Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Adv Clin Exp Med ; 2023 12 12.
Article in English | MEDLINE | ID: mdl-38085007

ABSTRACT

BACKGROUND: The number and activity of osteoblasts and osteoclasts play an important role in skeletal biology, especially in bone reconstruction. Scientific and rational regulation of osteoclast formation and activity has become a critical strategy aimed at inhibiting the loss of bone mass in the body and alleviating the occurrence of bone diseases. Currently, there are only a few reports related to hesperetin-regulated osteoclast differentiation. OBJECTIVES: To investigate the influence of hesperetin on osteoclast-like cell differentiation and formation, and determine whether the MAPK signaling pathway is involved in the differentiation process. MATERIAL AND METHODS: The RAW264.7 cells were induced and cultured in vitro to promote their differentiation into osteoclast-like cells. Tetrazolium bromide was utilized to determine the effects of different concentrations (100, 200, 400, and 600 µM) of hesperetin on the proliferation of osteoclast-like cell precursors. Osteoclast-like cell differentiation was conducted using tartrate-resistant acid phosphatase (TRAP) staining assay. The status of nuclei and actin filaments of differentiated osteoclast-like cells was observed with the use of 4',6-diamidino-2-phenylindole dihydrochloride (DAPI) and actin-tracker green staining experiments. Changes in key proteins of the MAPK signaling pathway were detected using western blot. RESULTS: The results of TRAP staining experiments showed that the number of osteoclast-like cells decreased with the increase in hesperetin concentration. The DAPI and actin-tracker green staining demonstrated that the nuclei of differentiated osteoclast-like cells reduced in size with the increase in hesperetin concentration, and the osteoclast-like cells became smaller. Western blot for key MAPK signaling pathway proteins revealed that phospho-ERK and phospho-p38 protein levels were not significantly inhibited, but phospho-JNK protein levels were reduced. CONCLUSIONS: Hesperetin inhibits the differentiation of osteoclast-like cells. Further studies revealed that hesperetin also affects the activation level of phospho-JNK, a key signaling protein of the MAPK signaling pathway, in the induced differentiation of osteoclast-like cells.

2.
Animals (Basel) ; 13(24)2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38136842

ABSTRACT

Primordial germ cells (PGCs) play a crucial role in preserving poultry genetic resources and conducting transgenic research. A system for the rapid isolation of PGCs from single chicken embryonic blood was established in this paper. We found that PGCs can migrate to the lower layer of chicken embryonic fibroblasts (CEFs) through pores smaller than their diameter, while blood cells cannot, when co-cultured with CEFs of passages two to three. Based on the characteristics of PGCs, we developed a new PGC isolation method (cell culture insert/CEF adhesion method) that utilizes a 3 µm cell culture insert and CEFs of passages two to three. Using this method, approximately 700 PGCs can be isolated from the blood of a single chicken embryo at Hamburger and Hamilton (H&H) stage 17 of development. The separation rate achieved was 87.5%, with a separation purity of 95%. The separation rate of this method was 41.4% higher than the common Percoll density gradient centrifugation method and 33.6% higher than lysis with ACK buffer. PGCs isolated from embryonic blood could proliferate 37-fold within 2 weeks when cultured in a feeder-free culture system. They also continued to express the SSEA-1 and DAZL proteins and retained the ability to migrate in vivo. Overall, PGCs separated using cell culture inserts/CEF adhesion method retain their stem cell characteristics and migration ability. PGCs also exhibit good proliferation efficiency, making them suitable for subsequent transgenic experiments or genetic resource preservation.

3.
Adv Healthc Mater ; 11(21): e2201457, 2022 11.
Article in English | MEDLINE | ID: mdl-36027596

ABSTRACT

Bone regeneration involves a cascade of sophisticated, multiple-staged cellular and molecular events, where early-phase stem cell recruitment mediated by chemokines and late-phase osteo-differentiation induced by pro-osteogenic factors play the crucial roles. Herein, enlightened by a bone physiological and regenerative mechanism, the multilayer nanofibrous membranes (PLLA@SDF-1α@MT01) consisting of PLLA/MT01 micro-sol electrospun nanofibers as intima and PLLA/PEG/SDF-1α electrospun nanofibers as adventitia are fabricated through micro-sol electrospinning and manual multi-layer stacking technologies. In vitro releasing profiles show that PLLA@SDF-1α@MT01 represents the rapid release of stromal cell-derived SDF-1α (SDF-1α) in the outer layers, while with long-term sustained release of MT01 in the inner layer. Owing to interconnected porosity like the natural bone extracellular matrix and improved hydrophilia, PLLA@SDF-1α@MT01 manifests good biocompatibility both in vitro and in vivo. Furthermore, PLLA@SDF-1α@MT01 can promote bone marrow mesenchymal stem cells (BMSCs) migration by amplifying the SDF-1α/CXCR4 axis and stimulating BMSCs osteo-differentiation via activating the MAPK pathway in vitro. PLLA@SDF-1α@MT01, with a programmed dual-delivery system, exhibits the synergetic promotion of bone regeneration and vascularization by emulating key characteristics of the staged bone repair in vivo. Overall, PLLA@SDF-1α@MT01 that mimics the endogenous cascades of bone regeneration can enrich the physiology-mimetic staged regenerative strategy and represent a promising tissue-engineered scaffold for the bone defect.


Subject(s)
Mesenchymal Stem Cells , Nanofibers , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Bone Regeneration , Osteogenesis
4.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-33576450

ABSTRACT

Abnormal osteoclastic activation and secretion of cysteine proteinases result in excessive bone resorption, which is one of the primary factors in the development of bone metabolic disorders, such as rheumatoid arthritis and osteoporosis. Mammalian cystatins have been demonstrated to restrain osteoclastic bone resorption and to alleviate severe osteolytic destruction via blocking the activity of cysteine proteinases. However, the specific effects of parasite cystatins on the formation and function of osteoclasts remain unclear. The purpose of the current study was to explore the effects of cystatins from Schistosoma japonicum (Sj­Cys) on macrophage colony­stimulating factor (M­CSF) and receptor activator of NF­κB ligand (RANKL)­induced osteoclast differentiation, as well as the underlying molecular mechanisms. Recombinant Sj­Cys (rSj­Cys) dose­dependently restrained osteoclast formation, with a half­maximal inhibitory concentration (IC50) value of 0.3 µM, and suppressed osteoclastic bone resorptive capability in vitro. The findings were based on tartrate resistant acid phosphatase (TRAP) staining and bone resorption assays, respectively. However, the cell viability assay showed that the repression of rSj­Cys on osteoclast formation did not depend on effects on cell viability or apoptosis. Based on the results of reverse transcription­quantitative PCR and western blot analysis, it was found that rSj­Cys downregulated the expression levels of osteoclastogenesis­related genes and proteins, by interfering with M­CSF and RANKL­induced NF­κB signaling and downstream transcription factors during early­phase osteoclastogenesis. Overall, the results of the present study revealed that rSj­Cys exerted an inhibitory role in osteoclast differentiation and could be a prospective biotherapeutic candidate for the treatment and prevention of bone metabolic disorders.


Subject(s)
Cystatins/pharmacology , Helminth Proteins/pharmacology , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , Animals , Bone Resorption/metabolism , Cell Differentiation/drug effects , Cystatins/genetics , Helminth Proteins/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/pharmacology , RAW 264.7 Cells , Recombinant Proteins/pharmacology , Schistosoma japonicum/genetics , Schistosoma japonicum/metabolism
5.
In Vitro Cell Dev Biol Anim ; 56(7): 533-542, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32839904

ABSTRACT

MicroRNAs (miRNAs) modulate gene expression and regulate many physiological and pathological conditions. However, their modulation and effect in osteoclastogenesis remain unknown. In this study, we investigated the role of miR-346-3p in regulating the osteoclast differentiation from RAW264.7 cells. We used the miRNA microarray assay, miR-346-3p mimic transfection, tartrate resistant acid phosphatase (TRAP) staining, bone resorption assay, qRT-PCR, and western blot. Our results showed that the expression of miR-346-3p was significantly upregulated during osteoclast differentiation. Further, by transfecting cells with miR-346-3p mimic, we observed an increased number of TRAP-positive multinucleated cells, increased pit area caused by bone resorption, and enhanced expression of osteoclast-specific genes and proteins. Conversely, miR-346-3p inhibition attenuated the osteoclast differentiation and function. Software-mediated prediction and validation using luciferase reporter assay showed that TRAF3, a negative regulator of osteoclast differentiation, was inhibited by miR-346-3p overexpression. Our results showed that miR-346-3p directly targeted TRAF3 mRNA via binding to its 3'-UTR and inhibited the expression of TRAF3 protein. Taken together, our results revealed that miR-346-3p promotes the regulation of osteoclastogenesis by suppressing the TRAF3 gene. In conclusion, miR-346-3p could be a novel therapeutic target for bone loss-related pathogenesis.


Subject(s)
MicroRNAs/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , TNF Receptor-Associated Factor 3/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Bone Resorption/genetics , Bone Resorption/pathology , HEK293 Cells , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice , MicroRNAs/genetics , RANK Ligand/pharmacology , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
6.
In Vitro Cell Dev Biol Anim ; 55(4): 243-251, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30887211

ABSTRACT

This study determined the effects of miR-338-3p on osteoclast (OC) differentiation and activation. The change levels of miR-338-3p in differentiated OCs were investigated by microRNA microarray assay and quantitative real-time PCR analysis. The effects of miR-338-3p on the differentiation and activation of OCs were determined by tartrate-resistant acid phosphatase staining resorption activity assay and Western blot. Target genes of miR-338-3p were identified by target gene prediction and dual-luciferase reporter gene detection assay as well as Western blot. Results showed that miR-338-3p was markedly downregulated in differentiated OCs. miR-338-3p could inhibit the formation and absorption activity of OCs. Western blot showed that miR-338-3p could influence the change levels of OC differentiation-related proteins. Dual-luciferase reporter gene detection assay and Western blot both showed that miR-338-3p directly targeted IKKß gene. In conclusion, miR-338-3p may affect the formation and activity of OCs by targeting the IKKß gene.


Subject(s)
Gene Expression Regulation , I-kappa B Kinase/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Animals , Base Sequence , Cell Differentiation/genetics , Down-Regulation , I-kappa B Kinase/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , MicroRNAs/genetics , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/pharmacology , RAW 264.7 Cells , Transcription Factors/metabolism
7.
Poult Sci ; 98(4): 1854-1860, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30590735

ABSTRACT

This study determined the influence of 3-(4-methoxyl)-1-(2-(4-coumarin)prop)-2-en-1-one (MCPEO) on the differentiation of Gaoyou duck embryo osteoclasts cultured in vitro. Bone marrow mononuclear cells (BM-MNCs) were harvested from 23-day-old Gaoyou duck embryos and induced by receptor activator of nuclear factor κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) in the presence of MCPEO at different concentrations (i.e., 1, 5, 10, 20, and 40 µM). Cell viability measurement, tartrate-resistant acid phosphatase (TRAP) staining, resorption activity assay, and co-staining with Tetramethylrhodamine (TRITC)-conjugated phalloidin and Hoechst 33,258 were conducted. Results indicated that MCPEO influenced the cell viability of the M-CSF + RANKL-induced BM-MNCs in a concentration-dependent manner, reduced the formation of positive multinucleated cells, and restrained the resorption capability of osteoclasts. Microfilament and nuclear staining indicated that MCPEO restricted the differentiation of BM-MNCs into large multinucleated osteoclasts. In short, MCPEO can inhibit the differentiation of BM-MNCs into mature osteoclasts in duck embryos. Therefore, MCPEO is a promising agent for the treatment of poultry osteoporosis.


Subject(s)
Cell Differentiation/drug effects , Coumarins/pharmacology , Ducks/embryology , Embryo, Nonmammalian/embryology , Osteoclasts/metabolism , Animals , Embryo, Nonmammalian/drug effects , In Vitro Techniques , Osteoclasts/drug effects
8.
Int J Mol Med ; 42(4): 2294-2302, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30066838

ABSTRACT

The aim of the present study was to determine the effects of the Ca2+/calmodulin­dependent protein kinase pathway inhibitor KN93 on osteoclastogenesis. RAW264.7 cells were incubated with macrophage colony­stimulating factor (M­CSF) + receptor activator of nuclear factor kappa­light­chain­enhancer of activated B cells ligand (RANKL) to stimulate osteoclastogenesis and then treated with 10 µM KN93. The methods included tartrate­resistant acid phosphatase (TRAP) staining, bone resorption activity assays, filamentous (F)­actin staining, determination of intracellular calcium ([Ca2+]i) levels, monitoring of osteoclast­specific gene expression levels and measurement of key transcription factors protein levels. The results suggested that KN93 inhibited the formation of TRAP­positive multinucleated cells, shaping of F­actin rings and resorption activity of the cells. In addition, KN93 decreased the concentration of [Ca2+]i, expression levels of osteoclast specific genes and protein levels of critical transcription factors in the M­CSF + RANKL­induced osteoclast model. In summary, KN93 may directly affect the differentiation and activation of osteoclasts, potentially through the Ca2+/calmodulin­dependent protein kinase signaling pathway.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Calmodulin/metabolism , Osteoclasts/metabolism , Protease Inhibitors/pharmacology , Animals , Mice , Osteoclasts/cytology , RANK Ligand/metabolism , RAW 264.7 Cells
9.
Int J Oncol ; 50(4): 1160-1174, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28350074

ABSTRACT

Emerging evidence demonstrates that the stromal derived factor-1 (SDF-1α)/CXCR4 axis is associated with tumor aggressiveness and metastasis, including glioma, the most common brain cancer. In the present study, we demonstrated that a novel designed peptide NT21MP of viral macrophage inflammatory protein II, targeting CXCR4 inhibits SDF-1α-induced activation in glioma. The effects of NT21MP on CXCR4 expression, cell survival and migration were assessed on the human glioma cell line U251 and SHG-44 exposed to SDF-1α, by western blotting, MTT assay, flow cytometry and transwell migration assay. Our results illustrated that NT21MP inhibited SDF-1α induced proliferation, migration and invasion by upregulated pro-apoptotic genes (Bak1 and caspase-3) and downregulated Bcl-2/Bax as well as cell cycle regulators (cyclin D1 and CDK4) to arrest cell cycle in G0/G1 phase and promote apoptosis. By RT-qPCR and immunofluorescence we found that CXCR4 was highly expressed in SHG-44 cells. Our results from wound healing and transwell invasion assays indicated silencing of CXCR4 significantly inhibited the SDF-1α­induced migration and invasion; similarly, flow cytometry showed that treatment with si-CXCR4 affected cell cycle and induced cell apoptosis in SHG-44. However, these effects were significantly weakened by NT21MP. In conclusion, the present study indicates that NT21MP plays a regulatory role in the SDF-1α/CXCR4 axis and further manages the invasion, migration, apoptosis and cell cycle of glioma cells. Thus, NT21MP might represent a novel therapeutic approach against glioma.

10.
Mol Med Rep ; 12(5): 6939-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26329402

ABSTRACT

The present study aimed to determine whether the mitogen­activated protein kinase (MAPK) signaling pathway is involved in the osteoprotegerin (OPG)­mediated inhibition of osteoclast differentiation and maturation. RAW264.7 cells were incubated with macrophage colony­stimulating factor (M­CSF) and receptor activator of nuclear factor­κB ligand (RANKL) to stimulate osteoclastogenesis and treated with various concentrations of OPG, an inhibitor of osteoclast differentiation. The differentiation and activation of osteoclasts were monitored by tartrate­resistant acid phosphatase staining and bone resorption assays. The phosphorylation levels of p38­MAPK, c­Jun N­terminal kinase (JNK)­MAPK and extracellular signal­regulated kinase (ERK)­MAPK in the different treatment groups were determined by western blot analysis. The results confirmed that M­CSF + RANKL stimulated the differentiation and activation of osteoclasts as well as the phosphorylation of p38­MAPK, JNK­MAPK and ERK­MAPK in osteoclasts, which was attenuated by OPG treatment. These findings indicated that the MAPK signaling pathway is involved in the regulation of osteoclastogenesis and in the OPG­mediated inhibition of osteoclast differentiation and activation.


Subject(s)
Cell Differentiation , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/cytology , Osteoprotegerin/metabolism , Signal Transduction , Animals , Bone Resorption/metabolism , Cell Line , Mice
11.
J Vet Sci ; 16(2): 151-6, 2015.
Article in English | MEDLINE | ID: mdl-25549213

ABSTRACT

The purpose of this study was to determine whether the Ca(2+) signaling pathway is involved in the ability of osteoprotegerin (OPG) to inhibit osteoclast differentiation and maturation. RAW264.7 cells were incubated with macrophage colony-stimulating factor (M-CSF) + receptor activator of nuclear factor-κB ligand (RANKL) to stimulate osteoclastogenesis and then treated with different concentrations of OPG, an inhibitor of osteoclast differentiation. The intracellular Ca(2+) concentration [Ca(2+)]i and phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) in the different treatment groups were measured by flow cytometry and Western blotting, respectively. The results confirmed that M-CSF + RANKL significantly increased [Ca(2+)]i and CaMKII phosphorylation in osteoclasts (p < 0.01), and that these effects were subsequently decreased by OPG treatment. Exposure to specific inhibitors of the Ca(2+) signaling pathway revealed that these changes varied between the different OPG treatment groups. Findings from the present study indicated that the Ca(2+) signaling pathway is involved in both the regulation of osteoclastogenesis as well as inhibition of osteoclast differentiation and activation by OPG.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Differentiation , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Macrophage Colony-Stimulating Factor/metabolism , Mice , RANK Ligand/metabolism
12.
Vet J ; 200(1): 181-5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24630041

ABSTRACT

This study aimed to determine the in vitro effect of cadmium on the differentiation of duck embryonic bone marrow cells into osteoclasts. Bone marrow cells were harvested from 23-day old Gaoyou duck embryos and were cultured with either 50 nmol/L cadmium alone or different cadmium concentrations (0, 5, 10, 20 and 50 nmol/L) in combination with macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANKL). Tartrate-resistant acid phosphatase (TRAP) staining, pit formation assay with bovine cortical bone slices, and co-staining with tetramethyl rhodamine isothiocyanate (TRITC)-conjugated phalloidin and Hoechst 33258 were performed to determine the number of TRAP-positive cells and bone resorption activity. Cadmium at a concentration ⩾ 10 nmol/L in the presence of M-CSF and RANKL significantly increased in a concentration-dependent manner both the number of TRAP-positive cells (35-160%) and bone resorption activity (36-261%) (P<0.05). High cadmium concentrations in the presence of M-CSF and RANKL markedly promoted the formation of filamentous (F)-actin rings in differentiated osteoclasts. In conclusion, cadmium promotes in vitro the differentiation of duck embryonic osteoclasts in the presence of M-CSF and RANKL.


Subject(s)
Bone Marrow Cells/drug effects , Cadmium/toxicity , Cell Differentiation/drug effects , Ducks/embryology , Osteoclasts/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Osteoclasts/cytology
13.
J Vet Sci ; 14(4): 405-12, 2013.
Article in English | MEDLINE | ID: mdl-23820214

ABSTRACT

The purpose of this study was to determine whether osteoprotegerin (OPG) could affect osteoclat differentiation and activation under serum-free conditions. Both duck embryo bone marrow cells and RAW264.7 cells were incubated with macrophage colony stimulatory factor (M-CSF) and receptor activator for nuclear factor kB ligand (RANKL) in serum-free medium to promote osteoclastogenesis. During cultivation, 0, 10, 20, 50, and 100 ng/mL OPG were added to various groups of cells. Osteoclast differentiation and activation were monitored via tartrate-resistant acid phosphatase (TRAP) staining, filamentous-actin rings analysis, and a bone resorption assay. Furthermore, the expression osteoclast-related genes, such as TRAP and receptor activator for nuclear factor κB (RANK), that was influenced by OPG in RAW264.7 cells was examined using real-time polymerase chain reaction. In summary, findings from the present study suggested that M-CSF with RANKL can promote osteoclast differentiation and activation, and enhance the expression of TRAP and RANK mRNA in osteoclasts. In contrast, OPG inhibited these activities under serum-free conditions.


Subject(s)
Avian Proteins/pharmacology , Bone Marrow Cells/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/pharmacology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Bone Marrow Cells/drug effects , Cells, Cultured , Ducks , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Osteoclasts/cytology , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tartrate-Resistant Acid Phosphatase
14.
Poult Sci ; 92(6): 1613-20, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23687158

ABSTRACT

ABSTRACT The aim of this study was to determine the influence of osteoprotegerin (OPG) on the differentiation, activation, and apoptosis of Gaoyou duck embryo osteoclasts cultured in vitro. Bone marrow cells were harvested from 23-d-old Gaoyou duck embryos and cultured in the presence of different concentrations of OPG (group A: no added factors, group B: 30 ng/mL of OPG, and group C: 100 ng/mL of OPG). Tartrate-resistant acid phosphatase (TRAP) staining, pit formation assay, and co-staining with tetramethylrhodamine isothiocyanate (TRITC)-conjugated phalloidin and Hoechst 33258 were all performed to determine the number of TRAP-positive cells, bone resorption activity, and the level of apoptosis, respectively. The number of TRAP-positive cells and the net expansion of pit formations area peaked on d 7 of culture in all 3 groups. The number of osteoclasts and the total volume of pit formations in OPG-treated groups were significantly lower compared with group A (P < 0.05). At each time point, the net expansion of pit formations area correlated with the number of TRAP-positive cells. The OPG inhibited the de novo formation of filamentous (F)-actin rings and promoted the disruption of existing F-actin rings in mature osteoclasts. In addition, OPG induced apoptosis in mature osteoclasts, as demonstrated by morphological changes in the nuclei. In osteoclast precursors, OPG inhibited differentiation and downregulated the formation of F-actin rings. In mature osteoclasts, OPG suppressed activation and enhanced the development of apoptosis, observed as a decrease in the number of TRAP-positive cells, the disruption of F-actin rings and morphological changes of the nuclei.


Subject(s)
Apoptosis/drug effects , Ducks/embryology , Osteoclasts/drug effects , Osteoprotegerin/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Osteoclasts/cytology , Osteoclasts/physiology
15.
Int J Mol Med ; 31(6): 1411-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23563320

ABSTRACT

The aim of the present study was to determine whether osteoprotegerin (OPG) influences the bone resorption activity of osteoclasts. RAW264.7 cells were induced by macrophage colony-stimulating factor (M-CSF) + receptor activator of nuclear factor-κB ligand (RANKL) and 0, 10, 20, 50 and 100 ng/ml OPG were added into various groups in the presence of the two cytokines. The OPG treatment was continued for 24 h. Osteoclast differentiation and activation were estimated via TRAP staining assay, TRITC-conjugated phalloidin staining, resorption activity analysis. Furthermore, the expression levels of the osteoclastic bone resorption-related genes MMP-9, cathepsin K and carbonic anhydrase II (CA II) were examined using real-time polymerase chain reaction (PCR). The data demonstrated that high concentrations of OPG could inhibit the differentiation and activation of osteoclasts. Furthermore, real-time PCR analysis illustrated that OPG decreased the expression of MMP-9 and cathepsin K in different concentrations of OPG and it decreased the expression of CA II genes at 10 and 20 ng/ml concentrations of OPG. For the time gradient study, OPG decreased the expression of MMP-9 and CA II genes but not that of the cathepsin K gene. In summary, the resorption activity of osteoclasts was suppressed by high concentrations of OPG and, at the molecular level, OPG decreased the expression of osteoclastic bone resorption-related genes.


Subject(s)
Bone Resorption/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/pharmacology , Acid Phosphatase/metabolism , Actins/metabolism , Animals , Bone Resorption/genetics , Carbonic Anhydrase II/genetics , Carbonic Anhydrase II/metabolism , Cathepsin K/genetics , Cathepsin K/metabolism , Cell Line , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Osteoclasts/cytology , RANK Ligand/metabolism , RNA, Messenger/genetics , Tartrate-Resistant Acid Phosphatase
16.
Zhong Yao Cai ; 34(10): 1615-7, 2011 Oct.
Article in Chinese | MEDLINE | ID: mdl-22372155

ABSTRACT

OBJECTIVE: To prepare the curcumin inclusion complex and study its stability. METHODS: Beta-cyclodextrin (beta-CD) was chosen as inclusion material and orthogonal design method was used. UV-Vis spectrophotometer was used to determine the solubility and content of different samples. They were placed under conditions of light and the solution containing Fe(3+) to determine their stability. RESULTS: The optimum preparation procedure were : curcumin and beta-cyclodextrin feed molar ratio of 1: 1, the concentration of ethanol was 40%, 40 degrees C inclusion temperature and 1 h reaction time. CONCLUSION: Inclusion complex prepared under eptimal condition is stable.


Subject(s)
Curcuma/chemistry , Curcumin/chemistry , Technology, Pharmaceutical/methods , beta-Cyclodextrins/chemistry , Curcumin/administration & dosage , Drug Carriers , Drug Compounding , Drug Stability , Ethanol/chemistry , Rhizome/chemistry , Solubility , Spectrophotometry, Ultraviolet , Temperature , Time Factors , beta-Cyclodextrins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...