Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Cell Biochem ; 120(8): 13861-13866, 2019 08.
Article in English | MEDLINE | ID: mdl-30989711

ABSTRACT

Periodontitis is characterized by increased levels of proinflammatory factors, such as interleukin-17 (IL-17) and IL-35. In this study, the expression of microRNA-146a (miRNA-146a), IL-17, and IL-35 in the plasma of patients with periodontitis and healthy controls were detected by quantitative reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. miRNA-146a mimic was transfected into periodontal ligament stem cells (PDLSCs) isolated from periodontitis-affected teeth and healthy teeth. Cell proliferation and expression of IL-17 and IL-35 were detected by cell counting kit-8 assay and Western blot analysis, respectively. It was observed that miRNA-146a was downregulated but IL-17 and IL-35 were upregulated in the plasma of patients with periodontitis than in healthy controls. miRNA-146a was inversely correlated with IL-17 and IL-35 in patients with periodontitis. miRNA-146a overexpression inhibited proliferation of PDLSCs derived from both periodontitis-affected teeth and healthy teeth. miRNA-146a overexpression led to downregulated IL-17 and IL-35 expression in PDLSCs isolated from periodontitis-affected teeth. We, therefore, conclude that miRNA-146a may improve periodontitis by downregulating IL-17 and IL-35 expression and inhibiting proliferation of human PDLSCs.


Subject(s)
Down-Regulation/genetics , Interleukin-17/genetics , Interleukins/genetics , MicroRNAs/metabolism , Periodontal Ligament/cytology , Stem Cells/cytology , Cell Proliferation , Humans , Interleukin-17/blood , Interleukin-17/metabolism , Interleukins/blood , Interleukins/metabolism , MicroRNAs/blood , MicroRNAs/genetics , Periodontitis/blood , Periodontitis/genetics , Tooth/pathology
2.
Mol Cell Biochem ; 411(1-2): 83-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26369531

ABSTRACT

Human periodontal ligament fibroblasts (hPLFs) are exposed to oxidative stress during periodontal inflammation and dental treatments. It is hypothesized that hydrogen peroxide (H2O2)-mediated oxidative stress decreases survival and osteogenic differentiation of hPLFs, whereas these decreases are prevented by activation of the Wnt pathway. However, there has been a lack of reports that define the exact roles of canonical Wnt/ß-catenin signaling in H2O2-exposed hPLFs. Treatment with H2O2 reduced viability and proliferation in hPLFs in a dose- and time-dependent manner and led to mitochondria-mediated apoptosis. Pretreatment with lithium chloride (LiCl) or Wnt1 inhibited the oxidative damage that occurred in H2O2-exposed hPLFs. However, knockout of ß-catenin or treatment with DKK1 facilitated the H2O2-induced decreases in viability, mitochondrial membrane potential, and Bcl-2 induction. Osteoblastic differentiation of hPLFs was also inhibited by combined treatment with 100 µM H2O2, as evidenced by the decreases in alkaline phosphatase (ALP) activity and mineralization. H2O2-mediated inhibition of osteoblast differentiation in hPLFs was significantly attenuated in the presence of 500 ng/ml Wnt1 or 20 mM LiCl. In particular, H2O2 stimulated the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) at protein and mRNA levels in hPLFs, whereas the induction was almost completely suppressed in the presence of Wnt1 or LiCl. Furthermore, siRNA-mediated silencing of Nrf2 blocked H2O2-induced decreases in ALP activity and mineralization of hPLFs with the concomitant restoration of runt-related transcription factor 2 and osteocalcin mRNA expression and ALP activity. Collectively, these results suggest that activation of the Wnt/ß-catenin pathway improves proliferation and mineralization in H2O2-exposed hPLFs by downregulating Nrf2.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Hydrogen Peroxide/pharmacology , Periodontal Ligament/drug effects , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Adult , Alkaline Phosphatase/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Male , Periodontal Ligament/cytology , Periodontal Ligament/enzymology , Young Adult , beta Catenin/genetics
3.
Mol Cell Biochem ; 382(1-2): 37-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24026476

ABSTRACT

Fibroblast growth factor-7 (FGF7) is known to regulate proliferation and differentiation of cells; however, little information is available on how FGF7 affects the differentiation of embryonic stem cells (ESCs). We examined the effects of FGF7 on proliferation and osteogenic differentiation of mouse ESCs. Exogenous FGF7 addition did not change the proliferation rate of mouse ESCs. In contrast, the addition of FGF7 facilitated the dexamethasone, ascorbic acid, and ß-glycerophosphate (DAG)-induced increases in bone-like nodule formation and calcium accumulation. FGF7 also augmented mRNA expression of runt-related transcription factor-2 (Runx2), osterix, bone sialoprotein (BSP), and osteocalcin (OC) in the presence of DAG. FGF7-mediated increases in the mineralization and bone-specific gene expression were almost completely attenuated by pretreating with anti-FGF7 antibody. FGF7 treatment accelerated the DAG-induced activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in the cells. A pharmacological inhibitor specific to ERK, but not to JNK or p38 kinase, dramatically suppressed FGF7-mediated mineralization and accumulation of collagen and OC in the presence of DAG. This suppression was accompanied by the reduction in Runx2, osterix, BSP, and OC mRNA levels, which were increased by FGF7 in the presence of DAG. Collectively, our results suggest that FGF7 stimulates osteogenic differentiation, but not proliferation, in ESCs, by activating ERK/Runx2 signaling.


Subject(s)
Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 7/pharmacology , Osteogenesis/drug effects , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Gene Expression Regulation/drug effects , Glycerophosphates/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Organ Specificity/drug effects , Organ Specificity/genetics , Osteogenesis/genetics , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Dent Mater J ; 42(4): 610-616, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37438118

ABSTRACT

Recently, the use of orthodontic mini-screws as an anchorage for orthodontic treatment is increasing, and the degree of osseointegration of the mini-screws affects the performance of orthodontic treatment. This study aimed to evaluate the biocompatibility and osseointegration of Titanium 6Aluminum 4Vanadium (Ti-6Al-4V) alloy orthodontic mini-screws with an ibandronate-loaded TiO2 nanotube (TNT) layer. The TNT layer was formed on the surface of the Ti-6Al-4V alloy orthodontic mini-screws and loaded with ibandronate. The TNT formed by anodic oxidation formed a completely self-organized and compact structure and was stably released for 7 days after loading with ibandronate. Mini-screws loaded with ibandronate were implanted into both tibias of rats, confirming rapid initial bone regeneration. We demonstrate that the release of stable ibandronate from the TNT layer of Ti-6Al-4V alloy orthodontic mini-screws can effectively improve biocompatibility and osseointegration.


Subject(s)
Dental Implants , Nanotubes , Rats , Animals , Titanium/chemistry , Osseointegration , Ibandronic Acid , Alloys , Bone Screws , Surface Properties
5.
J Cell Biochem ; 113(4): 1426-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22173791

ABSTRACT

Numerous studies have shown that hydrogen peroxide (H(2)O(2)) inhibits proliferation and osteoblastic differentiation in bone-like cells. Human periodontal ligament fibroblasts (PLF) are capable of differentiating into osteoblasts and are exposed to oxidative stress during periodontal inflammation. However, the cellular responses of PLF to H(2)O(2) have not been identified. In this study, we examined how H(2)O(2) affects the viability and proliferation of PLF by exposing the cells to glucose oxidase (GO) or direct addition of H(2)O(2). We also explored the effects of GO on the osteoblastic differentiation of PLF and the mechanisms involved. The viability and proliferation in PLF were increased with the addition of 10 mU/ml GO but not by volumes greater than 15 mU/ml or by H(2)O(2) itself. GO-stimulated DNA synthesis was correlated with the increase in cyclin E protein levels in the cells. Osteoblastic differentiation of PLF was also augmented by combined treatment with GO, as evidenced by the increases in alkaline phosphatase activity, mineralization, collagen synthesis, and osteocalcin content in the cells. The inductions of runt-related transcription factor 2 and osterix mRNA and proteins were further increased in PLF incubated in combination with GO compared to those in untreated cells. These results demonstrate that the continuous presence of H(2)O(2) stimulates the proliferation of PLF and augments their potential to differentiate into osteoblasts through the up-regulation of bone-specific transcription factors. Collectively, we suggest that H(2)O(2) may elicit the functions of PLF in maintaining the dimensions of the periodontal ligament and in mediating a balanced metabolism in alveolar bone.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Hydrogen Peroxide/pharmacology , Osteoblasts/drug effects , Periodontal Ligament/drug effects , Adult , Base Sequence , Blotting, Western , Collagen/metabolism , Culture Media , DNA Primers , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Male , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Real-Time Polymerase Chain Reaction
6.
Toxicol Appl Pharmacol ; 259(3): 329-37, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22285274

ABSTRACT

Sodium fluoride (NaF) is used as a source of fluoride ions in diverse applications. Fluoride salt is an effective prophylactic for dental caries and is an essential element required for bone health. However, fluoride is known to cause cytotoxicity in a concentration-dependent manner. Further, no information is available on the effects of NaF on mouse embryonic stem cells (mESCs). We investigated the mode of cell death induced by NaF and the mechanisms involved. NaF treatment greater than 1mM reduced viability and DNA synthesis in mESCs and induced cell cycle arrest in the G(2)/M phase. The addition of NaF induced cell death mainly by apoptosis rather than necrosis. Catalase (CAT) treatment significantly inhibited the NaF-mediated cell death and also suppressed the NaF-mediated increase in phospho-c-Jun N-terminal kinase (p-JNK) levels. Pre-treatment with SP600125 or z-VAD-fmk significantly attenuated the NaF-mediated reduction in cell viability. In contrast, intracellular free calcium chelator, but not of sodium or calcium ion channel blockers, facilitated NaF-induced toxicity in the cells. A JNK specific inhibitor (SP600125) prevented the NaF-induced increase in growth arrest and the DNA damage-inducible protein 45α. Further, NaF-mediated loss of mitochondrial membrane potential was apparently inhibited by pifithrin-α or CAT inhibitor. These findings suggest that NaF affects viability of mESCs in a concentration-dependent manner, where more than 1mM NaF causes apoptosis through hydroxyl radical-dependent and caspase- and JNK-mediated pathways.


Subject(s)
Apoptosis/drug effects , Cariostatic Agents/toxicity , Embryonic Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Sodium Fluoride/toxicity , Animals , Cariostatic Agents/administration & dosage , Catalase/metabolism , Cell Survival/drug effects , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Embryonic Stem Cells/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Hydroxyl Radical/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Sodium Fluoride/administration & dosage
7.
Mater Sci Eng C Mater Biol Appl ; 135: 112673, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35581065

ABSTRACT

Scaffolds combined with bioactive agents can enhance bone regeneration at therapeutic sites. We explore whether combined supplementation with coumaric acid and recombinant human-cartilage oligomeric matrix protein-angiopoietin 1 (rhCOMP-Ang1) is an ideal approach for bone tissue engineering. We developed coumaric acid-conjugated absorbable collagen scaffold (CA-ACS) and investigated whether implanting CA-ACS in combination with rhCOMP-Ang1 facilitates ACS- or CA-ACS-mediated bone formation using a rat model of critically sized mandible defects. We examined the mechanisms by which coumaric acid and rhCOMP-Ang1 regulate behaviors of human periodontal ligament fibroblasts (hPLFs). The CA-ACS exhibits greater anti-degradation and mechanical strength properties than does ACS alone. Implanting CA-ACS loaded with rhCOMP-Ang1 greatly enhances bone regeneration at the defect via the activation of angiogenic, osteogenic, and anti-osteoclastic responses compared with other rat groups implanted with an ACS alone or CA-ACS. Treatment with both rhCOMP-Ang1 and coumaric acid increases proliferation, mineralization, and migration of cultured hPLFs via activation of the Ang1/Tie2 signaling axis at a greater rate than treatment with either of them alone. Collectively, this study demonstrates that CA-ACS impregnated with rhCOMP-Ang1 enhances bone regeneration at therapeutic sites, and this enhancement is associated with a synergistic interaction between rhCOMP-Ang1-mediated angiogenesis and coumaric acid-related antioxidant responses.


Subject(s)
Angiopoietin-1 , Antioxidants , Angiopoietin-1/metabolism , Angiopoietin-1/pharmacology , Animals , Antioxidants/pharmacology , Cartilage Oligomeric Matrix Protein , Collagen/pharmacology , Coumaric Acids , Mandible , Rats
8.
Prog Orthod ; 23(1): 11, 2022 Apr 04.
Article in English | MEDLINE | ID: mdl-35368222

ABSTRACT

BACKGROUND: Mini-screws are widely used as temporary anchorages in orthodontic treatment, but have the disadvantage of showing a high failure rate of about 10%. Therefore, orthodontic mini-screws should have high biocompatibility and retention. Previous studies have demonstrated that the retention of mini-screws can be improved by imparting bioactivity to the surface. The method for imparting bioactivity proposed in this paper is to sequentially perform anodization, periodic pre-calcification, and heat treatments with a Ti-6Al-4V ELI alloy mini-screw. MATERIALS AND METHODS: A TiO2 nanotube-structured layer was formed on the surface of the Ti-6Al-4V ELI alloy mini-screw through anodization in which a voltage of 20 V was applied to a glycerol solution containing 20 wt% H2O and 1.4 wt% NH4F for 60 min. Fine granular calcium phosphate precipitates of HA and octacalcium phosphate were generated as clusters on the surface through the cyclic pre-calcification and heat treatments. The cyclic pre-calcification treatment is a process of immersion in a 0.05 M NaH2PO4 solution and a saturated Ca(OH)2 solution at 90 °C for 1 min each. RESULTS: It was confirmed that the densely structured protrusions were precipitated, and Ca and P concentrations, which bind and concentrate endogenous bone morphogenetic proteins, increased on the surface after simulated body fluid (SBF) immersion test. In addition, the removal torque of the mini-screw fixed into rabbit tibias for 4 weeks was measured to be 8.70 ± 2.60 N cm. CONCLUSIONS: A noteworthy point in this paper is that the Ca and P concentrations, which provide a scaffold suitable for endogenous bone formation, further increased over time after SBF immersion of the APH group specimens. The other point is that our mini-screws have a significantly higher removal torque compared to untreated mini-screws. These results represent that the mini-screw proposed in this paper can be used as a mini-screw for orthodontics.


Subject(s)
Hot Temperature , Osseointegration , Alloys , Animals , Biocompatible Materials , Bone Screws , Humans , Rabbits , Titanium
9.
Mol Cell Biochem ; 335(1-2): 263-72, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19798549

ABSTRACT

The precise mechanism by which Rho kinase translates the mechanical signals into OPN up-regulation in force-exposed fibroblasts has not been elucidated. Human periodontal ligament fibroblasts (hPLFs) were exposed to mechanical force by centrifuging the culture plates at a magnitude of 50 g/cm(2) for 60 min. At various times of the force application, they were processed for analyzing cell viability, trypan blue exclusion, and OPN expression at protein and RNA levels. Cellular mechanism(s) of the force-induced OPN up-regulation was also examined using various kinase inhibitors or antisense oligonucleotides specific to mechanosensitive factors. Centrifugal force up-regulated OPN expression and induced a rapid and transient increase in the phosphorylation of focal adhesion kinase (FAK), extracellular signal-regulated kinase (ERK), and Elk1. Pharmacological blockade of RhoA/Rho-associated coiled coil-containing kinase (ROCK) signaling markedly reduced force-induced FAK and ERK1/2 phosphorylation. Transfecting hPLFs with FAK antisense oligonucleotide diminished ERK1/2 activation and force-induced OPN expression. Further, ERK inhibitor inhibited significantly OPN expression, Elk1 phosphorylation, and activator protein-1 (AP-1)-DNA binding activation, but not FAK phosphorylation, in the force-applied cells. These results demonstrate that FAK signaling plays critical roles in force-induced OPN expression in hPLFs through interaction with Rho/ROCK as upstream effectors and ERK-Elk1/ERK-c-Fos as downstream effectors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Osteopontin/genetics , Periodontal Ligament/metabolism , rhoA GTP-Binding Protein/metabolism , Adult , Fibroblasts/cytology , Humans , Male , Osteopontin/metabolism , Periodontal Ligament/cytology , RNA, Messenger/metabolism , Stress, Mechanical , Transcription Factor AP-1/metabolism , Transfection , rho-Associated Kinases/metabolism
10.
Korean J Orthod ; 50(3): 206-215, 2020 May 25.
Article in English | MEDLINE | ID: mdl-32475848

ABSTRACT

Osteochondroma is a common benign tumor of bones, but it is rare in the mandibular condyle. With its outgrowth it manifests clinically as deviation of the mandible limitation of mouth opening, and facial asymmetry. After the tumor is diagnosed on the basis of clinical symptoms and radiographic examination including cone-beam computed tomography (CBCT) analysis, an appropriate surgery and treatment plan should be formulated. Herein, we present the case of a 44-year-old female patient who visited our dental hospital because her chin point had been deviating to the left side slowly but progressively over the last 3 years and she had difficulty masticating. Based on CBCT, she was diagnosed with skeletal Class III malocclusion accompanied by osteochondroma of the right mandibular condyle. Maxillary occlusal cant with the right side down was observed, but it was confirmed to be an extrusion of the molars associated with dental compensation. Therefore, after intrusion of the right molars with the use of temporary anchorage devices, sagittal split ramus osteotomy was used to remove the tumor and perform orthognathic surgery simultaneously. During 6 months after the surgery, continuous bone resorption and remodeling were observed in the condyle of the affected side, which led to a change in occlusion. During the postoperative orthodontic treatment, intrusive force and buccal torque were applied to the molars on the affected side, and a proper buccal overjet was created. After 18 months, CBCT revealed that the rate of bone absorption was continuously reduced, bone corticalization appeared, and good occlusion and a satisfying facial profile were achieved.

11.
Korean J Orthod ; 50(2): 136-144, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32257939

ABSTRACT

Hemifacial microsomia (HFM) patients may experience emotional withdrawal during their growth period due to their abnormal facial appearance. Distraction osteogenesis at an early age to improve their appearance can encourage these patients. Some abnormalities of the affected side can be overcome by distraction osteogenesis at an early age. However, differences in the growth rate between the affected and unaffected sides during the rest of the growth period are inevitable due to the characteristics of HFM. Therefore, re-evaluation should be performed after completion of growth in order to achieve stable occlusion through either orthognathic surgery or camouflage orthodontic treatment. An eight-year-old patient visited the clinic exhibiting features of HFM with slight mandibular involvement. He received phase I treatment with distraction osteogenesis and a functional appliance. Distraction osteogenesis was performed at the right ramus, which resulted in an open bite at the right posterior dentition. After distraction osteogenesis, a functional appliance and partial fixed appliance were used to achieve extrusion of the affected posterior dentition and settlement of the occlusion adjustment on the unaffected posterior dentition. The patient visited the clinic regularly for follow-up assessments, and at the age of 20 years, he showed facial asymmetry of the mandible, which had deviated to the right side. He received orthodontic treatment to improve the occlusion of his posterior dentition after the growth period. Without orthognathic surgery, stable occlusion and a satisfactory facial appearance were obtained through camouflage orthodontic treatment.

12.
J Cell Biochem ; 106(6): 1010-9, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19206164

ABSTRACT

Periodontal ligament and gingival fibroblasts play important roles in bone remodeling. Periodontal ligament fibroblasts stimulate bone remodeling while gingival fibroblasts protect abnormal bone resorption. However, few studies had examined the differences in stimulation of osteoclast formation between the two fibroblast populations. The precise effect of mechanical forces on osteoclastogenesis of these populations is also unknown. This study revealed that more osteoclast-like cells were induced in the co-cultures of bone marrow cells with periodontal ligament than gingival fibroblasts, and this was considerably increased when anti-osteoprotegerin (OPG) antibody was added to the co-cultures. mRNA levels of receptor activator of nuclear factor-kappaB ligand (RANKL) were increased in both populations when they were cultured with dexamethasone and vitamin D(3). Centrifugal forces inhibited osteoclastogenesis of both populations, and this was likely related to the force-induced OPG up-regulation. Inhibition of extracellular signal-regulated kinase (ERK) signaling by a pharmacological inhibitor (10 microM PD98059) or by siERK transfection suppressed the force-induced OPG up-regulation along with the augmentation of osteoclast-like cells that were decreased by the force. These results suggest that periodontal ligament fibroblasts are naturally better at osteoclast induction than gingival fibroblasts, and that centrifugal force inhibited osteoclastogenesis of the periodontal fibroblasts through OPG production and ERK activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/physiology , MAP Kinase Signaling System/physiology , Osteoclasts/metabolism , Osteoprotegerin/metabolism , Periodontal Ligament/cytology , Stress, Mechanical , Adult , Bone Marrow Cells/cytology , Bone Remodeling/physiology , Bone Resorption/metabolism , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Fibroblasts/cytology , Gingiva/cytology , Humans , Male , Osteoclasts/cytology , Osteoprotegerin/genetics , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Young Adult
13.
Mol Cell Biochem ; 320(1-2): 45-52, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18682895

ABSTRACT

In addition to periodontal ligament, the gingival plays an important role in alveolar bone remodeling induced by physiological and mechanical stimuli. However, there are few reports showing the cellular responses of human gingival fibroblasts (HGF) to a mechanical force. This study examined the effects of centrifugal force on the proliferation of the bone tissue components, such as type I collagen (COL I), osteopontin (OPN), and osteonectin (ONN) in the HGF. The roles of extracellular signal-regulated kinase (ERK), c-Jun-N-terminal kinase (JNK), and p-38 kinase were also investigated. Centrifugal force induced cell cycle arrest in the G(1) phase without any cytotoxic effects and increased the levels of COL I and OPN expression in the cells but had no effect on ONN. The force-induced up-regulation of COL I was found to be mediated by both the ERK-c-Fos-COL I and JNK-c-Jun-COL I pathways, while that of OPN was mediated only by the ERK-mediated pathway. Our present findings suggest that centrifugal force up-regulates COL I and OPN expression in HGF, where both ERK and JNK play indispensable roles.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/metabolism , Gingiva/cytology , Mitogen-Activated Protein Kinases/metabolism , Osteopontin/metabolism , Stress, Mechanical , Adult , Cell Cycle/physiology , Cells, Cultured , Collagen Type I/genetics , Fibroblasts/cytology , Humans , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinases/genetics , Osteopontin/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation , Young Adult
14.
Eur J Pharmacol ; 579(1-3): 26-33, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-17988664

ABSTRACT

The bioflavonoid, quercetin, is believed to inhibit bone loss by regulating many systemic and local factors including hormones and cytokines. However, our previous findings revealed that quercetin did not inhibit but facilitate the tumor necrosis factor (TNF)-alpha-mediated apoptosis of MC3T3-E1 osteoblastic cells. Therefore, this study was carried out to examine the cellular mechanisms for how quercetin accelerates TNF-alpha-mediated apoptosis, and to determine whether the accelerating effect of quercetin is a general effect in osteoblastic cells. Quercetin promoted the TNF-alpha-induced apoptosis of MC3T3-E1 cells through both the mitochondrial-mediated and caspase-dependent mechanisms. Quercetin also augmented the TNF-alpha-mediated apoptosis by activating c-Jun N-terminal kinase (JNK) with the attendant activation of activator protein-1, where the nuclear translocation of c-Jun protein appeared to be a critical event responsible for the accelerating action of quercetin. However, TNF-alpha-mediated apoptosis and its acceleration by quercetin were not observed in primary osteoblasts. These results strongly suggest that quercetin accelerates TNF-alpha-mediated apoptosis of osteoblasts through caspase-dependent and JNK-mediated pathways, and that the cellular responses of osteoblasts to TNF-alpha and/or quercetin might differ according to their origins.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Quercetin/pharmacology , Tumor Necrosis Factor-alpha/drug effects , 3T3 Cells , Animals , Caspases/drug effects , Caspases/metabolism , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Dent Mater ; 24(8): 1036-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18164755

ABSTRACT

OBJECTIVE: This study examined the effects of blue light exposure on the proliferation and cytotoxicity of human gingival fibroblasts (HGF). Cellular mechanism by which blue light causes cytotoxic effects was also investigated. METHODS: HGF were exposed to the plasma-arc generated blue light with various energy densities ranging from 2 to 48J/cm(2). After light exposure of the cells, they were processed for analyzing tritium incorporation, succinate dehydrogenase (SDH) activity, trypan blue exclusion, and DNA fragmentation. In addition, possible mechanism of the light-mediated cytotoxicity was investigated through flow cytometric and Western blot analyses. RESULTS: Blue light exposure significantly inhibited proliferation and SDH activity of HGF in a dose-dependent manner; exposure more than 12J/cm(2) had a toxic effect on the cells. The blue light-induced cytotoxicity of the cells resulted from apoptosis, as proven by the migration of many cells to the sub-G(1) phase of cell cycle and the appearance of DNA ladders. Additional experiments revealed that blue light induces apoptosis of HGF through mitochondrial stress and poly (ADP ribose) polymerase cleavage. SIGNIFICANCE: This study suggests that plasma-arc generated blue light exerts some harm to cells, particularly damaging effect to DNA, and thus a long curing time more than recommended can cause biological damage on the oral tissue.


Subject(s)
Apoptosis/radiation effects , Fibroblasts/radiation effects , Gingiva/radiation effects , Lighting/instrumentation , Blotting, Western , Cell Proliferation/radiation effects , Cells, Cultured , Coloring Agents , DNA Fragmentation/radiation effects , Dose-Response Relationship, Radiation , Electrophoresis, Agar Gel , Fibroblasts/cytology , Flow Cytometry , Gingiva/cytology , Humans , Light , Materials Testing , Mitochondria/radiation effects , Poly(ADP-ribose) Polymerases/radiation effects , Radiation Dosage , Radiopharmaceuticals , Succinate Dehydrogenase/analysis , Thymidine , Tritium , Trypan Blue
16.
Biochim Biophys Acta ; 1726(3): 309-16, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16213662

ABSTRACT

Many studies have suggested that dietary flavonoids are anticancer agents that induce the apoptosis of cancer cells. However, the effects of flavonoids on the induction of apoptosis in osteosarcoma cells are unclear. Previously, a flavonoid fraction, consisting mainly of protocatechuic acid, fustin, fisetin, sulfuretin, and butein, herein named RCMF (the RVS chloroform-methanol fraction), was prepared from a crude acetone extract of Rhus verniciflua Stokes (RVS). This study evaluated the effects of RCMF on the proliferation and apoptosis using human osteosarcoma (HOS) cells. The mechanism of growth inhibition of the HOS cells by the flavonoid fraction, RCMF, was also assessed. The results demonstrated that RCMF exhibited sensitive growth inhibition and induced apoptosis in HOS cells. PARP cleavage was closely associated with the RCMF-induced apoptosis of the HOS cells. Furthermore, the activation of caspase 8 and Bax, the inhibition of Bcl-2 expression, and the release of cytochrome c are believed to be involved in the RCMF-mediated apoptosis. Collectively, these findings suggest that RCMF is an agent which may be capable of inducing sensitive growth inhibition and apoptosis in HOS cells.


Subject(s)
Anticarcinogenic Agents/toxicity , Apoptosis , Flavonoids/toxicity , Anticarcinogenic Agents/isolation & purification , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Flavonoids/isolation & purification , Humans , Mitochondria/metabolism , Osteosarcoma/pathology , Plant Extracts/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Rhus/chemistry
17.
Eur J Pharmacol ; 529(1-3): 24-32, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16316652

ABSTRACT

The bioflavonoid quercetin is believed to play an important role in preventing bone loss by affecting osteoclastogenesis and regulating many systemic and local factors including hormones and cytokines. This study examined how quercetin acts on tumor necrosis factor-alpha (TNF-alpha)-mediated growth inhibition and apoptosis in MC3T3-E1 osteoblastic cells. Tritium uptake assay showed that a quercetin treatment accelerated TNF-alpha-induced inhibition of DNA synthesis in the cells in a dose-dependent manner. Both the 3-(4,5-dimethylthiazol-2yl-)-2,5-diphenyl tetrazolium bromide and trypan blue staining assays also showed the quercetin-mediated facilitation of TNF-alpha-induced cytotoxicity in the cells. Apoptosis assays revealed an accelerating effect of quercetin on TNF-alpha-induced apoptosis in MC3T3-E1 cells. In addition, Fas activation and poly (ADP ribose) polymerase cleavage are thought to be closely associated with the TNF-alpha-induced apoptosis and its acceleration by the quercetin treatment in the cells. Collectively, this study showed that quercetin accelerates the TNF-alpha-induced growth inhibition and apoptosis in MC3T3-E1 osteoblastic cells.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Osteoblasts/drug effects , Quercetin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Membrane Potentials/drug effects , Mice , Mitochondria/drug effects , Mitochondria/physiology , Osteoblasts/cytology , Osteoblasts/metabolism , Time Factors
18.
J Biochem Mol Biol ; 38(6): 755-62, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16336792

ABSTRACT

Epstein-Barr virus (EBV) infects more than 90 % of the world's population and has a potential oncogenic nature. A histone deacetylase (HDAC) inhibitor, trichostatin A (TSA), has shown potential ability in cancer chemoprevention and treatment, but its effect on EBV-infected Akata cells has not been examined. This study investigated the effect of TSA on the proliferation and apoptosis of the cells. TSA inhibited cell growth and induced cytotoxicity in the EBV-infected Akata cells. TSA treatment sensitively induced apoptosis in the cell, which was demonstrated by the increased number of positively stained cells in the TUNEL assay, the migration of many cells to the sub-G0/G1 phase in flow cytometric analysis, and the ladder formation of genomic DNA. Western blot analysis showed that caspase-dependent pathways are involved in the TSA-induced apoptosis of EBV-infected Akata cells. Overall, this study shows that EBV-infected B lymphomas are quite sensitive to TSA-provoked apoptosis.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Herpesvirus 4, Human/metabolism , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Caspase 8/metabolism , Cell Cycle , Cell Line, Tumor , DNA/metabolism , DNA Fragmentation , Enzyme Activation , Humans , In Situ Nick-End Labeling , Jurkat Cells , Propidium/pharmacology
19.
Toxicol Lett ; 155(1): 115-25, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15585366

ABSTRACT

Considerable attention is being concentrated on dietary flavonoids in developing novel cancer-preventive approaches due to their potential ability to induce selective apoptosis of cancer cells. In this study, we prepared a flavonoid-containing fraction from a crude acetone extract of Rhus verniciflua Stokes (RVS), traditionally used as a food additive and as an herbal medicine, and named RVS chloroform-methanol fraction (RCMF). We evaluated the effects of RCMF on proliferation and apoptosis using mouse embryonic primary hepatic cells (MPHC), embryonic normal hepatic cell line (BNL CL.2), and its SV40-mediated transformed cell line (BNL SV A.8). We also investigated the effects of RCMF on the antioxidant defense system in those cells. This study demonstrated that RCMF exhibited a selective growth inhibition and apoptosis induction on transformed cells. BNL SV A.8 cells were more sensitive to RCMF-mediated cytotoxicity than were MPHC or BNL CL.2. RCMF-mediated reduction of MnSOD activity and glutathione (GSH) content in BNL SV A.8 cells is thought to be associated with RCMF-induced apoptosis. Our findings suggest that RCMF is an agent which may be capable of inducing growth inhibition and apoptosis of hepatic tumor cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Liver/cytology , Rhus/chemistry , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , DNA/biosynthesis , DNA Fragmentation/drug effects , Flavonoids/chemistry , Flavonoids/isolation & purification , Hepatocytes/drug effects , In Situ Nick-End Labeling , Liver/drug effects , Mice , Mice, Inbred BALB C , Structure-Activity Relationship
20.
Mol Cells ; 18(2): 163-70, 2004 Oct 31.
Article in English | MEDLINE | ID: mdl-15528991

ABSTRACT

Excessive breakdown of extracellular matrix by metalloproteinases (MMPs) occurs in many pathological conditions. Consequently, methods for inhibiting MMP activity have therapeutic potential. In this study, we investigated the effect of G-120, a 120 kDa glycoprotein purified from the Oriental herbal plant, Ulmus davidiana Nakai (UDN), on the activity and production of several MMPs by evaluating its growth inhibitory effect on NIH 3T3 cells. Tritium uptake assays showed that proliferation of NIH 3T3 cells was strongly suppressed, and G-120-mediated inhibition of DNA synthesis proved to involve a cytostatic, rather than a cytotoxic, effect, as shown by cytotoxicity and apoptosis assays. More importantly, G-120 strongly reduced the gelatinolytic and collagenase activities of MMP proteins, as well as expression of MMP-2 and MMP-9. Electrophoretic mobility shift assays revealed that it suppressed the DNA binding activity of NF-kappaB. Collectively, our observations show that G-120 strongly inhibits the activation of MMPs and NF-kappaB.


Subject(s)
Cell Proliferation/drug effects , Matrix Metalloproteinase Inhibitors , Plant Proteins/pharmacology , Ulmus/chemistry , Animals , DNA/biosynthesis , Dose-Response Relationship, Drug , Glycoproteins/pharmacology , Matrix Metalloproteinases/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NIH 3T3 Cells , Plants, Medicinal/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL