J Bone Metab.  2014 Feb;21(1):8-20.

Bone Biology and Anabolic Therapies for Bone: Current Status and Future Prospects

Affiliations
  • 1Department of Medicine, St Vincent's Institute of Medical Research, University of Melbourne, Melbourne, VIC, Australia. jmartin@svi.edu.au

Abstract

Bone is continuously remodelled at many sites asynchronously throughout the skeleton, with bone formation and resorption balanced at these sites to retain bone structure. Negative balance resulting in bone loss and osteoporosis, with consequent fractures, has mainly been prevented or treated by anti-resorptive drugs that inhibit osteoclast formation and/or activity, with new prospects now of anabolic treatments that restore bone that has been lost. The anabolic effectiveness of parathyroid hormone has been established, and an exciting new prospect is presented of neutralising antibody against the osteocyte protein, sclerostin. The cellular actions of these two anabolic treatments differ, and the mechanisms will need to be kept in mind in devising their best use. On present evidence it seems likely that treatment with either of these anabolic agents will need to be followed by anti-resorptive treatment in order to maintain bone that has been restored. No matter how effective anabolic therapies for the skeleton become, it seems highly likely that there will be a continuing need for safe, effective anti-resorptive drugs.

Keyword

Anabolic agents; Bone and bones; Bone density conservation agents

MeSH Terms

Anabolic Agents
Bone and Bones
Bone Density Conservation Agents
Osteoclasts
Osteocytes
Osteogenesis
Osteoporosis
Parathyroid Hormone
Skeleton
Anabolic Agents
Bone Density Conservation Agents
Parathyroid Hormone

Figure

  • Fig. 1 Cellular events in the basic multicellular unit (BMU). Under the canopy generated by bone lining cells, osteoclasts are formed from hemopoietic precursors supplied by marrow and the bloodstream, and from partially differentiated osteoclasts termed quiescent osteoclast precursors (QoP). On the right side of the diagram, precursors of osteoblasts come from mesenchymal stem cells in the marrow and from blood, and from pericytes, and differentiate within the BMU through the osteoblast precursor stage to fully functional synthesizing osteoblasts; lining cells may also differentiate into active osteoblasts. Osteocytes communicate with the surface cells, particularly osteoblasts, through their canaliculae.

  • Fig. 2 Anabolic action of parathyroid hormone (PTH) through remodelling. PTH promotes differentiation of committed osteoblast precursors, activation of osteoclasts that produce coupling activities (see text for details), promotes survival of osteomlasts, and osteocytes, and inhibits sclerostin production by osteocytes.

  • Fig. 3 Possible mechanisms of parathyroid hormone (PTH) anabolic action in presence of resorption inhibition. PTH inhibits sclerostin production by osteocytes, promotes survival of osteoblasts and osteocytes, and promotes osteoblast differentiation in partly filled basic multicellular units (see text for details).

  • Fig. 4 Wnt signaling as a pathway to increase bone formation. (A) Wnt signalling pathway in the inactive state, showing a ligand Wnt inhibited by a decoy secreted frizzled-related protein (sFRP), the co-receptor, lipoprotein receptor-related protein (LRP)5/6, bound by either inhibitory protein, sclerostin or dickkopf (Dkk), and glycogen synthase kinase (GSK)-3β in the active state, resulting in proteasomal degradation of β-catenin (see text for details). (B) Active Wnt signalling, with LRP5/6 engaging in receptor complex after Wnt binding, disruption of GSK-3β inhibitory complex, stabilization of β-catenin and its translocation to nucleus where it activates transcription. Letters 1 to 5 indicate possible target sites for drug development (see text).

  • Fig. 5 Anabolic action of anti-sclerostin. Blockade of sclerostin acts predominantly through modelling-increasing bone on quiescent surfaces (left). On pre-resorbed surfaces (remodelling), it overfills resorbed sites and extends to adjacent quiescent surfaces (right). See text for details.


Reference

1. Aubin JE. Advances in the osteoblast lineage. Biochem Cell Biol. 1998; 76:899–910.
Article
2. Aubin JE. Regulation of osteoblast formation and function. Rev Endocr Metab Disord. 2001; 2:81–94.
3. Stein GS, Lian JB. Molecular mechanisms mediating proliferation/differentiation interrelationships during progressive development of the osteoblast phenotype. Endocr Rev. 1993; 14:424–442.
Article
4. Kim SW, Pajevic PD, Selig M, et al. Intermittent parathyroid hormone administration converts quiescent lining cells to active osteoblasts. J Bone Miner Res. 2012; 27:2075–2084.
Article
5. Baron R. Molecular mechanisms of bone resorption: therapeutic implications. Rev Rhum Engl Ed. 1996; 63:633–638.
6. Bruzzaniti A, Baron R. Molecular regulation of osteoclast activity. Rev Endocr Metab Disord. 2006; 7:123–139.
Article
7. Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003; 423:337–342.
Article
8. Suda T, Takahashi N, Udagawa N, et al. Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev. 1999; 20:345–357.
Article
9. Hauge EM, Qvesel D, Eriksen EF, et al. Cancellous bone remodeling occurs in specialized compartments lined by cells expressing osteoblastic markers. J Bone Miner Res. 2001; 16:1575–1582.
Article
10. Kristensen HB, Andersen TL, Marcussen N, et al. Increased presence of capillaries next to remodeling sites in adult human cancellous bone. J Bone Miner Res. 2013; 28:574–585.
Article
11. Eghbali-Fatourechi GZ, Modder UI, Charatcharoenwitthaya N, et al. Characterization of circulating osteoblast lineage cells in humans. Bone. 2007; 40:1370–1377.
Article
12. Eriksen EF, Eghbali-Fatourechi GZ, Khosla S. Remodeling and vascular spaces in bone. J Bone Miner Res. 2007; 22:1–6.
Article
13. Fujikawa Y, Quinn JM, Sabokbar A, et al. The human osteoclast precursor circulates in the monocyte fraction. Endocrinology. 1996; 137:4058–4060.
Article
14. Yamamoto Y, Udagawa N, Matsuura S, et al. Osteoblasts provide a suitable microenvironment for the action of receptor activator of nuclear factor-kappaB ligand. Endocrinology. 2006; 147:3366–3374.
Article
15. Mizoguchi T, Muto A, Udagawa N, et al. Identification of cell cycle-arrested quiescent osteoclast precursors in vivo. J Cell Biol. 2009; 184:541–554.
Article
16. Muto A, Mizoguchi T, Udagawa N, et al. Lineage-committed osteoclast precursors circulate in blood and settle down into bone. J Bone Miner Res. 2011; 26:2978–2990.
Article
17. Doherty MJ, Ashton BA, Walsh S, et al. Vascular pericytes express osteogenic potential in vitro and in vivo. J Bone Miner Res. 1998; 13:828–838.
Article
18. Howson KM, Aplin AC, Gelati M, et al. The postnatal rat aorta contains pericyte progenitor cells that form spheroidal colonies in suspension culture. Am J Physiol Cell Physiol. 2005; 289:C1396–C1407.
Article
19. Matsumoto T, Kawamoto A, Kuroda R, et al. Therapeutic potential of vasculogenesis and osteogenesis promoted by peripheral blood CD34-positive cells for functional bone healing. Am J Pathol. 2006; 169:1440–1457.
Article
20. Mödder UI, Khosla S. Skeletal stem/osteoprogenitor cells: current concepts, alternate hypotheses, and relationship to the bone remodeling compartment. J Cell Biochem. 2008; 103:393–400.
Article
21. Otsuru S, Tamai K, Yamazaki T, et al. Circulating bone marrow-derived osteoblast progenitor cells are recruited to the bone-forming site by the CXCR4/stromal cell-derived factor-1 pathway. Stem Cells. 2008; 26:223–234.
Article
22. Frost HM. Dynamics of bone remodeling. In : Frost HM, editor. Bone biodynamics. Boston, MA: Little, Brown and Company;1964. p. 315–333.
23. Parfitt AM. The coupling of bone formation to bone resorption: a critical analysis of the concept and of its relevance to the pathogenesis of osteoporosis. Metab Bone Dis Relat Res. 1982; 4:1–6.
Article
24. Eriksen EF. Normal and pathological remodeling of human trabecular bone: three dimensional reconstruction of the remodeling sequence in normals and in metabolic bone disease. Endocr Rev. 1986; 7:379–408.
Article
25. Martin T, Gooi JH, Sims NA. Molecular mechanisms in coupling of bone formation to resorption. Crit Rev Eukaryot Gene Expr. 2009; 19:73–88.
Article
26. Cummings SR, San Martin J, McClung MR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009; 361:756–765.
Article
27. Gelb BD, Shi GP, Chapman HA, et al. Pycnodysostosis, a lysosomal disease caused by cathepsin K deficiency. Science. 1996; 273:1236–1238.
Article
28. Saftig P, Hunziker E, Wehmeyer O, et al. Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin-K-deficient mice. Proc Natl Acad Sci U S A. 1998; 95:13453–13458.
Article
29. Pennypacker B, Shea M, Liu Q, et al. Bone density, strength, and formation in adult cathepsin K (-/-) mice. Bone. 2009; 44:199–207.
Article
30. Lotinun S, Kiviranta R, Matsubara T, et al. Osteoclast-specific cathepsin K deletion stimulates S1P-dependent bone formation. J Clin Invest. 2013; 123:666–681.
Article
31. Pennypacker BL, Duong le T, Cusick TE, et al. Cathepsin K inhibitors prevent bone loss in estrogen-deficient rabbits. J Bone Miner Res. 2011; 26:252–262.
Article
32. Cusick T, Chen CM, Pennypacker BL, et al. Odanacatib treatment increases hip bone mass and cortical thickness by preserving endocortical bone formation and stimulating periosteal bone formation in the ovariectomized adult rhesus monkey. J Bone Miner Res. 2012; 27:524–537.
Article
33. Masarachia PJ, Pennypacker BL, Pickarski M, et al. Odanacatib reduces bone turnover and increases bone mass in the lumbar spine of skeletally mature ovariectomized rhesus monkeys. J Bone Miner Res. 2012; 27:509–523.
Article
34. Eisman JA, Bone HG, Hosking DJ, et al. Odanacatib in the treatment of postmenopausal women with low bone mineral density: three-year continued therapy and resolution of effect. J Bone Miner Res. 2011; 26:242–251.
Article
35. Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001; 344:1434–1441.
Article
36. Frolik CA, Black EC, Cain RL, et al. Anabolic and catabolic bone effects of human parathyroid hormone (1-34) are predicted by duration of hormone exposure. Bone. 2003; 33:372–379.
Article
37. Ma YL, Cain RL, Halladay DL, et al. Catabolic effects of continuous human PTH (1--38) in vivo is associated with sustained stimulation of RANKL and inhibition of osteoprotegerin and gene-associated bone formation. Endocrinology. 2001; 142:4047–4054.
Article
38. Gowen M, Stroup GB, Dodds RA, et al. Antagonizing the parathyroid calcium receptor stimulates parathyroid hormone secretion and bone formation in osteopenic rats. J Clin Invest. 2000; 105:1595–1604.
Article
39. Fox J, Miller MA, Stroup GB, et al. Plasma levels of parathyroid hormone that induce anabolic effects in bone of ovariectomized rats can be achieved by stimulation of endogenous hormone secretion. Bone. 1997; 21:163–169.
Article
40. John MR, Widler L, Gamse R, et al. ATF936, a novel oral calcilytic, increases bone mineral density in rats and transiently releases parathyroid hormone in humans. Bone. 2011; 49:233–241.
Article
41. Misof BM, Roschger P, Cosman F, et al. Effects of intermittent parathyroid hormone administration on bone mineralization density in iliac crest biopsies from patients with osteoporosis: a paired study before and after treatment. J Clin Endocrinol Metab. 2003; 88:1150–1156.
Article
42. Ma YL, Zeng Q, Donley DW, et al. Teriparatide increases bone formation in modeling and remodeling osteons and enhances IGF-II immunoreactivity in postmenopausal women with osteoporosis. J Bone Miner Res. 2006; 21:855–864.
Article
43. Jilka RL. Molecular and cellular mechanisms of the anabolic effect of intermittent PTH. Bone. 2007; 40:1434–1446.
Article
44. Keller H, Kneissel M. SOST is a target gene for PTH in bone. Bone. 2005; 37:148–158.
Article
45. Martin TJ, Sims NA. Osteoclast-derived activity in the coupling of bone formation to resorption. Trends Mol Med. 2005; 11:76–81.
Article
46. Gooi JH, Pompolo S, Karsdal MA, et al. Calcitonin impairs the anabolic effect of PTH in young rats and stimulates expression of sclerostin by osteocytes. Bone. 2010; 46:1486–1497.
Article
47. Tang Y, Wu X, Lei W, et al. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med. 2009; 15:757–765.
Article
48. Xian L, Wu X, Pang L, et al. Matrix IGF-1 maintains bone mass by activation of mTOR in mesenchymal stem cells. Nat Med. 2012; 18:1095–1101.
Article
49. Canalis E, Giustina A, Bilezikian JP. Mechanisms of anabolic therapies for osteoporosis. N Engl J Med. 2007; 357:905–916.
Article
50. Horwitz MJ, Tedesco MB, Garcia-Ocaña A, et al. Parathyroid hormone-related protein for the treatment of postmenopausal osteoporosis: defining the maximal tolerable dose. J Clin Endocrinol Metab. 2010; 95:1279–1287.
Article
51. Henry JG, Mitnick M, Dann PR, et al. Parathyroid hormone-related protein-(1-36) is biologically active when administered subcutaneously to humans. J Clin Endocrinol Metab. 1997; 82:900–906.
Article
52. Ferrandon S, Feinstein TN, Castro M, et al. Sustained cyclic AMP production by parathyroid hormone receptor endocytosis. Nat Chem Biol. 2009; 5:734–742.
Article
53. Horwitz MJ, Augustine M, Kahn L, et al. A comparison of parathyroid hormone-related protein (1-36) and parathyroid hormone (1-34) on markers of bone turnover and bone density in postmenopausal women: the PrOP study. J Bone Miner Res. 2013; 28:2266–2276.
Article
54. Kurland ES, Heller SL, Diamond B, et al. The importance of bisphosphonate therapy in maintaining bone mass in men after therapy with teriparatide [human parathyroid hormone(1-34)]. Osteoporos Int. 2004; 15:992–997.
Article
55. Black DM, Bilezikian JP, Ensrud KE, et al. One year of alendronate after one year of parathyroid hormone (1-84) for osteoporosis. N Engl J Med. 2005; 353:555–565.
Article
56. Bilezikian JP, Rubin MR. Combination/sequential therapies for anabolic and antiresorptive skeletal agents for osteoporosis. Curr Osteoporos Rep. 2006; 4:5–13.
Article
57. Delmas PD, Vergnaud P, Arlot ME, et al. The anabolic effect of human PTH (1-34) on bone formation is blunted when bone resorption is inhibited by the bisphosphonate tiludronate--is activated resorption a prerequisite for the in vivo effect of PTH on formation in a remodeling system? Bone. 1995; 16:603–610.
Article
58. Black DM, Greenspan SL, Ensrud KE, et al. The effects of parathyroid hormone and alendronate alone or in combination in postmenopausal osteoporosis. N Engl J Med. 2003; 349:1207–1215.
Article
59. Finkelstein JS, Hayes A, Hunzelman JL, et al. The effects of parathyroid hormone, alendronate, or both in men with osteoporosis. N Engl J Med. 2003; 349:1216–1226.
Article
60. Tsai JN, Uihlein AV, Lee H, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet. 2013; 382:50–56.
Article
61. Cosman F, Eriksen EF, Recknor C, et al. Effects of intravenous zoledronic acid plus subcutaneous teriparatide [rhPTH (1-34)] in postmenopausal osteoporosis. J Bone Miner Res. 2011; 26:503–511.
Article
62. Pierroz DD, Bonnet N, Baldock PA, et al. Are osteoclasts needed for the bone anabolic response to parathyroid hormone? A study of intermittent parathyroid hormone with denosumab or alendronate in knock-in mice expressing humanized RANKL? J Biol Chem. 2010; 285:28164–28173.
Article
63. Vahle JL, Sato M, Long GG, et al. Skeletal changes in rats given daily subcutaneous injections of recombinant human parathyroid hormone (1-34) for 2 years and relevance to human safety. Toxicol Pathol. 2002; 30:312–321.
Article
64. Tashjian AH Jr, Goltzman D. On the interpretation of rat carcinogenicity studies for human PTH(1-34) and human PTH(1-84). J Bone Miner Res. 2008; 23:803–811.
Article
65. Cosman F, Nieves J, Zion M, et al. Daily and cyclic parathyroid hormone in women receiving alendronate. N Engl J Med. 2005; 353:566–575.
Article
66. Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005; 280:19883–19887.
Article
67. Semënov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005; 280:26770–26775.
Article
68. Rawadi G, Vayssière B, Dunn F, et al. BMP-2 controls alkaline phosphatase expression and osteoblast mineralization by a Wnt autocrine loop. J Bone Miner Res. 2003; 18:1842–1853.
Article
69. Gong Y, Slee RB, Fukai N, et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001; 107:513–523.
70. Babij P, Zhao W, Small C, et al. High bone mass in mice expressing a mutant LRP5 gene. J Bone Miner Res. 2003; 18:960–974.
Article
71. Kato M, Patel MS, Levasseur R, et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002; 157:303–314.
Article
72. Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling. J Clin Invest. 2006; 116:1202–1209.
Article
73. Baron R, Rawadi G. Targeting the Wnt/beta-catenin pathway to regulate bone formation in the adult skeleton. Endocrinology. 2007; 148:2635–2643.
Article
74. Ke HZ, Richards WG, Li X, et al. Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocr Rev. 2012; 33:747–783.
Article
75. Yaccoby S, Ling W, Zhan F, et al. Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood. 2007; 109:2106–2111.
Article
76. Clément-Lacroix P, Ai M, Morvan F, et al. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005; 102:17406–17411.
Article
77. Kulkarni NH, Onyia JE, Zeng Q, et al. Orally bioavailable GSK-3alpha/beta dual inhibitor increases markers of cellular differentiation in vitro and bone mass in vivo. J Bone Miner Res. 2006; 21:910–920.
Article
78. Loots GG, Kneissel M, Keller H, et al. Genomic deletion of a long-range bone enhancer misregulates sclerostin in Van Buchem disease. Genome Res. 2005; 15:928–935.
Article
79. Balemans W, Ebeling M, Patel N, et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum Mol Genet. 2001; 10:537–543.
Article
80. Balemans W, Patel N, Ebeling M, et al. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002; 39:91–97.
Article
81. van Bezooijen RL, Papapoulos SE, Löwik CW. Bone morphogenetic proteins and their antagonists: the sclerostin paradigm. J Endocrinol Invest. 2005; 28:15–17.
82. Li X, Ominsky MS, Warmington KS, et al. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Miner Res. 2009; 24:578–588.
Article
83. Tian X, Jee WS, Li X, et al. Sclerostin antibody increases bone mass by stimulating bone formation and inhibiting bone resorption in a hindlimb-immobilization rat model. Bone. 2011; 48:197–201.
Article
84. Padhi D, Jang G, Stouch B, et al. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J Bone Miner Res. 2011; 26:19–26.
Article
85. McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014; 370:412–420.
Article
86. Li X, Ominsky MS, Warmington KS, et al. Increased bone formation and bone mass induced by sclerostin antibody is not affected by pretreatment or cotreatment with alendronate in osteopenic, ovariectomized rats. Endocrinology. 2011; 152:3312–3322.
Article
87. Silvestrini G, Ballanti P, Leopizzi M, et al. Effects of intermittent parathyroid hormone (PTH) administration on SOST mRNA and protein in rat bone. J Mol Histol. 2007; 38:261–269.
Article
88. Leupin O, Kramer I, Collette NM, et al. Control of the SOST bone enhancer by PTH using MEF2 transcription factors. J Bone Miner Res. 2007; 22:1957–1967.
Article
89. Dong J, Peng J, Zhang H, et al. Role of glycogen synthase kinase 3beta in rapamycin-mediated cell cycle regulation and chemosensitivity. Cancer Res. 2005; 65:1961–1972.
Article
Full Text Links
  • JBM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr