Parathyroid hormone: anabolic and catabolic actions on the skeleton

https://doi.org/10.1016/j.coph.2015.03.005Get rights and content

Highlights

  • Parathyroid hormone has dual anabolic and catabolic actions on the skeleton.

  • The OPG–RANKL–RANK pathway is the main mediator of the catabolic actions of PTH.

  • PTH inhibits sclerostin, a major mechanism that leads to its osteoanabolic actions.

  • Osteocytes are critical effectors of PTH actions to increase bone remodeling.

  • PTH-induced bone formation may occur independent of bone resorption, through a mechanism that stimulates bone modeling.

Parathyroid hormone (PTH) is essential for the maintenance of calcium homeostasis through, in part, its actions to regulate bone remodeling. While PTH stimulates both bone formation and bone resorption, the duration and periodicity of exposure to PTH governs the net effect on bone mass, that is whether it is catabolic or anabolic. PTH receptor signaling in osteoblasts and osteocytes can increase the RANKL/OPG ratio, increasing both osteoclast recruitment and osteoclast activity, and thereby stimulating bone resorption. In contrast, PTH-induced bone formation is explained, at least in part, by its ability to downregulate SOST/sclerostin expression in osteocytes, permitting the anabolic Wnt signaling pathway to proceed. The two modes of administration of PTH, that is, continuous vs. intermittent, can regulate, in bone cells, different sets of genes; alternatively, the same sets of genes exposed to PTH in sustained vs. transient way, will favor bone resorption or bone formation, respectively. This article reviews the effects of PTH on bone cells that lead to these dual catabolic and anabolic actions on the skeleton.

Introduction

Parathyroid hormone (PTH) is an 84-amino acid peptide hormone synthesized in the chief cells of the parathyroid glands. It is essential for the maintenance of serum calcium concentration within narrow limits through direct actions on bone and kidney, and indirectly through actions on the gastrointestinal tract [1]. PTH also regulates phosphorus metabolism [2]. It decreases serum phosphorus levels through the inhibition of renal phosphate reabsorption in both proximal and distal tubules, although the proximal effect is quantitatively the more important [3].

PTH is released from parathyorid cells tonically, with circadian dynamics and in a stochastically pulsatile fashion. The synthesis and secretion of PTH are controlled by the calcium-sensing receptor (CaSR) expressed in the parathyroid cell membrane [4]. The signal for PTH production and secretion is a reduced extracellular ionized calcium concentration, while the signal for a reduction in PTH production and secretion is an increase in extracellular ionized calcium concentration. In these two situations, the signaling pathways triggered or inhibited by the CaSR are governed by the state of occupancy of CaSR by calcium ion. Of less importance, PTH secretion can also be stimulated by an increase in phosphorus levels either directly or through a stoichiometric reduction in calcium levels [3].

One of the key mechanisms by which PTH regulates calcium homeostasis is related to its actions to stimulate bone remodeling, a feat that is achieved by the direct actions of PTH on osteoblasts and osteocytes, and indirect effects on osteoclasts through its actions on osteoblasts and osteocytes. While PTH stimulates both bone resorption and bone formation, the final outcome on bone mass, either catabolic or anabolic, will depend on the dose and periodicity of the PTH signal. Continuous exposure to PTH result in catabolic effects on the skeleton, while intermittent, low doses of PTH result in osteoanabolic effects [5]. In human subjects, the catabolic effect of PTH is best represented by the classic disorder of PTH excess, primary hyperparathyroidism (PHPT). Even in the asymptomatic form of this disease, bone loss can be appreciated in both cortical and trabecular compartments of the skeleton [6, 7, 8•, 9, 10•]. Conversely, the foreshortened amino terminal peptide of PTH, teriparatide [PTH(1–34)] and the full length molecule [PTH(1–84)] are osteoanabolic when administered once daily in low doses for the treatment of osteoporosis [11, 12].

PTH actions are mediated primarily by a PTH receptor known as PTH1R. The two modes of administration of PTH, that is, continuous vs. intermittent, can regulate, in bone cells, different sets of genes or, alternatively, the same sets of genes in a sustained vs. transient manner, favoring bone resorption or bone formation, respectively [13, 14]. This article reviews the effects of PTH on bone cells that lead to these dual catabolic and anabolic actions on the skeleton.

Section snippets

PTH receptor and PTH signaling

The amino-terminal domain of PTH interacts with the PTH1R, a G-protein-coupled receptor encoded by a 14-exon gene located on chromosome 3. PTH1R is expressed on the surface of osteoblasts and osteocytes in bone, and tubular cells in the kidney [15, 16]. Stimulation of the PTH1R leads to the Gαs-mediated activation of the adenylyl cyclase/cyclic AMP (cAMP)/protein kinase A (PKA) signaling pathway [17]. The PTH1R is also coupled to Gαq-mediated activation of the phospholipase/protein kinase C

Catabolic actions of PTH: increased bone resorption

The PTH-induced increase in bone resorption is mediated, in vivo, by increased activity of the bone-resorbing cell, the osteoclast. However, in vitro and in vivo studies indicate that PTH does not directly activate osteoclasts; rather, the effect of PTH to enhance bone resorption appears to be indirect, through its actions on osteoblasts and osteocytes [26, 27, 28, 29].

Anabolic actions of PTH: increase in bone formation

Pre-clinical studies have demonstrated that the intermittent administration of PTH has anabolic effects on the skeleton [56, 57, 58]. These observations led to the clinical investigation of the biologically active but foreshortened amino-terminal fragment of PTH, PTH(1–34) and, later, the full length hormone [PTH(1–84)] as an anabolic approach to the treatment for osteoporosis. In fact, PTH therapy was eventually demonstrated to be effective for the treatment of osteoporosis [11, 12],

PTH actions on osteoblasts

Receptors for PTH are found in preosteoblasts, osteoblasts, lining cells, and osteocytes [3]. Intermittent PTH administration directly acts on osteoblasts to promote osteoblastogenesis, reduce osteoblast apoptosis, and re-activates quiescent lining cells [66, 67, 68]. An increase in osteoblast differentiation rather than osteoblast proliferation appears to be the main mechanism by which PTH stimulates osteoblastogenesis. Indeed, PTH arrests the cell cycle progression of osteoblasts, increasing

Sclerostin

Sclerostin is a secreted glycoprotein, primarily produced by osteocytes, that acts as an inhibitor of bone formation [81, 82]. Reduced sclerostin concentration and/or activity in human subjects leads to two genetic diseases known as van Buchem's Disease and sclerosteosis, characterized by generalized and progressive overgrowth of bone and sclerosis of the skeleton [83, 84, 85]. In mice, deletion or overexpression of the Sost gene encoding sclerostin leads, respectively, to high bone mass or

T-cells play a permissive role in anabolic and catabolic actions of PTH

T lymphocytes express the PTH1R, and may have a role in both the catabolic and anabolic actions of PTH in bone [123, 124]. Continuous PTH treatment of mice lacking T cells failed to increase osteoclast formation, bone resorption and cortical bone loss [125]. The lack of PTH-induced bone resorption in these mice appeared to be mediated by the CD40 ligand, a surface molecule present on activated T cells. This surface molecule induces the CD40 signaling in stromal cells, that stimulates RANKL/OPG

Is bone resorption required for the PTH-induced bone anabolism?

Evidence from pre-clinical and clinical studies suggests that osteoclastic resorption is required for the osteoanabolic effect of PTH [54, 130, 131]. In contrast, studies of Rhee et al. [132] suggest that PTH-induced bone formation may be distinctly affected by bone resorption depending upon the bone compartment. In this study, transgenic mice with increased bone mass due to activation of PTH receptor signaling in osteocytes were either treated with alendronate to block resorption-dependent

Conclusion

This review has focused on the catabolic and anabolic actions of PTH on the skeleton. Direct effects of PTH on osteoblasts and osteocytes, and indirect actions on osteoclasts, promote both bone formation and bone resorption, and the final effect on bone mass, either anabolic or catabolic, appear to depend on the duration and periodicity of the PTH exposure. While PTH stimulates bone remodeling overall, bone resorption predominates when continuous exposure to high levels of PTH ensues, whereas

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Conflict of interest statement

Nothing declared.

Acknowledgement

NIH grants: DK32333 and DK069350 to JPB (PI).

References (133)

  • 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)
  • Y. Rhee et al.

    PTH receptor signaling in osteocytes governs periosteal bone formation and intracortical remodeling

    J Bone Miner Res

    (2011)
  • T. Nakashima et al.

    Evidence for osteocyte regulation of bone homeostasis through RANKL expression

    Nat Med

    (2011)
  • J. Xiong et al.

    Matrix-embedded cells control osteoclast formation

    Nat Med

    (2011)
  • L.S. Stilgren et al.

    Skeletal changes in osteoprotegerin and receptor activator of nuclear factor-kappaB ligand mRNA levels in primary hyperparathyroidism: effect of parathyroidectomy and association with bone metabolism

    Bone

    (2004)
  • H. Patel et al.

    Serum monocyte chemokine protein-1 levels before and after parathyroidectomy in patients with primary hyperparathyroidism

    Endocr Pract

    (2014)
  • T.J. Wronski et al.

    Parathyroid hormone is more effective than estrogen or bisphosphonates for restoration of lost bone mass in ovariectomized rats

    Endocrinology

    (1993)
  • M.R. Rubin et al.

    Parathyroid hormone as an anabolic skeletal therapy

    Drugs

    (2005)
  • R.L. Jilka

    Molecular and cellular mechanisms of the anabolic effect of intermittent PTH

    Bone

    (2007)
  • M. Ogita et al.

    Differentiation and proliferation of periosteal osteoblast progenitors are differentially regulated by estrogens and intermittent parathyroid hormone administration

    Endocrinology

    (2008)
  • T. Ishizuya et al.

    Parathyroid hormone exerts disparate effects on osteoblast differentiation depending on exposure time in rat osteoblastic cells

    J Clin Invest

    (1997)
  • M. Schnoke et al.

    Parathyroid hormone suppresses osteoblast apoptosis by augmenting DNA repair

    Bone

    (2009)
  • S. Sharma et al.

    Cell cycle and apoptosis regulatory protein (CARP)-1 is expressed in osteoblasts and regulated by PTH

    Biochem Biophys Res Commun

    (2013)
  • H. Dobnig et al.

    Evidence that intermittent treatment with parathyroid hormone increases bone formation in adult rats by activation of bone lining cells

    Endocrinology

    (1995)
  • K.E. Poole et al.

    Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation

    FASEB J

    (2005)
  • A.G. Costa et al.

    Sclerostin: therapeutic horizons based upon its actions

    Curr Osteoporos Rep

    (2012)
  • Y. Cui et al.

    Lrp5 functions in bone to regulate bone mass

    Nat Med

    (2011)
  • M.K. Chang et al.

    Reversing LRP5-dependent osteoporosis and SOST-deficiency induced sclerosing bone disorders by altering WNT signaling activity

    J Bone Miner Res

    (2014)
  • D.A. Hanley et al.

    Pharmacological mechanisms of therapeutics: parathyroid hormone

  • R. Civitelli et al.

    Calcium and phosphate homeostasis: concerted interplay of new regulators

    J Endocrinol Invest

    (2011)
  • F.R. Bringhurst et al.

    Hormones and disorders of mineral metabolism

  • H. Dobnig et al.

    The effects of programmed administration of human parathyroid hormone fragment (1–34) on bone histomorphometry and serum chemistry in rats

    Endocrinology

    (1997)
  • M.R. Rubin et al.

    The natural history of primary hyperparathyroidism with or without parathyroid surgery after 15 years

    J Clin Endocrinol Metab

    (2008)
  • S. Hansen et al.

    Effects on bone geometry, density, and microarchitecture in the distal radius but not the tibia in women with primary hyperparathyroidism: a case–control study using HR-pQCT

    J Bone Miner Res

    (2010)
  • E.M. Stein et al.

    Primary hyperparathyroidism is associated with abnormal cortical and trabecular microstructure and reduced bone stiffness in postmenopausal women

    J Bone Miner Res

    (2013)
  • S.J. Silverberg et al.

    Current issues in the presentation of asymptomatic primary hyperparathyroidism: proceedings of the Fourth International Workshop

    J Clin Endocrinol Metab

    (2014)
  • J.P. Bilezikian et al.

    Guidelines for the management of asymptomatic primary hyperparathyroidism: summary statement from the Fourth International Workshop

    J Clin Endocrinol Metab

    (2014)
  • R.M. Neer et al.

    Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis

    N Engl J Med

    (2001)
  • S.L. Greenspan et al.

    Effect of recombinant human parathyroid hormone (1–84) on vertebral fracture and bone mineral density in postmenopausal women with osteoporosis: a randomized trial

    Ann Intern Med

    (2007)
  • J.E. Onyia et al.

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

    J Cell Biochem

    (2005)
  • R.M. Locklin et al.

    Mediators of the biphasic responses of bone to intermittent and continuously administered parathyroid hormone

    J Cell Biochem

    (2003)
  • N.S. Datta et al.

    PTH and PTHrP signaling in osteoblasts

    Cell Signal

    (2009)
  • B. Fermor et al.

    PTH/PTHrP receptor expression on osteoblasts and osteocytes but not resorbing bone surfaces in growing rats

    J Bone Miner Res

    (1995)
  • S. Kousteni et al.

    The cell biology of parathyroid hormone in osteoblasts

    Curr Osteoporos Rep

    (2008)
  • Q. Fu et al.

    Parathyroid hormone stimulates receptor activator of NFkappa B ligand and inhibits osteoprotegerin expression via protein kinase A activation of cAMP-response element-binding protein

    J Biol Chem

    (2002)
  • I. Kramer et al.

    Does osteocytic SOST suppression mediate PTH bone anabolism?

    Trends Endocrinol Metab

    (2010)
  • P.M. McSheehy et al.

    Osteoblastic cells mediate osteoclastic responsiveness to parathyroid hormone

    Endocrinology

    (1986)
  • H. Kaji et al.

    Involvement of dual signal transduction systems in the stimulation of osteoclast-like cell formation by parathyroid hormone and parathyroid hormone-related peptide

    Biochem Biophys Res Commun

    (1993)
  • J. Xiong et al.

    Osteocyte RANKL: new insights into the control of bone remodeling

    J Bone Miner Res

    (2012)
  • M. Kanzawa et al.

    Involvement of osteoprotegerin/osteoclastogenesis inhibitory factor in the stimulation of osteoclast formation by parathyroid hormone in mouse bone cells

    Eur J Endocrinol

    (2000)
  • Cited by (328)

    • Phosphate: An underrated component of primary hyperparathyroidism

      2024, Best Practice and Research: Clinical Endocrinology and Metabolism
    View all citing articles on Scopus
    View full text