Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 22-28, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38650160

ABSTRACT

This study aimed to evaluate the physiological role of NAMPT associated with MDPC-23 odontoblast cell proliferation. Cell viability was measured using the (DAPI) staining, caspase activation analysis and immunoblotting were performed. Visfatin promoted MDPC-23 odontoblast cell growth in a dose-dependent manner. Furthermore, the up-regulation of Visfatin promoted odontogenic differentiation and accelerated mineralization through an increase in representative odontoblastic biomarkers in MDPC-23 cells. However, FK-866 cell growth in a dose-dependent manner induced nuclear condensation and fragmentation. FK-866-treated cells showed H&E staining and increased apoptosis compared to control cells. The expression of anti-apoptotic factors components of the mitochondria-dependent intrinsic apoptotic pathway significantly decreased following FK-866 treatment. The expression of pro-apoptotic increased upon FK-866 treatment. In addition, FK-866 activated caspase-3 and PARP to induce cell death. In addition, after treating FK-866 for 72 h, the 3/7 activity of MDPC-23 cells increased in a concentration-dependent manner, and the IHC results also confirmed that Caspase-3 increased in a concentration-dependent. Therefore, the presence or absence of NAMPT expression in dentin cells was closely related to cell proliferation and formation of extracellular substrates.


Subject(s)
Apoptosis , Cell Proliferation , Nicotinamide Phosphoribosyltransferase , Odontoblasts , Nicotinamide Phosphoribosyltransferase/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Odontoblasts/drug effects , Odontoblasts/cytology , Odontoblasts/metabolism , Animals , Mice , Cell Line , Cytokines/metabolism , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Acrylamides/pharmacology , Odontogenesis/drug effects
2.
Biol Pharm Bull ; 47(2): 539-546, 2024.
Article in English | MEDLINE | ID: mdl-38417906

ABSTRACT

Metformin is an anti-diabetic drug that exerts protective effects against neurodegenerative diseases. In this study, we investigated the protective effects of metformin against manganese (Mn)-induced cytotoxicity associated with Parkinson's disease-like symptoms in N27-A dopaminergic (DA) cells. Metformin (0.1-1 mM) suppressed Mn (0.4 mM)-induced cell death in a concentration-dependent manner. Metformin pretreatment effectively suppressed the Mn-mediated increase in the levels of oxidative stress markers, such as reactive oxygen species (ROS) and thiobarbituric acid reactive substances. Moreover, metformin restored the levels of the antioxidants, superoxide dismutase, intracellular glutathione, and glutathione peroxidase, which were reduced by Mn. Metformin (0.5 mM) significantly attenuated the decrease in sirtuin-1 (SIRT1) and peroxisome proliferator activated receptor gamma coactivator-1 alpha levels, which were increased by Mn (0.4 mM). In addition, metformin inhibited the expression of microRNA-34a, which directly targeted SIRT1. Metformin also inhibited the loss of Mn-induced mitochondrial membrane potential (ΔΨm) and activation of the apoptosis marker, caspase-3. Furthermore, metformin-mediated inhibition of ROS generation and caspase-3 activation, recovery of ΔΨm, and restoration of cell viability were partially reversed by the SIRT1 inhibitor, Ex527. These results suggest that metformin may protects against Mn-induced DA neuronal cell death mediated by oxidative stress and mitochondrial dysfunction possibly via the regulation of SIRT1 pathway.


Subject(s)
Manganese , Metformin , Manganese/toxicity , Manganese/metabolism , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , Metformin/pharmacology , Sirtuin 1/metabolism , Apoptosis , Oxidative Stress , Dopaminergic Neurons
3.
Mol Cells ; 46(4): 245-255, 2023 04 30.
Article in English | MEDLINE | ID: mdl-36896597

ABSTRACT

This study aimed to exploring the pathophysiological mechanism of 7α,25-dihydroxycholesterol (7α,25-DHC) in osteoarthritis (OA) pathogenesis. 7α,25-DHC accelerated the proteoglycan loss in ex vivo organ-cultured articular cartilage explant. It was mediated by the decreasing extracellular matrix major components, including aggrecan and type II collagen, and the increasing expression and activation of degenerative enzymes, including matrix metalloproteinase (MMP)-3 and -13, in chondrocytes cultured with 7α,25-DHC. Furthermore, 7α,25-DHC promoted caspase dependent chondrocytes death via extrinsic and intrinsic pathways of apoptosis. Moreover, 7α,25-DHC upregulated the expression of inflammatory factors, including inducible nitric oxide synthase, cyclooxygenase-2, nitric oxide, and prostaglandin E2, via the production of reactive oxygen species via increase of oxidative stress in chondrocytes. In addition, 7α,25-DHC upregulated the expression of autophagy biomarker, including beclin-1 and microtubule-associated protein 1A/1B-light chain 3 via the modulation of p53-Akt-mTOR axis in chondrocytes. The expression of CYP7B1, caspase-3, and beclin-1 was elevated in the degenerative articular cartilage of mouse knee joint with OA. Taken together, our findings suggest that 7α,25-DHC is a pathophysiological risk factor of OA pathogenesis that is mediated a chondrocytes death via oxiapoptophagy, which is a mixed mode of apoptosis, oxidative stress, and autophagy.


Subject(s)
Osteoarthritis , Proto-Oncogene Proteins c-akt , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Chondrocytes/metabolism , Beclin-1/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , TOR Serine-Threonine Kinases/metabolism , Cells, Cultured
4.
Korean J Physiol Pharmacol ; 26(6): 447-456, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36302620

ABSTRACT

The present study was carried out to investigate the effect of Arctigenin on cell growth and the mechanism of cell death elicited by Arctigenin were examined in FaDu human pharyngeal carcinoma cells. To determine the apoptotic activity of Arctigenin in FaDu human pharyngeal carcinoma cells, cell viability assay, DAPI staining, caspase activation analysis, and immunoblotting were performed. Arctigenin inhibited the growth of cells in a dose-dependent manner and induced nuclear condensation and fragmentation. Arctigenin-treated cells showed caspase-3/7 activation and increased apoptosis versus control cells. FasL, a death ligand associated with extrinsic apoptotic signaling pathways, was up-regulated by Arctigenin treatment. Moreover, caspase-8, a part of the extrinsic apoptotic pathway, was activated by Arctigenin treatments. Expressions of anti-apoptotic factors such as Bcl-2 and Bcl-xL, components of the mitochondria-dependent intrinsic apoptosis pathway, significantly decreased following Arctigenin treatment. The expressions of pro-apoptotic factors such as BAX, BAD and caspase-9, and tumor suppressor -53 increased by Arctigenin treatments. In addition, Arctigenin activated caspase-3 and poly (ADP-ribose) polymerase (PARP) induced cell death. Arctigenin also inhibited the proliferation of FaDu cells by the suppression of p38, NF-κB, and Akt signaling pathways. These results suggest that Arctigenin may inhibit cell proliferation and induce apoptotic cell death in FaDu human pharyngeal carcinoma cells through both the mitochondria-mediated intrinsic pathway and the death receptor-mediated extrinsic pathway.

5.
Korean J Physiol Pharmacol ; 26(1): 37-45, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34965994

ABSTRACT

The aim of the present study was to investigate the physiological role of nicotinamide phosphoribosyltransferase (NAMPT) associated with odontogenic differentiation during tooth development in mice. Mouse dental papilla cell-23 (MDPC- 23) cells cultured in differentiation media were stimulated with the specific NAMPT inhibitor, FK866, and Visfatin (NAMPT) for up to 10 days. The cells were evaluated after 0, 4, 7, and 10 days. Cell viability was measured using the 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide assay. The mineralization assay was performed by staining MDPC-23 cells with Alizarin Red S solution. After cultivation, MDPC-23 cells were harvested for quantitative PCR or Western blotting. Analysis of variance was performed using StatView 5.0 software (SAS Institute Inc., Cary, NC, USA). Statistical significance was set at p < 0.05. The expression of NAMPT increased during the differentiation of murine odontoblast-like MDPC-23 cells. Furthermore, the up-regulation of NAMPT promoted odontogenic differentiation and accelerated mineralization through an increase in representative odontoblastic biomarkers, such as dentin sialophosphoprotein, dentin matrix protein-1, and alkaline phosphatase in MDPC-23 cells. However, treatment of the cells with the NAMPT inhibitor, FK866, attenuated odontogenic differentiation, as evidenced by the suppression of odontoblastic biomarkers. These data indicate that NAMPT regulated odontoblastic differentiation through the regulation of odontoblastic biomarkers. The increase in NAMPT expression in odontoblasts was closely related to the formation of the extracellular matrix and dentin via the Runx signaling pathway. Therefore, these data suggest that NAMPT is a critical regulator of odontoblast differentiation during tooth development.

6.
Oxid Med Cell Longev ; 2021: 8684725, 2021.
Article in English | MEDLINE | ID: mdl-33833854

ABSTRACT

Osteoarthritis (OA) is the most common degenerative joint disease with chronic joint pain caused by progressive degeneration of articular cartilage at synovial joints. Acteoside, a caffeoylphenylethanoid glycoside, has various biological activities such as antimicrobial, anti-inflammatory, anticancer, antioxidative, cytoprotective, and neuroprotective effect. Further, oral administration of acteoside at high dosage does not cause genotoxicity. Therefore, the aim of present study is to verify the anticatabolic effects of acteoside against osteoarthritis and its anticatabolic signaling pathway. Acteoside did not decrease the viabilities of mouse fibroblast L929 cells used as normal cells and primary rat chondrocytes. Acteoside counteracted the IL-1ß-induced proteoglycan loss in the chondrocytes and articular cartilage through suppressing the expression and activation of cartilage-degrading enzyme such as matrix metalloproteinase- (MMP-) 13, MMP-1, and MMP-3. Furthermore, acteoside suppressed the expression of inflammatory mediators such as inducible nitric oxide synthase, cyclooxygenase-2, nitric oxide, and prostaglandin E2 in the primary rat chondrocytes treated with IL-1ß. Subsequently, the expression of proinflammatory cytokines was decreased by acteoside in the primary rat chondrocytes treated with IL-1ß. Moreover, acteoside suppressed not only the phosphorylation of mitogen-activated protein kinases in primary rat chondrocytes treated with IL-1ß but also the translocation of NFκB from the cytosol to the nucleus through suppression of its phosphorylation. Oral administration of 5 and 10 mg/kg acteoside attenuated the progressive degeneration of articular cartilage in the osteoarthritic mouse model generated by destabilization of the medial meniscus. Our findings indicate that acteoside is a promising potential anticatabolic agent or supplement to attenuate or prevent progressive degeneration of articular cartilage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucosides/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-1beta/metabolism , Osteoarthritis/metabolism , Phenols/pharmacology , Signal Transduction , Animals , Anti-Inflammatory Agents/therapeutic use , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Line , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Glucosides/therapeutic use , Immunosuppressive Agents/therapeutic use , Matrix Metalloproteinases/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/drug therapy , Phenols/therapeutic use , Rats , Rats, Sprague-Dawley
7.
Neuroreport ; 30(17): 1222-1229, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31651708

ABSTRACT

Fusidic acid, a steroidal antibiotic, possesses antimicrobial, antioxidant, and anti-inflammatory properties, but the effect of fusidic acid against neurodegenerative disease-related cell death has not been studied. Here, we investigated the protective effects of fusidic acid on sodium nitroprusside (SNP)-induced toxicity in C6 glial cells. Fusidic acid (5-20 µM) prevented SNP (100 µM)-induced cell death dose dependently, and effectively attenuated SNP-induced generation of nitric oxide (NO), total reactive oxygen species (ROS), and peroxynitrite (ONOO). Fusidic acid (20 µM) pretreatment significantly suppressed SNP (100 µM)-induced apoptotic events, such as nuclear condensation and caspase-3 activation. In addition, fusidic acid effectively attenuated SNP-induced endoplasmic reticulum (ER) stress markers, such as GRP78, IRE1, ATF6, PERK, XBP1s, eIF2α, CHOP, and caspase-12. A specific adenosine monophosphate-activated protein kinase (AMPK) inhibitor, compound C (10 µM), reversed the preventive effects of fusidic acid against SNP-induced cytotoxicity, CHOP elevation, and caspase-3 activation. These results suggest that fusidic acid can protect C6 glial cells against cytotoxicity, through the regulation of AMPK pathway and apoptotic events.


Subject(s)
Apoptosis/drug effects , Fusidic Acid/pharmacology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Endoplasmic Reticulum Stress/drug effects , Nitroprusside/toxicity , Rats
8.
Genes Genomics ; 41(6): 657-665, 2019 06.
Article in English | MEDLINE | ID: mdl-30953339

ABSTRACT

BACKGROUND: The 2-(4-morpholinoanilino)-6-cyclohexylaminopurine (reversine) acts as a chemopreventive agent and induces apoptotic cell death in various cancer cells. However, the anticancer effects of reversine on osteosarcoma cells are not clearly established. OBJECTIVE: The purpose of this study was to investigate the effect of reversine on cell proliferation and induction of apoptosis in human osteosarcoma cells. METHODS: Cell viability assay, histological analysis, DAPI staining, caspase activation analysis, flow cytometric analysis and immunoblotting were carried out in MG-63 osteosarcoma cells. RESULTS: Reversine inhibited the growth of cells in a dose-dependent manner and induced nuclear condensation and fragmentation. Reversine-treated cells showed caspase-3/7 activation and increased apoptosis versus control cells. FasL, a death ligand associated with extrinsic apoptotic signaling pathways, was significantly up-regulated by reversine treatment. Moreover, the caspase-8, a part of the extrinsic apoptotic pathway, was activated by reversine treatments. Expressions of anti-apoptotic factors such as Bcl-2 and Bcl-xL, components of the mitochondria dependent intrinsic apoptosis pathway, significantly decreased following reversine treatment. The expressions of pro-apoptotic factors such as BAX, BAD and caspase-9 increased by reversine treatments. In addition, reversine activated caspase-3 and Poly (ADP-ribose) polymerase (PARP) to induce cell death. The Z-VAD-fmk significantly inhibited cell death through the suppression of caspase-3 expression in MG-63 cells treated with reversine. CONCLUSION: These results suggest that the reversine may inhibit cell proliferation and induce apoptotic cell death in MG-63 osteosarcoma cells through both the mitochondria-mediated intrinsic pathway and the death receptor-mediated extrinsic pathway, and may have potential properties for the discovery of anti-cancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Morpholines/pharmacology , Osteosarcoma/metabolism , Purines/pharmacology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Signal Transduction/drug effects
9.
Biol Pharm Bull ; 42(1): 94-102, 2019.
Article in English | MEDLINE | ID: mdl-30606992

ABSTRACT

Glutamate-mediated cytotoxicity has been implicated in the pathogenesis of neurological diseases, including Parkinson's disease, Alzheimer's disease, and stroke. In this study, we investigated the protective effects of alpha-lipoic acid (ALA), a naturally occurring thiol antioxidant, on glutamate-induced cytotoxicity in cultured C6 astroglial cells. Exposure to high-dose glutamate (10 mM) caused oxidative stress and mitochondrial dysfunction through the elevation of reactive oxygen species, depletion of glutathione, and loss of the mitochondrial membrane potential (ΔΨm). Pretreatment with ALA (200 µM), however, significantly inhibited the glutamate-induced oxidative stress and mitochondrial dysfunction. ALA pretreatment dose-dependently suppressed glutamate-induced apoptotic events including altered nuclear morphology and activation of caspase-3. In addition, ALA significantly attenuated glutamate-induced endoplasmic reticulum (ER) stress markers; namely, glucose-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), protein kinase regulated by RNA (PKR)-like ER-associated kinase (PERK), eukaryotic translation initiation factor 2 alpha (eIF2α), inositol-requiring enzyme 1 (IRE1), CCAAT/enhancer binding protein homologous protein (CHOP), and caspase-12. We confirmed that CHOP and caspase-12 are key mediators of glutamate-induced ER stress. Furthermore, exposure of the cells to a caspase-12-specific inhibitor and CHOP small interfering RNAs (siRNAs) led to restoration of the ΔΨm that was damaged by glutamate treatment. These results suggest that ALA can effectively suppress oxidative stress, mitochondrial dysfunction, and ER stress in astroglial cells.


Subject(s)
Cytoprotection/drug effects , Cytotoxins/toxicity , Glioma/metabolism , Glutamic Acid/toxicity , Oxidative Stress/drug effects , Thioctic Acid/pharmacology , Animals , Antioxidants/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cytoprotection/physiology , Dose-Response Relationship, Drug , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Oxidative Stress/physiology , Rats , Reactive Oxygen Species/metabolism
10.
Anticancer Res ; 38(6): 3477-3485, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29848700

ABSTRACT

BACKGROUND/AIM: MicroRNAs (miRNAs) are closely associated with a number of cellular processes, including cell development, differentiation, proliferation, carcinogenesis, and apoptosis. The aim of the present study was to elucidate the molecular mechanisms underlying the tumor suppressor activity of miRNA-203 (miR-203) in YD-38 human oral cancer cells. MATERIALS AND METHODS: Polymerase chain reaction analysis, MTT assay, DNA fragmentation assay, fluorescence-activated cell-sorting analysis, gene array, immunoblotting, and luciferase assay were carried out in YD-38 cells. RESULTS: miR-203 expression was significantly down-regulated in YD-38 cells compared to expression levels in normal human oral keratinocytes. miR-203 decreased the viability of YD-38 cells in a time- and dose-dependent manner. In addition, over-expression of miR-203 significantly increased not only DNA segmentation, but also the apoptotic population of YD-38 cells. These results indicate that miR-203 overexpression induces apoptosis in YD-38 cells. Target gene array analysis revealed that the expression of the polycomb complex protein gene Bmi-1, a representative oncogene, was significantly down-regulated by miR-203 in YD-38 cells. Moreover, both mRNA and protein levels of Bmi-1 were significantly reduced in YD-38 cells transfected with miR-203. These results indicate that Bmi-1 is a target gene of miR-203. A luciferase reporter assay confirmed that miR-203 suppressed Bmi-1 expression by directly targeting the 3'-untranslated region. CONCLUSION: miR-203 induces apoptosis in YD-38 cells by directly targeting Bmi-1, which suggests its possible application as an anti-cancer therapeutic.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Polycomb Repressive Complex 1/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Down-Regulation , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Polycomb Repressive Complex 1/metabolism
11.
J Med Food ; 21(6): 585-595, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29377739

ABSTRACT

Anthriscus sylvestris (L.) Hoffm. is a common perennial herb that is widely distributed in Europe, Korea, and New Zealand. The root of A. sylvestris has been used in Korean traditional medicine as an antitussive and cough remedy. However, the physiologically active function of A. sylvestris leaves is not yet known. In this study, we evaluated the anti-inflammatory effects, as well as the underlying molecular mechanisms of an aqueous extract of A. sylvestris leaves (AE-ASL) in vitro and in vivo. Our results indicated that pretreatment with AE-ASL significantly inhibited the lipopolysaccharide (LPS)-induced secretion of nitric oxide (NO) and prostaglandin E2 in RAW264.7 cells, without showing cytotoxicity. In addition, the LPS-induced mRNA and protein expression of inducible NO synthase, cyclooxygenase-2, and inflammatory mediators such as tumor necrosis factor alpha interleukin (IL)-1ß, and IL-6 was attenuated by pretreatment with AE-ASL in a dose-dependent manner. Therefore, we investigated the activation of nuclear factor (NF)-κB, a transcription factor regulating the expression of inflammation-related genes. AE-ASL inhibited the nuclear translocation of the NF-κB p65 subunit by suppressing the phosphorylation and degradation of the inhibitor of NF-κB (IκBα). Further, AE-ASL inhibited the LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs) in RAW264.7 cells. Orally administered AE-ASL (50, 100, and 200 mg/kg of body weight [BW]) suppressed the development of carrageenan-induced rat paw edema by 15%, 31%, and 40%, respectively, after 4 h. Altogether, our results suggest that AE-ASL possesses anti-inflammatory activity, based on the suppression of NF-κB and MAPK pathways in vitro and inhibition of the carrageenan-induced paw edema in vivo.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apiaceae/chemistry , Edema/drug therapy , Plant Extracts/administration & dosage , Animals , Anti-Inflammatory Agents/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dinoprostone/immunology , Edema/genetics , Edema/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Nitric Oxide/immunology , Plant Extracts/chemistry , Plant Leaves/chemistry , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley
12.
Biomed Pharmacother ; 97: 264-270, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29091874

ABSTRACT

BACKGROUND: Codium fragile (Suringar) Hariot has been used as a potential remedy in traditional medicine because of its anti-inflammatory and anti-oxidant effects. Osteoarthritis is a chronic progressive joint disease, characterized by complex mechanisms related to inflammation and degeneration of articular cartilage. In this study, we aimed to evaluate the cartilage protective effect of an aqueous extract of Codium fragile (AECF) using rat primary chondrocytes and the osteoarthritis animal model induced by destabilization of the medial meniscus (DMM). METHODS: In vitro, rat primary cultured chondrocytes were pre-treated with AECF (0.5, 1, and 2mg/mL) for 1h and then incubated with interleukin-1ß (10ng/mL) for 24h. Nitrite production was detected by the Griess reagent. Alteration of the protein levels of iNOS, MMP-13, ADAMTS-4, ADAMTS-5, mitogen-activated protein kinases (MAPKs), and nuclear factor-κB (NF-κB) was detected by western blotting. In vivo, osteoarthritis was induced by DMM of Sprague Dawley (SD) rats. The rats subjected to destabilization of the medial meniscus (DMM) surgery were orally administered with AECF (50, 100, and 200mg/kg bodyweight) or distilled water for 8w. The severity of cartilage lesions was evaluated by safranin O staining and the Osteoarthritis Research Society International (OARSI) score. RESULTS: These results demonstrated that AECF significantly inhibited nitrite production and inhibited the levels of iNOS, MMP-13, ADAMTS-4, and ADAMTS-5 in interleukin-1ß-induced rat primary cultured chondrocytes. Moreover, AECF suppressed interleukin-1ß-induced NF-κB activation in the nucleus and phosphorylation of ERK1/2 and JNK in the cytosol. In vivo, the cartilage lesions in AECF-treated osteoarthritis rats exhibited less proteoglycan loss and lower OARSI scores. CONCLUSIONS: These results suggested that AECF is a potential therapeutic agent for the alleviation of osteoarthritis progression.


Subject(s)
Chlorophyta , Chondrocytes/metabolism , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Animals , Anti-Inflammatory Agents , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/drug effects , Disease Models, Animal , Interleukin-1beta/toxicity , MAP Kinase Signaling System/drug effects , Male , NF-kappa B/antagonists & inhibitors , Osteoarthritis/chemically induced , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Water/pharmacology
13.
Anticancer Res ; 37(10): 5425-5433, 2017 10.
Article in English | MEDLINE | ID: mdl-28982852

ABSTRACT

BACKGROUND/AIM: The purpose of this study was to elucidate the molecular mechanism underlying regulation of semaphorin-6A (SEMA6A) involving microRNA-203 (miR-203) as a tumor suppressor in YD-38 human oral cancer cells. MATERIALS AND METHODS: miRNA arrays, polymerase chain reaction analyses, MTT assays, immunoblotting, and luciferase assays were carried out in YD-38 cells. RESULTS: MiRNA microarray results showed that expression of miR-203 was significantly down-regulated in YD-38 cells compared to normal human oral keratinocytes. The viability of YD-38 cells was reduced by miR-203 in time- and dose-dependent manners. Overexpression of miR-203 increased the nuclear condensation of YD-38 cells and activated the apoptotic signaling pathway by up-regulating pro-apoptotic factors, such as BCL-2-associated X protein (BAX) and BCL-2 homologous antagonist killer (BAK), and the active forms of caspase-9, caspase-3, and poly-(ADP-ribose)-polymerase (PARP). Furthermore, target gene array analyses revealed that the expression of class 6 semaphorin A (SEMA6A) was down-regulated by miR-203 in YD-38 cells. Both the mRNA and protein levels of SEMA6A were reduced in YD-38 cells transfected with miR-203. Luciferase activity assay confirmed that miR-203 directly targets the SEMA6A 3'-untranslated region to suppress gene expression. CONCLUSION: Our results indicate that miR-203 induces the apoptosis of YD-38 human oral cancer cells by directly targeting SEMA6A, suggesting its potential application in anticancer therapeutics.


Subject(s)
MicroRNAs/metabolism , Mouth Neoplasms/metabolism , Oncogenes , Semaphorins/metabolism , 3' Untranslated Regions , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Semaphorins/genetics , Signal Transduction , Time Factors , Transfection
14.
Biol Pharm Bull ; 40(8): 1275-1281, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28529240

ABSTRACT

Curcumin, a bioactive component in tumeric, has been shown to exert antioxidant, anti-inflammatory, anticarcinogenic, hepatoprotective, and neuroprotective effects, but the effects of curcumin against manganese (Mn)-mediated neurotoxicity have not been studied. This study examined the protective effects of curcumin on Mn-induced cytotoxicity in BV-2 microglial cells. Curcumin (0.1-10 µM) dose-dependently prevented Mn (250 µM)-induced cell death. Mn-induced mitochondria-related apoptotic characteristics, such as caspase-3 and -9 activation, cytochrome c release, Bax increase, and Bcl-2 decrease, were significantly suppressed by curcumin. In addition, curcumin significantly increased intracellular glutathione (GSH) and moderately potentiated superoxide dismutase (SOD), both which were diminished by Mn treatment. Curcumin pretreatment effectively suppressed Mn-induced upregulation of malondialdehyde (MDA), total reactive oxygen species (ROS). Moreover, curcumin markedly inhibited the Mn-induced mitochondrial membrane potential (MMP) loss. Furthermore, curcumin was able to induce heme oxygenase (HO)-1 expression. Curcumin-mediated inhibition of ROS, down-regulation of caspases, restoration of MMP, and recovery of cell viability were partially reversed by HO-1 inhibitor (SnPP). These results suggest the first evidence that curcumin can prevent Mn-induced microglial cell death through the induction of HO-1 and regulation of oxidative stress, mitochondrial dysfunction, and apoptotic events.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Microglia/drug effects , Mitochondria/drug effects , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Curcuma/chemistry , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Glutathione/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Malondialdehyde/metabolism , Manganese/toxicity , Membrane Potential, Mitochondrial/drug effects , Metalloporphyrins/pharmacology , Mice , Mitochondria/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Up-Regulation
15.
Neurochem Res ; 42(2): 656-666, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27900599

ABSTRACT

Melatonin, a naturally occurring neurohormone in the pineal gland, has been shown to exert antioxidant and anti-inflammatory effects. This study examined the effects of melatonin on manganese (Mn) and/or lipopolysaccharide (LPS)-induced microglial activation. Melatonin (10 µM) inhibited Mn (100 µM) and/or LPS (0.5 µg/ml)-induced phagocytotic activity of activated BV2 microglia. It also inhibited the lipid peroxidation and intracellular reduced glutathione (GSH) depletion induced by Mn and/or LPS. Melatonin effectively suppressed the upregulation of interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) at both mRNA and protein levels in Mn and/or LPS-stimulated BV2 microglia. In addition, melatonin pretreatment attenuated Mn and/or LPS-induced degradation of IκB-α, nuclear translocation of nuclear factor-κB (NF-κB) and its activation, and the expressions of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) in BV2 microglial cells. These results suggest that melatonin can effectively modulate phagocytosis and expression of proinflammatory mediators, and can prevent neuroinflammatory disorders accompanied by microglial activation.


Subject(s)
Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Manganese/toxicity , Melatonin/pharmacology , Microglia/metabolism , Phagocytosis/physiology , Animals , Cell Line , Mice , Microglia/drug effects , Phagocytosis/drug effects
16.
Neuroreport ; 27(15): 1140-6, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27513201

ABSTRACT

Dieldrin, an organochlorine pesticide still used in several developing countries, has been proposed as a risk factor for Parkinson's disease. Quercetin is one of the potent bioactive flavonoids present in numerous plants. In this study, we investigated the protective effects of quercetin on neurotoxicity induced by dieldrin in cultured dopaminergic SN4741 cells. Our initial experiments showed that quercetin (10-40 µM) dose dependently prevented dieldrin (20 µM)-induced cytotoxicity in SN4741 cells. Pretreatment for 1 h with quercetin before dieldrin application could significantly suppress dieldrin-induced apoptotic characteristics, including nuclear condensation, DNA fragmentation, and caspase-3/7 activation. Results showed that dieldrin-induced markers of endoplasmic reticulum (ER) stress response such as chaperone GRP78, heme oxygenase-1, and phosphorylation of the α subunit of eukaryotic initiation factor 2. In addition, dieldrin reduced antiapoptotic Bcl-2 expression, but significantly elevated a proapoptotic transcription factor CHOP. Furthermore, RNA interference to CHOP almost completely repressed dieldrin-induced apoptotic cell death. Interestingly, quercetin prevented the changes in dieldrin-induced ER stress markers. These results suggest that quercetin may suppress the ER stress-CHOP pathway and dieldrin-induced apoptosis in dopaminergic neurons.


Subject(s)
Apoptosis/drug effects , Dieldrin/pharmacology , Dopaminergic Neurons/drug effects , Endoplasmic Reticulum Stress/drug effects , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Animals , Caspases/metabolism , Cell Line , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/metabolism , Heme Oxygenase-1/metabolism , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Mice , Necrosis/chemically induced , Necrosis/prevention & control , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factor CHOP/metabolism
17.
J Biosci Bioeng ; 120(4): 351-8, 2015 10.
Article in English | MEDLINE | ID: mdl-25910964

ABSTRACT

The purpose of this study was to elucidate the molecular mechanisms of microRNA-203 (miR-203) as a tumor suppressor in KB human oral cancer cells. MicroRNA microarray results showed that the expression of miR-203 was significantly down-regulated in KB cells compared with normal human oral keratinocytes. The viability of KB cells was decreased by miR-203 in the time- and dose-dependent manners. In addition, over-expressed miR-203 not only increased the nuclear condensation but also significantly increased the apoptotic population of KB cells. These results indicated that the over-expression of miR-203 induced apoptosis of KB cells. Furthermore, the target gene array analyses revealed that the expression of Yes-1, a member of the Src family kinases (SFKs), was significantly down-regulated by miR-203 in KB cells. Moreover, both the mRNA and protein levels of Yes-1 were strongly reduced in KB cells transfected with miR-203. Therefore, these results indicated that Yes-1 is predicted to be a potential target gene of miR-203. Through a luciferase activity assay, miR-203 was confirmed to directly targets the Yes-1 3' untranslated region (UTR) to suppress gene expression. Therefore, our findings indicate that miR-203 induces the apoptosis of KB cells by directly targeting Yes-1, suggesting its application in anti-cancer therapeutics.


Subject(s)
Apoptosis/genetics , Down-Regulation , Genes, Tumor Suppressor , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-yes/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/therapeutic use , Mouth Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-yes/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection
18.
Neuroreport ; 26(3): 144-51, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25569790

ABSTRACT

The neurotrophin-inducible gene VGF plays an important role in the maintenance of organismal energy balance and in the mediation of hippocampal synaptic activity. The regulatory mechanism of VGF transcription is not fully understood. The neuron-restrictive silencer factor (NRSF) binds with the neuron-restrictive silencer element (NRSE), thereby suppressing the transcription of NRSE-containing genes. In this study, we show that the NRSE sequence of the VGF gene critically regulates the repression of VGF expression in NMB cells. Sequence analysis also establishes the presence of two putative NRSEs (NRSE-1 and NRSE-2) in the promoter region of the VGF gene. In reporter gene experiments, a more than eight-fold increase in the promoter activity was observed when both NRSE-1 and NRSE-2 were deleted. Deletion of NRSE-2 alone did not affect the promoter activity, thus indicating that NRSE-1 could be solely responsible for the repression of VGF gene expression. Mutations in the NRSE-1 sequence increased promoter activity. However, no change in activity was observed when NRSE-1 was coexpressed with dominant-negative NRSF, thereby suggesting that endogenous NRSF interacts with NRSE-1. Binding of NRSF to NRSE in a sequence-specific manner was confirmed with chromatin immunoprecipitation assays, respectively. Furthermore, the overexpressed NRSF in PC12 cells significantly suppressed the VGF gene expression by interacting with the NRSE located in the VGF promoter region. Our results indicate that NRSF plays an important role as a repressor of VGF gene regulation in NMB cells through a mechanism that is dependent on VGF-NRSE.


Subject(s)
Gene Expression Regulation/physiology , Nerve Growth Factors/metabolism , Neuropeptides/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Animals , Cell Line, Tumor , Humans , Mutation , PC12 Cells , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats
19.
Biochem Biophys Res Commun ; 446(4): 894-900, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24642258

ABSTRACT

MicroRNAs (miRNAs) regulate cell differentiation by inhibiting mRNA translation or by inducing its degradation. However, the role of miRNAs in odontogenic differentiation is largely unknown. In this present study, we observed that the expression of miR-663 increased significantly during differentiation of MDPC-23 cells to odontoblasts. Furthermore, up-regulation of miR-663 expression promoted odontogenic differentiation and accelerated mineralization without proliferation in MDPC-23 cells. In addition, target gene prediction for miR-663 revealed that the mRNA of the adenomatous polyposis coli (APC) gene, which is associated with the Wnt/ß-catenin signaling pathway, has a miR-663 binding site in its 3'-untranslated region (3'UTR). Furthermore, APC expressional was suppressed significantly by miR-663, and this down-regulation of APC expression triggered activation of Wnt/ß-catenin signaling through accumulation of ß-catenin in the nucleus. Taken together, these findings suggest that miR-663 promotes differentiation of MDPC-23 cells to odontoblasts by targeting APC-mediated activation of Wnt/ß-catenin signaling. Therefore, miR-663 can be considered a critical regulator of odontoblast differentiation and can be utilized for developing miRNA-based therapeutic agents.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Down-Regulation , Genes, APC , MicroRNAs/metabolism , Odontogenesis , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Line , Mice , Wnt Proteins/metabolism , beta Catenin/metabolism
20.
Oncol Rep ; 31(5): 2286-92, 2014 May.
Article in English | MEDLINE | ID: mdl-24604218

ABSTRACT

The aim of the present study was to investigate and compare the effects of diferuloylmethane (curcumin) and diphenyldifluoroketone (EF-24) on cell growth and apoptosis induction in human osteogenic sarcoma cells. This was examined by MTT assay, nuclear DAPI staining, caspase-activation assay, flow cytometry analysis and immunoblotting in Saos2 human osteogenic sarcoma cells. Curcumin and EF-24 inhibited the growth of Saos2 cells in a dose-dependent manner. The apparent potency of EF-24 was more than 3-fold higher that of curcumin. Treatment with curcumin or EF-24 resulted in nuclear condensation and fragmentation in the cells. The caspase-3/-7 activities were detected in living cells treated with curcumin or EF-24. Flow cytometry showed that the rate of apoptosis was increased by curcumin and EF-24 compared to the control. Curcumin and EF-24 promoted the proteolytic cleavages of procaspase-3/-7/-8/-9 with increases in the amount of cleaved caspase-3/-7/-8/-9. The curcumin- or EF-24-induced apoptosis in the Saos2 cells was mediated by the expression of Fas and activation of caspase-8, caspase-3 and poly(ADP-ribose) polymerase. Immunoblotting revealed the Bid and Bcl-2 proteins to be downregulated, and truncated-Bid, Bax and p53 proteins to be upregulated by curcumin and EF-24. Curcumin and EF-24 increased the Bax/Bcl-2 ratio significantly. These results suggest that the curcumin and EF-24 inhibit cell proliferation and induce apoptotic cell death in Saos2 human osteogenic sarcoma cells via both the mitochondria-mediated intrinsic pathway and the death receptor-mediated extrinsic pathway, and may have potential properties for anti-osteosarcoma drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Bone Neoplasms/drug therapy , Curcumin/pharmacology , Osteosarcoma/drug therapy , Piperidones/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Caspase 3/biosynthesis , Caspase 3/metabolism , Caspase 7/biosynthesis , Caspase 7/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein/biosynthesis , Humans , Poly(ADP-ribose) Polymerases/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , bcl-2-Associated X Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...