Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Dent Sci ; 18(2): 923, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37021247
2.
Medicine (Baltimore) ; 102(8): e32961, 2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36827024

ABSTRACT

Periodontal disease is often neglected and overlooking its initial symptoms can lead to tooth loss and systemic diseases. Patients with otitis media are at high risk of vestibular and balance dysfunction, consequently, and vertigo. Vertigo and dizziness are conditions with high reported incidences; they worsen with age and can burden health systems. The present study investigated whether periodontal disease causes dizziness. Research data covering 2008 through 2013 were retrieved from the National Health Insurance Research Database of Taiwan. Patients who were newly diagnosed as having periodontal disease or dizziness after at least 1 hospital admission or 3 outpatient visits were enrolled as participants. For our controls, we randomly selected individuals without periodontal disease who were sex- and age-matched with the investigated participants. In total, we enrolled 445 patients with periodontal disease and 1780 controls. The Kaplan-Meier curve indicated that the cumulative incidence of dizziness was significantly higher among the patients with periodontal disease relative to the controls. After adjustment for sex, age, income level, urbanization level, month of onset, and comorbidities, Cox proportional-hazards analysis revealed that patients with periodontal disease had an increased risk of dizziness (hazard ratio [HR]: 1.306, 95% confidence interval (CI): 1.155, 1.475). Compared with the controls, the risk of dizziness among patients with periodontal disease was higher for both female (HR: 1.439, 95%: 1.203, 1.720) and male patients (HR: 1.284, 95%: 1.123, 1.468); this risk was higher even when January (HR: 1.302, 95% CI: 1.145, 1.480), February (HR: 1.337, 95% CI: 1.178, 1.518), or March was excluded (HR: 1.308, 95% CI: 1.151, 1.487) and for patients without Ménière disease. Therefore, periodontal disease is not only a risk factor for dizziness but also an independent risk factor for dizziness. Future studies could clarify the mechanisms linking periodontal disease to dizziness.


Subject(s)
Dizziness , Periodontal Diseases , Adult , Female , Humans , Male , Cohort Studies , Comorbidity , Incidence , Periodontal Diseases/epidemiology , Proportional Hazards Models , Retrospective Studies , Risk Factors , Taiwan/epidemiology , Vertigo , Case-Control Studies
3.
Environ Toxicol ; 38(3): 628-634, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36413001

ABSTRACT

Bisphenol A-glycidyl methacrylate (BisGMA) is a methacrylate monomer that is mainly used in three-dimensional structures to reconstruct dental and bony defects. BisGMA has toxic and proinflammatory effects on macrophages. Rutin is a natural flavonol glycoside that is present in various plants and has useful biological effects, such as anti-inflammatory, anticancer, and antioxidative effects. The aim of this study was to investigate the anti-inflammation of rutin in macrophages after exposure to BisGMA. Pretreatment of the RAW264.7 macrophage with rutin at 0, 10, 30, and 100 µM for 30 min before being incubated with BisGMA at 0 or 3 µM. Proinflammatory cytokines and prostaglandin (PG) E2 were detected by enzyme-linked immunosorbent assay (ELISA). Nitric oxide (NO) was detected by the Griess assay. Expression and phosphorylation of proteins were measured by Western blot assay. Pretreatment with rutin inhibited the BisGMA-induced generation of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and PGE2, in macrophages. Rutin also suppressed the BisGMA-induced secretion of NO and expression of inducible nitric oxide synthase (iNOS) in a concentration-dependent manner. Furthermore, rutin suppressed the mitogen-activated protein kinase (MAPK) phosphorylation in a concentration-dependent manner. Finally, rutin suppressed the BisGMA-induced phosphorylation of nuclear factor (NF)-κB p65 and degradation of inhibitor of κB (IκB). These results indicate that the concentration of rutin has an inhibitory effect on proinflammatory mediator generation, MAPK phosphorylation, NF-κB p65 phosphorylation, and IκB degradation. In conclusion, rutin is a potential anti-inflammatory agent for BisGMA-stimulated macrophages through NF-κB p65 phosphorylation and IκB degradation resulting from MAPK phosphorylation.


Subject(s)
Mitogen-Activated Protein Kinases , NF-kappa B , NF-kappa B/metabolism , Mitogen-Activated Protein Kinases/metabolism , Bisphenol A-Glycidyl Methacrylate/metabolism , Bisphenol A-Glycidyl Methacrylate/pharmacology , Rutin/pharmacology , Macrophages , Cytokines/metabolism , Tumor Necrosis Factor-alpha/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism
4.
Int J Mol Sci ; 23(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36233071

ABSTRACT

Rutin, also called quercetin-3-rhamnosyl glucoside, is a natural flavonol glycoside present in many plants. Rutin is used to treat various diseases, such as inflammation, diabetes, and cancer. For polymeric biomaterials, triethylene glycol dimethacrylate (TEGDMA) is the most commonly used monomer and serves as a restorative resin, a dentin bonding agent and sealant, and a bone cement component. Overall, TEGDMA induces various toxic effects in macrophages, including cytotoxicity, apoptosis, and genotoxicity. The aim of this study was to investigate the protective mechanism of rutin in alleviating TEGDMA-induced toxicity in RAW264.7 macrophages. After treatment with rutin, we assessed the cell viability and apoptosis of TEGDMA-induced RAW264.7 macrophages using an methylthiazol tetrazolium (MTT) assay and Annexin V-FITC/propidium iodide assay, respectively. Subsequently, we assessed the level of genotoxicity using comet and micronucleus assays, assessed the cysteinyla aspartate specific proteinases (caspases) and antioxidant enzyme (AOE) activity using commercial kits, and evaluated the generation of reactive oxygen species (ROS) using a dichlorodihydrofluorescein diacetate (DCFH-DA) assay. We evaluated the expression of heme oxygenase (HO)-1, the expression of nuclear factor erythroid 2 related factor (Nrf-2), and phosphorylation of AMP activated protein kinase (AMPK) using the Western blot assay. The results indicated that rutin substantially reduced the level of cytotoxicity, apoptosis, and genotoxicity of TEGDMA-induced RAW264.7 macrophages. Rutin also blocked the activity of caspase-3, caspase-8, and caspase-9 in TEGDMA-stimulated RAW264.7 macrophages. In addition, it decreased TEGDMA-induced ROS generation and AOE deactivation in macrophages. Finally, we found that TEGDMA-inhibited slightly the HO-1 expression, Nrf-2 expression, and AMPK phosphorylation would be revered by rutin. In addition, the HO-1 expression, Nrf-2 expression, and AMPK phosphorylation was enhanced by rutin. These findings indicate that rutin suppresses TEGDMA-induced caspase-mediated toxic effects through ROS generation and antioxidative system deactivation through the Nrf-2/AMPK pathway. Therefore, rutin has the potential to serve as a novel antitoxicity agent for TEGDMA in RAW264.7 macrophages.


Subject(s)
AMP-Activated Protein Kinases , Rutin , AMP-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Apoptosis , Aspartic Acid , Biocompatible Materials/pharmacology , Bone Cements/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Caspases/metabolism , Dentin-Bonding Agents , Glucosides/pharmacology , Glycosides/pharmacology , Macrophages/metabolism , Polyethylene Glycols , Polymethacrylic Acids , Propidium , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Rutin/pharmacology
5.
Environ Toxicol ; 37(12): 3007-3012, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36178853

ABSTRACT

Composites, resins, and sealants that are commonly used in orthopedics and dentistry are based on 2,2-bis[p-(2'-hydroxy-3'-methacryloxypropoxy)phenylene]propane (BisGMA), which induces proinflammatory responses in macrophages. The present study aimed to explore the anti-inflammatory responses of wogonin, which is a natural dihydroxyl flavonoid compound, in BisGMA-treated macrophages. According to the findings, wogonin exhibits anti-inflammatory, antiallergic, anticancer, and antioxidative properties. The generation of nitric oxide (NO) and the expression of inducible nitric oxide synthase (iNOS) were noted to be inhibited by wogonin in BisGMA-treated macrophages. Furthermore, the production of proinflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 was reduced. In addition, BisGMA-induced nuclear factor (NF)-κB p65 phosphorylation and inhibitor of κB (IκB) degradation were inhibited. Finally, the BisGMA-induced phosphorylation of mitogen-activated protein kinases (MAPKs), including p38 MAPK, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) was inhibited. All these effects were induced by wogonin in the macrophages in a concentration-dependent manner. Similar inhibitory effects of wogonin were observed on the production of NO and proinflammatory cytokines, expression of iNOS, phosphorylation of NF-κB p65 and MAPK, and degradation of IκB. These results indicated that rutin is a potential anti-inflammatory agent for BisGMA-treated macrophages that undergo NFκB p65 phosphorylation and IκB degradation through upstream MAPK phosphorylation. Therefore, wogonin inhibits BisGMA-induced proinflammatory responses in macrophages through the regulation of the NFκB pathway and its upstream factor, MAPK.


Subject(s)
Lipopolysaccharides , NF-kappa B , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/metabolism , Macrophages , Anti-Inflammatory Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Nitric Oxide/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphorylation , Cytokines/metabolism , Cyclooxygenase 2/metabolism
6.
Anticancer Res ; 41(12): 6095-6104, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34848464

ABSTRACT

BACKGROUND/AIM: This study investigated the anti-metastatic effects of cetyltrimethylammonium bromide (CTAB) on tongue squamous cell carcinoma (TSCC) SCC4 cells. MATERIALS AND METHODS: Cell morphology, viability, cell cycle distribution, adhesion, migration, invasion and the expression levels of associated proteins were examined using microscopy, WST-1, wound-healing, Boyden chamber assays, and western blotting, respectively. RESULTS: CTAB significantly affected SCC4 cell morphology from spindle-shaped to cobblestone-shaped and resulted in loss of adherence. CTAB significantly inhibited cell adhesion, migration, and invasion of SCC4 cells, independent of cell viability. CTAB reduced expression of matrix metalloproteinases (MMPs) such as MMP3, MMP7, and MMP14 in a concentration-dependent manner, while it increased expression of tissue inhibitors of metalloproteinase 3 (TIMP3). In addition, CTAB reduced the phosphorylation of mothers against decapentaplegic homolog 2/3 (Smad2/3) proteins, which mediated CTAB-inhibited migration and invasion in SCC4 cells. These effects were reversed by TGF-ß1. CONCLUSION: CTAB attenuates the mesenchymal characteristics through upregulation of TIMP3 by inhibiting the canonical TGF-ß/Smad/miR-181b/TIMP3 signaling involved in extracellular matrix remodeling in SCC4 cells and might be a promising anti-metastatic therapeutic agent for TSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cetrimonium/therapeutic use , Smad2 Protein/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tongue Neoplasms/drug therapy , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Cetrimonium/pharmacology , Humans , Signal Transduction , Transfection
7.
Anticancer Res ; 41(8): 3789-3799, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34281838

ABSTRACT

BACKGROUND/AIM: Cetyltrimethylammonium bromide (CTAB), a quaternary ammonium surfactant, was shown to have antitumor effects in a cellular mode of head and neck squamous cell carcinoma (HNSCC), modulating apoptotic and cytotoxic processes. However, the mechanisms by which CTAB exerts its effects against the epithelial- mesenchymal transition in HNSCC remain poorly understood. In the present study, we investigated whether CTAB inhibits cellular mobility and invasiveness of hypopharyngeal squamous cell carcinoma (HPSCC) cells. MATERIALS AND METHODS: WST-1, cell-cycle phase distribution, and wound healing, as well as transwell assays were conducted. Changes in protein expression patterns and related signaling pathways involved in effects of CTAB on HPSCC cell lines were evaluated by western blotting. RESULTS: Treatment of human HPSCC cell lines with CTAB significantly altered their morphology from spindle-like to cobblestone-like by diminishing mesenchymal-like phenotypic characteristics. CTAB also hindered cell functional properties, including migration and invasion, independently of cell viability. In addition, western blot results demonstrated that treatment with CTAB reduced expression of mesenchymal markers. Further investigation showed that CTAB treatment suppressed the phosphorylation of extracellular-regulated kinase 1/2, mechanistic target of rapamycin kinase and AKT serine/threonine kinase 1. CTAB also repressed the expression and phosphorylation levels of epidermal growth factor receptor (EGFR) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), and the partial restoration of mesenchymal phenotype by EGF addition confirmed that CTAB inhibited migration and invasion in HPSCC cells by blocking the EGFR signaling pathway. CONCLUSION: Our results suggest that CTAB is involved in the suppression of EGFR-mediated mesenchymal phenotype and the molecular mechanism by which CTAB obstructs HPSCC cell metastasis may represent a promising strategy for use in HPSCC treatment.


Subject(s)
Cetrimonium/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Head and Neck Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/physiology , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , TOR Serine-Threonine Kinases/metabolism
8.
Environ Toxicol ; 36(1): 45-54, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32830914

ABSTRACT

Bisphenol-A-glycidyldimethacrylate (BisGMA) is a resin monomer frequently used in dentin restorative treatments. The leakage of BisGMA monomer from BisGMA-based polymeric resins can lead to cytotoxicity in macrophages. Rutin has various beneficial bioeffects, including antioxidation and antiinflammation. In this study, we found that pretreatment of RAW264.7 macrophages with rutin-inhibited cytotoxicity induced by BisGMA in a concentration-dependent manner. BisGMA-induced apoptosis, which was detected by levels of phosphatidylserine from the internal to the external membrane and formation of sub-G1, and genotoxicity, which was detected by cytokinesis-blocked micronucleus and single-cell gel electrophoresis assays, were inhibited by rutin in a concentration-dependent manner. Rutin suppressed the BisGMA-induced activation of caspase-3 and -9 rather than caspase-8. Rutin inhibited the activation of the mitochondrial apoptotic pathway, including cytochrome C release and mitochondria disruption, after macrophages were treated with BisGMA. Finally, BisGMA-induced reactive oxygen species (ROS) generation and antioxidant enzyme (AOE) deactivation could be reversed by rutin. Parallel trends were observed in the elevation of AOE activation and inhibition of ROS generation, caspase-3 activity, mitochondrial apoptotic pathway activation, and genotoxicity. These results suggested that rutin suppressed BisGMA-induced cytotoxicity through genotoxicity, the mitochondrial apoptotic pathway, and relatively upstream factors, including reduction of ROS generation and induction of AOE.

9.
Anticancer Res ; 40(9): 5059-5069, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878794

ABSTRACT

BACKGROUND/AIM: Liver cancer is the fourth leading cause of cancer-related mortality globally, of which hepatocellular carcinoma (HCC) accounts for 85-90% of total primary liver cancer. A drug shortage for HCC therapy triggered us to screen the small-molecule database with a high-throughput cellular screening system. Herein, we examined whether cetyltrimethylammonium bromide (CTAB) inhibits cellular mobility and invasiveness of Mahlavu HCC cells. MATERIALS AND METHODS: The effects of CTAB on cell viability were assessed using WST-1 assay, cell-cycle distribution using flow cytometric analysis, migration/invasion using woundhealing and transwell assays, and associated protein levels using western blotting. RESULTS: Treatment of Mahlavu cells with CTAB transformed its mesenchymal spindle-like morphology. In addition, CTAB exerted inhibitory effects on the migration and invasion of Mahlavu cells dose-dependently. CTAB also reduced the protein levels of matrix metalloproteinase-2 (MMP2), MMP9, RAC family small GTPase 1, SNAIL family transcriptional repressor 1 (SNAI1), SNAI2, TWIST family basic helix-loop-helix transcription factor 1 (TWIST1), vimentin, N-cadherin, phospho-fibroblast growth factor (FGF) receptor, phospho-phosphoinositide 3-kinase, phospho-v-Akt murine thymoma viral oncogene and phospho-signal transducer and activator of transcription 3 but increased the protein levels of tissue inhibitor of metalloproteinases-1/2 and E-cadherin. Rescue experiments proved that CTAB induced mesenchymal-epithelial transition in Mahlavu cells and this was significantly dose-dependently mitigated by basic FGF. CONCLUSION: CTAB suppressed the migration and invasion of Mahlavu cells through inhibition of the FGF signaling pathway. CTAB seems to be a potential agent for preventing metastasis of hepatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cetrimonium/pharmacology , Fibroblast Growth Factors/metabolism , Signal Transduction/drug effects , Animals , Biomarkers , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays
10.
Anticancer Res ; 40(8): 4513-4522, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32727781

ABSTRACT

BACKGROUND/AIM: Hepatocellular carcinoma (HCC) arises from hepatocytes, and is the most frequently occurring malignancy of primary liver cancer. In this study, we investigated the anti-metastatic effects of the quaternary ammonium compound, cetyltrimethylammonium bromide (CTAB), on HA22T/VGH HCC cells. MATERIALS AND METHODS: According to our preliminary data, the effect of CTAB on cell cycle distribution, migration, invasion and the associated protein levels was examined using flow cytometry, wound-healing migration, Matrigel transwell invasion assay and western blotting under sub-lethal concentrations. RESULTS: CTAB treatment of HA22T/VGH cells casued dose-dependent mesenchymal-epithelial transition (MET)-like changes and impaired migration and invasion capabilities. In addition, CTAB reduced the levels of metastasis-related proteins including c-Met, phosphoinositide 3-kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), ribosomal protein S6 kinase (p70S6K), Twist, N-cadherin, and Vimentin. Moreover, pretreatment with hepatocyte growth factor (HGF) rescued CTAB-mediated effects. CONCLUSION: CTAB exhibited potent anti-EMT and anti-metastatic activities through the inhibition of migration and invasion of HA22T/VGH cells. CTAB interrupted the mesenchymal characteristics of HA22T/VGH cells, which were significantly alleviated by HGF in a dose-dependent manner. CTAB has the potential to evolve as a therapeutic agent for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cetrimonium/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/metabolism , Signal Transduction/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
Article in English | MEDLINE | ID: mdl-32290568

ABSTRACT

Multiple sclerosis (MS) is an inflammatory neurological disease characterized by autoimmune-mediated demyelination of the central nervous system. Genetic and environmental factors may contribute to the development of MS. This has not been confirmed yet. Dental amalgam has long been controversial in MS due to its mercury content but the toxicological implications of mercury-containing amalgam fillings (AMF) for MS remain to be elucidated. We conducted a case-control study to investigate the association between AMF and the risk of MS from the Taiwanese National Health Insurance Research Database (NHIRD). Case (n = 612) and control (n = 612) groups were matched by sex, age, urbanization level, monthly income, and Charlson comorbidity index by propensity score matched with a 1:1 ratio from 2000 to 2013. Differences between cases and controls was not statistically significant (OR: 0.82, 95% CI = 0.65-1.05). In subjects stratified by gender, MS was also not associated with AMF for women (OR: 0.743, 95% CI = 0.552-1.000) and men (OR: 1.006, 95% CI = 0.670-1.509), respectively. In summary, this Taiwanese nationwide population-based case-control study did not find an association between MS and AMF.


Subject(s)
Dental Amalgam , Mercury , Multiple Sclerosis , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Dental Amalgam/toxicity , Female , Humans , Male , Mercury/analysis , Mercury/toxicity , Middle Aged , Multiple Sclerosis/chemically induced , Multiple Sclerosis/epidemiology , Taiwan/epidemiology , Young Adult
12.
Anticancer Res ; 39(8): 4149-4164, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31366500

ABSTRACT

BACKGROUND/AIM: Signaling regulation of myeloid zinc finger 1 (MZF1) has been implicated in the progression of many human malignancies; however, the mechanistic action of MZF1 in triple-negative breast cancer (TNBC) progression remains elusive. In this study, the aim was to investigate the molecular mechanisms of MZF1 and its functional role in TNBC cellular migration and invasion. MATERIALS AND METHODS: Hs578T and MDA-MB-231 cells were transfected to stably express the acidic domain of MZF1 (MZF160-72), or were transfected with MZF1-specific or ELK1-specific short hairpin RNA (shRNA). Changes in cell morphology and distributions of cellular proteins were observed and subsequently migration and invasion were measured by wound healing and transwell assays. Expression levels of epithelial-mesenchymal transition (EMT)-related genes were carried out using immunoblotting and quantitative reverse transcription-polymerase chain reaction (RT-PCR) assays. Data of transcriptional regulation were obtained from promoter-luciferase reporter and chromatin immunoprecipitation (ChIP) assays. RESULTS: Herein, we found that MZF1 in high-level MZF1-expressing TNBC cells is associated with cell migration, invasion, and mesenchymal phenotype. MZF1 interacted with the promoter region of insulin-like growth factor 1 receptor (IGF1R) to drive invasion and metastasis of high-level MZF1-expressing TNBC cells. Exogenous expression of the acidic domain of MZF1 repressed the binding of endogenous MZF1 to IGF1R promoter via blocking the interaction with ETS-like gene 1 (ELK1). This blockage not only caused MZF1 protein degradation, but also restrained ELK1 nuclear localization in high-level MZF1-expressing TNBC cells. MZF1, but not ELK1, was necessary for the retention of mesenchymal phenotype by repressing IGF1R promoter activity in TNBC cells expressing high levels of MZF1. Activation of the IGF1R-driven p38MAPK-ERα-slug-E-cadherin signaling axis mediated the conversion of mesenchymal cell to epithelial phenotype, caused by MZF1 destabilization. These results suggest that MZF1 is an oncogenic inducer. CONCLUSION: Blocking of the MZF1/ELK1 interaction to reduce MZF1 protein stability by saturating the endogenous MZF1/ELK1 binding domains might be a promising therapeutic strategy for the treatment of high-level MZF1-expressing TNBC.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Receptors, Somatomedin/genetics , Triple Negative Breast Neoplasms/genetics , ets-Domain Protein Elk-1/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement/genetics , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic/genetics , Protein Domains/genetics , Receptor, IGF Type 1 , Signal Transduction/genetics , Triple Negative Breast Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/genetics
13.
Anticancer Res ; 39(7): 3621-3631, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262888

ABSTRACT

BACKGROUND/AIM: Cetrimonium bromide (CTAB), a quaternary ammonium surfactant, is an antiseptic agent against bacteria and fungi. However, the mechanisms by which its pharmacological actions affect epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) cells, such as adenocarcinoma in SK-HEP-1 cells, have not been investigated. We, thereby, investigated whether CTAB inhibits cellular mobility and invasiveness of human hepatic adenocarcinoma in SK-HEP-1 cells. MATERIALS AND METHODS: SK-HEP-1 cells were treated with CTAB, and subsequent migration and invasion were measured by wound healing and transwell assays. Protein expression was detected by immunoblotting analysis. RESULTS: Our data revealed that treatment of SK-HEP-1 cells with CTAB altered their mesenchymal spindle-like morphology. CTAB exerted inhibitory effects on the migration and invasion of SK-HEP-1 cells dose-dependently, and reduced protein levels of matrix metalloproteinase-2 (MMP-2), MMP-9, snail, slug, twist, vimentin, fibronectin, N-cadherin, Smad2, Smad3, Smad4, phosphoinositide-3-kinase (PI3K), p-PI3K, Akt, p-Akt, ß-catenin, mammalian target of rapamycin (mTOR), p-mTOR, p-p70S6K, p-extracellular signal-regulated kinases (ERK)1/2, p-p38 mitogen-activated protein kinase (MAPK) and p-c-Jun N-terminal kinase (JNK), but increased protein levels of tissue inhibitor matrix metalloproteinase-1 (TIMP-1), TIMP-2, claudin-1 and p-GSK3ß. Based on these observations, we suggest that CTAB not only inhibits the canonical transforming growth factor-ß (TGF-ß) signaling pathway though reducing SMADs (an acronym from the fusion of Caenorhabditis elegans Sma genes and the Drosophila Mad, Mothers against decapentaplegic proteins), but also restrains the non-canonical TGF-ß signaling including MAPK pathways like ERK1/2, p38 MAPK, JNK and PI3K. CONCLUSION: CTAB is involved in the suppression of TGF-ß-mediated mesenchymal phenotype and could be a potent medical agent for use in controlling the migration and invasion of hepatic adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Cetrimonium/pharmacology , Liver Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Adenocarcinoma/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Humans , Liver Neoplasms/drug therapy , Neoplasm Invasiveness , Signal Transduction/drug effects
14.
Environ Toxicol ; 34(4): 486-494, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30609183

ABSTRACT

Macrophages not only play an important role in the innate immune response but also participate in many inflammatory and infectious diseases including asthma, diabetes, obesity, cardiovascular diseases, and cancers. Bisphenol A (BPA) is the most commonly used component for plastic products. However, BPA is an endocrine disruptor for mammals and participates in several inflammatory and infectious diseases. Up until now, there are no researches demonstrated the potential role of BPA in macrophage activation and its relative mechanism. BPA promoted the generation of proinflammatory cytokines IL-1ß, IL-6, and TNFα in a concentration-dependent manner (P < 0.05). BPA was identified to increase the expression of proinflammatory mediators NO and PGE2, and its upstream factors iNOS, COX2, and cPLA2 in a concentration-dependent manner (P < 0.05). Phosphorylation and nuclear translocation of NF-κB p65 were significantly induced by BPA via IκB degradation (P < 0.05). In addition, phosphorylation of ERK significantly induced by BPA at a concentration which was less than that for phosphorylation of p38 MAPK and JNK (P < 0.05). Furthermore, phosphorylation of STAT3 significantly induced by BPA at a concentration lower than that for phosphorylation of STAT1 (P < 0.05). Phosphorylation of JAK1 and JAK2 was also significantly induced by BPA in a concentration-dependent manner (P < 0.05).


Subject(s)
Benzhydryl Compounds/toxicity , Cytokines/genetics , Janus Kinase 1/drug effects , Janus Kinase 2/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Phenols/toxicity , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Animals , Dose-Response Relationship, Drug , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Phosphorylation , RAW 264.7 Cells
15.
Food Chem Toxicol ; 122: 215-224, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30312649

ABSTRACT

Bisphenol A (BPA) is primarily used in production of polycarbonate plastics and epoxy resins including plastic containers. BPA is an endocrine disruptor and supposes to induce asthma and cancer. However, so far only a few evidences have shown the BPA-induced toxic effect and its related mechanism in macrophages. BPA demonstrated cytotoxic effect on RAW264.7 macrophages in a concentration and time-dependent manner. BPA induces necrosis, apoptosis, and genotoxicity in a concentration-dependent manner. Phosphorylation of cytochrome C (cyto C) and p53 was due to mitochondrial disruption via BCL2 and BCL-XL downregulation and BAX, BID, and BAD upregulation. Both caspase-dependent, including caspase-9, caspase-3, and PARP-1 cleavage, and caspase-independent, such as nuclear translocation of AIF, pathways were activated by BPA. Furthermore, generation of reactive oxygen species (ROS) and reduction of antioxidative enzyme activities were induced by BPA. Parallel trends were observed in the effect of BPA on cytotoxicity, apoptosis, genotoxicity, p53 phosphorylation, BCL2 family expression exchange, caspase-dependent and independent apoptotic pathways, and ROS generation in RAW264.7 macrophages. Finally, BPA-exhibited cytotoxicity, apoptosis, and genotoxicity could be inhibited by N-acetylcysteine. These results indicated that the toxic effect of BPA was functioning via oxidative stress-associated mitochondrial apoptotic pathway in macrophages.


Subject(s)
Apoptosis/drug effects , Benzhydryl Compounds/toxicity , Cell Survival/drug effects , Endocrine Disruptors/toxicity , Macrophages/drug effects , Mitochondria/drug effects , Mutagens/toxicity , Oxidative Stress/drug effects , Phenols/toxicity , Acetylcysteine/pharmacology , Animals , Apoptosis Inducing Factor/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Catalase/metabolism , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Glutathione Peroxidase/metabolism , Macrophages/metabolism , Mice , Mitochondria/enzymology , Mitochondria/metabolism , Phosphorylation , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Stability , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism , RAW 264.7 Cells , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/metabolism
16.
J Formos Med Assoc ; 115(8): 602-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27287534

ABSTRACT

BACKGROUND/PURPOSE: Gingival overgrowth occurs as a side effect of systemic medication with immunosuppressant cyclosporine A (CsA). Slug, a master regulator of epithelial-mesenchymal transition, is dramatically upregulated in a variety of fibrotic diseases. The aim of this study is to investigate the role of epithelial-mesenchymal transition marker Slug in the pathogenesis of CsA-induced gingival overgrowth. METHODS: Clinically healthy gingiva and CsA-induced gingival overgrowth specimens were analyzed by immunohistochemistry. The effect of CsA on normal human gingival fibroblasts (HGFs) was used to elucidate whether Slug expression could be affected by CsA by real-time reverse transcription-polymerase chain reaction and western blot. Cell proliferation in CsA-treated HGFs with Slug lentiviral-mediated shRNAi knockdown was evaluated by tetrazolium bromide reduction assay. RESULTS: Slug expression was higher in CsA-induced gingival overgrowth specimens than in clinical healthy gingiva (p < 0.05). Slug expression was significantly higher in CsA-induced gingival overgrowth specimens with higher levels of inflammatory infiltrates (p < 0.05). CsA was found to increase Slug transcript and protein expression in HGFs in a dose-dependent manner (p < 0.05). In addition, knockdown of Slug significantly suppressed CsA-induced cell proliferation in HGFs (p < 0.05). CONCLUSION: Taken together, upregulation of Slug in HGFs stimulated by CsA may play an important role in the pathogenesis of CsA-induced gingival overgrowth.


Subject(s)
Cyclosporine/adverse effects , Epithelial-Mesenchymal Transition/genetics , Gingival Overgrowth/chemically induced , Gingival Overgrowth/genetics , Immunosuppressive Agents/adverse effects , Snail Family Transcription Factors/genetics , Blotting, Western , Case-Control Studies , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Gene Knockdown Techniques , Gingiva/pathology , Humans , Taiwan , Up-Regulation/drug effects
17.
Environ Toxicol ; 31(2): 176-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26756871

ABSTRACT

Bisphenol-A-glycidyldimethacrylate (BisGMA) is a frequently used monomer in dental restorative resins. However, BisGMA could leach from dental restorative resins after polymerization leading to inflammation in the peripheral environment. Wogonin, a natural flavone derivative, has several benefits, such as antioxidative, anti-inflammatory and neuroprotective properties. Pretreatment of macrophage RAW264.7 cells with wogonin inhibited cytotoxicity which is induced by BisGMA in a concentration-dependent manner. BisGMA induced apoptotic responses, such as redistribution of phosphatidylserine from the internal to the external membrane and DNA fragmentation, were decreased by wogonin in a concentration-dependent manner. In addition, BisGMA-induced genotoxicity, which detected by cytokinesis-blocked micronucleus and single-cell gel electrophoresis assays, were inhibited by wogonin in a concentration-dependent manner. Furthermore, wogonin suppressed BisGMA-induced activation of intrinsic caspase pathways, such as caspases-3 and -8. Parallel trends were observed in inhibition of caspase-3 and -8 activities, apoptosis, and genotoxicity. These results indicate wogonin suppressed the BisGMA-induced apoptosis and genotoxicity mainly via intrinsic caspase pathway in macrophages.


Subject(s)
Antimutagenic Agents/pharmacology , Bisphenol A-Glycidyl Methacrylate/toxicity , Caspases/drug effects , Cell Survival/drug effects , Flavanones/pharmacology , Free Radical Scavengers/pharmacology , Macrophages/drug effects , Mutagens/toxicity , Resins, Synthetic/toxicity , Animals , DNA Fragmentation , Mice , Micronucleus Tests , Phosphatidylserines/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
18.
Clin Oral Investig ; 20(5): 1029-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26336810

ABSTRACT

OBJECTIVES: Transglutaminase-2 (TGM-2) protein is involved in the cross-linking of matrix proteins resulting in several fibrotic disorders and can be induced by reactive oxygen species (ROS). Little is known about its role in the development of oral submucocal fibrosis (OSF). Hence, we hypothesize that TGM-2 may have a role in the pathogenesis of areca quid chewing-associated OSF and arecoline, a major areca nut alkaloid, could regulate TGM-2 via ROS generation. MATERIALS: Forty OSF specimens from areca quid chewing-associated OSF and ten normal buccal mucosa biopsy samples without areca quid chewing were analyzed by immunohistochemistry. The expression of TGM-2 from fibroblasts cultured from OSF and normal buccal mucosa was evaluated by Western blot. The effect of arecoline on normal buccal mucosa fibroblasts (BMFs) was used to elucidate whether TGM-2 expression could be affected by arecoline by using 2', 7'-dichlorofluorescein diacetate assay and Western blot. In addition, glutathione precursor N-acetyl-L-cysteine (NAC) and epigallocatechin-3 gallate (EGCG) were added to find the possible regulatory mechanisms. RESULTS: TGM-2 expression was significantly higher in OSF specimens than normal specimens (p < 0.05). Fibroblasts derived from OSF were found to exhibit higher TGM-2 expression than BMFs in protein levels (p < 0.05). Arecoline significantly upregulated the intracellular ROS generation in a dose-dependent manner (p < 0.05). TGM-2 protein induced by arecoline was found in BMFs in a dose-dependent manner (p < 0.05). Treatment with NAC and EGCG markedly inhibited TGM-2 expression induced by arecoline (p < 0.05). CONCLUSIONS: Our results suggest that TGM-2 expression is significantly upregulated in OSF tissues from areca quid chewers. Arecoline-upregulated TGM-2 expression may be mediated by ROS generation. CLINICAL RELEVANCE: TGM-2 protein is upregulated in areca quid chewing-associated OSF. Using this in vitro model, antioxidants could inhibit arecoline-upregulated TGM-2 expression. NAC and EGCG may serve as a useful agent in controlling OSF.


Subject(s)
Areca , GTP-Binding Proteins/metabolism , Oral Submucous Fibrosis/enzymology , Oral Submucous Fibrosis/prevention & control , Reactive Oxygen Species/metabolism , Transglutaminases/metabolism , Acetylcysteine/pharmacology , Arecoline/pharmacology , Blotting, Western , Catechin/analogs & derivatives , Catechin/pharmacology , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Protein Glutamine gamma Glutamyltransferase 2
20.
J Dent Sci ; 11(4): 463-467, 2016 Dec.
Article in English | MEDLINE | ID: mdl-30895013

ABSTRACT

BACKGROUND/PURPOSE: Fibroblast growth factor (FGF)-2 is known as a signaling molecule that induces tissue regeneration. Little is known about the effect of FGF-2 on cementoblasts for periodontal and periapical regeneration. The aim of this study was to investigate the effects of FGF-2 on murine immortalized cementoblast cell line (OCCM.30). MATERIALS AND METHODS: Cell growth and proliferation was judged by using alamar blue reduction assay. Flow cytometry analysis was used to evaluate Stro-1 positive cells expression with or without FGF-2. Western blot was used to evaluate the expression of phosphorylated serine-threonine kinase Akt (p-Akt) and extracellular signal-regulated protein kinase (p-ERK) in cementoblasts. RESULTS: FGF-2 was found to increase cell growth in a dose-dependent manner (P < 0.05). The concentration of 10 ng/mL FGF-2 enhanced cell proliferation in a time-dependent manner (P < 0.05). In addition, 10 ng/mL FGF-2 significantly increased the number of Stro-1 positive cells in the first 24 hours (P < 0.05). Moreover, 10 ng/mL FGF-2 was found to upregulate p-Akt and p-ERK in a time-dependent manner (P < 0.05). CONCLUSION: Taken together, FGF-2 could increase cementoblast growth, proliferation, and Stro-1 positive cells. These enhancements are associated with the upregulation of p-Akt and p-ERK expression. The application of FGF-2 may provide benefit for periodontal and periapical regeneration during the early phase of wound healing.

SELECTION OF CITATIONS
SEARCH DETAIL
...