Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 94(2): 669-677, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34978813

RESUMEN

The current methods for diagnosis of acute and chronic infections are complex and skill-intensive. For complex clinical biofilm infections, it can take days from collecting and processing a patient's sample to achieving a result. These aspects place a significant burden on healthcare providers, delay treatment, and can lead to adverse patient outcomes. We report the development and application of a novel multi-excitation Raman spectroscopy-based methodology for the label-free and non-invasive detection of microbial pathogens that can be used with unprocessed clinical samples directly and provide rapid data to inform diagnosis by a medical professional. The method relies on the differential excitation of non-resonant and resonant molecular components in bacterial cells to enhance the molecular finger-printing capability to obtain strain-level distinction in bacterial species. Here, we use this strategy to detect and characterize the respiratory pathogens Pseudomonas aeruginosa and Staphylococcus aureus as typical infectious agents associated with cystic fibrosis. Planktonic specimens were analyzed both in isolation and in artificial sputum media. The resonance Raman components, excited at different wavelengths, were characterized as carotenoids and porphyrins. By combining the more informative multi-excitation Raman spectra with multivariate analysis (support vector machine) the accuracy was found to be 99.75% for both species (across all strains), including 100% accuracy for drug-sensitive and drug-resistant S. aureus. The results demonstrate that our methodology based on multi-excitation Raman spectroscopy can underpin the development of a powerful platform for the rapid and reagentless detection of clinical pathogens to support diagnosis by a medical expert, in this case relevant to cystic fibrosis. Such a platform could provide translatable diagnostic solutions in a variety of disease areas and also be utilized for the rapid detection of anti-microbial resistance.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Esputo , Antibacterianos , Bacterias , Pseudomonas aeruginosa , Espectrometría Raman/métodos , Esputo/microbiología , Staphylococcus aureus/química
2.
J Allergy Clin Immunol ; 145(1): 147-159, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31254531

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) with nasal polyps is a common chronic condition. The exact cause of nasal polyps remains unknown. Recently, we made the novel observation of intracellular localization of Staphylococcus aureus within mast cells in nasal polyps. OBJECTIVE: This follow-up study aimed to further characterize interactions between S aureus and mast cells in this setting and elucidate potential internalization mechanisms with particular emphasis on the role of staphylococcal enterotoxin B (SEB). METHODS: A prospective study was performed using an explant tissue model with ex vivo inferior turbinate mucosa obtained from patients with chronic rhinosinusitis with nasal polyps (n = 7) and patients without CRS (n = 5). Immunohistochemistry was used to characterize S aureus uptake into mast cells and investigate the effects of SEB on this process. An in vitro cell-culture model was used to investigate mast cell-S aureus interactions by using a combination of fluorescent in situ hybridization, confocal laser scanning microscopy, scanning electron microscopy, transmission electron microscopy, and proliferation assays. RESULTS: S aureus was captured by extracellular traps and entered mast cells through phagocytosis. Proliferating intracellular S aureus led to the expansion and eventual rupture of mast cells, resulting in release of viable S aureus into the extracellular space. The presence of SEB appeared to promote internalization of S aureus into mast cells. CONCLUSION: This study provides new insights into the interactions between S aureus and mast cells, including the internalization process, and demonstrates a prominent role for SEB in promoting uptake of the bacteria into these cells.


Asunto(s)
Enterotoxinas/inmunología , Mastocitos , Pólipos Nasales , Fagocitosis , Staphylococcus aureus , Adulto , Anciano , Línea Celular , Femenino , Humanos , Masculino , Mastocitos/inmunología , Mastocitos/microbiología , Mastocitos/ultraestructura , Persona de Mediana Edad , Pólipos Nasales/inmunología , Pólipos Nasales/microbiología , Pólipos Nasales/ultraestructura , Estudios Prospectivos , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Técnicas de Cultivo de Tejidos
3.
J Antimicrob Chemother ; 75(1): 117-125, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682251

RESUMEN

OBJECTIVES: The cephalosporin nitric oxide (NO)-donor prodrug DEA-C3D ('DiEthylAmin-Cephalosporin-3'-Diazeniumdiolate') has been shown to initiate the dispersal of biofilms formed by the Pseudomonas aeruginosa laboratory strain PAO1. In this study, we investigated whether DEA-C3D disperses biofilms formed by clinical cystic fibrosis (CF) isolates of P. aeruginosa and its effect in combination with two antipseudomonal antibiotics, tobramycin and colistin, in vitro. METHODS: ß-Lactamase-triggered release of NO from DEA-C3D was confirmed using a gas-phase chemiluminescence detector. MICs for P. aeruginosa clinical isolates were determined using the broth microdilution method. A crystal violet staining technique and confocal laser scanning microscopy were used to evaluate the effects of DEA-C3D on P. aeruginosa biofilms alone and in combination with tobramycin and colistin. RESULTS: DEA-C3D was confirmed to selectively release NO in response to contact with bacterial ß-lactamase. Despite lacking direct, cephalosporin/ß-lactam-based antibacterial activity, DEA-C3D was able to disperse biofilms formed by three P. aeruginosa clinical isolates. Confocal microscopy revealed that DEA-C3D in combination with tobramycin produces similar reductions in biofilm to DEA-C3D alone, whereas the combination with colistin causes near complete eradication of P. aeruginosa biofilms in vitro. CONCLUSIONS: DEA-C3D is effective in dispersing biofilms formed by multiple clinical isolates of P. aeruginosa and could hold promise as a new adjunctive therapy to patients with CF.


Asunto(s)
Biopelículas/efectos de los fármacos , Cefalosporinas/farmacología , Fibrosis Quística/microbiología , Donantes de Óxido Nítrico/farmacología , Profármacos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Adolescente , Antibacterianos/farmacología , Sinergismo Farmacológico , Humanos , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Adulto Joven
4.
Mol Ther ; 25(9): 2104-2116, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28750737

RESUMEN

Despite aggressive antibiotic therapy, bronchopulmonary colonization by Pseudomonas aeruginosa causes persistent morbidity and mortality in cystic fibrosis (CF). Chronic P. aeruginosa infection in the CF lung is associated with structured, antibiotic-tolerant bacterial aggregates known as biofilms. We have demonstrated the effects of non-bactericidal, low-dose nitric oxide (NO), a signaling molecule that induces biofilm dispersal, as a novel adjunctive therapy for P. aeruginosa biofilm infection in CF in an ex vivo model and a proof-of-concept double-blind clinical trial. Submicromolar NO concentrations alone caused disruption of biofilms within ex vivo CF sputum and a statistically significant decrease in ex vivo biofilm tolerance to tobramycin and tobramycin combined with ceftazidime. In the 12-patient randomized clinical trial, 10 ppm NO inhalation caused significant reduction in P. aeruginosa biofilm aggregates compared with placebo across 7 days of treatment. Our results suggest a benefit of using low-dose NO as adjunctive therapy to enhance the efficacy of antibiotics used to treat acute P. aeruginosa exacerbations in CF. Strategies to induce the disruption of biofilms have the potential to overcome biofilm-associated antibiotic tolerance in CF and other biofilm-related diseases.


Asunto(s)
Antibacterianos/administración & dosificación , Biopelículas/efectos de los fármacos , Fibrosis Quística/complicaciones , Óxido Nítrico/administración & dosificación , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Adolescente , Adulto , Carga Bacteriana , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Infecciones por Pseudomonas/sangre , Ensayos Clínicos Controlados Aleatorios como Asunto , Esputo/microbiología , Factores de Tiempo , Adulto Joven
5.
Artículo en Inglés | MEDLINE | ID: mdl-27919896

RESUMEN

PYRRO-C3D is a cephalosporin-3-diazeniumdiolate nitric oxide (NO) donor prodrug designed to selectively deliver NO to bacterial infection sites. The objective of this study was to assess the activity of PYRRO-C3D against nontypeable Haemophilus influenzae (NTHi) biofilms and examine the role of NO in reducing biofilm-associated antibiotic tolerance. The activity of PYRRO-C3D on in vitro NTHi biofilms was assessed through CFU enumeration and confocal microscopy. NO release measurements were performed using an ISO-NO probe. NTHi biofilms grown on primary ciliated respiratory epithelia at an air-liquid interface were used to investigate the effects of PYRRO-C3D in the presence of host tissue. Label-free liquid chromatography-mass spectrometry (LC/MS) proteomic analyses were performed to identify differentially expressed proteins following NO treatment. PYRRO-C3D specifically released NO in the presence of NTHi, while no evidence of spontaneous NO release was observed when the compound was exposed to primary epithelial cells. NTHi lacking ß-lactamase activity failed to trigger NO release. Treatment significantly increased the susceptibility of in vitro NTHi biofilms to azithromycin, causing a log fold reduction (10-fold reduction or 1-log-unit reduction) in viability (P < 0.05) relative to azithromycin alone. The response was more pronounced for biofilms grown on primary respiratory epithelia, where a 2-log-unit reduction was observed (P < 0.01). Label-free proteomics showed that NO increased expression of 16 proteins involved in metabolic and transcriptional/translational functions. NO release from PYRRO-C3D enhances the efficacy of azithromycin against NTHi biofilms, putatively via modulation of NTHi metabolic activity. Adjunctive therapy with NO mediated through PYRRO-C3D represents a promising approach for reducing biofilm-associated antibiotic tolerance.


Asunto(s)
Compuestos Azo/farmacología , Biopelículas/efectos de los fármacos , Cefalosporinas/farmacología , Haemophilus influenzae/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Profármacos/farmacología , Antibacterianos/farmacología , Azitromicina/farmacología , Cromatografía Liquida , Farmacorresistencia Bacteriana , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Óxidos de Nitrógeno/metabolismo , Proteómica , beta-Lactamasas/metabolismo
6.
Thorax ; 72(10): 946-949, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28159773

RESUMEN

The impact of immunosuppression on interferon-γ release assays and novel cytokine biomarkers of TB infection, mycobacteria-specific IL-2, IP-10 and TNF-α responses was investigated in an ex vivo model. Cytokine responses in standard QuantiFERON-TB Gold in-Tube (QFT-GIT) assays were compared with duplicate assays containing dexamethasone or infliximab. Dexamethasone converted QFT-GIT results from positive to negative in 30% of participants. Antigen-stimulated interferon-γ, IL-2 and TNF-α responses were markedly reduced, but IP-10 responses were preserved. Infliximab caused QFT-GIT result conversion in up to 30% of participants and substantial reductions in all cytokine responses. Therefore, corticosteroids and anti-TNF-α agents significantly impair interferon-γ release assay performance. IP-10 may be a more robust TB biomarker than interferon-γ in patients receiving corticosteroids.


Asunto(s)
Corticoesteroides/farmacología , Antirreumáticos/farmacología , Infliximab/farmacología , Ensayos de Liberación de Interferón gamma , Tuberculosis Latente/diagnóstico , Adulto , Anciano , Dexametasona/farmacología , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Masculino , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/metabolismo
7.
Microbiology (Reading) ; 163(7): 1093-1104, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28699879

RESUMEN

Non-typeable Haemophilus influenzae (NTHi) is an opportunistic pathogen that plays a major role in a number of respiratory tract infections, including otitis media, cystic fibrosis and chronic obstructive pulmonary disease. Biofilm formation has been implicated in both NTHi colonization and disease, and is responsible for the increased tolerance of this pathogen towards antibiotic treatment. Targeting metabolic pathways that are important in NTHi biofilm formation represents a potential strategy to combat this antibiotic recalcitrance. A previous investigation demonstrated increased expression of a putative d-methionine uptake protein following exposure of NTHi biofilms to the ubiquitous signalling molecule, nitric oxide. We therefore hypothesized that treatment with exogenous d-methionine would impact on NTHi biofilm formation and increase antibiotic sensitivity. Treatment of NTHi during the process of biofilm formation resulted in a reduction in biofilm viability, increased biomass, changes in the overall biofilm architecture and the adoption of an amorphous cellular morphology. Quantitative proteomic analyses identified 124 proteins that were differentially expressed following d-methionine treatment, of which 51 (41 %) were involved in metabolic and transport processes. Nine proteins involved in peptidoglycan synthesis and cell division showed significantly increased expression. Furthermore, d-methionine treatment augmented the efficacy of azithromycin treatment and highlighted the potential of d-methionine as an adjunctive therapeutic approach for NTHi biofilm-associated infections.


Asunto(s)
Biopelículas , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/metabolismo , Metionina/metabolismo , Peptidoglicano/biosíntesis , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/genética , Humanos
8.
Eur Respir J ; 50(3)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28890436

RESUMEN

Non-typeable Haemophilus influenzae (NTHi) is the most common pathogen in primary ciliary dyskinesia (PCD) patients. We hypothesised that abnormal ciliary motility and low airway nitric oxide (NO) levels on airway epithelial cells from PCD patients might be permissive for NTHi colonisation and biofilm development.We used a primary epithelial cell co-culture model to investigate NTHi infection. Primary airway epithelial cells from PCD and non-PCD patients were differentiated to ciliation using an air-liquid interface culture and then co-cultured with NTHi.NTHi adherence was greater on PCD epithelial cells compared to non-PCD cells (p<0.05) and the distribution of NTHi on PCD epithelium showed more aggregated NTHi in biofilms (p<0.001). Apart from defective ciliary motility, PCD cells did not significantly differ from non-PCD epithelial cells in the degree of ciliation and epithelial integrity or in cytokine, LL-37 and NO production. Treatment of PCD epithelia using exogenous NO and antibiotic significantly reduced NTHi viability in biofilms compared with antibiotic treatment alone.Impaired ciliary function was the primary defect in PCD airway epithelium underlying susceptibility to NTHi biofilm development compared with non-PCD epithelium. Although NO responses were similar, use of targeted NO with antibiotics enhanced killing of NTHi in biofilms, suggesting a novel therapeutic approach.


Asunto(s)
Células Epiteliales/microbiología , Infecciones por Haemophilus/fisiopatología , Síndrome de Kartagener/microbiología , Óxido Nítrico/farmacología , Adolescente , Adulto , Antibacterianos/farmacología , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Estudios de Casos y Controles , Niño , Preescolar , Citocinas/metabolismo , Femenino , Haemophilus influenzae/patogenicidad , Haemophilus influenzae/fisiología , Humanos , Síndrome de Kartagener/fisiopatología , Masculino , Persona de Mediana Edad , Cultivo Primario de Células , Adulto Joven
9.
Nitric Oxide ; 65: 43-49, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28235635

RESUMEN

Bacterial biofilms show high tolerance towards antibiotics and are a significant problem in clinical settings where they are a primary cause of chronic infections. Novel therapeutic strategies are needed to improve anti-biofilm efficacy and support reduction in antibiotic use. Treatment with exogenous nitric oxide (NO) has been shown to modulate bacterial signaling and metabolic processes that render biofilms more susceptible to antibiotics. We previously reported on cephalosporin-3'-diazeniumdiolates (C3Ds) as NO-donor prodrugs designed to selectively deliver NO to bacterial infection sites following reaction with ß-lactamases. With structures based on cephalosporins, C3Ds could, in principal, also be triggered to release NO following ß-lactam cleavage mediated by transpeptidases/penicillin-binding proteins (PBPs), the antibacterial target of cephalosporin antibiotics. Transpeptidase-reactive C3Ds could potentially show both NO-mediated anti-biofilm properties and intrinsic (ß-lactam-mediated) antibacterial effects. This dual-activity concept was explored using Streptococcus pneumoniae, a species that lacks ß-lactamases but relies on transpeptidases for cell-wall synthesis. Treatment with PYRRO-C3D (a representative C3D containing the diazeniumdiolate NO donor PYRRO-NO) was found to significantly reduce viability of planktonic and biofilm pneumococci, demonstrating that C3Ds can elicit direct, cephalosporin-like antibacterial activity in the absence of ß-lactamases. While NO release from PYRRO-C3D in the presence of pneumococci was confirmed, the anti-pneumococcal action of the compound was shown to arise exclusively from the ß-lactam component and not through NO-mediated effects. The compound showed similar potency to amoxicillin against S. pneumoniae biofilms and greater efficacy than azithromycin, highlighting the potential of C3Ds as new agents for treating pneumococcal infections.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Compuestos Azo/farmacología , Biopelículas/efectos de los fármacos , Cefalosporinas/farmacología , Donantes de Óxido Nítrico/farmacología , Profármacos/farmacología , Streptococcus pneumoniae/efectos de los fármacos , Amoxicilina/farmacología , Antiinflamatorios no Esteroideos/química , Azitromicina/farmacología , Compuestos Azo/química , Cefalosporinas/química , Óxido Nítrico/análisis , Donantes de Óxido Nítrico/química , Penicilinasa/química , Plancton/microbiología , Profármacos/química
10.
Antimicrob Agents Chemother ; 60(4): 2456-66, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26856845

RESUMEN

Streptococcus pneumoniaeis one of the key pathogens responsible for otitis media (OM), the most common infection in children and the largest cause of childhood antibiotic prescription. Novel therapeutic strategies that reduce the overall antibiotic consumption due to OM are required because, although widespread pneumococcal conjugate immunization has controlled invasive pneumococcal disease, overall OM incidence has not decreased. Biofilm formation represents an important phenotype contributing to the antibiotic tolerance and persistence ofS. pneumoniaein chronic or recurrent OM. We investigated the treatment of pneumococcal biofilms with nitric oxide (NO), an endogenous signaling molecule and therapeutic agent that has been demonstrated to trigger biofilm dispersal in other bacterial species. We hypothesized that addition of low concentrations of NO to pneumococcal biofilms would improve antibiotic efficacy and that higher concentrations exert direct antibacterial effects. Unlike in many other bacterial species, low concentrations of NO did not result inS. pneumoniaebiofilm dispersal. Instead, treatment of bothin vitrobiofilms andex vivoadenoid tissue samples (a reservoir forS. pneumoniaebiofilms) with low concentrations of NO enhanced pneumococcal killing when combined with amoxicillin-clavulanic acid, an antibiotic commonly used to treat chronic OM. Quantitative proteomic analysis using iTRAQ (isobaric tag for relative and absolute quantitation) identified 13 proteins that were differentially expressed following low-concentration NO treatment, 85% of which function in metabolism or translation. Treatment with low-concentration NO, therefore, appears to modulate pneumococcal metabolism and may represent a novel therapeutic approach to reduce antibiotic tolerance in pneumococcal biofilms.


Asunto(s)
Combinación Amoxicilina-Clavulanato de Potasio/farmacología , Biopelículas/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/farmacología , Streptococcus pneumoniae/efectos de los fármacos , Tonsila Faríngea/efectos de los fármacos , Tonsila Faríngea/microbiología , Antibacterianos/farmacología , Biopelículas/crecimiento & desarrollo , Niño , Preescolar , Farmacorresistencia Bacteriana/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Hidrazinas/química , Hidrazinas/farmacología , Nitratos/química , Nitratos/farmacología , Óxido Nítrico/química , Donantes de Óxido Nítrico/química , Nitroprusiato/química , Nitroprusiato/farmacología , Otitis Media/tratamiento farmacológico , Otitis Media/microbiología , Otitis Media/patología , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Biosíntesis de Proteínas , Nitrito de Sodio/química , Nitrito de Sodio/farmacología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Transcripción Genética/efectos de los fármacos
12.
J Bacteriol ; 193(9): 2268-75, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21378197

RESUMEN

Spores of Bacillus subtilis require the GerAA, GerAB, and GerAC receptor proteins for L-alanine-induced germination. Mutations in gerAA, both random and site directed, result in phenotypes that identify amino acid residues important for receptor function in broad terms. They highlight the functional importance of two regions in the central, integral membrane domain of GerAA. A P324S substitution in the first residue of a conserved PFPP motif results in a 10-fold increase in a spore's sensitivity to alanine; a P326S change results in the release of phase-dark spores, in which the receptor may be in an "activated" or "quasigerminated" state. Substitutions in residues 398 to 400, in a short loop between the last two likely membrane-spanning helices of this central domain, all affect the germination response, with the G398S substitution causing a temperature-sensitive defect. In others, there are wider effects on the receptor: if alanine is substituted for conserved residue N146, H304, or E330, a severe defect in L-alanine germination results. This correlates with the absence of GerAC, suggesting that the assembly or stability of the entire receptor complex has been compromised by the defect in GerAA. In contrast, severely germination-defective mutants such as E129K, L373F, S400F, and M409N mutants retain GerAC at normal levels, suggesting more local and specific effects on the function of GerAA itself. Further interpretation will depend on progress in structural analysis of the receptor proteins.


Asunto(s)
Alanina/metabolismo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Esporas Bacterianas/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Secuencia Conservada , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Anotación de Secuencia Molecular , Fenotipo , Mutación Puntual
13.
NPJ Biofilms Microbiomes ; 6(1): 46, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33127897

RESUMEN

Pseudomonas aeruginosa MPAO1 is the parental strain of the widely utilized transposon mutant collection for this important clinical pathogen. Here, we validate a model system to identify genes involved in biofilm growth and biofilm-associated antibiotic resistance. Our model employs a genomics-driven workflow to assemble the complete MPAO1 genome, identify unique and conserved genes by comparative genomics with the PAO1 reference strain and genes missed within existing assemblies by proteogenomics. Among over 200 unique MPAO1 genes, we identified six general essential genes that were overlooked when mapping public Tn-seq data sets against PAO1, including an antitoxin. Genomic data were integrated with phenotypic data from an experimental workflow using a user-friendly, soft lithography-based microfluidic flow chamber for biofilm growth and a screen with the Tn-mutant library in microtiter plates. The screen identified hitherto unknown genes involved in biofilm growth and antibiotic resistance. Experiments conducted with the flow chamber across three laboratories delivered reproducible data on P. aeruginosa biofilms and validated the function of both known genes and genes identified in the Tn-mutant screens. Differential protein abundance data from planktonic cells versus biofilm confirmed the upregulation of candidates known to affect biofilm formation, of structural and secreted proteins of type VI secretion systems, and provided proteogenomic evidence for some missed MPAO1 genes. This integrated, broadly applicable model promises to improve the mechanistic understanding of biofilm formation, antimicrobial tolerance, and resistance evolution in biofilms.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana , Genes Esenciales , Pseudomonas aeruginosa/fisiología , Biopelículas/clasificación , Secuencia Conservada , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Genómica , Técnicas Analíticas Microfluídicas , Mutagénesis Insercional , Fenotipo , Proteogenómica , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/genética
14.
J Clin Pathol ; 71(6): 554-558, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29449345

RESUMEN

The opportunistic pathogen non-typeable Haemophilus influenzae (NTHi) plays an important role in many chronic respiratory diseases including otitis media, chronic rhinosinusitis, cystic fibrosis and chronic obstructive pulmonary disease. Biofilm formation has been implicated in NTHi colonisation, persistence of infection and recalcitrance towards antimicrobials. There is therefore a pressing need for the development of novel treatment strategies that are effective against NTHi biofilm-associated diseases. SurgihoneyRO is a honey-based product that has been bioengineered to enable the slow release of H2O2, a reactive oxygen species to which H. influenzae is susceptible. Treatment of established NTHi biofilms with SurgihoneyRO significantly reduced biofilm viability through enhanced H2O2 production and was shown to be more effective than the conventional antibiotic co-amoxiclav.


Asunto(s)
Antibacterianos/farmacología , Bioingeniería , Biopelículas/efectos de los fármacos , Haemophilus influenzae/efectos de los fármacos , Miel , Combinación Amoxicilina-Clavulanato de Potasio/farmacología , Antibacterianos/metabolismo , Biopelículas/crecimiento & desarrollo , Preescolar , Relación Dosis-Respuesta a Droga , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Viabilidad Microbiana/efectos de los fármacos
15.
Nat Rev Microbiol ; 15(12): 740-755, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28944770

RESUMEN

Biofilm formation is a key virulence factor for a wide range of microorganisms that cause chronic infections. The multifactorial nature of biofilm development and drug tolerance imposes great challenges for the use of conventional antimicrobials and indicates the need for multi-targeted or combinatorial therapies. In this Review, we focus on current therapeutic strategies and those under development that target vital structural and functional traits of microbial biofilms and drug tolerance mechanisms, including the extracellular matrix and dormant cells. We emphasize strategies that are supported by in vivo or ex vivo studies, highlight emerging biofilm-targeting technologies and provide a rationale for multi-targeted therapies aimed at disrupting the complex biofilm microenvironment.


Asunto(s)
Antibacterianos/farmacología , Bacterias/clasificación , Fenómenos Fisiológicos Bacterianos , Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana
16.
J Glob Antimicrob Resist ; 8: 186-191, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28213334

RESUMEN

Reactive oxygen species (ROS) is a novel therapeutic strategy for topical or local application to wounds, mucosa or internal structures where there may be heavy bacterial bioburden with biofilm and chronic inflammation. Bacterial biofilms are a significant problem in clinical settings owing to their increased tolerance towards conventionally prescribed antibiotics and their propensity for selection of further antibacterial resistance. There is therefore a pressing need for the development of alternative therapeutic strategies that can improve antibiotic efficacy towards biofilms. ROS has been successful in treating chronic wounds and in clearing multidrug-resistant organisms, including methicillin-resistant Staphylococcus aureus (MRSA), and carbapenemase-producing isolates from wounds and vascular line sites. There is significant antifungal activity of ROS against planktonic and biofilm forms. Nebulised ROS has been evaluated in limited subjects to assess reductions in bioburden in chronically colonised respiratory tracts. The antibiofilm activity of ROS could have great implications for the treatment of a variety of persistent respiratory conditions. Use of ROS on internal prosthetic devices shows promise. A variety of novel delivery mechanisms are being developed to apply ROS activity to different anatomical sites.


Asunto(s)
Antiinfecciosos Locales/uso terapéutico , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Biopelículas/efectos de los fármacos , Especies Reactivas de Oxígeno/uso terapéutico , Infección de Heridas/tratamiento farmacológico , Administración Tópica , Animales , Evaluación Preclínica de Medicamentos , Hongos/efectos de los fármacos , Humanos
17.
Genome Biol Evol ; 8(5): 1316-26, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27190203

RESUMEN

Streptococcus pneumoniae is a commensal human pathogen and the causative agent of various invasive and noninvasive diseases. Carriage of the pneumococcus in the nasopharynx is thought to be mediated by biofilm formation, an environment where isogenic populations frequently give rise to morphological colony variants, including small colony variant (SCV) phenotypes. We employed metabolic characterization and whole-genome sequencing of biofilm-derived S. pneumoniae serotype 22F pneumococcal SCVs to investigate diversification during biofilm formation. Phenotypic profiling revealed that SCVs exhibit reduced growth rates, reduced capsule expression, altered metabolic profiles, and increased biofilm formation compared to the ancestral strain. Whole-genome sequencing of 12 SCVs from independent biofilm experiments revealed that all SCVs studied had mutations within the DNA-directed RNA polymerase delta subunit (RpoE). Mutations included four large-scale deletions ranging from 51 to 264 bp, one insertion resulting in a coding frameshift, and seven nonsense single-nucleotide substitutions that result in a truncated gene product. This work links mutations in the rpoE gene to SCV formation and enhanced biofilm development in S. pneumoniae and therefore may have important implications for colonization, carriage, and persistence of the organism. Furthermore, recurrent mutation of the pneumococcal rpoE gene presents an unprecedented level of parallel evolution in pneumococcal biofilm development.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Evolución Biológica , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/genética , Proteínas Bacterianas/genética , Genoma Bacteriano , Humanos , Mutación/genética , Streptococcus pneumoniae/aislamiento & purificación
20.
J Infect ; 69 Suppl 1: S47-52, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25240819

RESUMEN

Bacteria causing chronic infections predominately grow as surface-attached, sessile communities known as biofilms. Biofilm-related infections including cystic fibrosis lung infection, chronic and recurrent otitis media, chronic wounds and implant- and catheter-associated infections, are a significant cause of morbidity and mortality and financial cost. Chronic biofilm-based infections are recalcitrant to conventional antibiotic therapy and are often unperturbed by host immune responses such as phagocytosis, despite a sustained presence of host inflammation. The diagnosis of clinically important biofilm infections is often difficult as Koch's postulates are rarely met. If treatment is required, surgical removal of the infected implant, or debridement of wound or bone, is the most efficient means of eradicating a clinically significant biofilm. New approaches to treatment are under investigation.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Fenómenos Fisiológicos Bacterianos , Biopelículas , Antibacterianos/farmacología , Infecciones Bacterianas/diagnóstico , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA