Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Biofouling ; 36(3): 245-255, 2020 03.
Article in English | MEDLINE | ID: mdl-32326753

ABSTRACT

Dental plaque is a biofilm composed of a complex oral microbial community. The accumulation of plaque in the pit and fissures of dental elements often leads to the development of tooth decay (dental caries). Here, potent anti-biofilm materials were developed by incorporating zinc methacrylates or di-n-butyl-dimethacrylate-tin into the light-curable sealant and their physical, mechanical, and biological properties were evaluated. The data revealed that 5% di-n-butyl-dimethacrylate-tin (SnM 5%) incorporated sealant showed strong anti-biofilm efficacy against various single-species (Streptococcus mutans or Streptococcus oralis or Candida albicans) and S. mutans-C. albicans cross-kingdom dual-species biofilms without either impairing the mechanical properties of the sealant or causing cytotoxicities against mouse fibroblasts. The findings indicate that the incorporation of SnM 5% in the experimental pit and fissure self-adhesive sealant may have the potential to be part of current chemotherapeutic strategies to prevent the formation of cariogenic oral biofilms that cause dental caries.


Subject(s)
Adhesives/pharmacology , Biofilms/drug effects , Dental Caries/prevention & control , Pit and Fissure Sealants/pharmacology , Zinc/chemistry , Adhesives/chemistry , Animals , Biofilms/growth & development , Candida albicans/drug effects , Candida albicans/growth & development , Dental Caries/microbiology , Humans , Methacrylates/chemistry , Mice , Microbiota/drug effects , Pit and Fissure Sealants/chemistry , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Streptococcus oralis/drug effects , Streptococcus oralis/growth & development
2.
Nanomedicine ; 14(7): 2307-2316, 2018 10.
Article in English | MEDLINE | ID: mdl-29410321

ABSTRACT

'Soft' nanomaterials have the potential to produce substantive antibiofilm effects. The aim of this study was to understand the oral antimicrobial activity of soft nanomaterials generated from alpha-tocopherol (α-T) and alpha-tocopherol phosphate (α-TP). (+) α-TP formed planar bilayer islands (175 ± 21 nm, -14.9 ± 3.5 mV) in a Trizma® buffer, whereas (+) α-T formed spherical liposomes (563 ± 1 nm, -10.5 ± 0.2 mV). The (+) α-TP bilayers displayed superior Streptococcus oralis biofilm growth retardation, a more substantive action, generated a superior adsorption to hydroxyapatite and showed an enhanced inhibition of multi-species bacterial saliva biofilm growth (38 ± 7µm vs 58 ± 18 µm, P ˂ 0.05) compared to (+) α-T. Atomic force microscopy data indicated that the ability of the 'soft' α-TP nanomaterials to transition into planar bilayer structures upon contact with interfaces facilitated their adhesive properties and substantive antimicrobial effects.


Subject(s)
Anti-Infective Agents/administration & dosage , Biofilms/drug effects , Lipid Bilayers/chemistry , Saliva/microbiology , Streptococcus mutans/drug effects , Streptococcus oralis/drug effects , alpha-Tocopherol/analogs & derivatives , Adhesives , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Biofilms/growth & development , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Microscopy, Atomic Force , Mouth/microbiology , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
3.
Proc Natl Acad Sci U S A ; 111(10): 3835-40, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24567409

ABSTRACT

The formation of dental plaque, a highly complex biofilm that causes gingivitis and periodontitis, requires specific adherence among many oral microbes, including the coaggregation of Actinomyces oris with Streptococcus oralis that helps to seed biofilm development. Here, we report the discovery of a key coaggregation factor for this process. This protein, which we named coaggregation factor A (CafA), is one of 14 cell surface proteins with the LPXTG motif predicted in A. oris MG1, whose function was hitherto unknown. By systematic mutagenesis of each of these genes and phenotypic characterization, we found that the Actinomyces/Streptococcus coaggregation is only abolished by deletion of cafA. Subsequent biochemical and cytological experiments revealed that CafA constitutes the tip of a unique form of the type 2 fimbria long known for its role in coaggregation. The direct and predominant role of CafA in adherence is evident from the fact that CafA or an antibody against CafA inhibits coaggregation, whereas the shaft protein FimA or a polyclonal antibody against FimA has no effect. Remarkably, FimA polymerization was blocked by deletion of genes for both CafA and FimB, the previously described tip protein of the type 2 fimbria. Together, these results indicate that some surface proteins not linked to a pilus gene cluster in Gram-positive bacteria may hijack the pilus. These unique tip proteins displayed on a common pilus shaft may serve distinct physiological functions. Furthermore, the pilus shaft assembly in Gram-positive bacteria may require a tip, as is true for certain Gram-negative bacterial pili.


Subject(s)
Actinomyces/metabolism , Bacterial Proteins/metabolism , Biofilms/growth & development , Dental Plaque/microbiology , Fimbriae, Bacterial/physiology , Membrane Proteins/metabolism , Streptococcus oralis/metabolism , Actinomyces/growth & development , Amino Acid Motifs/genetics , Bacterial Proteins/genetics , Blotting, Western , Cell Fractionation , Escherichia coli , Humans , Membrane Proteins/genetics , Microscopy, Immunoelectron , Multigene Family/genetics , Mutagenesis , Streptococcus oralis/growth & development
4.
Appl Microbiol Biotechnol ; 100(15): 6767-6777, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27102127

ABSTRACT

Lactic acid bacteria (LAB) can interfere with pathogens through different mechanisms; one is the production of biosurfactants, a group of surface-active molecules, which inhibit the growth of potential pathogens. In the present study, biosurfactants produced by Lactobacillus reuteri DSM 17938, Lactobacillus acidophilus DDS-1, Lactobacillus rhamnosus ATCC 53103, and Lactobacillus paracasei B21060 were dialyzed (1 and 6 kDa) and characterized in term of reduction of surface tension and emulsifying activity. Then, aliquots of the different dialyzed biosurfactants were added to Streptococcus mutans ATCC 25175 and Streptococcus oralis ATCC 9811 in the culture medium during the formation of biofilm on titanium surface and the efficacy was determined by agar plate count, biomass analyses, and flow cytometry. Dialyzed biosurfactants showed abilities to reduce surface tension and to emulsifying paraffin oil. Moreover, they significantly inhibited the adhesion and biofilm formation on titanium surface of S. mutans and S. oralis in a dose-dependent way, as demonstrated by the remarkable decrease of cfu/ml values and biomass production. The antimicrobial properties observed for dialyzed biosurfactants produced by the tested lactobacilli opens future prospects for their use against microorganisms responsible of oral diseases.


Subject(s)
Anti-Bacterial Agents/metabolism , Biofilms/growth & development , Lacticaseibacillus paracasei/metabolism , Lacticaseibacillus rhamnosus/metabolism , Lactobacillus acidophilus/metabolism , Limosilactobacillus reuteri/metabolism , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , Surface-Active Agents/metabolism , Bacterial Adhesion/drug effects , Microbial Sensitivity Tests , Streptococcus mutans/drug effects , Streptococcus oralis/drug effects , Surface Tension/drug effects , Surface-Active Agents/pharmacology , Titanium
5.
Biofouling ; 32(9): 1079-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27643392

ABSTRACT

Despite the widespread use of fluoride for the prevention of dental caries, few studies have demonstrated the effects of fluoride on the bacterial composition of dental biofilms. This study investigated whether fluoride affects the proportion of Streptococcus mutans and S. oralis in mono- and dual-species biofilm models, via microbiological, biochemical, and confocal fluorescence microscope studies. Fluoride did not affect the bacterial count and bio-volume of S. mutans and S. oralis in mono-species biofilms, except for the 24-h-old S. mutans biofilms. However, fluoride reduced the proportion and bio-volume of S. mutans but did not decrease those of S. oralis during both S. oralis and S. mutans dual-species biofilm formation, which may be related to the decrease in extracellular polysaccharide formation by fluoride. These results suggest that fluoride may prevent the shift in the microbial proportion to cariogenic bacteria in dental biofilms, subsequently inhibiting the cariogenic bacteria dominant biofilm formation.


Subject(s)
Antibiosis/drug effects , Biofilms/drug effects , Fluorides/pharmacology , Streptococcus mutans/drug effects , Streptococcus oralis/drug effects , Bacterial Load/drug effects , Dental Caries/microbiology , Dose-Response Relationship, Drug , Humans , Models, Biological , Streptococcus mutans/growth & development , Streptococcus mutans/physiology , Streptococcus oralis/growth & development , Streptococcus oralis/physiology
6.
Odontology ; 104(3): 310-7, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26319990

ABSTRACT

Streptococcus gordonii, a bacterium involved in the initial colonization of tooth surfaces, contributes to dental biofilm formation and is an important cause of infective endocarditis. This study aimed to investigate the influence of surface reaction-type pre-reacted glass ionomer (S-PRG) filler on oral bacterial growth and aggregation of S. gordonii. The effect of various concentrations of S-PRG eluate on the growth and the biofilm formation of S. gordonii and other oral microorganisms (Streptococcus mutans, Streptococcus oralis, Lactobacillus acidophilus, and Candida albicans) was assessed. In addition, the effect of S-PRG eluate on coaggregation of S. gordonii with both S. oralis and Fusobacterium nucleatum was assessed. The effect of S-PRG eluate treatment on autoaggregation of S. gordonii was also evaluated. Our results indicate that S-PRG eluate treatment reduced both for the growth and for biofilm of all organisms in a dose-dependent manner. Coaggregation of S. gordonii with both S. oralis and F. nucleatum was inhibited by S-PRG eluate, whereas autoaggregation of S. gordonii increased at certain concentrations of S-PRG eluate. These results indicate that the S-PRG filler possesses antimicrobial activity that is mediated by inhibiting growth and biofilm of oral microorganisms, and by suppressing coaggregation of S. gordonii. In addition, these findings indicate that coaggregation of S. gordonii with other bacteria is inhibited by increased autoaggregation of S. gordonii.


Subject(s)
Glass Ionomer Cements/pharmacology , Streptococcus gordonii/growth & development , Bacterial Adhesion , Biofilms , Candida albicans/growth & development , Fusobacterium nucleatum/growth & development , Glass Ionomer Cements/chemistry , Lactobacillus acidophilus/growth & development , Materials Testing , Spectrophotometry, Atomic , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , Surface Properties
7.
PLoS Pathog ; 8(4): e1002623, 2012.
Article in English | MEDLINE | ID: mdl-22496649

ABSTRACT

Virulent biofilms are responsible for a range of infections, including oral diseases. All biofilms harbor a microbial-derived extracellular-matrix. The exopolysaccharides (EPS) formed on tooth-pellicle and bacterial surfaces provide binding sites for microorganisms; eventually the accumulated EPS enmeshes microbial cells. The metabolic activity of the bacteria within this matrix leads to acidification of the milieu. We explored the mechanisms through which the Streptococcus mutans-produced EPS-matrix modulates the three-dimensional (3D) architecture and the population shifts during morphogenesis of biofilms on a saliva-coated-apatitic surface using a mixed-bacterial species system. Concomitantly, we examined whether the matrix influences the development of pH-microenvironments within intact-biofilms using a novel 3D in situ pH-mapping technique. Data reveal that the production of the EPS-matrix helps to create spatial heterogeneities by forming an intricate network of exopolysaccharide-enmeshed bacterial-islets (microcolonies) through localized cell-to-matrix interactions. This complex 3D architecture creates compartmentalized acidic and EPS-rich microenvironments throughout the biofilm, which triggers the dominance of pathogenic S. mutans within a mixed-species system. The establishment of a 3D-matrix and EPS-enmeshed microcolonies were largely mediated by the S. mutans gtfB/gtfC genes, expression of which was enhanced in the presence of Actinomyces naeslundii and Streptococcus oralis. Acidic pockets were found only in the interiors of bacterial-islets that are protected by EPS, which impedes rapid neutralization by buffer (pH 7.0). As a result, regions of low pH (<5.5) were detected at specific locations along the surface of attachment. Resistance to chlorhexidine was enhanced in cells within EPS-microcolony complexes compared to those outside such structures within the biofilm. Our results illustrate the critical interaction between matrix architecture and pH heterogeneity in the 3D environment. The formation of structured acidic-microenvironments in close proximity to the apatite-surface is an essential factor associated with virulence in cariogenic-biofilms. These observations may have relevance beyond the mouth, as matrix is inherent to all biofilms.


Subject(s)
Biofilms/growth & development , Mouth/microbiology , Polysaccharides/metabolism , Streptococcus mutans , Streptococcus oralis , Animals , Humans , Hydrogen-Ion Concentration , Streptococcus mutans/growth & development , Streptococcus mutans/metabolism , Streptococcus mutans/pathogenicity , Streptococcus oralis/growth & development , Streptococcus oralis/metabolism , Streptococcus oralis/pathogenicity , Virulence Factors/metabolism
8.
J Periodontal Res ; 49(3): 323-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23815431

ABSTRACT

BACKGROUND AND OBJECTIVE: Bacteria in the oral cavity grow in the form of biofilms; these structures are subject to constant saliva or gingival crevicular fluid flow conditions. The aims of this study were: (i) to develop and to characterize an in-vitro biofilm model with oral bacteria growing under flow and shear conditions; and (ii) to demonstrate the usefulness of the model for evaluating the activity of three antiplaque agents. MATERIAL AND METHODS: We used a bioreactor to grow the oral bacteria Streptococcus oralis, Actinomyces naeslundii, Veillonella parvula, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis under planktonic conditions. Biofilms were established using a modified Robbins device on hydroxyapatite (HAP) discs. Three- to 7-d-old biofilms were analysed using culture methods, scanning electron microscopy, Live/Dead staining and fluorescence in-situ hybridization (confocal laser scanning microscopy). Finally, we assessed the antimicrobial activity of three mouthrinses [0.12% chlorhexidine (CHX), 0.12% chlorhexidine and sodium fluoride (CHX+NaF) and 0.12% chlorhexidine and 0.05% cetylpyridinium chloride (CHX+CPC)] using a planktonic test (short interval-killing test) and in our 4-d biofilm model. RESULTS: The viable cell counts showed that each species was consistently found in the biofilms throughout the study. The architecture and cell distribution were similar to those described for biofilms in situ, with the exception of a thin layer of living cells that was found close to the HAP. The effectiveness test of the mouthwashes demonstrated that cells in biofilms showed more tolerance compared with planktonic cells. Moreover, it was observed that in 4-d biofilm formed in vitro, CHX+CPC caused significantly higher mortality compared with CHX (p = 0.003) and CHX+NaF (p < 0.001). CONCLUSION: Our results suggest that we have a highly reproducible system for multispecies oral biofilm formation and that it is a useful tool for assessing antibacterial molecules before their clinical evaluation. It also has great potential to be used in basic research on supragingival and subgingival biofilms.


Subject(s)
Biofilms/growth & development , Bioreactors , Mouth/microbiology , Actinomyces/growth & development , Aggregatibacter actinomycetemcomitans/growth & development , Anti-Infective Agents, Local/pharmacology , Bacterial Load/drug effects , Bacteriological Techniques , Cetylpyridinium/pharmacology , Chlorhexidine/pharmacology , Durapatite/chemistry , Fusobacterium nucleatum/growth & development , Humans , In Situ Hybridization, Fluorescence , Microbial Sensitivity Tests , Microbial Viability , Microscopy, Confocal , Microscopy, Electron, Scanning , Mouthwashes/pharmacology , Porphyromonas gingivalis/growth & development , Saliva/physiology , Sodium Fluoride/pharmacology , Streptococcus oralis/growth & development , Veillonella/growth & development
9.
J Periodontal Res ; 48(4): 517-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23278531

ABSTRACT

BACKGROUND AND OBJECTIVE: Subgingival biofilms are the prime etiological factor of periodontal disease. Owing to their complex polymicrobial nature, quantification of individual bacterial species within the biofilm for research and diagnostic purposes can be methodologically challenging. The aims of this study were to establish a quantitative real-time PCR (qPCR) assay to quantify the bacteria used in our 10-species in vitro 'subgingival' biofilm model and to compare the quantitative outcome with fluorescence microscopy and colony-forming unit (CFU) counts on selective agar plates. MATERIAL AND METHODS: The 10 species included in the in vitro biofilm were Streptococcus oralis, Streptococcus anginosus, Veillonella dispar, Fusobacterium nucleatum, Treponema denticola, Tannerella forsythia, Actinomyces oris, Campylobacter rectus, Porphyromonas gingivalis and Prevotella intermedia. The numbers of each species were quantified at two time points using qPCR, microscopy counting following fluorescence in-situ hybridization (FISH) or immunofluorescence staining, and counting of CFUs after growth on selective agar plates. RESULTS: All 10 species were successfully quantified using qPCR and FISH or immunofluorescence, and the eight species culturable on selective agar plates were also quantified by counting the numbers of CFUs after growth on selective agar. In early biofilm cultures, all methods showed a significant correlation, although the absolute numbers differed between methods. In late biofilm cultures, measurements obtained using qPCR and FISH or immunofluorescence, but not by CFU counts, maintained significant correlation. CFU counts yielded lower values than did measurements made using the other two methods. CONCLUSION: Quantitative PCR and epifluorescence microscopy can be easily combined with each other to determine species-specific bacterial numbers within biofilms. However, conventional bacterial cultures cannot be as efficiently combined using these molecular detection methods. This may be crucial in designing and selecting appropriate clinical diagnostic methods for subgingival biofilm samples.


Subject(s)
Bacterial Load/methods , Biofilms/classification , Gingiva/microbiology , Microscopy, Fluorescence/methods , Real-Time Polymerase Chain Reaction/methods , Actinomyces/growth & development , Actinomyces/isolation & purification , Agar , Bacteriological Techniques , Bacteroides/growth & development , Bacteroides/isolation & purification , Biofilms/growth & development , Campylobacter rectus/growth & development , Campylobacter rectus/isolation & purification , Culture Media , Fluorescent Antibody Technique , Fusobacterium nucleatum/growth & development , Fusobacterium nucleatum/isolation & purification , Humans , In Situ Hybridization, Fluorescence , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/isolation & purification , Prevotella intermedia/growth & development , Prevotella intermedia/isolation & purification , Streptococcus anginosus/growth & development , Streptococcus anginosus/isolation & purification , Streptococcus oralis/growth & development , Streptococcus oralis/isolation & purification , Time Factors , Treponema denticola/growth & development , Treponema denticola/isolation & purification , Veillonella/growth & development , Veillonella/isolation & purification
10.
Microbiology (Reading) ; 158(Pt 7): 1783-1795, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22493304

ABSTRACT

Streptococcus gordonii and Streptococcus oralis are among the first bacterial species to colonize clean tooth surfaces. Both produce autoinducer-2 (AI-2): a family of inter-convertible cell-cell signal molecules synthesized by the LuxS enzyme. The overall aim of this work was to determine whether AI-2 alters interspecies interactions between S. gordonii DL1 and S. oralis 34 within dual-species biofilms in flowing human saliva. Based upon AI-2 bioluminescence assays, S. gordonii DL1 produced more AI-2 activity than S. oralis 34 in batch culture, and both were able to remove AI-2 activity from solution. In single-species, saliva-fed flowcell systems, S. oralis 34 formed scant biofilms that were similar to the luxS mutant. Conversely, S. gordonii DL1 formed confluent biofilms while the luxS mutant formed architecturally distinct biofilms that possessed twofold greater biovolume than the wild-type. Supplementing saliva with 0.1-10 nM chemically synthesized AI-2 (csAI-2) restored the S. gordonii DL1 luxS biofilm phenotype to that which was similar to the wild-type; above or below this concentration range, biofilms were architecturally similar to that formed by the luxS mutant. In dual-species biofilms, S. gordonii DL1 was always more abundant than S. oralis 34. Compared with dual-species, wild-type biofilms, the biovolume occupied by S. oralis 34 was reduced by greater than sevenfold when neither species produced AI-2. The addition of 1 nM csAI-2 to the dual-species luxS-luxS mutant biofilms re-established the biofilm phenotype to resemble that of the wild-type pair. Thus, this work demonstrates that AI-2 can alter the biofilm structure and composition of pioneering oral streptococcal biofilms. This may influence the subsequent succession of other species into oral biofilms and the ecology of dental plaque.


Subject(s)
Biofilms/growth & development , Homoserine/analogs & derivatives , Lactones/metabolism , Microbial Interactions , Streptococcus gordonii/physiology , Streptococcus oralis/physiology , Homoserine/metabolism , Humans , Saliva/microbiology , Streptococcus gordonii/growth & development , Streptococcus gordonii/metabolism , Streptococcus oralis/growth & development , Streptococcus oralis/metabolism
11.
J Periodontal Res ; 46(2): 252-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21261622

ABSTRACT

BACKGROUND AND OBJECTIVE: There are few in vitro models available in the scientific literature for study of the structure, formation and development of the subgingival biofilm. The purpose of this study was to develop and validate an in vitro biofilm model, using representative selected bacteria from the subgingival microbiota. MATERIAL AND METHODS: Six standard reference strains were used to develop biofilms over sterile ceramic calcium hydroxyapatite discs coated with saliva within the wells of presterilized polystyrene tissue culture plates. The selected species represent initial (Streptococcus oralis and Actinomyces naeslundii), early (Veillonella parvula), secondary (Fusobacterium nucleatum) and late colonizers (Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans). The structure of the biofilm obtained was studied using a vital fluorescence technique in conjunction with confocal laser scanning microscopy. The biofilm bacterial kinetics were studied by terminal restriction fragment length polymorphism analysis. RESULTS: After 12 h, initial and early colonizers were the first microorganisms detected adhering to the calcium hydroxyapatite discs. The intermediate colonizer F. nucleatum was not detected in the model until 24 h of incubation. Late colonizers A. actinomycetemcomitans and P. gingivalis could be measured inside the biofilm after 48 h. The biofilm reached its steady state between 72 and 96 h after inoculation, with bacterial vitality increasing from the hydroxyapatite surface to the central part of the biofilm. CONCLUSION: An in vitro biofilm model was developed and validated, demonstrating a pattern of bacterial colonization and maturation similar to the in vivo development of the subgingival biofilm.


Subject(s)
Biofilms/growth & development , Dental Plaque/microbiology , Gingiva/microbiology , Actinomyces/growth & development , Actinomyces/physiology , Aggregatibacter actinomycetemcomitans/growth & development , Aggregatibacter actinomycetemcomitans/physiology , Bacterial Adhesion/physiology , Bacterial Physiological Phenomena , Bacteriological Techniques , Culture Media , Durapatite , Fluorescence , Fusobacterium nucleatum/growth & development , Fusobacterium nucleatum/physiology , Humans , Microbial Viability , Microscopy, Confocal , Polymorphism, Restriction Fragment Length , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/physiology , Saliva/microbiology , Streptococcus oralis/growth & development , Streptococcus oralis/physiology , Time Factors , Veillonella/growth & development , Veillonella/physiology
12.
Molecules ; 16(5): 3530-43, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21525793

ABSTRACT

Surface colonization is an essential step in biofilm development. The ability of oral pathogens to adhere to tooth surfaces is directly linked with the presence of specific molecules at the bacterial surface that can interact with enamel acquired pellicle ligands. In light of this, the aim of this study was to verify inhibitory and antibiofilm action of lectins from the Diocleinaesubtribe against Streptococcus mutans and Streptococcus oralis. The inhibitory action against planctonic cells was assessed using lectins from Canavaliaensi formis (ConA), Canavalia brasiliensis (ConBr), Canavalia maritima (ConM), Canavalia gladiata (CGL) and Canavalia boliviana (ConBol). ConBol, ConBr and ConM showed inhibitory activity on S. mutans growth. All lectins, except ConA, stimulated significantly the growth of S. oralis. To evaluate the effect on biofilm formation, clarified saliva was added to 96-well, flat-bottomed polystyrene plates, followed by the addition of solutions containing 100 or 200 µg/mL of the selected lectins. ConBol, ConM and ConA inhibited the S. mutans biofilms. No effects were found on S. oralis biofilms. Structure/function analysis were carried out using bioinformatics tools. The aperture and deepness of the CRD (Carbohydrate Recognition Domain) permit us to distinguish the two groups of Canavalia lectins in accordance to their actions against S. mutans and S. oralis. The results found provide a basis for encouraging the use of plant lectins as biotechnological tools in ecological control and prevention of caries disease.


Subject(s)
Biofilms/drug effects , Plant Lectins/pharmacology , Streptococcus/drug effects , Streptococcus/growth & development , Concanavalin A/pharmacology , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Streptococcus oralis/drug effects , Streptococcus oralis/growth & development
13.
mSphere ; 6(6): e0087521, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34908459

ABSTRACT

As common commensals residing on mucosal tissues, Lactobacillus species are known to promote health, while some Streptococcus species act to enhance the pathogenicity of other organisms in those environments. In this study, we used a combination of in vitro imaging of live biofilms and computational modeling to explore biofilm interactions between Streptococcus oralis, an accessory pathogen in oral candidiasis, and Lactobacillus paracasei, an organism with known probiotic properties. A computational agent-based model was created where the two species interact only by competing for space, oxygen and glucose. Quantification of bacterial growth in live biofilms indicated that S. oralis biomass and cell numbers were much lower than predicted by the model. Two subsequent models were then created to examine more complex interactions between these species, one where L. paracasei secretes a surfactant, and another where L. paracasei secretes an inhibitor of S. oralis growth. We observed that the growth of S. oralis could be affected by both mechanisms. Further biofilm experiments support the hypothesis that L. paracasei may secrete an inhibitor of S. oralis growth, although they do not exclude that a surfactant could also be involved. This contribution shows how agent-based modeling and experiments can be used in synergy to address multiple species biofilm interactions, with important roles in mucosal health and disease. IMPORTANCE We previously discovered a role of the oral commensal Streptococcus oralis as an accessory pathogen. S. oralis increases the virulence of Candida albicans infections in murine oral candidiasis and epithelial cell models through mechanisms which promote the formation of tissue-damaging biofilms. Lactobacillus species have known inhibitory effects on biofilm formation of many microbes, including Streptococcus species. Agent-based modeling has great advantages as a means of exploring multifaceted relationships between organisms in complex environments such as biofilms. Here, we used an iterative collaborative process between experimentation and modeling to reveal aspects of the mostly unexplored relationship between S. oralis and L. paracasei in biofilm growth. The inhibitory nature of L. paracasei on S. oralis in biofilms may be exploited as a means of preventing or alleviating mucosal fungal infections.


Subject(s)
Biofilms/growth & development , Lacticaseibacillus paracasei/growth & development , Streptococcus oralis/growth & development , Systems Analysis , Virulence
14.
BMC Microbiol ; 10: 111, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20398271

ABSTRACT

BACKGROUND: Microbial cell-cell interactions in the oral flora are believed to play an integral role in the development of dental plaque and ultimately, its pathogenicity. The effects of other species of oral bacteria on biofilm formation and virulence gene expression by Streptococcus mutans, the primary etiologic agent of dental caries, were evaluated using a dual-species biofilm model and RealTime-PCR analysis. RESULTS: As compared to mono-species biofilms, biofilm formation by S. mutans was significantly decreased when grown with Streptococcus sanguinis, but was modestly increased when co-cultivated with Lactobacillus casei. Co-cultivation with S. mutans significantly enhanced biofilm formation by Streptococcus oralis and L. casei, as compared to the respective mono-species biofilms. RealTime-PCR analysis showed that expression of spaP (for multi-functional adhesin SpaP, a surface-associated protein that S. mutans uses to bind to the tooth surface in the absence of sucrose), gtfB (for glucosyltransferase B that synthesizes alpha1,6-linked glucan polymers from sucrose and starch carbohydrates) and gbpB (for surface-associated protein GbpB, which binds to the glucan polymers) was decreased significantly when S. mutans were co-cultivated with L. casei. Similar results were also found with expression of spaP and gbpB, but not gtfB, when S. mutans was grown in biofilms with S. oralis. Compared to mono-species biofilms, the expression of luxS in S. mutans co-cultivated with S. oralis or L. casei was also significantly decreased. No significant differences were observed in expression of the selected genes when S. mutans was co-cultivated with S. sanguinis. CONCLUSIONS: These results suggest that the presence of specific oral bacteria differentially affects biofilm formation and virulence gene expression by S. mutans.


Subject(s)
Antibiosis , Biofilms/growth & development , Lacticaseibacillus casei/physiology , Streptococcus mutans/growth & development , Streptococcus mutans/pathogenicity , Streptococcus oralis/physiology , Streptococcus sanguis/physiology , Adhesins, Bacterial/biosynthesis , Bacterial Proteins/biosynthesis , Gene Expression Profiling , Humans , Lacticaseibacillus casei/growth & development , Streptococcus oralis/growth & development , Streptococcus sanguis/growth & development , Virulence , Virulence Factors/biosynthesis
15.
J Clin Periodontol ; 37(6): 563-73, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20507380

ABSTRACT

BACKGROUND: Peri-implantitis is common in patients with dental implants. We performed a single-blinded longitudinal randomized study to assess the effects of mechanical debridement on the peri-implant microbiota in peri-implantitis lesions. MATERIALS AND METHODS: An expanded checkerboard DNA-DNA hybridization assay encompassing 79 different microorganisms was used to study bacterial counts before and during 6 months following mechanical treatment of peri-implantitis in 17 cases treated with curettes and 14 cases treated with an ultrasonic device. Statistics included non-parametric tests and GLM multivariate analysis with p<0001 indicating significance and 80% power. RESULTS: At selected implant test sites, the most prevalent bacteria were: Fusobacterium nucleatum sp., Staphylococci sp., Aggregatibacter actinomycetemcomitans, Helicobacter pylori, and Tannerella forsythia. 30 min. after treatment with curettes, A. actinomycetemcomitans (serotype a), Lactobacillus acidophilus, Streptococcus anginosus, and Veillonella parvula were found at lower counts (p<0.001). No such differences were found for implants treated with the ultrasonic device. Inconsistent changes occurred following the first week. No microbiological differences between baseline and 6-month samples were found for any species or between treatment study methods in peri-implantitis. CONCLUSIONS: Both methods failed to eliminate or reduce bacterial counts in peri-implantitis. No group differences were found in the ability to reduce the microbiota in peri-implantitis.


Subject(s)
Bacteria/growth & development , Dental Implants/microbiology , Dental Prophylaxis/methods , Periodontitis/microbiology , Ultrasonic Therapy/methods , Aggregatibacter actinomycetemcomitans/growth & development , Bacteria/classification , Bacteroides/growth & development , Capnocytophaga/growth & development , Colony Count, Microbial , Female , Follow-Up Studies , Fusobacterium nucleatum/growth & development , Helicobacter pylori/growth & development , Humans , Lactobacillus/growth & development , Lactobacillus acidophilus/growth & development , Longitudinal Studies , Male , Periodontal Pocket/microbiology , Periodontal Pocket/therapy , Periodontitis/therapy , Single-Blind Method , Smoking , Staphylococcus/growth & development , Streptococcus/growth & development , Streptococcus anginosus/growth & development , Streptococcus gordonii/growth & development , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , Treatment Outcome , Veillonella/growth & development
16.
Clin Oral Investig ; 14(4): 403-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19565279

ABSTRACT

In vitro plaque removal studies require biofilm models that resemble in vivo dental plaque. Here, we compare contact and non-contact removal of single and dual-species biofilms as well as of biofilms grown from human whole saliva in vitro using different biofilm models. Bacteria were adhered to a salivary pellicle for 2 h or grown after adhesion for 16 h, after which, their removal was evaluated. In a contact mode, no differences were observed between the manual, rotating, or sonic brushing; and removal was on average 39%, 84%, and 95% for Streptococcus mutans, Streptococcus oralis, and Actinomyces naeslundii, respectively, and 90% and 54% for the dual- and multi-species biofilms, respectively. However, in a non-contact mode, rotating and sonic brushes still removed considerable numbers of bacteria (24-40%), while the manual brush as a control (5-11%) did not. Single A. naeslundii and dual-species (A. naeslundii and S. oralis) biofilms were more difficult to remove after 16 h growth than after 2 h adhesion (on average, 62% and 93% for 16- and 2-h-old biofilms, respectively), while in contrast, biofilms grown from whole saliva were easier to remove (97% after 16 h and 54% after 2 h of growth). Considering the strong adhesion of dual-species biofilms and their easier more reproducible growth compared with biofilms grown from whole saliva, dual-species biofilms of A. naeslundii and S. oralis are suggested to be preferred for use in mechanical plaque removal studies in vitro.


Subject(s)
Biofilms/growth & development , Dental Pellicle/microbiology , Dental Plaque/microbiology , Dental Plaque/therapy , Models, Biological , Toothbrushing/methods , Actinomyces/growth & development , Analysis of Variance , Bacterial Adhesion , Female , Humans , Male , Saliva/microbiology , Sonication , Streptococcus mutans/growth & development , Streptococcus oralis/growth & development , Toothbrushing/instrumentation
17.
J Bacteriol ; 191(22): 6804-11, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19749049

ABSTRACT

Porphyromonas gingivalis is present in dental plaque as early as 4 h after tooth cleaning, but it is also associated with periodontal disease, a late-developing event in the microbial successions that characterize daily plaque development. We report here that P. gingivalis ATCC 33277 is remarkable in its ability to interact with a variety of initial, early, middle, and late colonizers growing solely on saliva. Integration of P. gingivalis into multispecies communities was investigated by using two in vitro biofilm models. In flow cells, bacterial growth was quantified using fluorescently conjugated antibodies against each species, and static biofilm growth on saliva-submerged polystyrene pegs was analyzed by quantitative real-time PCR using species-specific primers. P. gingivalis could not grow as a single species or together with initial colonizer Streptococcus oralis but showed mutualistic growth when paired with two other initial colonizers, Streptococcus gordonii and Actinomyces oris, as well as with Veillonella sp. (early colonizer), Fusobacterium nucleatum (middle colonizer), and Aggregatibacter actinomycetemcomitans (late colonizer). In three-species flow cells, P. gingivalis grew with Veillonella sp. and A. actinomycetemcomitans but not with S. oralis and A. actinomycetemcomitans. Also, it grew with Veillonella sp. and F. nucleatum but not with S. oralis and F. nucleatum, indicating that P. gingivalis and S. oralis are not compatible. However, P. gingivalis grew in combination with S. gordonii and S. oralis, demonstrating its ability to overcome the incompatibility when cultured with a second initially colonizing species. Collectively, these data help explain the observed presence of P. gingivalis at all stages of dental plaque development.


Subject(s)
Biofilms/growth & development , Porphyromonas gingivalis/growth & development , Actinomyces/growth & development , Aggregatibacter actinomycetemcomitans/growth & development , Dental Enamel/microbiology , Fusobacterium nucleatum/growth & development , Humans , Polymerase Chain Reaction , Saliva/microbiology , Streptococcus gordonii/growth & development , Streptococcus oralis/growth & development , Veillonella/growth & development
18.
Appl Environ Microbiol ; 75(10): 3250-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19286780

ABSTRACT

Formation of dental plaque is a developmental process involving initial and late colonizing species that form polymicrobial communities. Fusobacteria are the most numerous gram-negative bacteria in dental plaque, but they become prevalent after the initial commensal colonizers, such as streptococci and actinomyces, have established communities. The unusual ability of these bacteria to coaggregate with commensals, as well as pathogenic late colonizers, has been proposed to facilitate colonization by the latter organisms. We investigated the integration of Fusobacterium nucleatum into multispecies communities by employing two in vitro models with saliva as the sole nutritional source. In flow cell biofilms, numbers of cells were quantified using fluorescently conjugated antibodies against each species, and static biofilms were analyzed by quantitative real-time PCR (q-PCR) using species-specific primers. Unable to grow as single-species biofilms, F. nucleatum grew in two-species biofilms with Actinomyces naeslundii but not with Streptococcus oralis. However, enhanced growth of fusobacteria was observed in three-species biofilms, indicating that there was multispecies cooperation. Importantly, these community dynamics yielded an 18-fold increase in the F. nucleatum biomass between 4 h and 18 h in the flow cell inoculated with three species. q-PCR analysis of static biofilms revealed that maximum growth of the three species occurred at 24 h to 36 h. Lower numbers of cells were observed at 48 h, suggesting that saliva could not support higher cell densities as the sole nutrient. Integration of F. nucleatum into multispecies commensal communities was evident from the interdigitation of fusobacteria in coaggregates with A. naeslundii and S. oralis and from the improved growth of fusobacteria, which was dependent on the presence of A. naeslundii.


Subject(s)
Actinomyces/growth & development , Fusobacterium nucleatum/growth & development , Saliva/microbiology , Streptococcus oralis/growth & development , Biofilms/growth & development , Biomass , Colony Count, Microbial/methods , Dental Plaque/microbiology , Microscopy, Confocal
19.
Probiotics Antimicrob Proteins ; 11(4): 1219-1226, 2019 12.
Article in English | MEDLINE | ID: mdl-30535674

ABSTRACT

Changes in bacterial composition of nasal microbiota may alter the host's susceptibility to several infectious and allergic diseases such as chronic rhinosinusitis and allergic rhinitis. The aim of this study was to evaluate the effects of 1-week administration of a probiotic product, composed by a combination of Streptococcus salivarius 24SMBc and Streptococcus oralis 89a, on the nostril microbiota. Differences in the nasal microbiota composition were investigated by using a next-generation sequencing approach. A strong and significant decrease in Staphylococcus aureus abundance was detected immediately after the bacterial administration. Moreover, comparing the microbial networks of nostril microbiota before and 1 month after the end of treatment, we detected an increase in the total number of both bacterial nodes and microbial correlations, with particular regard to the beneficial ones. Furthermore, a less abundance of microbial genera commonly associated to potential harmful bacteria has been observed. These results suggest a potential ability of S. salivarius 24SMBc and S. oralis 89a to regulate and reorganize the nasal microbiota composition, possibly favoring those microorganisms that may be able to limit the overgrowth of potential pathogens.


Subject(s)
Microbiota , Nose/microbiology , Streptococcus oralis/physiology , Streptococcus salivarius/physiology , Administration, Intranasal , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Female , Humans , Male , Probiotics/administration & dosage , Streptococcus oralis/growth & development , Streptococcus salivarius/growth & development
20.
Colloids Surf B Biointerfaces ; 173: 392-399, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30317126

ABSTRACT

The extracellular polymer substances (EPS) generated by biofilms confers resistance to antimicrobial agents through electrostatic and steric interactions that hinder molecular diffusion. This resistance mechanism is particularly evident for antibacterial nanomaterials, which inherently diffuse more slowly compared to small organic antibacterial agents. The aim of this study was to determine if a biofilm's resistance to antibacterial nanomaterial diffusion could be diminished using electrolytes to screen the EPS's electrostatic interactions. Anionic (+) alpha-tocopherol phosphate (α-TP) liposomes were used as the antimicrobial nanomaterials in the study. They self-assembled into 700 nm sized structures with a zeta potential of -20 mV that were capable of killing oral bacteria (S. oralis growth inhibition time of 3.34 ± 0.52 h). In a phosphate (-ve) buffer the -ve α-TP liposomes did not penetrate multispecies oral biofilms, but in a Tris (hydroxymethyl)aminomethane (+ve) buffer they did (depth - 12.4 ± 3.6 µm). The Tris did not modify the surface charge of the α-TP nanomaterials, rather it facilitated the α-TP-biofilm interactions through electrolyte screening (Langmuir modelled surface pressure increase of 2.7 ± 1.8 mN/ m). This data indicated that EPS resistance was mediated through charge repulsion and that this effect could be diminished through the co-administration of cationic electrolytes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Electrolytes/chemistry , Nanostructures/chemistry , Streptococcus oralis/drug effects , alpha-Tocopherol/analogs & derivatives , Anti-Bacterial Agents/chemistry , Biofilms/growth & development , Buffers , Drug Resistance, Bacterial/drug effects , Extracellular Polymeric Substance Matrix/chemistry , Liposomes/chemistry , Particle Size , Permeability , Static Electricity , Streptococcus oralis/chemistry , Streptococcus oralis/growth & development , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL