Runx-2 is not essential for the vitamin D-regulated expression of RANKL and osteoprotegerin in osteoblastic cells

https://doi.org/10.1016/j.bbrc.2004.09.101Get rights and content

Abstract

The differentiation and activity of osteoclasts are positively and negatively controlled by receptor activator of nuclear factor-κB ligand (RANKL), which is expressed on the surface of osteoblasts and stromal cells, and its decoy receptor osteoprotegerin (OPG), which is secreted by osteoblasts and stromal cells, respectively. The expression of the genes for RANKL and OPG is regulated by 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3]. Runt-related gene-2 (Runx-2) is essential for osteoblast differentiation and there are several reports that Runx-2 is involved in osteoclast formation. Therefore, to clarify the role of Runx-2 in osteoclastogenesis, we designed a series of experiments using C2 cells and C6 cells, which are derived from calvariae of runx2-deficient mice. Treatment of C2 cells and C6 cells with 1α,25(OH)2D3 for 2–4 days increased and decreased the levels of expression of the mRNAs for RANKL and OPG, respectively, and the effects were dose-dependent. However, by day 8, the level of RANKL mRNA had fallen and that of OPG mRNA had risen. Furthermore, C6 cells induced the differentiation of mouse spleen cells into tartrate-resistant acid phosphatase-positive (TRAP-positive) multinucleated cells (osteoclast-like cells) in the presence of 10−7 M 1α,25(OH)2D3. Such formation of osteoclast-like cells was inhibited by exogenous OPG in a dose-dependent manner. Thus, our findings indicate that Runx-2 is not essential for the expression of RANKL and OPG, and the formation of osteoclast-like cells.

Section snippets

Materials and methods

Materials. α-Modified minimum essential medium (α-MEM), fetal bovine serum, and penicillin/streptomycin antibiotic mixture were purchased from Life Technologies (Grand Island, NY, USA). 1α,25(OH)2D3 and ISOGEN were purchased from Wako Pure Chemicals (Osaka, Japan). Recombinant human OPG was obtained from PeproTech EC (London, UK).

Reverse transcriptase-polymerase chain reaction. We used reverse transcriptase-polymerase chain reaction (RT-PCR) to detect mRNAs for Runx-2, RANKL, and OPG in runx-2

Results

Both C2 cells and C6 cells were established from calvariae of runx-2-deficient mice. Although these cells exhibit very low level of alkaline phosphatase activity and no osteocalcin synthesis when they were cultured in the control medium, the treatment with BMP-2 increased alkaline phosphatase activity and induced osteocalcin synthesis in these cells, indicating that C2 and C6 cells are capable of differentiating into osteoblastic cells in the presence of BMP-2 (unpublished data). Analysis by

Discussion

The formation of osteoclasts from hematopoietic cells is regulated by various factors, such as RANKL, OPG, and M-CSF, that are produced by cells of the osteoblastic lineage. Recently, we demonstrated that premyoblastic C2C12 cells differentiate into osteoblast-like cells upon treatment with bone morphogenetic protein-2 (BMP-2) and synchronously induce the formation of osteoclasts from spleen cells in vitro [29]. These observations suggest that osteoblastic differentiation might be associated

Acknowledgments

This work was supported by Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Sports and Culture of Japan and by grants from the Japan Science Society and the Smoking Research Foundation.

References (32)

Cited by (22)

  • The role of vitamin D in the endocrinology controlling calcium homeostasis

    2017, Molecular and Cellular Endocrinology
    Citation Excerpt :

    RANKL action can be antagonized by osteoprotegerin (OPG), a secreted protein produced by osteoblasts that binds to RANK, blocks RANKL binding to RANK, and prevents RANKL-mediated activation of osteoclast differentiation. 1,25(OH)2 D activates this system by inducing RANKL expression (Kitazawa et al., 2008) and suppressing OPG expression (Notoya et al., 2004; Lee et al., 2002). Thus, VDR deletion in osteoblasts could increase bone mass by reducing osteoclast production.

  • Control of RANKL gene expression

    2010, Bone
    Citation Excerpt :

    Specifically, expression of a dominant-negative form of Runx2 in a stromal/osteoblastic cell line had no effect on RANKL expression [96]. Moreover, even though osteoclasts are reduced in Runx2-deficient mice, cell lines derived from these mice express RANKL in response to 1,25(OH)2D3[38,99]. Consistent with this, mouse embryonic fibroblasts (MEFs) from Runx2-deficient mice and wild type littermates express equivalent amounts of RANKL in response to activation of the cAMP-PKA pathway [100].

  • Commitment to the osteoblast lineage is not required for RANKL gene expression

    2009, Journal of Biological Chemistry
    Citation Excerpt :

    Another possibility is that Runx2 is required for expression of RANKL in response to stimuli that were not addressed in our study. However, the present work, together with previous studies examining the requirement of Runx2 for stimulation of RANKL by 1,25(OH)2D3 (28), demonstrates that Runx2 is not required for control of RANKL by two of the major pathways known to control this gene in bone. In conclusion, we have shown that commitment to the osteoblast lineage is not a prerequisite for the ability of fibroblastic cell types to express RANKL and that mature osteoblasts are not a significant source of RANKL in bone.

  • EBF2 regulates osteoblast-dependent differentiation of osteoclasts

    2005, Developmental Cell
    Citation Excerpt :

    However, Opg is efficiently expressed in a Runx2−/− calvaria-derived cell line, and forced expression of Runx2 via adenoviral transduction of these cell results in suppression of Opg and activation of Rankl expression (Enomoto et al., 2003). In addition, it has been shown that Runx2 is not essential for vitamin D-regulated expression of Rankl and Opg in osteoblastic cells (Notoya et al., 2004). Moreover, our transfection experiments in HeLa cells indicate that Runx2 has only a modest effect on the activity of the OPG promoter construct, although Runx2 could augment the expression of the endogenous OPG gene.

View all citing articles on Scopus
View full text