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











Publication year range
1.
Front Pharmacol ; 15: 1395160, 2024.
Article in English | MEDLINE | ID: mdl-39135784

ABSTRACT

Introduction: Endometriosis (EMs) is characterized by ectopic growth of active endometrial tissue outside the uterus. The Luoshi Neiyi prescription (LSNYP) has been extensively used for treating EMs in China. However, data on the active chemical components of LSNYP are insufficient, and its pharmacological mechanism in EMs treatment remains unclear. This study aimed to explore the potential mechanism of LSNYP for EMs through network pharmacology based on the components absorbed into the blood. Methods: Ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry was used to analyze blood components, and a series of network pharmacology strategies were utilized to predict targets of these components and EMs. Protein-protein interaction (PPI) network analysis, component-target-disease network construction, gene ontology (GO) functional enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. Additionally, molecular docking, molecular dynamics simulations, and in vitro and in vivo experiments were conducted to validate the HIF1A/EZH2/ANTXR2 pathway associated with hypoxic pathology in EMs. Results: Thirty-four absorbed components suitable for network pharmacology analysis were identified, and core targets, such as interleukin 6, EGFR, HIF1A, and EZH2, were founded. Enrichment results indicated that treatment of EMs with LSNYP may involve the regulation of hypoxia and inflammatory-related signaling pathways and response to oxidative stress and transcription factor activity. Experimental results demonstrated that LSNYP could decrease the expression of HIF1A, ANTXR2, YAP1, CD44, and ß-catenin, and increased EZH2 expression in ectopic endometrial stromal cells and endometriotic tissues. Molecular docking and molecular dynamics simulations manifested that there was stable combinatorial activity between core components and key targets of the HIF1A/EZH2/ANTXR2 pathway. Conclusion: LSNYP may exert pharmacological effects on EMs via the HIF1A/EZH2/ANTXR2 pathway; hence, it is a natural herb-related therapy for EMs.

2.
J Ethnopharmacol ; 335: 118659, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39098622

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Endometriosis (EMS) is a common gynecological disease that causes dysmenorrhea, chronic pelvic pain and infertility. Luoshi Neiyi Prescription (LSNYP), a traditional Chinese medicine (TCM) formula, is used to relieve EMS in the clinic. AIMS: This study aimed to examine the active components of LSNYP and the possible mechanism involved in its treatment of EMS. MATERIALS AND METHODS: Ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS) was used to identify the chemical components of LSNYP. Human primary ectopic endometrial stromal cells (ecESCs) and eutopic endometrial stromal cells (euESCs) were isolated, and the expression levels of hypoxia inducible factor 1A (HIF1A), enhancer of zeste homolog 2 (EZH2) and steroidogenic factor 1 (SF-1) were detected by immunofluorescence and qPCR. Cobalt chloride (CoCl2) was utilized to construct an in vitro hypoxic environment, and lentiviruses were engineered to downregulate HIF1A and EZH2 and upregulate EZH2. Subsequently, the expression levels of HIF1A, EZH2, and SF-1 were measured using qPCR or western blotting. The binding of EZH2 to the SF-1 locus in ESCs was examined via ChIP. Furthermore, the effects of LSNYP on the HIF1A/EZH2/SF-1 pathway were evaluated both in vitro and in vivo. RESULTS: A total of 185 components were identified in LSNYP. The protein and gene expression levels of HIF1A and SF-1 were increased, whereas those of EZH2 were decreased in ecESCs. After treating euESCs with 50 µmol L-1 CoCl2 for 24 h, cell viability and estradiol (E2) production were enhanced. Hypoxia decreased EZH2 protein expression, while si-HIF1A increased it. SF-1 was increased when EZH2 was downregulated in normal and hypoxic environments, whereas the overexpression of EZH2 led to a decrease in SF-1 expression. ChIP revealed that hypoxia reduced EZH2 binding to the SF-1 locus in euESCs. In vitro, LSNYP-containing serum decreased E2 and prostaglandin E2 (PGE2) production, inhibited cell proliferation and invasion, and reduced the expression of HIF1A, SF-1, steroidogenic acute regulatory protein (StAR), and aromatase cytochrome P450 (P450arom). In vivo, LSNYP suppressed inflammation and adhesion and inhibited the HIF1A/EZH2/SF-1 pathway in endometriotic tissues. CONCLUSIONS: LSNYP may exert pharmacological effects on EMS by inhibiting E2 synthesis and inflammation through regulation of the HIF1A/EZH2/SF-1 pathway. These results suggest that LSNYP may be a promising candidate for the treatment of EMS.


Subject(s)
Drugs, Chinese Herbal , Endometriosis , Enhancer of Zeste Homolog 2 Protein , Estradiol , Hypoxia-Inducible Factor 1, alpha Subunit , Endometriosis/drug therapy , Endometriosis/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Drugs, Chinese Herbal/pharmacology , Estradiol/pharmacology , Animals , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Signal Transduction/drug effects , Mice , Adult , Cells, Cultured , Inflammation/drug therapy , Stromal Cells/drug effects , Stromal Cells/metabolism
3.
Front Mol Biosci ; 10: 1298457, 2023.
Article in English | MEDLINE | ID: mdl-38370978

ABSTRACT

Background: Endometriosis (EM) is a long-lasting inflammatory disease that is difficult to treat and prevent. Existing research indicates the significance of immune infiltration in the progression of EM. Efferocytosis has an important immunomodulatory function. However, research on the identification and clinical significance of efferocytosis-related genes (EFRGs) in EM is sparse. Methods: The EFRDEGs (differentially expressed efferocytosis-related genes) linked to datasets associated with endometriosis were thoroughly examined utilizing the Gene Expression Omnibus (GEO) and GeneCards databases. The construction of the protein-protein interaction (PPI) and transcription factor (TF) regulatory network of EFRDEGs ensued. Subsequently, machine learning techniques including Univariate logistic regression, LASSO, and SVM classification were applied to filter and pinpoint diagnostic biomarkers. To establish and assess the diagnostic model, ROC analysis, multivariate regression analysis, nomogram, and calibration curve were employed. The CIBERSORT algorithm and single-cell RNA sequencing (scRNA-seq) were employed to explore immune cell infiltration, while the Comparative Toxicogenomics Database (CTD) was utilized for the identification of potential therapeutic drugs for endometriosis. Finally, immunohistochemistry (IHC) and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were utilized to quantify the expression levels of biomarkers in clinical samples of endometriosis. Results: Our findings revealed 13 EFRDEGs associated with EM, and the LASSO and SVM regression model identified six hub genes (ARG2, GAS6, C3, PROS1, CLU, and FGL2). Among these, ARG2, GAS6, and C3 were confirmed as diagnostic biomarkers through multivariate logistic regression analysis. The ROC curve analysis of GSE37837 (AUC = 0.627) and GSE6374 (AUC = 0.635), along with calibration and DCA curve assessments, demonstrated that the nomogram built on these three biomarkers exhibited a commendable predictive capacity for the disease. Notably, the ratio of nine immune cell types exhibited significant differences between eutopic and ectopic endometrial samples, with scRNA-seq highlighting M0 Macrophages, Fibroblasts, and CD8 Tex cells as the cell populations undergoing the most substantial changes in the three biomarkers. Additionally, our study predicted seven potential medications for EM. Finally, the expression levels of the three biomarkers in clinical samples were validated through RT-qPCR and IHC, consistently aligning with the results obtained from the public database. Conclusion: we identified three biomarkers and constructed a diagnostic model for EM in this study, these findings provide valuable insights for subsequent mechanistic research and clinical applications in the field of endometriosis.

4.
Zhongguo Zhong Yao Za Zhi ; 47(11): 3073-3087, 2022 Jun.
Article in Chinese | MEDLINE | ID: mdl-35718532

ABSTRACT

Network Meta-analysis was performed to systematically compare the efficacy of different Chinese patent medicines for activating blood and resolving stasis in the treatment of endometriosis and to provide evidence-based references for clinical medication regimens. The relevant randomized controlled trials(RCTs) involving Chinese patent medicines combined with conventional treatment(experimental group) vs conventional treatment(control group) were retrieved from Chinese and English literature databases. The bias risk assessment tool recommended in Cochrane handbook 5.3 was used to evaluate the quality of the included studies. The result data of each outcome index was extracted for network Meta-analysis in Stata 15.0. A total of 44 RCTs were included in this study, involving 4 345 patients and 9 Chinese patent medicines. The network Meta-analysis revealed the following trends.(1)In terms of reducing the visual analogue scale(VAS) scores, Dan'e Fukang Plaster+conventional treatment>Xuefu Zhuyu Capsules+conventional treatment>Gongliuxiao Capsules+conventional treatment.(2)In terms of reducing cancer antigen CA125, Xiaojin Capsules+conventional treatment>Shaofu Zhuyu Granules+conventional treatment>Dan'e Fukang Plaster+conventional treatment.(3)In terms of reducing estradiol(E_2), Gongliuxiao Capsules+conventional treatment>Xiaojin Capsules+conventional treatment>Sanjie Zhentong Capsules+conventional treatment.(4) In terms of reducing recurrence rate, Guizhi Fuling Capsules+conventional treatment>Xuefu Zhuyu Capsules+conventional treatment>Dan'e Fukang Plaster+conventional treatment. The peroral Chinese patent medicines for activating blood and resolving stasis combined with conventional treatment have better efficacy in the treatment of endometriosis than conventional treatment. However, considering the low quality of the included literature, large-scale high-quality clinical trials are needed in the future research.


Subject(s)
Drugs, Chinese Herbal , Endometriosis , Capsules , China , Drugs, Chinese Herbal/therapeutic use , Endometriosis/drug therapy , Female , Humans , Network Meta-Analysis , Nonprescription Drugs/therapeutic use
5.
BMC Oral Health ; 21(1): 560, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732192

ABSTRACT

BACKGROUND: Nicotine is an important risk factor and the main toxic component associated with periodontitis. However, the mechanism of nicotine induced periodontitis is not clear. To investigated the mechanism through which nicotine regulates autophagy of human periodontal ligament cells (hPDLCs) through the alpha7 nicotinic acetylcholine receptor (α7 nAChR) and how autophagy further regulates the release of IL-1ß and IL-8 secretion in hPDLCs. METHODS: HPDLCs were obtained from root of extracted teeth and pre-incubated in alpha-bungarotoxin (α-BTX) or 3-Methyladenine (3-MA), followed by culturing in nicotine. We used a variety of experimental detection techniques including western blotting, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), transmission electron microscopy (TEM) and RT-qPCR to assess the expression of the LC3 protein, autolysosome, and release of IL-1ß and IL-8 from hPDLCs. RESULTS: Western blots, immunofluorescence and TEM results found that the nicotine significantly increased the autophagy expression in hPDLCs that was time and concentration dependent and reversed by α-BTX treatment (p < 0.05). RT-qPCR and ELISA results revealed a noticeable rise in the release of inflammatory factors IL-1ß and IL-8 from hPDLCs in response to nicotine. RT-qPCR and ELISA results showed that nicotine can significantly up-regulate the release of inflammatory factors IL-1ß and IL-8 in hPDLCs, and this effect can be inhibited by 3-MA (p < 0.05). CONCLUSIONS: Nicotine regulated autophagy of hPDLCs through α7 nAChR and in turn the regulation of the release of inflammatory factors 1L-1ß and 1L-8 by hPDLCs.


Subject(s)
Interleukin-8 , Nicotine , Autophagy , Cells, Cultured , Humans , Interleukin-8/genetics , Nicotine/toxicity , Periodontal Ligament
6.
Biochem Cell Biol ; 98(3): 354-361, 2020 06.
Article in English | MEDLINE | ID: mdl-31689122

ABSTRACT

The consumption of nicotine via smoking tobacco has been reported to stimulate the occurrence and progression of periodontitis. Many studies have demonstrated that nicotine prevents the regeneration of periodontal tissues primarily by inhibiting the proliferation of human periodontal ligament (PDL) cells. However, the mechanisms underlying this process are still unclear. Therefore, we investigated whether nicotine-upregulated miR-30a inhibited the proliferation of human PDL cells by downregulating cyclin E2 (CCNE2), in vitro. Quantitative real-time PCR analysis revealed that nicotine upregulated the expression of miR-30a in human PDL cells. In addition, nicotine inhibited the proliferation of human PDL cells by inducing cell cycle arrest. To support this hypothesis, we showed that nicotine downregulated the expression of CCNE2 in human PDL cells, whereas inhibition of miR-30a restored CCNE2 expression that had been downregulated by nicotine. Furthermore, using luciferase reporter assays, we found that miR-30a directly interacts with the CCNE2 3'UTR. In conclusion, these findings indicate that nicotine-upregulated miR-30a inhibits the proliferation of human PDL cells by downregulating the expression of CCNE2.


Subject(s)
Cyclins/metabolism , MicroRNAs/genetics , Nicotine/pharmacology , Periodontal Ligament/drug effects , Periodontitis/metabolism , 3' Untranslated Regions , Adolescent , Cell Cycle Checkpoints , Cell Proliferation , Cells, Cultured , Child , G1 Phase , Humans , Male , Periodontal Ligament/cytology , Real-Time Polymerase Chain Reaction , Up-Regulation
7.
Cancer Manag Res ; 12: 13357-13368, 2020.
Article in English | MEDLINE | ID: mdl-33402847

ABSTRACT

BACKGROUND: Long non-coding RNA (lncRNA) is a key part of non-coding RNA, and more and more evidence has revealed that it plays a vital role in tumors. NEAT1 is a lncRNA discovered in the early stage. However, it is still unclear whether NEAT1 and miR-204 play a regulatory role in lung cancer (LC). This research aimed to determine the biological function of NEAT1/miR-204 in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS: In order to research the function of NEAT1 in NSCLC, RT-PCR, Western blot, luciferase assay and RNA immunoprecipitation assay were used to determine the relationship between NEAT1, miR-204 and NUAK1. CCK8 test, cell migration and invasion test were used to explore the influence of NEAT1 on proliferation and metastasis of LC cells. Tumor allotransplantation was used to detect the influence of NEAT1 on the growth of LC. RESULTS: The results revealed that NEAT1 was obviously enhanced in LC cell lines. Further functional analysis showed that low expression of NEAT1 obviously suppressed the growth, migration and invasion of NSCLC and facilitated cell apoptosis. Determination of luciferase reporter gene revealed that miR-204 was the direct target of NEAT1 in LC. In addition, NUAK1 was called the direct target of miR-204, and miR-204/NUAK1 had saved the role of NEAT1 in NSCLC cells. Tumor allotransplantation experiments showed that knocking down NEAT1 could inhibit the growth of LC. CONCLUSION: In summary, our results showed that the down-regulation of NEAT1 in NSCLC inhibited its growth, migration and invasion through the miR-204/NUAK1 axis.

8.
Stem Cells Dev ; 26(16): 1186-1198, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28494644

ABSTRACT

The physiological root resorption of deciduous teeth is a normal phenomenon, but the mechanisms underlying this process are still unclear. In this study, deciduous dental pulp stem cells (DDPSCs) and permanent dental pulp stem cells (DPSCs) were derived from deciduous teeth and normal permanent teeth at different stages of resorption. In the middle stage of root resorption, DDPSCs exhibited an increase in the ability to induce osteoclast differentiation. Activation of the alpha 7 nicotinic acetylcholine receptor (α7 nAChR) by secretory mammalian Ly-6 urokinase-type plasminogen activator receptor-associated protein 1 (SLURP-1) caused a significant increase in the expression levels of NF-κB, receptor activator of nuclear factor-kappa B ligand (RANKL), and the ratio of RANKL/osteoprotegerin (OPG). These effects were inhibited by alpha-bungarotoxin (α-BTX). Furthermore, the expression levels of RANKL/OPG were significantly reduced following inhibition of NF-κB. High-strength, dynamic positive pressure increased the expression of SLURP-1 and α7 nAChR in DDPSCs in the stable stage. These data indicated that mechanical stress stimulated the expression of SLURP-1 and α7 nAChR in DDPSCs. Additionally, SLURP-1 activated α7 nAChR, thereby upregulating the expression of NF-κB and enhancing its activity, thus regulating RANKL/OPG expression and affecting the ability of DDPSCs to influence osteoclastogenesis, which likely enhances root resorption and leads to the physiological loss of deciduous teeth.


Subject(s)
Adult Stem Cells/metabolism , Dental Pulp/cytology , Osteogenesis , Root Resorption , Tooth, Deciduous/cytology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Adult Stem Cells/cytology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Line , Cells, Cultured , Child , Dental Pulp/metabolism , Dental Pulp/physiology , Humans , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Tooth, Deciduous/metabolism , Tooth, Deciduous/physiology , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics
9.
Hum Exp Toxicol ; 35(9): 983-90, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26553320

ABSTRACT

Periodontitis is a common infectious disease associated with destruction of periodontal ligaments and alveolar bones. CD4(+) T cell-mediated immune response is involved in the progression of periodontitis. Tobacco consumption increases the risk of periodontal disease. However, the impact of nicotine on the interaction between human periodontal ligament (PDL) cells and CD4(+) T cells remains unrevealed. Our study aims to investigate the effect of nicotine on PDL cells and the cocultured CD4(+) T cells. The PDL cell cultures were established by explants from healthy individuals, exposed to nicotine or α-bungarotoxin (α-BTX), and incubated solely or in combination with CD4(+) T cells. Afterwards, cell viability, secreted cytokines, and matrix metalloproteinases (MMPs) were evaluated. In monoculture of PDL cells, nicotine dramatically repressed cell viability and increased apoptosis. Meanwhile, α-BTX largely reversed the nicotine-induced apoptosis and increased viability of PDL cells. Compared with the monoculture, MMP-1, MMP-3, interleukin (IL)-1ß, IL-6, IL-17, and IL-21 in supernatant of cocultures were markedly elevated after treatment with nicotine. Moreover, α-BTX significantly attenuated nicotine-triggered production of these components either in mono- or co-cultures. In addition, PDL cell-derived CXCL12 following nicotine treatment recruited CD4(+) T cells. Above all, nicotine deteriorated periodontitis partially by promoting PDL cell-CD4(+) T cell-mediated inflammatory response and matrix degradation.


Subject(s)
Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , Cell Communication/drug effects , Nicotine/toxicity , Periodontal Ligament/drug effects , Bungarotoxins/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Matrix Metalloproteinases/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/immunology , Periodontitis/chemically induced , Periodontitis/immunology
11.
Cell Physiol Biochem ; 34(2): 423-31, 2014.
Article in English | MEDLINE | ID: mdl-25059554

ABSTRACT

BACKGROUND/AIMS: Tobacco smoking is a major risk factor for the occurrence and progression of periodontitis. We previously demonstrated that nicotine could induce the expression of α7 nicotinic acetylcholine receptors (α7 nAChR) in human and rat periodontal tissues. To further examine the signal pathways mediated by α7 nAChR in periodontal ligament (PDL) cells, we investigated whether nicotine affects interleukin-1ß (IL-1ß) and interleukin-8 (IL-8) via the α7 nAChR/NF-κB pathway in human PDL cells. METHODS: Human PDL cells were pre-incubated with alpha-bungarotoxin (α-BTX) or pyrrolidine dithiocarbamate (PDTC), then cultured with nicotine. Then, we used western blotting, a dual-luciferase reporter, real-time quantitative PCR and an enzyme-linked immunoassay to assess expression of the NF-κB p65 subunit, NF-κB activity and production of IL-1ß and IL-8 in human PDL cells. RESULTS: Compared with the control group, nicotine could significantly induce production of IL-1ß and IL-8 in human PDL cells and cause the similar effects on the expression of the NF-κB p65 subunit and NF-κB activity. CONCLUSION: This study demonstrates that nicotine could induce production of IL-1ß and IL-8 via the α7 nAChR/NF-κB pathway in human PDL cells, providing data for a better understanding of the relationships among smoking, nicotine, and periodontitis.


Subject(s)
Interleukin-1beta/biosynthesis , Interleukin-8/biosynthesis , NF-kappa B/metabolism , Nicotine/pharmacology , Periodontal Ligament/drug effects , Signal Transduction , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Base Sequence , Blotting, Western , Cells, Cultured , Child , DNA Primers , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Real-Time Polymerase Chain Reaction
12.
Stem Cells Dev ; 23(20): 2524-34, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24827498

ABSTRACT

Physiological primary teeth exfoliation is a normal phenomenon during teeth development. However, retained primary teeth can often be observed in the patients with cleidocranial dysplasia (CCD) caused by mutation of Runx2. The potential regulative mechanism is still unknown. In the present study, periodontal ligament stem cells (PDLSCs) were derived from different resorbed stages of primary teeth and permanent teeth from normal patients and primary teeth from CCD patient. The proliferative, osteogenic and osteoclast-inductive capacities of PDLSCs from each group were detected. We demonstrated here that the proliferative ability of PDLSCs was reduced while the osteogenic and the osteoclast-inductive capacity of PDLSCs were enhanced during root resorption. The results also showed that PDLSCs from permanent teeth and CCD patient expressed low level of Runx2 and RANKL while high level of OPG. However, expression of Runx2 and RANKL were increased while expression of OPG was decreased in PDLSCs derived from resorbed teeth. Furthermore, Runx2 regulating the expression of RANKL and OPG and the osteoclast-inductive capacity of PDLSCs were confirmed by gain or loss of function assay. These data suggest that PDLSCs promote osteoclast differentiation via Runx2 upregulating RANKL and downregulating OPG, leading to enhanced root resorption that results in physiological exfoliation of primary teeth.


Subject(s)
Core Binding Factor Alpha 1 Subunit/biosynthesis , Osteoprotegerin/biosynthesis , Periodontal Ligament/metabolism , RANK Ligand/biosynthesis , Root Resorption/metabolism , Stem Cells/metabolism , Tooth, Deciduous/metabolism , Cells, Cultured , Child , Child, Preschool , Female , Gene Expression Regulation/physiology , Humans , Male , Osteoclasts/cytology , Osteoclasts/metabolism , Periodontal Ligament/cytology , Stem Cells/cytology , Tooth Exfoliation/metabolism , Tooth, Deciduous/cytology
13.
PLoS One ; 8(12): e83102, 2013.
Article in English | MEDLINE | ID: mdl-24376645

ABSTRACT

AIMS: Cigarette smoking is one of the high risk factors of adult chronic periodontitis and nicotine is the well established toxic substance in cigarette. However, the mechanism of nicotine induced periodontitis is still unknown. Here we studied whether nicotine impaired the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) through activating α7 nicotinic acetylcholine receptor (α7 nAChR). METHODS: hPDLSCs with multi differentiation potential and surface makers for mesenchymal stem cells were harvested by limiting dilution technique. The level of mineralized nodule formation was assessed by alizarin red S staining. Expression level of ostegenic related genes and proteins were detected by real-time PCR and western blot analysis. The expression of α7 nAChR and its downstream signaling pathway were examined by western blot. The role of the receptor and related signaling pathway in nicotine impairing the osteogenic potential of hPDLSCs were also studied in different levels. RESULTS: Nicotine deteriorated the ostegenic differentiation of hPDLSCs in a dose dependent manner. Activation of α7 nAChR by nicotine treatment activated wnt/ß-catenin signaling pathway, leading to osteogenic deficiency of hPDLSCs. Blockage of α7 nAChR and wnt pathway inhibitor treatment rescued nicotine induced osteogenic differentiation deficiency. CONCLUSIONS: These data suggested that nicotine activated α7 nAChR expressed on PDLSCs and further activated wnt signaling downstream, thus deteriorating the osteogenic potential of PDLSCs. The impairment of osteogenic differentiation of PDLSCs by nicotine might lead to cigarette smoking related periodontitis.


Subject(s)
Nicotine/pharmacology , Osteogenesis/drug effects , Periodontal Ligament/drug effects , Stem Cells/drug effects , Wnt Proteins/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Adolescent , Cell Differentiation/drug effects , Child , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Primary Cell Culture , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/agonists , Wnt Proteins/metabolism , Young Adult , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , beta Catenin/genetics , beta Catenin/metabolism
14.
Int J Mol Med ; 31(4): 938-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23443505

ABSTRACT

Periodontitis, which is the main cause of tooth loss, is one of the most common chronic oral diseases in adults. Tooth loss is mainly a result of alveolar bone resorption, which reflects an increased osteoclast formation and activation. Osteoclast formation in periodontal tissue is a multistep process driven by osteoclastogenesis supporting cells such as human periodontal ligament (PDL) cells and CD4(+) T cells. Inflammatory cytokines, such as interleukin-1ß (IL-1ß), can induce osteoclastogenesis by affecting the expression of receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG) in human PDL cells. Nicotine, the major component in tobacco smoking and a specific agonist of the α7 nicotinic acetylcholine receptor (α7 nAChR), has been proven to regulate the expression of inflammatory cytokines in smoking-associated periodontitis. In this study, we investigated the mechanism(s) through which nicotine affects osteoclastogenesis in human PDL cells co-cultured/non-co-cultured with CD4(+) T cells. Human PDL cells were stimulated with nicotine (10-5 M) and/or α-bungarotoxin (α-BTX, specific antagonist of α7 nAChR, 10-8 M) before being co-cultured with CD4(+) T cells. Compared with mono-culture systems, stimulation with nicotine caused an increased secretion of IL-1ß in serum of human PDL cell-CD4(+) T cell co-culture, and the expression of RANKL in human PDL cells was further upregulated co-cultured with CD4(+) T cells, while no differences were observed in the expression of OPG between the co-culture and mono-culture systems. Our data suggested that nicotine upregulated IL-1ß secretion, further upregulated RANKL expression in smoking-associated periodontitis, which may aid in the better understanding of the relationship between nicotine and alveolar bone resorption.


Subject(s)
CD4-Positive T-Lymphocytes , Interleukin-1beta/metabolism , Nicotine/pharmacology , Osteoclasts , Periodontal Ligament/cytology , Analysis of Variance , Bungarotoxins , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Humans , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/analysis , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/analysis , RANK Ligand/genetics , RANK Ligand/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL