Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Saudi Dent J ; 36(3): 492-497, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38525187

ABSTRACT

Background: Although the role of chlorhexidine and other mouthwashes in periodontal therapy has been elucidated, little information is available on their use as routine preoperative mouth rinses before surgery, especially in periodontal procedures such as dental implant surgery. Objective: This study aimed to compare the efficacy of preoperative chlorhexidine, essential oil, and cetylpyridinium chloride mouthwashes in reducing bacterial contamination at the time of implant placement. Materials and Methods: Eligible patients who underwent dental implant surgery were randomly divided into four groups based on the mouthwash used: (1) 0.12 % chlorhexidine, (2) essential oil, (3) cetylpyridinium chloride, and (4) saline (served as the control group). All the patients of each group rinsed preoperatively with 15 mL of the respective mouthwash for 60 s. Saliva samples before (pre) and immediately after rinsing with the mouthwash (post) and after suturing the flap (end) were collected on the day of the implant placement. Real-time quantitative polymerase chain reaction (qPCR) was performed to analyze the samples and quantify the targeted periodontal pathogens using a propidium monoazide (PMA) dye. Results: Forty patients were included in the study. Real-time qPCR demonstrated a significant reduction in the number of pathogens in the saliva samples of the mouthwash groups compared to that of the control group. A statistically significant difference was observed between the groups for the pre-post and pre-end samples (p < 0.001) but not for the post-end samples (p = 0.203). A statistically significant difference was observed between the chlorhexidine, essential oil, and cetylpyridinium chloride mouthwash groups and the saline group (P < 0.001). The bacterial counts significantly differed with and without the use of the PMA dye. Conclusions: Preoperative chlorhexidine, essential oil, and cetylpyridinium chloride mouthwashes can reduce the bacterial load at the time of implant placement, thereby reducing the incidence of implant-related complications.

2.
J Cell Mol Med ; 27(12): 1750-1756, 2023 06.
Article in English | MEDLINE | ID: mdl-37170687

ABSTRACT

Bone remodelling is mediated by orchestrated communication between osteoclasts and osteoblasts which, in part, is regulated by coupling and anti-coupling factors. Amongst formally known anti-coupling factors, Semaphorin 4D (Sema4D), produced by osteoclasts, plays a key role in downmodulating osteoblastogenesis. Sema4D is produced in both membrane-bound and soluble forms; however, the mechanism responsible for producing sSema4D from osteoclasts is unknown. Sema4D, TACE and MT1-MMP are all expressed on the surface of RANKL-primed osteoclast precursors. However, only Sema4D and TACE were colocalized, not Sema4D and MT1-MMP. When TACE and MT1-MMP were either chemically inhibited or suppressed by siRNA, TACE was found to be more engaged in shedding Sema4D. Anti-TACE-mAb inhibited sSema4D release from osteoclast precursors by ~90%. Supernatant collected from osteoclast precursors (OC-sup) suppressed osteoblastogenesis from MC3T3-E1 cells, as measured by alkaline phosphatase activity, but OC-sup harvested from the osteoclast precursors treated with anti-TACE-mAb restored osteoblastogenesis activity in a manner that compensates for diminished sSema4D. Finally, systemic administration of anti-TACE-mAb downregulated the generation of sSema4D in the mouse model of critical-sized bone defect, whereas local injection of recombinant sSema4D to anti-TACE-mAb-treated defect upregulated local osteoblastogenesis. Therefore, a novel pathway is proposed whereby TACE-mediated shedding of Sema4D expressed on the osteoclast precursors generates functionally active sSema4D to suppress osteoblastogenesis.


Subject(s)
Osteoclasts , Semaphorins , Animals , Mice , Disease Models, Animal , Matrix Metalloproteinase 14/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Semaphorins/genetics , Semaphorins/metabolism
3.
Int J Mol Sci ; 23(10)2022 May 18.
Article in English | MEDLINE | ID: mdl-35628440

ABSTRACT

It is well known that Semaphorin 4D (Sema4D) inhibits IGF-1-mediated osteogenesis by binding with PlexinB1 expressed on osteoblasts. However, its elevated level in the gingival crevice fluid of periodontitis patients and the broader scope of its activities in the context of potential upregulation of osteoclast-mediated periodontal bone-resorption suggest the need for further investigation of this multifaceted molecule. In short, the pathophysiological role of Sema4D in periodontitis requires further study. Accordingly, attachment of the ligature to the maxillary molar of mice for 7 days induced alveolar bone-resorption accompanied by locally elevated, soluble Sema4D (sSema4D), TNF-α and RANKL. Removal of the ligature induced spontaneous bone regeneration during the following 14 days, which was significantly promoted by anti-Sema4D-mAb administration. Anti-Sema4D-mAb was also suppressed in vitro osteoclastogenesis and pit formation by RANKL-stimulated BMMCs. While anti-Sema4D-mAb downmodulated the bone-resorption induced in mouse periodontitis, it neither affected local production of TNF-α and RANKL nor systemic skeletal bone remodeling. RANKL-induced osteoclastogenesis and resorptive activity were also suppressed by blocking of CD72, but not Plexin B2, suggesting that sSema4D released by osteoclasts promotes osteoclastogenesis via ligation to CD72 receptor. Overall, our data indicated that ssSema4D released by osteoclasts may play a dual function by decreasing bone formation, while upregulating bone-resorption.


Subject(s)
Alveolar Bone Loss , Periodontitis , Alveolar Bone Loss/etiology , Animals , Antigens, CD , Bone Regeneration , Disease Models, Animal , Mice , Periodontitis/pathology , Semaphorins , Tumor Necrosis Factor-alpha
4.
FASEB J ; 32(7): 4016-4030, 2018 07.
Article in English | MEDLINE | ID: mdl-29533736

ABSTRACT

Cell fusion-mediated formation of multinuclear osteoclasts (OCs) plays a key role in bone resorption. It is reported that 2 unique OC-specific fusogens [ i.e., OC-stimulatory transmembrane protein (OC-STAMP) and dendritic cell-specific transmembrane protein (DC-STAMP)], and permissive fusogen CD9, are involved in OC fusion. In contrast to DC-STAMP-knockout (KO) mice, which show the osteopetrotic phenotype, OC-STAMP-KO mice show no difference in systemic bone mineral density. Nonetheless, according to the ligature-induced periodontitis model, significantly lower level of bone resorption was found in OC-STAMP-KO mice compared to WT mice. Anti-OC-STAMP-neutralizing mAb down-modulated in vitro: 1) the emergence of large multinuclear tartrate-resistant acid phosphatase-positive cells, 2) pit formation, and 3) mRNA and protein expression of CD9, but not DC-STAMP, in receptor activator of NF-κB ligand (RANKL)-stimulated OC precursor cells (OCps). While anti-DC-STAMP-mAb also down-regulated RANKL-induced osteoclastogenesis in vitro, it had no effect on CD9 expression. In our mouse model, systemic administration of anti-OC-STAMP-mAb suppressed the expression of CD9 mRNA, but not DC-STAMP mRNA, in periodontal tissue, along with diminished alveolar bone loss and reduced emergence of CD9+ OCps and tartrate-resistant acid phosphatase-positive multinuclear OCs. The present study demonstrated that OC-STAMP partners CD9 to promote periodontal bone destruction by up-regulation of fusion during osteoclastogenesis, suggesting that anti-OC-STAMP-mAb may lead to the development of a novel therapeutic regimen for periodontitis.-Ishii, T., Ruiz-Torruella, M., Ikeda, A., Shindo, S., Movila, A., Mawardi, H., Albassam, A., Kayal, R. A., Al-Dharrab, A. A., Egashira, K., Wisitrasameewong, W., Yamamoto, K., Mira, A. I., Sueishi, K., Han, X., Taubman, M. A., Miyamoto, T., Kawai, T. OC-STAMP promotes osteoclast fusion for pathogenic bone resorption in periodontitis via up-regulation of permissive fusogen CD9.


Subject(s)
Alveolar Bone Loss/metabolism , Membrane Proteins/genetics , Osteoclasts/metabolism , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Cells, Cultured , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Tetraspanin 29/genetics , Tetraspanin 29/metabolism , Up-Regulation
5.
Immunology ; 150(1): 74-86, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27599164

ABSTRACT

Osteopontin (OPN) is a pro-inflammatory protein that paradoxically protects against inflammation and bone destruction in a mouse model of endodontic infection. Here we have tested the hypothesis that this effect of OPN is mediated by effects on migration of innate immune cells to the site of infection. Using the air pouch as a model of endodontic infection in mice, we showed that neutrophil accumulation at the site of infection with a mixture of endodontic pathogens is significantly reduced in OPN-deficient mice. Reduced neutrophil accumulation in the absence of OPN was accompanied by an increase in bacterial load. OPN-deficiency did not affect neutrophil survival, CXCR2 ligand expression, or the production of inflammatory cytokines in the air pouch. In vitro, OPN enhanced neutrophil migration to CXCL1, whereas in vivo, inhibition of CXCR2 suppressed cellular infiltration in air pouches of infected wild-type mice by > 50%, but had no effect in OPN-deficient mice. OPN increased cell surface expression of CXCR2 on bone marrow neutrophils in an integrin-αv -dependent manner, and suppressed the internalization of CXCR2 in the absence of ligand. Together, these results support a model where the protective effect of OPN results from enhanced initial neutrophil accumulation at sites of infection resulting in optimal bacterial killing. We describe a novel mechanism for this effect of OPN: integrin-αv -dependent suppression of CXCR2 internalization in neutrophils, which increases the ability of these cells to migrate to sites of infection in response to CXCR2 ligands.


Subject(s)
Bacterial Infections/immunology , Integrin alpha5/metabolism , Neutrophils/immunology , Osteopontin/metabolism , Pulpitis/immunology , Animals , Bacterial Load , Cell Movement , Chemokine CXCL1/metabolism , Disease Models, Animal , Humans , Immunity, Innate/genetics , Integrin alpha5/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/genetics , Receptors, Interleukin-8B/metabolism
6.
Infect Immun ; 84(10): 2802-12, 2016 10.
Article in English | MEDLINE | ID: mdl-27456834

ABSTRACT

In response to a defined panel of stimuli, immature macrophages can be classified into two major phenotypes: proinflammatory (M1) and anti-inflammatory (M2). Although both phenotypes have been implicated in several chronic inflammatory diseases, their direct role in bone resorption remains unclear. The present study investigated the possible effects of M1 and M2 macrophages on RANKL-induced osteoclastogenesis. In osteoclastogenesis assays using RAW264.7 cells or bone marrow cells as osteoclast precursors, addition of M1 macrophages significantly suppressed RANKL-induced osteoclastogenesis compared to nonstimulated conditions (M0), addition of M2 macrophages, or no macrophage addition (P < 0.05), suggesting that M1 macrophages can downregulate osteoclastogenesis. This effect was maintained when direct contact between M1 and osteoclast precursors was interrupted by cell culture insertion, indicating engagement of soluble factors released from M1. M1 macrophages developed from interferon gamma (IFN-γ) knockout (IFN-γ-KO) mice lost the ability to downregulate osteoclastogenesis. Antibody-based neutralization of interleukin-12 (IL-12), but not IL-10, produced by M1 macrophages also abrogated M1-mediated downregulation of osteoclastogenesis. Real-time PCR analyses showed that IFN-γ suppressed gene expression of NFATc1, a master regulator of osteoclastogenesis, whereas IL-12 increased the apoptosis of osteoclasts, suggesting molecular mechanisms underlying the possible roles of IFN-γ or IL-12 in M1-mediated inhibition of osteoclastogenesis. These findings were confirmed in an in vivo ligature-induced mouse periodontitis model in which adoptive transfer of M1 macrophages showed a significantly lower level of bone loss and less tartrate-resistant acid phosphatase (TRAP)-positive cell induction than M0 or M2 macrophage transfer. In conclusion, by its secretion of IFN-γ and IL-12, M1, but not M0 or M2, was demonstrated to inhibit osteoclastogenesis.


Subject(s)
Macrophages/physiology , Osteoclasts/physiology , Osteogenesis/physiology , RANK Ligand/physiology , Analysis of Variance , Animals , Bone Marrow Cells , Cell Differentiation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/metabolism , Interleukin-12/metabolism , Mice , Mice, Knockout , Periodontitis/physiopathology , Polymerase Chain Reaction/methods , Tartrate-Resistant Acid Phosphatase/metabolism
7.
J Bone Miner Res ; 31(9): 1688-700, 2016 09.
Article in English | MEDLINE | ID: mdl-27082509

ABSTRACT

By binding to its chemokine receptor CXCR4 on osteoclast precursor cells (OCPs), it is well known that stromal cell-derived factor-1 (SDF-1) promotes the chemotactic recruitment of circulating OCPs to the homeostatic bone remodeling site. However, the engagement of circulating OCPs in pathogenic bone resorption remains to be elucidated. The present study investigated a possible chemoattractant role of macrophage migration inhibitory factor (MIF), another ligand for C-X-C chemokine receptor type 4 (CXCR4), in the recruitment of circulating OCPs to the bone lytic lesion. To accomplish this, we used Csf1r-eGFP-knock-in (KI) mice to establish an animal model of polymethylmethacrylate (PMMA) particle-induced calvarial osteolysis. In the circulating Csf1r-eGFP+ cells of healthy Csf1r-eGFP-KI mice, Csf1r+/CD11b+ cells showed a greater degree of RANKL-induced osteoclastogenesis compared to a subset of Csf1r+/RANK+ cells in vitro. Therefore, Csf1r-eGFP+/CD11b+ cells were targeted as functionally relevant OCPs in the present study. Although expression of the two cognate receptors for MIF, CXCR2 and CXCR4, was elevated on Csf1r+/CD11b+ cells, transmigration of OCPs toward recombinant MIF in vitro was facilitated by ligation with CXCR4, but not CXCR2. Meanwhile, the level of PMMA-induced bone resorption in calvaria was markedly greater in wild-type (WT) mice compared to that detected in MIF-knockout (KO) mice. Interestingly, in contrast to the elevated MIF, diminished SDF-1 was detected in a particle-induced bone lytic lesion of WT mice in conjunction with an increased number of infiltrating CXCR4+ OCPs. However, such diminished SDF-1 was not found in the PMMA-injected calvaria of MIF-KO mice. Furthermore, stimulation of osteoblasts with MIF in vitro suppressed their production of SDF-1, suggesting that MIF can downmodulate SDF-1 production in bone tissue. Systemically administered anti-MIF neutralizing monoclonal antibody (mAb) inhibited the homing of CXCR4+ OCPs, as well as bone resorption, in the PMMA-injected calvaria, while increasing locally produced SDF-1. Collectively, these data suggest that locally produced MIF in the inflammatory bone lytic site is engaged in the chemoattraction of circulating CXCR4+ OCPs. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Osteoclasts/metabolism , Osteolysis/metabolism , Osteolysis/pathology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bone and Bones/pathology , CD11b Antigen/metabolism , Cell Line , Cell Separation , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Gene Knock-In Techniques , Green Fluorescent Proteins/metabolism , Immunophenotyping , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/diagnostic imaging , Phenotype , Polymethyl Methacrylate , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, CXCR4/metabolism , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...