Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Anal Chem ; 94(2): 669-677, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34978813

ABSTRACT

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.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Sputum , Anti-Bacterial Agents , Bacteria , Pseudomonas aeruginosa , Spectrum Analysis, Raman/methods , Sputum/microbiology , Staphylococcus aureus/chemistry
3.
NPJ Biofilms Microbiomes ; 6(1): 46, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127897

ABSTRACT

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.


Subject(s)
Biofilms/growth & development , Drug Resistance, Bacterial , Genes, Essential , Pseudomonas aeruginosa/physiology , Biofilms/classification , Conserved Sequence , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genomics , Microfluidic Analytical Techniques , Mutagenesis, Insertional , Phenotype , Proteogenomics , Pseudomonas aeruginosa/classification , Pseudomonas aeruginosa/genetics
6.
J Allergy Clin Immunol ; 145(1): 147-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31254531

ABSTRACT

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.


Subject(s)
Enterotoxins/immunology , Mast Cells , Nasal Polyps , Phagocytosis , Staphylococcus aureus , Adult , Aged , Cell Line , Female , Humans , Male , Mast Cells/immunology , Mast Cells/microbiology , Mast Cells/ultrastructure , Middle Aged , Nasal Polyps/immunology , Nasal Polyps/microbiology , Nasal Polyps/ultrastructure , Prospective Studies , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Tissue Culture Techniques
7.
J Antimicrob Chemother ; 75(1): 117-125, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31682251

ABSTRACT

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.


Subject(s)
Biofilms/drug effects , Cephalosporins/pharmacology , Cystic Fibrosis/microbiology , Nitric Oxide Donors/pharmacology , Prodrugs/pharmacology , Pseudomonas aeruginosa/drug effects , Adolescent , Anti-Bacterial Agents/pharmacology , Drug Synergism , Humans , Microbial Sensitivity Tests , Middle Aged , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Young Adult
8.
J Clin Pathol ; 71(6): 554-558, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29449345

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bioengineering , Biofilms/drug effects , Haemophilus influenzae/drug effects , Honey , Amoxicillin-Potassium Clavulanate Combination/pharmacology , Anti-Bacterial Agents/metabolism , Biofilms/growth & development , Child, Preschool , Dose-Response Relationship, Drug , Haemophilus influenzae/growth & development , Haemophilus influenzae/metabolism , Humans , Hydrogen Peroxide/metabolism , Microbial Viability/drug effects
9.
Nat Rev Microbiol ; 15(12): 740-755, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28944770

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/classification , Bacterial Physiological Phenomena , Biofilms/growth & development , Drug Resistance, Bacterial
10.
Eur Respir J ; 50(3)2017 09.
Article in English | MEDLINE | ID: mdl-28890436

ABSTRACT

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.


Subject(s)
Epithelial Cells/microbiology , Haemophilus Infections/physiopathology , Kartagener Syndrome/microbiology , Nitric Oxide/pharmacology , Adolescent , Adult , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion , Bacterial Proteins/metabolism , Biofilms/growth & development , Case-Control Studies , Child , Child, Preschool , Cytokines/metabolism , Female , Haemophilus influenzae/pathogenicity , Haemophilus influenzae/physiology , Humans , Kartagener Syndrome/physiopathology , Male , Middle Aged , Primary Cell Culture , Young Adult
11.
Mol Ther ; 25(9): 2104-2116, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28750737

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biofilms/drug effects , Cystic Fibrosis/complications , Nitric Oxide/administration & dosage , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Adolescent , Adult , Bacterial Load , Dose-Response Relationship, Drug , Humans , Middle Aged , Nitric Oxide/metabolism , Pseudomonas Infections/blood , Randomized Controlled Trials as Topic , Sputum/microbiology , Time Factors , Young Adult
12.
Microbiology (Reading) ; 163(7): 1093-1104, 2017 07.
Article in English | MEDLINE | ID: mdl-28699879

ABSTRACT

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.


Subject(s)
Biofilms , Haemophilus Infections/microbiology , Haemophilus influenzae/growth & development , Haemophilus influenzae/metabolism , Methionine/metabolism , Peptidoglycan/biosynthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Haemophilus influenzae/drug effects , Haemophilus influenzae/genetics , Humans
13.
Thorax ; 72(10): 946-949, 2017 10.
Article in English | MEDLINE | ID: mdl-28159773

ABSTRACT

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.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Antirheumatic Agents/pharmacology , Infliximab/pharmacology , Interferon-gamma Release Tests , Latent Tuberculosis/diagnosis , Adult , Aged , Dexamethasone/pharmacology , Female , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Middle Aged , Tumor Necrosis Factor-alpha/metabolism
14.
J Glob Antimicrob Resist ; 8: 186-191, 2017 03.
Article in English | MEDLINE | ID: mdl-28213334

ABSTRACT

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.


Subject(s)
Anti-Infective Agents, Local/therapeutic use , Bacteria/drug effects , Bacterial Infections/drug therapy , Biofilms/drug effects , Reactive Oxygen Species/therapeutic use , Wound Infection/drug therapy , Administration, Topical , Animals , Drug Evaluation, Preclinical , Fungi/drug effects , Humans
15.
Nitric Oxide ; 65: 43-49, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28235635

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azo Compounds/pharmacology , Biofilms/drug effects , Cephalosporins/pharmacology , Nitric Oxide Donors/pharmacology , Prodrugs/pharmacology , Streptococcus pneumoniae/drug effects , Amoxicillin/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Azithromycin/pharmacology , Azo Compounds/chemistry , Cephalosporins/chemistry , Nitric Oxide/analysis , Nitric Oxide Donors/chemistry , Penicillinase/chemistry , Plankton/microbiology , Prodrugs/chemistry
16.
Article in English | MEDLINE | ID: mdl-27919896

ABSTRACT

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.


Subject(s)
Azo Compounds/pharmacology , Biofilms/drug effects , Cephalosporins/pharmacology , Haemophilus influenzae/drug effects , Nitric Oxide Donors/pharmacology , Prodrugs/pharmacology , Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Chromatography, Liquid , Drug Resistance, Bacterial , Mass Spectrometry , Microbial Sensitivity Tests , Nitrogen Oxides/metabolism , Proteomics , beta-Lactamases/metabolism
17.
Genome Biol Evol ; 8(5): 1316-26, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27190203

ABSTRACT

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.


Subject(s)
Biofilms/growth & development , Biological Evolution , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/genetics , Bacterial Proteins/genetics , Genome, Bacterial , Humans , Mutation/genetics , Streptococcus pneumoniae/isolation & purification
18.
Antimicrob Agents Chemother ; 60(4): 2456-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26856845

ABSTRACT

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.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination/pharmacology , Biofilms/drug effects , Gene Expression Regulation, Bacterial/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Streptococcus pneumoniae/drug effects , Adenoids/drug effects , Adenoids/microbiology , Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Child , Child, Preschool , Drug Resistance, Bacterial/drug effects , Drug Synergism , Drug Therapy, Combination , Humans , Hydrazines/chemistry , Hydrazines/pharmacology , Nitrates/chemistry , Nitrates/pharmacology , Nitric Oxide/chemistry , Nitric Oxide Donors/chemistry , Nitroprusside/chemistry , Nitroprusside/pharmacology , Otitis Media/drug therapy , Otitis Media/microbiology , Otitis Media/pathology , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Protein Biosynthesis , Sodium Nitrite/chemistry , Sodium Nitrite/pharmacology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Transcription, Genetic/drug effects
20.
J Infect ; 69 Suppl 1: S47-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25240819

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Biofilms , Anti-Bacterial Agents/pharmacology , Bacterial Infections/diagnosis , Biofilms/drug effects , Biofilms/growth & development , Drug Resistance, Bacterial , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...