Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 455
Filter
1.
BMC Oral Health ; 24(1): 525, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702623

ABSTRACT

OBJECTIVE: To evaluate the antibacterial effectiveness of a combination of ε-poly-L-lysine (ε-PL), funme peptide (FP) as well as domiphen against oral pathogens, and assess the efficacy of a BOP® mouthwash supplemented with this combination in reducing halitosis and supragingival plaque in a clinical trial. MATERIALS AND METHODS: The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the compound against Fusobacterium nucleatum, Porphyromonas gingivalis, Streptococcus mutans, and Aggregatibacter actinomycetemcomitans were determined by the gradient dilution method. Subsequently, the CCK-8 assay was used to detect the toxicity of mouthwash on human gingival fibroblastst, and the effectiveness in reducing halitosis and supragingival plaque of the mouthwash supplemented with the combination was analyzed by a randomized, double-blind, parallel-controlled clinical trial. RESULTS: The combination exhibited significant inhibitory effects on tested oral pathogens with the MIC < 1.56% (v/v) and the MBC < 3.13% (v/v), and the mouthwash containing this combination did not inhibit the viability of human gingival fibroblasts at the test concentrations. The clinical trial showed that the test group displayed notably lower volatile sulfur compounds (VSCs) at 0, 10, 24 h, and 7 d post-mouthwash (P < 0.05), compared with the baseline. After 7 days, the VSC levels of the and control groups were reduced by 50.27% and 32.12%, respectively, and notably cutting severe halitosis by 57.03% in the test group. Additionally, the Plaque Index (PLI) of the test and control group decreased by 54.55% and 8.38%, respectively, and there was a significant difference in PLI between the two groups after 7 days (P < 0.01). CONCLUSIONS: The combination of ε-PL, FP and domiphen demonstrated potent inhibitory and bactericidal effects against the tested oral pathogens, and the newly formulated mouthwash added with the combination exhibited anti-dental plaque and anti-halitosis properties in a clinical trial and was safe. TRIAL REGISTRATION: The randomized controlled clinical trial was registered on Chinese Clinical Trial Registry (No. ChiCTR2300073816, Date: 21/07/2023).


Subject(s)
Dental Plaque , Halitosis , Mouthwashes , Polylysine , Humans , Halitosis/prevention & control , Halitosis/drug therapy , Halitosis/microbiology , Mouthwashes/therapeutic use , Dental Plaque/microbiology , Dental Plaque/prevention & control , Double-Blind Method , Male , Female , Polylysine/therapeutic use , Adult , Microbial Sensitivity Tests , Young Adult , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Porphyromonas gingivalis/drug effects , Fusobacterium nucleatum/drug effects , Fibroblasts/drug effects , Peptides/therapeutic use , Peptides/pharmacology , Aggregatibacter actinomycetemcomitans/drug effects , Streptococcus mutans/drug effects
2.
Front Cell Infect Microbiol ; 14: 1368684, 2024.
Article in English | MEDLINE | ID: mdl-38779565

ABSTRACT

Introduction: Quorum-quenching enzyme Est816 hydrolyzes the lactone rings of N-acyl homoserine lactones, effectively blocking the biofilm formation and development of Gram-negative bacteria. However, its applications in the oral field is limited. This study aimed to evaluate the efficacy of enzyme Est816 in combination with antibiotics against periodontitis induced by Aggregatibacter actinomycetemcomitans in vitro and in vivo. Methods: The antimicrobial efficacy of enzyme Est816 in combination with minocycline, metronidazole, and amoxicillin was determined using the minimum inhibitory concentration test. The anti-biofilm effect of enzyme Est816 was assessed using scanning electron microscopy, live/dead bacterial staining, crystal violet staining, and real-time quantitative PCR. Biocompatibility of enzyme Est816 was assessed in human gingival fibroblasts (HGF) by staining. A rat model of periodontitis was established to evaluate the effect of enzyme Est816 combined with minocycline using micro-computed tomography and histological staining. Results: Compared to minocycline, metronidazole, and amoxicillin treatment alone, simultaneous treatment with enzyme Est816 increased the sensitivity of biofilm bacteria to antibiotics. Enzyme Est816 with minocycline exhibited the highest rate of biofilm clearance and high biocompatibility. Moreover, the combination of enzyme Est816 with antibiotics improved the antibiofilm effects of the antibiotics synergistically, reducing the expression of the virulence factor leukotoxin gene (ltxA) and fimbria-associated gene (rcpA). Likewise, the combination of enzyme Est816 with minocycline exhibited a remarkable inhibitory effect on bone resorption and inflammation damage in a rat model of periodontitis. Discussion: The combination of enzyme Est816 with antibiotics represents a prospective anti-biofilm strategy with the potential to treat periodontitis.


Subject(s)
Aggregatibacter actinomycetemcomitans , Anti-Bacterial Agents , Biofilms , Disease Models, Animal , Metronidazole , Microbial Sensitivity Tests , Periodontitis , Quorum Sensing , Animals , Aggregatibacter actinomycetemcomitans/drug effects , Biofilms/drug effects , Anti-Bacterial Agents/pharmacology , Periodontitis/drug therapy , Periodontitis/microbiology , Rats , Humans , Metronidazole/pharmacology , Quorum Sensing/drug effects , Minocycline/pharmacology , Amoxicillin/pharmacology , Rats, Sprague-Dawley , Male , Fibroblasts/drug effects , Gingiva/microbiology
3.
Oral Health Prev Dent ; 22: 171-180, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687029

ABSTRACT

PURPOSE: To investigate the microbiological outcomes obtained with either subgingival debridement (SD) in conjunction with a gel containing sodium hypochlorite and amino acids followed by subsequent application of a cross-linked hyaluronic acid gel (xHyA) gel, or with SD alone. MATERIALS AND METHODS: Forty-eight patients diagnosed with stages II-III (grades A/B) generalised periodontitis were randomly treated with either SD (control) or SD plus adjunctive sodium hypochlorite/amino acids and xHyA gel (test). Subgingival plaque samples were collected from the deepest site per quadrant in each patient at baseline and after 3 and 6 months. Pooled sample analysis was performed using a multiplex polymerase chain reaction (PCR)-based method for the identification of detection frequencies and changes in numbers of the following bacteria: Aggregatibacter actinomycetemcomitans (A.a), Porphyromonas gingivalis (P.g), Tannerella forsythia (T.f), Treponema denticola (T.d), and Prevotella intermedia (P.i). RESULTS: In terms of detection frequency, in the test group, statistically significant reductions were found for P.g, T.f, T.d and P.i (p < 0.05) after 6 months. In the control group, the detection frequencies of all investigated bacterial species at 6 months were comparable to the baseline values (p > 0.05). The comparison of the test and control groups revealed statistically significant differences in detection frequency for P.g (p = 0.034), T.d (p < 0.01) and P.i (p = 0.02) after 6 months, favouring the test group. Regarding reduction in detection frequency scores, at 6 months, statistically significant differences in favour of the test group were observed for all investigated bacterial species: A.a (p = 0.028), P.g (p = 0.028), T.f (p = 0.004), T.d (p <0.001), and P.i (p = 0.003). CONCLUSIONS: The present microbiological results, which are related to short-term outcomes up to 6 months post-treatment, support the adjunctive subgingival application of sodium hypochlorite/amino acids and xHyA to subgingival debridement in the treatment of periodontitis.


Subject(s)
Aggregatibacter actinomycetemcomitans , Amino Acids , Dental Plaque , Hyaluronic Acid , Porphyromonas gingivalis , Prevotella intermedia , Sodium Hypochlorite , Tannerella forsythia , Treponema denticola , Humans , Hyaluronic Acid/therapeutic use , Sodium Hypochlorite/therapeutic use , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/isolation & purification , Porphyromonas gingivalis/drug effects , Female , Middle Aged , Male , Prevotella intermedia/drug effects , Tannerella forsythia/drug effects , Treponema denticola/drug effects , Adult , Dental Plaque/microbiology , Amino Acids/therapeutic use , Periodontal Debridement/methods , Bacterial Load/drug effects , Gels , Combined Modality Therapy , Follow-Up Studies , Cross-Linking Reagents/therapeutic use , Periodontal Pocket/microbiology , Periodontal Pocket/therapy , Periodontitis/microbiology , Periodontitis/therapy , Periodontitis/drug therapy
4.
Colloids Surf B Biointerfaces ; 238: 113870, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555763

ABSTRACT

Antibiotic resistance has become an urgent threat to health care in recent years. The use of drug delivery systems provides advantages over conventional administration of antibiotics and can slow the development of antibiotic resistance. In the current study, we developed a toxin-triggered liposomal antibiotic delivery system, in which the drug release is enabled by the leukotoxin (LtxA) produced by the Gram-negative pathogen, Aggregatibacter actinomycetemcomitans. LtxA has previously been shown to mediate membrane disruption by promoting a lipid phase change in nonlamellar lipids, such as 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-methyl (N-methyl-DOPE). In addition, LtxA has been observed to bind strongly and nearly irreversibly to membranes containing large amounts of cholesterol. Here, we designed a liposomal delivery system composed of N-methyl-DOPE and cholesterol to take advantage of these interactions. Specifically, we hypothesized that liposomes composed of N-methyl-DOPE and cholesterol, encapsulating antibiotics, would be sensitive to LtxA, enabling controlled antibiotic release. We observed that liposomes composed of N-methyl-DOPE were sensitive to the presence of low concentrations of LtxA, and cholesterol increased the extent and kinetics of content release. The liposomes were stable under various storage conditions for at least 7 days. Finally, we showed that antibiotic release occurs selectively in the presence of an LtxA-producing strain of A. actinomycetemcomitans but not in the presence of a non-LtxA-expressing strain. Together, these results demonstrate that the designed liposomal vehicle enables toxin-triggered delivery of antibiotics to LtxA-producing strains of A. actinomycetemcomitans.


Subject(s)
Aggregatibacter actinomycetemcomitans , Anti-Bacterial Agents , Liposomes , Liposomes/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Aggregatibacter actinomycetemcomitans/drug effects , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Drug Liberation , Cholesterol/chemistry , Cholesterol/metabolism , Microbial Sensitivity Tests , Exotoxins/metabolism , Exotoxins/chemistry , Phosphatidylethanolamines/chemistry , Drug Delivery Systems
5.
J Infect Dev Ctries ; 15(11): 1685-1693, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34898497

ABSTRACT

INTRODUCTION: Development of bacterial resistance and antimicrobial side-effect has shifted the focus of research toward Ethnopharmacology. A biologically active compound derived from the plants may increase the effectiveness of antibiotic when used in combination. The present study aims to determine the synergistic antibacterial effect of ethanolic extracts of Punica granatum (pericarp), Commiphora molmol, Azadirachta indica (bark) in combination with amoxicillin, metronidazole, tetracycline, and azithromycin on periodontopathic bacteria: Porphyromonas gingivalis, Tannerella forsythia, Treponema denticola and Aggregatibacter actinomycetemcomitans. METHODOLOGY: Periodontopathic bacterial strains were isolated from the plaque sample that was collected from periodontitis patients and grown under favorable conditions. Susceptibility of bacteria to the antibiotics and extracts was determined by disc diffusion method by measuring the diameter of the inhibition zones. Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of plant extracts were evaluated against each bacterium. Synergistic effect of plant extract in combination with antibiotics was tested against each bacterium by measuring the diameter of zone of inhibition (ZOI). RESULTS: Findings revealed that all plant extracts exhibited an inhibitory effects on the proliferation and growth of periodontopathic bacteria. The maximum antibacterial effect was exhibited by C. molmol on P. gingivalis (ZOI = 20 ± 0.55 mm, MIC = 0.53 ± 0.24 mg/mL and MBC = 5.21 ± 1.81 mg/mL) (p < 0.05), meanwhile, no antibacterial activity was exhibited by P. granatum on T. forsythia. Synergistic antibacterial effect was recorded when plant extracts were used in combination with antibiotics. The best synergism was exhibited by P. granatum with amoxicillin against A. actinomycetemcomitans (24 ± 1.00 mm) (p < 0.05). CONCLUSIONS: The synergistic test showed significant antibacterial activity when plant extracts were combined with antibiotics against all the experimented bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Periodontitis/microbiology , Plant Extracts/pharmacology , Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/therapeutic use , Drug Therapy, Combination , Humans , Microbial Sensitivity Tests , Periodontitis/drug therapy , Plant Extracts/therapeutic use , Porphyromonas gingivalis/drug effects , Tannerella forsythia/drug effects
6.
Food Funct ; 12(21): 10470-10483, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34554173

ABSTRACT

Cranberries are widely recognized as a functional food that can promote oral health. However, the high concentration of organic acids in cranberry juice can cause tooth enamel erosion. Electrodialysis with bipolar membrane (EDBM) is a process used for the deacidification of cranberry juice. The present study investigated whether the removal of organic acids (0%, 19%, 42%, 60%, and 79%) from cranberry juice by EDBM affects its antibacterial activity against major periodontopathogens as well as its anti-inflammatory properties in an oral epithelial cell model. A deacidification rate ≥60% attenuated the bactericidal effect against planktonic and biofilm-embedded Aggregatibacter actinomycetemcomitans but had no impact on Porphyromonas gingivalis and Fusobacterium nucleatum. Cranberry juice increased the adherence of A. actinomycetemcomitans and P. gingivalis to oral epithelial cells, but reduced the adherence of F. nucleatum by half regardless of the deacidification rate. F. nucleatum produced more hydrogen sulfide when it was exposed to deacidified cranberry juice with a deacidification rate ≥42% compared to the raw beverage. Interestingly, the removal of organic acids from cranberry juice lowered the cytotoxicity of the beverage for oral epithelial cells. Deacidification attenuated the anti-inflammatory effect of cranberry juice in an in vitro oral epithelial cell model. The secretion of IL-6 by lipopolysaccharide (LPS)-stimulated oral epithelial cells exposed to cranberry juice increased proportionally with the deacidification rate. No such effect was observed with respect to the production of IL-8. This study provided evidence that organic acids, just like phenolic compounds, might contribute to the health benefits of cranberry juice against periodontitis.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Fusobacterium nucleatum/drug effects , Plant Extracts/pharmacology , Porphyromonas gingivalis/drug effects , Vaccinium macrocarpon/chemistry , Acids/chemistry , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Biofilms , Cells, Cultured , Electrochemical Techniques/methods , Epithelial Cells/drug effects , Fruit and Vegetable Juices , Plant Extracts/chemistry
7.
J Pharm Pharmacol ; 73(4): 505-514, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33793838

ABSTRACT

OBJECTIVES: We and others have previously shown that epigallocatechin gallate (EGCg) inhibits the activity of an important virulence factor, leukotoxin (LtxA), produced by the oral bacterium Aggregatibacter actinomycetemcomitans, suggesting the potential use of this molecule as an anti-virulence strategy to treat periodontal infections. Here, we sought to better understand the effects of EGCg on toxin secretion and A. actinomycetemcomitans pathogenicity in a co-culture model. METHODS: We used a quantitative immunoblot assay to determine the concentrations of LtxA in the bacterial supernatant and on the bacterial cell surface. Using a co-culture model, consisting of A. actinomycetemcomitans and THP-1 cells, we studied the impact of EGCg-mediated changes in LtxA secretion on the toxicity of A. actinomycetemcomitans. KEY FINDINGS: EGCg increased production of LtxA and changed the localization of secreted LtxA from the supernatant to the surface of the bacterial cells. In the co-culture model, a single low dose of EGCg did not protect host THP-1 cells from A. actinomycetemcomitans-mediated cytotoxicity, but a multiple dosing strategy had improved effects. CONCLUSIONS: Together, these results demonstrate that EGCg has important, but complicated, effects on toxin secretion and activity; new dosing strategies and comprehensive model systems may be required to properly develop these anti-virulence activities.


Subject(s)
Aggregatibacter actinomycetemcomitans , Catechin/analogs & derivatives , Exotoxins , Periodontitis , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/metabolism , Aggregatibacter actinomycetemcomitans/pathogenicity , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Catechin/pharmacology , Coculture Techniques/methods , Dose-Response Relationship, Drug , Exotoxins/antagonists & inhibitors , Exotoxins/metabolism , Humans , Periodontitis/drug therapy , Periodontitis/microbiology , Virulence/drug effects
8.
Int J Biol Macromol ; 173: 99-108, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33460660

ABSTRACT

The present investigation reports an in-vitro study using combination of laccase and an enhancer capable of inhibiting the growth of pathogenic microorganisms, preventing biofilm formation, and whitening teeth. Laccase-cinnamic acid system remarkably inhibited the growth of Aggregatibacter actinomycetemcomitans, Candida albicans, S. aureus, and Streptococcus mutans whilst showed no significant effects on Gram-negative bacteria. Data presented that cinnamic acid (10 mM) with laccase (0.125 U ml-1) led to a maximum decrease of about 90%, in S. mutans biofilm formation. The confocal laser scanning microscopy showed considerable detachment of S. mutans cells from glass substratum. The combined laccase-cinnamic acid system could remove teeth discoloration caused by coffee. SEM of the teeth surface exhibited no damages such as surface cracking or fracture. Liquid chromatography-tandem mass spectrometry (LC-MS) and cyclic voltammetry (CV) studies showed that laccase can catalyze the one-electron oxidation of cinnamic acid to the respective radical. This radical can then undergo several fates, including recombination with another radical to form a dimeric species, dismutation of the radical back to cinnamic acid or decarboxylation to give various reduced oxygen species. Therefore, the redox potential values of phenolic monomers/oligomers are related with their biological activities.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Cinnamates/pharmacology , Fungal Proteins/pharmacology , Hericium/chemistry , Laccase/pharmacology , Aggregatibacter actinomycetemcomitans/growth & development , Biofilms/drug effects , Biofilms/growth & development , Caffeic Acids/pharmacology , Candida albicans/drug effects , Candida albicans/growth & development , Catechols/pharmacology , Drug Synergism , Escherichia coli/drug effects , Escherichia coli/growth & development , Fungal Proteins/isolation & purification , Gallic Acid/pharmacology , Hericium/enzymology , Hydroquinones/pharmacology , Laccase/isolation & purification , Lactobacillus/drug effects , Lactobacillus/growth & development , Microbial Sensitivity Tests , Oxidation-Reduction , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Tooth Bleaching Agents/pharmacology
9.
J Biomed Mater Res A ; 109(5): 713-721, 2021 05.
Article in English | MEDLINE | ID: mdl-32654391

ABSTRACT

The prevalence of periodontal disease poses a significant global health burden. Treatments for these diseases, primarily focused on removal and eradication of dental plaque biofilms, are challenging due to limited access to periodontal pockets where these oral pathogens reside. Herein, we report on the development and characterization of nitric oxide (NO)-releasing carboxymethylcellulose (CMC) derivatives and evaluate their in vitro bactericidal efficacy against planktonic Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans, two prominent periodontopathogens. Bactericidal exposure assays revealed that three of the synthesized NO-releasing polymers were capable of reducing bacterial viability of both species by 99.9% in 2 hr at concentrations of 4 mg ml-1 or lower, reflecting NO's potent and rapid bactericidal action. The NO-releasing CMCs elicited minimal toxicity to human gingival fibroblasts at their bactericidal concentrations following 24-hr exposure.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Azo Compounds/pharmacology , Carboxymethylcellulose Sodium , Ethanolamines/pharmacology , Nitric Oxide/pharmacology , Periodontal Diseases/microbiology , Polyamines/pharmacology , Porphyromonas gingivalis/drug effects , Propylamines/pharmacology , Anti-Bacterial Agents/administration & dosage , Azo Compounds/administration & dosage , Azo Compounds/chemistry , Biopolymers , Cell Line , Diamines/chemistry , Drug Carriers , Drug Delivery Systems , Ethanolamines/administration & dosage , Ethanolamines/chemistry , Fibroblasts/drug effects , Gingiva/cytology , Humans , Molecular Structure , Nitric Oxide/administration & dosage , Nitric Oxide/toxicity , Polyamines/administration & dosage , Polyamines/chemistry , Propylamines/administration & dosage , Propylamines/chemistry , Species Specificity , Viscosity
10.
Arch Oral Biol ; 122: 104989, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33249361

ABSTRACT

OBJECTIVE: To determine the mechanism of growth inhibition of Aggregatibacter actinomycetemcomitans by Maillard reaction products (MRP). DESIGN: Growth and cell viabilities in the presence or absence of MRP were measured for both the rough and smooth variants of the bacteria. Effects of addition of ferrous and ferric ions on the inhibition of the bacteria by MRP were determined. RESULTS: MRPs decreased the extent of complex formation of Chrome Azurol S with iron suggesting that MRPs can chelate iron effectively. The chelation causes growth inhibition of both the rough and smooth strains. At low concentrations of the inhibitor, lag time was extended by approximately 12 h while at high concentrations, cells were killed, decreasing cell viability by up to 8 orders of magnitude. Growth of both the rough and smooth strains could be restored to original level by addition of iron. For the rough strain, both ferrous and ferric ions could relieve the inhibition by MRP while for the smooth strain only ferrous ion was effective. CONCLUSION: MRPs inhibit the growth of A. actinomycetemcomitans by chelating iron and the inhibition can be relieved by addition of iron.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Chelating Agents/pharmacology , Glycation End Products, Advanced , Iron/pharmacology , Aggregatibacter actinomycetemcomitans/growth & development , Hydroxybenzoates/pharmacology , Maillard Reaction
11.
J Biomater Appl ; 35(6): 643-654, 2021 01.
Article in English | MEDLINE | ID: mdl-33045872

ABSTRACT

Dental implants are the most innovative and superior treatment modality for tooth replacement. However, titanium implants still suffer from insufficient antibacterial capability and peri-implant diseases remain one of the most common and intractable complications. To prevent peri-implant diseases, a composite coating containing a new antibacterial agent, (Z-)-4-bromo-5-(bromomethylene)-2(5H)-furanone (BBF) was fabricated on titanium. This study was designed to investigate the antibacterial activity of the composite coating against two common peri-implant pathogens (Porphyromonas gingivalis and Actinobacillus actinomycetemcomitans). The morphology of the composite coating showed that BBF-loaded poly(L-lactic acid) nanospheres were well-distributed in the pores of the microarc oxidation coating, and cross-linked with each other and the wall pores by gelatin. A release study indicated that the antibacterial coating could sustain the release of BBF for 60 d, with a slight initial burst release occurring during the first 4 h. The antibacterial rate of the composite coating for adhering bacteria was the highest (over 97%) after 1 d and over 90% throughout a 30-day incubation period. The total fluorescence intensity of the composite coating was the lowest, and the vast majority of the fluorescence was red (dead bacteria). Moreover, real-time polymerase chain reaction analysis confirmed that the relative gene expression of the adherent bacteria on the composite coating was down-regulated. It was therefore concluded that the composite coating fabricated on titanium, which showed excellent and relatively long-term antibacterial activity against Porphyromonas gingivalis and Actinobacillus actinomycetemcomitans, is a potential and promising strategy to be applied on dental implants for the prevention of peri-implant diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Coated Materials, Biocompatible/pharmacology , Dental Implants , Furans/pharmacology , Nanoparticles/chemistry , Polyesters/chemistry , Titanium/chemistry , Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/chemistry , Coated Materials, Biocompatible/chemistry , Cross-Linking Reagents/chemistry , Drug Liberation , Furans/chemistry , Gene Expression Regulation/drug effects , Humans , Oxidation-Reduction , Porphyromonas gingivalis/drug effects
12.
Mol Med Rep ; 22(5): 4143-4150, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33000248

ABSTRACT

Periodontitis is a chronic inflammatory disease caused by various periodontal pathogens. Weissella cibaria CMU (oraCMU) is a probiotic that promotes oral health. However, its anti­inflammatory effects against periodontal pathogens have not yet been investigated. The present study evaluated the anti­inflammatory effects of live oraCMU against stimulation with the formalin­inactivated periodontal pathogen Aggregatibacter actinomycetemcomitans in RAW 264.7 macrophages. Cell viability was analyzed by the MTS assay in a dose­dependent manner (at multiplicities of infection of 0.1, 1, 10, 100 and 1,000). Nitric oxide (NO) was monitored using the Griess test. The mRNA expression of proinflammatory cytokines such as interleukin (IL)1ß and IL6 was assessed by reverse transcription­quantitative PCR. Western blotting was used to examine the effects of oraCMU on the phosphorylation of NF­κB inhibitor α (IκBα) and IκBα kinase (IKK), the nuclear translocation of the NF­κB subunit p65 and the expression of inducible NO synthase (iNOS). Live oraCMU had no cytotoxic effects on RAW 264.7 macrophages. In A. actinomycetemcomitans­stimulated RAW 264.7 macrophages, oraCMU reduced NO production by suppressing iNOS expression and downregulating the mRNA expression of proinflammatory cytokines in a dose­dependent manner. IKK phosphorylation and IκBα degradation were dose­dependently inhibited by oraCMU and the nuclear translocation of p65 via the canonical NF­κB pathway was simultaneously reduced. The results indicated that oraCMU possessed anti­inflammatory activity associated with the inhibition of NF­κB signal activation in response to periodontal pathogens. This suggests that oraCMU is a beneficial anti­inflammatory probiotic that can aid in the maintenance of oral health.


Subject(s)
Aggregatibacter actinomycetemcomitans/pathogenicity , Anti-Inflammatory Agents/pharmacology , Cytokines/genetics , Macrophages/cytology , Probiotics/pharmacology , Weissella/physiology , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/immunology , Animals , Cell Survival , Dose-Response Relationship, Drug , Formaldehyde/adverse effects , Gene Expression Regulation/drug effects , Interleukin-1beta/genetics , Interleukin-6/genetics , Macrophages/immunology , Mice , NF-kappa B/metabolism , Phosphorylation , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Biomed Res Int ; 2020: 7289208, 2020.
Article in English | MEDLINE | ID: mdl-32908908

ABSTRACT

Periodontal diseases are mainly the results of infections and inflammation of the gum and bone that surround and support the teeth. In this study, the alveolar bone destruction in periodontitis is hypothesized to be treated with novel Mg-Cu alloy grafts due to their antimicrobial and osteopromotive properties. In order to study this new strategy using Mg-Cu alloy grafts as a periodontal bone substitute, the in vitro degradation and antibacterial performance were examined. The pH variation and Mg2+ and Cu2+ release of Mg-Cu alloy extracts were measured. Porphyromonas gingivalis (P. gingivalis) and Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans), two common bacteria associated with periodontal disease, were cultured in Mg-Cu alloy extracts, and bacterial survival rate was evaluated. The changes of bacterial biofilm and its structure were revealed by scanning electron microscopy (SEM) and transmission electronic microscopy (TEM), respectively. The results showed that the Mg-Cu alloy could significantly decrease the survival rates of both P. gingivalis and A. actinomycetemcomitans. Furthermore, the bacterial biofilms were completely destroyed in Mg-Cu alloy extracts, and the bacterial cell membranes were damaged, finally leading to bacterial apoptosis. These results indicate that the Mg-Cu alloy can effectively eliminate periodontal pathogens, and the use of Mg-Cu in periodontal bone grafts has a great potential to prevent infections after periodontal surgery.


Subject(s)
Alloys/pharmacology , Anti-Bacterial Agents/pharmacology , Bone Transplantation/adverse effects , Copper/pharmacology , Magnesium/physiology , Periodontal Diseases/drug therapy , Periodontitis/drug therapy , Aggregatibacter actinomycetemcomitans/drug effects , Biofilms/drug effects , Humans , Periodontal Diseases/microbiology , Periodontics/methods , Periodontitis/microbiology , Porphyromonas gingivalis/drug effects
14.
Sci Rep ; 10(1): 8553, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444858

ABSTRACT

This study investigated the bactericidal effect, the underlying mechanisms of treatment, and recovery of biocompatibility of the infected titanium surface using a combination treatment of silver ion application and ultraviolet-A (UV-A) light irradiation. Streptococcus mutans and Aggregatibacter actinomycetemcomitans were used in suspension and as a biofilm on a titanium surface to test for the bactericidal effect. The bactericidal effect of the combination treatment was significantly higher than that of silver ion application or UV-A light irradiation alone. The bactericidal effect of the combination treatment was attributable to hydroxyl radicals, which generated from the bacterial cell wall and whose yield increased with the silver concentration. To assess the biocompatibility, proliferation and calcification of MC3T3E1 cells were evaluated on the treated titanium surface. The treated titanium screws were implanted into rat tibias and the removal torques were measured 28 days post-surgery. The titanium surface that underwent the combination treatment exhibited recovery of biocompatibility by allowing cellular proliferation or calcification at levels observed in the non-infected titanium surfaces. The removal torque 28 days after surgery was also comparable to the control values. This approach is a novel treatment option for peri-implantitis.


Subject(s)
Aggregatibacter actinomycetemcomitans/growth & development , Anti-Bacterial Agents/administration & dosage , Biofilms/growth & development , Hydroxyl Radical/chemistry , Pasteurellaceae Infections/prevention & control , Silver/administration & dosage , Streptococcus mutans/growth & development , Titanium/chemistry , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/radiation effects , Animals , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Biofilms/radiation effects , Mice , Pasteurellaceae Infections/microbiology , Peri-Implantitis/microbiology , Peri-Implantitis/therapy , Rats , Rats, Wistar , Silver/chemistry , Streptococcus mutans/drug effects , Streptococcus mutans/radiation effects , Tibia/microbiology , Tibia/surgery , Ultraviolet Rays
15.
Colloids Surf B Biointerfaces ; 193: 111074, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32416519

ABSTRACT

Bacteria easily adhere, colonize, and form biofilm on oral implants subsequently causing periimplantation periarthritis and mechanical loosening. Previous studies show that a high potential surface on polymeric implants can achieve surface bacteriostasis without side effects. In this study, a high surface potential is introduced to zirconia ceramics to mitigate bacterial infection. Carbon and nitrogen plasma immersion ion implantation (C-PIII and N-PIII) are conducted on zirconia ceramic samples sequentially to elevate the surface potential. The surface with a high potential but without ion leaching exhibits excellent antibacterial effects against oral bacteria and little bacterial resistance is triggered. The surface also has high strength and excellent biocompatibility. The nitrogen-containing inorganic structure with high potential can actualize bacteriostasis and biocompatibility on zirconia ceramics simultaneously and this new strategy can enhance the antibacterial ability of oral implants.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biocompatible Materials/pharmacology , Ceramics/pharmacology , Zirconium/pharmacology , Aggregatibacter actinomycetemcomitans/drug effects , Animals , Anti-Bacterial Agents/chemistry , Biocompatible Materials/chemistry , Cells, Cultured , Ceramics/chemistry , Materials Testing , Mice , Microbial Sensitivity Tests , Particle Size , Porphyromonas gingivalis/drug effects , Staphylococcus aureus/drug effects , Streptococcus mutans/drug effects , Surface Properties , Zirconium/chemistry
16.
Drug Des Devel Ther ; 14: 233-242, 2020.
Article in English | MEDLINE | ID: mdl-32021107

ABSTRACT

PURPOSE: Electrospun PLA fiber devices were investigated in the form of fiber mats and disks. Metronidazole was used as an active agent; its concentration was 12.2 and 25.7 wt% in the devices. METHODS: The structure was studied by X-ray diffraction and scanning electron microscopy, drug release by dissolution measurements, while the antimicrobial efficiency was tested on five bacterial strains. RESULTS: The XRD study showed that the polymer was partially crystalline in both devices, but a part of metronidazole precipitated and was in the form of crystals among and within the fibers. Liquid penetration and dissolution were different in the two devices, they were faster in disks and slower in fiber mats, due to the morphology of the device and the action of capillary forces. Disks released the drug much faster than fiber mats. Although the release study indicated fast drug dissolution, the concentration achieved a plateau value in 24 hrs for the disks; the inhibition effect lasted much longer, 13 days for bacteria sensitive to metronidazole. The longer inhibition period could be explained by the slower diffusion of metronidazole located inside the fibers of the device. CONCLUSION: The results suggest that the devices may be effective in the treatment of periodontitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Metronidazole/pharmacology , Periodontal Diseases/drug therapy , Polyesters/chemistry , Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/chemistry , Drug Liberation , Eikenella corrodens/drug effects , Firmicutes/drug effects , Fusobacterium nucleatum/drug effects , Humans , Metronidazole/chemistry , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Particle Size , Periodontal Diseases/microbiology , Prevotella intermedia/drug effects , Surface Properties , X-Ray Diffraction
17.
Anaerobe ; 62: 102153, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31962136

ABSTRACT

For dental implants the accumulation of anaerobic bacteria is a main reason for peri-implant inflammation, which untreated can lead to implant loss. Oxygen releasing substances may act as antibacterial agents. In this study glucose-1-phosphate (Glc-1P) biofunctionalized zinc peroxide (ZnO2) nanoparticles of four different synthesis ratio (1-10:1) and sizes (4-5 nm) were tested against the anaerobes Fusobacterium nucleatum, Porphyromonas gingivalis, and Prevotella intermedia, as well as against Aggregatibacter actinomycetemcomitans, Enterococcus faecalis, Staphylococcus aureus, Lactobacillus paracasei, and the yeast Candida albicans. Nanoparticles stabilized with o-phosphorylethanolamine, bis[2-(methacryloyloxy)ethyl] phosphate, or dioctyl sulfosuccinate instead of glucose were used as controls. For every combination of test strain and nanoparticle both, the minimal inhibitory (MIC) and minimal microbicidal concentration (MBC or MFC) were determined under different pH conditions in microtiter plates. Furthermore, transmission electron (TEM) and fluorescence microscopy after live-dead-staining was performed on selected combinations of pathogen and nanoparticle in order to visualize the interactions. The ZnO2/Glc-1P nanoparticles had an inhibitory effect on gram-negative anaerobes and on A. actinomycetemcomitans with a pH-dependent MIC ≥25 µg/ml and MBC ≥50 µg/ml, while the gram-positive species tested and C. albicans were not inhibited. In TEM images, attachment of nanoparticle-chains to the bacterial outer membrane and subsequent penetration was found together with an intracellular oxygen release. For nanoparticles with other stabilizers than glucose an invasion was only seen in elongated, dividing cells, possibly because of the more porous cell wall in the parting layer. Decorating ZnO2 by glucose-1-phosphate is a Trojan horse approach to permit their uptake in gram-negative oxygen-sensitive bacterial cells.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Nanoparticles/chemistry , Peri-Implantitis/etiology , Peroxides , Zinc , Humans , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Nanoparticles/ultrastructure , Peroxides/chemistry , Sensitivity and Specificity , Zinc/chemistry
18.
Lett Appl Microbiol ; 70(4): 310-317, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31955445

ABSTRACT

Probiotics can stabilize gut flora, regulate intestinal immunity and protect the host from enteric diseases; however, their roles in oral health have received little attention compared to their roles in gut health. Nowadays, the prevalence of sugar-sweetened foods and abuse of antibiotics contribute towards dysbiosis of oral microbiota and drug resistance development in oral pathogens, resulting in various intractable oral diseases. We screened the antibacterial activities of viable and heat-killed probiotic strains against the oral pathogens Streptococcus mutans, Porphyromonas gingivalis, Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans. The probiotic strains Lactobacillus salivarius subsp. salicinius AP-32, L. rhamnosus CT-53, L. paracasei ET-66 and Bifidobacterium animalis subsp. lactis CP-9 displayed strong antipathogenic activities, whereas heat-killed AP-32, CT-53 and ET-66 displayed high levels of pathogen inhibition. The antibacterial activities of these probiotics were not associated with their H2 O2 production; L. acidophilus TYCA02 produced high levels of H2 O2 but merely exhibited moderate antibacterial activities. Oral tablets containing probiotics showed positive inhibitory effects against oral pathogens, particularly those containing viable probiotics. Our results indicate that probiotics prevent the growth of oral pathogens and improve oral health, providing insights into the antipathogenic efficacy of different probiotic species and their potential role in functional foods that improve oral health. SIGNIFICANCE AND IMPACT OF THE STUDY: Our study provides insights into the antipathogenic efficacy of different probiotic species and their potential roles in developing functional foods to improve oral health. We showed that the probiotic strains Lactobacillus salivarius subsp. salicinius AP-32, L. rhamnosus CT-53, L. paracasei ET-66 and Bifidobacterium animalis subsp. lactis CP-9 have great potential for use in the development of functional foods to improve oral health. Since active probiotics may provide strong and long-term protection, the development of functional food products should favour the use of viable bacteria.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Antibiosis , Fusobacterium nucleatum/drug effects , Ligilactobacillus salivarius/physiology , Mouth/microbiology , Porphyromonas gingivalis/drug effects , Probiotics/pharmacology , Streptococcus mutans/physiology , Aggregatibacter actinomycetemcomitans/physiology , Fusobacterium nucleatum/physiology , Humans , Microbiota , Porphyromonas gingivalis/physiology , Streptococcus mutans/drug effects
19.
Braz. arch. biol. technol ; 63: e20190196, 2020. tab, graf
Article in English | LILACS | ID: biblio-1132166

ABSTRACT

Abstract Endodontic infections result from oral pathogenic bacteria which reach and infect dental pulp, as well as surrounding tissues, through cracks, unrepaired caries and failed caries restorations. This study aims to determine the chemical composition of essential oil from Psidium cattleianum leaves (PC-EO) and to assess its antibacterial activity against endodontic bacteria. Antibacterial activity of PC-EO was evaluated in terms of its minimum inhibitory concentration (MIC) values by the broth microdilution method on 96-well microplates. Bacteria Porphyromonas gingivalis (MIC = 20 µg/mL), Prevotella nigrescens (MIC = 62.5 µg/mL), Fusobacterium nucleatum (MIC = 12.5 µg/mL), Actinomyces naeslundii (MIC = 50 µg/mL), Bacteroides fragilis (MIC = 12.5 µg/mL), Aggregatibacter actinomycetemcomitans (MIC = 6.25 µg/mL) and Peptostreptococcus anaerobius (MIC = 62.5 µg/mL) were evaluated and compared to chlorhexidine dihydrochloride (CDH), the positive control. PC-EO was obtained by hydrodistillation with the use of a Clevenger-type apparatus whereas its chemical composition was analyzed by gas chromatography-flame ionization detection (GC-FID) and gas chromatography-mass spectrometry (GC-MS). Viridiflorol (17.9%), β-caryophyllene (11.8%), 1,8-cineole (10.8%) and β-selinene (8.6%) were the major constituents found in PC-EO, which exhibited high antibacterial activity against all endodontic pathogens under investigation. Therefore, PC-EO, a promising source of bioactive compounds, may provide therapeutic solutions for the field of endodontics.


Subject(s)
Oils, Volatile/pharmacology , Chlorhexidine/pharmacology , Psidium/chemistry , Anti-Bacterial Agents/pharmacology , Peptostreptococcus/drug effects , Bacteroides fragilis/drug effects , Actinomyces/drug effects , Microbial Sensitivity Tests , Fusobacterium nucleatum/drug effects , Aggregatibacter actinomycetemcomitans/drug effects , Porphyromonas gingivalis/drug effects , Prevotella nigrescens/drug effects , Gas Chromatography-Mass Spectrometry
20.
BMJ Case Rep ; 12(9)2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31551318

ABSTRACT

A 45-year-old- man presented with left chest wall pain, swelling and cough. Over a 2-month period he developed abscesses in the right foot, right anterior thigh, left buttock and left chest. Incision and drainage of the soft tissue abscesses and video-assisted thoracoscopic surgery to drain the loculated empyema contiguous with the chest wall abscess were performed as surgical management. Gram stain showed beaded Gram-positive rods and the culture initially grew Aggregatibacter actinomycetemcomitans and Eikenella corrodens Pathological evaluation of the pleura showed sulfur granules and organisms consistent with Actinomyces spp. on Gomori methenamine silver stain; Actinomyces israelii was recovered in culture with extended incubation. The patient was treated for 3 weeks with ceftriaxone and oral metronidazole, followed by oral amoxicillin. Culture of A. actinomycetemcomitans with other findings consistent with actinomycosis warrants 6-12 months of antibiotic therapy.


Subject(s)
Abscess/microbiology , Actinomycosis/microbiology , Empyema, Pleural/microbiology , Lung Diseases/microbiology , Pasteurellaceae Infections/microbiology , Skin Diseases, Bacterial/microbiology , Actinomyces/drug effects , Actinomycosis/drug therapy , Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/therapeutic use , Chest Pain , Coinfection/microbiology , Cough , Empyema, Pleural/surgery , Humans , Male , Middle Aged , Pasteurellaceae Infections/drug therapy , Skin Diseases, Bacterial/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...