Icariin Stimulates hFOB 1.19 Osteoblast Proliferation and Differentiation via OPG/RANKL Mediated by the Estrogen Receptor

2020 ◽  
Vol 22 (1) ◽  
pp. 168-175 ◽  
Author(s):  
Lin-Jun Sun ◽  
Chong Li ◽  
Xiang-hao Wen ◽  
Lu Guo ◽  
Zi-Fen Guo ◽  
...  

Background:: Icariin (ICA), one of the main effective components isolated from the traditional Chinese herb Epimedium brevicornu Maxim., has been reported to possess extensive pharmacological actions, including enhanced sexual function, immune regulation, anti-inflammation, and antiosteoporosis. Methods:: Our study was designed to investigate the effect of ICA on cell proliferation and differentiation and the molecular mechanism of OPG/RANKL mediated by the Estrogen Receptor (ER) in hFOB1.19 human osteoblast cells. Results:: The experimental results show that ICA can stimulate cell proliferation and increase the activity of Alkaline Phosphatase (ALP), Osteocalcin (BGP) and I Collagen (Col I) and a number of calcified nodules. Furthermore, the mRNA and protein expression of OPG and RANKL and the OPG/ RANKL mRNA and protein expression ratios were upregulated by ICA. The above-mentioned results indicated that the optimal concentration of ICA for stimulating osteogenesis was 50ng/mL. Subsequent mechanistic studies comparing 50ng/mL ICA with an estrogen receptor antagonist demonstrated that the effect of the upregulated expression is connected with the estrogen receptor. In conclusion, ICA can regulate bone formation by promoting cell proliferation and differentiation and upregulating the OPG/RANKL expression ratio by the ER in hFOB1.19 human osteoblast cells.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4711-4711
Author(s):  
Jie Sun ◽  
He Huang ◽  
Weijun Zhong ◽  
Shan Fu

Abstract Abstract 4711 Promyelocytic leukemia protein (PML) was found in discrete, punctuate subnuclear structures known as PML nuclear bodies (PML NBs) in normal somatic cells and is an important factor acting downstream of wnt/β-catenin in the regulation of cell proliferation and differentiation. It is well-known that PML can bind cbp/p300 to promote TCF/LEF dependent transcription of downstream genes in 293T cells. However, its function in human mesenchymal stem cells (hMSCs) remains unclear. In our former studies, we found PML NBs in mitosis metaphase of hMSCs were more abundant than in Hela cells and 293T cells, which indicates PML may have special functions in hMSCs compared with tumor cells and normal somatic cells. The aim of our study is to investigate PML's function during proliferation and differentiation in hMSCs in contrast to Hela cells and 293T cells. hMSCs were isolated from bone marrow of healthy volunteers by density gradient centrifugation using Ficoll-paque and cultured (LG-DMEM, 10%FBS) with their characteristic adherence and morphology. hMSC immunophenotype was analyzed by flow cytometry and demonstrated uniform positivity for CD29, CD44, CD166 and CD105, and negativity for CD34, CD45 and HLA-DR. MTT assay and flow cytometry were used to evaluate proliferation of hMSCs. Osteogenic differentiation of hMSCs was perfoprmed and differentiated cells were identified with biochemical and morphological approaches. The levels of PML mRNA and protein expression during proliferation and differentiation were detected by RT-PCR, western blot and immunofluorescence while Hela cells and 293T cells were used as controls. Results showed the mean levels of mRNA and protein expression significantly increased during proliferation of hMSCs and 293T cells and positively correlated with the proliferation status of hMSCs. However, the expression of PML decreased in proliferating Hela cells (Fig. 1A, B). Intranuclear NBs of MSCs immunostained with PML monoclonal antibody and showed increased levels during proliferation, but remained very low in Hela cells throughout their proliferation. (Fig2). The above results indicate PML may take part in regulating the proliferation of hMSCs, and the mechanism involved is different from that in tumor cells. In tumor cells, the main function of PML maybe a suppressor of malignant cell growth. And PML protein is reduced or almost completely lost by post-transcriptional mechanisms. This loss is associated with tumor cell proliferation while regulation of cell proliferation and differentiation is the more important function of PML in hMSCs. During osteogenic differentiation of MSCs, ALP which is an early marker of osteogenic differentiation increased while PML's expression accordingly increased as detected by RT-PCR. (Fig 1C). Intranuclear NBs during differentiation are bigger but unequal in size compared with controls. (Fig3) These changes indicated that PML may interact with other molecules in NBs and act as a complex to regulate the process of osteogneic differentiation, but the exact mechanism involved is unclear and needs further study. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 21 (1) ◽  
Author(s):  
Gan Xiaoling ◽  
Liu Shuaibin ◽  
Liang Kailu

Abstract Background To investigated the role of miR-19b-3p in regulating bone marrow mesenchymal stem cell (BMSC) proliferation and osteoblast differentiation. Methods The expression of miR-19b-3p and lncRNA H19 were measured in postmenopausal osteoporosis patients and BMP-22 induced BMSCs using qRT-PCR. MiR-19b-3p mimic or inhibitor was transfected into BMP-2 induced BMSCs. Cell proliferation was measured by BrdU method. Protein expression of RUNX2 and COL1A1 were measured by western blot. PcDNA3.1-lncRNA H19 with or without miR-19b-3p mimic was transfected into BMP-2 induced BMSCs. Results The expression of miR-19b-3p was significantly up-regulated in postmenopausal osteoporosis patients and BMP-2 induced BMSCs. MiR-19b-3p overexpression dramatically elevated, while miR-19b-3p inhibition decreased cell proliferation of BMSCs. Additionally, protein expression levels of RUNX2 and COL1A1, as well as ALP activity were significantly promoted by miR-19b-3p mimic transfection and inhibited by miR-19b-3p inhibitor transfection. LncRNA H19 was obviously down-regulated in postmenopausal osteoporosis patients. H19 overexpression significantly decreased cell proliferation and differentiation by down-regulating miR-19b-3p. Moreover, the expression of miR-19b-3p was inhibited, while H19 elvated in 17β-estradiol (E2) treated BMSCs in a dose-dependent manner. Conclusion These data were the first to reveal the critical role of H19/miR-19b-3p in postmenopausal osteoporosis, and provided a new therapeutic target for OP.


1998 ◽  
Vol 333 (3) ◽  
pp. 645-654 ◽  
Author(s):  
Judit GARRIGA ◽  
Ana LIMÓN ◽  
Xavier MAYOL ◽  
Sushil G. RANE ◽  
Jeffrey H. ALBRECHT ◽  
...  

In the present study we have analysed the regulation of pocket protein expression and post-transcriptional modifications on cell proliferation and differentiation, both in vivo and in vitro. There are marked changes in pocket protein levels during these transitions, the most striking differences being observed between p130 and p107. The mechanisms responsible for regulating pocket protein levels seem to be dependent on both cell type and pocket protein, in addition to their dependence on the cell growth status. Changes in retinoblastoma protein and p107 levels are independent of their state of phosphorylation. However, whereas p130 phosphorylation to forms characteristic of quiescent/differentiated cells results in the accumulation of p130 protein, phosphorylation of p130 to one or more forms characteristic of cycling cells is accompanied by down-regulation of its protein levels. We also show here that the phosphorylation status and protein levels of p130 and p107 are regulated in vivo as in cultured cells. In vivo, changes in p130 forms are correlated with changes in E2F complexes. Moreover, the modulation of p130 and p107 status during cell differentiation in vitro is consistent with the patterns of protein expression and phosphorylation status found in mouse tissues. Thus in addition to the direct disruption of pocket protein/E2F complexes induced by cyclin/cyclin-dependent kinase, the results we report here indicate that the differential modulation of pocket protein levels constitutes a major mechanism that regulates the pool of each pocket protein that is accessible to E2F and/or other transcription factors.


2005 ◽  
Vol 63 (3) ◽  
pp. 362-369 ◽  
Author(s):  
Takahiro Kanno ◽  
Tetsu Takahashi ◽  
Toshiyuki Tsujisawa ◽  
Wataru Ariyoshi ◽  
Tatsuji Nishihara

Sign in / Sign up

Export Citation Format

Share Document