Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Tissue Eng Regen Med ; 19(3): 565-575, 2022 06.
Article in English | MEDLINE | ID: mdl-34973125

ABSTRACT

BACKGROUND: The use of mouse bone marrow mesenchymal stem cells (mBMSCs) represents a promising strategy for performing preclinical studies in the field of cell-based regenerative medicine; however, mBMSCs obtained via conventional isolation methods have two drawbacks, i.e., (i) they are heterogeneous due to frequent macrophage contamination, and (ii) they require long-term culturing for expansion. METHODS: In the present study, we report a novel strategy to generate highly pure mBMSCs using liposomal clodronate. This approach is based on the properties of the two cell populations, i.e., BMSCs (to adhere to the plasticware in culture dishes) and macrophages (to phagocytose liposomes). RESULTS: Liposomal clodronate added during the first passage of whole bone marrow culture was selectively engulfed by macrophages in the heterogeneous cell population, resulting in their effective elimination without affecting the MSCs. This method allowed the generation of numerous high-purity Sca-1+CD44+F4/80- mBMSCs (> 95%) with just one passaging. Comparative studies with mBMSCs obtained using conventional methods revealed that the mBMSCs obtained in the present study had remarkably improved experimental utilities, as demonstrated by in vitro multilineage differentiation and in vivo ectopic bone formation assays. CONCLUSION: Our newly developed method, which enables the isolation of mBMSCs using simple and convenient protocol, will aid preclinical studies based on the use of MSCs.


Subject(s)
Clodronic Acid , Mesenchymal Stem Cells , Animals , Cell Differentiation , Clodronic Acid/pharmacology , Liposomes , Macrophages , Mice
2.
Biomater Res ; 25(1): 30, 2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34565474

ABSTRACT

OBJECTIVE: Various surface modification techniques that can further improve the function and usability of stainless steel as a medical device have been reported. In the present study, the physical and biological properties of nanoporous stainless steel as well as its usefulness for drug delivery were assessed. METHODS: The specimen was prepared with a circular disk shape (15 mm in diameter and 1 mm in thickness). The disk was subjected to electropolishing at a constant voltage of 20 V and 10 A for 10 min in an acidic environment (50% H2SO4). Everolimus (EVL) was used as a testing drug for drug-loading capacity of the material surface and release kinetics. The physiobiological properties of the material were assessed using platelet adhesion, and smooth muscle cell (SMC) adhesion, migration, and proliferation assays. RESULTS: The surface roughness of the postpolishing group was greater than that of the nonpolishing group. Platelet adhesion and SMC adhesion and migration were inhibited in the postpolishing group compared to those in the prepolishing group. In the postpolishing group, the total amount of EVL on the surface (i.e., drug storage rate) was higher and the drug release rate was lower, with half the amount of the EVL released within 4 days compared with only 1 day for that of the prepolishing group. CONCLUSION: Taken together, this stainless steel with a nanoporous surface could be used as a medical device for controlling cellular responses and carrying drugs.

3.
Tissue Eng Regen Med ; 18(1): 155-164, 2021 02.
Article in English | MEDLINE | ID: mdl-33058016

ABSTRACT

BACKGROUND: The delivery of recombinant human bone morphogenetic protein 2 (rhBMP2) by using various carriers has been used to successfully induce bone formation in many animal models. However, the effect of multiple administration of rhBMP2 on bone formation and BMP2 antibody production has not been determined. Our aim was to examine the bone formation activity of rhBMP2 and serum levels of anti-BMP2 antibodies following the repeated administration of rhBMP2 in mice. METHODS: Absorbable collagen sponges or polyphosphazene hydrogels containing rhBMP2 were subcutaneously implanted or injected into one side on the back of six-week-old C57BL/6 mice. Three or 4 weeks later, the same amount of rhBMP2 was administered again with the same carrier into the subcutaneous regions on the other side of the back or into calvarial defects. The effects of a single administration of rhBMP2 on the osteoinductive ability in the ectopic model were compared with those of repeated administrations. In vivo ectopic or orthotopic bone formation was evaluated using microradiography and histological analyses. Serum concentrations of anti-rhBMP2 antibodies were measured by ELISAs. RESULTS: Re-administration of the same amount of rhBMP2 into the subcutaneous area showed a comparable production of ectopic bone as after the first administration. The bone forming ability of repeated rhBMP2 administrations was equal to that of single rhBMP2 administration. The administration of rhBMP2 into calvarial defects, following the first subcutaneous administration of rhBMP2 on the back, completely recovered the defect area with newly regenerated bone within 3 weeks. Repeated administration of rhBMP2 at 4-week intervals did not significantly alter the serum levels of anti-BMP2 antibodies and did not induce any inflammatory response. The serum obtained from rhBMP2-exposed mice had no effect on the ability of rhBMP2 to induce osteogenic gene expressions in MC3T3-E1. CONCLUSION: We suggest that the osteoinductive ability of rhBMP2 is not compromised by repeated administrations. Thus, rhBMP2 can be repeatedly used for bone regeneration at various sites within a short duration.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration , Osteogenesis , Animals , Bone Morphogenetic Protein 2/administration & dosage , Bone and Bones , Mice , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage
4.
J Bone Metab ; 27(4): 267-279, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33317230

ABSTRACT

BACKGROUND: Macrophage-stimulating protein (MSP; also known as macrophage stimulating 1 and hepatocyte growth factor-like protein) has been shown to play a crucial role in calcium homeostasis and skeletal mineralization in zebrafish. However, the precise role of MSP in osteoblasts has not been elucidated. In this study, we investigated the effect of MSP on osteoblast differentiation of pre-osteoblast cells. METHODS: Osteoblast differentiation upon MSP treatment was evaluated by analyzing the osteogenic gene expression, alkaline phosphatase (ALP) activity, and mineralized nodule formation. To assess changes in the MSP-RON signaling pathway, knockdown of Ron gene was performed using siRNA and pharmacological inhibitor treatment. RESULTS: Expression of the tyrosine kinase receptor RON, a receptor of MSP, was found to be significantly increased during osteoblast differentiation. MSP treatment significantly upregulated the expression of osteogenic marker genes and remarkably increased ALP activity and mineralized nodule formation. Conversely, knockdown of Ron significantly attenuated the expression of osteogenic marker genes and ALP activity that were induced upon MSP treatment. Mechanistically, MSP treatment significantly enhanced the phosphorylation of extracellular signal-regulated kinase (ERK); however, additional treatment with the selective ERK inhibitor PD98059 attenuated the effect of MSP on osteoblast differentiation. CONCLUSIONS: Altogether, these results indicate that the MSP-RON axis is involved in promoting osteoblast differentiation via activation of the ERK signaling pathway.

5.
Cell Death Dis ; 11(5): 344, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393737

ABSTRACT

The CUE domain-containing 2 (CUEDC2) protein plays critical roles in many biological processes, such as the cell cycle, inflammation, and tumorigenesis. However, whether CUEDC2 is involved in osteoblast differentiation and plays a role in bone regeneration remains unknown. This study investigated the role of CUEDC2 in osteogenesis and its underlying molecular mechanisms. We found that CUEDC2 is expressed in bone tissues. The expression of CUEDC2 decreased during bone development and BMP2-induced osteoblast differentiation. The overexpression of CUEDC2 suppressed the osteogenic differentiation of precursor cells, while the knockdown of CUEDC2 showed the opposite effect. In vivo studies showed that the overexpression of CUEDC2 decreased bone parameters (bone volume, bone area, and bone mineral density) during ectopic bone formation, whereas its knockdown increased bone volume and the reconstruction percentage of critical-size calvarial defects. We found that CUEDC2 affects STAT3 activation by regulating SOCS3 protein stability. Treatment with a chemical inhibitor of STAT3 abolished the promoting effect of CUEDC2 silencing on osteoblast differentiation. Together, we suggest that CUEDC2 functions as a key regulator of osteoblast differentiation and bone formation by targeting the SOCS3-STAT3 pathway. CUEDC2 manipulation could serve as a therapeutic strategy for controlling bone disease and regeneration.


Subject(s)
Cell Differentiation , Osteoblasts/metabolism , Osteogenesis , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , Skull/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , 3T3 Cells , Animals , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Osteoblasts/pathology , Phosphorylation , Protein Stability , Repressor Proteins/genetics , Signal Transduction , Skull/pathology , Skull/surgery
6.
Stem Cell Res ; 45: 101799, 2020 05.
Article in English | MEDLINE | ID: mdl-32339903

ABSTRACT

The primary cilium acts as a sensory organelle with diverse receptors and ion channels to detect extracellular cues and regulate cellular functions, including cell migration. The migration of mesenchymal stem cells (MSCs) to bone remodeling sites is important for bone homeostasis. Recently, we have suggested that osteopontin (OPN) is a significant chemoattractant in MSC migration to bone remodeling sites. The objective of this study was to determine whether the primary cilium acts as a chemoattractant sensory unit to detect OPN cues and control MSC migration. We found that the loss of primary cilium induced by silencing of IFT88 reduced OPN-induced migration of MSCs. The effect of IFT88 silencing on cellular attachment, spreading, and proliferation was negligible. The loss of primary cilium did not affect the level of integrinß1 or CD44, two known receptors for OPN. Interestingly, CD44 was localized to the primary cilium by OPN stimulus. Knockdown of IFT88 or CD44 dysregulated OPN-induced signaling activation and abolished OPN-induced Cdc42 activation. Our findings suggest that the primary cilium acts as a chemoattractant sensor for OPN to regulate MSC migration by controlling not only CD44-mediated OPN signaling, but also Cdc42-mediated actin cytoskeleton rearrangement.


Subject(s)
Mesenchymal Stem Cells , Osteopontin , Cell Movement , Cilia , Hyaluronan Receptors/genetics , Osteopontin/genetics , Signal Transduction
7.
Biochem Biophys Res Commun ; 520(2): 459-465, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31607484

ABSTRACT

The peroxisome proliferator-activated receptor (PPAR)-α agonist fenofibrate is used as a lipid-lowering agent to reduce cholesterol and triglyceride in blood. In this study, we investigated whether fenofibrate affects osteoblast differentiation of osteogenic precursor cells. Quantitative real-time PCR and alkaline phosphatase (ALP) staining assays revealed that fenofibrate can enhance the osteoblast differentiation of C3H10T1/2 and MC3T3-E1 cells. In contrast with fenofibrate, the PPARγ agonist rosiglitazone decreased or did not affect the expression of osteogenic genes in these cells. Fenofibrate dose- and time-dependently increased PPARα expression, and concomitantly increased the expression of bone morphogenetic protein 2 (BMP2). Knockdown of PPARα abolished fenofibrate-induced BMP2 expression, activity of the BMP2 promoter gene, and calcium deposition. The chromatin immunoprecipitation assay demonstrated that fenofibrate increased BMP2 expression by inducing direct binding of PPARα to the BMP2 promoter region. Taken together, we suggest that fenofibrate has a stimulatory effect on osteoblast differentiation via the elevation of PPARα levels and the PPARα-mediated BMP2 expression. Our findings provide fenofibrate as a useful agent for controlling hypercholesterolemic patients with osteoporosis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Fenofibrate/pharmacology , Osteoblasts/drug effects , PPAR alpha/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Gene Expression Regulation/drug effects , Mice , Osteoblasts/cytology , Osteoblasts/physiology , PPAR alpha/agonists , PPAR alpha/genetics , Promoter Regions, Genetic , Transcription, Genetic
8.
Exp Mol Med ; 50(11): 1-16, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30393382

ABSTRACT

Supplementation of mesenchymal stem cells (MSCs) at sites of bone resorption is required for bone homeostasis because of the non-proliferation and short lifespan properties of the osteoblasts. Calcium ions (Ca2+) are released from the bone surfaces during osteoclast-mediated bone resorption. However, how elevated extracellular Ca2+ concentrations would alter MSCs behavior in the proximal sites of bone resorption is largely unknown. In this study, we investigated the effect of extracellular Ca2+ on MSCs phenotype depending on Ca2+ concentrations. We found that the elevated extracellular Ca2+ promoted cell proliferation and matrix mineralization of MSCs. In addition, MSCs induced the expression and secretion of osteopontin (OPN), which enhanced MSCs migration under the elevated extracellular Ca2+ conditions. We developed in vitro osteoclast-mediated bone resorption conditions using mouse calvaria bone slices and demonstrated Ca2+ is released from bone resorption surfaces. We also showed that the MSCs phenotype, including cell proliferation and migration, changed when the cells were treated with a bone resorption-conditioned medium. These findings suggest that the dynamic changes in Ca2+ concentrations in the microenvironments of bone remodeling surfaces modulate MSCs phenotype and thereby contribute to bone regeneration.


Subject(s)
Calcium/pharmacology , Cell Movement , Cell Proliferation , Mesenchymal Stem Cells/drug effects , Animals , Calcium/metabolism , Cell Line, Tumor , Cells, Cultured , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Osteopontin/genetics , Osteopontin/metabolism
9.
J Cell Physiol ; 233(2): 1512-1522, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28574578

ABSTRACT

Differentiated ameloblasts secret enamel matrix proteins such as amelogenin, ameloblastin, and enamelin. Expression levels of these proteins are regulated by various factors. To find a new regulatory factor for ameloblast differentiation, we performed 2D-PAGE analysis using mouse ameloblast lineage cell line (mALCs) cultured with mineralizing medium. Of identified proteins, family with sequence similarity 50 member A (Fam50a) was significantly increased during differentiation of mALCs. Fam50a protein was also highly expressed in secretory ameloblasts of mouse tooth germs. In mALCs cultures, forced expression of Fam50a up-regulated the expression of enamel matrix protein genes such as amelogenin, ameloblastin, and enamelin. In addition, up-regulation of Fam50a also increased ALP activity and mineralized nodule formation in a dose-dependent manner. In contrast, knockdown of Fam50a decreased expression levels of enamel matrix protein genes, ALP activity, and mineralized nodule formation. By fluorescence microscopy, endogenous Fam50a protein was found to be localized to the nucleus of ameloblasts. In addition, Fam50a synergistically increased Ambn transactivation by Runx2. Moreover, Fam50a increased binding affinity of Runx2 to Ambn promoter by physically interacting with Runx2. Taken together, these results suggest Fam50a might be a new positive regulator of ameloblast differentiation.


Subject(s)
Ameloblasts/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/metabolism , Molar/metabolism , Nuclear Proteins/metabolism , Alkaline Phosphatase/metabolism , Amelogenin/genetics , Amelogenin/metabolism , Animals , Binding Sites , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , DNA-Binding Proteins/genetics , Dental Enamel Proteins/genetics , Dental Enamel Proteins/metabolism , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA-Binding Proteins , Signal Transduction , Time Factors , Tooth Calcification , Transcription, Genetic , Transcriptional Activation , Transfection
10.
PLoS One ; 12(11): e0188749, 2017.
Article in English | MEDLINE | ID: mdl-29182679

ABSTRACT

Epidermal growth factor-like repeats and discoidin I-like domain 3 (Edil3) is an extracellular matrix protein containing an Arg-Gly-Asp (RGD) motif that binds integrin. Recently, Edil3 has been implicated in various biological processes, including angiogenesis and cellular differentiation. It can inhibit inflammatory bone destruction. The objective of this study was to explore the role of Edil3 in osteoblast differentiation and its underlying molecular mechanisms. In wild-type mice, high expression levels of Edil3 mRNA were observed in isolated calvaria and tibia/femur bones. Immunohistochemical analysis showed that Edil3 protein was localized along periosteum and calcified regions surrounding bone tissues. When murine calvaria-derived MC3T3-E1 cells were cultured in osteogenic medium containing 50 µg/ml ascorbic acid and 5 mM ß-glycerophosphate, Edil3 mRNA and protein expression levels were increased. Treatment with Edil3 protein in growth media increased expression levels of alkaline phosphatase and osteocalcin gene and phosphorylation level of extracellular signal-regulated kinase (ERK). Edil3 treatment with osteogenic medium induced mineralization. Treatment with a neutralizing antibody against α5ß1 and MEK inhibitor U0126 inhibited Edil3-enhanced osteogenic marker gene expression and mineral deposition. Edil3 increased protein expression levels of transcription factor runt-related transcription factor2 (Runx2). Edil3-induced Runx2 protein expression was suppressed by pretreatment with U0126. Taken together, these results suggest that Edil3 may stimulate osteoblast differentiation and matrix mineralization by increasing expression of Runx2 through α5ß1 integrin /ERK pathway.


Subject(s)
Carrier Proteins/physiology , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin alpha5beta1/metabolism , Osteoblasts/cytology , Alkaline Phosphatase/genetics , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules , Cell Line , Culture Media , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteocalcin/genetics , Phosphorylation , Signal Transduction
11.
Eur J Pharmacol ; 806: 10-17, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28322831

ABSTRACT

The c-Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), have been recently introduced to negatively regulate bone morphogenetic protein (BMP)-induced osteogenesis. However, the effect of chemical inhibitors of c-Met receptor on osteoblast differentiation process has not been examined, especially the applicability of c-Met chemical inhibitors on in vivo bone regeneration. In this study, we demonstrated that chemical inhibitors of c-Met receptor tyrosine kinase, SYN1143 and SGX523, could potentiate the differentiation of precursor cells to osteoblasts and stimulate regeneration in calvarial bone defects of mice. Treatment with SYN1143 or SGX523 inhibited HGF-induced c-Met phosphorylation in MC3T3-E1 and C3H10T1/2 cells. Cell proliferation of MC3T3-E1 or C3H10T1/2 was not significantly affected by the concentrations of these inhibitors. Co-treatment with chemical inhibitor of c-Met and osteogenic inducing media enhanced osteoblast-specific genes expression and calcium nodule formation accompanied by increased Runx2 expression via c-Met receptor-dependent but Erk-Smad signaling independent pathway. Notably, the administration of these c-Met inhibitors significantly repaired critical-sized calvarial bone defects. Collectively, our results suggest that chemical inhibitors of c-Met receptor tyrosine kinase might be used as novel therapeutics to induce bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , 3T3-L1 Cells , Animals , Benzothiazoles/pharmacology , Calcification, Physiologic/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Pyridazines/pharmacology , Skull/cytology , Skull/drug effects , Skull/metabolism , Skull/physiology , Triazoles/pharmacology
12.
PLoS One ; 11(7): e0159234, 2016.
Article in English | MEDLINE | ID: mdl-27404388

ABSTRACT

Chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) is an orphan nuclear receptor that regulates many key biological processes, including organ development and cell fate determination. Although the biological functions of COUP-TFII have been studied extensively, little is known about what regulates its gene expression, especially the role of inducible extracellular factors in triggering it. Here we report that COUP-TFII expression is regulated specifically by fibroblast growth factor 2 (FGF2), which mediates activation of the MEK1/2 pathway in mesenchymal lineage C3H10T1/2 cells. Although FGF2 treatment increased cell proliferation, the induction of COUP-TFII expression was dispensable. Instead, FGF2-primed cells in which COUP-TFII expression was induced showed a low potential for osteoblast differentiation, as evidenced by decreases in alkaline phosphatase activity and osteogenic marker gene expression. Reducing COUP-TFII by U0126 or siRNA against COUP-TFII prevented the anti-osteogenic effect of FGF2, indicating that COUP-TFII plays a key role in the FGF2-mediated determination of osteoblast differentiation capability. This report is the first to suggest that FGF2 is an extracellular inducer of COUP-TFII expression and may suppress the osteogenic potential of mesenchymal cells by inducing COUP-TFII expression prior to the onset of osteogenic differentiation.


Subject(s)
COUP Transcription Factor II/genetics , Cell Differentiation/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Osteoblasts/cytology , 3T3 Cells , Animals , Cell Lineage/drug effects , Cell Proliferation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Time Factors
13.
Biomed Res Int ; 2015: 926291, 2015.
Article in English | MEDLINE | ID: mdl-26491693

ABSTRACT

Recently a submicron particle of biphasic calcium phosphate ceramic (BCP) with through-hole (donut-shaped BCP (d-BCP)) was developed for improving the osteoconductivity. This study was performed to examine the usefulness of d-BCP for the delivery of osteoinductive rhBMP2 and the effectiveness on cranial bone regeneration. The d-BCP was soaked in rhBMP2 solution and then freeze-dried. Scanning electron microscope (SEM), energy dispersive spectroscopy (EDS), and Raman spectroscopy analyses confirmed that rhBMP2 was well delivered onto the d-BCP surface and the through-hole. The bioactivity of the rhBMP2/d-BCP composite was validated in MC3T3-E1 cells as an in vitro model and in critical-sized cranial defects in C57BL/6 mice. When freeze-dried d-BCPs with rhBMP2 were placed in transwell inserts and suspended above MC3T3-E1, alkaline phosphatase activity and osteoblast-specific gene expression were increased compared to non-rhBMP2-containing d-BCPs. For evaluating in vivo effectiveness, freeze-dried d-BCPs with or without rhBMP2 were implanted into critical-sized cranial defects. Microcomputed tomography and histologic analysis showed that rhBMP2-containing d-BCPs significantly enhanced cranial bone regeneration compared to non-rhBMP2-containing control. These results suggest that a combination of d-BCP and rhBMP2 can accelerate bone regeneration, and this could be used to develop therapeutic strategies in hard tissue healing.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Ceramics/pharmacology , Hydroxyapatites/pharmacology , Skull/injuries , Animals , Humans , Male , Mice , Recombinant Proteins/pharmacology
14.
J Biol Chem ; 290(21): 13556-66, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25873397

ABSTRACT

Endoplasmic reticulum (ER) stress transducers, such as old astrocyte specifically induced substance (OASIS) and activating transcription factor 6 (ATF6), which are induced by bone morphogenetic protein 2 (BMP2), regulate bone formation and osteoblast differentiation. Here, we examined the role of cAMP response element-binding protein H (CREBH), a member of the same family of ER membrane-bound basic leucine zipper (bZIP) transcription factors as OASIS and ATF6, in osteoblast differentiation and bone formation. Proinflammatory cytokine TNFα increased CREBH expression by up-regulating the nuclear factor-κB (NF-κB) signaling pathway in osteoblasts, increased the level of N-terminal fragment of CREBH in the nucleus, and inhibited BMP2 induction of osteoblast specific gene expression. Overexpression of CREBH suppressed BMP2-induced up-regulation of the osteogenic markers runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and osteocalcin (OC) in MC3T3-E1 cells and primary osteoblasts, as well as BMP2-induced ALP activity and OC protein production. In contrast, knockdown of CREBH attenuated the inhibitory effect of TNFα on BMP2-induced osteoblast differentiation. Mechanistic studies revealed that CREBH increased the expression of Smad ubiquitination regulatory factor 1 (Smurf1), leading to ubiquitin-dependent degradation of Smad1, whereas knockdown of CREBH inhibited TNFα-mediated degradation of Smad1 by Smurf1. Consistent with these in vitro findings, administration of Ad-CREBH inhibited BMP2-induced ectopic and orthotopic bone formation in vivo. Taken together, these results suggest that CREBH is a novel negative regulator of osteoblast differentiation and bone formation.


Subject(s)
Cell Differentiation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Osteoblasts/cytology , Osteogenesis/drug effects , Smad1 Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/genetics , Endoplasmic Reticulum Stress , Humans , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Proteolysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
J Bone Miner Res ; 29(7): 1586-96, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24643989

ABSTRACT

Relaxin (Rln), a polypeptide hormone of the insulin superfamily, is an ovarian peptide hormone that is involved in a diverse range of physiological and pathological reactions. In this study, we investigated the effect of Rln on bone morphogenetic protein 2 (BMP-2)-induced osteoblast differentiation and bone formation. Expression of Rln receptors was examined in the primary mouse bone marrow stem cells (BMSCs) and mouse embryonic fibroblast cell line C3H/10T1/2 cells by RT-PCR and Western blot during BMP-2-induced osteoblast differentiation. The effect of Rln on osteoblast differentiation and mineralization was evaluated by measuring the alkaline phosphatase activity, osteocalcin production, and Alizarin red S staining. For the in vivo evaluation, BMP-2 and/or Rln were administered with type I collagen into the back of mice, and after 3 weeks, bone formation was analyzed by micro-computed tomography (µCT). Western blot was performed to determine the effect of Rln on osteoblast differentiation-related signaling pathway. Expression of Rxfp 1 in BMSCs and C3H/10T1/2 cells was significantly increased by BMP-2. In vitro, Rln augmented BMP-2-induced alkaline phosphatase expression, osteocalcin production, and matrix mineralization in BMSCs and C3H/10T1/2 cells. In addition, in vivo administration of Rln enhanced BMP-2-induced bone formation in a dose-dependent manner. Interestingly, Rln synergistically increased and sustained BMP-2-induced Smad, p38, and transforming growth factor-ß activated kinase (TAK) 1 phosphorylation. BMP-2-induced Runx 2 expression and activity were also significantly augmented by Rln. These results show that Rln enhanced synergistically BMP-2-induced osteoblast differentiation and bone formation through its receptor, Rxfp 1, by augmenting and sustaining BMP-2-induced Smad and p38 phosphorylation, which upregulate Runx 2 expression and activity. These results suggest that Rln might be useful for therapeutic application in destructive bone diseases.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteogenesis/drug effects , Relaxin/pharmacology , Animals , Calcification, Physiologic/drug effects , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/enzymology , Phosphorylation/drug effects , Protein Binding/drug effects , Receptors, G-Protein-Coupled/metabolism , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Biol Chem ; 287(23): 18888-99, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22493443

ABSTRACT

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is an orphan nuclear receptor of the steroid-thyroid hormone receptor superfamily. COUP-TFII is widely expressed in multiple tissues and organs throughout embryonic development and has been shown to regulate cellular growth, differentiation, and organ development. However, the role of COUP-TFII in osteoblast differentiation has not been systematically evaluated. In the present study, COUP-TFII was strongly expressed in multipotential mesenchymal cells, and the endogenous expression level decreased during osteoblast differentiation. Overexpression of COUP-TFII inhibited bone morphogenetic protein 2 (BMP2)-induced osteoblastic gene expression. The results of alkaline phosphatase, Alizarin Red staining, and osteocalcin production assay showed that COUP-TFII overexpression blocks BMP2-induced osteoblast differentiation. In contrast, the down-regulation of COUP-TFII synergistically induced the expression of BMP2-induced osteoblastic genes and osteoblast differentiation. Furthermore, the immunoprecipitation assay showed that COUP-TFII and Runx2 physically interacted and COUP-TFII significantly impaired the Runx2-dependent activation of the osteocalcin promoter. From the ChIP assay, we found that COUP-TFII repressed DNA binding of Runx2 to the osteocalcin gene, whereas Runx2 inhibited COUP-TFII expression via direct binding to the COUP-TFII promoter. Taken together, these findings demonstrate that COUP-TFII negatively regulates osteoblast differentiation via interaction with Runx2, and during the differentiation state, BMP2-induced Runx2 represses COUP-TFII expression and promotes osteoblast differentiation.


Subject(s)
COUP Transcription Factor II/metabolism , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , Matrix Metalloproteinase 2/metabolism , Osteoblasts/metabolism , Animals , COUP Transcription Factor II/genetics , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation/physiology , Humans , Matrix Metalloproteinase 2/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Binding
17.
Pharmacology ; 86(5-6): 327-35, 2010.
Article in English | MEDLINE | ID: mdl-21109762

ABSTRACT

BACKGROUND/AIMS: Cartilage oligomeric matrix protein (COMP)-angiopoietin 1 (Ang1) is a soluble and stable form of Ang1 which plays important roles in vessel formation and the survival of endothelial cells, neurons and cardiomyocytes. However, the effects of COMP-Ang1 on the survival of mesenchymal cells are unknown. Mesenchymal cells have been transplanted with some scaffolds for bone tissue regeneration, but they occasionally underwent cell death due to a lack of nutrient supply. This study examined the effects of COMP-Ang1 on the survival of mesenchymal cells under nutrient-deprived conditions. METHODS: Primary and C3H10T1/2 mesenchymal cells were cultured under serum deprivation with or without COMP-Ang1. The effects of COMP-Ang1 on mesenchymal cell survival and its molecular mechanism were determined using a viability test, RT-PCR, Western blotting and fluorescence-activated cell sorting analysis. RESULTS AND CONCLUSION: COMP-Ang1 inhibited the nutrient-deprived apoptotic cell death of mesenchymal cells through the Akt, p38 and extracellular-signal-regulated kinase (ERK) pathways. In addition, COMP-Ang1 reversed the nutrient-deprived suppression of cyclin D1 mRNA expression. These results suggest that COMP-Ang1 has a protective role in the survival of nutrient-deprived mesenchymal cells. The use of COMP-Ang1 with some scaffolds might be useful for bone tissue engineering.


Subject(s)
Apoptosis/drug effects , Mesenchymal Stem Cells/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cyclin D1/genetics , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
J Immunol ; 182(12): 7957-62, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494320

ABSTRACT

The nonessential amino acid L-glutamine (Gln) is the most abundant amino acid in plasma. Clinical trials have demonstrated that Gln therapy is safe and improves clinical outcomes in critically ill patients. We have previously shown that Gln protect animals from endotoxic shock through the inhibition of cytosolic phospholipase A(2) activity. In this study, we investigated how Gln regulates MAPK activation, as the molecular mechanism underlying Gln-induced cytosolic phospholipase A(2) inactivation. Gln rapidly (within 10 min) inactivated p38 and JNK, but not ERK, by dephosphorylating them only when these MAPKs were phosphorylated in response to LPS in vivo as well as in vitro. Western blot analysis revealed that Gln administration resulted in rapid ( approximately 5 min) phosphorylation and protein induction of MAP kinase phosphatase-1 (MKP-1). MKP-1 siRNA abrogated the Gln-mediated 1) inactivation of p38 and JNK, 2) induction of MKP-1, and 3) protection against endotoxic shock. The ERK inhibitor U0126 blocked Gln-induced MKP-1 phosphorylation and protein induction, as well as Gln's protective activity against endotoxic shock. These data suggest that Gln exerts a beneficial effect on endotoxic shock by inactivating p38 and JNK via a rapid induction of MKP-1 protein in an ERK-dependent way.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Glutamine/pharmacology , Shock, Septic/enzymology , Shock, Septic/pathology , Animals , Cell Line , Dual Specificity Phosphatase 1/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Enzymologic , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , RNA, Small Interfering/genetics , Shock, Septic/genetics , Shock, Septic/prevention & control , Survival Rate , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Immunology ; 124(3): 428-35, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18397269

ABSTRACT

Platelet-activating factor (PAF) has been demonstrated to augment resistance against Candida albicans infection. In this study, the role of nitric oxide (NO) in PAF-induced resistance in the kidneys was investigated. Pretreatment of the C. albicans-infected mice with PAF resulted in strong expression of messenger RNA (mRNA) and the protein synthesis of inducible nitric oxide synthase (iNOS). These PAF effects were inhibited to a significant degree by pretreatment with the nuclear factor-kappaB inhibitor, pyrrolidinedithiocarbamate. Pretreatment with PAF protected the mice from death caused by C. albicans infection and reduced the growth of fungus in the kidneys. The protective activity of PAF was abrogated by pretreatment with the iNOS inhibitor, aminoguanidine, and in the iNOS(-/-) mice. The PAF markedly increased the infiltration of neutrophils, but not macrophages, and also enhanced the mRNA expression levels of the CXC chemokine, keratinocyte-derived chemokine, in C. albicans-infected kidneys. These effects of PAF were attenuated in the aminoguanidine-treated mice and the iNOS(-/-) mice. These data show that NO plays an important role in PAF-induced protection against C. albicans.


Subject(s)
Candidiasis/prevention & control , Nitric Oxide/immunology , Platelet Activating Factor/immunology , Animals , Candida albicans/growth & development , Candidiasis/immunology , Female , Guanidines/pharmacology , Kidney/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...