Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Transplant Proc ; 49(9): 2176-2182, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29149979

ABSTRACT

OBJECTIVE: Oral mucositis (OM) caused by infection facilitated by myelosuppression and immunosuppression can be controlled through oral care. We investigated changes in oral anaerobic bacterial flora during the onset of OM with hematopoietic stem cell transplantation (HSCT). METHODS: This study included 19 patients who underwent HSCT. All received professional oral care before initiating the preparative regimen. We assessed OM, oral health and obtained microbial samples from the oral cavity during 5 assessment points: before initiating the preparative regimen; the day before HSCT (day 1); and at 7, 14, and 30 days after HSCT. Microbial species were identified by using a mass spectrometer. RESULTS: The number of patients with serious OM increased initially after HSCT and decreased thereafter. Many Streptococcus species were identified before HSCT, but these gradually decreased and were replaced by coagulase-negative staphylococci. An increase in Candida species after HSCT and the identification of Enterococcus species were significantly associated with OM. Nutritional status recovery and prognosis were significantly worse in patients who developed OM. CONCLUSIONS: To the best of our knowledge, this study is the first which shows that anaerobic bacteria were identified in patients' oral flora before and after HSCT by using a mass spectrometer. These results indicate that Enterococcus species and Candida species may have been associated with OM. OM affected the patients' improvement in nutritional status and their prognosis. We concluded that it is important to provide more complete oral care instructions and interventions to prevent these bacterial infections.


Subject(s)
Bacteria, Anaerobic/growth & development , Hematopoietic Stem Cell Transplantation/adverse effects , Mouth Mucosa/microbiology , Postoperative Complications/microbiology , Stomatitis/microbiology , Adult , Aged , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Mass Spectrometry , Middle Aged , Oral Hygiene , Postoperative Complications/prevention & control , Preoperative Period , Stomatitis/prevention & control
2.
J Periodontal Res ; 51(4): 462-70, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26432443

ABSTRACT

BACKGROUND AND OBJECTIVE: Periodontal disease is dental plaque-induced inflammatory disease of the periodontal tissues that results in bone loss in the affected teeth. During bone resorption, receptor activator of nuclear factor kappa B ligand (RANKL) is an essential factor that regulates osteoclastogenesis. Recently, we found that gingival epithelial cells (GECs) in periodontal tissue produce RANKL, the expression of which is regulated by tumor necrosis factor-α and protein kinase A signaling. In this study, we asked whether RANKL-producing GECs induce bone marrow macrophages (BMMs) to form osteoclasts in a co-culture system. MATERIAL AND METHODS: Ca9-22 GECs and osteoclast precursor BMMs were co-cultured with or without the protein kinase A signaling activator forskolin or inhibitor H89 to examine whether the RANKL-producing GECs could be induced to form osteoclasts, as determined using a pit formation assay. RESULTS: Osteoclasts formed spontaneously in co-cultures of Ca9-22 cells and BMMs, even in the absence of RANKL. The cells were cultured on bone slices for 14 d, at which time resorption pits were observed. Forskolin treatment significantly increased osteoclast numbers in these co-cultures, but forskolin alone did not induce osteoclast formation by BMMs. CONCLUSION: GECs producing RANKL are able to support osteoclastogenesis in an in vitro co-culture system using GECs and BMMs, in a process promoted by forskolin.


Subject(s)
Bone Marrow Cells/metabolism , Epithelial Cells/metabolism , Gingiva/cytology , Macrophages/metabolism , Osteoclasts/physiology , Osteogenesis/physiology , RANK Ligand/biosynthesis , Cells, Cultured , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/metabolism , Gingiva/metabolism , Humans
3.
J Periodontal Res ; 49(4): 508-17, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24102429

ABSTRACT

OBJECTIVE AND BACKGROUND: Periodontitis is an inflammatory disorder of the supporting tissue of teeth, which is composed of gingival soft tissue, cementum covering the tooth root, alveolar bone and periodontal ligament. The receptor activator of nuclear factor kappa B ligand (RANKL) is known to be an essential factor for osteoclastogenesis. Recent clinical studies indicate that levels of RANKL in the gingival crevicular fluid are increased while levels of its decoy receptor, osteoprotegerin (OPG), are decreased in patients with periodontitis. Although the gingival sulcus is composed of gingival tissue, RANKL and OPG expression in gingival epithelial cells is not fully understood. The aim of this study is to investigate the expression of RANKL and OPG in gingival tissue and which factors regulate RANKL expression in gingival epithelial cells. MATERIAL AND METHODS: Reverse transcriptase polymerase chain reaction analysis, western blotting and immunohistochemistry were performed to confirm RANKL and OPG expression in gingival epithelial cells (GECs) and in gingival tissue. Immunostaining was also examined to confirm tumor necrosis factor (TNF)-α and TNF receptor type 1 (TNFR1) expression in gingival tissue. Ca9-22 cells, a human gingival epithelial cell line and human primary GECs were treated with TNF-α. Ca9-22 cells were treated by antibodies against TNF receptors, an inhibitor and an activator of protein kinase A (PKA) signaling and inhibitors of p38, Erk and NF-κB signaling to examine TNF-α-RANKL signaling pathways. RESULTS: RANKL mRNA and protein were expressed in GECs. Immunohistochemistry also showed RANKL expression in gingival tissue. On the other hand, the reverse transcriptase polymerase chain reaction and immunohistochemistry assay showed that GECs did not express OPG. In addition, TNF-α and TNFR1 proteins were expressed in junctional epithelium. TNF-α increased RANKL expression in GECs. TNF-α-induced RANKL expression was inhibited by an antibody against TNFR1 and an inhibitor of PKA signaling. Surprisingly, forskolin, a PKA activator, increased TNF-α-induced RANKL expression. CONCLUSION: RANKL, TNF and TNFR1 were coexpressed in junctional epithelium of gingival tissue. TNF-α induced RANKL expression via TNFR1 and PKA signaling in GECs of junctional epithelium.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/drug effects , Gingiva/drug effects , RANK Ligand/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Culture Techniques , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Epithelial Attachment/drug effects , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Gingiva/cytology , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred ICR , NF-kappa B/antagonists & inhibitors , Osteoprotegerin/drug effects , Receptors, Tumor Necrosis Factor, Type I/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
4.
J Dent Res ; 88(12): 1137-41, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19892918

ABSTRACT

Genetic variants at multiple loci have been shown to be associated with susceptibility to periodontitis. To better assess the genetic risk factors for periodontitis, we performed a case-control study in 319 Japanese individuals with periodontitis (172 aggressive and 147 chronic disease) and 303 race-matched healthy control individuals. Thirty-five functional gene polymorphisms that had been previously associated with immune responses were genotyped. For all gene polymorphisms tested, no significant differences were observed in the allele frequencies of persons with aggressive, chronic, and combined (aggressive and chronic) periodontitis, compared with control individuals. Multiple logistic regression analysis revealed a significant association of the vitamin D receptor +1056 T/C polymorphism with susceptibility to chronic periodontitis, after adjustment for age, gender, and smoking status (P = 0.002). These results suggest that none of the polymorphisms tested was strongly associated with periodontitis in a Japanese population. However, the vitamin D receptor +1056 polymorphism may be related to chronic periodontitis.


Subject(s)
Periodontitis/genetics , Adult , Age Factors , Aggressive Periodontitis/genetics , Alveolar Bone Loss/genetics , Case-Control Studies , Chronic Periodontitis/genetics , Cytosine , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Japan , Male , Middle Aged , Periodontal Attachment Loss/genetics , Periodontal Pocket/genetics , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Calcitriol/genetics , Risk Factors , Sex Factors , Smoking , Thymine
5.
Oral Microbiol Immunol ; 23(3): 259-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18402614

ABSTRACT

INTRODUCTION: Collagen phagocytosis by fibroblasts is involved in the intracellular pathway related to collagen breakdown in soft connective tissues. The possible role of lipopolysaccharide (LPS) in regulating this fibroblast function has not been elucidated so we investigated the effect of LPS from Actinobacillus actinomycetemcomitans, a periodontopathic bacterium, on collagen phagocytic activity in human gingival fibroblasts and associated regulatory mechanisms. METHODS: LPS pretreatment stimulated binding of collagen-coated beads to cells and, subsequently, their internalization. RESULTS: The LPS-activated collagen phagocytic process was enhanced in the presence of the soluble form of CD14 (sCD14) or LPS-binding protein (LBP), while the LPS/LBP treatment activated Akt and induced actin reorganization. Furthermore, these LPS/LBP-induced effects were partially suppressed by adding phosphatidyl-inositol-3 kinase (PI3K) inhibitors. CONCLUSION: These results suggest that A. actinomycetemcomitans LPS disturbs the homeostasis of collagen metabolism within gingival tissue by facilitating collagen phagocytosis by gingival fibroblasts, and serum sCD14 and LBP positively regulate the action of LPS. In addition, the PI3K/Akt signaling is thought to partially mediate the LPS/LBP-stimulated collagen phagocytic pathway, which may be dependent on actin cytoskeletal rearrangement.


Subject(s)
Aggregatibacter actinomycetemcomitans , Collagen/metabolism , Fibroblasts/drug effects , Gingiva/drug effects , Lipopolysaccharides/pharmacology , Phagocytosis/drug effects , Actins/drug effects , Acute-Phase Proteins/pharmacology , Adult , Androstadienes/pharmacology , Carrier Proteins/pharmacology , Cells, Cultured , Chromones/pharmacology , Cytochalasin D/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli , Fibroblasts/physiology , Gingiva/cytology , Humans , Lipopolysaccharide Receptors/pharmacology , Male , Membrane Glycoproteins/pharmacology , Morpholines/pharmacology , Phagocytosis/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/drug effects , Wortmannin
6.
J Periodontal Res ; 42(4): 367-76, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559635

ABSTRACT

BACKGROUND AND OBJECTIVE: Interleukin-1beta-stimulated receptor activator of nuclear factor-kappaB ligand (RANKL) expression in human periodontal ligament cells is partially mediated by endogenous prostaglandin E2, whereas mitogen-activated protein kinases (MAPKs) are implicated in regulating various interleukin-1-responsive genes. We investigated herein the involvement of MAPKs in interleukin-1beta-stimulated RANKL expression in human periodontal ligament cells. MATERIAL AND METHODS: Human periodontal ligament cells were pretreated separately with specific inhibitors of MAPKs, including extracellular signal-regulated kinase, p38 MAPK and c-Jun N-terminal kinase, and subsequently treated with interleukin-1beta. Following each treatment, the phosphorylation of each MAPK, the expression of RANKL, and the production of prostaglandin E2 were determined. RANKL activity was evaluated using an assay to determine the survival of prefusion osteoclasts. RESULTS: Interleukin-1beta induced RANKL expression at the mRNA and protein levels, as well as RANKL activity in human periodontal ligament cells. Interleukin-1beta also activated extracellular signal-regulated kinase, p38 MAPK, and c-Jun N-terminal kinase. Pretreatment with each MAPK inhibitor partially, but significantly, suppressed interleukin-1beta-induced RANKL expression and its activity, as well as prostaglandin E2 production. CONCLUSION: In human periodontal ligament cells, three types of MAPK inhibitor may abrogate RANKL expression and activity induced by interleukin-1beta, directly or indirectly through partial suppression of prostaglandin E2 synthesis. In addition, extracellular signal-regulated kinase, p38 MAPK, and c-Jun N-terminal kinase signals may co-operatively mediate interleukin-1beta-stimulated RANKL expression and its activity in those cells.


Subject(s)
Interleukin-1beta/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Periodontal Ligament/enzymology , RANK Ligand/metabolism , Adolescent , Adult , Animals , Cattle , Dinoprostone/analysis , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Osteoblasts/enzymology , Osteoclasts/enzymology , Periodontal Ligament/cytology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Inflamm Res ; 55(12): 534-42, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17221168

ABSTRACT

OBJECTIVE: We investigated the possible roles of interferon-gamma (IFN-gamma) in modulation of extracellular and intracellular routes of collagen digestion by human fibroblasts. METHODS: Human gingival fibroblasts were treated with IFN-gamma, after which matrix metalloproteinase-1 (MMP-1) activation was determined. Following the IFN-gamma treatment, cells were further incubated with either activating antibody for beta1 integrin or actin monomer-sequestering agent latrunculin B before incubation with collagen-coated fluorescent beads. Thereafter, the binding and internalization of the beads were assessed. RESULTS: IFN-gamma had no significant effect on MMP-1 activation, however, it reduced the binding of collagen-coated beads in the minimum affinity range and, subsequently, internalization of the beads. The inhibitory effects of IFN-gamma were partially reversed by adding either the beta1 integrin activating antibody or latrunculin B. CONCLUSIONS: Although IFN-gamma does not appreciably moderate the extracellular route of collagen digestion by human fibroblasts, the reduced level of collagen phagocytosis by IFN-gamma in the cells may contribute to fibrosis in inflamed connective tissues. Further, IFN-gamma may decrease the binding of collagen and following phagocytosis in cells by inducing a subcortical actin assembly and reducing the ability of beta1 integrin to bind to collagen.


Subject(s)
Actins , Integrin beta1 , Actins/metabolism , Cells, Cultured , Collagen/metabolism , Fibroblasts/drug effects , Humans , Interferon-gamma/metabolism , Phagocytosis/drug effects
8.
J Periodontal Res ; 38(1): 1-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558931

ABSTRACT

Previous studies have provided the biological basis for the therapeutic use of enamel matrix derivative (EMD) at sites of periodontal regeneration. A purpose of this study is to determine effects of EMD on cell growth, osteoblastic differentiation and insulin-like growth factor-I (IGF-I) and transforming growth factor-beta 1 (TGF-beta 1) production in human periodontal ligament cells (HPLC). We also examined participation of endogenous IGF-I and TGF-beta 1 with EMD-stimulated cell growth in these cells. HPLCs used in this study were treated with EMD alone or in combination with antihuman IGF-I antibody (anti-hIGF-I) or anti-hTGF-beta 1, recombinant human bone morphogenetic protein-2 (rhBMP-2), 1,25-dihydroxyvitamin D3[1,25(OH)2D3], rhTGF-beta 1 or rhIGF-I. After each treatment, cell growth, the production of IGF-I and TGF-beta 1 and the expression of osteoblastic phenotypes were evaluated. EMD stimulated cell growth in dose-dependent and time-dependent manners. EMD was also stimulated to express IGF-I and TGF-beta 1 at protein and mRNA levels. The EMD-stimulated cell growth was partially suppressed by cotreatment with anti-hIGF-I or anti-hTGF-beta 1, and cell growth was also stimulated by treatment with rhIGF-I or rhTGF-beta 1. rhBMP-2 stimulated alkaline phosphatase (ALPase) activity and ALPase mRNA expression, and 1,25(OH)2D3 stimulated ALPase and osteocalcin mRNA expression. However, EMD showed no effect on the osteoblastic phenotypes expression. These results demonstrated that EMD has no appreciable effect on osteoblastic differentiation, however it stimulates cell growth and IGF-I and TGF-beta 1 production in HPLC, and that these endogenous growth factors partially relate to the EMD-stimulated cell growth in HPLC.


Subject(s)
Dental Enamel Proteins/pharmacology , Insulin-Like Growth Factor I/physiology , Periodontal Ligament/drug effects , Transforming Growth Factor beta/physiology , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/genetics , Analysis of Variance , Antibodies , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Calcitriol/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Dose-Response Relationship, Drug , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Insulin-Like Growth Factor I/genetics , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/genetics , Periodontal Ligament/cytology , Phenotype , RNA, Messenger/genetics , Recombinant Proteins , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
9.
J Dent Res ; 78(4): 840-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10326728

ABSTRACT

Interleukin-1 (IL-1) may play a critical role in immune and inflammatory responses in inflamed gingiva, and it is synthesized by a wide variety of host cells. In this study, we examined the regulatory effects of various cytokines on bioactive membrane IL-1 and intracellular IL-1 alpha production in cultured human gingival fibroblasts (HGF). Recombinant human (rh) IL-1 beta stimulated membrane IL-1 activity, which was mainly attributed to IL-1 alpha. rhIL-1 beta and rh tumor necrosis factor (TNF)-alpha stimulated HGF to produce intracellular IL-1 alpha, whereas rh interleukin-6 (IL-6), rh interleukin-4 (IL-4), and rh interferon (IFN)-gamma did not do so. Intracellular IL-1 alpha production induced by rhIL-1 beta or rhTNF-alpha may be partially related to protein kinase C (PKC) activation, because rhIL-1 beta or rhTNF-alpha-induced intracellular IL-1 alpha production was stimulated by pre-treatment with 12-o-tetradecanoylphorbol 13-acetate (TPA), a PKC activator, but was suppressed by the pre-treatment with 1-(5-isoquinoline-sulfonyl) -2-methylpiperazine dihydrochloride (H-7), which is a PKC inhibitor. rhIL-4 inhibited rhIL-1 beta- or rhTNF-alpha-induced intracellular IL-1 alpha production, but rhIL-6 had no effect on this production. Pre-treatment with rh IFN-gamma remarkably enhanced intracellular IL-1 alpha production induced by subsequent treatment with rhIL-1 beta or rhTNF-alpha. Simultaneous treatment with rhIFN-gamma and rhIL-1 beta inhibited rhIL-1 beta-induced intracellular IL-1 alpha production, but co-treatment with rhIFN-gamma and rhTNF-alpha enhanced rhTNF-alpha-induced intracellular IL-1 alpha production. These results suggest that in inflamed gingiva, pro-inflammatory cytokines such as IL-1 beta and TNF-alpha may induce bioactive intracellular IL-1 alpha production in human gingival fibroblasts and that this production can be differentially modulated by T-cell-derived cytokines such as IFN-gamma or IL4.


Subject(s)
Cytokines/pharmacology , Gingiva/metabolism , Interleukin-1/biosynthesis , Adult , Analysis of Variance , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Gingiva/chemistry , Gingiva/cytology , Gingiva/drug effects , Humans , Interleukin-1/analysis , Recombinant Proteins/pharmacology , Time Factors
10.
J Periodontal Res ; 32(7): 559-69, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9401927

ABSTRACT

Interleukin-6 (IL-6) is thought to be a major mediator of the host's defense against infection, and it regulates immune responses in inflamed tissue. In this study, we investigated the regulation of IL-6 production in human gingival fibroblasts (HGF) and human periodontal ligament fibroblasts (HPLF). Pro-inflammatory cytokines including interleukin (IL)-1 alpha, IL-1 beta and tumor necrosis factor (TNF)-alpha stimulated IL-6 production in HGF and HPLF in a time- and dose-dependent manner. This IL-1 alpha, IL-1 beta, or TNF-alpha-induced IL-6 production was enhanced, but the cAMP accumulation they induced was inhibited by the addition of indomethacin. This result suggests that endogenous prostaglandin E2 (PGE2) partially inhibits IL-1 or TNF-alpha-induced IL-6 production and that the enhancement of IL-6 production by IL-1 or TNF-alpha may not be caused through endogenous PGE2-induced cAMP-dependent pathway. Dexamethasone (DEX), a glucocorticoid which is a inhibitor of nuclear factor kappa B (NF-kappa B activation, markedly inhibited IL-1 (alpha or beta) or TNF-alpha-induced IL-6 production; so this production may be partially mediated through NF-kappa B. IL-1 (alpha or beta) and TNF-alpha enhanced IL-6 production synergistically. IL-6 production in HGF or HPLF stimulated with IL-1 beta was augmented by the addition of interferon (IFN)-gamma, but was slightly suppressed by the addition of IL-4. Endogenous IL-6 enhanced IL-1 (alpha or beta)-induced IL-6 production in the presence of IL-6 soluble receptor (IL-6sR). Accordingly, in inflamed periodontal tissues, gingival fibroblasts and periodontal ligament fibroblasts stimulated with pro-inflammatory cytokines such as IL-1 or TNF-alpha, may produce IL-6, and this production can be differentially modulated by endogenous PGE2, IL-6sR, T cell-derived cytokines such as IFN-gamma or IL-4, and glucocorticoids.


Subject(s)
Cytokines/pharmacology , Dexamethasone/pharmacology , Fibroblasts/immunology , Gingiva/immunology , Glucocorticoids/pharmacology , Interleukin-6/biosynthesis , Periodontal Ligament/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/biosynthesis , Cytokines/administration & dosage , Dexamethasone/administration & dosage , Dinoprostone/biosynthesis , Dinoprostone/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gingiva/cytology , Glucocorticoids/administration & dosage , Humans , Indomethacin/pharmacology , Interferon-gamma/pharmacology , Interleukin-1/administration & dosage , Interleukin-1/pharmacology , Interleukin-4/pharmacology , Interleukin-6/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Periodontal Ligament/cytology , Receptors, Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology
11.
J Periodontal Res ; 31(5): 355-64, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8858540

ABSTRACT

Previous observations suggest that interleukin-1 (IL-1) may play an important role in the progression of periodontitis. In the present study, we investigated whether a cell-associated IL-1 alpha (CAIL-1 alpha) produced in human gingival fibroblasts (HGF) induces biological activities related to the progression of periodontitis. HGF were treated with recombinant human IL-1 beta (rhIL-1 beta) for 12 h. After that, the cell layers of HGF were washed 3 times with fresh medium and were then fixed with 1% paraformaldehyde. The fixed cell layers of HGF were used for assays for bone resorbing activity, prostaglandin E2 (PGE2) production and collagenase activity. Fixed cell layers of HGF treated with rhIL-1 beta enhanced not only calcium release from BALB/c mouse calvaria but also PGE2 production and collagenase activity in HGF and human periodontal ligament fibroblasts (HPLF) cultured on the fixed cell layers. These activities were neutralized by treatment with monoclonal mouse anti-human IL-1 alpha antibody, but monoclonal mouse anti-human IL-1 beta antibody showed no effects on these activities. The induction of these activities by fixed cell layers of HGF required direct contact between the fixed cell layers and the calvaria, HGF, or HPLF. These results suggest that CAIL-1 alpha produced in HGF treated with rhIL-1 beta induces bone resorbing activity, PGE2 production and collagenase activity in the target cells by direct contact; CAIL-1 alpha may play an important role in the progression of periodontitis.


Subject(s)
Bone Resorption/physiopathology , Gingiva/metabolism , Interleukin-1/physiology , Periodontitis/physiopathology , Animals , Antibodies, Monoclonal , Bone Resorption/immunology , Cell Communication , Cells, Cultured , Collagenases/metabolism , Dinoprostone/biosynthesis , Disease Progression , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/metabolism , Gingiva/cytology , Gingiva/drug effects , Gingiva/immunology , Humans , Interleukin-1/biosynthesis , Interleukin-1/pharmacology , Mice , Periodontal Ligament/immunology , Periodontal Ligament/metabolism , Periodontal Ligament/physiology , Periodontitis/immunology , Recombinant Proteins/pharmacology
12.
Periodontal Clin Investig ; 18(1): 26-38, 1996.
Article in English | MEDLINE | ID: mdl-9116461

ABSTRACT

The purpose of this study was to determine the effects of a fibrin tissue adhesive material (FAM) on periodontal tissue regeneration. In an in vitro study comparing osteogenic cells with gingival fibroblasts, it was shown that the degradation of FAM adjacent to the osteogenic cells was faster than that adjacent to the gingival fibroblasts. In two in vivo studies in dogs where surgical bony defects were created, it was shown through histometric measurements that in the sites where FAM was applied, more new bone was found than in the control sites. It was concluded that FAM may enhance periodontal tissue regeneration.


Subject(s)
Alveolar Bone Loss/surgery , Bone Regeneration/drug effects , Fibrin Tissue Adhesive/pharmacology , Guided Tissue Regeneration, Periodontal/methods , Membranes, Artificial , Animals , Cells, Cultured , Dogs , Gingiva/cytology , Male , Mandible , Osteoblasts , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...