Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 836
Filter
1.
Sci Rep ; 14(1): 6719, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38509204

ABSTRACT

Alveolar bone loss caused by periodontal disease eventually leads to tooth loss. Periodontal ligament stem cells (PDLSCs) are the tissue-specific cells for maintaining and repairing the periodontal ligament, cementum, and alveolar bone. Here, we investigated the role of erythropoietin receptor (EPOR), which regulates the microenvironment-modulating function of mesenchymal stem cells, in PDLSC-based periodontal therapy. We isolated PDLSCs from patients with chronic periodontal disease and healthy donors, referred to as PD-PDLSCs and Cont-PDLSCs, respectively. PD-PDLSCs exhibited reduced potency of periodontal tissue regeneration and lower expression of EPOR compared to Cont-PDLSCs. EPOR-silencing suppressed the potency of Cont-PDLSCs mimicking PD-PDLSCs, whereas EPO-mediated EPOR activation rejuvenated the reduced potency of PD-PDLSCs. Furthermore, we locally transplanted EPOR-silenced and EPOR-activated PDLSCs into the gingiva around the teeth of ligament-induced periodontitis model mice and demonstrated that EPOR in PDLSCs participated in the regeneration of the periodontal ligament, cementum, and alveolar bone in the ligated teeth. The EPOR-mediated paracrine function of PDLSCs maintains periodontal immune suppression and bone metabolic balance via osteoclasts and osteoblasts in the periodontitis model mice. Taken together, these results suggest that EPOR signaling is crucial for PDLSC-based periodontal regeneration and paves the way for the development of novel options for periodontal therapy.


Subject(s)
Periodontal Diseases , Periodontitis , Humans , Mice , Animals , Periodontal Ligament , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Cells, Cultured , Cell Differentiation , Stem Cells , Periodontal Diseases/therapy , Periodontal Diseases/metabolism , Periodontitis/therapy , Periodontitis/metabolism , Ligaments , Osteogenesis/physiology
2.
Clin Rheumatol ; 43(1): 95-102, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37507614

ABSTRACT

OBJECTIVE: To investigate the association among rheumatoid arthritis (RA), saliva production, and periodontal status. METHODS: An observational study was carried out on 103 subjects with RA and 103 without RA matched by sex and age. Rheumatologic evaluation included serological and clinical variables. A full mouth periodontal examination was performed according to the American Academy of Periodontology (1999). Resting and stimulated whole salivary flows were determined after spiting during 5 min. RESULTS: RA was associated with a higher prevalence of severe periodontitis (12% vs. 4%), with a marked reduction in resting and stimulated saliva production, and with a higher prevalence of resting (19% vs. 0%) and also stimulated hyposalivation (54% vs. 10%), compared with the control group. The differences in mean resting and stimulated salivary flows between RA and control groups persisted after the exclusion of people with hyposalivation. Saliva production was not associated with the presence or the severity of periodontal disease, or with the rheumatic clinical characteristics of the patients. CONCLUSIONS: More than 50% of people with RA have some degree of reduction in their salivary flows, an affection not associated with the periodontal status or rheumatic activity, which are the expression of the two related inflammatory diseases. The influence of autonomic dysfunction on hyposalivation can be considered. While periodontitis would be a disease-associated comorbidity of RA, poor saliva production should be included among the extra-articular manifestations. Key Points • Rheumatoid arthritis patients are more prone to suffer from periodontitis and/or hyposalivation. • Periodontal disease is more prevalent in people with rheumatoid arthritis and also an association was found between the severities of both pathologies. • More than 50% of people with RA would have some degree of reduction in their salivary flows, an affection not associated with the periodontal status or rheumatic activity. • Reduced saliva production in rheumatoid arthritis patients should be included among the extra-articular manifestations.


Subject(s)
Arthritis, Rheumatoid , Periodontal Diseases , Periodontitis , Xerostomia , Humans , Periodontal Diseases/complications , Periodontal Diseases/epidemiology , Periodontal Diseases/metabolism , Periodontitis/complications , Periodontitis/epidemiology , Xerostomia/epidemiology , Saliva/metabolism
3.
J Periodontol ; 95(1): 64-73, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37436713

ABSTRACT

BACKGROUND: Mitochondria and endoplasmic reticulum are key cellular organelles and create contact sites (mitochondria-endoplasmic reticulum contact [MERC]), which plays a major role in calcium metabolism, apoptotic processes, and inflammation. Previously, proteins that have been associated with these MERC contact sites mitofusin-1 (MFN1) and mitofusin-2 (MFN2) have been found to be downregulated in periodontal disease in vitro. Therefore, the aim of the current study was to evaluate MFN1 and MFN2 in gingival crevicular fluid (GCF) of patients with periodontal disease compared with healthy controls clinically. METHODS: A total of 48 participants were divided into three groups including periodontally healthy (n = 16), patients with gingivitis (n = 16), and patients with stage 3 grade B periodontitis (n = 16). GCF levels of MFN1, MFN2, calcium (Ca), caspase-1, and tumor necrosis factor-alpha (TNF-α) were determined via enzyme-linked immunosorbent assay (ELISA). Results were calculated as total amount and concentration. RESULTS: MFN1 levels (total amount) were significantly higher in patients with periodontitis and gingivitis when compared with healthy controls (p < 0.05). However, concentration levels of MFN1, MFN2, Ca, caspase-1, TNF-α significantly decreased in periodontal disease groups compared with healthy controls (p < 0.05). A positive correlation was detected among all evaluated markers (p < 0.05). CONCLUSION: The MERC protein MFN1 may have a role in the pathogenesis of periodontal disease due to its increase in GCF of patients with periodontitis and gingivitis.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Humans , Tumor Necrosis Factor-alpha/metabolism , Calcium/metabolism , Periodontal Diseases/metabolism , Periodontitis/metabolism , Gingivitis/metabolism , Caspases/metabolism , Gingival Crevicular Fluid
4.
J Periodontal Res ; 59(2): 387-394, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38126217

ABSTRACT

OBJECTIVE: The purpose of the present research is to evaluate the salivary levels of leucine-rich alpha-2 glycoprotein (LRG) and C-reactive protein (CRP) in periodontal health and disease (gingivitis and stage III periodontitis) and also to compare the discriminative efficiencies of both biomarkers in periodontal disease. BACKGROUND: LRG is a new acute-phase protein whose functions are still being investigated. LRG and CRP are both biomarkers that are increased by inflammation. No clinical study has yet investigated the comparison of the level of LRG and CRP in periodontal health, gingivitis and periodontitis in saliva samples. METHODS: A total of 60 individuals, including 20 periodontally healthy (control group/group C), 20 with gingivitis (group G), and 20 with Stage III periodontitis (group P), who were systemically healthy and non-smokers, participated in this study. Periodontal charts were used for recording clinical periodontal parameters and saliva LRG and CRP levels were measured by ELISA. Analyzing the area under the curve (AUC) was performed by the receiver-operating characteristics curve. RESULTS: Salivary levels of LRG and CRP were significantly higher in disease groups than in group C (p < .05). Positive statistically significant correlations were observed between both biomarkers and clinical parameters (p < .05). There was also a strong positive correlation between two biomarkers (p < .05). In distinguishing periodontal disease from periodontal health, LRG (AUC = 0.833) and CRP (AUC = 0.826) were found to have similar accuracy (p = .923). CONCLUSION: LRG and CRP may be useful and similarly effective biomarkers in the diagnosis of periodontal diseases based on the findings of this study.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Humans , C-Reactive Protein/metabolism , Leucine/metabolism , Glycoproteins/metabolism , Periodontal Diseases/diagnosis , Periodontal Diseases/metabolism , Biomarkers/metabolism , Gingivitis/diagnosis , Gingivitis/metabolism , Periodontitis/metabolism , Saliva/chemistry
5.
Int J Mol Sci ; 24(19)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37834046

ABSTRACT

Periodontitis is one of the primary causes of tooth loss, and is also related to various systemic diseases. Early detection of this condition is crucial when it comes to preventing further oral damage and the associated health complications. This study offers a systematic review of the literature published up to April 2023, and aims to clearly explain the role of proteomics in identifying salivary biomarkers for periodontitis. Comprehensive searches were conducted on PubMed and Web of Science to shortlist pertinent studies. The inclusion criterion was those that reported on mass spectrometry-driven proteomic analyses of saliva samples from periodontitis cohorts, while those on gingivitis or other oral diseases were excluded. An assessment for risk of bias was carried out using the Newcastle-Ottawa Scale and Quality Assessment of Diagnostic Accuracy Studies or the NIH quality assessment tool, and a meta-analysis was performed for replicable candidate biomarkers, i.e., consistently reported candidate biomarkers (in specific saliva samples, and periodontitis subgroups, reported in ≥2 independent cohorts/reports) were identified. A Gene Ontology enrichment analysis was conducted using the Database for Annotation, Visualization, and Integrated Discovery bioinformatics resources, which consistently expressed candidate biomarkers, to explore the predominant pathway wherein salivary biomarkers consistently manifested. Of the 15 studies included, 13 were case-control studies targeting diagnostic biomarkers for periodontitis participants (periodontally healthy/diseased, n = 342/432), while two focused on biomarkers responsive to periodontal treatment (n = 26 participants). The case-control studies were considered to have a low risk of bias, while the periodontitis treatment studies were deemed fair. Summary estimate and confidence/credible interval, etc. determination for the identified putative salivary biomarkers could not be ascertained due to the low number of studies in each case. The results from the included case-control studies identified nine consistently expressed candidate biomarkers (from nine studies with 230/297 periodontally healthy/diseased participants): (i) those that were upregulated: alpha-amylase, serum albumin, complement C3, neutrophil defensin, profilin-1, and S100-P; and (ii) those that were downregulated: carbonic anhydrase 6, immunoglobulin J chain, and lactoferrin. All putative biomarkers exhibited consistent regulation patterns. The implications of the current putative marker proteins identified were reviewed, with a focus on their potential roles in periodontitis diagnosis and pathogenesis, and as putative therapeutic targets. Although in its early stages, mass spectrometry-based salivary periodontal disease biomarker proteomics detection appeared promising. More mass spectrometry-based proteomics studies, with or without the aid of already available clinical biochemical approaches, are warranted to aid the discovery, identification, and validation of periodontal health/disease indicator molecule(s). Protocol registration number: CRD42023447722; supported by RD-02-202410 and GRF17119917.


Subject(s)
Periodontal Diseases , Periodontitis , Humans , Proteomics/methods , Periodontitis/diagnosis , Periodontitis/metabolism , Mass Spectrometry/methods , Biomarkers/metabolism , Proteins/metabolism , Periodontal Diseases/metabolism , Saliva/metabolism
6.
J Periodontal Res ; 58(6): 1272-1280, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37787434

ABSTRACT

OBJECTIVE: The aim of this study was to investigate metabolomics markers in the saliva of patients with periodontal health, gingivitis and periodontitis. BACKGROUND: The use of metabolomics for diagnosing and monitoring periodontitis is promising. Although several metabolites have been reported to be altered by inflammation, few studies have examined metabolomics in saliva collected from patients with different periodontal phenotypes. METHODS: Saliva samples collected from a total of 63 patients were analysed by nuclear magnetic resonance (NMR) followed by ELISA for interleukin (IL)-1ß. The patient sample, well-characterised clinically, included periodontal health (n = 8), gingivitis (n = 19) and periodontitis (n = 36) cases, all non-smokers and not diabetic. RESULTS: Periodontal diagnosis (healthy/gingivitis/periodontitis) was not associated with any salivary metabolites in this exploratory study. Periodontal staging showed nominal associations with acetoin (p = .030) and citrulline (p = .047). Among other investigated variables, the use of systemic antibiotics in the previous 3 months was associated with higher values of the amino acids taurine, glycine and ornithine (p = .002, p = .05 and p = .005, respectively, at linear regression adjusted for age, gender, ethnicity, body mass index and staging). CONCLUSION: While periodontal staging was marginally associated with some salivary metabolites, other factors such as systemic antibiotic use may have a much more profound effect on the microbial metabolites in saliva. Metabolomics in periodontal disease is still an underresearched area that requires further observational studies on large cohorts of patients, aiming to obtain data to be used for clinical translation.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Humans , Saliva/chemistry , Periodontitis/metabolism , Gingivitis/metabolism , Periodontal Diseases/metabolism , Biomarkers/metabolism
7.
PLoS Pathog ; 19(10): e1011743, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37871107

ABSTRACT

Gram-negative bacteria derived extracellular vesicles (EVs), also known as outer membrane vesicles, have attracted significant attention due to their pathogenic roles in various inflammatory diseases. We recently demonstrated that EVs secreted by the periodontopathogen Aggregatibacter actinomycetemcomitans (Aa) can cross the blood-brain barrier (BBB) and that their extracellular RNA cargo can promote the secretion of proinflammatory cytokines, such as IL-6 and TNF-α, in the brain. To gain more insight into the relationship between periodontal disease (PD) and neuroinflammatory diseases, we investigated the effect of Aa EVs in a mouse model of ligature-induced PD. When EVs were administered through intragingival injection or EV-soaked gel, proinflammatory cytokines were strongly induced in the brains of PD mice. The use of TLR (Toll-like receptor)-reporter cell lines and MyD88 knockout mice confirmed that the increased release of cytokines was triggered by Aa EVs via TLR4 and TLR8 signaling pathways and their downstream MyD88 pathway. Furthermore, the injection of EVs through the epidermis and gingiva resulted in the direct retrograde transfer of Aa EVs from axon terminals to the cell bodies of trigeminal ganglion (TG) neurons and the subsequent activation of TG neurons. We also found that the Aa EVs changed the action potential of TG neurons. These findings suggest that EVs derived from periodontopathogens such as Aa might be involved in pathogenic pathways for neuroinflammatory diseases, neuropathic pain, and other systemic inflammatory symptoms as a comorbidity of periodontitis.


Subject(s)
Extracellular Vesicles , Periodontal Diseases , Periodontitis , Mice , Animals , Neuroinflammatory Diseases , Trigeminal Ganglion , Myeloid Differentiation Factor 88/metabolism , Periodontitis/metabolism , Periodontal Diseases/metabolism , Blood-Brain Barrier/metabolism , Cytokines/metabolism , Mice, Knockout , Extracellular Vesicles/metabolism
8.
Tissue Cell ; 84: 102184, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37541115

ABSTRACT

Periodontitis is characterized by periodontal destruction triggered by chronic inflammation. The optimal treatment for periodontitis is to improve the periodontal microenvironment, reduce inflammation and achieve periodontal regeneration. Recently, the role of TRPM2 in inflammatory diseases has been reported. However, the function of TRPM2 in periodontal disease and the biological mechanism remain elusive. Therefore, this study aimed to identify the role and explore the underlying mechanisms of TRPM2 in periodontal disease. Here, we first identified the characterization of human periodontal ligament stem cells (PDLSCs). Oil Red O Staining and Alizarin Red mineralized matrix were used to evaluate the multi-differentiation capacity of cells. Flow cytometry was employed to detect MSC-specific surface markers of hPDLSCs. hPDLSCs were treated with 0, 5, 10 or 40 µg/mL of TNF-α for 72 h. Western blot assay were performed to examine the expression of Transient receptor potential cation channel, subfamily M, member 2 (TRPM2) in hPDLSCs. CCK8 and colony formation assays were used to detect the cell viability and proliferation of hPDLSCs, which revealed that TRPM2 knockdown promoted hPDLSCs proliferation. Then, ALP activity in hPDLSCs was detected by ALP activity detection kit. Next, the expression of ALP and Runx2 in hPDLSCs was detected by immunofluorescence staining. The result showed that TRPM2 knockdown promoted osteogenic differentiation and affected the genes expression of osteogenic. Finally, the expressions of p-p65, p65, p-IκBα, IκBα and NLRP3 in hPDLSCs were detected by western blot assay. Together, these results suggested that knockdown of TRPM2 accelerated osteogenic differentiation of hPDLSCs through mediating NF-κB /NLRP3 pathway.


Subject(s)
Periodontal Diseases , Periodontitis , TRPM Cation Channels , Humans , NF-kappa B/metabolism , Periodontal Ligament , NF-KappaB Inhibitor alpha/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Osteogenesis/genetics , Cell Differentiation/genetics , Periodontitis/metabolism , Inflammation/metabolism , Periodontal Diseases/metabolism , Stem Cells , Cells, Cultured , Cell Proliferation
9.
Nutrients ; 15(15)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37571281

ABSTRACT

Although numerous studies have been published investigating the relationship between various dietary components and inflammatory periodontal disease, it has not yet been possible to clearly distinguish between periodontally healthy and unhealthy diets. This clinical study aimed to assess the association of specific food ingredients and physical activity on local and systemic inflammatory signs in experimentally induced gingivitis. Thirty-nine non-smoking periodontally healthy volunteers (mean age 23.2 ± 3.8 years) refrained from oral hygiene in the right maxilla for 21 days to induce an experimental gingivitis. Clinical examination (baseline and day 21) included plaque index, bleeding on probing (BOP), gingival crevicular fluid volume and high sensitive C-reactive protein levels (blood sample). Accompanying the intervention, volunteers documented with validated questionnaires their physical activity converted into metabolic equivalent (MET) and their nutrition converted into the dietary inflammatory index (DII). Significantly lower BOP (p = 0.039) was found for subjects with a more anti-inflammatory DII than for those with a more pro-inflammatory DII; higher MET values were correlated with lower BOP at day 21 (correlation coefficient -0.36). The results show an influence of nutrition and physical activity on periodontal inflammation signs. The DII may be a suitable parameter to verify the relationship between nutrition and inflammatory periodontal diseases.


Subject(s)
Gingivitis , Periodontal Diseases , Humans , Young Adult , Adult , Periodontal Diseases/metabolism , Inflammation/metabolism , Gingival Crevicular Fluid/metabolism , Exercise
10.
J Periodontal Res ; 58(4): 715-722, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37186464

ABSTRACT

OBJECTIVES: To determine the abilities of salivary E-cadherin to differentiate between periodontal health and periodontitis and to discriminate grades of periodontitis. BACKGROUND: E-cadherin is the main protein responsible for maintaining the integrity of epithelial-barrier function. Disintegration of this protein is one of the events associated with the destructive forms of periodontal disease leading to increase concentration of E-cadherin in the oral biofluids. MATERIALS AND METHODS: A total of 63 patients with periodontitis (case) and 35 periodontally healthy subjects (control) were included. For each patient, periodontal parameters including bleeding on probing (BOP), probing pocket depth (PPD), and clinical attachment level (CAL) were recorded. Concentration of salivary E-cadherin was determined by ELISA. Receiver operating characteristic (ROC) curve and area under the curve (AUC) were used to determine the diagnostic potentials of E-cadherin. RESULTS: Level of salivary E-cadherin was significantly higher in periodontitis cases than controls. The ROC analysis showed that salivary E-cadherin exhibits excellent sensitivity and specificity (AUC 1.000) to differentiate periodontal health from periodontitis with a cutoff concentration equal to 1.325 ng/mL. The AUCs of E-cadherin to differentiate grade A from grade B and C periodontitis were 0.731 (cutoff point = 1.754 ng/mL) and 0.746 (cutoff point = 1.722 ng/mL), respectively. However, the AUC of salivary E-cadherin to differentiate grade B from grade C periodontitis was lower (0.541). Additionally, BOP and PPD were significantly and positively correlated with the concentration of salivary E-cadherin. CONCLUSION: Salivary E-cadherin exhibited excellent sensitivity and specificity to differentiate periodontitis from a healthy periodontium. The level of accuracy of E-cadherin was also sufficient to recognize grade A periodontitis from grade B and C periodontitis.


Subject(s)
Periodontal Diseases , Periodontitis , Humans , Periodontitis/diagnosis , Periodontitis/metabolism , Periodontal Diseases/metabolism , Periodontium/metabolism , Biomarkers/metabolism , Cadherins/metabolism , Saliva/chemistry
11.
Inflamm Res ; 72(4): 859-873, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36912916

ABSTRACT

INTRODUCTION: The role of suppressor of cytokine signaling 2 (SOCS2) in Aggregatibacter actinomycetemcomitans (Aa)-induced alveolar bone loss is unknown; thus, it was investigated in this study. METHODS: Alveolar bone loss was induced by infecting C57BL/6 wild-type (WT) and Socs2-knockout (Socs2-/-) mice with Aa. Bone parameters, bone loss, bone cell counts, the expression of bone remodeling markers, and cytokine profile were evaluated by microtomography, histology, qPCR, and/or ELISA. Bone marrow cells (BMC) from WT and Socs2-/- mice were differentiated in osteoblasts or osteoclasts for analysis of the expression of specific markers. RESULTS: Socs2-/- mice intrinsically exhibited irregular phenotypes in the maxillary bone and an increased number of osteoclasts. Upon Aa infection, SOCS2 deficiency resulted in the increased alveolar bone loss, despite decreased proinflammatory cytokine production, in comparison to the WT mice. In vitro, SOCS2 deficiency resulted in the increased osteoclasts formation, decreased expression of bone remodeling markers, and proinflammatory cytokines after Aa-LPS stimulus. CONCLUSIONS: Collectively, data suggest that SOCS2 is a regulator of Aa-induced alveolar bone loss by controlling the differentiation and activity of bone cells, and proinflammatory cytokines availability in the periodontal microenvironment and an important target for new therapeutic strategies. Thus, it can be helpful in preventing alveolar bone loss in periodontal inflammatory conditions.


Subject(s)
Alveolar Bone Loss , Periodontal Diseases , Mice , Animals , Alveolar Bone Loss/genetics , Aggregatibacter actinomycetemcomitans/metabolism , Mice, Inbred C57BL , Periodontal Diseases/metabolism , Osteoclasts/metabolism , Cytokines/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
12.
J Periodontal Res ; 58(2): 433-443, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36717759

ABSTRACT

OBJECTIVE: The objective of the study was to profile the expression level of histone deacetylase enzymes (HDACs) in human saliva in periodontal health, gingivitis and periodontitis. BACKGROUND: HDACs are epigenetic modulators and a group of enzymes that catalyse the removal of acetyl functional groups from the lysine residues of both histone and nonhistone proteins. HDACs have been detected in gingival tissues and may provide valuable insight into the periodontal inflammatory response. However, no studies have investigated the expression of HDACs in saliva from periodontitis-affected individuals and their capacity for periodontal diagnostics and screening. MATERIALS AND METHODS: Whole unstimulated saliva was collected from 53 participants (17 healthy, 14 gingivitis and 22 stages III/IV periodontitis). The expression of 11 HDACs in saliva samples was determined using RT-qPCR and diagnostic power was calculated using the receiver operating characteristic (ROC) curves and area under the ROC Curve (AUC). RESULTS: Relative to health, the expression of HDAC4, 8 and 10 was downregulated in gingivitis, and the expression of HDAC4, 6, 8 and 9 was downregulated in periodontitis. Increased HDAC1 and decreased HDAC9 expression were observed in periodontitis compared to gingivitis. Higher HDAC1 and lower HDAC6 and 9 expression was observed in periodontitis compared to non-periodontitis (combining health and gingivitis). Expression of HDAC3, 4, 8, 9 and 10 was significantly decreased in periodontal disease (combining gingivitis and periodontitis) compared to health. HDAC4 and 8 exhibited an excellent diagnostic capacity for distinguishing gingivitis and periodontal disease from health (AUC 0.79-0.86). HDAC9 showed an acceptable power in discriminating periodontitis from health, gingivitis and non-periodontitis (AUC 0.76-0.80). Salivary HDAC enzyme activity showed no significant difference among the groups. CONCLUSION: This pilot study has demonstrated the differential expression of HDACs in human saliva for the first time and identified HDAC4, 8 and 9 as potential biomarkers in periodontal diagnosis.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Humans , Pilot Projects , Cross-Sectional Studies , Periodontal Diseases/metabolism , Periodontitis/metabolism , Gingivitis/diagnosis , Gingivitis/metabolism , Biomarkers/metabolism , Saliva/chemistry
13.
J Periodontal Res ; 58(1): 131-142, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36445954

ABSTRACT

BACKGROUND AND OBJECTIVE: Teeth overeruption is a problem of clinical significance, but the underlying mechanism how changes in external occlusal force convert to the periodontium remodeling signals has been a largely under explored domain. And recently, periodontal ligament-associated protein-1 (PLAP-1)/asporin was found to play a pivotal role in maintaining periodontal homeostasis. The aim of this study was to explore the function of PLAP-1 in the periodontally hypofunctional tissue turnover. METHODS: After extracting left maxillary molars in mice, the left and right mandibular molars were distributed into hypofunction group (HG) and control group (CG), respectively. Mice were sacrificed for radiographic, histological, and molecular biological analyses after 1, 4 and 12 weeks. In vitro, dynamic compression was applied using Flexcell FX-5000 Compression System to simulate intermittent occlusal force. The expression of PLAP1 in loaded and unloaded human periodontal ligament cells (hPDLCs) was compared, and its molecular biological effects were further explored by small interfering RNA (siRNA) targeting PLAP1. RESULTS: In vivo, fiber disorder in periodontal ligament (PDL), bone apposition at furcation regions, and bone resorption in alveolar bone were illustrated in the HG compared with the CG. In addition, PLAP-1 positive area decreased significantly in PDL following occlusal unloading. In vitro, the loss of compressive loading relatively downregulated PLAP1 expression, which was essential to promote collagen I but inhibit osterix and osteocalcin expression in hPDLCs. CONCLUSIONS: PLAP-1 presumably plays a pivotal role in occlusal force-regulated periodontal homeostasis by facilitating collagen fiber synthesis in hPDLCs and suppressing excessive osteoblast differentiation, further preventing teeth from overeruption. Further evidence in PLAP-1 conditional knockout mice is needed.


Subject(s)
Periodontal Diseases , Tooth , Animals , Humans , Mice , Collagen/metabolism , Periodontal Diseases/metabolism , Periodontal Ligament , Periodontium
14.
Arch Oral Biol ; 145: 105574, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36395562

ABSTRACT

OBJECTIVES: Salivary statherin and alpha-amylase play significant roles in biofilm formation and pathogenic bacteria adhesion. Examination of these proteins may provide information on their roles in periodontal diseases. The present study was based on the hypothesis that; the salivary proteins -statherin and alpha-amylase- effective on biofilm formation, may play important roles in the etiology of periodontal disease. Therefore, we aimed to analyze the differences in periodontal diseases compared to periodontal health in order to search their roles in periodontal disease. METHODS: Patients with gingivitis (n = 26) and periodontitis (n = 20), and periodontally healthy individuals (n = 21) were included in this study. Unstimulated whole saliva samples were obtained from a total of 67 individuals. Salivary statherin level and alpha-amylase activity were determined using ELISA and enzymatic methods, respectively. RESULTS: Statherin levels in saliva were significantly higher in the periodontitis group compared to the gingivitis group (p = 0.014), while alpha-amylase activities and total protein levels were slightly higher in the periodontitis and gingivitis groups compared to controls, without significant differences among the groups (p = 0.295 and p = 0.019, respectively). Statherin levels showed positive correlations with gingival and plaque indices in the disease groups. CONCLUSIONS: The results suggest that statherin level in saliva increase to provide a protective effect against periodontitis, and higher salivary statherin level is related to the degree of gingival inflammation and plaque accumulation.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Humans , alpha-Amylases/metabolism , Bacterial Adhesion , Gingivitis/metabolism , Periodontal Diseases/metabolism , Periodontitis/metabolism , Saliva/metabolism
15.
Immunopharmacol Immunotoxicol ; 45(1): 94-101, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36053007

ABSTRACT

CONTEXT: Many reports in the literature have suggested the therapeutic value of carbon monoxide-releasing molecules (CORMs) against various diseases. However, to date, little is known about their possible influence on periodontal disease. OBJECTIVE: This study was performed to investigate the influence of CORM-401 on the generation of nitric oxide (NO) in murine macrophage cells activated with lipopolysaccharide (LPS) derived from Prevotella intermedia, a pathogen associated with periodontal disease. MATERIALS AND METHODS: LPS was isolated by the hot phenol-water method. Culture supernatants were analyzed for NO. Real-time PCR and immunoblotting were conducted to quantify mRNA and protein expression, respectively. NF-κB-dependent SEAP levels were estimated by reporter assay. DNA-binding of NF-κB was also analyzed. RESULTS: CORM-401 caused an apparent suppression of NO production through inhibition of iNOS at both the mRNA and protein levels in RAW264.7 cells stimulated with P. intermedia LPS. CORM-401 upregulated the expression of both the HO-1 gene and its protein in LPS-activated cells, and treatment with the HO-1 inhibitor significantly reversed the attenuating influence of CORM-401 against LPS-induced generation of NO. CORM-401 caused an apparent attenuation of NF-κB-dependent SEAP release induced by LPS. IκB-α degradation and nuclear translocation of NF-κB p50 subunit induced by LPS were significantly reduced by CORM-401. Additionally, CORM-401 significantly attenuated DNA-binding of p65 and p50 induced by LPS. CORM-401 attenuated NO generation induced by P. intermedia LPS independently of PPAR-γ, JNK, p38 and STAT1/3. CONCLUSION: The modulation of host inflammatory response by CORM-401 might be of help in the therapy of periodontal disease.


Subject(s)
NF-kappa B , Periodontal Diseases , Animals , Mice , NF-kappa B/metabolism , Lipopolysaccharides/toxicity , Manganese/metabolism , Prevotella intermedia/chemistry , Prevotella intermedia/metabolism , Nitric Oxide/metabolism , Carbon Monoxide/metabolism , Macrophages/metabolism , Periodontal Diseases/metabolism , RNA, Messenger/metabolism , DNA/metabolism , Nitric Oxide Synthase Type II/metabolism
16.
Front Immunol ; 13: 1044334, 2022.
Article in English | MEDLINE | ID: mdl-36341447

ABSTRACT

Aging is a gradual and progressive deterioration of integrity across multiple organ systems that negatively affects gingival wound healing. The cellular responses associated with wound healing, such as collagen synthesis, cell migration, proliferation, and collagen contraction, have been shown to be lower in gingival fibroblasts (the most abundant cells from the connective gingival tissue) in aged donors than young donors. Cellular senescence is one of the hallmarks of aging, which is characterized by the acquisition of a senescence-associated secretory phenotype that is characterized by the release of pro-inflammatory cytokines, chemokines, growth factors, and proteases which have been implicated in the recruitment of immune cells such as neutrophils, T cells and monocytes. Moreover, during aging, macrophages show altered acquisition of functional phenotypes in response to the tissue microenvironment. Thus, inflammatory and resolution macrophage-mediated processes are impaired, impacting the progression of periodontal disease. Interestingly, salivary antimicrobial peptides, such as histatins, which are involved in various functions, such as antifungal, bactericidal, enamel-protecting, angiogenesis, and re-epithelization, have been shown to fluctuate with aging. Several studies have associated the presence of Porphyromonas gingivalis, a key pathogen related to periodontitis and apical periodontitis, with the progression of Alzheimer's disease, as well as gut, esophageal, and gastric cancers. Moreover, herpes simplex virus types 1 and 2 have been associated with the severity of periodontal disease, cardiovascular complications, and nervous system-related pathologies. This review encompasses the effects of aging on periodontal tissues, how P. gingivalis and HSV infections could favor periodontitis and their relationship with other pathologies.


Subject(s)
Periodontal Diseases , Periodontitis , Humans , Gingiva/pathology , Porphyromonas gingivalis , Periodontium , Periodontal Diseases/metabolism
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 842-850, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-36224687

ABSTRACT

Objective: To analyze the salivary metabolic profile of patients with periodontitis through metabolomic techniques and to explore the metabolic patterns associated with periodontal diseases. Methods: Liquid chromatography/mass spectrometry (LC/MS) technique in conjunction with principal component analysis (PCA) analysis and orthogonal partial least squares identification (OPLS-DA) method was used to study the metabolomics of saliva samples from gingivitis patients, periodontitis patients, and healthy controls, with 10 samples for each group. We examined the correlation between migration in metabolic profile and the progression of periodontal diseases. Results: Saliva metabolite profiles of gingivitis and periodontitis patients was significantly different from those of the healthy controls. Significant differences were identified between the different groups for eight salivary metabolites, including arachidonic acid, tyramine, L-arginine, thymine, N-acetylgalactosamine sulfate, prostaglandin E2, L-phenylalanine, and 5-aminoimidazole-4-carboxamide-riboside (AICAR). In comparison with those of the health controls, the concentration of AICAR in patients with gingivitis and periodontitis was lower and the metabolic trend was down-regulated, while the other metabolites were up-regulated. Conclusion: Salivary metabolic profile changes along with the progression of periodontal diseases. Abnormal metabolism of the periodontal tissue and of pathogenic microorganisms related to periodontal diseases is one of the mechanisms involved in the pathogenesis, development and prognosis of periodontal diseases.


Subject(s)
Gingivitis , Periodontal Diseases , Periodontitis , Acetylgalactosamine , Arachidonic Acid , Arginine , Biomarkers/analysis , Dinoprostone , Gingivitis/metabolism , Humans , Metabolomics , Periodontal Diseases/metabolism , Periodontal Diseases/pathology , Periodontitis/metabolism , Phenylalanine , Sulfates , Thymine , Tyramine
18.
Front Immunol ; 13: 998244, 2022.
Article in English | MEDLINE | ID: mdl-36304447

ABSTRACT

Periodontitis involves the loss of connective tissue attachment and alveolar bone. Single cell RNA-seq experiments have provided new insight into how resident cells and infiltrating immune cells function in response to bacterial challenge in periodontal tissues. Periodontal disease is induced by a combined innate and adaptive immune response to bacterial dysbiosis that is initiated by resident cells including epithelial cells and fibroblasts, which recruit immune cells. Chemokines and cytokines stimulate recruitment of osteoclast precursors and osteoclastogenesis in response to TNF, IL-1ß, IL-6, IL-17, RANKL and other factors. Inflammation also suppresses coupled bone formation to limit repair of osteolytic lesions. Bone lining cells, osteocytes and periodontal ligament cells play a key role in both processes. The periodontal ligament contains cells that exhibit similarities to tendon cells, osteoblast-lineage cells and mesenchymal stem cells. Bone lining cells consisting of mesenchymal stem cells, osteoprogenitors and osteoblasts are influenced by osteocytes and stimulate formation of osteoclast precursors through MCSF and RANKL, which directly induce osteoclastogenesis. Following bone resorption, factors are released from resorbed bone matrix and by osteoclasts and osteal macrophages that recruit osteoblast precursors to the resorbed bone surface. Osteoblast differentiation and coupled bone formation are regulated by multiple signaling pathways including Wnt, Notch, FGF, IGF-1, BMP, and Hedgehog pathways. Diabetes, cigarette smoking and aging enhance the pathologic processes to increase bone resorption and inhibit coupled bone formation to accelerate bone loss. Other bone pathologies such as rheumatoid arthritis, post-menopausal osteoporosis and bone unloading/disuse also affect osteoblast lineage cells and participate in formation of osteolytic lesions by promoting bone resorption and inhibiting coupled bone formation. Thus, periodontitis involves the activation of an inflammatory response that involves a large number of cells to stimulate bone resorption and limit osseous repair processes.


Subject(s)
Bone Resorption , Periodontal Diseases , Periodontitis , Humans , Hedgehog Proteins/metabolism , Osteoblasts/metabolism , Bone Resorption/pathology , Periodontitis/pathology , Periodontal Diseases/metabolism
19.
J Periodontal Res ; 57(6): 1127-1138, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36070347

ABSTRACT

OBJECTIVES: Billions of individuals worldwide suffer from periodontal disease, an inflammatory disease that results in hard-tissue and soft-tissue destruction. A viable therapeutic option to treat periodontal disease may be via cannabinoids that exert immunomodulatory effects, and the endocannabinoid system (ECS) is readily present in periodontal tissues that exhibit cannabinoid type 1 and 2 receptors (CB1R and CB2R). Phytocannabinoids (pCBs), which are a part of a heterogeneous group of molecules acting on cannabinoid receptors (CBR) derived from the cannabis plants, have been attributed to a wide variety of effects including anti-inflammatory activity and some pro-inflammatory effects depending on the cell type. Thus, this study aims to examine the effects of pCBs on primary human gingival fibroblasts (HGFs) in IL-1ß stimulated (simulated periodontal disease) HGFs. MATERIALS AND METHODS: Human gingival fibroblasts (HGFs) obtained from ATCC were cultured per the manufacturer's recommendation. The functional activity of cannabinoid receptors was measured using ACTOne (cAMP)-based CB1R and CB2R assay. The effects of three pCBs (0.1-10 µg/ml or 10-4.5 -10-6.5  M) on cell viability were assessed using the CCK-8 cellular dehydrogenase assay. IL-1ß (1 ng/ml) was added an hour before the treatment to stimulate inflammation in the HGFs before the addition of cannabinoid ligands. After 24-h incubation, the production of INF-γ, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, and TNF-α was measured using Mesoscale Discovery (MSD) Human Pro-Inflammatory kit. To measure prostaglandin E 2 levels (PGE2), Cisbio HTRF PGE2 assay kit was used per the manufacturer's recommendation to measure after 24-h incubation. The data were analyzed using GraphPad Prism 6.0. The analytes for each group were compared using a one-way ANOVA test with Bonferroni's correction. RESULTS: Cannabidivarin (CBVN or CBDV) (EC50  = 12 nM) and cannabigerol (CBG) (EC50  = 30 nM) exhibited agonist activity on CB2R with intermediate efficacy. Cannabidiol (CBD) did not exhibit activation of the CB2R, and the CB1R activation was not observed with any of the pCBs. Cytotoxicity results showed that concentrations of 2.50 µg/ml or greater for the pCBs were toxic except for CBVN. Lower concentrations of CBD and CBG (0.1-0.75 µg/ml), and CBVN at 2.50 µg/ml exhibited significant effects on HGF proliferation. In IL-1ß-stimulated HGFs, prostaglandin E2 (PGE2) production was significantly suppressed only by CBG and CBVN. CBD and CBG treatment alone did, however, elevate PGE2 production significantly compared to control. IL-1ß stimulation resulted in a robust increase in the production of all cytokines tested. Treatment of IL-ß-stimulated HGF with the three pCBs (1 µg/ml) significantly reduced INF-É£, TNF-α, and IL-2. The significant suppression of IL-4 was seen with CBD and CBVN, while only CBVN exerted suppression of IL-13. The three pCBs significantly increased IL-6, IL-10, and IL-12 levels, while none of the pCBs reduced the expression of IL-8 in IL-1ß-stimulated HGF. CONCLUSION: The effective inhibition of IL-1ß-stimulated production of PGE2 and cytokines by the pCB in HGFs suggests that targeting the endocannabinoid system may lead to the development of therapeutic strategies for periodontal therapy. However, each pCB has its unique anti-inflammatory profile, in which certain pro-inflammatory activities are also exhibited. The pCBs alone or in combination may benefit and aid in improving public oral health.


Subject(s)
Cannabinoids , Periodontal Diseases , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Cannabinoids/pharmacology , Cells, Cultured , Cytokines/metabolism , Dinoprostone/metabolism , Endocannabinoids/pharmacology , Fibroblasts , Gingiva/metabolism , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-13/metabolism , Interleukin-13/pharmacology , Interleukin-1beta/pharmacology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-4 , Interleukin-6/metabolism , Interleukin-8/metabolism , Periodontal Diseases/metabolism , Receptors, Cannabinoid/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Sci Rep ; 12(1): 11284, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35788667

ABSTRACT

The objective of this pilot clinical study was to identify salivary biomarkers that are associated with periodontal disease and measures of diabetic autonomic dysfunction. Saliva samples from 32 participants were obtained from 3 groups: healthy (H), type 1 diabetes mellitus (DM), and type 1 diabetes mellitus with neuropathy (DMN). Based on the periodontal examination, individuals' mean Periodontal Screening and Recording scores were categorized into two groups (periodontally healthy and gingivitis), and correlated to specific salivary inflammatory biomarkers assessed by a customized protein array and enzyme assay. The mean salivary IgA level in DM was 9211.5 ± 4776.4 pg/ml, which was significantly lower than H (17,182.2 ± 8899.3 pg/ml). IgA in DMN with healthy periodontium was significantly lower (5905.5 ± 3124.8 pg/ml) compared to H, although IgA levels in DMN patients with gingivitis (16,894. 6 ± 7084.3) were not. According to the result of a logistic regression model, IgA and periodontal condition were the indicators of the binary response given by H versus DM, and H versus DMN, respectively. These data suggest that selected salivary biomarkers, such as IgA, combined with a periodontal examination prior to obtaining salivary samples can offer a non-invasive method to assess risk for developing diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetic Neuropathies , Gingivitis , Periodontal Diseases , Periodontitis , Biomarkers/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetic Neuropathies/complications , Diabetic Neuropathies/etiology , Gingivitis/complications , Humans , Immunoglobulin A/metabolism , Periodontal Diseases/metabolism , Periodontitis/complications , Periodontitis/diagnosis , Periodontitis/metabolism , Saliva/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...