Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.841
Filter
1.
BMC Oral Health ; 24(1): 763, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965550

ABSTRACT

BACKGROUND: There is insufficient clinical and microbiological evidence to support the use of diode laser and air-polishing with erythritol as supplements to scaling and root planning(SRP). The aim of the current study is to evaluate the clinical and microbiologic efficacy of erythritol subgingival air polishing and diode laser in treatment of periodontitis. METHODS: The study encompassed twenty-four individuals seeking periodontal therapy and diagnosed with stage I and stage II periodontitis. Eight patients simply underwent SRP. Eight more patients had SRP followed by erythritol subgingival air polishing, and eight patients had SRP followed by diode laser application. At baseline and six weeks, clinical periodontal parameters were measured, including Plaque Index (PI), Gingival Index (GI), periodontal Probing Depth (PPD), and Clinical Attachment Level (CAL). The bacterial count of Aggregatibacter actinomycetemcomitans(A.A), Porphyromonas gingivalis (P.G) was evaluated at different points of time. RESULTS: The microbiological assessment revealed significant differences in the count of A.A. between the laser and erythritol groups immediately after treatment, indicating a potential impact on microbial levels. However, the microbial levels showed fluctuations over the subsequent weeks, without statistically significant differences. Plaque indices significantly decreased post-treatment in all groups, with no significant inter-group differences. Gingival indices decreased, and the laser group showed lower values than erythritol and control groups. PPD and CAL decreased significantly across all groups, with the laser group exhibiting the lowest values. CONCLUSION: The supplementary use of diode laser and erythritol air polishing, alongside SRP, represents an expedited periodontal treatment modality. This approach leads to a reduction in bacteria and improvement in periodontal health. TRIAL REGISTRATION: This clinical trial was registered on Clinical Trials.gov (Registration ID: NCT06209554) and released on 08/01/2024.


Subject(s)
Aggregatibacter actinomycetemcomitans , Bacterial Load , Dental Plaque Index , Dental Scaling , Erythritol , Lasers, Semiconductor , Periodontal Index , Porphyromonas gingivalis , Root Planing , Adult , Female , Humans , Male , Middle Aged , Aggregatibacter actinomycetemcomitans/isolation & purification , Aggregatibacter actinomycetemcomitans/drug effects , Air Abrasion, Dental/methods , Bacterial Load/drug effects , Dental Scaling/methods , Erythritol/therapeutic use , Follow-Up Studies , Lasers, Semiconductor/therapeutic use , Periodontal Attachment Loss/therapy , Periodontal Attachment Loss/microbiology , Periodontal Pocket/therapy , Periodontal Pocket/microbiology , Periodontitis/microbiology , Periodontitis/therapy , Periodontitis/drug therapy , Porphyromonas gingivalis/isolation & purification , Porphyromonas gingivalis/drug effects , Root Planing/methods , Treatment Outcome
2.
Arch Microbiol ; 206(8): 354, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39017726

ABSTRACT

Titanium implants are subject to bacterial adhesion and peri-implantitis induction, and biosurfactants bring a new alternative to the fight against infections. This work aimed to produce and characterize the biosurfactant from Bacillus subtilis ATCC 19,659, its anti-adhesion and antimicrobial activity, and cell viability. Anti-adhesion studies were carried out against Streptococcus sanguinis, Staphylococcus aureus, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, and Proteus mirabilis as the minimum inhibitory concentration and the minimum bactericidal concentration. Cell viability was measured against osteoblast and fibroblast cells. The biosurfactant was classified as lipopeptide, with critical micelle concentration at 40 µg mL- 1, and made the titanium surface less hydrophobic. The anti-adhesion effect was observed for Staphylococcus aureus and Streptococcus sanguinis with 54% growth inhibition and presented a minimum inhibitory concentration of 15.7 µg mL- 1 for Streptococcus sanguinis and Aggregatibacter actinomycetemcomitans. The lipopeptide had no cytotoxic effect and demonstrated high potential application against bacterial biofilms.


Subject(s)
Bacterial Adhesion , Biofilms , Dental Implants , Lipopeptides , Microbial Sensitivity Tests , Titanium , Titanium/pharmacology , Titanium/chemistry , Biofilms/drug effects , Biofilms/growth & development , Bacterial Adhesion/drug effects , Dental Implants/microbiology , Lipopeptides/pharmacology , Humans , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Bacillus subtilis/drug effects , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/physiology , Porphyromonas gingivalis/growth & development , Aggregatibacter actinomycetemcomitans/drug effects , Surface Properties , Fibroblasts/drug effects , Fusobacterium nucleatum/drug effects , Cell Survival/drug effects , Osteoblasts/drug effects , Surface-Active Agents/pharmacology
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1159-1165, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977346

ABSTRACT

OBJECTIVE: To investigate the effect of Porphyromonas gingivalis (Pg) infection on immune escape of oesophageal cancer cells and the role of YTHDF2 and Fas in this regulatory mechanism. METHODS: We examined YTHDF2 and Fas protein expressions in esophageal squamous cell carcinoma (ESCC) tissues with and without Pg infection using immunohistochemistry and in Pg-infected KYSE150 cells using Western blotting. The interaction between YTHDF2 and Fas was investigated by co-immunoprecipitation (Co-IP). Pg-infected KYSE150 cells with lentivirus-mediated YTHDF2 knockdown were examined for changes in expression levels of YTHDF2, cathepsin B (CTSB), Fas and FasL proteins, and the effect of E64 (a cathepsin inhibitor) on these proteins were observed. After Pg infection and E64 treatment, KYSE150 cells were co-cultured with human peripheral blood mononuclear cells (PBMCs), and the expressions of T cell-related effector molecules were detected by flow cytometry. RESULTS: ESCC tissues and cells with Pg infection showed significantly increased YTHDF2 expression and lowered Fas expression. The results of Co-IP demonstrated a direct interaction between YTHDF2 and Fas. In Pg-infected KYSE150 cells with YTHDF2 knockdown, the expression of CTSB was significantly reduced while Fas and FasL expressions were significantly increased. E64 treatment of KYSE150 cells significantly decreased the expression of CTSB without affecting YTHDF2 expression and obviously increased Fas and FasL expressions. Flow cytometry showed that in Pg-infected KYSE150 cells co-cultured with PBMCs, the expressions of Granzyme B and Ki67 were significantly decreased while PD-1 expression was significantly enhanced. CONCLUSION: Pg infection YTHDF2-dependently regulates the expression of Fas to facilitate immune escape of esophageal cancer and thus promoting cancer progression, suggesting the key role of YTHDF2 in regulating immune escape of esophageal cancer.


Subject(s)
Esophageal Neoplasms , Porphyromonas gingivalis , RNA-Binding Proteins , fas Receptor , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Cell Line, Tumor , fas Receptor/metabolism , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Fas Ligand Protein/metabolism , Tumor Escape
4.
Mol Biol Rep ; 51(1): 814, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008163

ABSTRACT

Periodontitis is a severe gum infection that begins as gingivitis and can lead to gum recession, bone loss, and tooth loss if left untreated. It is primarily caused by bacterial infection, which triggers inflammation and the formation of periodontal pockets. Notably, periodontitis is associated with systemic health issues and has been linked to heart disease, diabetes, respiratory diseases, adverse pregnancy outcomes, and cancers. Accordingly, the presence of chronic inflammation and immune system dysregulation in individuals with periodontitis significantly contributes to the initiation and progression of various cancers, particularly oral cancers. These processes promote genetic mutations, impair DNA repair mechanisms, and create a tumor-supportive environment. Moreover, the bacteria associated with periodontitis produce harmful byproducts and toxins that directly damage the DNA within oral cells, exacerbating cancer development. In addition, chronic inflammation not only stimulates cell proliferation but also inhibits apoptosis, causes DNA damage, and triggers the release of pro-inflammatory cytokines. Collectively, these factors play a crucial role in the progression of cancer in individuals affected by periodontitis. Further, specific viral and bacterial agents, such as hepatitis B and C viruses, human papillomavirus (HPV), Helicobacter pylori (H. pylori), and Porphyromonas gingivalis, contribute to cancer development through distinct mechanisms. Bacterial infections have systemic implications for cancer development, while viral infections provoke immune and inflammatory responses that can lead to genetic mutations. This review will elucidate the link between periodontitis and cancers, particularly oral cancers, exploring their underlying mechanisms to provide insights for future research and treatment advancements.


Subject(s)
Mouth Neoplasms , Periodontitis , Humans , Periodontitis/complications , Periodontitis/microbiology , Mouth Neoplasms/microbiology , Mouth Neoplasms/genetics , Animals , Inflammation/complications , Porphyromonas gingivalis/pathogenicity
5.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 653-662, 2024 Jul 09.
Article in Chinese | MEDLINE | ID: mdl-38949133

ABSTRACT

Objective: To investigate the regulatory effect and mechanism of interleukin-22 (IL-22) on the gingival epithelial barrier in the context of periodontal inflammation. Methods: IL-22 knockout (IL-22 KO) mice were constructed, and periodontitis mice models were established through oral gavage with polymicrobial inoculation. DNAs were extracted from the oral plaques of IL-22 KO periodontitis mice group (n=7) and their wild-type littermates periodontitis group (n=7) to establish a periodontitis-related oral microbiota database"PD-RiskMicroDB", determining the relationship between changes in oral microbiota and microbial function in two groups using 16S rRNA sequencing results. Gingival epithelial cells (GEC) were cultured by modified trypsinization method, and were stimulated with 100 µg/L IL-22, Porphyromonas gingivalis (Pg) (multiplicity of infection:100), separately or together for 3 and 12 hours. The experimental groups were as follows: control group (no stimulation), IL-22 group, Pg group and Pg+IL-22 group. The expression of barrier protein E-cadherin in each group at 3 h was detected by immunofluorescence, real-time fluorescence quantitative PCR (RT-qPCR) and Western blotting. Fluorescein isothiocyanate-dextran-mediated epithelial cell permeability experiment was conducted to clarify the changes in permeability of GEC in each group at 3 and 12 h. The mRNA expressions of E-cadherin in the gingival epithelium of wild-type littermates periodontitis group and IL-22 KO periodontitis group were detected by RT-qPCR. Fifteen C57BL/6 wild-type mice were randomly divided into control group (n=5), periodontitis group (n=5) and periodontitis+IL-22 treatment group (n=5). RT-qPCR and immunohistochemistry (IHC) staining were used to detect the expression level of E-cadherin in the gingival epithelium of each group. Results: 16S rRNA sequencing results showed that the composition of oral microbiota changed in IL-22 KO periodontitis group, of which the abundance of bacterial genera related to periodontal tissue invasion was significantly increased (linear discriminant analysis score: 2.22, P=0.009), compared with wild-type littermates periodontitis group. In vitro cell experiments showed that after Pg infection for 3 hours, the cell connections of GEC in Pg group were interrupted, and the fluorescence intensity of E-cadherin was reduced in Pg group compared with the control group. Meanwhile, the mRNA and protein expression levels of E-cadherin (mRNA: 0.69±0.12; protein: 0.60±0.12) were downregulated compared with the control group [mRNA: 1.00±0.00 (P=0.043); protein: 1.04±0.08 (P=0.003)], respectively. The fluorescence intensity of E-cadherin in the Pg+IL-22 group was enhanced compared with Pg group, and expression levels of E-cadherin mRNA (1.16±0.10) and protein (0.98±0.07) in Pg+IL-22 group showed a significant increase compared with Pg group [mRNA: 0.69±0.12 (P=0.005); protein: 0.60±0.12 (P=0.007)]. The result of epithelial permeability test showed that there was no statistical difference in epithelial permeability among control group, Pg group, IL-22 group and Pg+IL-22 group with treatment for 3 hours (F=0.20, P=0.893). While when the treatment time turned to be 12 hours, the epithelial barrier permeability showed a significant increase in Pg group (1.39±0.15) compared with control group (1.00±0.00, P=0.027), and a decrease in Pg+IL-22 group (1.02±0.18) compared with Pg group (1.39±0.15, P=0.034). In vivo, the mRNA expression of E-cadherin in the gingival epithelium of IL-22 KO periodontitis group decreased significantly (0.32±0.21) compared with wild-type littermates periodontitis group (1.01±0.01) (t=5.70, P=0.005). Moreover, RT-qPCR and IHC staining results showed that the mRNA expression level of E-cadherin (0.40±0.07) and absorbance value of E-cadherin positive expression (0.02±0.00) in gingival epithelial tissue of periodontitis group were both significantly down-regulated compared with control group [mRNA: 1.00±0.00 (P=0.005); absorbance value of E-cadherin positive expression: 0.04±0.01 (P=0.006)]. Meanwhile, the mRNA expression level of E-cadherin (1.06±0.24) and the absorbance value of E-cadherin positive expression (0.03±0.01) were both observed increase in periodontitis+IL-22 treatment group compared with periodontitis group (P=0.003, P=0.039). Conclusions: IL-22 may exert a protective effect on the gingival epithelial barrier in an inflammatory environment by regulating the invasiveness of oral microbiota and the expression of host barrier protein.


Subject(s)
Cadherins , Gingiva , Interleukin-22 , Interleukins , Mice, Knockout , Microbiota , Periodontitis , Porphyromonas gingivalis , Animals , Interleukins/metabolism , Cadherins/metabolism , Gingiva/cytology , Gingiva/metabolism , Gingiva/microbiology , Mice , Periodontitis/microbiology , Periodontitis/metabolism , Epithelial Cells/metabolism , RNA, Ribosomal, 16S
6.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 672-680, 2024 Jul 09.
Article in Chinese | MEDLINE | ID: mdl-38949135

ABSTRACT

Objective: To investigate the effects of Porphyromonas gingivalis (Pg) persisters (Ps) on immuno-inflammatory responses in macrophages, and to explore the underlying mechanisms. Methods: Pg cells were cultured to the stationary phase (72 h), and subsequently treated by high concentration of metronidazole at 100 mg/L, amoxicillin at 100 mg/L and the combination of them for different time period, named as metronidazole group, amoxicillin group and (metronidazole+amoxicillin) group. Pg cells without treatment were used as Blank control. The survival profile of PgPs cells was measured by colony-forming unit assay. The living state of PgPs was observed by Live/Dead staining. Then, Pg and metronidazole-treated PgPs (M-PgPs) were used to treat macrophages, named as Pg group and M-PgPs group. Transmission electron microscopy (TEM) was used to observe the bacteria in the macrophages. The expression levels of proinflammatory cytokines in macrophages were determined by real-time fluorescence quantitative PCR and enzyme-linked immunosorbent assay. The location of forkhead box transcription factor 1 (FOXO1) was detected by confocal immunofluorescence microscopy. After inhibiting or enhancing the FOXO1 expressions using inhibitors (Fi) or activators (Fa) respectively, the macrophages were treated with Pg and M-PgPs, divided as Blank group, Pg group, M-PgPs group, Fi group, (Fi+Pg) group, (Fi+M-PgPs) group, Fa group, (Fa+Pg) group and (Fa+M-PgPs) group. Then, the expression pattens of proinflammatory cytokines were assessed. Results: Remarkable number of lived PgPs was observed, both in planktonic culture and Pg biofilms either treated with metronidazole, amoxicillin or both, and those persisters could form new colonies. Pg and M-PgPs were able to enter into the macrophages and the protein expression levels of interleukin (IL)-1ß, IL-6, IL-8 and tumor necrosis factor-α (TNF-α) [Pg group: (2 392±188), (162±29), (5 558±661), (789±155) µg/L; M-PgPs group: (2 415±420), (155±3), (5 732±782), (821±176) µg/L] were significantly upregulated than those in Blank group [(485±140), (21±9), (2 332±87), (77±7) µg/L] (P<0.01). Moreover, Pg and M-PgPs could facilitate the nuclear translocation and accumulation of FOXO1. In addition, the relative mRNA expression levels of FOXO1, B-cell lymphoma 6 and Krüppel-like factor 2 were upregulated when compared to Blank group (P<0.05). Furthermore, the protein expression levels of IL-1ß, IL-6, IL-8 and TNF-α in Fi+Pg group [(1 081±168), (70±8), (1 976±544), (420±47) µg/L] were remarkably lower than Pg group [(4 411±137), (179±6), (5 161±929), (934±24) µg/L] (P<0.05). Similarly, the protein expression levels of IL-1ß, IL-6, IL-8 and TNF-α in Fi+M-PgPs group [(1 032±237), (74±10), (1 861±614), (405±32) µg/L] were remarkably lower than M-PgPs group [(4 342±314), (164±17), (4 438±1 374), (957±25) µg/L] (P<0.05). On the contrary, the protein expression levels of IL-1ß, IL-6, IL-8 and TNF-α in Fa+Pg group [(8 198±1 825), (431±28), (8 919±650), (2 186±301) µg/L] and Fa+M-PgPs group [(8 159±2 627), (475±26), (8 995±653), (2 255±387) µg/L] were significantly higher than Pg group and M-PgPs group, respectively (P<0.05). Conclusions: PgPs are highly tolerant to metronidazole and amoxicillin. The M-PgPs could enhance the immuno-inflammatory responses in macrophages by upregulating the FOXO1 signaling pathway, while this effect exhibits no significant difference with Pg.


Subject(s)
Biofilms , Macrophages , Metronidazole , Porphyromonas gingivalis , Signal Transduction , Macrophages/metabolism , Metronidazole/pharmacology , Biofilms/drug effects , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Amoxicillin/pharmacology , Up-Regulation , Animals , Interleukin-1beta/metabolism , Mice , Forkhead Box Protein O1/metabolism , Interleukin-8/metabolism , Inflammation , Humans
7.
Clin Exp Dent Res ; 10(4): e903, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39031165

ABSTRACT

OBJECTIVES: To explore the antimicrobial potential of strontium (Sr)-functionalized wafers against multiple bacteria associated with per-implant infections, in both mono- and multispecies biofilms. MATERIALS AND METHODS: The bactericidal and bacteriostatic effect of silicon wafers functionalized with a strontium titanium oxygen coating (Sr-Ti-O) or covered only with Ti (controls) against several bacteria, either grown as a mono-species or multispecies biofilms, was assessed using a bacterial viability assay and a plate counting method. Mono-species biofilms were assessed after 2 and 24 h, while the antimicrobial effect on multispecies biofilms was assessed at Days 1, 3, and 6. The impact of Sr functionalization on the total percentage of Porphyromonas gingivalis in the multispecies biofilm, using qPCR, and gingipain activity was also assessed. RESULTS: Sr-functionalized wafers, compared to controls, were associated with statistically significant less viable cells in both mono- and multispecies tests. The number of colony forming units (CFUs) within the biofilm was significantly less in Sr-functionalized wafers, compared to control wafers, for Staphylococcus aureus at all time points of evaluation and for Escherichia coli at Day 1. Gingipain activity was less in Sr-functionalized wafers, compared to control wafers, and the qPCR showed that P. gingivalis remained below detection levels at Sr-functionalized wafers, while it consisted of 15% of the total biofilm on control wafers at Day 6. CONCLUSION: Sr functionalization displayed promising antimicrobial potential, possessing bactericidal and bacteriostatic ability against bacteria associated with peri-implantitis grown either as mono-species or mixed in a multispecies consortium with several common oral microorganisms.


Subject(s)
Biofilms , Peri-Implantitis , Porphyromonas gingivalis , Strontium , Titanium , Titanium/chemistry , Titanium/pharmacology , Biofilms/drug effects , Peri-Implantitis/microbiology , Peri-Implantitis/drug therapy , Strontium/pharmacology , Porphyromonas gingivalis/drug effects , Humans , Anti-Bacterial Agents/pharmacology , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Microbial Viability/drug effects , Dental Implants/microbiology
8.
BMC Oral Health ; 24(1): 668, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849764

ABSTRACT

BACKGROUND: Crohn's disease (CD)-associated periodontitis is common. However, the role of periodontal pathogens in the Coexistence of CD and periodontal disease remains unclear. METHODS: To investigate the potential relationship mediated by periodontal pathogens between periodontitis and CD, we collected salivary samples from healthy participants (H group, n = 12), patients with CD (Ch group, n = 10), patients with periodontitis (Ps group, n = 12), and patients with Coexistence of CD and periodontal disease (Cp group, n = 12) and analyzed them by 16 S rRNA sequencing. RESULTS: Patients with Coexistence of CD and periodontal disease had increased levels of Fusobacterium, Actinomyces, Leptotrichia, and Prevotella, which correlated with the severity of periodontitis. Conversely, the levels of Streptococcus, Neisseria, Haemophilus, and Gemella, which decreased in Coexistence of CD and periodontal disease, were negatively correlated with the severity of periodontitis. To further investigate the role of periodontal pathogens in CD development, representative periodontal pathogens causing periodontitis, Porphyromonas gingivalis and Fusobacterium nucleatum, were administered to mice. These pathogens migrate to, and colonize, the gut, accelerating CD progression and aggravating colitis, and even systemic inflammation. In vitro experiments using a Caco-2/periodontal pathogen coculture revealed that P. gingivalis and F. nucleatum increased intestinal permeability by directly disrupting the tight junctions of intestinal epithelial cells. CONCLUSION: Our findings strongly suggest that periodontal pathogens play a role in the relationship between periodontitis and CD. These results provide a basis for understanding the pathogenesis of Coexistence of CD and periodontal disease and may lead to the development of novel therapeutic strategies.


Subject(s)
Crohn Disease , Fusobacterium nucleatum , Periodontitis , Porphyromonas gingivalis , Humans , Crohn Disease/microbiology , Crohn Disease/complications , Periodontitis/microbiology , Periodontitis/complications , Animals , Mice , Male , Female , Adult , Fusobacterium nucleatum/isolation & purification , Caco-2 Cells , Saliva/microbiology , RNA, Ribosomal, 16S
9.
PeerJ ; 12: e17241, 2024.
Article in English | MEDLINE | ID: mdl-38854801

ABSTRACT

Tea tree (Melaleuca alternifolia) oil (TTO) is an antimicrobial agent, and hence, its use in fabricating nanoparticles (NP) may be useful in providing more efficacious antimicrobial agents. The current research aimed to test the antimicrobial efficacy of TTO and its TTO-Metal-NPs against oral microbes: Porphyromonas gingivalis, Enterococcus faecalis, and Streptococcus mutans. The antimicrobial activity of TTO and zinc (Zn) and iron (Fe) nanoparticles (NPs) and the combined effects of antimicrobial agents were investigated using agar well diffusion assays. Fourier-transform infrared spectroscopy (FT-IR) was used to identify the phyto-constituents of TTO. Field emission scanning electron microscopy (FE-SEM), dynamic light scatter (DLS), and zeta potential were utilized to analyze the biogenic nanoparticles' morphology, size, and potential. The antimicrobial mode of action was determined by assessing the morphological changes under scanning electron microscopy (SEM). The TTO extracts converted Zn and Fe ions to NPs, having an average size of 97.50 (ZnNPs) and 102.4 nm (FeNPs). All tested agents had significant antibacterial efficacy against the tested oral microbes. However, the TTO extract was more efficacious than the NPs. Combination treatment of TTO with antibiotics resulted in partial additive effects against P. gingivalis and partial antagonistic effects against E. faecalis, S. mutans, and common mouthwashes (Oral B and chlorhexidine). TTO and NP-treated bacteria underwent morphological changes on treatment. M. alternifolia phytochemicals could be useful for further research and development of antimicrobial NPs. The current study highlights the variance in activity observed for different types of bacteria and antagonistic effects seen with common mouthwashes, which represent a threat to therapeutic efficacy and heighten the risk of clinical microbial resistance.


Subject(s)
Metal Nanoparticles , Porphyromonas gingivalis , Streptococcus mutans , Tea Tree Oil , Tea Tree Oil/pharmacology , Tea Tree Oil/chemistry , Metal Nanoparticles/chemistry , Porphyromonas gingivalis/drug effects , Streptococcus mutans/drug effects , Microbial Sensitivity Tests , Enterococcus faecalis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mouth/microbiology , Microscopy, Electron, Scanning , Melaleuca/chemistry , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Humans , Iron , Spectroscopy, Fourier Transform Infrared
10.
J Appl Oral Sci ; 32: e20240047, 2024.
Article in English | MEDLINE | ID: mdl-38922243

ABSTRACT

OBJECTIVE: To assess the efficacy of Phyllanthus emblica extract in alleviating halitosis and reducing the inflammatory response to halitosis-related bacteria. METHODOLOGY: This investigation, using Phyllanthus emblica fruit extract (PE), involved four aspects. First, we evaluated the effect on growth and aggregation of halitosis-related bacteria, including Fusobacterium nucleatum, Porphyromonas gingivalis, and Solobacterium moorei, using a microdilution assay and scanning electron microscopy. Second, volatile sulfur compound (VSC) levels were measured on individuals with halitosis in randomized short-term (26 participants) and double-blind randomized long-term trials (18 participants in each group) after rinsing with PE for 3, 6, and 12 h, and 28 days. Third, we analyzed pro-inflammatory cytokine expression in TR146 cells using quantitative real-time PCR and enzyme-linked immunosorbent assays. Lastly, we assessed pro-inflammatory cytokine secretion and Toll-like receptor (TLR) 2 mRNA expression via the same experimental methods in a three-dimensional oral mucosal epithelial model (3D OMEM). RESULTS: PE extract dose-dependently inhibited the growth of F. nucleatum (50% inhibition concentration [IC50]=0.079%), P. gingivalis (IC50=0.65%), and S. moorei (IC50=0.07%) and effectively prevented bacterial aggregation. Furthermore, VSC contents decreased significantly at 3, 6, and 12 h after rinsing with 5% PE compared with those in the control. Long-term use of mouthwash containing 5% PE for 28 days led to a significant decrease in VSC contents. PE attenuated the F. nucleatum- or P. gingivalis-stimulated mRNA expression and protein release of interleukin (IL)-6 and IL-8 in TR146 cells. It also suppressed IL-8 and prostaglandin E2 secretion and TLR2 mRNA expression in F. nucleatum-induced OMEMs. CONCLUSION: Our findings support the use of PE in oral care products to alleviate halitosis and it may reduce inflammation.


Subject(s)
Cytokines , Enzyme-Linked Immunosorbent Assay , Fusobacterium nucleatum , Halitosis , Microscopy, Electron, Scanning , Phyllanthus emblica , Plant Extracts , Porphyromonas gingivalis , Real-Time Polymerase Chain Reaction , Phyllanthus emblica/chemistry , Halitosis/drug therapy , Halitosis/microbiology , Humans , Plant Extracts/pharmacology , Double-Blind Method , Fusobacterium nucleatum/drug effects , Porphyromonas gingivalis/drug effects , Female , Time Factors , Male , Treatment Outcome , Adult , Young Adult , Toll-Like Receptor 2/drug effects , Fruit/chemistry , Statistics, Nonparametric , Mouth Mucosa/drug effects , Mouth Mucosa/microbiology , Analysis of Variance , Sulfur Compounds/pharmacology , Sulfur Compounds/analysis
11.
Front Cell Infect Microbiol ; 14: 1418651, 2024.
Article in English | MEDLINE | ID: mdl-38933693

ABSTRACT

Background: This study unveils the intricate functional association between cyclic di-3',5'-adenylic acid (c-di-AMP) signaling, cellular bioenergetics, and the regulation of lipopolysaccharide (LPS) profile in Porphyromonas gingivalis, a Gram-negative obligate anaerobe considered as a keystone pathogen involved in the pathogenesis of chronic periodontitis. Previous research has identified variations in P. gingivalis LPS profile as a major virulence factor, yet the underlying mechanism of its modulation has remained elusive. Methods: We employed a comprehensive methodological approach, combining two mutants exhibiting varying levels of c-di-AMP compared to the wild type, alongside an optimized analytical methodology that combines conventional mass spectrometry techniques with a novel approach known as FLATn. Results: We demonstrate that c-di-AMP acts as a metabolic nexus, connecting bioenergetic status to nuanced shifts in fatty acid and glycosyl profiles within P. gingivalis LPS. Notably, the predicted regulator gene cdaR, serving as a potent regulator of c-di-AMP synthesis, was found essential for producing N-acetylgalactosamine and an unidentified glycolipid class associated with the LPS profile. Conclusion: The multifaceted roles of c-di-AMP in bacterial physiology are underscored, emphasizing its significance in orchestrating adaptive responses to stimuli. Furthermore, our findings illuminate the significance of LPS variations and c-di-AMP signaling in determining the biological activities and immunostimulatory potential of P. gingivalis LPS, promoting a pathoadaptive strategy. The study expands the understanding of c-di-AMP pathways in Gram-negative species, laying a foundation for future investigations into the mechanisms governing variations in LPS structure at the molecular level and their implications for host-pathogen interactions.


Subject(s)
Lipopolysaccharides , Porphyromonas gingivalis , Signal Transduction , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/genetics , Lipopolysaccharides/metabolism , Virulence Factors/metabolism , Gene Expression Regulation, Bacterial , Energy Metabolism , Dinucleoside Phosphates/metabolism , Fatty Acids/metabolism , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
12.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892314

ABSTRACT

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Subject(s)
Apolipoproteins E , Atherosclerosis , Periodontal Diseases , Porphyromonas gingivalis , Animals , Mice , Apolipoproteins E/deficiency , Periodontal Diseases/microbiology , Periodontal Diseases/prevention & control , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Atherosclerosis/microbiology , Telomerase/metabolism , Peptide Fragments/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Alzheimer Disease/microbiology , Periodontitis/microbiology , Periodontitis/prevention & control , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/prevention & control , Disease Models, Animal , Mice, Inbred C57BL , Male , Humans
13.
Open Biol ; 14(6): 230448, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862016

ABSTRACT

Gram-negative bacteria from the Bacteroidota phylum possess a type-IX secretion system (T9SS) for protein secretion, which requires cargoes to have a C-terminal domain (CTD). Structurally analysed CTDs are from Porphyromonas gingivalis proteins RgpB, HBP35, PorU and PorZ, which share a compact immunoglobulin-like antiparallel 3+4 ß-sandwich (ß1-ß7). This architecture is essential as a P. gingivalis strain with a single-point mutant of RgpB disrupting the interaction of the CTD with its preceding domain prevented secretion of the protein. Next, we identified the C-terminus ('motif C-t.') and the loop connecting strands ß3 and ß4 ('motif Lß3ß4') as conserved. We generated two strains with insertion and replacement mutants of PorU, as well as three strains with ablation and point mutants of RgpB, which revealed both motifs to be relevant for T9SS function. Furthermore, we determined the crystal structure of the CTD of mirolase, a cargo of the Tannerella forsythia T9SS, which shares the same general topology as in Porphyromonas CTDs. However, motif Lß3ß4 was not conserved. Consistently, P. gingivalis could not properly secrete a chimaeric protein with the CTD of peptidylarginine deiminase replaced with this foreign CTD. Thus, the incompatibility of the CTDs between these species prevents potential interference between their T9SSs.


Subject(s)
Bacterial Proteins , Bacterial Secretion Systems , Porphyromonas gingivalis , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/chemistry , Models, Molecular , Crystallography, X-Ray , Amino Acid Sequence , Protein Sorting Signals , Protein Domains , Bacteroidetes/metabolism , Bacteroidetes/genetics , Tannerella forsythia/metabolism , Tannerella forsythia/genetics , Tannerella forsythia/chemistry , Structure-Activity Relationship , Protein Conformation
14.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892350

ABSTRACT

Periodontitis is an inflammatory disease caused by Porphyromonas gingivalis (P. gingivalis) in the oral cavity. This periodontal disease causes damage to the periodontal ligament and alveolar bone and can cause tooth loss, but there is no definite treatment yet. In this study, we investigated the possibility of using no-ozone cold plasma to safely treat periodontitis in the oral cavity. First, human gingival fibroblasts (HGFs) were treated with P. gingivalis-derived lipopolysaccharide (PG-LPS) to induce an inflammatory response, and then the anti-inflammatory effect of NCP was examined, and a study was conducted to identify the mechanism of action. Additionally, the anti-inflammatory effect of NCP was verified in rats that developed an inflammatory response similar to periodontitis. When NCP was applied to PG-LPS-treated HGFs, the activities of inflammatory proteins and cytokines were effectively inhibited. It was confirmed that the process of denaturing the medium by charged particles of NCP is essential for the anti-inflammatory effect of NCP. Also, it was confirmed that repeated treatment of periodontitis rats with NCP effectively reduced the inflammatory cells and osteoclast activity. As a result, this study suggests that NCP can be directly helpful in the treatment of periodontitis in the future.


Subject(s)
Anti-Inflammatory Agents , Fibroblasts , Gingiva , Lipopolysaccharides , Periodontitis , Porphyromonas gingivalis , Animals , Periodontitis/microbiology , Periodontitis/drug therapy , Rats , Anti-Inflammatory Agents/pharmacology , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Ozone/pharmacology , Plasma Gases/pharmacology , Plasma Gases/therapeutic use , Male , Cytokines/metabolism , Disease Models, Animal , Nitric Oxide/metabolism , Cells, Cultured
15.
Front Cell Infect Microbiol ; 14: 1413787, 2024.
Article in English | MEDLINE | ID: mdl-38836053

ABSTRACT

Background: Trimethylamine-N-oxide (TMAO) is produced by hepatic flavin-containing monooxygenase 3 (FMO3) from trimethylamine (TMA). High TMAO level is a biomarker of cardiovascular diseases and metabolic disorders, and it also affects periodontitis through interactions with the gastrointestinal microbiome. While recent findings indicate that periodontitis may alter systemic TMAO levels, the specific mechanisms linking these changes and particular oral pathogens require further clarification. Methods: In this study, we established a C57BL/6J male mouse model by orally administering Porphyromonas gingivalis (P. gingivalis, Pg), Fusobacterium nucleatum (F. nucleatum, Fn), Streptococcus mutans (S. mutans, Sm) and PBS was used as a control. We conducted LC-MS/MS analysis to quantify the concentrations of TMAO and its precursors in the plasma and cecal contents of mice. The diversity and composition of the gut microbiome were analyzed using 16S rRNA sequencing. TMAO-related lipid metabolism and enzymes in the intestines and liver were assessed by qPCR and ELISA methods. We further explored the effect of Pg on FMO3 expression and lipid molecules in HepG2 cells by stimulating the cells with Pg-LPS in vitro. Results: The three oral pathogenic bacteria were orally administered to the mice for 5 weeks. The Pg group showed a marked increase in plasma TMAO, betaine, and creatinine levels, whereas no significant differences were observed in the gut TMAO level among the four groups. Further analysis showed similar diversity and composition in the gut microbiomes of both the Pg and Fn groups, which were different from the Sm and control groups. The profiles of TMA-TMAO pathway-related genera and gut enzymes were not significantly different among all groups. The Pg group showed significantly higher liver FMO3 levels and elevated lipid factors (IL-6, TG, TC, and NEFA) in contrast to the other groups. In vitro experiments confirmed that stimulation of HepG2 cells with Pg-LPS upregulated the expression of FMO3 and increased the lipid factors TC, TG, and IL-6. Conclusion: This study conclusively demonstrates that Pg, compared to Fn and Sm, plays a critical role in elevating plasma TMAO levels and significantly influences the TMA-TMAO pathway, primarily by modulating the expression of hepatic FMO3 and directly impacting hepatic lipid metabolism.


Subject(s)
Gastrointestinal Microbiome , Methylamines , Mice, Inbred C57BL , Oxygenases , Porphyromonas gingivalis , Animals , Male , Methylamines/metabolism , Methylamines/blood , Humans , Mice , Oxygenases/metabolism , Porphyromonas gingivalis/metabolism , Fusobacterium nucleatum/metabolism , Metabolic Networks and Pathways , Hep G2 Cells , Lipid Metabolism , Disease Models, Animal , Periodontitis/microbiology , Periodontitis/metabolism , Liver/metabolism , RNA, Ribosomal, 16S/genetics , Tandem Mass Spectrometry , Mouth/microbiology
16.
PLoS One ; 19(6): e0292830, 2024.
Article in English | MEDLINE | ID: mdl-38857232

ABSTRACT

Lipopolysaccharide (LPS), a component of the Gram-negative bacterial cell wall, activates Toll-like receptors (TLRs). Porphyromonas gingivalis (Pg) may be involved in the progression of periodontal disease. Mice exposed to a novel environment show hyperlocomotion that is inhibited by systemic administration of LPS derived from Escherichia coli (Ec-LPS). However, whether Pg-LPS influences novelty-induced locomotion is unknown. Accordingly, we carried out an open field test to analyse the effects of Pg-LPS. For comparison, effects of Ec-LPS were also studied. We additionally investigated the influence of systemic administration of Pg-LPS or Ec-LPS on IL-6, TNF-alpha, and IL-10 levels in blood, as they could be involved in the changes in locomotion. The TLR4 receptor antagonist TAK-242 was used to study the involvement of TLR4. Since Pg-LPS may block TLR4 in vitro, we analysed the effects of Pg-LPS on Ec-LPS-induced changes in behavioural and biochemical parameters. Male ddY mice were used. Pg- or Ec-LPS and TAK-242 were administered intraperitoneally. Ec-LPS (840 µg/kg), but not Pg-LPS (100, 500 and 840 µg/kg), inhibited novelty-induced locomotion, which was antagonized by TAK-242 (3.0 mg/kg). Ec-LPS (840 µg/kg) increased blood levels of IL-6 and IL-10, which were antagonized by TAK-242 (3.0 mg/kg). However, TAK-242 did not inhibit Ec-LPS-induced increases in TNF-alpha levels in blood. Pg-LPS (100, 500, and 840 µg/kg) did not alter blood IL-6, TNF-alpha, or IL-10 levels. The Ec-LPS-induced increase in blood IL-10, but not IL-6 and TNF-alpha, levels was inhibited by Pg-LPS (500 µg/kg). These results suggest that TLR4 stimulation mediates the inhibition of novel environment-induced locomotion in mice following systemic administration of Ec-LPS, while also increasing blood IL-6 and IL-10 levels. In contrast, Pg-LPS did not exhibit these effects. The present study also provides in vivo evidence that Pg-LPS can inhibit TLR4-mediated increases in blood levels of IL-10, a cytokine thought to prevent the development of periodontal disease.


Subject(s)
Escherichia coli , Lipopolysaccharides , Porphyromonas gingivalis , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Mice , Male , Locomotion/drug effects , Cytokines/blood , Cytokines/metabolism , Interleukin-6/blood , Interleukin-10/blood , Tumor Necrosis Factor-alpha/blood , Sulfonamides
17.
Sci Rep ; 14(1): 13969, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886503

ABSTRACT

Periodontitis is a chronic inflammatory disease that affects the periodontal tissues. Although it is associated with various systemic diseases, the impact of periodontitis on kidney transplantation (KT) outcomes, particularly allograft rejection, remains unclear. This study investigated the effect of periodontitis on transplant immunity, specifically examining Porphyromonas gingivalis-derived lipopolysaccharide (LPS-PG). In vitro experiments revealed that LPS-PG increased regulatory T cells (Tregs) in Lewis rat spleen cells. In a mixed lymphocyte reaction assay, concentrations of interferon-γ, indicative of alloreactivity, were lower than in controls when LPS-PG was added to the culture and when LPS-PG-administered Lewis rat spleen cells were used as responders. In a rat KT model, LPS-PG administration to recipients promoted mild tubulitis and low serum creatinine and blood urea nitrogen levels 5 days post-KT compared with PBS-administered controls. Furthermore, LPS-PG-administered recipients had an elevated Treg proportion in their peripheral blood and spleen cells, and increased infiltrating Tregs in kidney allografts, compared with controls. The elevated Treg proportion in peripheral blood and spleen cells had a significant negative correlation with serum creatinine, suggesting elevated Tregs modulated allograft rejection. These findings suggest that periodontitis might modulate alloimmune reactivity through LPS-PG and Tregs, offering insights to refine immunosuppressive strategies for KT recipients.


Subject(s)
Graft Rejection , Kidney Transplantation , Lipopolysaccharides , Porphyromonas gingivalis , Rats, Inbred Lew , T-Lymphocytes, Regulatory , Animals , Porphyromonas gingivalis/immunology , Kidney Transplantation/adverse effects , Rats , T-Lymphocytes, Regulatory/immunology , Male , Graft Rejection/immunology , Allografts , Periodontitis/immunology , Periodontitis/microbiology , Disease Models, Animal , Spleen/immunology
18.
Mol Biol Rep ; 51(1): 758, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874801

ABSTRACT

OBJECTIVES: This study aimed to evaluate the impact of scaling and root surface debridement (SRP) on salivary bacterial counts and systolic and diastolic blood pressure in hypertensive patients with chronic periodontitis, with a focus on clinical significance. METHODS: An observational trial included 24 chronic periodontitis patients, eleven of them were hypertensive patients. Non-surgical periodontal treatment was administered to all patients, with clinical parameters including gingival index (GI), plaque index (PI), and probing pocket depth (PPD) recorded. Saliva samples were collected before and after SRP to quantify total bacterial counts and specific bacterial counts. RESULTS: Two months following SRP, PI and PPD in every subject under study demonstrated good responses. In hypertension patients, the salivary bacterial count was significantly higher following SRP (P = 0.0221). The incidence of Porphyromonas gingivalis in hypertension patients significantly decreased after treatment (P = 0.0386). Despite this, there was no discernible decrease in blood pressure following treatment. CONCLUSIONS: SRP alone was ineffective in reducing overall bacterial counts, but P. gingivalis levels responded favorably. Regular periodontal assessment is crucial for hypertensive individuals to mitigate cardiovascular risk. CLINICAL SIGNIFICANCE: Periodontal therapy in hypertensive patients may improve oral health but might not significantly impact blood pressure. Regular periodontal evaluation is essential for managing cardiovascular risk in hypertension.


Subject(s)
Chronic Periodontitis , Dental Scaling , Hypertension , Saliva , Humans , Chronic Periodontitis/microbiology , Chronic Periodontitis/therapy , Chronic Periodontitis/complications , Hypertension/microbiology , Hypertension/complications , Hypertension/therapy , Female , Male , Middle Aged , Saliva/microbiology , Dental Scaling/methods , Adult , Porphyromonas gingivalis/isolation & purification , Bacterial Load , Blood Pressure/physiology , Periodontal Index , Debridement/methods , Aged
19.
Front Cell Infect Microbiol ; 14: 1421018, 2024.
Article in English | MEDLINE | ID: mdl-38938884

ABSTRACT

Introduction: Porphyromonas gingivalis and Porphyromonas endodontalis belong to the Bacteroidota phylum. Both species inhabit the oral cavity and can be associated with periodontal diseases. To survive, they must uptake heme from the host as an iron and protoporphyrin IX source. Among the best-characterized heme acquisition systems identified in members of the Bacteroidota phylum is the P. gingivalis Hmu system, with a leading role played by the hemophore-like HmuY (HmuYPg) protein. Methods: Theoretical analysis of selected HmuY proteins and spectrophotometric methods were employed to determine the heme-binding mode of the P. endodontalis HmuY homolog (HmuYPe) and its ability to sequester heme. Growth phenotype and gene expression analysis of P. endodontalis were employed to reveal the importance of the HmuYPe and Hmu system for this bacterium. Results: Unlike in P. gingivalis, where HmuYPg uses two histidines for heme-iron coordination, other known HmuY homologs use two methionines in this process. P. endodontalis HmuYPe is the first characterized representative of the HmuY family that binds heme using a histidine-methionine pair. It allows HmuYPe to sequester heme directly from serum albumin and Tannerella forsythia HmuYTf, the HmuY homolog which uses two methionines for heme-iron coordination. In contrast to HmuYPg, which sequesters heme directly from methemoglobin, HmuYPe may bind heme only after the proteolytic digestion of hemoglobin. Conclusions: We hypothesize that differences in components of the Hmu system and structure-based properties of HmuY proteins may evolved allowing different adaptations of Porphyromonas species to the changing host environment. This may add to the superior virulence potential of P. gingivalis over other members of the Bacteroidota phylum.


Subject(s)
Bacterial Proteins , Heme , Porphyromonas endodontalis , Porphyromonas gingivalis , Tannerella forsythia , Heme/metabolism , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/genetics , Tannerella forsythia/metabolism , Tannerella forsythia/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Porphyromonas endodontalis/metabolism , Porphyromonas endodontalis/genetics , Humans , Gene Expression Regulation, Bacterial , Protein Binding , Iron/metabolism
20.
Clin Exp Dent Res ; 10(3): e887, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38798089

ABSTRACT

OBJECTIVE: This study aimed to evaluate the impact of silver nanoparticles (AgNPs) synthesized from propolis on the formation of Porphyromonas gingivalis biofilms. MATERIAL AND METHODS: AgNPs were synthesized from propolis, and their inhibitory effect on P. gingivalis biofilm formation was assessed. Different concentrations of AgNPs (0.1%, 0.3%, and 0.5%) were tested to determine the dose-dependent antibacterial activity. RESULTS: The results of this study indicated that AgNPs exhibited an inhibitory effect on P. gingivalis biofilm formation. The antibacterial activity of AgNPs was dose-dependent, with concentrations of 0.1%, 0.3%, and 0.5% showing effectiveness. Notably, the concentration of 0.5% demonstrated the most significant anti-biofilm formation activity. CONCLUSION: The results of this study suggest that AgNPs synthesized from propolis have potential as an effective option for enhancing periodontal treatment outcomes. The inhibitory effect of AgNPs on P. gingivalis biofilm formation highlights their potential as alternative antimicrobial agents in the management of periodontal diseases.


Subject(s)
Anti-Bacterial Agents , Biofilms , Metal Nanoparticles , Porphyromonas gingivalis , Silver , Porphyromonas gingivalis/drug effects , Biofilms/drug effects , Silver/pharmacology , Silver/chemistry , Metal Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Green Chemistry Technology , Propolis/pharmacology , Propolis/chemistry , Microbial Sensitivity Tests , Dose-Response Relationship, Drug , Humans
SELECTION OF CITATIONS
SEARCH DETAIL