Elsevier

Bone

Volume 75, June 2015, Pages 96-104
Bone

Original Full Length Article
Estrogens antagonize RUNX2-mediated osteoblast-driven osteoclastogenesis through regulating RANKL membrane association

https://doi.org/10.1016/j.bone.2015.02.007Get rights and content

Highlights

  • Estradiol antagonizes RUNX2-mediated osteoblast-driven osteoclastogenesis.

  • RUNX2 regulates RANKL membrane association.

  • E2 counteracts RUNX2-driven RANKL membrane association.

  • SERMs mimic antagonistic action of E2 on RUNX2-induced osteoclastogenesis.

Abstract

In addition to its thoroughly investigated role in bone formation, the osteoblast master transcription factor RUNX2 also promotes osteoclastogenesis and bone resorption. Here we demonstrate that 17β-estradiol (E2), strongly inhibits RUNX2-mediated osteoblast-driven osteoclastogenesis in co-cultures. Towards deciphering the underlying mechanism, we induced premature expression of RUNX2 in primary murine pre-osteoblasts, which resulted in robust differentiation of co-cultured splenocytes into mature osteoclasts. This was attributable to RUNX2-mediated increase in RANKL secretion, determined by ELISA, as well as to RUNX2-mediated increase in RANKL association with the osteoblast membrane, demonstrated using confocal fluorescence microscopy. The increased association with the osteoblast membrane was recapitulated by transiently expressed GFP-RANKL. E2 abolished the RUNX2-mediated increase in membrane-associated RANKL and GFP-RANKL, as well as the concomitant osteoclastogenesis. RUNX2-mediated RANKL cellular redistribution was attributable in part to a decrease in Opg expression, but E2 did not influence Opg expression either in the presence or absence of RUNX2. Diminution of RUNX2-mediated osteoclastogenesis by E2 occurred regardless of whether the pre-osteoclasts were derived from wild type or estrogen receptor alpha (ERα)-knockout mice, suggesting that activated ERα inhibited osteoblast-driven osteoclastogenesis by acting in osteoblasts, possibly targeting RUNX2. Indeed, microarray analysis demonstrated global attenuation of the RUNX2 response by E2, including abrogation of Pstpip2 expression, which likely plays a critical role in membrane trafficking. Finally, the selective ER modulators (SERMs) tamoxifen and raloxifene mimicked E2 in abrogating the stimulatory effect of osteoblastic RUNX2 on osteoclast differentiation in the co-culture assay. Thus, E2 antagonizes RUNX2-mediated RANKL trafficking and subsequent osteoclastogenesis. Targeting RUNX2 and/or downstream mechanisms that regulate RANKL trafficking may lead to the development of improved SERMs and possibly non-hormonal therapeutic approaches to high turnover bone disease.

Introduction

Runx2 is as an osteoblast master regulator that is required for bone formation. Initially identified based on its interaction with the bone-specific Osteocalcin promoter in vitro [1], [2], [3], the pivotal role of Runx2 in osteogenesis in vivo was demonstrated by the absence of differentiated osteoblasts and failure of skeletal mineralization in Runx2-deficient mice [4], [5]. Furthermore, inhibition of Runx2 in vitro abrogates expression of osteoblast markers, and its forced expression in non-osteoblasts induces bone-like cellular phenotypes [2], [3]. Contrasting the role of Runx2 as a master regulator of osteoblast differentiation and embryonic bone development, its function in bone resorption is less appreciated. Over-expression of Runx2 in osteoblasts resulted in increased osteoclast differentiation from co-cultured pre-osteoclasts in vitro [6], [7], [8] and exaggerated bone resorption in vivo [6], [9]. Conversely, expression of a dominant negative RUNX2 decreased osteoclastogenesis in co-culture assays and decreased bone resorption in vivo [7]. Accordingly, osteoclast number decreased in mice with either global or osteoblast-specific Runx2 ablation [4], [10]. Thus, RUNX2 promotes both osteoblastogenesis and osteoblast-driven osteoclastogenesis.

Regulation of osteoclastogenesis by osteoblasts constitutes a fundamental principle in the coupling of bone resorption to bone formation [11]. Among osteoblast-borne signals mediating this coupling is the quintessential osteoclastogenic factor RANKL [12], [13], [14]. Although RUNX2 can increase RANKL mRNA levels in smooth muscle cells [15], stimulation of osteoclastogenesis by RUNX2 does not necessarily involve the regulation of Rankl mRNA levels in osteoblasts [8], [16], [17]. Indeed, using primary osteoblast cultures, the present work demonstrates that RUNX2 influences RANKL through regulating its association with the cell membrane without significantly affecting its mRNA expression.

Postmenopausal osteoporosis inflicts a pathological fracture on two in every five women over the age of fifty [18]. It is mostly attributable to reduced stimulation of estrogen receptor α (ERα) in osteoblasts and osteoclasts [19]. Accordingly, estrogens and selective estrogen receptor modulators (SERMs) constitute therapeutic options for the preservation of bone mass in postmenopausal women, and some SERMs have beneficial effects on the skeleton when used for the management of breast cancer [20]. Based on previous reports on inhibition of RUNX2 activity by ERα [21], as well as resistance to ovariectomy-induced bone loss in mice expressing of a dominant negative RUNX2 isoform [7], we hypothesized that activation of ERα in osteoblasts attenuates RUNX2-driven osteoclastogenic signal(s). Indeed, we show that estrogen signaling in osteoblasts abrogates RUNX2-mediated RANKL membrane association and differentiation of co-cultured splenocytes into mature osteoclasts.

Section snippets

Animals

C57BL/6 JAX® mice from Jackson Laboratory (Sacramento, CA) were used for the extraction of both osteoblasts and splenocytes without regard to mouse gender. Splenocytes were isolated from either wild type or ERα knockout (ERKO) animals. Mice were housed in microisolator-type cages at the vivaria of University of Southern California (USC) or University of California Los Angeles (UCLA). The respective Institutional Animal Care and Use Committees approved all experimental procedures with animals.

Reagents

Dox-inducible Runx2 expression in newborn mouse calvarial osteoblasts (NeMCOs)

RUNX2 promotes not only osteoblast differentiation and bone formation, but also osteoblast-driven osteoclastogenesis [6], [7], [8], [9], [10], [28]. Because estrogens inhibit RUNX2 activity [21], we asked whether they would inhibit osteoclast differentiation driven by expression of RUNX2 in co-cultured osteoblasts. First, we transduced newborn mouse calvarial osteoblasts (NeMCOs) with lentiviruses encoding doxycycline (dox)-inducible FLAG-RUNX2 [24] and treated the so-called NeMCO/Rx2dox cells

Discussion

It is well established that accelerated bone turnover increases fracture risk, with postmenopausal osteoporosis serving a prime example. Bone loss that occurs at physiological turnover rates is slow and usually transpires without pathological consequences because coupling mechanisms secure the replacement of most of the resorbed bone with newly deposited material. Adding to classical coupling mechanisms of signaling from osteoblasts to osteoclasts and back, the regulation of both osteoblast

Acknowledgments

This work was supported by grants RO1 DK071122 and RO1 DK071122S1 from NIDDK to BF, holder of the J. Harold and Edna L. LaBriola Chair in Genetic Orthopaedic Research. AM was supported by a training grant T32 DE021982 from NIDCR, and YG was supported by a Meyer Young Investigator Fellowship from The Arthritis Foundation. We thank Drs. Ebrahim Zandi (USC), Dr. Masashi Honma and Dr. Hiroshi Suzuki (University of Tokyo) for reagents, the UCLA Vector Core (supported by CURE/P30DK041301) for

References (62)

  • T. Nakashima et al.

    Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-kappaB ligand: modulation of the expression by osteotropic factors and cytokines

    Biochem Biophys Res Commun

    (2000)
  • V. Kartsogiannis et al.

    Localization of RANKL (receptor activator of NF kappa B ligand) mRNA and protein in skeletal and extraskeletal tissues

    Bone

    (1999)
  • I. Kalajzic et al.

    In vitro and in vivo approaches to study osteocyte biology

    Bone

    (2013)
  • S. Huang et al.

    The PR domain of the Rb-binding zinc finger protein RIZ1 is a protein binding interface and is related to the SET domain functioning in chromatin-mediated gene expression

    J Biol Chem

    (1998)
  • P. Ducy et al.

    Two distinct osteoblast-specific cis-acting elements control expression of a mouse osteocalcin gene

    Mol Cell Biol

    (1995)
  • C. Banerjee et al.

    Runt homology domain proteins in osteoblast differentiation: AML3/CBFA1 is a major component of a bone-specific complex

    J Cell Biochem

    (1997)
  • V. Geoffroy et al.

    High bone resorption in adult aging transgenic mice overexpressing cbfa1/runx2 in cells of the osteoblastic lineage

    Mol Cell Biol

    (2002)
  • Z. Maruyama et al.

    Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency

    Dev Dyn

    (2007)
  • S.K. Baniwal et al.

    Runx2 promotes both osteoblastogenesis and novel osteoclastogenic signals in ST2 mesenchymal progenitor cells

    Osteoporos Int

    (2011)
  • W. Liu et al.

    Overexpression of Cbfa1 in osteoblasts inhibits osteoblast maturation and causes osteopenia with multiple fractures

    J Cell Biol

    (2001)
  • M.D. Adhami et al.

    Loss of Runx2 in committed osteoblasts impairs postnatal skeletogenesis

    J Bone Miner Res

    (2014)
  • T. Suda et al.

    Modulation of osteoclast differentiation

    Endocr Rev

    (1992)
  • Y.Y. Kong et al.

    OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis

    Nature

    (1999)
  • H. Yasuda et al.

    Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL

    Proc Natl Acad Sci U S A

    (1998)
  • C.H. Byon et al.

    Runx2-upregulated receptor activator of nuclear factor kappaB ligand in calcifying smooth muscle cells promotes migration and osteoclastic differentiation of macrophages

    Arterioscler Thromb Vasc Biol

    (2011)
  • Services USDoHaH

    Bone health and osteoporosis: a report of the Surgeon General

    (2004)
  • S.C. Manolagas et al.

    The role of estrogen and androgen receptors in bone health and disease

    Nat Rev Endocrinol

    (2013)
  • S. Khosla

    Update on estrogens and the skeleton

    J Clin Endocrinol Metab

    (2010)
  • O. Khalid et al.

    Modulation of Runx2 activity by estrogen receptor-alpha: implications for osteoporosis and breast cancer

    Endocrinology

    (2008)
  • Y. Gabet et al.

    Developmentally regulated inhibition of cell cycle progression by glucocorticoids through repression of cyclin A transcription in primary osteoblast cultures

    J Cell Physiol

    (2011)
  • N. Udagawa et al.

    Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells

    Proc Natl Acad Sci U S A

    (1990)
  • Cited by (44)

    • Multiple roles of Runt-related transcription factor-2 in tooth eruption: bone formation and resorption

      2022, Archives of Oral Biology
      Citation Excerpt :

      Meanwhile, in the co-culture system, Runx2 induces RANKL secretion in osteoblast lineage cells and promotes the osteoclastogenesis from splenocytes (Geoffroy et al., 2002) or bone marrow osteoclast progenitors (Enomoto et al., 2003; Maruyama et al., 2007). This is due to Runx2 binding to the RANKL gene and transactivating its expression (Martin et al., 2015; O'Brien, 2010). RUNX2 exerts a pivotal influence on osteoclastogenesis by regulating the expression of OPG and RANKL and orchestrating the RANKL/RANK/OPG system (Fig. 3).

    • Withanolide B promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via ERK1/2 and Wnt/β-catenin signaling pathways

      2020, International Immunopharmacology
      Citation Excerpt :

      Osteoblasts are one of the most important cells in bone metabolism, and the process of osteogenic differentiation of stem cells involves many signaling pathways. Among them, signal transduction pathways such as ER signaling pathway [24], OPG/RANKL signaling pathway [25], Wnt signaling pathway [26], and MAPK signaling pathway [27], are involved in osteogenic differentiation [28]. As mentioned before, Withanolides regulate osteoclastogenesis through the NF-κB signaling pathway.

    • Osteoporosis in men: What is similar and what is different?

      2020, Marcus and Feldman’s Osteoporosis
    • Estrogens and selective estrogen receptor modulators differentially antagonize Runx2 in ST2 mesenchymal progenitor cells

      2018, Journal of Steroid Biochemistry and Molecular Biology
      Citation Excerpt :

      We next assessed the effect of E2 on development of the osteoblast phenotype in primary NeMCO cultures based on the activity of alkaline phosphatase (ALP). Contrasting the primarily negative regulation of Runx2 at the level of gene expression (Ref. [6] and Supplemental Fig. 1), E2 did not inhibit ALP activity in this culture model at either physiological (0.1–1 nM; Fig. 2A) or higher concentrations (data not shown). Similarly, E2 did not inhibit ALP activity in the MC3T3-E1 culture model of committed osteoblasts (Fig. 2B).

    View all citing articles on Scopus
    1

    Present Address: Department of Orthopaedic Surgery, University of Tennessee Health Science Center, 19 S. Manassas St, Memphis, TN 38163, USA.

    View full text