Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
J Cell Biochem ; 125(7): e30576, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38726711

ABSTRACT

Gingival epithelial cells (GECs) are physical and immunological barriers against outward pathogens while coping with a plethora of non-pathogenic commensal bacteria. GECs express several members of Toll-like receptors (TLRs) and control subsequent innate immune responses. TLR4 senses lipopolysaccharide (LPS) while TLR7/8 recognizes single-strand RNA (ssRNA) playing important roles against viral infection. However, their distinct roles in GECs have not been fully demonstrated. Here, we analyzed biological responses of GECs to  LPS and CL075, a TLR7/8 agonist. GE1, a mouse gingival epithelial cell line, constitutively express TLR4 and TLR7, but not TLR8, like primary skin keratinocytes. Stimulation of GE1 cells with CL075 induced cytokine, chemokine, and antimicrobial peptide  expressions, the pattern of which is rather different from that with LPS: higher mRNA levels of interferon (IFN) ß, CXCL10, and ß-defensin (BD) 14 (mouse homolog of human BD3); lower levels of tumor necrosis factor (TNF), CCL5, CCL11, CCL20, CXCL2, and CX3CL1. As for the intracellular signal transduction of GE1 cells, CL075 rapidly induced significant AKT phosphorylation but failed to activate IKKα/ß-NFκB pathway, whereas LPS induced marked IKKα/ß-NFκB activation without significant AKT phosphorylation. In contrast, both CL075 and LPS induced rapid IKKα/ß-NFκB activation and AKT phosphorylation in a macrophage cell line. Furthermore, specific inhibition of AKT activity abrogated CL075-induced IFNß, CXCL10, and BD14 mRNA expression in GE1 cells. Thus, TLR4/7 ligands appear to induce rather different host-defense responses of GECs through distinct intracellular signaling mechanisms.


Subject(s)
Epithelial Cells , Gingiva , Lipopolysaccharides , Toll-Like Receptor 4 , Toll-Like Receptor 7 , Mice , Animals , Gingiva/cytology , Gingiva/metabolism , Epithelial Cells/metabolism , Epithelial Cells/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/metabolism , Lipopolysaccharides/pharmacology , Signal Transduction , Cell Line , Immunity, Innate , Membrane Glycoproteins/metabolism , Humans , Sulfonamides
2.
J Oral Biosci ; 66(1): 26-34, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37949170

ABSTRACT

OBJECTIVE: Periodontal disease is a risk factor for preterm delivery, and elevated female hormone levels during pregnancy promote hormone-dependent periodontopathogenic bacterial growth and gingivitis. Although the saliva of pregnant women contains female hormones at elevated levels, their effects on the gingiva are poorly understood. Therefore, in this study, we investigated the effects of estradiol and progesterone stimulation on gingival epithelial cells via ingenuity pathway analysis. METHODS: Human gingival epithelial progenitors were cultured in a CnT-Prime medium; 17ß-estradiol (E2) and progesterone (P4) were used as the reagents. Cells treated with dimethyl sulfoxide alone were used as the control group. Cells in the control and experimental groups were incubated for 12 h. RNA was extracted from the cultured cells, RNA-Seq was performed, and pathway analysis was conducted. RESULTS: Differentially expressed genes were detected for 699 (over 2-fold increase) and 348 (decrease) genes in group E2 and for 1448 (increase) and 924 (decrease) genes in group P4 compared with those in the control group (FDR <0.05, n = 4). The z-scores of the pathways suggest that E2 and P4 increased the activity of the wound healing signaling pathway. The activation of this pathway was higher in the E2 and P4 groups than that in the control group. CONCLUSIONS: The results of this study suggest that estradiol and progesterone may affect gingival homeostasis and wound healing.


Subject(s)
Estradiol , Progesterone , Infant, Newborn , Female , Pregnancy , Humans , Progesterone/pharmacology , Progesterone/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Gingiva/metabolism , Epithelial Cells/metabolism , Cells, Cultured
3.
J Periodontal Res ; 58(3): 553-563, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36974375

ABSTRACT

OBJECTIVE AND BACKGROUND: Heated tobacco products have recently become commercially available. These products, as well as combustible cigarettes, produce aerosols; the risk of various diseases associated with heated tobacco products may be the same or higher than that with combustible cigarettes. In this study, we examined the effect of Ploom TECH+ extract on gingival epithelial cells. METHODS: Tobacco leaves from Ploom TECH+ tobacco capsules and water were mixed and heated; the supernatant subsequently collected was the heated tobacco product (HTP; control: HTP not added). Normal human gingival epithelial progenitors were cultured alternately with or without HTP for a total of 1 month. Subsequently, RNA, DNA, and proteins were isolated from these samples and comprehensively analyzed using RNA sequencing (RNA-seq), reduced representation bisulfite sequencing (RRBS), and western blotting, respectively. RESULTS: RNA-seq revealed that 284 genes showed a twofold increase and 145 genes showed a twofold decrease in gene expression. A heat map showed genetic differences between the control and HTP groups. A principal component analysis plot showed a clear genetic distribution between the control and HTP. Gene Ontology (GO) analysis showed that genes related to seven GO terms, including cornification and keratinization, were induced by long-term HTP stimulation. By contrast, GO pathways with a significant decrease in component expression were not detected. RRBS revealed that CpG island methylation increased more than twofold in 158 genes and decreased to less than twofold in 171 genes. Methylation of these CpG islands was not correlated with changes in gene expression levels. HTP treatment increased S100A7 expression. CONCLUSION: Long-term HTP stimulation affected epithelial differentiation and keratinization of gingival epithelial cells. Thus, habitual use of Ploom TECH+ may be a risk factor for tobacco-related oral mucosal diseases.


Subject(s)
Tobacco Products , Humans , Risk Factors , Hot Temperature , Epithelial Cells
4.
Arch Oral Biol ; 141: 105498, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35810494

ABSTRACT

OBJECTIVES: This study aims to investigate the effects of cannabis smoke condensate (CSC) on the adhesion, growth, and signaling pathways of human gingival epithelial cells. DESIGN: The effects of CSC on cell shape and adhesion, and viability were evaluated after 30 min, 60 min, 2 h, and 24 h of exposure using microscopic observation, cell metabolic activity, and lactate dehydrogenase activity assays. The effects of CSC on cell apoptosis, necrosis, autophagy, and oxidative stress were determined through flow cytometry, while apoptotic and autophagic gene expression were identified via an RT2-PCR array. Phosphorylated signaling pathway proteins were measured using flow cytometry. RESULTS: CSC deregulated gingival epithelial cell shape and adhesion, decreased cell viability, and increased lactate dehydrogenase release. Its toxic effects included apoptosis, autophagy, and oxidative stress. Moreover, it modulated seven specific apoptotic and six autophagic genes. Furthermore, it decreased phosphorylation in signaling proteins, such as STAT5, ERK12, P38, and nuclear factor κB. CONCLUSIONS: CSC has notable adverse effects on gingival epithelial cells. This finding indicates that cannabis smoke could impair gingival epithelial cell innate immune function, leading to gingivitis and periodontitis. Oral health professionals may need to document observed modifications in the oral cavity of patients who smoke cannabis and consider these potential changes during clinical care.


Subject(s)
Cannabis , Apoptosis , Autophagy , Epithelial Cells , Humans , Lactate Dehydrogenases , Oxidative Stress , Nicotiana
5.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 40(1): 93-99, 2022 Jan 25.
Article in English, Chinese | MEDLINE | ID: mdl-38596999

ABSTRACT

OBJECTIVES: This study aims to investigate the effect of calbindin 1 on the proliferation and apoptosis of gingival epithelial cells affected by Porphyromonas gingivalis (P. gingivalis)invitro. METHODS: A model of P. gingivalis infecting CA9-22 was established in vitro. At 24 h after infection, the expression of calbindin 1 (CALB1) was detected by real-time fluorescent quantitative polymerase chain reaction, Western blot, and immunofluorescence analyses. The expression of CALB1 was further inhibited by RNA interference. Cell proliferation was detected by BrdU analysis, and cell apoptosis was detected by caspase 3 activity. The expression of MDM2 and p53 was detected by Western blot analysis. RESULTS: P. gingivalis infection upregulated the expression of CALB1 in CA9-22 cells with multiplicity-dependent manner. CALB1 promoted the proliferation of CA9-22 cells, increased the expression of MDM2, and inhibited the expression of p53. Inhibiting CALB1 expression did not affect the inhibitory effect of P. gingivalis infection on CA9-22 apoptosis. CONCLUSIONS: P. gingivalis infection can promote the proliferation of CA9-22 cells by increasing CALB1 expression. The related mechanism may be associated with MDM2-p53.

6.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576085

ABSTRACT

Bitter-taste receptors (T2Rs) have emerged as key players in host-pathogen interactions and important modulators of oral innate immunity. Previously, we reported that T2R14 is expressed in gingival epithelial cells (GECs) and interacts with competence stimulating peptides (CSPs) secreted by the cariogenic Streptococcus mutans. The underlying mechanisms of the innate immune responses and physiological effects of T2R14 on Gram-positive bacteria are not well characterized. In this study, we examined the role of T2R14 in internalization and growth inhibitory effects on Gram-positive bacteria, namely Staphylococcus aureus and S. mutans. We utilized CRISPR-Cas9 T2R14 knockdown (KD) GECs as the study model to address these key physiological mechanisms. Our data reveal that the internalization of S. aureus is significantly decreased, while the internalization of S. mutans remains unaffected upon knockdown of T2R14 in GECs. Surprisingly, GECs primed with S. mutans CSP-1 resulted in an inhibition of growth for S. aureus, but not for S. mutans. The GECs infected with S. aureus induced T2R14-dependent human ß-defensin-2 (hBD-2) secretion; however, S. mutans-infected GECs did not induce hBD-2 secretion, but induced T2R14 dependent IL-8 secretion. Interestingly, our results show that T2R14 KD affects the cytoskeletal reorganization in GECs, thereby inhibiting S. aureus internalization. Our study highlights the distinct mechanisms and a direct role of T2R14 in influencing physiological responses to Gram-positive bacteria in the oral cavity.


Subject(s)
Endocytosis , Epithelial Cells/metabolism , Gingiva/cytology , Gram-Positive Bacteria/metabolism , Microbial Viability , Receptors, G-Protein-Coupled/metabolism , Taste , Actins/metabolism , Cell Line , Epithelial Cells/ultrastructure , Humans , Interleukin-8/metabolism , Models, Biological , Nitrates/metabolism , Nitrites/metabolism , Staphylococcus aureus/metabolism , Streptococcus mutans/metabolism , beta-Defensins/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
7.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34132371

ABSTRACT

MicroRNAs (miRNAs), a family of small non­coding RNAs, serve a pivotal role in the regulation of the inflammation by modulating the expression of various genes. However, the molecular mechanism by which miRNAs regulate inflammation­associated molecules in oral epithelial cells remains to be elucidated. The present study examined the biological function of miR­429 by performing the gain­/loss­of­function studies of miR­429 in a gingival squamous cell carcinoma line Ca9­22 cells that either over­ or under­expressed miR­429 through transient transfection with miR­429 mimic or miR­429 inhibitor, respectively. The results demonstrated that the over­expression of miR­429 suppressed the mRNA level of several interleukins, including IL­8. In addition, the over­expression of miR­429 reduced IL­8 secretion under the basal and TNF­α stimulated conditions, whereas the secretion of IL­8 was enhanced when miR­429 was under­expressed. The over­expression of miR­429 inhibited the activation of the transcription factor NF­κB. Furthermore, we found that miR­429 suppressed both mRNA and protein levels of IKKß via its direct binding to the 3'­untranslated region of IKKß mRNA. In addition, the downregulation of IKKß by small interfering RNA reduced both NF­kB activity and IL­8 production in Ca9­22 cells. Taken together, the findings revealed the molecular mechanism of miR­429 to regulate the inflammatory mediator in gingival cells and suggested that it could be useful as a therapeutic target of oral inflammatory diseases.


Subject(s)
Epithelial Cells/metabolism , Gingiva/metabolism , I-kappa B Kinase/metabolism , Interleukin-8/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , Anti-Inflammatory Agents/metabolism , Cell Line, Tumor , Gene Expression Regulation , Humans , I-kappa B Kinase/genetics , Inflammation/metabolism , MicroRNAs/genetics
8.
FASEB J ; 35(3): e21375, 2021 03.
Article in English | MEDLINE | ID: mdl-33559200

ABSTRACT

Host-pathogen interactions play an important role in defining the outcome of a disease. Recent studies have shown that the bacterial quorum sensing molecules (QSM) can interact with host cell membrane proteins, mainly G protein-coupled receptors (GPCRs), and induce innate immune responses. However, few studies have examined QSM-GPCR interactions and their influence on oral innate immune responses. In this study, we examined the role of bitter taste receptor T2R14 in sensing competence stimulating peptides (CSPs) secreted by cariogenic bacterium Streptococcus mutans and in mediating innate immune responses in gingival epithelial cells (GECs). Transcriptomic and western blot analyses identify T2R14 to be highly expressed in GECs. Our data show that only CSP-1 from S. mutans induces robust intracellular calcium mobilization compared to CSP-2 and CSP-3. By using CRISPR-Cas9, we demonstrate that CSP-1 induced calcium signaling and secretion of cytokines CXCL-8/IL-8, TNF-α, and IL-6 is mediated through T2R14 in GECs. Interestingly, the NF-kB signaling activated by CSP-1 in GECs was independent of T2R14. CSP-1-primed GECs attracted differentiated HL-60 immune cells (dHL-60) and this effect was abolished in T2R14 knock down GECs and also in cells primed with T2R14 antagonist 6-Methoxyflavone (6-MF). Our findings identify S. mutans CSP-1 as a peptide ligand for the T2R family. Our study establishes a novel host-pathogen interaction between cariogenic S. mutans CSP-1 and T2R14 in GECs leading to an innate immune response. Collectively, these findings suggest T2Rs as potential therapeutic targets to modulate innate immune responses upon oral bacterial infections.


Subject(s)
Bacterial Proteins/physiology , Gingiva/immunology , Host-Pathogen Interactions , Quorum Sensing/physiology , Receptors, G-Protein-Coupled/physiology , Streptococcus mutans/physiology , Calcium/metabolism , Cell Line , Cell Movement , Cytokines/biosynthesis , Epithelial Cells/immunology , Gingiva/cytology , Humans , Immunity, Innate , NF-kappa B/physiology , Phospholipase C beta/physiology
9.
Methods Mol Biol ; 2210: 215-224, 2021.
Article in English | MEDLINE | ID: mdl-32815142

ABSTRACT

Porphyromonas gingivalis is a major pathogen responsible for severe and chronic manifestations of periodontal disease, which is one of the most common infectious disorders of humans. Although human gingival epithelium prevents intrusions by periodontal bacteria, P. gingivalis is able to invade gingival epithelial cells. To study the dynamics and the fate of intracellular P. gingivalis, confocal laser scanning microscopy (CLSM) is a method of choice. Information gained with CLSM contains not only the number of P. gingivalis associated with gingival epithelial cells but also the bacterial localization on/inside the host cells, morphological change of host cells, and physical interaction between the bacteria and host organelle. In this chapter, we describe the protocols for microscopy techniques to morphologically study gingival epithelial cells infected by P. gingivalis.


Subject(s)
Bacteroidaceae Infections/pathology , Epithelial Cells/pathology , Gingiva/pathology , Periodontal Diseases/pathology , Porphyromonas gingivalis/physiology , Bacteroidaceae Infections/microbiology , Cell Culture Techniques/methods , Cell Line , Epithelial Cells/microbiology , Gingiva/cytology , Gingiva/microbiology , Humans , Microscopy, Confocal/methods , Periodontal Diseases/microbiology , Porphyromonas gingivalis/isolation & purification , Staining and Labeling/methods
10.
Article in English | MEDLINE | ID: mdl-32205352

ABSTRACT

Periodontitis as a biofilm-associated inflammatory disease is highly prevalent worldwide. It severely affects oral health and yet closely links to systemic diseases like diabetes and cardiovascular disease. Porphyromonas gingivalis as a "keystone" periodontopathogen drives the shift of microbe-host symbiosis to dysbiosis and critically contributes to the pathogenesis of periodontitis. Persisters represent a tiny subset of biofilm-associated microbes highly tolerant to lethal treatment of antimicrobials, and, notably, metronidazole-tolerant P. gingivalis persisters have recently been identified by our group. This study further explored the interactive profiles of metronidazole-treated P. gingivalis persisters (M-PgPs) with human gingival epithelial cells (HGECs). P. gingivalis cells (ATCC 33277) at stationary phase were treated with a lethal dosage of metronidazole (100 µg/ml, 6 h) for generating M-PgPs. The interaction of M-PgPs with HGECs was assessed by microscopy, flow cytometry, cytokine profiling, and quantitative PCR (qPCR). We demonstrated that the overall morphology and ultracellular structure of M-PgPs remained unchanged. Importantly, M-PgPs maintained the capabilities to adhere to and invade HGECs. Moreover, M-PgPs significantly suppressed proinflammatory cytokine expression in HGECs at a level comparable to that seen with the untreated P. gingivalis cells, through the thermosensitive components. The present report reveals that P. gingivalis persisters induced by lethal treatment of antibiotics were able to maintain their capabilities to adhere to and invade human gingival epithelial cells and to perturb the innate host responses. Novel strategies and approaches need to be developed for tackling P. gingivalis and favorably modulating the dysregulated immunoinflammatory responses for oral/periodontal health and general well-being.


Subject(s)
Periodontitis , Porphyromonas gingivalis , Cells, Cultured , Epithelial Cells , Gingiva , Humans , Metronidazole/pharmacology
11.
Front Immunol ; 10: 933, 2019.
Article in English | MEDLINE | ID: mdl-31114581

ABSTRACT

BET bromodomain proteins are important epigenetic regulators of gene expression that bind acetylated histone tails and regulate the formation of acetylation-dependent chromatin complexes. BET inhibitors suppress inflammatory responses in multiple cell types and animal models, and protect against bone loss in experimental periodontitis in mice. Here, we analyzed the role of BET proteins in inflammatory activation of gingival fibroblasts (GFs) and gingival epithelial cells (GECs). We show that the BET inhibitors I-BET151 and JQ1 significantly reduced expression and/or production of distinct, but overlapping, profiles of cytokine-inducible mediators of inflammation and bone resorption in GFs from healthy donors (IL6, IL8, IL1B, CCL2, CCL5, COX2, and MMP3) and the GEC line TIGK (IL6, IL8, IL1B, CXCL10, MMP9) without affecting cell viability. Activation of mitogen-activated protein kinase and nuclear factor-κB pathways was unaffected by I-BET151, as was the histone acetylation status, and new protein synthesis was not required for the anti-inflammatory effects of BET inhibition. I-BET151 and JQ1 also suppressed expression of inflammatory cytokines, chemokines, and osteoclastogenic mediators in GFs and TIGKs infected with the key periodontal pathogen Porphyromonas gingivalis. Notably, P. gingivalis internalization and intracellular survival in GFs and TIGKs remained unaffected by BET inhibitors. Finally, inhibition of BET proteins significantly reduced P. gingivalis-induced inflammatory mediator expression in GECs and GFs from patients with periodontitis. Our results demonstrate that BET inhibitors may block the excessive inflammatory mediator production by resident cells of the gingival tissue and identify the BET family of epigenetic reader proteins as a potential therapeutic target in the treatment of periodontal disease.


Subject(s)
Azepines/pharmacology , Epithelial Cells , Fibroblasts , Gingiva , Heterocyclic Compounds, 4 or More Rings/pharmacology , Periodontitis/drug therapy , Porphyromonas gingivalis/immunology , Triazoles/pharmacology , Animals , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/immunology , Fibroblasts/immunology , Fibroblasts/microbiology , Fibroblasts/pathology , Gingiva/immunology , Gingiva/microbiology , Gingiva/pathology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Mice , Periodontitis/immunology , Periodontitis/pathology
12.
Adv Med Sci ; 64(1): 54-57, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30472626

ABSTRACT

PURPOSE: This study aimed to evaluate the possible antagonistic effects of Lactobacillus acidophilus on Porphyromonas gingivalis, and detect inhibition of Lactobacillus acidophilus on Porphyromonas gingivalis when they are co-cultured with human gingival epithelial cells. MATERIALS AND METHODS: Human gingival epithelial cells were co-cultured with Lactobacillus acidophilus and Porphyromonas gingivalis alone or together. The amount of Porphyromonas gingivalis adhering to or invading the epithelial cells were determined by bacterial counts. The cellular proliferation was assayed by the MTT method. Apoptosis was detected by flow cytometry with apoptosis detection kit. RESULTS: On one hand, Lactobacillus acidophilus reduced the inhibitory effect of Porphyromonas gingivalis on the human gingival epithelial cells proliferation in a dose dependent manner. On the other hand, Porphyromonas gingivalis induced significant apoptosis on human gingival epithelial cells, and Lactobacillus acidophilus inhibited this apoptosis-inducing effect of Porphyromonas gingivalis in a dose dependent manner. CONCLUSIONS: Porphyromonas gingivalis inhibits the proliferation and induces the apoptosis of human gingival epithelial cells. Lactobacillus acidophilus could attenuate this effect in a dose-dependent manner, and it thus reduces the destruction from pathogens. Lactobacillus acidophilus could be an effective candidate for probiotic therapy in periodontal diseases.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Epithelial Cells/microbiology , Gingiva/cytology , Lactobacillus acidophilus/physiology , Porphyromonas gingivalis/physiology , Cell Proliferation , Coculture Techniques , Cytoprotection , Humans
13.
Jpn Dent Sci Rev ; 54(2): 66-75, 2018 May.
Article in English | MEDLINE | ID: mdl-29755617

ABSTRACT

Periodontal disease is a bacterial biofilm-associated inflammatory disease that has been implicated in many systemic diseases. A new preventive method for periodontal disease needs to be developed in order to promote the health of the elderly in a super-aged society. The gingival epithelium plays an important role as a mechanical barrier against bacterial invasion and a part of the innate immune response to infectious inflammation in periodontal tissue. The disorganization of cell-cell interactions and subsequent inflammation contribute to the initiation of periodontal disease. These make us consider that regulation of host defensive functions, epithelial barrier and neutrophil activity, may become novel preventive methods for periodontal inflammation. Based on this concept, we have found that several agents regulate the barrier function of gingival epithelial cells and suppress the accumulation of neutrophils in the gingival epithelium. We herein introduce the actions of irsogladine maleate, azithromycin, amphotericin B, and Houttuynia cordata (dokudami in Japanese), which is commonly used in traditional medicine, on the epithelial barrier and neutrophil migration in gingival epithelial cells in vivo and in vitro, in order to provide support for the clinical application of these agents to the prevention of periodontal inflammation.

14.
Mol Cell Biochem ; 436(1-2): 39-48, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28593565

ABSTRACT

Gingival epithelial cells form a physiological barrier against bacterial invasion. Excessive bacterial invasion destroys the attachment between the tooth surface and the epithelium, resulting in periodontitis. Integrins play a significant role in cell attachment; therefore, we hypothesized that bacterial infection might decrease the expressions of these integrins in gingival epithelial cells, resulting in reduced cell adhesion. Immortalized human gingival epithelial cells were co-cultured with Aggregatibacter actinomycetemcomitans Y4 (Aa Y4), and the gene expression levels of IL-8, proliferating cell nuclear antigen (PCNA), and integrins (α2, α3, α5, ß4, and ß6) were measured using quantitative reverse transcription polymerase chain reaction. Expression of PCNA and integrins, except integrin α5, was significantly downregulated, while expression of IL-8 and integrin α5 was significantly upregulated in the cells co-cultured with Aa Y4. The number of adherent cells significantly decreased when co-cultured with Aa Y4, as determined using cell adhesion assays. In the cells co-cultured with Aa Y4 and an integrin α5 neutralizing antibody, there was no effect on the expression of IL-8 and PCNA, while the expressions of integrins α2, α3, ß4, and ß6, and the number of adherent cells did not decrease. The number of invading bacteria in the cells was reduced in the presence of the antibody and increased in the presence of TLR2/4 inhibitor. Therefore, integrin α5 might be involved in Aa Y4 invasion into gingival epithelial cells, and the resulting signal transduction cascade reduces cell adhesion by decreasing the expression of integrins, while the TLR2/4 signaling cascade regulates IL-8 expression.


Subject(s)
Aggregatibacter actinomycetemcomitans/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Gingiva/metabolism , Integrin alpha Chains/metabolism , Pasteurellaceae Infections/metabolism , Adult , Cell Adhesion , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gingiva/microbiology , Gingiva/pathology , Humans , Male , Pasteurellaceae Infections/pathology
15.
J Periodontal Res ; 52(1): 127-134, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27016382

ABSTRACT

BACKGROUND AND OBJECTIVE: Hypoxia has been widely studied in inflammatory diseases as it can modulate the inflammatory response, mainly via the hypoxia-inducible factor (HIF). However, little is known about the effects of hypoxia and the role of HIF in the inflammatory responses to periodontitis. In this study, we focused on the gingival epithelium that is exposed to relatively low levels of oxygen. We investigated whether hypoxic conditions have an impact on inflammatory responses in human gingival epithelial cells (HGECs). MATERIAL AND METHODS: Pimonidazole HCl, which accumulates in hypoxic cells, was administered intraperitoneally to C57BL/6 mice with or without Porphyromonas gingivalis infection. Immunohistochemistry was then performed to detect the hypoxic cells in periodontal tissue. Immortalized HGECs were cultured under hypoxic conditions with or without interleukin (IL)-1ß, and the expression levels of IL-6 and IL-8 were measured by real-time reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. HIF-1α expression was detected by western blotting. The DNA-binding activity of HIF-1α was determined by a DNA-binding enzyme-linked immunosorbent assay. The involvement of HIF-1α in the hypoxic response was examined by transfection with HIF-1α siRNA. RESULTS: Immunohistochemistry revealed pimonidazole HCl accumulation in the gingival epithelium of both normal and P. gingivalis-infected mice, with a slightly stronger signal in the P. gingivalis-infected mice than in the normal mice. The IL-1ß-induced IL-6 and IL-8 production by HGECs was suppressed under hypoxic conditions. HIF-1α accumulated during hypoxia, and this accumulation was further enhanced by IL-1ß treatment. The hypoxia-dependent suppression of IL-6 and IL-8 expression was reversed by treating the cells with HIF-1α siRNA. CONCLUSION: Our results suggest that the gingival epithelium is exposed to low oxygen tension in periodontal tissue and that this hypoxic condition modulates the local inflammatory response of gingival epithelial cells in an HIF-1α-dependent manner.


Subject(s)
Epithelium/metabolism , Gingiva/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Male , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
16.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 35(6): 654-658, 2017 Dec 01.
Article in Chinese | MEDLINE | ID: mdl-29333782

ABSTRACT

Autophagy is an intracellular conservative degradation pathway. This event has been considered as a key step in host defense against bacterial infection. However, Porphyromonas gingivalis, as one of the evidence-sufficient periodontal pathogens, can utilize self-induced autophagy to achieve persistent intracellular survival and proliferation, which enable this organism to escape from host immune surveillance. This review focuses on molecular mechanism of P. gingivalis internalization and autophagy to illuminate its pathogenesis and to further explore the relationship between P. gingivalis and systemic diseases.


Subject(s)
Autophagy , Gingiva , Porphyromonas gingivalis , Gingiva/microbiology , Research
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-357431

ABSTRACT

Autophagy is an intracellular conservative degradation pathway. This event has been considered as a key step in host defense against bacterial infection. However, Porphyromonas gingivalis, as one of the evidence-sufficient periodontal pathogens, can utilize self-induced autophagy to achieve persistent intracellular survival and proliferation, which enable this organism to escape from host immune surveillance. This review focuses on molecular mechanism of P. gingivalis internalization and autophagy to illuminate its pathogenesis and to further explore the relationship between P. gingivalis and systemic diseases.

18.
Oral Dis ; 22(6): 578-84, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27171647

ABSTRACT

OBJECTIVE: Diabetes is often associated with increased prevalence and severity of periodontal disease. We hypothesized that gingival epithelial cells modify periodontal disease progression and predicted that hyperglycemia would activate an inflammatory response in human gingival epithelial cells (HGECs). MATERIALS AND METHODS: We tested our hypothesis in immortalized HGECs (epi 4 cells) isolated from periodontal tissue and transfected with the simian virus 40 T antigen. The epi 4 cells were cultured in high (25 mM, HG) and normal (6 mM, NG) glucose conditions. RESULTS: The epi 4 cells showed increased interleukin-8 (IL-8) protein secretion and mRNA expression when cultured in HG, compared with in NG. These effects were not associated with increased cell proliferation and were not observed in a hyperosmolar control group (normal glucose with 19 mM mannitol). Increased IL-8 secretion in HG was inhibited by pretreatment with an antioxidant, N-acetylcysteine, or a protein kinase C inhibitor, Ro31-8220. Hyperglycemia did not affect IL-8 secretion by gingival fibroblasts or periodontal ligament cells. In epi 4 cells, hyperglycemia also induced expression of toll-like receptor 2 (TLR2) but not TLR4. CONCLUSION: These findings suggest a potential participation of epithelial cells in periodontal disease during diabetes by evoking an excessive host inflammatory response.


Subject(s)
Epithelial Cells/physiology , Gingiva/cytology , Interleukin-8/biosynthesis , Oxidative Stress/physiology , Cells, Cultured , Diabetes Mellitus/metabolism , Humans
19.
Microb Pathog ; 94: 104-11, 2016 May.
Article in English | MEDLINE | ID: mdl-26806000

ABSTRACT

Host cell invasion is important for periodontal pathogens in evading host defenses and spreading into deeper areas of the periodontal tissue. Treponema denticola has been implicated in a number of potentially pathogenic processes, including periodontal tissue penetration. Here we tested the ability of T. denticola strains to invade human gingival epithelial cells (HGEC). After 2 h infection, intracellular location of T. denticola cells was confirmed by confocal laser scanning microscopy (CLSM). Results from an antibiotic protection assay following [(3)H]uridine labeling indicated that invasion efficiency reached a maximum at 2 h after infection. Internalized T. denticola cells were still observed in HGEC at 24 h by CLSM. A dentilisin deficient mutant exhibited significantly decreased invasion (p < 0.05) compared with the wild-type strain. In inhibition assays, phenylmethylsulfonyl fluoride and metabolic inhibitors such as methyl-ß-cyclodextrin and staurosporine significantly reduced T. denticola invasion. Under CLSM, T. denticola colocalized with GM-1 ganglioside-containing membrane microdomains in a cholesterol-dependent manner. These results indicated that T. denticola has the ability to invade into and survive within HGECs. Dentilisin activity of T. denticola and lipid rafts on HGEC appear to play important roles in this process.


Subject(s)
Epithelial Cells/microbiology , Gingiva/microbiology , Gingiva/pathology , Spirochaetales Infections/microbiology , Treponema denticola/pathogenicity , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Epithelial Cells/pathology , Host-Parasite Interactions , Humans , Membrane Microdomains/metabolism , Microbial Sensitivity Tests , Peptide Hydrolases/deficiency , Peptide Hydrolases/metabolism , Periodontitis/microbiology , Phenylmethylsulfonyl Fluoride/pharmacology , Staurosporine/pharmacology , Treponema denticola/drug effects , Treponema denticola/enzymology , beta-Cyclodextrins/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL