Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Antibiotics (Basel) ; 12(9)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37760729

ABSTRACT

Antifungal agents are widely used to specifically eliminate infections by fungal pathogens. However, the specificity of antifungal agents has been challenged by a few studies demonstrating antibacterial inhibitory effects against Mycobacteria and Streptomyces species. Here, we evaluated for the first time the potential effect of fluconazole, the most clinically used antifungal agent, on a human oral microbiota biofilm model. The results showed that biofilm viability on blood and mitis salivarius agar media was increased over time in the presence of fluconazole at clinically relevant concentrations, despite a reduction in biomass. Targeted PCR revealed a higher abundance of Veillonella atypica, Veillonella dispar, and Lactobacillus spp. in the fluconazole-treated samples compared to the control, while Fusobacterium nucleatum was reduced and Streptococcus spp were not significantly affected. Further, we tested the potential impact of fluconazole using single-species models. Our results, using Streptococcus mutans and Streptococcus mitis luciferase reporters, showed that S. mutans planktonic growth was not significantly affected by fluconazole, whereas for S. mitis, planktonic growth, but not biofilm viability, was inhibited at the highest concentration. Fluconazole's effects on S. mitis biofilm biomass were concentration and time dependent. Exposure for 48 h to the highest concentration of fluconazole was associated with S. mitis biofilms with the most increased biomass. Potential growth inhibitory effects were further tested using four non-streptococcal species. Among these, the planktonic growth of both Escherichia coli and Granulicatella adiacens was inhibited by fluconazole. The data indicate bacterial responses to fluconazole that extend to a broader range of bacterial species than previously anticipated from the literature, with the potential to disturb biofilm communities.

2.
Antibiotics (Basel) ; 12(2)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36830333

ABSTRACT

Emerging evidence suggests differential effects of therapeutic antibiotics on infant T cell responses to pathogens. In this study, we explored the impact of the treatment of mouse infants with amoxicillin and the human milk-derived antimicrobial HAMLET (human alpha-lactalbumin made lethal to tumor cells) on T cell responses to Streptococcus pneumoniae. Lung cells and splenocytes were isolated from the infant mice subjected to intranasal administration of amoxicillin, HAMLET, or a combination of HAMLET and amoxicillin, and cultured with S. pneumoniae to measure T cell responses. After in-vitro stimulation with S. pneumoniae, lung cells from amoxicillin- or amoxicillin plus HAMLET-treated mice produced lower levels of Th17 (IL-17A), but not Th1 (IFN-γ), cytokine than mice receiving HAMLET or PBS. IL-17A/IFN-γ cytokine levels produced by the stimulated splenocytes, on the other hand, revealed no significant difference among treatment groups. Further analysis of T cell cytokine profiles by flow cytometry showed that lung CD4+, but not CD8+, T cells from amoxicillin- or HAMLET plus amoxicillin-treated mice expressed decreased levels of IL-17A compared to those from HAMLET-exposed or control mice. Collectively, these results indicate that exposure of infant mice to amoxicillin, but not HAMLET, may suppress lung Th17 responses to S. pneumoniae.

3.
Gut Microbes ; 15(1): 2157200, 2023.
Article in English | MEDLINE | ID: mdl-36576106

ABSTRACT

The collateral impact of antibiotics on the microbiome has attained increasing attention. However, the ecological consequences of long-term antibiotic exposure on the gut microbiome, including antibiotic resistance, are still limited. Here, we investigated long-term exposure effects to amoxicillin on the human gut microbiome and resistome. Fecal samples were collected from 20 patients receiving 3-months of amoxicillin or placebo treatment as part of a Norwegian multicenter clinical trial on chronic low back pain (AIM study). Samples were collected at baseline, last day of treatment, and 9 months after antibiotic cessation. The abundance and diversity of microbial and resistome composition were characterized using whole shotgun and functional metagenomic sequencing data. While the microbiome profiles of placebo subjects were stable over time, discernible changes in diversity and overall microbiome composition were observed after amoxicillin treatment. In particular, health-associated short-chain fatty acid producing species significantly decreased in proportion. However, these changes were short-lived as the microbiome showed overall recovery 9 months post-treatment. On the other hand, exposure to long-term amoxicillin was associated with an increase in total antimicrobial resistance gene load and diversity of antimicrobial resistance genes, with persistent changes even at 9 months post-treatment. Additionally, beta-lactam resistance was the most affected antibiotic class, suggesting a targeted response to amoxicillin, although changes at the gene level varied across individuals. Overall, our results suggest that the impact of prolonged amoxicillin exposure was more explicit and long-lasting in the fecal resistome than in microbiome composition. Such information is relevant for designing rational administration guidelines for antibiotic therapies.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Amoxicillin/pharmacology , Amoxicillin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Feces
4.
Antimicrob Resist Infect Control ; 11(1): 158, 2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36503570

ABSTRACT

BACKGROUND: The antimicrobial resistance (AMR) crisis is a major global threat and one of its biggest drivers is the overuse of antibiotics in humans. Dentists are responsible for 5-10% antibiotic prescriptions worldwide and recent data suggest that knowledge and prescribing practices need improvement. METHODS: A cross-sectional web-survey was sent to dental students from six universities in Norway, Canada, and Brazil. Topics addressed covered awareness, confidence to prescribe antibiotics, and education needs. Data were presented descriptively and statistical testing was employed to compare group means when applicable. RESULTS: In total, 562 responses were collected across the three countries with a response rate of 28.6%. 'Antibiotic resistance' was among the highest priorities (scale 1-10) with an average of 8.86 (SEM ± 0.05), together with 'Gender inequality' (8.68 ± 0.07) and 'Climate change' (8.68 ± 0.07). Only 28.8% thought that Dentistry was engaged in national/international campaigns promoting awareness on the topic and 8.9% stated to have heard about the 'One Health' concept. Final year dental students showed an average confidence to prescribe antibiotics of 7.59 (± 0.14). Most students demonstrated interest in receiving additional education on all topics listed, with the three most pressing being 'antibiotic prescription for treatment of infections' (82.9%), 'drug interactions' (80.9%), and 'spread of antibiotic resistance' (79.6%). A trend was observed between higher awareness regarding the topic and higher confidence to prescribe. CONCLUSIONS: There is a need to revisit dental education on antibiotic resistance with a global perspective and to create more stewardship initiatives that promote awareness on the topic.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Humans , Anti-Bacterial Agents/therapeutic use , Cross-Sectional Studies , Surveys and Questionnaires , Prescriptions
5.
PLoS One ; 17(10): e0275737, 2022.
Article in English | MEDLINE | ID: mdl-36301847

ABSTRACT

Antibiotics seize an effect on bacterial composition and diversity and have been demonstrated to induce disruptions on gut microbiomes. This may have implications for human health and wellbeing, and an increasing number of studies suggest a link between the gut microbiome and several diseases. Hence, reducing antibiotic treatments may be beneficial for human health status. Further, antimicrobial resistance (AMR) is an increasing global problem that can be counteracted by limiting the usage of antibiotics. Longer antibiotic treatments have been demonstrated to increase the development of AMR. Therefore, shortening of antibiotic treatment durations, provided it is safe for patients, may be one measure to reduce AMR. In this study, the objective was to investigate effects of standard and reduced antibiotic treatment lengths on gut microbiomes using a murine model. Changes in the murine gut microbiome was assessed after using three different treatment durations of amoxicillin (3, 7 or 14 days) as well as a control group not receiving amoxicillin. Fecal samples were collected before and during the whole experiment, until three weeks past end of treatment. These were further subject for 16S rRNA Illumina MiSeq sequencing. Our results demonstrated significant changes in bacterial diversity, richness and evenness during amoxicillin treatment, followed by a reversion in terms of alpha-diversity and abundance of major phyla, after end of treatment. However, a longer restitution time was indicated for mice receiving amoxicillin for 14 days, and phylum Patescibacteria did not fully recover. In addition, an effect on the composition of Firmicutes was indicated to last for at least three weeks in mice treated with amoxicillin for 14 days. Despite an apparently reversion to a close to original state in overall bacterial diversity and richness, the results suggested more durable changes in lower taxonomical levels. We detected several families, genera and ASVs with significantly altered abundance three weeks after exposure to amoxicillin, as well as bacterial taxa that appeared significantly affected by amoxicillin treatment length. This may strengthen the argument for shorter antibiotic treatment regimens to both limit the emergence of antibiotic resistance and risk of gut microbiome disturbance.


Subject(s)
Amoxicillin , Microbiota , Humans , Mice , Animals , Amoxicillin/pharmacology , RNA, Ribosomal, 16S/genetics , Duration of Therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria
6.
Colloids Surf B Biointerfaces ; 219: 112813, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36084512

ABSTRACT

Oral biofilms can be a major health problem causing infections and chronic inflammation of mucosal tissue. While much effort is put in the investigation of bacteria in biofilms, the role of fungi is often neglected, despite Candida albicans playing a key role in the formation of multispecies oral biofilms. With the rise of antibiotic resistance, new strategies to reduce microbial growth need to be found. Therefore, plant derived polyphenolic molecules have been suggested to reduce both adhesion and growth of pathogenic bacteria and fungi. In this study, we investigated the use of polyphenolic coatings to reduce adhesion and biofilm formation of C. albicans BWP17 on titanium implants. Tannic acid and pyrogallol coatings altered the hydrophobic and charge properties of titanium surfaces, and both compounds were gradually released as active molecules over time. Despite such effects, we found no significant inhibition on growth and biofilm formation of C. Albicans, indicating that the release of active molecules from the coatings did not reach relevant inhibitory concentrations. However, a potential antibiofilm effect was observed by the pH-dependent disassembly of the polyphenolic layer, which caused the biofilm to detach. Hence, further efforts are required to create tailored implant surfaces, which sustainably reduce microbial growth and adhesion.

7.
Front Microbiol ; 13: 1038120, 2022.
Article in English | MEDLINE | ID: mdl-36620054

ABSTRACT

Introduction: Low microbial biomass and high human DNA content in nasopharyngeal aspirate samples hinder comprehensive characterization of microbiota and resistome. We obtained samples from premature infants, a group with increased risk of developing respiratory disorders and infections, and consequently frequent exposure to antibiotics. Our aim was to devise an optimal protocol for handling nasopharyngeal aspirate samples from premature infants, focusing on host DNA depletion and microbiome and resistome characterization. Methods: Three depletion and three DNA extraction protocols were compared, using RT-PCR and whole metagenome sequencing to determine the efficiency of human DNA removal, taxonomic profiling and assignment of antibiotic resistance genes. Protocols were tested using mock communities, as well as pooled and individual patient samples. Results: The only extraction protocol to retrieve the expected DNA yield from mock community samples was based on a lytic method to improve Gram positive recovery (MasterPure™). Host DNA content in non-depleted aliquots from pooled patient samples was 99%. Only samples depleted with MolYsis™ showed satisfactory, but varied reduction in host DNA content, in both pooled and individual patient samples, allowing for microbiome and resistome characterisation (host DNA content from 15% to 98%). Other depletion protocols either retrieved too low total DNA yields, preventing further analysis, or failed to reduce host DNA content. By using Mol_MasterPure protocol on aliquots from pooled patient samples, we increased the number of bacterial reads by 7.6 to 1,725.8-fold compared to non-depleted reference samples. PCR results were indicative of achieved microbial enrichment. Individual patient samples processed with Mol_MasterPure protocol varied greatly in total DNA yield, host DNA content (from 40% to 98%), species and antibiotic resistance gene richness. Discussion: Despite high human DNA and low microbial biomass content in nasopharynx aspirates of preterm infants, we were able to reduce host DNA content to levels compatible with downstream shotgun metagenomic analysis, including bacterial species identification and coverage of antibiotic resistance genes. Whole metagenomic sequencing of microbes colonizing the nasopharynx may contribute to explaining the possible role of airway microbiota in respiratory conditions and reveal carriage of antibiotic resistance genes.

8.
Head Neck ; 43(11): 3374-3385, 2021 11.
Article in English | MEDLINE | ID: mdl-34338386

ABSTRACT

BACKGROUND: This study aimed to evaluate the prognostic significance of expression levels of involucrin (IVL), cytokeratin (CK)-10 and -13 at different intratumor sites (tumor center and invading area) of oral tongue squamous cell carcinoma (OTSCC). METHODS: IVL, CK13 and CK10 expression levels were examined in a multicenter cohort of 146 OTSCCs using immunohistochemistry. External mRNA datasets were used for expression analysis and/or to validate survival associations. RESULTS: External transcriptomic datasets showed downregulation of IVL and KRT13 in oral malignancies including OTSCC as compared to normal controls. The combined loss of IVL and CK13 expression at the invading core but not at the center core was significantly associated with poor differentiation and reduced 5-year overall survival. Multivariate Cox analysis confirmed the loss of CK13 and IVL expression to be an independent prognostic factor. Transcriptomic dataset corroborated immunohistochemistry results. CONCLUSIONS: Combined expression levlels of IVL and CK13 might be useful as prognostic biomarkers in OTSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Keratin-13 , Protein Precursors , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms , Carcinoma, Squamous Cell/genetics , Humans , Keratin-13/genetics , Prognosis , Tongue Neoplasms/genetics
9.
Front Immunol ; 12: 676488, 2021.
Article in English | MEDLINE | ID: mdl-33953733

ABSTRACT

Recent studies have identified a clinical isolate of the commensal Streptococcus mitis that expresses Streptococcus pneumoniae serotype 5 capsule (S. mitis serotype 5) and shows serospecificity toward pneumococcal serotype 5. However, it remains unknown whether S. mitis serotype 5 induces protective immunity against pneumococcal serotype 5. In this study, we evaluated the ability of S. mitis serotype 5 to generate protective immunity in a mouse model of lung infection with pneumococcal serotype 5. Upon challenge infection with S. pneumoniae serotype 5, mice intranasally immunized with S. mitis serotype 5 exhibited reduced pneumococcal loads in the lungs, nasal wash, and bronchoalveolar lavage fluid compared with those receiving PBS (control). The immunized mice displayed significantly higher levels of IgG and IgA antibodies reactive to S. mitis serotype 5, S. pneumoniae serotype 5 or S. pneumoniae serotype 4 than the antibody levels in control mice. In vaccinated mice, the IgG/IgA antibody levels reactive to S. mitis serotype 5 or S. pneumoniae serotype 5 were higher than the levels reactive to S. pneumoniae serotype 4. Furthermore, in-vitro restimulation of the lung-draining mediastinal lymph node cells and splenocytes from immunized mice with killed S. mitis serotype 5, S. pneumoniae serotype 5 or S. pneumoniae serotype 4 showed enhanced Th17, but not Th1 and Th2, responses. Overall, our findings show that mucosal immunization with S. mitis serotype 5 protects against S. pneumoniae serotype 5 infection and induces Th17 and predominant serotype-specific IgG/IgA antibody responses against pneumococcal infection.


Subject(s)
Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Pneumococcal Vaccines/administration & dosage , Pneumonia, Pneumococcal/prevention & control , Polysaccharides, Bacterial/immunology , Serogroup , Streptococcus mitis/immunology , Streptococcus pneumoniae/immunology , Th17 Cells/immunology , Vaccination/methods , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Disease Models, Animal , Female , Mice , Pneumococcal Vaccines/immunology , Pneumonia, Pneumococcal/microbiology , Treatment Outcome
10.
Front Pediatr ; 8: 544460, 2020.
Article in English | MEDLINE | ID: mdl-33178650

ABSTRACT

Although antibiotics confer significant health benefits in treating or preventing bacterial infections, an accumulating wealth of evidence illustrates their detrimental effect on host-microbiota homeostasis, posing a serious menace to the global public health. In recent years, it is becoming evident that infants, who are subjected to frequent antibiotic exposures due to their vulnerability to infection, reflect increased susceptibility to a wide spectrum of diseases, including infection, in later life. Antibiotics induce perturbations of the microbiota or dysbiosis, which in turn alters the host immune responses against pathogens. In comparison with adults, antibiotic treatments in infants have disproportionate consequences because the infant microbiota represents an evolving system that is unstable and immature until 2-3 years of age. However, relatively less knowledge is available on how antibiotics affect the infant microbiota and immunity. In this review article, we focus on how antibiotic treatment regimens influence the infant innate and adaptive immunity to pathogens in humans and animal models, and make the host susceptible to infections in later life. There is a critical need to better understand the effect of antibiotics on infant immune function, which may have implications for developing effective prophylactics and therapeutics against diseases in infants and adults.

12.
Sci Rep ; 10(1): 8737, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457404

ABSTRACT

Helicobacter pylori (HP) infection is an established causative agent for gastric cancer. Although the oral cavity is a part of the gastrointestinal system, the presence and possible causative role of HP in oral squamous cell carcinoma (OSCC) is a subject of controversy. Therefore, the current study aimed to investigate HP infection in two cohorts of OSCC patients with different demographic characteristics, lifestyles and habitual risk factors. A total of 242 formalin-fixed paraffin-embedded OSCC specimens from two different patient cohorts (Norway, n = 171 and Nepal, n = 71) were used to examine HP using immunohistochemistry (IHC) and quantitative polymerase chain reaction (qPCR). Two different HP specific genes (23S rRNA and ureA) were used for TaqMan-based qPCR, and for subsequent verification using HP specific RIDAGENE HP kit and SYBR Green based qPCR. All of the OSCC specimens from both cohorts were found to be negative for HP infection with IHC and qPCR, although the positive control specimens tested positive. Our findings suggest that HP is absent in the examined OSCC cohorts, irrespective of race, lifestyle and habitual risk factors. This indicates that, in contrast to gastric cancer, HP is an unlikely contributing factor for OSCC pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Carcinoma, Squamous Cell/microbiology , Helicobacter Infections/diagnosis , Helicobacter pylori/isolation & purification , Mouth Neoplasms/microbiology , Aged , Case-Control Studies , Female , Helicobacter pylori/genetics , Humans , Life Style , Male , Middle Aged , Nepal , Norway , RNA, Ribosomal, 23S/genetics
14.
J Immunol Res ; 2019: 7906724, 2019.
Article in English | MEDLINE | ID: mdl-31583259

ABSTRACT

Here we show that mouse IgG2a and IgG1 antibodies specific for the commensal Streptococcus mitis cross-react with pathogen Streptococcus pneumoniae serotypes 2 and 4, although the cross-reactivity conferred by IgG2a is stronger than that by IgG1 antibodies. These findings may be important for understanding the S. mitis-induced IgG isotype responses and have consequences for the development of an effective pneumococcal vaccine.


Subject(s)
Antibodies, Bacterial/immunology , Cross Reactions/immunology , Immunoglobulin G/immunology , Streptococcal Infections/immunology , Streptococcus mitis/immunology , Streptococcus pneumoniae/immunology , Animals , Disease Models, Animal , Female , Mice , Streptococcal Infections/microbiology
15.
Front Immunol ; 10: 1203, 2019.
Article in English | MEDLINE | ID: mdl-31214175

ABSTRACT

A diverse community of trillions of commensal bacteria inhabits mucosal and epidermal surfaces in humans and plays an important role in defense against pathogens, including respiratory pathogens. Commensal bacteria act on the host's immune system to induce protective responses that prevent colonization and invasion by pathogens. On the other hand, these bacteria can directly inhibit the growth of respiratory pathogens by producing antimicrobial products/signals and competing for nutrients and adhesion sites. Such mechanisms preserve the niche for commensal bacteria and support the host in containing respiratory infections. Herein, we discuss current evidence on the role of commensal bacteria in conferring protection against respiratory pathogens and the underlying mechanisms by which these bacteria do so. A deeper knowledge of how commensal bacteria interact with the host and pathogens might provide new insights that are poised to aid in the development of vaccines and therapeutics that target infectious diseases.


Subject(s)
Bacterial Physiological Phenomena , Host-Pathogen Interactions , Microbiota , Respiratory Tract Infections/microbiology , Animals , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans
16.
Appl Environ Microbiol ; 85(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30683742

ABSTRACT

Streptococcus pneumoniae is a bacterial pathogen that causes various diseases of public health concern worldwide. Current pneumococcal vaccines target the capsular polysaccharide surrounding the cells. However, only up to 13 of more than 90 pneumococcal capsular serotypes are represented in the current conjugate vaccines. In this study, we used two experimental approaches to evaluate the potential of Streptococcus mitis, a commensal that exhibits immune cross-reactivity with S. pneumoniae, to confer protective immunity to S. pneumoniae lung infection in mice. First, we assessed the immune response and protective effect of wild-type S. mitis against lung infection by S. pneumoniae strains D39 (serotype 2) and TIGR4 (serotype 4). Second, we examined the ability of an S. mitis mutant expressing the S. pneumoniae type 4 capsule (S. mitis TIGR4cps) to elicit focused protection against S. pneumoniae TIGR4. Our results showed that intranasal immunization of mice with S. mitis produced significantly higher levels of serum IgG and IgA antibodies reactive to both S. mitis and S. pneumoniae, as well as enhanced production of interleukin 17A (IL-17A), but not gamma interferon (IFN-γ) and IL-4, compared with control mice. The immunization resulted in a reduced bacterial load in respiratory tissues following lung infection with S. pneumoniae TIGR4 or D39 compared with control mice. With S. mitis TIGR4cps, protection upon challenge with S. pneumoniae TIGR4 was superior. Thus, these findings show the potential of S. mitis to elicit natural serotype-independent protection against two pneumococcal serotypes and to provide the benefits of the well-recognized protective effect of capsule-targeting vaccines.IMPORTANCEStreptococcus pneumoniae causes various diseases worldwide. Current pneumococcal vaccines protect against a limited number of more than 90 pneumococcal serotypes, accentuating the urgent need to develop novel prophylactic strategies. S. pneumoniae and the commensal Streptococcus mitis share immunogenic characteristics that make S. mitis an attractive vaccine candidate against S. pneumoniae In this study, we evaluated the potential of S. mitis and its mutant expressing pneumococcal capsule type 4 (S. mitis TIGR4cps) to induce protection against S. pneumoniae lung infection in mice. Our findings show that intranasal vaccination with S. mitis protects against S. pneumoniae strains D39 (serotype 2) and TIGR4 (serotype 4) in a serotype-independent fashion, which is associated with enhanced antibody and T cell responses. Furthermore, S. mitis TIGR4cps conferred additional protection against S. pneumoniae TIGR4, but not against D39. The findings highlight the potential of S. mitis to generate protection that combines both serotype-independent and serotype-specific responses.


Subject(s)
Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Female , Humans , Immunization , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Lung/immunology , Lung/microbiology , Mice , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/immunology , Serogroup , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/genetics , T-Lymphocytes/immunology
17.
Biofouling ; 33(6): 451-459, 2017 07.
Article in English | MEDLINE | ID: mdl-28524724

ABSTRACT

The chemical decontamination of infected dental implants is essential for the successful treatment of peri-implantitis. The aim of this study was to assess the antibacterial effect of a hydrogen peroxide-titanium dioxide (H2O2-TiO2) suspension against Staphylococcus epidermidis biofilms. Titanium (Ti) coins were inoculated with a bioluminescent S. epidermidis strain for 8 h and subsequently exposed to H2O2 with and without TiO2 nanoparticles or chlorhexidine (CHX). Bacterial regrowth, bacterial load and viability after decontamination were analyzed by continuous luminescence monitoring, live/dead staining and scanning electron microscopy. Bacterial regrowth was delayed on surfaces treated with H2O2-TiO2 compared to H2O2. H2O2-based treatments resulted in a lower bacterial load compared to CHX. Few viable bacteria were found on surfaces treated with H2O2 and H2O2-TiO2, which contrasted with a uniform layer of dead bacteria for surfaces treated with CHX. H2O2-TiO2 suspensions could therefore be considered an alternative approach in the decontamination of dental implants.


Subject(s)
Anti-Bacterial Agents/pharmacology , Decontamination/methods , Dental Implants/microbiology , Hydrogen Peroxide/pharmacology , Staphylococcus epidermidis/drug effects , Titanium/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Load , Biofilms/drug effects , Chlorhexidine/pharmacology , Hydrogen Peroxide/chemistry , Peri-Implantitis/microbiology , Peri-Implantitis/prevention & control , Surface Properties , Suspensions , Titanium/chemistry
18.
Front Immunol ; 8: 1971, 2017.
Article in English | MEDLINE | ID: mdl-29387057

ABSTRACT

Commensal microbes are currently in the limelight in biomedical research because they play an important role in health and disease. Humans harbor an enormous diversity of commensals in various parts of the body, including the gastrointestinal and respiratory tracts. Advancement in metagenomic and other omic approaches, and development of suitable animal models have provided an unprecedented appreciation into the diversity of commensals, and the intricacies of their intimate communication with the host immune system. Most studies have focused on the host-commensal interaction in the gut, while less is known on this relationship in other sites of the body, such as the respiratory tract. In this article, we review emerging data from human and animal studies on the host responses to respiratory commensals, immune cross-reactivity between commensals and pathogens, and use of commensals as a vaccine delivery system. A better understanding of the delicate interplay between commensals and host may aid in efforts to develop effective vaccines and therapeutics.

19.
PLoS One ; 9(8): e104306, 2014.
Article in English | MEDLINE | ID: mdl-25119879

ABSTRACT

BACKGROUND: Carriage of and infection with Streptococcus pneumoniae is known to predominantly induce T helper 17 (Th17) responses in humans, but the types of Th cells showing reactivity towards commensal streptococci with low pathogenic potential, such as the oral commensals S. mitis and S. salivarius, remain uncharacterized. METHODS: Memory CD4(+) T helper (Th) cell subsets were isolated from healthy human blood donors according to differential expression of chemokine receptors, expanded in vitro using polyclonal stimuli and characterized for reactivity against different streptococcal strains. RESULTS: Th cells responding to S. mitis, S. salivarius and S. pneumoniae were predominantly in a CCR6(+)CXCR3(+) subset and produced IFN-γ, and in a CCR6(+)CCR4(+) subset and produced IL-17 and IL-22. Frequencies of S. pneumoniae-reactive Th cells were higher than frequencies of S. mitis- and S. salivarius-specific Th cells. S. mitis and S. pneumoniae isogenic capsule knock-out mutants and a S. mitis mutant expressing the serotype 4 capsule of S. pneumoniae showed no different Th cell responses as compared to wild type strains. S. mitis-specific Th17 cells showed cross-reactivity with S. pneumoniae. CONCLUSIONS: As Th17 cells partly control clearance of S. pneumoniae, cross-reactive Th17 cells that may be induced by commensal bacterial species may influence the immune response, independent of capsule expression.


Subject(s)
Mouth/microbiology , Streptococcus mitis/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Helper-Inducer/immunology , Cross Reactions/immunology , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukins/immunology , Interleukin-22
20.
FEMS Immunol Med Microbiol ; 65(2): 326-34, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22443118

ABSTRACT

Frequent use of medical implants has led Staphylococcus epidermidis to develop into an opportunistic pathogen. The virulence is mainly linked to biofilm formation. Infections associated with biofilms are difficult to treat owing to enhanced resistance to antibiotics. Therefore, new and alternative treatments are called for. Bacterial communication is one of the regulatory mechanisms suggested to be involved in coordinating biofilm formation. In this study, we compared three communication inhibitors for preventing in vitro biofilm formation: a synthetic furanone, and two synthetic thiophenones, which are sulphur analogues of furanones. Furanones naturally source from the red macro alga Delisea pulchra. We also investigated the effect of thiophenone on transcriptional levels of genes associated with biofilm formation. We found that thiophenones were more effective in inhibiting biofilm formation than furanone, also in presence of albumin. We furthermore found that the thiophenones inhibited biofilm formation and bacterial communication more than furanones, and were less cytotoxic. The expression of the icaC and the lrgB genes, which are associated with biofilm formation, were affected by the thiophenone.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Furans/pharmacology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/physiology , Sulfur Compounds/pharmacology , Bacterial Proteins/biosynthesis , Gene Expression Profiling , Humans , Virulence Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...