Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
1.
J Nanobiotechnology ; 22(1): 207, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664778

ABSTRACT

Several studies suggest that oral pathogenic biofilms cause persistent oral infections. Among these is periodontitis, a prevalent condition brought on by plaque biofilm. It can even result in tooth loss. Furthermore, the accumulation of germs around a dental implant may lead to peri-implantitis, which damages the surrounding bone and gum tissue. Furthermore, bacterial biofilm contamination on the implant causes soft tissue irritation and adjacent bone resorption, severely compromising dental health. On decontaminated implant surfaces, however, re-osseointegration cannot be induced by standard biofilm removal techniques such as mechanical cleaning and antiseptic treatment. A family of nanoparticles known as nanozymes (NZs) comprise highly catalytically active multivalent metal components. The most often employed NZs with antibacterial activity are those that have peroxidase (POD) activity, among other types of NZs. Since NZs are less expensive, more easily produced, and more stable than natural enzymes, they hold great promise for use in various applications, including treating microbial infections. NZs have significantly contributed to studying implant success rates and periodontal health maintenance in periodontics and implantology. An extensive analysis of the research on various NZs and their applications in managing oral health conditions, including dental caries, dental pulp disorders, oral ulcers, peri-implantitis, and bacterial infections of the mouth. To combat bacteria, this review concentrates on NZs that imitate the activity of enzymes in implantology and periodontology. With a view to the future, there are several ways that NZs might be used to treat dental disorders antibacterially.


Subject(s)
Anti-Bacterial Agents , Biofilms , Dental Implants , Peri-Implantitis , Periodontitis , Peri-Implantitis/drug therapy , Peri-Implantitis/microbiology , Humans , Periodontitis/drug therapy , Periodontitis/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Dental Implants/microbiology , Animals , Nanoparticles/chemistry , Bacteria/drug effects
2.
Gen Dent ; 72(3): 70-73, 2024.
Article in English | MEDLINE | ID: mdl-38640010

ABSTRACT

Treatment of peri-implant diseases focuses on reducing the bacterial load and consequent infection control. The use of local antimicrobials as an adjunct to mechanical therapy may result in a better outcome. Among antimicrobials, doxycycline stands out because of its local modulation of cytokines, microbial reduction, and clinical parameters in the treatment of periodontal diseases. The objective of this case report was to describe the combined application of mechanical debridement and bioresorbable doxycycline-loaded nanospheres for the treatment of peri-implantitis in a 71-year-old man. At the 3-year evaluation, the peri-implant tissues had improved, showing decreased probing depths, an absence of bleeding on probing, and no suppuration. This case report highlights the importance of supportive therapy, which is essential for the long-term success of peri-implantitis treatment.


Subject(s)
Anti-Infective Agents , Dental Implants , Nanospheres , Peri-Implantitis , Male , Humans , Aged , Peri-Implantitis/drug therapy , Peri-Implantitis/microbiology , Doxycycline/therapeutic use , Follow-Up Studies , Debridement , Absorbable Implants , Anti-Infective Agents/therapeutic use , Treatment Outcome
3.
BMC Oral Health ; 24(1): 105, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38233815

ABSTRACT

BACKGROUND: Peri-implantitis is a polybacterial infection that can lead to the failure of dental implant rehabilitation. This study aimed to profile the microbiome of the peri-implant plaque and estimate the effect of periodontitis on it among 40 Chinese participants with dental implant prostheses and presenting with varying peri-implant and periodontal health states. METHODS: Submucosal plaque samples were collected from four distinct clinical categories based on both their implant and periodontal health status at sampling point. Clinical examinations of dental implant and remaining teeth were carried out. Metagenomic analysis was then performed. RESULTS: The microbiome of the peri-implantitis sites differed from that of healthy implant sites, both taxonomically and functionally. Moreover, the predominant species in peri-implantitis sites were slightly affected by the presence of periodontitis. T. forsythia, P. gingivalis, T. denticola, and P. endodontalis were consistently associated with peri-implantitis and inflammatory clinical parameters regardless of the presence of periodontitis. Prevotella spp. and P. endodontalis showed significant differences in the peri-implantitis cohorts under different periodontal conditions. The most distinguishing function between diseased and healthy implants is related to flagellar assembly, which plays an important role in epithelial cell invasion. CONCLUSIONS: The composition of the peri-implant microbiome varied in the diseased and healthy states of implants and is affected by individual periodontal conditions. Based on their correlations with clinical parameters, certain species are associated with disease and healthy implants. Flagellar assembly may play a vital role in the process of peri-implantitis.


Subject(s)
Dental Implants , Dental Plaque , Microbiota , Peri-Implantitis , Periodontal Diseases , Periodontitis , Humans , Peri-Implantitis/microbiology , Dental Implants/microbiology , Cross-Sectional Studies , Dental Plaque/microbiology
4.
J Clin Periodontol ; 51(2): 209-221, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37941050

ABSTRACT

AIM: To compare individuals with a periodontitis background (Grade C, stage III/IV-formerly generalized aggressive periodontitis) (H-GAP) with periodontally healthy subjects (H-Health) in terms of molecular changes (immunological/microbiological) accompanying experimental peri-implant mucositis and gingivitis. MATERIALS AND METHODS: H-GAP and control (H-Health) subjects were recruited, and experimental mucositis/gingivitis was induced around a single screw-retained implant and one contralateral tooth. Participants refrained from oral hygiene for 21 days in the selected areas, followed by professional prophylaxis and hygiene instructions for 21 days. Clinical parameters, immunological markers (multiplex analysis) and microbial data (16S rRNA gene sequencing) were collected at baseline, during induction (7, 14 and 21 days) and following remission (42 days). RESULTS: Clinically, no significant differences were observed between the groups (n = 10/each group) (H-GAP vs. H-Health) (p > .05, Mann-Whitney test) and the type of site (tooth vs. implant) (p > .05, Wilcoxon test) at the time of onset and resolution, or severity of gingival/mucosal inflammation. H-GAP displayed lower concentrations of the cytokines interleukin (IL)-1B, IL-4, IL-17, tumor necrosis factor-α and interferon-γ around implants than H-Health at baseline and during induction of mucositis (p < .05, Mann-Whitney test). In both groups, implants showed significantly higher inflammatory background at baseline and all subsequent visits when compared with teeth (p < .05, Wilcoxon test). Alpha and ß-diversity metrics showed a significant shift in the microbiome composition and abundances of core species during induction and resolution of peri-implant mucositis and gingivitis (p < .05, restricted maximum likelihood method of Shannon and Bray-Curtis indices, respectively). Differences were not significant for these parameters between the H-Health and H-GAP groups when the periodontal and peri-implant microbiomes were compared separately; however, at each time point, the peri-implant microbiome differed significantly from the periodontal microbiome. CONCLUSIONS: Within the limitations of this pilot study (e.g. low power), it can be concluded that different microbial shifts contribute to the onset and progression of inflammatory responses around teeth and implants and that history of periodontal disease experience plays an additional role in modulating the immune response of peri-implant and periodontal tissues to biofilm accumulation.


Subject(s)
Aggressive Periodontitis , Dental Implants , Gingivitis , Mucositis , Peri-Implantitis , Humans , Mucositis/etiology , Pilot Projects , RNA, Ribosomal, 16S/genetics , Dental Implants/adverse effects , Dental Implants/microbiology , Peri-Implantitis/microbiology , Gingivitis/microbiology
5.
J Appl Microbiol ; 134(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37951291

ABSTRACT

AIM: Clinically relevant in-vitro biofilm models are essential and valuable tools for mechanistically dissecting the etiopathogenesis of infectious diseases and test new antimicrobial therapies. Thus, the aim of this study was to develop and test a clinically relevant in-vitro oral polymicrobial biofilm model that mimics implant-related infections in terms of microbial profile. METHODS AND RESULTS: For this purpose, 24-well plate system was used to model oral biofilms, using three different microbial inoculums to grow in-vitro biofilms: (1) human saliva from periodontally healthy patients; (2) saliva as in inoculum 1 + Porphyromonas gingivalis strain; and (3) supra and subgingival biofilm collected from peri-implant sites of patients diagnosed with peri-implantitis. Biofilms were grown to represent the dynamic transition from an aerobic to anaerobic community profile. Subsequently, biofilms were collected after each phase and evaluated for microbiological composition, microbial counts, biofilm biomass, structure, and susceptibility to chlorhexidine (CHX). Results showed higher live cell count (P < .05) for biofilms developed from patients' biofilm inoculum, but biomass volume, dry weight, and microbiological composition were similar among groups (P > .05). Interestingly, according to the checkerboard DNA-DNA hybridization results, the biofilm developed from stimulated human saliva exhibited a microbial composition more similar to the clinical subgingival biofilm of patients with peri-implantitis, with proportions of the main pathogens closer to those found in the disease. In addition, biofilm developed using saliva as inoculum was shown to be susceptible to CHX with significant reduction in bacteria compared with biofilms without exposure to CHX (P < .05). CONCLUSION: The findings suggested that the in-vitro polymicrobial biofilm developed from human saliva as inoculum is a suitable model and clinically relevant tool for mimicking the microbial composition of implant-related infections.


Subject(s)
Communicable Diseases , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , Biofilms , Chlorhexidine , Porphyromonas gingivalis , Disease Progression , DNA
6.
Clin Oral Implants Res ; 34(9): 958-966, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37392017

ABSTRACT

OBJECTIVES: The aim of this study was to assess the prevalence of certain microbiota and their potential correlation with clinical parameters, expression of proinflammatory cytokines, Notch signalling pathway molecules and bone remodelling mediators among different peri-implant conditions. MATERIALS AND METHODS: Included participants had at least one dental implant minimally 1 year in function. They were divided into peri-implantitis (PI), peri-implant mucositis (PM) and healthy implants (HIs) groups. Prevalence of P. ginigvalis, Fusobacterium spp., EBV and C. albicans was detected in participants' crevicular fluid (CF) using quantitative real-time polymerase chain reaction, different markers' expression, as well as clinical data, were correlated with the microbial presence. RESULTS: CF samples taken from one chosen implant from each of the 102 participants were analyzed. Significantly higher levels of P. gingivalis were found in PI compared with HI (p = .012) and PM (p = .026). Fusobacterium spp. was also more prevalent in PI (p = .041) and PM (0.008) than in HI. P. gingivalis was a predictor of PPDi (p = .011, R2 = 0.063) and CALi (p = .049, R2 = 0.038). A positive correlation was found in PI for the level of Fusobacterium spp. and TNFα expression (ρ = 0.419, p = .017) while in PM, P. gingivalis and Notch 2 expression were correlated (ρ = 0.316, p = .047). CONCLUSIONS: P. gingivalis appears to be involved in the osteolysis in patients with PI, while the positive correlation of its level with Notch 2 expression in patients with PM suggests a potential involvement of P. gingivalis in the progression of PM into PI.


Subject(s)
Dental Implants , Peri-Implantitis , Humans , Dental Implants/microbiology , Cross-Sectional Studies , Peri-Implantitis/microbiology
7.
Clin Oral Implants Res ; 34(9): 979-986, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37394702

ABSTRACT

OBJECTIVES: Previous studies have indicated a progressive internal bacterial colonization of implants and possible implications for peri-implant bone loss. The aim of this study was to evaluate a decontamination protocol, two disinfectants, and a sealant for their ability to prevent such a colonization. MATERIALS AND METHODS: Bacterial samples were harvested from the peri-implant sulcus (external) and following abutment removal from the implant cavity (internal) during routine supportive peri-implant care in 30 edentulous patients 2 years after they had obtained two implants. In a split-mouth design, implants were randomly assigned to receive either internal decontamination alone (10% H2 O2 , brush) or additional placement of either sealant (GS), disinfectant agent (CHX-varnish) or disinfectant gel (1% CHX-gel), in the internal cavity before remounting of abutment/suprastructure. Twelve months later, internal and external sampling was repeated. Total bacterial counts (TBCs) were determined using real-time PCR in a total of 240 samples (eight per patient). RESULTS: Total bacterial counts in the internal cavity significantly reduced overall treatment modalities 1 year after the treatments (4.0 [2.3-6.9]-fold reduction; p = .000). No significant differences between the four treatment types were found (p = .348). Comparison of internal and external sampling points revealed significant correlation (R2 = .366; p = .000) with systematically higher TBC counts in external samples. CONCLUSIONS: Within the limitations of the present study, it can be concluded that the use of disinfectant agents or a sealant did not show an additional benefit in the prevention of internal bacterial colonization of implants compared to a decontamination protocol alone.


Subject(s)
Dental Implants , Disinfectants , Peri-Implantitis , Humans , Dental Implants/microbiology , Dental Materials , Bacteria , Bacterial Load , Peri-Implantitis/microbiology
8.
Expert Opin Ther Targets ; 27(7): 627-637, 2023.
Article in English | MEDLINE | ID: mdl-37522314

ABSTRACT

INTRODUCTION: When collected in a standardized fashion, oral fluid analysis can refine the diagnosis of periodontal and peri-implant disease. In practice, dental professionals can perform active matrix metalloproteinase (aMMP-8) analysis chairside. AREAS COVERED: Periodontal tissues are mainly made up of type I collagen, and collagen breakdown is one of the main events in periodontal and peri-implantitis destructive lesions. In addition to traditional measurements, their diagnosis can be refined with tests utilizing oral fluids. The active matrix metalloproteinase-8 (aMMP-8) is possible to be determined from the gingival crevicular fluid (GCF), peri-implant sulcus fluid (PISF), and other oral fluids such as mouth rinse and saliva. We also investigated the applicability of aMMP-8 chair-side test kits in the evaluation of oral health benefits of different adjunctive host-modulating periodontal therapies including fermented lingonberry mouthwash (FLJ) and antibacterial photodynamic therapy (aPDT). EXPERT OPINION: The aMMP-8 levels can more reliably detect early activation of periodontal and peri-implant disease as compared to traditional diagnostic methods that assess the experienced health status or past disease, rather than the present or future pathology. Novel therapies like, fermented lingonberry juice as a mouthrinse or aPDT, are potential host-modulating adjunctive treatments to reduce the signs of oral inflammation and infection.


Subject(s)
Peri-Implantitis , Periodontitis , Humans , Peri-Implantitis/diagnosis , Peri-Implantitis/therapy , Peri-Implantitis/microbiology , Point-of-Care Systems , Periodontitis/diagnosis , Periodontitis/drug therapy , Gingival Crevicular Fluid/metabolism
9.
Clin Oral Implants Res ; 34(11): 1176-1187, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37523470

ABSTRACT

AIM: To answer the following PECO question: "In systemically healthy human subjects (P), which are the differences between peri-implantitis (E) and peri-implant health/mucositis (C) in terms of bacterial presence/count (O)?" MATERIALS AND METHODS: Cross-sectional studies fulfilling specific inclusion criteria established to answer the PECO question were included. Two review authors independently searched for studies, screened the titles and abstracts, did full-text analysis, extracted the data from the included reports, and performed the risk of bias assessment through an adaptation of the Newcastle/Ottawa tool for cross-sectional studies and of the JBI critical appraisal checklist. In case of disagreement, a third reviewer author took the final decision. Study results were summarized using random effects meta-analyses. RESULTS: A total of 12 studies were included, involving 1233 participants and 1513 implants. Peri-implantitis was associated with the presence of S. epidermidis (Odds ratio, OR = 10.28 [95% Confidence interval, CI: 1.26-83.98]), F. nucleatum (OR = 7.83 [95% CI: 2.24-27.36]), T. denticola (OR = 6.11 [95% CI: 2.72-13.76]), T. forsythia (OR = 4.25 [95% CI: 1.71-10.57]), P. intermedia (OR = 3.79 [95% CI: 1.07-13.35]), and P. gingivalis (OR = 2.46 [95% CI: 1.21-5.00]). Conversely, the presence of A. actinomycetemcomitans (OR = 3.82 [95% CI: 0.59-24.68]), S. aureus (OR = 1.05 [95% CI: 0.06-17.08]), and C. rectus (OR = 1.48 [95% CI: 0.69-3.17]) was not associated with peri-implantitis. CONCLUSIONS: Peri-implantitis is associated with the presence of S. epidermidis and specific periodontopathogens (P. gingivalis, T. forsythia, T. denticola, F. nucleatum, and P. intermedia). (CRD42021254589).


Subject(s)
Dental Implants , Microbiota , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , Staphylococcus aureus , Cross-Sectional Studies , Porphyromonas gingivalis , Dental Implants/adverse effects , Dental Implants/microbiology
10.
Clin Implant Dent Relat Res ; 25(5): 938-947, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37259674

ABSTRACT

INTRODUCTION: The peri-implant sulcus is a good niche for infectious colonization such as Candida spp. In this study, the level of Candida spp. fungal colonization is analyzed in patients with peri-implantitis under supportive peri-implant therapy, as well as its correlation with the main clinicopathological data. METHODS: A case-control study was carried out on 161 patients treated with dental implants, 80 with PI and 81 without PI, which corresponded to 91 women and 70 men, whose mean age was 60.90 years. A specific protocol was completed for the clinical and implant data. Microbiological samples were taken by oral rinse and with paper tips from the peri-implant sulcus. For the quantitative and qualitative analysis Candida Chromogenic Agar/CONDA plates were incubated for 72 h at 36 + 1°C. Fungal growth was considered active when having more than 50 CFU. Specific Candida spp. cultures were later confirmed by API ID 32C and PCR. RESULTS: Fungal growth was achieved in 28% of oral rinse and 6.75% of peri-implant fluid samples. No significant differences were recognized between study groups. Most of the cultures (>65%) showed more than 50 CFU. The most frequent species were Candida albicans and Candida parapsilosis. There was no association between different PI risk factors and fungal data. The presence of Candida spp. in the oral cavity of patients with dental implants was related to total edentulism and the use of implant-fixed complete prosthesis implant-retained removable prosthesis. CONCLUSIONS: These results suggest that there is no link between PI and presence of Candida in patients with dental implants undergoing regular supportive periodontal therapy.


Subject(s)
Dental Implants , Peri-Implantitis , Male , Humans , Female , Middle Aged , Peri-Implantitis/microbiology , Dental Implants/adverse effects , Spain , Candida , Case-Control Studies
11.
Clin Implant Dent Relat Res ; 25(4): 767-781, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37386807

ABSTRACT

BACKGROUND: Antibiotics are the most effective adjuncts in the treatment of periodontitis. However, the benefits of these agents in treating peri-implantitis are still debatable and demand further analysis. PURPOSE: The aim of this review was to critically appraise the literature on the use of antibiotics to treat peri-implantitis, with the ultimate goal of supporting evidence-based clinical recommendations, defining gaps in knowledge and guiding future studies on this topic. METHODS: A systematized literature search was conducted in MEDLINE/PubMed and Cochrane Library databases for randomized clinical trials (RCTs) on patients with peri-implantitis treated by mechanical debridement-only or with adjunctive use of local or systemic antibiotics. Clinical and microbiological data were extracted from the RCTs included. The findings were critically reviewed, interpreted, and discussed. An overview of antibiotic-loaded dental implant materials in peri-implantitis treatment was also provided. RESULTS: Twelve RCTs testing local/systemic antibiotics were included. Although not always statistically significant, all antibiotic-treated groups had greater reductions in mean PD than those treated by mechanical debridement-only. The only clinically relevant antibiotic protocol supported by one RCT with low risk of bias and long-lasting benefits was systemic metronidazole (MTZ). Studies using ultrasonic debridement reported better outcomes. No RCTs to date have tested MTZ-only or with amoxicillin (AMX) as adjuncts to open-flap implant debridement. In vitro/animal studies suggested that biomaterials with antimicrobial properties are promising to treat peri-implantitis. CONCLUSION: There are insufficient data to support a particular evidence-based antibiotic protocol to treat peri-implantitis using surgical or nonsurgical therapy, but some conclusions may be drawn. Systemic MTZ adjunct to ultrasonic debridement is an effective protocol to improve the outcomes of nonsurgical treatment. Future studies should assess the clinical and microbiological effects of MTZ and MTZ + AMX as adjuncts to optimal nonsurgical implant decontamination protocols or open-flap debridement. In addition, new locally delivered drugs and antibiotic-loaded surfaces should be assessed by RCTs.


Subject(s)
Dental Implants , Peri-Implantitis , Periodontitis , Humans , Anti-Bacterial Agents/therapeutic use , Peri-Implantitis/drug therapy , Peri-Implantitis/surgery , Peri-Implantitis/microbiology , Amoxicillin/therapeutic use , Metronidazole/therapeutic use , Periodontitis/drug therapy , Periodontitis/surgery , Dental Implants/adverse effects
12.
Clin Oral Implants Res ; 34(9): 947-957, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37358250

ABSTRACT

OBJECTIVES: This study aimed to investigate the relationship between microbial communities and the severity of peri-implant mucosal bleeding in peri-implant mucositis. MATERIALS AND METHODS: Submucosal plaque samples were collected from 54 implants divided into the healthy implant (HI) group, peri-implant mucositis (PM) group, and peri-implantitis (PI) group. Sequencing of 16S rRNA was performed using the Illumina MiSeq platform. Alpha diversity (i.e., Shannon and Chao index) and beta diversity were used to measure microbial diversity within and between microbial communities, respectively. Differences in microbial taxa between groups were assessed via linear discriminate analysis effect size. Correlation between the modified sulcus bleeding index (mSBI) and microbial dysbiosis index (MDI) was examined using Spearman correlation analysis and linear models. RESULTS: The submucosal bacterial richness (Chao index) was positively correlated with the mean mSBI in the PM group. As the mean mSBI increased in the PM group, the beta diversity became closer to that of the PI group. In the PM group, the abundances of 47 genera were significantly correlated with the mean mSBI, and the MDI was positively associated with the mean mSBI. Fourteen of the forty-seven genera were discriminative taxa between the HI and PI groups, and the abundances of these biomarkers became closer to those in the PI group in the progression of peri-implant disease. CONCLUSIONS: A higher mSBI value corresponded to a higher risk of microbial dysbiosis in peri-implant mucositis. The biomarkers identified may be useful for monitoring the progression of peri-implant disease.


Subject(s)
Dental Implants , Mucositis , Peri-Implantitis , Periodontitis , Humans , Peri-Implantitis/microbiology , Dental Implants/adverse effects , Dental Implants/microbiology , Mucositis/microbiology , Dysbiosis , RNA, Ribosomal, 16S/genetics , Biomarkers
13.
New Microbiol ; 46(2): 202-206, 2023 May.
Article in English | MEDLINE | ID: mdl-37247241

ABSTRACT

Microbial biofilm is of paramount importance in the development of mucositis or peri-implantitis in patients with dental implants. This study was designed to investigate whether an electromagnetic field at high frequency waves directly applied on 33 titanium implants could remove experimentally-induced Enterococcus faecalis bacterial biofilm. A specially designed device (X-IMPLANT) was used to generate the electromagnetic field, with output power of 8 W, supply frequency (action/pause) 3/2s, and an output frequency of 625±5% kHz in plastic devices containing the biofilm-covered implants immersed in sterile saline. The bacterial biofilm on both treated and untreated control implants was quantitatively measured by phenol red-based Bio-Timer-Assay reagent. The kinetic analysis of the curves showed that the electrical treatment generated by the X-IMPLANT device completely removed the bacterial biofilm after 30 minutes of treatment (p<0.01). Elimination of the biofilm was also confirmed by chromatic observation in the macro-method. Our data seem to indicate that the procedure could be considered for clinical application in peri-implantitis to counteract bacterial biofilm on dental implants.


Subject(s)
Dental Implants , Peri-Implantitis , Humans , Peri-Implantitis/therapy , Peri-Implantitis/microbiology , Titanium , Electromagnetic Fields , Kinetics , Bacteria , Biofilms
14.
Clin Oral Implants Res ; 34(4): 393-403, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36856540

ABSTRACT

OBJECTIVES: This randomized clinical trial assessed changes in protein biomarker levels and bacterial profiles after surgical reconstructive therapy of peri-implantitis and investigated whether the adjunctive use of Er:YAG laser impacts protein biomarker and microbial outcomes. MATERIALS AND METHODS: Twenty-four patients received surgical reconstructive therapy for peri-implantitis with guided bone regeneration following mechanical debridement with (test) or without (control) the adjunctive irradiation of Er:YAG laser. Bacterial and peri-implant crevicular fluid (PICF) samples were collected over 6 months and analyzed with bacterial qPCR and luminex multiplex assays. RESULTS: Surgical reconstructive treatment significantly affected the concentration of PICF protein biomarkers, including a 50% reduction in IL-1ß between 2 and 4 weeks (p < .0001). Both MMP-9 (p < .001) and VEGF (p < .05) levels steadily decreased after treatment. In the laser group, the peak increase in IL-1ß was attenuated at 2 weeks, followed by significant reduction in MMP-9 (p < .01) and VEGF (p < .05) across all follow-up appointments compared with the control nonlaser group. The total bacterial load was reduced 2 weeks after treatment, especially in the laser group, but recolonized to presurgical levels after 4 weeks in both groups (p < .01). The composition of selective pathogens varied significantly over the follow-up, but recolonization patterns did not differ between groups. CONCLUSIONS: Reconstructive therapy of peri-implantitis significantly altered PICF protein biomarker and microbial levels during the healing process. The adjunctive use of Er:YAG laser significantly modulated the inflammatory response through reduced levels of MMP-9 and VEGF during the postsurgical period. The bacterial load was reduced immediately after therapy, but recolonization was observed by 4 weeks in both groups.


Subject(s)
Dental Implants , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , Matrix Metalloproteinase 9 , Bacterial Load , Vascular Endothelial Growth Factor A , Biomarkers/analysis , Bacteria , Lasers
15.
Beijing Da Xue Xue Bao Yi Xue Ban ; 55(1): 30-37, 2023 Feb 18.
Article in Chinese | MEDLINE | ID: mdl-36718686

ABSTRACT

OBJECTIVE: To describe the submucosal microbial profiles of peri-implantitis and healthy implants, and to explore bacteria that might be correlated with clinical parameters. METHODS: In the present cross-sectional study, 49 patients were recruited. Each patient contributed with one implant, submucosal biofilms were collected from 20 healthy implants and 29 implants with peri-implantitis. DNA was extracted and bacterial 16S ribosomal RNA (16S rRNA) genes were amplified. Submucosal biofilms were analyzed using 16S rRNA sequencing at Illumina MiSeq platform. Differences between the groups were determined by analyzing α diversity, microbial component and microbial structure. The potential correlation between the bacteria with pocket probing depth (PPD) of peri-implant calculated by Spearman correlation analysis. RESULTS: The α diversity of submucosal microbial of health group was significantly lower than that in peri-implantitis group (Chao1 index: 236.85±66.13 vs. 150.54±57.43, P < 0.001; Shannon index: 3.42±0.48 vs. 3.02±0.65, P=0.032). Principal coordinated analysis showed that the submucosal microbial structure had significant difference between healthy and peri-implantitis groups [R2=0.243, P=0.001, analysis of similarities (ANOSIM)]. Compared with healthy implants, relative abundance of periodontal pathogens were higher in peri-implantitis, including members of the red complex (Porphyromonas gingivalis, Tannerella forsythia, Treponema denticola) and some members of orange complex (Precotella intermedia, Eubacterium nodatum, Parvimonas micra), as well as some new periodontal pathogens, such as Fillifactor alocis, Fretibacterium fastidiosum, Desulfobulbus sp._HMT_041, and Porphyromonas endodontalis. Spearman correlation analysis revealed that the relative abundance of Treponema denticola (r=0.686, P < 0.001), Tannerella forsythia (r=0.675, P < 0.001), Fretibacterium sp. (r=0.671, P < 0.001), Desulfobulbus sp._HMT_041 (r=0.664, P < 0.001), Filifactor alocis (r=0.642, P < 0.001), Fretibacterium fastidiosum (r=0.604, P < 0.001), Porphyromonas gingivalis (r=0.597, P < 0.001), Porphyromonas endodontalis (r=0.573, P < 0.001) were positive correlated with PPD. While the relative abundance of Rothia aeria (r=-0.615, P < 0.001) showed negatively correlation with PPD. CONCLUSION: Marked differences were observed in the microbial profiles of healthy implants and peri-implantitis. The members of red and orange complex as well as some new periodontal pathogens seem to play an important role in peri-implant disease. Compared with healthy implants, the submucosal microbial of peri-implantitis were characterized by high species richness and diversity.


Subject(s)
Dental Implants , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , Cross-Sectional Studies , RNA, Ribosomal, 16S/genetics , Bacterial Load , Porphyromonas gingivalis
16.
J Prosthodont ; 32(9): 783-792, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36691777

ABSTRACT

PURPOSE: To characterize the microbiome composition within dental implants of peri-implantitis subjects and healthy controls using 16S rRNA gene sequencing. MATERIALS AND METHODS: Twenty-three subjects with healthy (n = 11 implants) and diseased (peri-implantitis, n = 21) implants were included in this controlled clinical cross-sectional study. Samples were obtained from internal surfaces of dental implants using sterile paper points for microbiological analysis. DNA was extracted, and the16S rRNA gene was amplified using universal primers targeting the V3-V4 regions. The resulting 16S polymerize chain reaction amplicons were sequenced on Illumina MiSeq, and the sequences were processed using DADA2 and the Human Oral Microbiome Database (HOMD) as references. Alpha and Beta diversity, as well as core microbiome and differential abundance analyses were then performed using the MicrobiomeAnalyst workflow. RESULTS: A significant increase in microbial diversity was observed in the internal implant surface of healthy implants compared with the internal surfaces of peri-implantitis (Shannon p = 0.02). Bacterial community structure was significantly different among groups (p = 0.012). High levels of Gram-positive bacteria were detected inside implants with peri-implantitis compared to healthy implants, especially Enterococci. CONCLUSIONS: There is a shift in bacterial diversity inside implants with peri-implantitis from the healthy control. The microbial colonization within that space might contribute to the etiology of peri-implant disease.


Subject(s)
Dental Implants , Microbiota , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , Dental Implants/adverse effects , RNA, Ribosomal, 16S/genetics , Cross-Sectional Studies , Microbiota/genetics
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-971270

ABSTRACT

OBJECTIVE@#To describe the submucosal microbial profiles of peri-implantitis and healthy implants, and to explore bacteria that might be correlated with clinical parameters.@*METHODS@#In the present cross-sectional study, 49 patients were recruited. Each patient contributed with one implant, submucosal biofilms were collected from 20 healthy implants and 29 implants with peri-implantitis. DNA was extracted and bacterial 16S ribosomal RNA (16S rRNA) genes were amplified. Submucosal biofilms were analyzed using 16S rRNA sequencing at Illumina MiSeq platform. Differences between the groups were determined by analyzing α diversity, microbial component and microbial structure. The potential correlation between the bacteria with pocket probing depth (PPD) of peri-implant calculated by Spearman correlation analysis.@*RESULTS@#The α diversity of submucosal microbial of health group was significantly lower than that in peri-implantitis group (Chao1 index: 236.85±66.13 vs. 150.54±57.43, P < 0.001; Shannon index: 3.42±0.48 vs. 3.02±0.65, P=0.032). Principal coordinated analysis showed that the submucosal microbial structure had significant difference between healthy and peri-implantitis groups [R2=0.243, P=0.001, analysis of similarities (ANOSIM)]. Compared with healthy implants, relative abundance of periodontal pathogens were higher in peri-implantitis, including members of the red complex (Porphyromonas gingivalis, Tannerella forsythia, Treponema denticola) and some members of orange complex (Precotella intermedia, Eubacterium nodatum, Parvimonas micra), as well as some new periodontal pathogens, such as Fillifactor alocis, Fretibacterium fastidiosum, Desulfobulbus sp._HMT_041, and Porphyromonas endodontalis. Spearman correlation analysis revealed that the relative abundance of Treponema denticola (r=0.686, P < 0.001), Tannerella forsythia (r=0.675, P < 0.001), Fretibacterium sp. (r=0.671, P < 0.001), Desulfobulbus sp._HMT_041 (r=0.664, P < 0.001), Filifactor alocis (r=0.642, P < 0.001), Fretibacterium fastidiosum (r=0.604, P < 0.001), Porphyromonas gingivalis (r=0.597, P < 0.001), Porphyromonas endodontalis (r=0.573, P < 0.001) were positive correlated with PPD. While the relative abundance of Rothia aeria (r=-0.615, P < 0.001) showed negatively correlation with PPD.@*CONCLUSION@#Marked differences were observed in the microbial profiles of healthy implants and peri-implantitis. The members of red and orange complex as well as some new periodontal pathogens seem to play an important role in peri-implant disease. Compared with healthy implants, the submucosal microbial of peri-implantitis were characterized by high species richness and diversity.


Subject(s)
Humans , Peri-Implantitis/microbiology , Cross-Sectional Studies , RNA, Ribosomal, 16S/genetics , Bacterial Load , Porphyromonas gingivalis , Dental Implants
18.
Article in English | MEDLINE | ID: mdl-36497685

ABSTRACT

The implementation of adjunctive antibiotics has been recommended for the therapy of peri-implantitis (PI). In this review, antibiotic resistance patterns in PI patients were assessed. A systematic scoping review of observational studies and trials was established in conjunction with the PRISMA extension for scoping reviews. The SCOPUS, PubMed/MEDLINE, EMBASE, SCIELO, Web of Science, and LILACS databases were reviewed along with the gray literature. The primary electronic examination produced 139 investigations. Finally, four observational studies met the selection criteria. These studies evaluated 214 implants in 168 patients. Porphyromonas gingivalis and Fusobacterium nucleatum mainly presented high resistance to tetracycline, metronidazole, and erythromycin in PI patients. Similarly, Aggregatibacter actinomycetemcomitans was also highly resistant to clindamycin and doxycycline. Other microorganisms such as Tannerella forsythia, Parvimonas micra, and Prevotella intermedia/nigrescens also presented significant levels of resistance to other antibiotics including amoxicillin, azithromycin, and moxifloxacin. However, most microorganisms did not show resistance to the combination amoxicillin metronidazole. Although the management of adjunctive antimicrobials in the therapy of PI is controversial, in this review, the resistance of relevant microorganisms to antibiotics used to treat PI, and usually prescribed in dentistry, was observed. Clinicians should consider the antibiotic resistance demonstrated in the treatment of PI patients and its public health consequences.


Subject(s)
Peri-Implantitis , Humans , Peri-Implantitis/drug therapy , Peri-Implantitis/microbiology , Aggregatibacter actinomycetemcomitans , Drug Resistance, Microbial , Fusobacterium nucleatum , Porphyromonas gingivalis , Amoxicillin , Metronidazole , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology
19.
Article in English | MEDLINE | ID: mdl-36360970

ABSTRACT

Peri-implantitis is a disease influenced by dysbiotic microbial communities that play a role in the short- and long-term outcomes of its clinical treatment. The ecological triggers that establish the progression from peri-implant mucositis to peri-implantitis remain unknown. This investigation describes the development of a novel in vitro microcosm biofilm model. Biofilms were grown over 30 days over machined titanium discs in a constant depth film fermentor (CDFF), which was inoculated (I) with pooled human saliva. Following longitudinal biofilm sampling across peri-implant health (PH), peri-implant mucositis (PM), and peri-implantitis (PI) conditions, the characterisation of the biofilms was performed. The biofilm analyses included imaging by confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM), selective and non-selective culture media of viable biofilms, and 16S rRNA gene amplification and sequencing. Bacterial qualitative shifts were observed by CLSM and SEM across conditions, which were defined by characteristic phenotypes. A total of 9 phyla, 83 genera, and 156 species were identified throughout the experiment. The phyla Proteobacteria, Bacteroidetes, Firmicutes, Fusobacteria, and Actinobacteria showed the highest prevalence in PI conditions. This novel in vitro microcosm model provides a high-throughput alternative for growing microcosm biofilms resembling an in vitro progression from PH-PM-PI conditions.


Subject(s)
Microbiota , Mucositis , Peri-Implantitis , Humans , Peri-Implantitis/microbiology , RNA, Ribosomal, 16S/genetics , Biofilms , Microbiota/genetics , Bacteria/genetics
20.
Biofouling ; 38(8): 814-823, 2022 09.
Article in English | MEDLINE | ID: mdl-36250998

ABSTRACT

This systematic review evaluated the features of the progression of experimentally induced gingivitis and peri-implant mucositis in humans. Included were studies that evaluated clinical, immunological, or microbiological responses between experimentally induced gingivitis and peri-implant mucositis in periodontally healthy patients. A total of 887 articles were initially identified, but only 12 were included in the final analysis. Implants accumulate less biofilm and suffer the most heterogeneous alterations in the microbiota, in the abstinence of oral hygiene, compared with the tooth. Interestingly, although dental implants presented less biofilm accumulation, the peri-implant mucosa showed a more exacerbated clinical response than the gingival tissue. The risk of bias of the selected studies was moderate to low, with one study presenting serious risk. The progression events of peri-implant mucositis were similar to those of experimental gingivitis but led to a different host response. This review was registered in the PROSPERO database CRD420201 123360.


Subject(s)
Dental Implants , Gingivitis , Mucositis , Peri-Implantitis , Humans , Mucositis/microbiology , Biofilms , Peri-Implantitis/microbiology , Gingivitis/microbiology , Dental Implants/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...