Review
Does osteocytic SOST suppression mediate PTH bone anabolism?

https://doi.org/10.1016/j.tem.2009.12.002Get rights and content

Parathyroid hormone (PTH) has bone anabolic activity when administered intermittently, affecting cells of the osteoblastic lineage at various stages, yet much remains to be learned about precisely how PTH promotes osteoblastic bone formation. Recent discoveries revealed that PTH causes transcriptional suppression of the osteocyte marker gene SOST, which encodes the potent secreted bone formation inhibitor, sclerostin. This review addresses whether osteocytes, terminally differentiated cells of the osteoblastic lineage, which are entrapped within the mineralized bone matrix, contribute to PTH-induced bone formation responses via regulation of sclerostin levels, and discusses recent evidence on how the bone anabolic responses elicited by intermittent PTH treatment or by sclerostin inhibition overlap and diverge.

Section snippets

Intermittent parathyroid hormone (PTH) treatment is bone anabolic, whereas continuous treatment is bone catabolic

PTH is currently the only anabolic bone treatment option for patients with low bone mass conditions such as severe osteoporosis (OP) (Box 1). It has received much attention in recent years because of the finding that intermittent treatment (i.e. daily injection) causes a significant increase in bone mass 1, 2, 3. The intact hormone PTH (1–84) and its N-terminal fragment (1–34) have been developed as treatments for established osteoporosis. Nonetheless, despite PTH being a well-recognized bone

Osteocytes express PTH1R and secrete the bone formation inhibitor, sclerostin

Osteoblasts differentiate from mesenchymal bone marrow progenitor cells into bone forming cells that reside on the bone surface and deposit new bone matrix. A small fraction of osteoblasts further differentiates into osteocytes, terminally differentiated cells of the osteoblastic lineage, which represent >90% of all bone cells in the adult skeleton [17]. They reside within lacunae, small cavities within the mineralized bone matrix, and extend long, thin cellular protrusions termed dendrites,

PTH suppresses osteocytic Sost expression

The localization and abundance of osteocytes within bone, their expression of PTH1R and their ability to secrete the potent bone formation inhibitor sclerostin indicate their potential role as important regulators of bone formation. This hypothesis is supported by the findings that PTH suppresses Sost transcription in vitro 32, 33. Moreover, PTH treatment decreases Sost expression in vivo 32, 33, 34, resulting in fewer numbers of osteocytes expressing sclerostin 33, 34. Sost expression was

PTH-induced Sost inhibition contributes to PTH bone anabolic action in vivo

Recently, the role of PTH signaling in osteocytes in vivo was addressed in mice overexpressing a constitutively active PTH1R variant selectively in osteocytes, mimicking part of the effects triggered by continuous systemic PTH exposure [39]. In these mice, Sost expression is suppressed by about 80% and consistently, the number of sclerostin positive osteocytes is dramatically reduced. These mice further display a bone overgrowth phenotype characterized by elevated bone turnover caused by

PTH signaling activates canonical Wnt signaling

Transgenic mice selectively expressing constitutively active PTH1R in osteocytes display decreased Sost expression and increased canonical Wnt signaling [39]. As described, sclerostin is currently speculated to function as a secreted inhibitor of canonical Wnt/β-catenin signaling by binding to Lrp5 and Lrp6 Wnt co-receptors, preventing their association with the Wnt-Frizzled (Fzd) receptor complex [28] (Figure 2a). Interestingly, simultaneous deletion of Lrp5 abolished the increased bone mass

Intermittent PTH treatment and sclerostin inhibition: two converging and diverging bone anabolic principles

It is worth pointing out that bone anabolism induced by intermittent PTH treatment or sclerostin inhibition overlap in several, but not in all aspects. The bone forming effects of an anti-sclerostin antibody in rodents [53] resemble in some ways those of high-dose intermittent PTH treatment [54]. Either anabolic agent increases both the extent of bone forming surfaces and osteoblastic bone matrix synthesis. Likewise, all skeletal envelopes respond to bone anabolic intermittent PTH treatment and

Concluding remarks

PTH exerts complex and only partially elucidated cellular and molecular actions in bone, directly affecting bone marrow stromal cells and osteoblasts at all stages of their life cycle. In addition, PTH indirectly regulates osteoclast differentiation and activity through cells of the osteoblastic lineage via various paracrine cytokines and growth factors. Recent studies in mice demonstrate that bone anabolic pathways induced by PTH or sclerostin inhibition partially overlap, because loss or gain

Acknowledgements

We thank Frederic Bassilana and Klaus Seuwen for critically reviewing the manuscript.

References (79)

  • M.V. Semenov et al.

    LRP5 Mutations linked to high bone mass diseases cause reduced LRP5 binding and inhibition by SOST

    J. Biol. Chem.

    (2006)
  • V.K. Yadav

    Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum

    Cell

    (2008)
  • H. Keller et al.

    SOST is a target gene for PTH in bone

    Bone

    (2005)
  • M.A. Arnold

    MEF2C transcription factor controls chondrocyte hypertrophy and bone development

    Dev. Cell

    (2007)
  • A.G. Robling

    Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin

    J. Biol. Chem.

    (2008)
  • A. Moustafa

    The mouse fibula as a suitable bone for the study of functional adaptation to mechanical loading

    Bone

    (2009)
  • J.A. Robinson

    Wnt/β-catenin signaling is a normal physiological response to mechanical loading in bone

    J. Biol. Chem.

    (2006)
  • K. Sawakami

    The Wnt co-receptor LRP5 is essential for skeletal mechanotransduction but not for the anabolic bone response to parathyroid hormone treatment

    J. Biol. Chem.

    (2006)
  • D.A. Glass

    Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation

    Dev. Cell

    (2005)
  • S.L. Holmen

    Essential role of β-catenin in postnatal bone acquisition

    J. Biol. Chem.

    (2005)
  • E. Seeman et al.

    Reconstructing the skeleton with intermittent parathyroid hormone

    Trends Endocrinol. Metab.

    (2001)
  • J. Fox

    Effects of daily treatment with parathyroid hormone 1-84 for 16 months on density, architecture and biomechanical properties of cortical bone in adult ovariectomized rhesus monkeys

    Bone

    (2007)
  • S.L. Bonnick

    Osteoporosis in men and women

    Clin. Cornerstone

    (2006)
  • K. Henriksen

    Local communication on and within bone controls bone remodeling

    Bone

    (2009)
  • F. Liu

    CREM deficiency in mice alters the response of bone to intermittent parathyroid hormone treatment

    Bone

    (2007)
  • M.M. Hurley

    Impaired bone anabolic response to parathyroid hormone in Fgf2-/- and Fgf2+/- mice

    Biochem. Biophys, Res. Comm.

    (2006)
  • Terauchi

    T Lymphocytes amplify the anabolic activity of parathyroid hormone through Wnt10b signaling

    Cell Metabolism

    (2009)
  • N. Bonnet

    The matricellular protein Periostin is required for Sclerostin inhibition and the anabolic response to mechanical loading and physical activity

    J. Biol. Chem.

    (2009)
  • T. Sugiyama

    Mechanical loading enhances the anabolic effects of intermittent parathyroid hormone (1-34) on trabecular and cortical bone in mice

    Bone

    (2008)
  • M.D. Roberts

    Local bone formation due to combined mechanical loading and intermittent hPTH-(1-34) treatment and its correlation to mechanical signal distributions

    J. Biomech.

    (2009)
  • J. Compston

    Recombinant parathyroid hormone in the management of osteoporosis

    Calcif. Tiss. Int.

    (2005)
  • A.B. Hodsman

    Parathyroid hormone and teriparatide for the treatment of osteoporosis: a review of the evidence and suggested guidelines for its use

    Endocr. Rev.

    (2005)
  • M.R. Rubin et al.

    Parathyroid Hormone as an anabolic skeletal therapy

    Drugs

    (2005)
  • N.H. Kulkarni

    Effects of parathyroid hormone on wnt signaling pathway in bone

    J. Cell Biochem.

    (2005)
  • H. Dobnig

    Early changes in biochemical markers of bone formation correlate with improvements in bone structure during teriparatide therapy

    J. Clin. Endocrinol. Metab.

    (2005)
  • M.R. McClung

    Opposite bone remodeling effects of teriparatide and alendronate in increasing bone mass

    Arch. Intern. Med.

    (2005)
  • D.C. Bauer

    Short-term changes in bone turnover markers and BMD Response to parathyroid hormone in postmenopausal women with osteoporosis

    J. Clin. Endocrinol. Metab.

    (2006)
  • J.E. Onyia

    Molecular profile of catabolic versus anabolic treatment regimens of parathyroid hormone (PTH) in rat bone: an analysis by DNA microarray

    J. Cell Biochem.

    (2005)
  • L.F. Bonewald

    Osteocytes as dynamic multifunctional cells

    Ann. N. Y. Acad. Sci.

    (2007)
  • Cited by (114)

    • Receptors | Parathyroid hormone/parathyroid hormone-related protein receptor

      2021, Encyclopedia of Biological Chemistry: Third Edition
    • Parathyroid hormone

      2021, Advances in Clinical Chemistry
    • WNT signaling in skeletal homeostasis and diseases

      2020, Marcus and Feldman’s Osteoporosis
    View all citing articles on Scopus
    View full text