Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
Arch Oral Biol ; 142: 105513, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35939865

RESUMEN

OBJECTIVES: The aims of this study were to examine the effects of probiotic strains on the expression of cytokines and human ß-defensins-2-4 in human gingival epithelial cells and to investigate the in vivo efficacy of probiotics in a dog model. DESIGN: Selected probiotics, Lacticaseibacillus paracasei SD1, L. rhamnosus SD4, L. rhamnosus SD11 and Limosilactobacillus fermentum SD7, were examined for the expression of human ß-defensins-2-4, and cytokine responses after stimulated by various periodontopathogens. Subsequently, an in vivo study was set as a randomized, double-blind, placebo-controlled intervention in a dog model. A total of 20 dogs with mild gingivitis were randomly assigned to either the probiotic or control group. The effects of probiotics on periodontopathogenic- and cytokine levels were analyzed after 4 weeks of intervention. RESULTS: It showed that all probiotics could induce human ß-defensins-2-4, interleukin-1ß, interleukin-6, interleukin-8, and tumor necrosis factor-α expressions in human gingival epithelial cells; however, a significant difference had been found among the strains. When individual probiotic strain was combined to periodontopathogens, a significant reduction of IL-8 expression was found. Results of the in vivo study demonstrated that the bacterial levels, especially Porphyromonas gingivalis, and the interleukin-8 levels were significantly decreased after receiving the probiotic products compared to the controls. CONCLUSION: The results of the in vitro and in vivo studies in dogs are encouraging to support the effectiveness of a mixture of probiotic treatments for improvement of periodontal health by reducing periodontopathogens and interleukin-8 levels. Therefore, such probiotics preparation could possibly have therapeutic potential in human periodontitis.


Asunto(s)
Probióticos , beta-Defensinas , Animales , Citocinas , Perros , Células Epiteliales , Humanos , Interleucina-8 , Probióticos/farmacología , Probióticos/uso terapéutico , beta-Defensinas/farmacología
2.
Front Cell Infect Microbiol ; 12: 879423, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573793

RESUMEN

In subgingival plaque biofilms, Fusobacterium nucleatum is closely related to the occurrence and development of periodontitis. Streptococcus gordonii, as an accessory pathogen, can coaggregate with periodontal pathogens, facilitating the subgingival colonization of periodontal pathogens. Studies have shown that F. nucleatum can coaggregate with S. gordonii and colonize the subgingival plaque. However, most studies have focused on monocultures or coinfection of species and the potential impact of coaggregation between the two species on periodontal interactions to human gingival epithelial cells (hGECs) remains poorly understood. The present study explored the effect of coaggregation between F. nucleatum and S. gordonii on subgingival synergistic virulence to hGECs. The results showed that coaggregation inhibited the adhesion and invasion of F. nucleatum to hGECs compared with that in the F. nucleatum monoculture and coinfection group. Coaggregation and coinfection with F. nucleatum both enhanced S. gordonii adhesion to hGECs, but neither of the two groups affected S. gordonii invasion to hGECs compared with S. gordonii monoculture. The gene expression levels of TLR2 and TLR4 in hGECs in the coaggregation group were higher than those in the monoculture groups but lower than those in the coinfection group. Compared with coinfection, the coaggregation inhibited apoptosis of hGECs and promoted the secretion of the proinflammatory cytokines TNF-α and IL-6 by hGECs, showed a synergistic inflammatory effect, while coaggregation inhibited the secretion of the anti-inflammatory cytokine TGF-ß1. Coaggregation enhanced the phosphorylation of p65, p38, and JNK proteins and therefore activated the NF-κB and MAPK signaling pathways. Pretreatment with a pathway antagonist/inhibitor decreased the phosphorylation levels of proteins and the secretion of TNF-α and IL-6. In conclusion, coaggregation inhibited the adhesion and invasion of F. nucleatum to hGECs. However, it enhanced the adhesion of S. gordonii to hGECs. Compared with coinfection, coaggregation inhibited the apoptosis of hGECs. The coaggregation coordinately promoted the secretion of TNF-α and IL-6 by hGECs through the TLR/NF-κB and TLR/MAPK signaling pathways while inhibiting the secretion of TGF-ß1, thus aggravating the inflammatory response of hGECs.


Asunto(s)
Coinfección , Fusobacterium nucleatum , Adhesión Bacteriana , Células Epiteliales/microbiología , Humanos , Interleucina-6 , FN-kappa B , Streptococcus gordonii/genética , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa/farmacología , Virulencia
3.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628542

RESUMEN

Porphyromonas gingivalis as the keystone periodontopathogen plays a critical role in the pathogenesis of periodontitis, and crucially accounts for inflammatory comorbidities such as cardiovascular disease and Alzheimer's disease. We recently identified the existence of P. gingivalis persisters and revealed the unforeseen perturbation of innate response in human gingival epithelial cells (HGECs) due to these noxious persisters. Herein, RNA sequencing revealed how P. gingivalis persisters affected the expression profile of cytokine genes and related signaling pathways in HGECs. Results showed that metronidazole-treated P. gingivalis persisters (M-PgPs) impaired the innate host defense of HGECs, in a similar fashion to P. gingivalis. Notably, over one thousand differentially expressed genes were identified in HGECs treated with M-PgPs or P. gingivalis with reference to the controls. Gene Ontology and KEGG pathway analysis demonstrated significantly enriched signaling pathways, such as FOXO. Importantly, the FOXO1 inhibitor rescued the M-PgP-induced disruption of cytokine expression. This study suggests that P. gingivalis persisters may perturb innate host defense, through the upregulation of the FOXO signaling pathway. Thus, the current findings could contribute to developing new approaches to tackling P. gingivalis persisters for the effective control of periodontitis and P. gingivalis-related inflammatory comorbidities.


Asunto(s)
Periodontitis , Porphyromonas gingivalis , Citocinas/metabolismo , Células Epiteliales/metabolismo , Humanos , Periodontitis/tratamiento farmacológico , Periodontitis/genética , Periodontitis/metabolismo , Porphyromonas gingivalis/fisiología , Análisis de Secuencia de ARN , Transducción de Señal , Regulación hacia Arriba
4.
BMC Oral Health ; 22(1): 144, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35473620

RESUMEN

BACKGROUND: Oxidative stress mediated by hyperglycemia damages cell-reparative processes such as mitophagy. Down-regulation of mitophagy is considered to be a susceptible factor for diabetes mellitus (DM) and its complications. However, the role of mitophagy in DM-associated periodontitis has not been fully elucidated. Apoptosis of human gingival epithelial cells (hGECs) is one of the representative events of DM-associated periodontitis. Thus, this study aimed to investigate PTEN-induced putative kinase 1 (PINK1)-mediated mitophagy activated in the process of high glucose (HG)-induced hGECs apoptosis. METHODS: For dose-response studies, hGECs were incubated in different concentrations of glucose (5.5, 15, 25, and 50 mmol/L) for 48 h. Then, hGECs were challenged with 25 mmol/L glucose for 12 h and 48 h, respectively. Apoptosis was detected by TdT-mediated dUTP nick end labeling (TUNEL), caspase 9 and mitochondrial membrane potential (MMP). Subsequently, autophagy was evaluated by estimating P62, LC3 II mRNA levels, LC3 fluorescent puncta and LC3-II/I ratio. Meanwhile, the involvement of PINK1-mediated mitophagy was assessed by qRT-PCR, western blotting and immunofluorescence. Finally, hGECs were transfected with shPINK1 and analyzed by MMP, caspase 9 and annexin V-FITC apoptosis. RESULTS: The number of TUNEL-positive cells and caspase 9 protein were significantly increased in cells challenged with HG (25 mmol/L) for 48 h (HG 48 h). MMP was impaired both at HG 12 h and HG 48 h, but the degree of depolarization was more serious at HG 48 h. The autophagy improved as the amount of LC3 II increased and p62 decreased in HG 12 h. During this process, HG 12 h treatment induced PINK1-mediated mitophagy. PINK1 silencing with HG 12 h resulted in MMP depolarization and cell apoptosis. CONCLUSIONS: These results suggested that loss of the PINK1 gene may cause mitochondrial dysfunction and increase sensitivity to HG-induced apoptosis of hGECs at the early stage. PINK1 mediated mitophagy attenuates early apoptosis of gingival epithelial cells induced by high glucose.


Asunto(s)
Glucosa , Mitofagia , Proteínas Quinasas , Humanos , Apoptosis/efectos de los fármacos , Caspasa 9/metabolismo , Células Epiteliales , Glucosa/farmacología , Mitofagia/fisiología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
5.
Odontology ; 110(3): 557-568, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35179670

RESUMEN

Junction epithelium (JE) is located apical to the bottom of the gingival sulcus and binds enamel to hemidesmosomes to protect the periodontal tissue from bacterial infection. Function of odontogenic ameloblast-associated protein (ODAM) is suggested by its expression sites (JE and maturation stage ameloblasts) to be involved in the adhesion between the JE and enamel, and odontogenesis. To analyze the changes in ODAM gene and protein expressions in inflamed gingiva, Ca9-22 gingival epithelial cells were stimulated with 1 ng/ml interleukin-1ß (IL-1ß) for 3-24 h, and ODAM mRNA and protein levels were analyzed by real-time PCR and Western blotting. Luciferase (LUC) constructs were made ligating various lengths of human ODAM gene promoters and performed LUC analyses in Ca9-22 cells. Gel shift and chromatin immunoprecipitation (ChIP) assays were performed. IL-1ß induced ODAM mRNA and protein levels at 6-24 h. IL-1ß increased LUC activities of the ODAM gene promoter constructs from - 85 to - 950. These activities were blocked by protein kinase A, tyrosine kinase, mitogen-activated protein (MAP) kinase kinase and phosphoinositide 3-kinase inhibitors. Gel shift and ChIP assays showed that IL-1ß induced CCAAT/enhancer-binding protein (C/EBP) ß and Yin Yang1 (YY1) binding to C/EBP1, 2, 3, and YY1 elements. These data indicate that IL-1ß stimulates ODAM gene transcription mediated through C/EBP1, C/EBP2, C/EBP3, and YY1 elements in the human ODAM gene promoter.


Asunto(s)
Ameloblastos , Encía , Ameloblastos/metabolismo , Células Epiteliales/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Odontogénesis , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética
6.
Inflamm Res ; 71(1): 119-129, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34787682

RESUMEN

OBJECTIVE: Odontogenic ameloblast-associated protein (ODAM) is produced by maturation stage ameloblasts and junctional epithelium (JE). The function of ODAM is thought to be involved in the attachment of teeth and JE. To elucidate transcriptional regulation of human ODAM gene in inflamed gingiva, we have analyzed the effects of TNF-α on the expression of ODAM gene in Ca9-22 and Sa3 gingival epithelial cells. MATERIALS AND METHODS: Total RNAs were extracted from Ca9-22 and Sa3 cells after stimulation by TNF-α (10 ng/ml). ODAM mRNA and protein levels were analyzed by qPCR and Western blotting. Luciferase (LUC) analyses were performed using LUC constructs inserted in various lengths of ODAM gene promoter. Gel shift and chromatin immunoprecipitation (ChIP) assays were carried out. RESULTS: TNF-α increased ODAM mRNA and protein levels at 3 to 24 h. TNF-α induced LUC activities of the ODAM gene promoter constructs, and the activities were inhibited by protein kinase A, tyrosine kinase, MEK1/2, PI3-kinase and NF-κB inhibitors. Gel shift and ChIP assays revealed that TNF-α increased CCAAT/enhancer-binding protein (C/EBP) ß and Yin Yang1 (YY1) binding to three kinds of C/EBPs and YY1 elements. CONCLUSION: These results demonstrate that TNF-α stimulates ODAM gene transcription via C/EBPs and YY1 elements in the human ODAM gene promoter.


Asunto(s)
Ameloblastos , Factor de Necrosis Tumoral alfa , Ameloblastos/metabolismo , Inserción Epitelial/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas I-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
7.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 40(3): 285-292, 2022 May 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38597008

RESUMEN

OBJECTIVES: This work aimed to study the biological behavior of human gingival epithelial cells (HGECs) irradiated by non-thermal atmospheric plasma (NTAP) on a titanium surface. METHODS: Cultured HGECs (3⁃5 generations) with the best activity were digested and treated for varying times (0, 10, 20, 30, and 60 s) by NTAP and then seeded on the surface of a titanium disc. The HGECs were cultured in oral keratinocyte medium and 1% penicillin/streptomycin solution. The cells were kept in an atmosphere of 5% CO2 at 37 ℃ and incubated for different times (4, 12, 24, and 48 h; n=5). Cell counting kit-8 (CCK-8) was used to detect cell adhesion capacity. Scanning electron microscopy (SEM) was conducted to observe the morphology of cells on titanium plates. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to evaluate the gene expression of adhesion-related molecules, such as Laminin α3, Integrin ß4, and Plectin. RESULTS: The number of adhered cells increased at 0­20 s, whereas that gradually decreased at 20⁃60 s. Therefore, cell culture at the two time points showed that HGECs adhesion reached the maximum when NATP was irradiated for 20 s. Compared with the control group, more cells in the treatment group adhered to the titanium surface at each time point (P<0.05). Cells in the treatment group showed more irregular polygons, more protrusions and pseudopods, and a larger cell diffusion area on the titanium surface than those in the control group. qRT-PCR showed that the expression levels of Laminin α3, Integrin ß4, and Plectin adhesion-related genes on the titanium surface in the treatment group were higher than those in the control group at each culture time point (P<0.05). Western blot showed that the expression levels of Laminin α3, Integrin ß4, and Plectin adhesion-related proteins on the titanium surface were higher in the treatment group than in the control group at 4 and 12 h. CONCLUSIONS: After NTAP treatment, the results showed that 20 s of treatment time could maximize the number of adhered cells on the titanium surface; change the cell adhesion morphology; and significantly upregulate the expression of adhesion-related genes and proteins of Laminin α3, Integrin ß4, and Plectin. Furthermore, it could promote the biological sealing effect of HGECs on the titanium surface.

8.
Artif Cells Nanomed Biotechnol ; 48(1): 1167-1177, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32924619

RESUMEN

PURPOSE: To explore the feasibility of constructing tissue-engineered vascularised oral mucosa-like structures with rabbit ACVM-0.25% HLC-I scaffold and human gingival fibroblasts (HGFs), human gingival epithelial cells (HGECs) and vascular endothelial-like cells (VEC-like cells). METHOD: Haematoxylin and Eosin (H&E) staining, immunohistochemical, immunofluorescence, 5-ethynyl-2'-deoxyuridine (EdU) staining and scanning electron microscope (SEM) were performed to detect the growth status of cells on the scaffold complex. After the scaffold complex implanted into nude mice for 28 days, tissues were harvested to observe the cell viability and morphology by the same method as above. Additionally, biomechanical experiments were used to assess the stability of composite scaffold. RESULTS: Immunofluorescence and Immunohistochemistry showed positive expression of Vimentin, S100A4 and CK, and the induced VEC-like cells had the ability to form tubule-like structures. In vitro observation results showed that HGFs, HGECs and VEC-like had good compatibility with ACVM-0.25% HLC-I and could be layered and grow in the scaffold. After implanted, the mice had no immune rejection and no obvious scar repair on the body surface. The biomfechanical test results showed that the composite scaffold has strong stability. CONCLUSION: The tissue-engineered vascularised complexes constructed by HGFs, HGECs, VEC-like cells and ACVM-0.25% HLC-I has good biocompatibility and considerable strength.


Asunto(s)
Aciclovir/análogos & derivados , Materiales Biomiméticos/farmacología , Mucosa Bucal/metabolismo , Neovascularización Fisiológica , Albúmina Sérica/farmacología , Ingeniería de Tejidos , Aciclovir/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Encía/citología , Encía/metabolismo , Conejos , Andamios del Tejido/química
9.
Cells ; 9(8)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32824595

RESUMEN

The role of the adaptor molecule MyD88 is thought to be independent of Toll-like receptor 3 (TLR3) signaling. In this report, we demonstrate a previously unknown role of MyD88 in TLR3 signaling in inducing endogenous ligands of TLR2 to elicit innate immune responses. Of the various TLR ligands examined, the TLR3-specific ligand polyinosinic:polycytidylic acid (poly I:C), significantly induced TNF production and the upregulation of other TLR transcripts, in particular, TLR2. Accordingly, TLR3 stimulation also led to a significant upregulation of endogenous TLR2 ligands mainly, HMGB1 and Hsp60. By contrast, the silencing of TLR3 significantly downregulated MyD88 and TLR2 gene expression and pro-inflammatory IL1ß, TNF, and IL8 secretion. The silencing of MyD88 similarly led to the downregulation of TLR2, IL1ß, TNF and IL8, thus suggesting MyD88 to somehow act downstream of TLR3. Corroborating in vitro data, Myd88-/- knockout mice downregulated TNF, CXCL1; and phospho-p65 and phospho-IRF3 nuclear localization, upon poly I:C treatment in a mouse model of skin infection. Taken together, we identified a previously unknown role for MyD88 in the TLR3 signaling pathway, underlying the importance of TLRs and adapter protein interplay in modulating endogenous TLR ligands culminating in pro-inflammatory cytokine regulation.


Asunto(s)
Inmunidad Innata , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Encía/citología , Voluntarios Sanos , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 3/genética , Transfección
10.
Cells ; 9(5)2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32429151

RESUMEN

Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.


Asunto(s)
Biopelículas , Células Epiteliales/microbiología , Fibroblastos/microbiología , Prótesis e Implantes/microbiología , Streptococcus oralis/fisiología , Adhesión Celular , Forma de la Célula , Supervivencia Celular , Citocinas/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Encía/patología , Humanos , Mediadores de Inflamación/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética
11.
Mol Immunol ; 111: 27-31, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30954023

RESUMEN

The oral cavity is home for a plethora of bacteria and viruses. Epithelial barriers encounter these micro-organisms and recognize them via pathogen recognition receptors (PRRs) that instigate antibacterial and antiviral responses. We and others have shown that human gingival epithelial cells (HGECs) express PRRs to defend invading pathogens. Among these PRRs, TLR2, TLR3 and TLR4 are highly expressed in HGECs and appear to be important based on our previous findings. IFN-ß is one of the major type 1 interferons induced to defend viral attack. In this report, we sought to dissect TLR3 and TLR4 mediated secretion of IFN-ß in HGECs. We stimulated HGECs with ultrapure LPS (TLR4 ligand) and Poly I:C (TLR3 ligand) for 24 h and supernatant was used to determine IFN-ß secretion. We show that cells treated with Poly I:C induced IFN-ß secretion but not cells treated with LPS. In addition, silencing of TLR3 prior to Poly I:C stimulation significantly downregulated IFN-ß secretion. On the contrary, overexpression of MD2 and TLR4 in HGECs restored IFN-ß secretion. Upon further evaluation, we found that TLR3 stimulation but not TLR4 induced the phosphorylation of interferon regulatory factor 3 (IRF3), which is critical for IFN-ß secretion. We conclude that IFN-ß secretion is through TLR3 and not via TLR4 in HGECs.


Asunto(s)
Células Epiteliales/metabolismo , Encía/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Células Cultivadas , Humanos , Interferón beta/metabolismo , Poli I-C/metabolismo , Transducción de Señal/fisiología
12.
J Cell Biochem ; 119(2): 1591-1603, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28771806

RESUMEN

Accumulation of advanced glycation end-products (AGEs) in periodontal tissues of patients with diabetes mellitus aggravates periodontitis, but the mechanisms are unknown. Calprotectin, a heterocomplex of S100A8 and S100A9 proteins, is a constitutive cytoplasmic component of healthy gingival epithelial cells. This study aimed at investigating the effects of AGE and Porphyromonas gingivalis lipopolysaccharide (PgLPS) on calprotectin expression in the human gingival epithelial cell line OBA-9. AGE and PgLPS increased the expression of S100A8 and S100A9 mRNAs, and AGE+PgLPS co-stimulation amplified their expression in OBA-9 cells. A higher concentration of calprotectin in cell lysates was also induced by stimulation with AGE and/or PgLPS. S100A8 was mainly translocated from the nucleus to the cytoplasm by AGE stimulation, while cytoplasmic localization of S100A9 was not altered following stimulation with AGE and/or PgLPS. Calprotectin was found in the cytoplasm of BSA-treated cells, but cytoplasmic and nuclear localization was observed following stimulation with AGE and/or PgLPS. AGE-induced S100A8, and S100A9 mRNA expression was partially suppressed by RAGE-specific siRNA. In contrast, PgLPS-induced S100A8 and S100A9 mRNA expression was strongly suppressed by TLR2-specific siRNA. Furthermore, the inhibition of p38, JNK MAPK, and NF-κB attenuated AGE- and PgLPS-induced S100A8 and S100A9 mRNA expression. Taken together, these results demonstrate that AGE acts in synergy with PgLPS to stimulate RAGE and TLR2 expression and activate p38, JNK MAPK, and NF-κB signaling pathways, resulting in increased activation of calprotectin (S100A8/S100A9) in human gingival epithelial cells. Our results suggest that calprotectin may be involved in the pathogenesis of diabetic periodontitis.


Asunto(s)
Calgranulina A/genética , Calgranulina B/genética , Encía/metabolismo , Productos Finales de Glicación Avanzada/efectos adversos , Lipopolisacáridos/efectos adversos , Porphyromonas gingivalis/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Encía/citología , Encía/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas , Periodontitis/genética , Periodontitis/metabolismo , Regulación hacia Arriba
13.
J Oral Microbiol ; 9(1): 1334503, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28748038

RESUMEN

Periodontitis is an inflammatory disease induced by pathogenic bacteria such as Porphyromonas gingivalis. Little is known about epidermal growth factor (EGF) signals in human gingival epithelial cells (HGEC), which are major targets of P. gingivalis, and how the expression of proteins participating in EGF signaling-that is, EGF-receptor (EGFR), suppressor of cytokine signaling-3 (SOCS-3), interferon regulatory factor-1 (IRF-1), and signal transducers and activators of transcription (STAT-3)-are modified. This study aimed to assess the effects of P. gingivalis and its purified lipopolysaccharide (LPS-Pg) on EGF signaling. HGEC were infected for 2 h in a dose-dependent manner with P. gingivalis and with heat-killed P. gingivalis, and activated for 2 and 24 h by 1 µg/mL of purified LPS-Pg. Quantitative reverse transcription polymerase chain reaction and Western blotting were performed to measure mRNA and protein levels for SOCS-3, IRF-1 EGF, EGFR, and STAT-3. The tyrosine-phosphorylation status of STAT-3 was also examined. The results showed that infection of HGEC cells with P. gingivalis, but not with heat-killed P. gingivalis, led to significant reductions in expression levels of mRNAs and proteins for SOCS-3, IRF-1, and EGFR, while LPS-Pg over time significantly increased the expression of these mRNAs and proteins. Tyrosine-phosphorylation of STAT-3 was significantly increased during infection with P. gingivalis and activation by LPS-Pg but not modified during infection with heat-killed P. gingivalis. This study highlights that P. gingivalis and its purified LPS differentially modulated the expression of proteins (SOCS-3, IRF-1, EGFR, and STAT-3) interfering with EGF signaling.

14.
Arch Oral Biol ; 62: 64-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26655749

RESUMEN

OBJECTIVE: The gingival epithelium plays an important role in protecting against the invasion of periodontal pathogens, and the permeability of gingival epithelial cells has been implicated in the initiation of periodontitis. Azithromycin (AZM) has been used in the treatment of chronic inflammatory airway diseases because it regulates cell-cell contact in airway epithelial cells. Therefore, AZM may also regulate barrier function in gingival epithelial cells. In the present study, we examined the effects of AZM on the permeability of human gingival epithelial cells (HGEC) under inflammatory conditions in vitro. MATERIALS AND METHODS: HGEC were stimulated by tumor necrosis factor-α (TNF-α) in the presence of AZM or p38 MAP kinase and ERK inhibitors. Permeability was assessed based on transepithelial electrical resistance (TER). The expression of E-cadherin, phosphorylated p38 MAP kinase, and ERK was analyzed by Western blotting. RESULTS: TNF-α decreased TER in HGEC, and AZM and the p38 MAP kinase and ERK inhibitors recovered this decrease. AZM inhibited the phosphorylation of ERK and p38 MAP kinase in TNF-α-stimulated HGEC. Furthermore, AZM recovered the decrease in E-cadherin expression in HGEC stimulated with TNF-α. CONCLUSIONS: These results suggested that AZM regulated gingival epithelial permeability through p38 MAP kinase and ERK signaling, and may contribute to suppress the inflammation in gingival tissue.


Asunto(s)
Azitromicina/farmacología , Células Epiteliales/efectos de los fármacos , Encía/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Cadherinas/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Encía/citología , Encía/metabolismo , Gingivitis/metabolismo , Humanos , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Cytokine ; 75(1): 165-73, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25882870

RESUMEN

Periodontitis is the most prevalent infectious disease caused by periodontopathic bacteria and is also a chronic inflammatory disease. Gingival crevicular fluid (GCF) is an inflammatory exudate that seeps into the gingival crevices or periodontal pockets around teeth with inflamed gingiva, and contains various materials including leukocytes and cytokines. Since gingival epithelial cells, which form a barrier against bacterial challenges, are affected by GCF, cytokines or other materials contained within GCF are engaged in the maintenance and disruption of the epithelial barrier. Accordingly, its compositional pattern has been employed as a reliable objective index of local inflammation. Transforming growth factor ß1 (TGF-ß1) levels in GCF were previously shown to be markedly higher in patients with periodontitis than in healthy subject. However, it currently remains unclear how TGF-ß1 affects gingival epithelial cell growth or apoptosis; therefore, elucidating the mechanism responsible may lead to a deeper understanding of pathogenic periodontitis. In the present study, the human gingival epithelial cell line, OBA9 cells were stimulated with recombinant TGF-ß1. Apoptosis-related protein levels were determined by Western blotting. Caspase-3/7 activity was measured with a Caspase-Glo assay kit. Surviving and apoptotic cells were detected using an MTS assay and TUNEL staining, respectively. TGF-ßRI siRNA and smad2 siRNA were transfected into cells using the lipofectamine RNAiMAX reagent. TGF-ß1 elevated caspase-3 activity and the number of TUNEL-positive apoptotic cells in OBA9 cells. Furthermore, while the levels of the pro-apoptotic proteins Bax, Bak, Bim, and Bad were increased in OBA9 cells stimulated with TGF-ß1, the TGF-ß1 treatment also decreased the levels of anti-apoptotic proteins such as Bcl-2 and Bcl-xL in a time-dependent manner. Additionally, TGF-ß1 up-regulated the protein levels of cleaved caspase-9. These results indicated that TGF-ß1-induced apoptosis was involved in a mitochondria-related intrinsic pathway. TGF-ß1 phosphorylated smad2 in OBA9 cells and this phosphorylation was clearly reduced by SB431542 (a TGF-ß type I receptor inhibitor). Consistent with this result, SB431542 or smad2 siRNA-induced reductions in smad2 protein expression levels attenuated TGF-ß1-induced apoptosis. On the other hand, the ligation of TGF-ß1 on its receptor also stimulated the phosphorylation of Erk and Akt, which are smad2-independent pathways. However, the inhibition of Erk/Akt signaling pathways by U0126, a MEK-Erk inhibitor and LY294002, a PI3Kinase-Akt inhibitor, augmented TGF-ß1-induced apoptosis in OBA9 cells. Taken together, the results of present study demonstrated that TGF-ß1 activated both the smad2 and Erk/Akt cascades via its receptor on gingival epithelial cells, even though these two pathways have opposite roles in cell death and survival, and the culmination of these signaling events induced mitochondria-dependent apoptosis in gingival epithelial cells. Based on the results of the present study, we herein proposed for the first time, that TGF-ß1 is a novel target cytokine for monitoring the progression of periodontal disease.


Asunto(s)
Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Encía/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Acetilcisteína/metabolismo , Apoptosis , Benzamidas/química , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Dioxoles/química , Humanos , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , Periodontitis/metabolismo , Fosforilación , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
Artículo en Coreano | WPRIM (Pacífico Occidental) | ID: wpr-145425

RESUMEN

The gingival epithelium of the oral cavity is constantly exposed to exogenous stimuli such as bacterial toxins, allergens, and thermal changes. These exogenous stimuli are resisted by innate host defense in gingival epithelial cells. However, it is unclear exactly how the exogenous stimuli affect detrimentally on the human gingival epithelial cells. Here, we investigated whether the allergen, such as house dust mite (HDM) extract, is linked to Ca2+ signaling and proinflammatory cytokine expression in primary cultured human gingival epithelial cells. HDM extract induced an increase in intracellular Ca2+ concentration ([Ca2+]i) in a dose-dependent manner. Extracellular Ca2+ depletion did not affected on the HDM extract-induced increase in [Ca2+]i. The HDM extract-induced increase in [Ca2+]i was abolished by the treatment with U73122 and 2-APB, which are inhibitors of phospholipase C (PLC) and inositol 1,4,5-trisphosphate (IP3) receptor. Moreover, HDM extract induced the mRNA expression of pro-inflammatory cytokine, interleukin (IL)-8. These results suggest that HDM extract triggers PLC/IP3-dependent Ca2+ signaling and IL-8 mRNA expression in primary cultured human gingival epithelial cells.


Asunto(s)
Humanos , Alérgenos , Toxinas Bacterianas , Células Epiteliales , Epitelio , Inositol 1,4,5-Trifosfato , Interleucina-8 , Interleucinas , Boca , Pyroglyphidae , ARN Mensajero , Fosfolipasas de Tipo C
17.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-537630

RESUMEN

objective: To investigate the effect of human serum albumin(HSA)on the attachment of human gingival epithelial cells(HGEs)to commercial pure titanium(cpTi).Methods: HGEs were cultured, the cells of passage 2~5 were seeded on to cpTi samples which were preincubated in keratinocyte serum free medium with 50 mg/ml HSA or without HSA. Cell attachment was studied by immunofluorescence analysis.Results: HGEs were cultured and identified by positive expression of cytokeratin. 4,12 and 24 h after seeding attached cells on HSA preincubated cpTi were 218,730 and 849, those on the control 417,745 and 864, respectively.Conclusion: The early stage of cell attachment of HGEs on to cpTi may be inhibited by HSA.

18.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-670925

RESUMEN

Objective: To determine if the hemagglutinin A (HagA) of Porphyromonas gingivalis could be involved in the adhesion and invasion in human gingival epithelial cells (HGEC). Methods:P. gingivalis 381 hagA mutant was constructed by conjugation method. The whole length of hagA gene was cloned into pYA292 in Salmonella typhimurium x4072 (S. typhimurium-hagA). The strains were used to test their ability of adhesion and invasion into HGEC using a standard antibiotic protection assay. S. typhimurium x4072 strains containing empty vectors were used as negative control. HagA expression in S. typhimurium-hagA was confirmed by Western blot. Results:Although there were no significant differences between P. gingivalis 381 hagA mutant and wild type in adhesion and invasion into HGEC, the adhesion values of S. typhimurium-hagA to HGEC were increased by 3 times compared to their respective controls, while the invasion ability of S. typhimurium-hagA was 4 times greater than that of the negative controls. Conclusion: These results suggest that HagA may participate in P. gingivalis adhesion and invasion into HGEC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA