Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Periodontal Res ; 53(2): 200-209, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29063603

ABSTRACT

BACKGROUND AND OBJECTIVE: Single nucleotide polymorphisms (SNPs) of paraoxonase 1 (PON1) are known to be associated with the pathogenesis of osteoporosis and periodontitis. However, the effects of PON1 on the osteoblastic differentiation of periodontal ligament (PDL) cells are unclear. In this study, we examined the effects of PON1 on the osteoblastic differentiation of PDL cells, and analysed the role of PON1 SNPs on the pathogenesis of aggressive periodontitis (AgP) in the Japanese population. MATERIAL AND METHODS: Human PDL (HPDL) cells were exposed to the PON1 plasmid and PON1 inhibitor, 2-hydroxyquinoline, and cultured in mineralization medium. Expression of calcification-related genes and calcified nodule formation were assessed by real-time PCR, an alkaline phosphatase (ALPase) activity assay and Alizarin red staining. Sanger sequencing was performed to evaluate whether PON1 SNPs are associated with the pathogenesis of AgP in Japanese people. RESULTS: During osteoblastic differentiation of HPDL cells, expression of PON1 mRNA increased in a time-dependent manner. PON1 stimulated an increase in expression of mRNA for calcification-related genes, as well as ALPase activity. In contrast, 2-hydroxyquinoline clearly inhibited the expression of calcification-related genes, ALPase activity and calcified nodule formation in HPDL cells. Moreover, there was a statistically significant difference in the minor allele frequency of PON1 SNP rs854560 between the Japanese control database and patients with AgP in the Japanese population (P = .0190). CONCLUSION: PON1 induced cytodifferentiation and mineralization of HPDL cells, and PON1 SNP rs854560 may be associated with the pathogenesis of AgP in the Japanese population.


Subject(s)
Aggressive Periodontitis/pathology , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/pharmacology , Cell Differentiation/drug effects , Periodontal Ligament/drug effects , Periodontal Ligament/pathology , Adult , Aggressive Periodontitis/enzymology , Alkaline Phosphatase/analysis , Aryldialkylphosphatase/genetics , Bone Resorption , Calcification, Physiologic , Cells, Cultured , Female , Gene Expression , Gene Frequency , Humans , Hydroxyquinolines/antagonists & inhibitors , Japan , Male , Osteoblasts/drug effects , Periodontal Pocket , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism
2.
J Dent Res ; 96(3): 339-346, 2017 03.
Article in English | MEDLINE | ID: mdl-28221099

ABSTRACT

Sphingomyelin phosphodiesterase 3 ( Smpd3), which encodes neutral sphingomyelinase 2 (nSMase2), is a key molecule for skeletal development as well as for the cytodifferentiation of odontoblasts and alveolar bone. However, the effects of nSMase2 on the cytodifferentiation of periodontal ligament (PDL) cells are still unclear. In this study, the authors analyzed the effects of Smpd3 on the cytodifferentiation of human PDL (HPDL) cells. The authors found that Smpd3 increases the mRNA expression of calcification-related genes, such as alkaline phosphatase (ALPase), type I collagen, osteopontin, Osterix (Osx), and runt-related transcription factor (Runx)-2 in HPDL cells. In contrast, GW4869, an inhibitor of nSMase2, clearly decreased the mRNA expression of ALPase, type I collagen, and osteocalcin in HPDL cells, suggesting that Smpd3 enhances HPDL cytodifferentiation. Next, the authors used exome sequencing to evaluate the genetic variants of Smpd3 in a Japanese population with aggressive periodontitis (AgP). Among 44 unrelated subjects, the authors identified a single nucleotide polymorphism (SNP), rs145616324, in Smpd3 as a putative genetic variant for AgP among Japanese people. Moreover, Smpd3 harboring this SNP did not increase the sphingomyelinase activity or mRNA expression of ALPase, type I collagen, osteopontin, Osx, or Runx2, suggesting that this SNP inhibits Smpd3 such that it has no effect on the cytodifferentiation of HPDL cells. These data suggest that Smpd3 plays a crucial role in maintaining the homeostasis of PDL tissue.


Subject(s)
Aggressive Periodontitis/genetics , Periodontal Ligament/cytology , Sphingomyelin Phosphodiesterase/physiology , Adult , Aggressive Periodontitis/enzymology , Alkaline Phosphatase/metabolism , Calcification, Physiologic , Cell Differentiation , Cell Line , Cells, Cultured , Collagen Type I/metabolism , Female , Gene Expression , Genome-Wide Association Study , Humans , Immunoblotting , Japan , Male , Osteocalcin/metabolism , Osteopontin/metabolism , Phenotype , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Sphingomyelin Phosphodiesterase/genetics
3.
J Dent Res ; 95(9): 1026-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27129490

ABSTRACT

Periodontal ligaments (PDLs) play an important role in remodeling the alveolar bond and cementum. Characterization of the periodontal tissue transcriptome remains incomplete, and an improved understanding of PDL features could aid in developing new regenerative therapies. Here, we aimed to generate and analyze a large human PDL transcriptome. We obtained PDLs from orthodontic treatment patients, isolated the RNA, and used a vector-capping method to make a complementary DNA library from >20,000 clones. Our results revealed that 58% of the sequences were full length. Furthermore, our analysis showed that genes expressed at the highest frequencies included those for collagen type I, collagen type III, and proteases. We also found 5 genes whose expressions have not been previously reported in human PDL. To access which of the highly expressed genes might be important for PDL cell differentiation, we used real-time polymerase chain reaction to measure their expression in differentiating cells. Among the genes tested, the cysteine protease cathepsin K had the highest upregulation, so we measured its relative expression in several tissues, as well as in osteoclasts, which are known to express high levels of cathepsin K. Our results revealed that PDL cells express cathepsin K at similar levels as osteoclasts, which are both expressed at higher levels than those of the other tissues tested. We also measured cathepsin K protein expression and enzyme activity during cell differentiation and found that both increased during this process. Immunocytochemistry experiments revealed that cathepsin K localizes to the interior of lysosomes. Last, we examined the effect of inhibiting cathepsin K during cell differentiation and found that cathepsin K inhibition stimulated calcified nodule formation and increased the levels of collagen type I and osteocalcin gene expression. Based on these results, cathepsin K seems to regulate collagen fiber accumulation during human PDL cell differentiation into hard tissue-forming cells.


Subject(s)
Cathepsin K/metabolism , Periodontal Ligament/metabolism , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , Humans , Periodontal Ligament/cytology , Periodontal Ligament/growth & development , RNA/genetics , RNA/metabolism , Real-Time Polymerase Chain Reaction , Transcriptome
4.
J Periodontal Res ; 50(2): 248-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24948468

ABSTRACT

BACKGROUND AND OBJECTIVE: The proteasome inhibitor, bortezomib, is known to induce osteoblastic differentiation in a number of cell lines, such as mesenchymal stem cells and osteoblastic precursor cells. As periodontal ligament (PDL) cells are multipotent, we examined whether bortezomib may induce the differentiation of PDL cells into hard-tissue-forming cells. MATERIAL AND METHODS: A mouse PDL clone cell line, MPDL22 cells, was cultured in mineralization medium in the presence or absence of bortezomib. Expression of calcification-related genes and calcified-nodule formation were evaluated by real-time PCR and Alizarin Red staining, respectively. RESULTS: Bortezomib increased the expression of calcification-related mRNAs, such as tissue nonspecific alkaline phosphatase isoenzyme (ALPase), bone sialoprotein (Bsp), runt-related transcription factor 2 (Runx2) and osteopontin, and calcified-nodule formation in MPDL22 cells. These effects were induced, in part, by increasing the cytosolic accumulation and nuclear translocation of ß-catenin, leading to an increase in expression of bone morphogenetic protein (Bmp)-2, -4 and -6 mRNAs. In addition, bortezomib enhanced BMP-2-induced expression of Bsp and osteopontin mRNAs and increased calcified-nodule formation in MPDL22 cells. CONCLUSION: Bortezomib induced cytodifferentiation and mineralization of PDL cells by enhancing the accumulation of ß-catenin within the cytosol and the nucleus and increasing the expression of Bmp-2, -4 and -6 mRNAs. Moreover, bortezomib enhanced the BMP-2-induced cytodifferentiation and mineralization of PDL cells, suggesting that bortezomib may be efficacious for use in periodontal regeneration therapy.


Subject(s)
Bortezomib/pharmacology , Calcification, Physiologic/drug effects , Periodontal Ligament/drug effects , Proteasome Inhibitors/pharmacology , Alkaline Phosphatase/drug effects , Animals , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 4/drug effects , Bone Morphogenetic Protein 6/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Core Binding Factor Alpha 1 Subunit/drug effects , Cytosol/drug effects , Isoenzymes/drug effects , Mice , Osteopontin/drug effects , Periodontal Ligament/cytology , beta Catenin/drug effects
5.
Oncogene ; 28(4): 619-24, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18978812

ABSTRACT

Nitric oxide (NO) is a major effector molecule in cancer prevention. A number of studies have shown that NO prodrug JS-K (O(2)-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate) induces apoptotic cell death in vitro and in vivo, indicating that it is a promising new therapeutic for cancer. However, the mechanism of its tumor-killing activity remains unclear. Ubiquitin plays an important role in the regulation of tumorigenesis and cell apoptosis. Our earlier report has shown that inactivation of the ubiquitin system through blocking E1 (ubiquitin-activating enzyme) activity preferentially induces apoptosis in p53-expressing transformed cells. As E1 has an active cysteine residue that could potentially interact with NO, we hypothesized that JS-K could inactivate E1 activity. E1 activity was evaluated by detecting ubiquitin-E1 conjugates through immunoblotting. JS-K strikingly inhibits the ubiquitin-E1 thioester formation in cells in a dose-dependent manner with an IC(50) of approximately 2 microM, whereas a JS-K analog that cannot release NO did not affect these levels in cells. Moreover, JS-K decreases total ubiquitylated proteins and increases p53 levels, which is mainly regulated by ubiquitin and proteasomal degradation. Furthermore, JS-K preferentially induces cell apoptosis in p53-expressing transformed cells. These findings indicate that JS-K inhibits E1 activity and kills transformed cells harboring wild-type p53.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azo Compounds/pharmacology , Neoplasms/metabolism , Nitric Oxide/metabolism , Piperazines/pharmacology , Prodrugs/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Azo Compounds/therapeutic use , Cell Line, Transformed , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Piperazines/therapeutic use , Prodrugs/therapeutic use , Tumor Suppressor Protein p53 , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination/drug effects
6.
Article in English | MEDLINE | ID: mdl-19202599

ABSTRACT

The ubiquitin conjugating system represents a rich source of potential molecular targets for cancer and other diseases. One target of great interest is the RING finger ubiquitin ligase (E3) Hdm2/Mdm2, which is frequently overexpressed in cancer and is a critical E3 for the tumor suppressor p53. For those 50% of tumors that express wild-type p53, agents that inhibit Hdm2 have great potential clinical utility. We summarize our ongoing efforts to identify inhibitors of Hdm2 E3 activity by high-throughput screening of both defined small molecules and natural product extracts. Employing a strategy using both enzymatic and cell-based assays, we have identified inhibitors that block the E3 activity of Hdm2, activate a p53 response, preferentially kill p53-expressing cells, and have the capacity to differentially cause death of transformed cells. Therefore, screening for inhibitors of Hdm2 ubiquitin ligase activity through in vitro assays represents a powerful means of identifying molecules that activate p53 in cancer cells to induce apoptosis. We also discuss the potential of inhibitors of ubiquitin-activating enzyme (E1) that were discovered during these screens. E1 inhibitors may similarly serve as the basis for novel therapeutics. Additionally, they represent unique tools for providing new insights into the ubiquitin conjugating system.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , Protein Binding , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors
7.
Osteoarthritis Cartilage ; 11(1): 6-15, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12505482

ABSTRACT

OBJECTIVE: Runx2 (also known as Cbfa1) is a transcription factor required for chondrocyte maturation and osteoblast differentiation. While there is information on the regulation of its expression during osteogenesis, much less is known about it during cartilage maturation. Here we asked whether Runx2 expression and function are affected by retinoic acid (RA) and parathyroid hormone-related peptide (PTHrP), which represent an important stimulator and inhibitor of chondrocyte maturation, respectively. DESIGN: We first cloned and characterized Runx2 expressed by chick chondrocytes (cRunx2). We then constructed expression vectors of cRunx2 and a dominant-negative form (DN-cRunx2) and determined their effects on chondrocyte maturation in culture before and during retinoid and PTHrP treatment. RESULTS: cRunx2 showed similar transactivation activity to that of its mammalian counterparts although it has a very short QA domain and lacks a small portion of the PST domain. cRunx2 over-expression stimulated chondrocyte maturation, as indicated by increases in alkaline phosphatase activity (APase), mineralization, and type X collagen and MMP-13 expression, and by maintenance of Indian hedgehog (Ihh) expression. RA treatment stimulated cRunx2 gene expression and boosted its pro-maturation effects. PTHrP treatment blocked Runx2 expression and its pro-maturation effects. Over-expression of DN-cRunx2 inhibited maturation and even prevented RA from exerting its pro-maturation role. CONCLUSIONS: As previously indicated by mammalian studies, cRunx2 has chondrocyte pro-maturation activity. Its expression and roles are favorably modulated by retinoid signaling but are completely inhibited by PTHrP. A model integrating cRunx2 with PTHrP, Ihh and retinoid signaling and operating during skeletogenesis is proposed.


Subject(s)
Chondrocytes/metabolism , Keratolytic Agents/pharmacology , Neoplasm Proteins , Peptide Hormones/pharmacology , Transcription Factors/metabolism , Tretinoin/pharmacology , Animals , Blotting, Northern , Cartilage/cytology , Cartilage/metabolism , Cells, Cultured , Chick Embryo , Chondrocytes/drug effects , Core Binding Factor Alpha 1 Subunit , In Situ Hybridization , Parathyroid Hormone-Related Protein , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Analysis, Protein , Transcription Factors/drug effects , Transcription Factors/genetics
8.
Osteoarthritis Cartilage ; 11(1): 36-43, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12505485

ABSTRACT

OBJECTIVE: Members of the Wnt signaling protein family are expressed during cartilage development and skeletogenesis, but their roles and mechanisms of action in those processes remain unclear. Recently, we found that beta-catenin-LEF/TCF-dependent Wnt signaling stimulates chondrocyte maturation and hypertrophy and extracellular matrix calcification in vitro, events normally associated with cartilage-to-bone transition during skeletogenesis. Thus, we tested here whether activation of this pathway promotes endochondral ossification. DESIGN: Chick chondrocytes were infected with avian retroviral expression vectors encoding constitutive-active (CA) or dominant-negative (DN) forms of LEF, which activate or block beta-catenin-dependent Wnt signaling respectively. These cells and companion uninfected control cells were seeded into type I collagen gels and transplanted intramuscularly into nude mice. The resulting ectopic tissue masses forming over time in vivo were subjected to histological and molecular biological analyses. RESULTS: Transplantation of chick chondrocytes induced de novo endochondral bone formation. In situ hybridization and RT-PCR using species-specific probes and primers showed that the ectopic cartilaginous tissue was avian and thus donor-derived, whereas the bone tissue was mouse and thus host-derived. CA-LEF-expressing ectopic tissue masses contained abundant bone and marrow, while DN-LEF-expressing masses contained little bone and lacked marrow. CONCLUSIONS: Activation of beta-catenin-LEF/TCF-dependent Wnt signaling accelerates chondrocyte maturation and replacement of cartilage by bone.


Subject(s)
Chondrocytes/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Ossification, Heterotopic/etiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Zebrafish Proteins , Animals , Cells, Cultured , Chick Embryo , Lymphoid Enhancer-Binding Factor 1 , Mice , Ossification, Heterotopic/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...