in 35 pOXA-48-carrying isolates recovered from hospitalized patients. Our results revealed that pOXA-48 produces variable effects across the collection of wild-type enterobacterial strains naturally carrying the plasmid, ranging from fitness costs to fitness benefits. Importantly, the plasmid was only associated with a significant fitness reduction in four out of 35 clones, and produced no significant changes in fitness in the great majority of isolates. Our results suggest that plasmids produce neutral fitness effects in most native bacterial hosts, helping to explain the great prevalence of plasmids in natural microbial communities.


Asunto(s)
Bacterias , Enterobacteriaceae , Humanos , Enterobacteriaceae/genética , Plásmidos/genética , Bacterias/genética , Antibacterianos/farmacología , beta-Lactamasas/genética
2.
PLoS Pathog ; 12(4): e1005576, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27124727

RESUMEN

Many bacterial species actively take up and recombine homologous DNA into their genomes, called natural competence, a trait that offers a means to identify the genetic basis of naturally occurring phenotypic variation. Here, we describe "transformed recombinant enrichment profiling" (TREP), in which natural transformation is used to generate complex pools of recombinants, phenotypic selection is used to enrich for specific recombinants, and deep sequencing is used to survey for the genetic variation responsible. We applied TREP to investigate the genetic architecture of intracellular invasion by the human pathogen Haemophilus influenzae, a trait implicated in persistence during chronic infection. TREP identified the HMW1 adhesin as a crucial factor. Natural transformation of the hmw1 operon from a clinical isolate (86-028NP) into a laboratory isolate that lacks it (Rd KW20) resulted in ~1,000-fold increased invasion into airway epithelial cells. When a distinct recipient (Hi375, already possessing hmw1 and its paralog hmw2) was transformed by the same donor, allelic replacement of hmw2AHi375 by hmw1A86-028NP resulted in a ~100-fold increased intracellular invasion rate. The specific role of hmw1A86-028NP was confirmed by mutant and western blot analyses. Bacterial self-aggregation and adherence to airway cells were also increased in recombinants, suggesting that the high invasiveness induced by hmw1A86-028NP might be a consequence of these phenotypes. However, immunofluorescence results found that intracellular hmw1A86-028NP bacteria likely invaded as groups, instead of as individual bacterial cells, indicating an emergent invasion-specific consequence of hmw1A-mediated self-aggregation.


Asunto(s)
Adhesinas Bacterianas/genética , Perfilación de la Expresión Génica/métodos , Infecciones por Haemophilus/microbiología , Western Blotting , Células Epiteliales/microbiología , Haemophilus influenzae/genética , Humanos , Espacio Intracelular/microbiología , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa
3.
Nat Commun ; 15(1): 4150, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755164

RESUMEN

Age-related neurodegenerative diseases involving amyloid aggregation remain one of the biggest challenges of modern medicine. Alterations in the gastrointestinal microbiome play an active role in the aetiology of neurological disorders. Here, we dissect the amyloidogenic properties of biofilm-associated proteins (BAPs) of the gut microbiota and their implications for synucleinopathies. We demonstrate that BAPs are naturally assembled as amyloid-like fibrils in insoluble fractions isolated from the human gut microbiota. We show that BAP genes are part of the accessory genomes, revealing microbiome variability. Remarkably, the abundance of certain BAP genes in the gut microbiome is correlated with Parkinson's disease (PD) incidence. Using cultured dopaminergic neurons and Caenorhabditis elegans models, we report that BAP-derived amyloids induce α-synuclein aggregation. Our results show that the chaperone-mediated autophagy is compromised by BAP amyloids. Indeed, inoculation of BAP fibrils into the brains of wild-type mice promote key pathological features of PD. Therefore, our findings establish the use of BAP amyloids as potential targets and biomarkers of α-synucleinopathies.


Asunto(s)
Amiloide , Biopelículas , Caenorhabditis elegans , Neuronas Dopaminérgicas , Microbioma Gastrointestinal , Enfermedad de Parkinson , alfa-Sinucleína , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Humanos , Biopelículas/crecimiento & desarrollo , Amiloide/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/microbiología , Enfermedad de Parkinson/patología , Ratones , Neuronas Dopaminérgicas/metabolismo , Autofagia , Enfermedades Neurodegenerativas/metabolismo , Ratones Endogámicos C57BL , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Encéfalo/metabolismo , Encéfalo/patología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
4.
Nat Commun ; 15(1): 4731, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830889

RESUMEN

Major antibiotic groups are losing effectiveness due to the uncontrollable spread of antimicrobial resistance (AMR) genes. Among these, ß-lactam resistance genes -encoding ß-lactamases- stand as the most common resistance mechanism in Enterobacterales due to their frequent association with mobile genetic elements. In this context, novel approaches that counter mobile AMR are urgently needed. Collateral sensitivity (CS) occurs when the acquisition of resistance to one antibiotic increases susceptibility to another antibiotic and can be exploited to eliminate AMR selectively. However, most CS networks described so far emerge as a consequence of chromosomal mutations and cannot be leveraged to tackle mobile AMR. Here, we dissect the CS response elicited by the acquisition of a prevalent antibiotic resistance plasmid to reveal that the expression of the ß-lactamase gene blaOXA-48 induces CS to colistin and azithromycin. We next show that other clinically relevant mobile ß-lactamases produce similar CS responses in multiple, phylogenetically unrelated E. coli strains. Finally, by combining experiments with surveillance data comprising thousands of antibiotic susceptibility tests, we show that ß-lactamase-induced CS is pervasive within Enterobacterales. These results highlight that the physiological side-effects of ß-lactamases can be leveraged therapeutically, paving the way for the rational design of specific therapies to block mobile AMR or at least counteract their effects.


Asunto(s)
Antibacterianos , Escherichia coli , Pruebas de Sensibilidad Microbiana , beta-Lactamasas , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Antibacterianos/farmacología , Sensibilidad Colateral al uso de Fármacos/genética , Plásmidos/genética , Azitromicina/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Resistencia betalactámica/genética
5.
Front Microbiol ; 14: 1253623, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38179447

RESUMEN

Antibiotic resistance is a major Public Health challenge worldwide. Mechanisms other than resistance are described as contributors to therapeutic failure. These include heteroresistance and tolerance, which escape the standardized procedures used for antibiotic treatment decision-making as they do not involve changes in minimal inhibitory concentration (MIC). Haemophilus influenzae causes chronic respiratory infection and is associated with exacerbations suffered by chronic obstructive pulmonary disease (COPD) patients. Although resistance to imipenem is rare in this bacterial species, heteroresistance has been reported, and antibiotic tolerance cannot be excluded. Moreover, development of antibiotic heteroresistance or tolerance during within-host H. influenzae pathoadaptive evolution is currently unknown. In this study, we assessed imipenem resistance, heteroresistance and tolerance in a previously sequenced longitudinal collection of H. influenzae COPD respiratory isolates. The use of Etest, disc diffusion, population analysis profiling, tolerance disc (TD)-test methods, and susceptibility breakpoint criteria when available, showed a significant proportion of imipenem heteroresistance with differences in terms of degree among strains, absence of imipenem tolerance, and no specific trends among serial and clonally related strains could be established. Analysis of allelic variation in the ftsI, acrA, acrB, and acrR genes rendered a panel of polymorphisms only found in heteroresistant strains, but gene expression and genome-wide analyses did not show clear genetic traits linked to heteroresistance. In summary, a significant proportion of imipenem heteroresistance was observed among H. influenzae strains isolated from COPD respiratory samples over time. These data should be useful for making more accurate clinical recommendations to COPD patients.

6.
Microbiol Spectr ; 11(6): e0099323, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37795992

RESUMEN

IMPORTANCE: Genomic diversity of nontypeable H. influenzae strains confers phenotypic heterogeneity. Multiple strains of H. influenzae can be simultaneously isolated from clinical specimens, but we lack detailed information about polyclonal infection dynamics by this pathogen. A long-term barrier to our understanding of this host-pathogen interplay is the lack of genetic tools for strain engineering and differential labeling. Here, we present a novel plasmid toolkit named pTBH (toolbox for Haemophilus), with standardized modules for fluorescent or bioluminescent labeling, adapted to H. influenzae requirements but designed to be versatile so it can be utilized in other bacterial species. We present detailed experimental and quantitative image analysis methods, together with proof-of-principle examples, and show the ample possibilities of 3D microscopy, combined with quantitative image analysis, to model H. influenzae polyclonal infection lifestyles and unravel the co-habitation and co-infection dynamics of this respiratory pathogen.


Asunto(s)
Infecciones por Haemophilus , Haemophilus influenzae , Humanos , Haemophilus influenzae/genética , Sistema Respiratorio , Infecciones por Haemophilus/microbiología , Microscopía
7.
mBio ; 12(3): e0078921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34154422

RESUMEN

Genetic variants arising from within-patient evolution shed light on bacterial adaptation during chronic infection. Contingency loci generate high levels of genetic variation in bacterial genomes, enabling adaptation to the stringent selective pressures exerted by the host. A significant gap in our understanding of phase-variable contingency loci is the extent of their contribution to natural infections. The human-adapted pathogen nontypeable Haemophilus influenzae (NTHi) causes persistent infections, which contribute to underlying disease progression. The phase-variable high-molecular-weight (HMW) adhesins located on the NTHi surface mediate adherence to respiratory epithelial cells and, depending on the allelic variant, can also confer high epithelial invasiveness or hyperinvasion. In this study, we characterize the dynamics of HMW-mediated hyperinvasion in living cells and identify a specific HMW binding domain shared by hyperinvasive NTHi isolates of distinct pathological origins. Moreover, we observed that HMW expression decreased over time by using a longitudinal set of persistent NTHi strains collected from chronic obstructive pulmonary disease (COPD) patients, resulting from increased numbers of simple-sequence repeats (SSRs) downstream of the functional P2hmw1A promoter, which is the one primarily driving HMW expression. Notably, the increased SSR numbers at the hmw1 promoter region also control a phenotypic switch toward lower bacterial intracellular invasion and higher biofilm formation, likely conferring adaptive advantages during chronic airway infection by NTHi. Overall, we reveal novel molecular mechanisms of NTHi pathoadaptation based on within-patient lifestyle switching controlled by phase variation. IMPORTANCE Human-adapted bacterial pathogens have evolved specific mechanisms to colonize their host niche. Phase variation is a contingency strategy to allow adaptation to changing conditions, as phase-variable bacterial loci rapidly and reversibly switch their expression. Several NTHi adhesins are phase variable. These adhesins are required for colonization but also immunogenic, in such a way that bacteria with lower adhesin levels are better equipped to survive an immune response, making their contribution to natural infections unclear. We show here that the major NTHi adhesin HMW1A displays allelic variation, which can drive a phase-variable epithelial hyperinvasion phenotype. Over time, hmw1A phase variation lowers adhesin expression, which controls an NTHi lifestyle switch from high epithelial invasiveness to lower invasion and higher biofilm formation. This reversible loss of function aligns with the previously stated notion that epithelial infection is essential for NTHi infection establishment, but once established, persistence favors gene inactivation, in this case facilitating biofilm growth.


Asunto(s)
Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Variación Genética , Genoma Bacteriano , Haemophilus influenzae/genética , Haemophilus influenzae/metabolismo , Adaptación Fisiológica/genética , Adhesinas Bacterianas/clasificación , Adhesión Bacteriana/genética , Adhesión Bacteriana/fisiología , Biopelículas , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Humanos , Regiones Promotoras Genéticas
8.
Biomolecules ; 9(12)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31861238

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by abnormal inflammation and impaired airway immunity, providing an opportunistic platform for nontypeable Haemophilus influenzae (NTHi) infection. In this context, therapies targeting not only overactive inflammation without significant adverse effects, but also infection are of interest. Increasing evidence suggests that polyphenols, plant secondary metabolites with anti-inflammatory and antimicrobial properties, may be protective. Here, a Cistus salviifolius plant extract containing quercetin, myricetin, and punicalagin was shown to reduce NTHi viability. Analysis of these polyphenols revealed that quercetin has a bactericidal effect on NTHi, does not display synergies, and that bacteria do not seem to develop resistance. Moreover, quercetin lowered NTHi airway epithelial invasion through a mechanism likely involving inhibition of Akt phosphorylation, and reduced the expression of bacterially-induced proinflammatory markers il-8, cxcl-1, il-6, pde4b, and tnfα. We further tested quercetin's effect on NTHi murine pulmonary infection, showing a moderate reduction in bacterial counts and significantly reduced expression of proinflammatory genes, compared to untreated mice. Quercetin administration during NTHi infection on a zebrafish septicemia infection model system showed a bacterial clearing effect without signs of host toxicity. In conclusion, this study highlights the therapeutic potential of the xenohormetic molecule quercetin against NTHi infection.


Asunto(s)
Antibacterianos/farmacología , Infecciones por Haemophilus/tratamiento farmacológico , Haemophilus influenzae/efectos de los fármacos , Extractos Vegetales/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Quercetina/farmacología , Células A549 , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Cistus/química , Modelos Animales de Enfermedad , Femenino , Infecciones por Haemophilus/microbiología , Humanos , Inmunomodulación/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Quercetina/química , Quercetina/aislamiento & purificación , Células Tumorales Cultivadas , Pez Cebra
9.
Virulence ; 10(1): 315-333, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30973092

RESUMEN

Nutrient iron sequestration is the most significant form of nutritional immunity and causes bacterial pathogens to evolve strategies of host iron scavenging. Cigarette smoking contains iron particulates altering lung and systemic iron homeostasis, which may enhance colonization in the lungs of patients suffering chronic obstructive pulmonary disease (COPD) by opportunistic pathogens such as nontypeable. NTHi is a heme auxotroph, and the NTHi genome contains multiple heme acquisition systems whose role in pulmonary infection requires a global understanding. In this study, we determined the relative contribution to NTHi airway infection of the four heme-acquisition systems HxuCBA, PE, SapABCDFZ, and HbpA-DppBCDF that are located at the bacterial outer membrane or the periplasm. Our computational studies provided plausible 3D models for HbpA, SapA, PE, and HxuA interactions with heme. Generation and characterization of single mutants in the hxuCBA, hpe, sapA, and hbpA genes provided evidence for participation in heme binding-storage and inter-bacterial donation. The hxuA, sapA, hbpA, and hpe genes showed differential expression and responded to heme. Moreover, HxuCBA, PE, SapABCDFZ, and HbpA-DppBCDF presented moonlighting properties related to resistance to antimicrobial peptides or glutathione import, together likely contributing to the NTHi-host airway interplay, as observed upon cultured airway epithelia and in vivo lung infection. The observed multi-functionality was shown to be system-specific, thus limiting redundancy. Together, we provide evidence for heme uptake systems as bacterial factors that act in a coordinated and multi-functional manner to subvert nutritional- and other sources of host innate immunity during NTHi airway infection.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Haemophilus influenzae/patogenicidad , Hemo/metabolismo , Interacciones Huésped-Patógeno , Pulmón/microbiología , Infecciones del Sistema Respiratorio/microbiología , Células A549 , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Sitios de Unión , Simulación por Computador , Femenino , Proteínas de Unión al Hemo/genética , Proteínas de Unión al Hemo/metabolismo , Humanos , Ratones , Simulación del Acoplamiento Molecular
10.
mBio ; 9(5)2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254117

RESUMEN

Tracking bacterial evolution during chronic infection provides insights into how host selection pressures shape bacterial genomes. The human-restricted opportunistic pathogen nontypeable Haemophilus influenzae (NTHi) infects the lower airways of patients suffering chronic obstructive pulmonary disease (COPD) and contributes to disease progression. To identify bacterial genetic variation associated with bacterial adaptation to the COPD lung, we sequenced the genomes of 92 isolates collected from the sputum of 13 COPD patients over 1 to 9 years. Individuals were colonized by distinct clonal types (CTs) over time, but the same CT was often reisolated at a later time or found in different patients. Although genomes from the same CT were nearly identical, intra-CT variation due to mutation and recombination occurred. Recurrent mutations in several genes were likely involved in COPD lung adaptation. Notably, nearly a third of CTs were polymorphic for null alleles of ompP1 (also called fadL), which encodes a bifunctional membrane protein that both binds the human carcinoembryonic antigen-related cell adhesion molecule 1 (hCEACAM1) receptor and imports long-chain fatty acids (LCFAs). Our computational studies provide plausible three-dimensional models for FadL's interaction with hCEACAM1 and LCFA binding. We show that recurrent fadL mutations are likely a case of antagonistic pleiotropy, since loss of FadL reduces NTHi's ability to infect epithelia but also increases its resistance to bactericidal LCFAs enriched within the COPD lung. Supporting this interpretation, truncated fadL alleles are common in publicly available NTHi genomes isolated from the lower airway tract but rare in others. These results shed light on molecular mechanisms of bacterial pathoadaptation and guide future research toward developing novel COPD therapeutics.IMPORTANCE Nontypeable Haemophilus influenzae is an important pathogen in patients with chronic obstructive pulmonary disease (COPD). To elucidate the bacterial pathways undergoing in vivo evolutionary adaptation, we compared bacterial genomes collected over time from 13 COPD patients and identified recurrent genetic changes arising in independent bacterial lineages colonizing different patients. Besides finding changes in phase-variable genes, we found recurrent loss-of-function mutations in the ompP1 (fadL) gene. We show that loss of OmpP1/FadL function reduces this bacterium's ability to infect cells via the hCEACAM1 epithelial receptor but also increases its resistance to bactericidal fatty acids enriched within the COPD lung, suggesting a case of antagonistic pleiotropy that restricts ΔfadL strains' niche. These results show how H. influenzae adapts to host-generated inflammatory mediators in the COPD airways.


Asunto(s)
Adaptación Biológica , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/genética , Neumonía Bacteriana/microbiología , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Anciano , Anciano de 80 o más Años , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Biología Computacional , Proteínas de Transporte de Ácidos Grasos/química , Proteínas de Transporte de Ácidos Grasos/genética , Variación Genética , Genoma Bacteriano , Haemophilus influenzae/clasificación , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Mutación , Recombinación Genética , Análisis de Secuencia de ADN , Esputo/microbiología , Secuenciación Completa del Genoma
11.
Sci Rep ; 8(1): 6872, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29720703

RESUMEN

Airway infection by nontypeable Haemophilus influenzae (NTHi) associates to chronic obstructive pulmonary disease (COPD) exacerbation and asthma neutrophilic airway inflammation. Lipids are key inflammatory mediators in these disease conditions and consequently, NTHi may encounter free fatty acids during airway persistence. However, molecular information on the interplay NTHi-free fatty acids is limited, and we lack evidence on the importance of such interaction to infection. Maintenance of the outer membrane lipid asymmetry may play an essential role in NTHi barrier function and interaction with hydrophobic molecules. VacJ/MlaA-MlaBCDEF prevents phospholipid accumulation at the bacterial surface, being the only system involved in maintaining membrane asymmetry identified in NTHi. We assessed the relationship among the NTHi VacJ/MlaA outer membrane lipoprotein, bacterial and exogenous fatty acids, and respiratory infection. The vacJ/mlaA gene inactivation increased NTHi fatty acid and phospholipid global content and fatty acyl specific species, which in turn increased bacterial susceptibility to hydrophobic antimicrobials, decreased NTHi epithelial infection, and increased clearance during pulmonary infection in mice with both normal lung function and emphysema, maybe related to their shared lung fatty acid profiles. Altogether, we provide evidence for VacJ/MlaA as a key bacterial factor modulating NTHi survival at the human airway upon exposure to hydrophobic molecules.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidad , Lipoproteínas/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Ácidos Grasos/metabolismo , Femenino , Infecciones por Haemophilus/microbiología , Humanos , Ratones , Mucosa Respiratoria/microbiología
12.
Sci Rep ; 7(1): 12860, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29038519

RESUMEN

The respiratory pathogen nontypeable Haemophilus influenzae (NTHi) is an important cause of acute exacerbation of chronic obstructive pulmonary disease (AECOPD) that requires efficient treatments. A previous screening for host genes differentially expressed upon NTHi infection identified sirtuin-1, which encodes a NAD-dependent deacetylase protective against emphysema and is activated by resveratrol. This polyphenol concomitantly reduces NTHi viability, therefore highlighting its therapeutic potential against NTHi infection at the COPD airway. In this study, resveratrol antimicrobial effect on NTHi was shown to be bacteriostatic and did not induce resistance development in vitro. Analysis of modulatory properties on the NTHi-host airway epithelial interplay showed that resveratrol modulates bacterial invasion but not subcellular location, reduces inflammation without targeting phosphodiesterase 4B gene expression, and dampens ß defensin-2 gene expression in infected cells. Moreover, resveratrol therapeutics against NTHi was evaluated in vivo on mouse respiratory and zebrafish septicemia infection model systems, showing to decrease NTHi viability in a dose-dependent manner and reduce airway inflammation upon infection, and to have a significant bacterial clearing effect without signs of host toxicity, respectively. This study presents resveratrol as a therapeutic of particular translational significance due to the attractiveness of targeting both infection and overactive inflammation at the COPD airway.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/fisiología , Factores Inmunológicos/uso terapéutico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología , Resveratrol/uso terapéutico , Células A549 , Administración Intranasal , Animales , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Células Cultivadas , Farmacorresistencia Bacteriana/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Infecciones por Haemophilus/patología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/crecimiento & desarrollo , Humanos , Factores Inmunológicos/farmacología , Interleucina-8/metabolismo , Ratones , Enfermedad Pulmonar Obstructiva Crónica/patología , Infecciones del Sistema Respiratorio/patología , Resveratrol/administración & dosificación , Resveratrol/farmacología , Pez Cebra , beta-Defensinas/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-28676846

RESUMEN

Antibacterial treatment with cotrimoxazol (TxS), a combination of trimethoprim and sulfamethoxazole, generates resistance by, among others, acquisition of thymidine auxotrophy associated with mutations in the thymidylate synthase gene thyA, which can modify the biology of infection. The opportunistic pathogen non-typeable Haemophilus influenzae (NTHi) is frequently encountered in the lower airways of chronic obstructive pulmonary disease (COPD) patients, and associated with acute exacerbation of COPD symptoms. Increasing resistance of NTHi to TxS limits its suitability as initial antibacterial against COPD exacerbation, although its relationship with thymidine auxotrophy is unknown. In this study, the analysis of 2,542 NTHi isolates recovered at Bellvitge University Hospital (Spain) in the period 2010-2014 revealed 119 strains forming slow-growing colonies on the thymidine low concentration medium Mueller Hinton Fastidious, including one strain isolated from a COPD patient undergoing TxS therapy that was a reversible thymidine auxotroph. To assess the impact of thymidine auxotrophy in the NTHi-host interplay during respiratory infection, thyA mutants were generated in both the clinical isolate NTHi375 and the reference strain RdKW20. Inactivation of the thyA gene increased TxS resistance, but also promoted morphological changes consistent with elongation and impaired bacterial division, which altered H. influenzae self-aggregation, phosphorylcholine level, C3b deposition, and airway epithelial infection patterns. Availability of external thymidine contributed to overcome such auxotrophy and TxS effect, potentially facilitated by the nucleoside transporter nupC. Although, thyA inactivation resulted in bacterial attenuation in a lung infection mouse model, it also rendered a lower clearance upon a TxS challenge in vivo. Thus, our results show that thymidine auxotrophy modulates both the NTHi host airway interplay and antibiotic resistance, which should be considered at the clinical setting for the consequences of TxS administration.


Asunto(s)
Farmacorresistencia Microbiana/efectos de los fármacos , Infecciones por Haemophilus/tratamiento farmacológico , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/metabolismo , Timidilato Sintasa/genética , Células A549 , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Línea Celular Tumoral , ADN Bacteriano , Femenino , Genes Bacterianos/genética , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/patología , Haemophilus influenzae/citología , Haemophilus influenzae/genética , Interacciones Huésped-Patógeno , Humanos , Interleucina-8/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Microscopía Electrónica de Transmisión , Mutación , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/patología , España , Sulfametoxazol/farmacología , Timidina/metabolismo , Trimetoprim/farmacología , Combinación Trimetoprim y Sulfametoxazol/farmacología , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA