Elsevier

Bone

Volume 46, Issue 2, February 2010, Pages 274-280
Bone

Review
NOTCHing the bone: Insights into multi-functionality

https://doi.org/10.1016/j.bone.2009.05.027Get rights and content

Abstract

Evolutionarily conserved Notch signaling plays a critical role during embryonic and postnatal life. The importance of Notch signaling in the determination of cell fate, and the spatio-temporal regulation of proliferation, differentiation and apoptosis has been demonstrated in various different organ systems. However, how Notch signaling affects the bone development was unknown until now. The in vivo effects of Notch signaling in lineage commitment, bone formation and bone resorption were demonstrated in recent studies. In addition to regulation of osteoblastogenesis, osteoblast directed osteoclastogenesis by Notch signaling revealed a dimorphic effect for this signaling pathway providing another example of such in bone development. Moreover, identification of the cross talk between the hematopoietic stem cell niche and osteoblasts through Notch signaling also suggested another important role for Notch signaling, i.e., the coupling of cellular components of the bone microenvironment. The association between the gain and loss of function of Notch activity in bone pathology highlights Notch as a potentially novel therapeutic target for the treatment of metabolic bone disease and bone cancer. In this review, we will focus primarily on the regulation of bone cells, i.e., osteoblasts and osteoclasts by Notch signaling. We will also review the importance of Notch in specifying bone–hematopoietic stem cell niche interactions within the bone microenvironment. Finally, we will discuss potential clinical implications and future directions for this field.

Section snippets

Mammalian Notch signaling

Evolutionarily conserved Notch signaling plays an important role in developmental processes and adult tissue homeostasis by regulating cell fate determination, proliferation, differentiation and apoptosis in a spatio-temporal manner. Altered Notch signaling has been associated with many different diseases including cancers of epithelial and hematopoietic origins. The Notch receptor and its ligands are transmembrane proteins whose signaling requires cell to cell contact between neighboring

Bone formation and remodeling

Skeleton forms through two different mechanisms; endochondral and intramembranous ossification [6], [7]. During endochondral ossification, cells located in the center of mesenchymal condensations differentiate into chondrocytes. Later, chondrocytes at the growth plate undergo well-ordered and controlled phases of cell proliferation, maturation, and apoptosis in order to form the future skeletal elements [8]. During intramembranous ossification, mesenchymal cells give rise to pre-osteoblasts to

Notch signaling during skeletogenesis

Until recently, the role of Notch signaling during skeletogenesis has mainly been limited to its role in patterning and somitogenesis. The involvement of the Notch-signaling pathway in somitogenesis was first revealed by the finding that Notch1 and its ligand Dll1 were highly expressed in the presomitic mesoderm (PSM) of mouse embryos. Subsequently, additional cycling genes, such as Lunatic fringe (Lfng), Hes7, and Hes1 were identified in different species to be components of this pathway [9],

Notch regulates osteoblast commitment, proliferation and differentiation

The importance of Notch function in osteoblast was demonstrated in vivo by studies in which tissue specific gain and loss of function mutants were generated. We generated an osteoblast specific over-expression of Notch1 ICD by using 2.3 kb collagen type 1 (Col1a1) promoter to drive the expression of activated Notch receptor specifically in committed osteoblasts [29]. These transgenic mice showed a dramatic increase in osteoblast number, proliferation and formation resulting in a severe

Notch regulates osteoclastogenesis directly and indirectly

The animal models generated above indicated that Notch could also regulate osteoblast dependent formation of osteoclasts. It has been shown that in PNN mice while RANKL expression was up-regulated, Opg levels were decreased. Osteoblast specific deletion of Presenilins via the 2.3 kb Col1a1-Cre resulted in decreased levels of Opg mRNA and protein as well. This demonstrates that Notch signaling may regulate osteoclastogenesis in a non-cell-autonomous manner through Opg regulation in osteoblasts (

Osteoblast regulation of hematopoietic stem cells via Notch signaling

In bone marrow, osteoblasts of the trabecular bone are in close physical association with hematopoietic stem cells and blood vessels, suggesting that the bone marrow microenvironment may provide regulatory signals for hematopoietic cells. Emerging data support the notion that osteoblasts lining the endosteal surface of trabecular bone can provide important cues to regulate/support the hematopoietic stem cell (HSC) niche. Transgenic mice expressing Parathyroid Hormone Receptor (PTH1R) under the

Notch as a therapeutic target for bone diseases

Loss of function studies of Notch signaling indicated an age dependent osteoporotic phenotype in mice. There are few anabolic bone agents for the treatment of osteoporosis, with most therapies targeted at inhibition of bone resorption. Up-regulation of Notch signaling may represent a potential approach for increasing bone formation over bone resorption as well as for inhibiting osteoclastogenesis. However, Notch's effects on other cellular compartments such as the mesenchymal stem cell pool

Future directions

Notch has a dual function in bone development in regulating both osteoblastogenesis and osteoclastogenesis. Cells in osteoclast and osteoblast lineages communicate with each other through cell to cell contact. Although osteoblast dependent activation of osteoclasts through Notch signaling has been demonstrated, whether there is reverse signaling between these two types of bone cells needs to be further investigated. Such cross talk and coupling might also exist within the bone marrow

References (46)

  • T. Yamada et al.

    Regulation of osteoclast development by Notch signaling directed to osteoclast precursors and through stromal cells

    Blood

    (2003)
  • K. Nakanishi et al.

    Notch signaling is required for the chondrogenic specification of mouse mesencephalic neural crest cells

    Mech. Dev.

    (2007)
  • H. Enomoto et al.

    Induction of osteoclast differentiation by Runx2 through receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin regulation and partial rescue of osteoclastogenesis in Runx2−/− mice by RANKL transgene

    J. Biol. Chem.

    (2003)
  • S. Bai et al.

    NOTCH1 regulates osteoclastogenesis directly in osteoclast precursors and indirectly via osteoblast lineage cells

    J. Biol. Chem.

    (2008)
  • C. Zhao et al.

    Bidirectional ephrinB2–EphB4 signaling controls bone homeostasis

    Cell Metab.

    (2006)
  • S. Lymperi et al.

    Strontium can increase some osteoblasts without increasing hematopoietic stem cells

    Blood

    (2008)
  • D. Edbauer et al.

    Reconstitution of gamma-secretase activity

    Nat. Cell Biol.

    (2003)
  • P. Zhang et al.

    Critical role of notch signaling in osteosarcoma invasion and metastasis

    Clin. Cancer Res.

    (2008)
  • C. Colnot

    Cellular and molecular interactions regulating skeletogenesis

    J. Cell. Biochem.

    (2005)
  • R.A. Conlon et al.

    Notch1 is required for the coordinate segmentation of somites

    Development

    (1995)
  • C. Oka et al.

    Disruption of the mouse RBP-J kappa gene results in early embryonic death

    Development

    (1995)
  • P.D. Turnpenny et al.

    Abnormal vertebral segmentation and the notch signaling pathway in man

    Dev. Dyn.

    (2007)
  • L. Li et al.

    Alagille syndrome is caused by mutations in human Jagged1, which encodes a ligand for Notch1

    Nat. Genet.

    (1997)
  • Cited by (70)

    • ADAM10 is indispensable for longitudinal bone growth in mice

      2020, Bone
      Citation Excerpt :

      Consistently, our gene expression analysis revealed that the transcripts of molecules downstream of Notch signaling, including Hes1, Hey1, and Heyl, were all expressed at lower levels in Adam10ΔCol2 mice than those in Ctrl mice (Fig. 3). The role of Notch signaling in chondrogenesis and skeletal development has been widely investigated [24–27]. The manipulation of Notch signaling in vivo proves that Notch signaling plays a crucial role in skeletal development.

    View all citing articles on Scopus
    View full text