Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Heliyon ; 9(2): e13236, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36798766

ABSTRACT

Bone remodelling is the process of bone resorption and formation, necessary to maintain bone structure or for adaptation to new conditions. Mechanical loadings, such as exercise, weight bearing and orthodontic force, play important roles in bone remodelling. During the remodelling process, osteocytes play crucial roles as mechanosensors to regulate osteoblasts and osteoclasts. However, the precise molecular mechanisms by which the mechanical stimuli affect the function of osteocytes remain unclear. In the present study, we analysed viability, cell cycle distribution and gene expression pattern of murine osteocyte-like MLO-Y4 cells exposed to tension force (TF). Cells were subjected to TF with 18% elongation at 6 cycles/min for 24 h using Flexcer Strain Unit (FX-3000). We found that TF stimulation induced cell cycle arrest at G2/M phase but not cell death in MLO-Y4 cells. Differentially expressed genes (DEGs) between TF-stimulated and unstimulated cells were identified by microarray analysis, and a marked increase in glutathione-S-transferase α (GSTA) family gene expression was observed in TF-stimulated cells. Enrichment analysis for the DEGs revealed that Gene Ontology (GO) terms and Kyoto Encyclopedia Genes and Genomes (KEGG) pathways related to the stress response were significantly enriched among the upregulated genes following TF. Consistent with these results, the production of reactive oxygen species (ROS) was elevated in TF-stimulated cells. Activation of the tumour suppressor p53, and upregulation of its downstream target GADD45A, were also observed in the stimulated cells. As GADD45A has been implicated in the promotion of G2/M cell cycle arrest, these observations may suggest that TF stress leads to G2/M arrest at least in part in a p53-dependent manner.

2.
J Bone Miner Metab ; 40(3): 434-447, 2022 May.
Article in English | MEDLINE | ID: mdl-35195777

ABSTRACT

INTRODUCTION: The detailed mechanism of the process during bone healing of drill-hole injury has been elucidated, but a crucial factor in regulating drill-hole healing has not been identified. The transcription factor p53 suppresses osteoblast differentiation through inhibition of osterix expression. In present study, we demonstrate the effects of p53 deficiency on the capacity of MSCs and osteoblasts during drill-hole healing. MATERIALS AND METHODS: Mesenchymal stromal cells (MSCs) and osteoblasts were collected from bone marrow and calvaria of p53 knockout (KO) mice, respectively. The activities of cell mobility, cell proliferation, osteoblast differentiation, and wound healing of MSCs and/or osteoblasts were determined by in vitro experiments. In addition, bone healing of drill-hole injury in KO mice was examined by micro-CT and immunohistological analysis using anti-osterix, Runx2, and sclerostin antibodies. RESULTS: KO MSCs stimulated cell mobility, cell proliferation, and osteoblast differentiation. Likewise, KO osteoblasts enhanced cell proliferation and wound healing. KO MSCs and osteoblasts showed high potency in the inflammation and callus formation phases compared to those from wild-type (WT) mice. In addition, increased expression of osterix and Runx2 was observed in KO MSCs and osteoblasts that migrated in the drill-hole. Conversely, sclerostin expression was inhibited in KO mice. Eventually, KO mice exhibited high repairability of drill-hole injury, suggesting a novel role of p53 in MSCs and osteoblasts in improving bone healing. CONCLUSION: p53 Deficiency promotes bone healing of drill-hole injury by enhancing the bone-regenerative ability of MSCs and osteoblasts.


Subject(s)
Bone Regeneration , Core Binding Factor Alpha 1 Subunit , Mesenchymal Stem Cells , Osteoblasts , Tumor Suppressor Protein p53 , Animals , Bone Regeneration/physiology , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Cell Tissue Res ; 375(3): 655-664, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30284086

ABSTRACT

Nuclear protein, lamin A, which is a component of inner membrane on nucleoplasm, plays a role in nuclear formation and cell differentiation. The expression of mutated lamin A, termed progerin, causes a rare genetic aging disorder, Hutchinson-Gilford progeria syndrome, which shows abnormal bone formation with the decrease in a number of osteoblasts and osteocytes. However, exact molecular mechanism how progerin exerts depressive effects on osteogenesis has not been fully understood. Here, we created mouse lamin A dC50 cDNA encoding progerin that lacks 50 amino acid residues at C-terminus, transfected it in mouse preosteoblast-like MC3T3-E1 cells, and examined the changes in osteoblast phenotype. When lamin A dC50-expressed cells were cultured with differentiation-inductive medium, alkaline phosphatase (ALP) activity and mRNA levels of major osteoblast markers, type I collagen (Col1), bone sialoprotein (BSP), dentine matrix protein 1 (DMP1), and Runx2 were significantly decreased, and no mineralized nodules were detected as seen in control cells expressing empty vector. In the culture with mineralization-inductive medium, mRNA levels of BSP, osteocalcin, DMP1, Runx2, and osterix were strongly decreased parallel with loss of mineralization in lamin A dC50-expressed cells, while mineralized nodules appear at 21 days in control cells. Furthermore, lamin A dC50 expression was depressed nuclear localization of ß-catenin with the decrease of GSK-3ß phosphorylation level. These results suggest that lamin A dC50 depresses osteoblast differentiation in both early and late stages, and it negatively regulates ß-catenin activity interacting with GSK-3ß in cytoplasm.


Subject(s)
Cell Differentiation , Lamin Type A/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Signal Transduction , beta Catenin/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Collagen Type I/metabolism , Deoxycholic Acid/pharmacology , Humans , Indoles/pharmacology , Lamin Type A/chemistry , Maleimides/pharmacology , Mice , Osteoblasts/drug effects , Signal Transduction/drug effects
4.
Mol Cell Biochem ; 435(1-2): 97-108, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28534121

ABSTRACT

Tetrahydrobiopterin (BH4) is a common coenzyme of phenylalanine-, tyrosine-, and tryptophan hydroxylases, alkylglycerol monooxygenase, and NO synthases (NOS). Synthetic BH4 is used medicinally for BH4-responsive phenylketonuria and inherited BH4 deficiency. BH4 supplementation has also drawn attention as a therapy for various NOS-related cardio-vascular diseases, but its use has met with limited success in decreasing BH2, the oxidized form of BH4. An increase in the BH2/BH4 ratio leads to NOS dysfunction. Previous studies revealed that BH4 supplementation caused a rapid urinary loss of BH4 accompanied by an increase in the blood BH2/BH4 ratio and an involvement of probenecid-sensitive but unknown transporters was strongly suggested in these processes. Here we show that OAT1 and OAT3 enabled cells to take up BP (BH4 and/or BH2) in a probenecid-sensitive manner using rat kidney slices and transporter-expressing cell systems, LLC-PK1 cells and Xenopus oocytes. Both OAT1 and OAT3 preferred BH2 and sepiapterin as their substrate roughly 5- to 10-fold more than BH4. Administration of probenecid acutely reduced the urinary exclusion of endogenous BP accompanied by a rise in blood BP in vivo. These results indicated that OAT1 and OAT3 played crucial roles: (1) in determining baseline levels of blood BP by excluding endogenous BP through the urine, (2) in the rapid distribution to organs of exogenous BH4 and the exclusion to urine of a BH4 excess, particularly when BH4 was administered, and (3) in scavenging blood BH2 by cellular uptake as the gateway to the salvage pathway of BH4, which reduces BH2 back to BH4.


Subject(s)
Biopterins/analogs & derivatives , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Biopterins/genetics , Biopterins/metabolism , Cell Line , Oocytes , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Probenecid/pharmacology , Rats , Rats, Sprague-Dawley , Xenopus
5.
Biochem Biophys Res Commun ; 488(4): 664-670, 2017 07 08.
Article in English | MEDLINE | ID: mdl-28237702

ABSTRACT

Lamin A/C is a component of the nuclear lamina, which is involved in cellular proliferation and differentiation. However, the mechanism by which lamin A regulates osteoblast differentiation is not well understood. In this study, we investigated lamin A/C expression during osteoblast differentiation in a preosteoblastic cell line, MC3T3-E1. Real-time PCR analysis showed that lamin A/C mRNA expression was upregulated during BMP-2 induced osteoblast differentiation. Treatment with the estrogen receptor antagonist, fulvestrant, inhibited osteoblast differentiation and the upregulation of lamin A/C mRNA and protein expressions in the presence of BMP-2. These results clearly demonstrated that lamin A/C expression correlates with osteoblast differentiation. To determine the roles of lamin A expression in osteoblast differentiation, MC3T3-E1 cells were transfected with a vector overexpressing lamin A. Results showed that lamin A overexpression promoted osteoblast differentiation and calcification by inducing the expression of alkaline phosphatase, type 1 collagen, BSP, osteocalcin, and DMP-1 in the presence of BMP-2. Furthermore, lamin A overexpression partially restored osteoblastic capacity in the presence of fulvestrant by increasing the expression of BSP, osteocalcin, and DMP-1. These results suggest that lamin A plays important roles in maintaining the osteoblast differentiation and function.


Subject(s)
Calcification, Physiologic , Cell Differentiation , Lamin Type A/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Animals , Cells, Cultured , Lamin Type A/metabolism , Mice , Mice, Inbred C57BL
6.
PLoS One ; 11(10): e0164305, 2016.
Article in English | MEDLINE | ID: mdl-27711248

ABSTRACT

Tetrahydrobiopterin (BH4) is an essential cofactor of nitric oxide synthase (NOS) and aromatic amino acid hydroxylases. BH4 and 7,8-dihydrobiopterin (BH2) are metabolically interchangeable at the expense of NADPH. Exogenously administered BH4 can be metabolized by the body, similar to vitamins. At present, synthetic BH4 is used as an orphan drug for patients with inherited diseases requiring BH4 supplementation. BH4 supplementation has also drawn attention as a means of treating certain cardiovascular symptoms, however, its application in human patients remains limited. Here, we tracked biopterin (BP) distribution in blood, bile, urine, liver, kidney and brain after BH4 administration (5 mg/kg rat, i.v.) with or without prior treatment with probenecid, a potent inhibitor of uptake transporters particularly including organic anion transporter families such as OTA1 and OAT3. The rapid excretion of BP in urine was driven by elevated blood concentrations and its elimination reached about 90% within 120 min. In the very early period, BP was taken up by the liver and kidney and gradually released back to the blood. BH4 administration caused a considerable decrease in the BH4% in blood BP as an inevitable compensatory process. Probenecid treatment slowed down the decrease in blood BP and simultaneously inhibited its initial rapid excretion in the kidney. At the same time, the BH4% was further lowered, suggesting that the probenecid-sensitive BP uptake played a crucial role in BH2 scavenging in vivo. This suggested that the overproduced BH2 was taken up by organs by means of the probenecid-sensitive process, and was then scavenged by counter-conversion to BH4 via the BH4 salvage pathway. Taken together, BH4 administration was effective at raising BP levels in organs over the course of hours but with extremely low efficiency. Since a high BH2 relative to BH4 causes NOS dysfunction, the lowering of the BH4% must be avoided in practice, otherwise the desired effect of the supplementation in ameliorating NOS dysfunction would be spoiled.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/analysis , Kidney/drug effects , Liver/drug effects , Probenecid/pharmacology , Animals , Bile/chemistry , Bile/drug effects , Bile/metabolism , Biopterins/blood , Biopterins/chemistry , Biopterins/metabolism , Biopterins/pharmacology , Biopterins/urine , Brain/drug effects , Brain/metabolism , Cyclosporine/pharmacology , Erythrocytes/chemistry , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Male , Nitric Oxide Synthase/metabolism , Rats
7.
Mol Cell Endocrinol ; 417: 114-23, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26419928

ABSTRACT

Glucocorticoids (GCs) affect the proliferation of growth plate chondrocytes. In this study, we investigated the role of the GC-inducible promyelocytic leukemia zinc finger (PLZF) gene in chondrocyte differentiation by using the chondrogenic cell line ATDC5. PLZF overexpression suppressed cell cycle progression (p < 0.01) and promoted differentiation into hypertrophic chondrocytes by inducing mRNA expression of alkaline phosphatase (p < 0.01), and the cyclin-dependent kinase (CDK) inhibitor p21 (p < 0.01). In contrast, PLZF knockdown impaired differentiation into hypertrophic chondrocytes and promoted cell cycle progression (p < 0.01). Treatment with the GC analogue dexamethasone (10(-6) M) suppressed cell cycle progression in ATDC5 cells. PLZF shRNA attenuated dexamethasone-induced cell cycle arrest (p < 0.01) by downregulating the mRNA expression of the CDK inhibitors p21 and p57 (p < 0.01). These results clearly indicated that PLZF promoted differentiation into hypertrophic chondrocytes and mediated dexamethasone-induced cell cycle arrest by regulating CDK inhibitors.


Subject(s)
Cell Cycle Checkpoints/drug effects , Chondrocytes/drug effects , Glucocorticoids/pharmacology , Kruppel-Like Transcription Factors/metabolism , Alkaline Phosphatase/genetics , Animals , Cell Differentiation/drug effects , Cell Line , Chondrocytes/cytology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Kruppel-Like Transcription Factors/genetics , Mice , Promyelocytic Leukemia Zinc Finger Protein
8.
Histochem Cell Biol ; 144(3): 261-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26105025

ABSTRACT

Glucocorticoids (GCs) regulate proliferation and differentiation in cultured mesenchymal cells through the modulation of various molecules. However, the relationship between growth factor signaling and GCs in differentiating chondrocytes has not been elucidated. In this study, we examined the effects of Wnt/ß-catenin signaling on chondrocyte differentiation and the effects of a GC analogue, dexamethasone (Dex), on Wnt/ß-catenin signaling activity by using a chondrocyte progenitor cell line ATDC5. Western blot analysis and TCF/LEF-optimized promoter EGFP (TOPEGFP) assay showed that both ß-catenin protein levels and TCF/LEF transcription were up-regulated during insulin-transferrin-sodium selenite (ITS)-induced chondrogenic differentiation. Morphological analysis showed that TCF/LEF transcription activity was most prominent in cartilage nodule-like structures. Furthermore, a ß-catenin mutant with constitutive transcriptional activity (ΔN90) showed increased Alcian blue staining intensity and mRNA expression of Sox9, Col2a, aggrecan, Col10, and alkaline phosphatase, even in the absence of ITS stimulation. In contrast, Dex suppressed formation of ITS-induced cartilage nodule-like structures, TCF/LEF-mediated transcription, and ß-catenin protein levels. Real-time PCR analysis showed that Dex increased the mRNA expression levels of secreted frizzled-related protein 1 (sFRP1) and Axin2. Furthermore, treatment with a sFRP1 inhibitor or the ΔN90 ß-catenin mutant transfection attenuated Dex-induced suppression of cartilage matrix production by increasing Sox9 mRNA levels. These results suggest that Dex inhibits chondrocyte differentiation via down-regulation of Wnt/ß-catenin signaling, which promotes chondrocyte differentiation in ATDC5 cells.


Subject(s)
Chondrocytes/drug effects , Dexamethasone/pharmacology , Wnt Proteins/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Line , Chondrogenesis/drug effects , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Plasmids/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
9.
Differentiation ; 87(3-4): 111-8, 2014.
Article in English | MEDLINE | ID: mdl-24582280

ABSTRACT

The 75 kDa transmembrane protein, p75(NTR), is a marker of mesenchymal stem cells (MSCs). Isolated MSCs are capable of differentiating into osteoblasts, but the molecular function of p75(NTR) in MSCs and osteoblasts is poorly understood. The aim of this study was to examine the function of p75(NTR) in the human MG63 osteoblast cell line compared to the murine MC3T3E-1 pre-osteoblast cell line. MG63 cells and MC3T3-E1 cells expressing exogenous p75(NTR) protein (denoted as p75-MG63 and p75GFP-E1, respectively) were generated to compare osteogenic differentiation and cell proliferation abilities. Overexpression of p75(NTR) induced alkaline phosphatase activity and the mRNA expression of osteoblast-related genes such as osterix and bone sialoprotein in both p75-MG63 and p75GFP-E1. Interestingly, exogenous p75(NTR) stimulated cell proliferation and cell cycle progression in p75GFP-E1, but not in p75-MG63. To elucidate any different effects of p75(NTR) expression on osteogenic differentiation and cell proliferation, we examined the mRNA expression of tropomyosin receptor kinase (trk) genes (trkA, trkB, trkC) and Nogo receptor (NgR), which are binding partners of p75(NTR). Although trkA, trkB, and trkC were detected in both p75-MG63 and p75GFP-E1, only NgR was detected in p75-MG63. We then used the K252a inhibitor of the trks to identify the signaling pathway for osteogenic differentiation and cell proliferation. Inhibition of trks by K252a suppressed p75(NTR)-mediated osteogenic differentiation of p75GFP-E1, whereas deletion of the GDI domain in P75(NTR) from the p75-MG63 produced enhanced cell proliferation compared to p75-MG63. These results suggest that p75(NTR) signaling associated with trk receptors promotes both cell proliferation and osteoblast differentiation, but that p75(NTR)-mediated proliferation may be suppressed by signaling from the p75(NTR)/NgR complex.


Subject(s)
Cell Proliferation , Osteoblasts/metabolism , Receptor, Nerve Growth Factor/metabolism , Alkaline Phosphatase/metabolism , Carbazoles/pharmacology , Cell Line , Humans , Indole Alkaloids/pharmacology , Osteoblasts/cytology , Osteogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Nerve Growth Factor/genetics , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Signal Transduction , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cell Tissue Res ; 354(3): 761-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23996200

ABSTRACT

Dexamethasone (Dex) regulates osteoblastic and adipocytic differentiation in mesenchymal progenitor cells through regulation of Wnt/ß-catenin signaling. To elucidate the regulatory mechanisms underlying the effects of Dex, we examine the expression of Axin2, which is an intracellular inhibitor of Wnt/ß-catenin signaling, in ROB-C26 clonal mesenchymal progenitor cells (C26). We observed the induction of Axin2 mRNA in C26 cells in response to Dex treatment. Treatment with a glucocorticoid receptor (GR) antagonist, mifepristone, showed that Dex-induced up-regulation of Axin2 is mediated by the GR. In the absence of Dex, gene silencing by using Axin2-targeted short hairpin RNA increased the number of alkaline phosphatase (ALP)-positive and nuclear ß-catenin-positive cells and ALP activity. In the presence of Dex, Axin2 knockdown resulted in an increased number of ALP-positive and nuclear ß-catenin-positive cells. Furthermore, Axin2 knockdown in Dex-treated cells suppressed adipocyte differentiation (as determined by reduced Oil Red O staining), reduced the number of PPARγ-positive and aP2-positive cells and decreased the mRNA expression of PPARγ2 and aP2. These results suggest that Axin2 plays a key role in adipocyte and osteoblastic differentiation by controlling ß-catenin expression.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Carrier Proteins/metabolism , Dexamethasone/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Adaptor Proteins, Signal Transducing , Adipocytes/cytology , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured , Gene Knockdown Techniques , Gene Silencing , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Rats , Transfection , Up-Regulation/drug effects , beta Catenin/biosynthesis , beta Catenin/genetics
11.
Histochem Cell Biol ; 138(6): 833-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22886144

ABSTRACT

Dexamethasone (Dex) stimulates the differentiation of mesenchymal progenitor cells into adipocytes and osteoblasts. However, the mechanisms underlying Dex-induced differentiation have not been clearly elucidated. We examined the effect of Dex on the expression and activity of Wnt/ß-catenin signal-related molecules in a clonal mesenchymal progenitor cell line, ROB-C26 (C26). Dex induced the mRNA expression of Wnt antagonists, dickkopf-1 (Dkk-1), and Wnt inhibitory factor (WIF)-1. Immunocytochemical analysis showed that the downregulation of ß-catenin protein expression by Dex occured concomitantly with the increased expression of the PPARγ protein. Dex decreased phosphorylation of Ser9-GSK3ß and expression of active ß-catenin protein. To examine the effects of Dex on Wnt/ß-catenin activity, we used immunocytochemistry to analyze TCF/LEF-mediated transcription during Dex-induced adipogenesis in Wnt indicator (TOPEGFP) C26 cells. Our results demonstrated that Dex repressed TCF/LEF-mediated transcription, but induced adipocyte differentiation. Treatment with a GSK3ß inhibitor attenuated Dex-induced inhibition of TCF/LEF-mediated transcriptional activity, but suppressed Dex-induced adipocyte differentiation, indicating that adipocyte differentiation and inhibition of Wnt/ß-catenin activity by Dex are mediated by GSK3ß activity. Furthermore, ß-catenin knockdown not only suppressed Dex-induced ALP-positive osteoblasts differentiation but also promoted Dex-induced adipocytes differentiation. These results suggest that inhibition of ß-catenin expression by Dex promotes the differentiation of mesenchymal progenitor cells into adipocytes.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Dexamethasone/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Signal Transduction/drug effects , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Adipocytes/cytology , Animals , Cells, Cultured , Down-Regulation/drug effects , Gene Silencing , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Osteoblasts/cytology , Osteoblasts/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Structure-Activity Relationship , Wnt Proteins/metabolism , beta Catenin/metabolism
12.
Differentiation ; 84(5): 392-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22906707

ABSTRACT

While the role of p75(NTR) signaling in the regulation of nerve-related cell growth and survival has been well documented, its actions in osteoblasts are poorly understood. In this study, we examined the effects of p75(NTR) on osteoblast proliferation and differentiation using the MC3T3-E1 pre-osteoblast cell line. Proliferation and osteogenic differentiation were significantly enhanced in p75(NTR)-overexpressing MC3T3-E1 cells (p75GFP-E1). In addition, expression of osteoblast-specific osteocalcin (OCN), bone sialoprotein (BSP), and osterix mRNA, ALP activity, and mineralization capacity were dramatically enhanced in p75GFP-E1 cells, compared to wild MC3T3-E1 cells (GFP-E1). To determine the binding partner of p75(NTR) in p75GFP-E1 cells during osteogenic differentiation, we examined the expression of trkA, trkB, and trkC that are known binding partners of p75(NTR), as well as NgR. Pharmacological inhibition of trk tyrosine kinase with the K252a inhibitor resulted in marked reduction in the level of ALPase under osteogenic conditions. The deletion of the GDI binding domain in the p75(NTR)-GFP construct had no effect on mineralization. Taken together, our studies demonstrated that p75(NTR) signaling through the trk tyrosine kinase pathway affects osteoblast functions by targeting osteoblast proliferation and differentiation.


Subject(s)
Cell Differentiation , Osteoblasts/cytology , Receptors, Nerve Growth Factor/metabolism , Animals , Calcification, Physiologic , Carbazoles , Cell Culture Techniques , Cell Proliferation , Gene Expression Regulation , Indole Alkaloids , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Mice , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Protein Binding , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Histochem Cell Biol ; 137(2): 235-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22119912

ABSTRACT

Lamin A/C gene encodes a nuclear membrane protein, and mutations in this gene are associated with diverse degenerative diseases that are linked to premature aging. While lamin A/C is involved in the regulation of tissue homeostasis, the distinct expression patterns are poorly understood in the mesenchymal cells differentiating into adipocytes. Here, we examined the expression of lamin A/C in a rat mesenchymal progenitor cell-line, ROB-C26 (C26). Immunocytochemical analysis showed that lamin A/C was transiently down-regulated in immature adipocytes, but its expression increased with terminal differentiation. To elucidate the role of lamin A/C expression on mesenchymal cell differentiation, lamin A/C expression was suppressed using short hairpin RNA (shRNA) molecules in C26 cells. In the absence of adipogenic stimuli, lamin A/C shRNA decreased alkaline phosphatase (ALP) activity, but induced preadipocyte factor -1 (Pref-1) mRNA expression. In the presence of adipogenic stimuli, lamin A/C knockdown promotes adipocytes differentiation, as assessed by the detection of an increase in Oil Red O staining. RT-PCR analysis showed that lamin A/C shRNA resulted in increased mRNA expression of PPARγ2 and aP2 during adipocyte differentiation. These results suggest that decreased lamin A/C expression levels not only suppress osteoblast phenotypes but also promote adipocyte differentiation in C26 cells.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/physiology , Lamin Type A/metabolism , Mesenchymal Stem Cells/cytology , Adaptor Protein Complex 2/metabolism , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Female , Gene Knockdown Techniques , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nuclear Lamina/metabolism , Osteoblasts , PPAR gamma/metabolism , RNA, Small Interfering/metabolism , Rats , Stem Cells/metabolism
15.
J Pharmacol Sci ; 117(3): 204-7, 2011.
Article in English | MEDLINE | ID: mdl-22027096

ABSTRACT

Streptococcus mutans is a bacterial cause of dental caries that is resistant to bacitracin. The aim of this study was to elucidate the mbrABCD-related bacitracin resistance mechanism of S. mutans. Transcriptome data demonstrated that the expression levels of 33 genes were induced more than twofold by bacitracin. Fourteen genes were selected from the upregulated genes, and defective mutants of these genes were constructed for measurement of their sensitivity to bacitracin. Among the mutants, only the mbrA- or mbrB-deficient mutants exhibited 100- to 121-fold greater sensitivity to bacitracin when compared with the wild-type strain. Moreover, knockout of the mbrC and mbrD genes abolished the bacitracin-induced mbrAB upregulation. These results suggest that both mbrC and mbrD are required for mbrAB upregulation that confers the bacitracin-resistant phenotype on S. mutans.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacitracin/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Gene Expression Regulation, Bacterial/drug effects , Streptococcus mutans/genetics , Dental Caries/genetics , Dental Caries/microbiology , Real-Time Polymerase Chain Reaction , Streptococcus mutans/physiology , Transcriptome/drug effects , Up-Regulation
16.
Calcif Tissue Int ; 88(4): 336-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21258786

ABSTRACT

Runx2, a transcription factor, is essential for osteoblastic differentiation, bone formation, and maintenance. We examined the effect of Runx2 on transdifferentiation of 3T3-L1 preadipocytes into functional, mature osteoblasts. Forced expression of exogenous Runx2 using a retroviral gene-delivery system showed increases of alkaline phosphatase (ALP) activity and expression of the osteoblastic marker genes osteocalcin (OC), bone sialoprotein (BSP), and osterix (Osx), accompanied by low-level matrix mineralization. In contrast, adipocytic differentiation was completely blocked with downregulation of adipogenic transcription factors PPARγ2, C/EBPα, and C/EBPδ. Treatment of dexamethasone (Dex), a synthetic glucocorticoid, stimulated the formation of mineralized nodules in Runx2-overexpressing 3T3-L1 cells with increases of ALP, OC, BSP, and Osx expression. Here, we focused on a dual specific phosphatase, mitogen-activated protein kinase (MKP-1), since Dex significantly increased MKP-1 expression in Runx2-overexpressing 3T3-L1 cells. Forced expression of exogenous MKP-1 resulted in accumulation of robust matrix mineralization in parallel with induction of ALP activity and expression of OC, BSP, and Osx in Runx2-overexpressing 3T3-L1 cells. These results suggest that simultaneous overexpression of Runx2 and MKP-1 is effective for transdifferentiation of preadipocytes into fully differentiated bone-forming osteoblasts and provide a novel strategy for cell-based therapeutic applications requiring significant numbers of osteogenic cells to synthesize mineralized constructs for the treatment of large bone defects.


Subject(s)
Bone and Bones/cytology , Core Binding Factor Alpha 1 Subunit/metabolism , Dual Specificity Phosphatase 1/metabolism , Osteoblasts/cytology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Bone and Bones/metabolism , Cell Differentiation , Cell Transdifferentiation , Dexamethasone/pharmacology , Down-Regulation , Gene Transfer Techniques , Glucocorticoids/pharmacology , Mice , Plasmids/metabolism , Retroviridae/genetics
17.
Cell Biol Int ; 32(2): 239-46, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17950631

ABSTRACT

Dentin matrix protein 1 (DMP1) is an acidic phosphorylated extracellular protein and essential for mineralization of dentin and bone; however, the precise mechanism regulating DMP1 expression is not fully understood. A synthetic glucocorticoid (GC), dexamethasone (Dex), promotes an early osteoblast differentiation of a mesenchymal progenitor, ROB-C26 (C26), in parallel with inductive expression of an osteoblast-specific transcription factor, Runx2, and other extracellular matrix proteins such as osteocalcin and bone sialoprotein (BSP). We have examined the effect of Dex on DMP1 expression via induction of Runx2 in C26 cells. Real time RT-PCR showed that Dex increases DMP1 mRNA expression levels at time- and dose-dependent manners and a GC antagonist, RU486, drastically inhibited DMP1 mRNA expression levels. Furthermore, Dex increased the luciferase activity of six-repeated osteoblast-specific cis-acting element 2 (6 x OSE2), which is the binding sequence of Runx2, suggesting that Dex stimulates DMP1 expression via activation of Runx2. However, unexpected results showed that overexpression of exogenous Runx2 depressed DMP1 mRNA expression level, even after cells had been treated with Dex, while downregulated expression of endogenous Runx2 enhanced Dex-induced DMP1 mRNA expression level. These results imply that large amounts of exogenous Runx2 inhibit DMP1 expression, whereas small amounts are more effective for Dex-induced DMP1 expression in C26 cells. Therefore, Dex may activate some factors that inhibit negative action of Runx2 on DMP1 expression. Since mitogen-activating protein kinase (MAPK) phosphatase-1 (MKP-1) has been reported to affect the Dex-induced osteoblast differentiation via decrease of Runx2-phosphorylation, we focus on the relationship between MKP-1 and DMP1 expression. Dex increases MKP-1 expression, and overexpression of exogenous MKP-1 showed significant increase of luciferase activity of 6 x OSE up to the level detected in Dex-treated C26 cells. However, no inductive DMP1 mRNA expression level was found in C26 cells unlike BSP and OPN. These results suggest that although MKP-1 increases DNA-binding activity of Runx2, DMP1 expression may require the collaboration of MKP-1 and additive factors to stimulate Runx2-mediated DMP1 expression in the post-transcriptional event of Dex-treated C26 cells.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Dexamethasone/pharmacology , Extracellular Matrix Proteins , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Mesenchymal Stem Cells/drug effects , Multipotent Stem Cells/drug effects , Phosphoproteins , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Dexamethasone/metabolism , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glucocorticoids/metabolism , Humans , Integrin-Binding Sialoprotein , Mesenchymal Stem Cells/physiology , Multipotent Stem Cells/physiology , Osteopontin/genetics , Osteopontin/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Glucocorticoid/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism
18.
Arch Oral Biol ; 53(1): 87-94, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17825242

ABSTRACT

OBJECTIVE: A zinc finger type transcription factor, AJ18, is thought to be a negative regulator of osteoblast differentiation, but its expression mechanism is not fully understood. Retinoic acid (RA) is a metabolite of vitamin A and involves the proliferation and differentiation in a variety of cells. To verify the effect of RA on osteoblast differentiation, AJ18 expression level was examined using a rat clonal preosteoblastic cell line, ROB-C20 (C20). DESIGN: Confluent C20 cells were treated with or without RA (10(-6)M) for several days. Northern, real time RT-PCR and Western blotting analyses were performed to examine AJ18 expression pattern in gene and protein levels. To identify the active promoter sequence of AJ18 gene, luciferase assay was designed. Furthermore, the effect of overexpressed AJ18 in C20 cells on alkaline phosphatase (ALP) mRNA expression and its activity was compared with that of RA-treated cells. RESULTS: RA increased the expression of AJ18 mRNA from 2 to 13 days as well as its protein production. However, no significant changes of Runx2 mRNA expression and undetectable osterix mRNA expression were observed in C20 cells treated with or without RA. Luciferase assay showed increases in promoter activities in some constructs of 5'-flanking region of AJ18 gene in RA-treated C20 cells. On the other hand, RA decreases enzymatic activity and mRNA expression level of ALP, but mRNA expression levels of bone sialoprotein and osteocalcin were not altered. Interestingly, reduced ALP activity and its mRNA expression level were detected in exogenous AJ18-overexpressing C20 cells. CONCLUSIONS: This study supports the hypothesis that RA may restrict to the differentiation of C20 cells into mature osteoblasts via inductive AJ18 expression with activation of multiple signal pathways.


Subject(s)
DNA-Binding Proteins/genetics , Osteoblasts/cytology , Promoter Regions, Genetic , Repressor Proteins/genetics , Tretinoin/pharmacology , Up-Regulation , 5' Untranslated Regions , Alkaline Phosphatase/analysis , Alkaline Phosphatase/genetics , Animals , Blotting, Northern/methods , Blotting, Western/methods , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Cell Differentiation/drug effects , Cell Line , DNA-Binding Proteins/analysis , Gene Expression/drug effects , Osteoblasts/metabolism , RNA, Messenger/analysis , Rats , Repressor Proteins/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Sulfanilamides
19.
Bone ; 40(1): 84-92, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16949358

ABSTRACT

To clarify the effects of glucocorticoids (GCs) on osteoblast and adipocyte differentiation, we investigated the effects of dexamethasone (Dex), a GC analogue on transcription factors for osteoblasts (Runx2, Dlx5 and Osterix) and adipocytes (C/EBPs such as C/EBPalpha, C/EBPbeta and C/EBPdelta, and PPARgamma2), late osteoblastic markers, bone sialoprotein (BSP) and osteocalcin (OC), and adipocyte differentiation-dependent protein, aP2 in a clonal mesenchymal progenitor cell line, ROB-C26 (C26). C26 cells were dose- and time-dependently responsive to Dex in terms of an increase in not only mRNA and protein expressions of the C/EBPs, PPARgamma2 and aP2, but also Runx2, Dlx5, BSP and OC with no induction of Osterix, which is considered to act mainly on terminal osteoblast differentiation. Cycloheximide pretreatment indicated that Dex signaling immediately increases expressions of the C/EBPs and Dlx5, while expressions of the rest of the genes require de novo protein synthesis. Continuous Dex treatment stimulated adipocyte formation, but failed to induce Osterix expression and mineralized matrix formation. However, BMP-2 treatment of Dex-treated cells induced Osterix expression and subsequent mineralized matrix formation. These results indicate that Dex up-regulates the C/EBPs followed by increasing PPARgamma2 and aP2 expressions in C26 cells to induce adipocyte differentiation, while Dex enhances Dlx5 followed by increasing Runx2, BSP and OC expressions at gene and protein levels, but cannot induce Osterix expression, suggesting that Dex does not promote their terminal osteoblast differentiation.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Dexamethasone/pharmacology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Transcription Factors/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Glucocorticoids/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/drug effects , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
20.
J Oral Sci ; 47(4): 199-207, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16415564

ABSTRACT

Runx2 is essential for osteoblast differentiation and gene expression of bone matrix proteins, however, little is known about the mechanism regulating its activity. In this study, the role of Runx2 on gene expression of transcription factors, AJ18, Msx2, and Dlx5, was examined in vitro. It is known that AJ18 and Msx2 act as repressors to inhibit activity of Runx2, whereas Dlx5 promotes its activity. An expression vector inserted Runx2 cDNA was transiently overexpressed in a rat multipotential mesenchymal cell line, ROB-C26 (C26). Real time reverse transcription-PCR analysis showed that, in exogenous Runx2-overexpressing C26 cells (C26-Rx), AJ18 expression increased 1.8-fold, Msx2 expression increased 3.0-fold, and Dlx5 expression increased 2.7-fold compared to the cells transfected with vector alone (C26-Co). Luciferase assay also showed that, in C26-Rx, AJ18 promoter activity increased 2.1-fold compared to C26-Co. Furthermore, gene expression of alkaline phosphatase (ALP) and bone matrix proteins including type I collagen (Col1), osteocalcin (OC), osteopontin (OPN), and matrix Gla protein (MGP) was examined. In C26-Rx, MGP expression increased 1.8-fold, and OPN expression increased 1.4-fold compared to C26-Co. However, no significant difference in Col1, ALP, and OC expressions was detected between C26-Rx and C26-Co. These results suggest that the existence of autoregulatory feed back loops, which inhibit Runx2 activity through the interaction of AJ18, Dlx5, and Msx2 cooperating with that of MGP and OPN, interferes with the differentiation of C26 cells toward mature osteoblasts.


Subject(s)
Bone Matrix/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation/genetics , Mesenchymal Stem Cells/physiology , Multipotent Stem Cells/physiology , Proteins/genetics , Transcription Factors/genetics , Alkaline Phosphatase/genetics , Animals , Cell Differentiation/genetics , Cell Line , Collagen Type I/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Homeostasis/genetics , Osteoblasts/physiology , Osteocalcin/genetics , Osteopontin , Phosphoproteins/genetics , Rats , Repressor Proteins/genetics , Sialoglycoproteins/genetics , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...