Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Mechanisms of bone development and repair

Abstract

Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment — including inflammatory, endothelial and Schwann cells — persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Bone homeostasis.
Fig. 2: Skeletal stem cell hierarchy.
Fig. 3: Long bone anatomy.
Fig. 4: Developmental signalling pathways regulating osteoblast differentiation.
Fig. 5: Continuum of bone disorders.

Similar content being viewed by others

References

  1. Ambrosi, T. H., Longaker, M. T. & Chan, C. K. F. A revised perspective of skeletal stem cell biology. Front. Cell Dev. Biol. 7, 189 (2019).

    PubMed  PubMed Central  Google Scholar 

  2. Murphy, M. P. et al. The role of skeletal stem cells in the reconstruction of bone defects. J. Craniofac. Surg. 28, 1136–1141 (2017).

    PubMed  PubMed Central  Google Scholar 

  3. Long, F. Building strong bones: molecular regulation of the osteoblast lineage. Nat. Rev. Mol. Cell Biol. 13, 27–38 (2012).

    CAS  Google Scholar 

  4. Bianco, P. & Robey, P. G. Skeletal stem cells. Development 142, 1023–1027 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Garnero, P., Sornay-Rendu, E., Chapuy, M. C. & Delmas, P. D. Increased bone turnover in late postmenopausal women is a major determinant of osteoporosis. J. Bone Miner. Res. 11, 337–349 (2009).

    Google Scholar 

  6. Soltanoff, C. S., Yang, S., Chen, W. & Li, Y. P. Signaling networks that control the lineage commitment and differentiation of bone cells. Crit. Rev. Eukaryot. Gene Expr. 19, 1–46 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Compton, J. T. & Lee, F. Y. Current concepts review: a review of osteocyte function and the emerging importance of sclerostin. J. Bone Joint Surg. Am. 96, 1659–1668 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Van Bezooijen, R. L. et al. Sclerostin Is an osteocyte-expressed negative regulator of bone formation, but not a classical BMP antagonist. J. Exp. Med. 199, 805–814 (2004).

    PubMed  PubMed Central  Google Scholar 

  9. Robling, A. G. et al. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J. Biol. Chem. 283, 5866–5875 (2008).

    CAS  PubMed  Google Scholar 

  10. Tatsumi, S. et al. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 5, 464–475 (2007).

    CAS  PubMed  Google Scholar 

  11. Jacome-Galarza, C. E., Lee, S. K., Lorenzo, J. A. & Aguila, H. L. Identification, characterization, and isolation of a common progenitor for osteoclasts, macrophages, and dendritic cells from murine bone marrow and periphery. J. Bone Miner. Res. 28, 1203–1213 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315–323 (1999).

    CAS  PubMed  Google Scholar 

  13. Dougall, W. C. et al. RANK is essential for osteoclast and lymph node development. Genes. Dev. 13, 2412–2424 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Xu, F. & Teitelbaum, S. L. Osteoclasts: new insights. Bone Res. 1, 11–26 (2013).

    CAS  PubMed Central  Google Scholar 

  15. Meyers, C. et al. Heterotopic ossification: a comprehensive review. JBMR Plus 3, e10172 (2019).

    PubMed  PubMed Central  Google Scholar 

  16. Dallas, S. L., Xie, Y., Shiflett, L. A. & Ueki, Y. Mouse Cre models for the study of bone diseases. Curr. Osteoporos. Rep. 16, 466–477 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999). This work establishes the potential for MSCs to differentiate into bone, cartilage and fat.

    CAS  PubMed  Google Scholar 

  18. Chen, Q. et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 23, 1128–1139 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Friedenstein, A. J., Chailakhjan, R. K. & Lalykina, K. S. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Prolif. 3, 393–403 (1970).

    CAS  Google Scholar 

  20. Friedenstein, A. J., Chailakhyan, R. K. & Gerasimov, U. V. Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Prolif. 20, 263–272 (1987).

    CAS  Google Scholar 

  21. Friedenstein, A. J. Osteogenic stem cells in the bone marrow. Bone Miner. Res. https://doi.org/10.1016/b978-0-444-81371-8.50012-1 (1990).

    Article  Google Scholar 

  22. Wei, J. et al. Glucose uptake and Runx2 synergize to orchestrate osteoblast differentiation and bone formation. Cell 161, 1576–1591 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang, T., Zhang, X. & Bikle, D. D. Osteogenic differentiation of periosteal cells during fracture healing. J. Cell Physiol. 232, 913–921 (2017).

    CAS  PubMed  Google Scholar 

  24. Ackema, K. B. & Charité, J. Mesenchymal stem cells from different organs are characterized by distinct topographic Hox codes. Stem Cell Dev. 17, 979–991 (2008).

    CAS  Google Scholar 

  25. Rux, D. R. et al. Regionally restricted Hox function in adult bone marrow multipotent mesenchymal stem/stromal cells. Dev. Cell 39, 653–666 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Nelson, L. T., Rakshit, S., Sun, H. & Wellik, D. M. Generation and expression of a Hoxa11eGFP targeted allele in mice. Dev. Dyn. 237, 3410–3416 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Swinehart, I. T., Schlientz, A. J., Quintanilla, C. A., Mortlock, D. P. & Wellik, D. M. Hox11 genes are required for regional patterning and integration of muscle, tendon and bone. Development 140, 4574–4582 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Pineault, K. M., Song, J. Y., Kozloff, K. M., Lucas, D. & Wellik, D. M. Hox11 expressing regional skeletal stem cells are progenitors for osteoblasts, chondrocytes and adipocytes throughout life. Nat. Commun. 10, 3168 (2019).

    PubMed  PubMed Central  Google Scholar 

  29. Rux, D. R. & Wellik, D. M. Hox genes in the adult skeleton: novel functions beyond embryonic development. Dev. Dyn. 246, 310–317 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Chan, C. K. F. et al. Identification and specification of the mouse skeletal stem cell. Cell 160, 285–298 (2015). The work is the first to isolate the SSC in mice, which has the differentiation capacity to be restricted to bone, cartilage,and bone stroma.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Chan, C. K. F. et al. Identification of the human skeletal stem cell. Cell 175, 43–56 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Sacchetti, B. et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131, 324–336 (2007).

    CAS  PubMed  Google Scholar 

  33. Kassem, M. & Bianco, P. Skeletal stem cells in space and time. Cell 160, 17–19 (2015).

    CAS  PubMed  Google Scholar 

  34. Bianco, P. Stem cells and bone: a historical perspective. Bone 70, 2–9 (2015).

    PubMed  Google Scholar 

  35. Ueno, H. & Weissman, I. L. Clonal analysis of mouse development reveals a polyclonal origin for yolk sac blood islands. Dev. Cell 11, 519–533 (2006).

    CAS  PubMed  Google Scholar 

  36. Worthley, D. L. et al. Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 160, 269–284 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Chan, C. K. F. et al. Clonal precursor of bone, cartilage, and hematopoietic niche stromal cells. Proc. Natl Acad. Sci. USA 110, 12643–12648 (2013).

    CAS  PubMed  Google Scholar 

  38. Berendsen, A. D. & Olsen, B. R. Bone development. Bone 80, 14–18 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Marecic, O. et al. Identification and characterization of an injury-induced skeletal progenitor. Proc. Natl Acad. Sci. USA 112, 9920–9925 (2015).

    CAS  PubMed  Google Scholar 

  40. Tevlin, R. et al. Pharmacological rescue of diabetic skeletal stem cell niches. Sci. Transl Med. 9, eaag2809 (2017).

    PubMed  PubMed Central  Google Scholar 

  41. Ransom, R. C. et al. Mechanoresponsive stem cells acquire neural crest fate in jaw regeneration. Nature 563, 514–521 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mizuhashi, K. et al. Resting zone of the growth plate houses a unique class of skeletal stem cells. Nature 563, 254–258 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Debnath, S. et al. Discovery of a periosteal stem cell mediating intramembranous bone formation. Nature 562, 133–139 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Jia, G. et al. Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement. Nat. Commun. 9, 4877 (2018).

    PubMed  PubMed Central  Google Scholar 

  45. Baker, S., Rogerson, C., Hayes, A., Sharrocks, A. & Rattray, M. Classifying cells with Scasat, a single-cell ATAC-seq analysis tool. Nucleic Acids Res. 47, e10 (2019).

    PubMed  Google Scholar 

  46. Le Douarin, N. M. & Smith, J. Development of the peripheral nervous system from the neural crest. Annu. Rev. Cell Biol. 4, 375–404 (1988).

    PubMed  Google Scholar 

  47. Long, F. & Ornitz, D. M. Development of the endochondral skeleton. Cold Spring Harb Perspect. Biol. 5, a008334 (2013).

    PubMed  PubMed Central  Google Scholar 

  48. Kronenberg, H. M. Developmental regulation of the growth plate. Nature. 423, 332–336 (2003).

    CAS  PubMed  Google Scholar 

  49. Maes, C. & Kronenberg, H. M. Postnatal bone growth: growth plate biology, bone formation, and remodeling. pediatric. Bone https://doi.org/10.1016/B978-0-12-382040-2.10004-8 (2012).

    Article  Google Scholar 

  50. Lefebvr, E. V. & Dvir-Ginzberg, M. SOX9 and the many facets of its regulation in the chondrocyte lineage. Connect. Tissue Res. 58, 2–14 (2017).

    Google Scholar 

  51. Lovell-Badge, R. The early history of the Sox genes. Int. J. Biochem. Cell Biol. 42, 378–380 (2010).

    CAS  PubMed  Google Scholar 

  52. Bi, W., Deng, J. M., Zhang, Z., Behringer, R. R. & De Crombrugghe, B. Sox9 is required for cartilage formation. Nat. Genet. 22, 85–89 (1999).

    CAS  PubMed  Google Scholar 

  53. Akiyama, H., Chaboissier, M. C., Martin, J. F., Schedl, A. & De Crombrugghe, B. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev. 16, 2813–2828 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Henry, S. P., Liang, S., Akdemir, K. C. & De Crombrugghe, B. The postnatal role of Sox9 in cartilage. J. Bone Miner. Res. 27, 2511–2525 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Schafer, A. J. et al. Campomelic dysplasia with XY sex reversal: diverse phenotypes resulting from mutations in a single gene. Ann. N. Y. Acad. Sci. 785, 137–149 (1996).

    CAS  PubMed  Google Scholar 

  56. Gentilin, B. et al. Phenotype of five cases of prenatally diagnosed campomelic dysplasia harboring novel mutations of the SOX9 gene. Ultrasound Obstet. Gynecol. 36, 315–323 (2010).

    CAS  PubMed  Google Scholar 

  57. Komori, T. Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res. 339, 189–195 (2010).

    CAS  PubMed  Google Scholar 

  58. Ducy, P., Zhang, R., Geoffroy, V., Ridall, A. L. & Karsenty, G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell 89, 747–754 (1997).

    CAS  PubMed  Google Scholar 

  59. Harada, H. et al. Cbfa1 isoforms exert functional differences in osteoblast differentiation. J. Biol. Chem. 274, 6972–6978 (1999).

    CAS  PubMed  Google Scholar 

  60. Komori, T. et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89, 755–764 (1997). This work establishes RUNX2 as an essential transcription factor for osteoblast differentiation.

    CAS  PubMed  Google Scholar 

  61. Otto, F. et al. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89, 765–771 (1997).

    CAS  PubMed  Google Scholar 

  62. Inada, M. et al. Maturational disturbance of chondrocytes in Cbfa1-deficient mice. Dev. Dyn. 214, 279–290 (1999).

    CAS  PubMed  Google Scholar 

  63. Takarada, T. et al. An analysis of skeletal development in osteoblast-specific and chondrocyte-specific runt-related transcription factor-2 (Runx2) knockout mice. J. Bone Miner. Res. 28, 2064–2069 (2013).

    CAS  PubMed  Google Scholar 

  64. Maruyama, Z. et al. Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev. Dyn. 236, 1876–1890 (2007).

    CAS  PubMed  Google Scholar 

  65. Sinha, K. M. & Zhou, X. Genetic and molecular control of osterix in skeletal formation. J. Cell Biochem. 114, 975–984 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Karsenty, G. Minireview: tranzscriptional control of osteoblast differentiation. Endocrinology 142, 2731–2733 (2001).

    CAS  PubMed  Google Scholar 

  67. Nakashima, K. & De Crombrugghe, B. Transcriptional mechanisms in osteoblast differentiation and bone formation. Trends Genet. 19, 458–466 (2003).

    CAS  PubMed  Google Scholar 

  68. Nakashima, K. et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108, 17–29 (2002). This work establishes the temporal coordination between OSX and RUNX2 activation for osteoblast differentiation.

    CAS  PubMed  Google Scholar 

  69. Yang, X. & Karsenty, G. Transcription factors in bone: developmental and pathological aspects. Trends Mol. Med. 8, 340–345 (2002).

    CAS  PubMed  Google Scholar 

  70. Zhou, X. et al. Multiple functions of osterix are required for bone growth and homeostasis in postnatal mice. Proc. Natl Acad. Sci. USA 107, 12919–12924 (2010).

    CAS  PubMed  Google Scholar 

  71. Liu, T. M. & Lee, E. H. Transcriptional regulatory cascades in Runx2-dependent bone development. Tissue Eng. Part B Rev. 19, 254–263 (2013).

    PubMed  Google Scholar 

  72. St-Arnaud, R. & Hekmatnejad, B. Combinatorial control of ATF4-dependent gene transcription in osteoblasts. Ann. N. Y. Acad. Sci. 1237, 11–18 (2011).

    CAS  PubMed  Google Scholar 

  73. Yang, X. et al. ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology: implication for Coffin-Lowry syndrome. Cell 117, 387–398 (2004).

    CAS  PubMed  Google Scholar 

  74. Jing, D. et al. The role of microRNAs in bone remodeling. Int. J. Oral. Sci. 7, 131–143 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Xiao, G. et al. Cooperative interactions between activating transcription factor 4 and Runx2/Cbfa1 stimulate osteoblast-specific osteocalcin gene expression. J. Biol. Chem. 280, 30689–30696 (2005).

    CAS  PubMed  Google Scholar 

  76. Wagner, E. F. Functions of AP1 (Fos/Jun) in bone development. Ann. Rheum. Dis. 61, ii40–ii42 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kenner, L. et al. Mice lacking JunB are osteopenic due to cell-autonomous osteoblast and osteoclast defects. J. Cell Biol. 164, 613–623 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Zambotti, A., Makhluf, H., Shen, J. & Ducy, P. Characterization of an osteoblast-specific enhancer element in the CBFA1. Gene 277, 41497–41506 (2002).

    CAS  Google Scholar 

  79. Jochum, W. et al. Increased bone formation and osteosclerosis in mice overexpressing the transcription factor Fra-1. Nat. Med. 6, 980–984 (2000).

    CAS  PubMed  Google Scholar 

  80. Bozec, A. et al. Fra-2/AP-1 controls bone formation by regulating osteoblast differentiation and collagen production. J. Cell Biol. 190, 1093–1106 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Nüsslein-volhard, C. & Wieschaus, E. Mutations affecting segment number and polarity in drosophila. Nature. 287, 795–801 (1980).

    PubMed  Google Scholar 

  82. McMahon, A. P., Ingham, P. W. & Tabin, C. J. 1 Developmental roles and clinical significance of Hedgehog signaling. Curr. Top. Dev. Biol. 53, 1–114 (2003).

    CAS  PubMed  Google Scholar 

  83. Ocbina, P. J. R. & Anderson, K. V. Intraflagellar transport, Cilia, and mammalian hedgehog signaling: analysis in mouse embryonic fibroblasts. Dev. Dyn. 237, 2030–2038 (2008).

    PubMed  PubMed Central  Google Scholar 

  84. Riddle, R. D., Johnson, R. L., Laufer, E. & Tabin, C. Sonic hedgehog mediates the polarizing activity of the ZPA. Cell 75, 1401–1416 (1993).

    CAS  PubMed  Google Scholar 

  85. Rohatgi, R., Milenkovic, L. & Scott, M. P. Patched1 regulates hedgehog signaling at the primary cilium. Science. 317, 372–376 (2007).

    CAS  PubMed  Google Scholar 

  86. Corbit, K. C. et al. Vertebrate Smoothened functions at the primary cilium. Nature. 437, 1018–1021 (2005).

    CAS  PubMed  Google Scholar 

  87. Towers, M., Mahood, R., Yin, Y. & Tickle, C. Integration of growth and specification in chick wing digit-patterning. Nature 452, 882–886 (2008).

    CAS  PubMed  Google Scholar 

  88. Chinnaiya, K., Tickle, C. & Towers, M. Sonic hedgehog-expressing cells in the developing limb measure time by an intrinsic cell cycle clock. Nat. Commun. 5, 4230 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Wang, B., Fallon, J. F. & Beachy, P. A. Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell 100, 423–434 (2000).

    CAS  PubMed  Google Scholar 

  90. Mo, R. et al. Specific and redundant functions of Gli2 and Gli3 zinc finger genes in skeletal patterning and development. Development 124, 113–123 (1997).

    CAS  PubMed  Google Scholar 

  91. Park, H. et al. Mouse Gli1 mutants are viable but have defects in SHH signaling in combination with a Gli2 mutation. Development 127, 1593–1605 (2000).

    CAS  PubMed  Google Scholar 

  92. Hojo, H. et al. Gli1 protein participates in hedgehog-mediated specification of osteoblast lineage during endochondral ossification. J. Biol. Chem. 287, 17860–17869 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Amano, K., Densmore, M., Nishimura, R. & Lanske, B. Indian hedgehog signaling regulates transcription and expression of collagen type X via Runx2/Smads interactions. J. Biol. Chem. 289, 24898–24910 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Jemtland, R., Divieti, P., Lee, K. & Segre, G. V. Hedgehog promotes primary osteoblast differentiation and increases PTHrP mRNA expression and iPTHrP secretion. Bone 32, 611–620 (2003).

    CAS  PubMed  Google Scholar 

  95. Long, F. & Linsenmayer, T. F. Regulation of growth region cartilage proliferation and differentiation by perichondrium. Development 125, 1067–1073 (1998).

    CAS  PubMed  Google Scholar 

  96. Mak, K. K., Chen, M. H., Day, T. F., Chuang, P. T. & Yang, Y. Wnt/β-catenin signaling interacts differentially with Ihh signaling in controlling endochondral bone and synovial joint formation. Development 133, 3695–3707 (2006).

    CAS  PubMed  Google Scholar 

  97. Day, T. F. & Yang, Y. Wnt and hedgehog signaling pathways in bone development. J. Bone Joint Surg. Ser. Am. 90, 19–24 (2008).

    Google Scholar 

  98. Hojo, H. et al. Hedgehog-Gli activators direct osteo-chondrogenic function of bone morphogenetic protein toward osteogenesis in the perichondrium. J. Biol. Chem. 288, 9924–9932 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Schroeter, E. H., Kisslinger, J. A. & Kopan, R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature 393, 382–386 (1998).

    CAS  PubMed  Google Scholar 

  100. Zanotti, S. & Canalis, E. Notch signaling and the skeleton. Endocr. Rev. 37, 223–253 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Tu, X. et al. Physiological Notch signaling maintains bone homeostasis via RBPjk and Hey upstream of NFATc1. PLoS Genet. 8, e1002577 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Hilton, M. J. et al. Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat. Med. 14, 306–314 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Engin, F. et al. Dimorphic effects of Notch signaling in bone homeostasis. Nat. Med. 14, 299–305 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Canalis, E., Parker, K., Feng, J. Q. & Zanotti, S. Osteoblast lineage-specific effects of notch activation in the skeleton. Endocrinology 154, 623–634 (2013).

    CAS  PubMed  Google Scholar 

  105. Zanotti, S. & Canalis, E. Notch1 and Notch2 expression in osteoblast precursors regulates femoral microarchitecture. Bone 62, 22–28 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Kim, J. B. et al. Bone regeneration is regulated by Wnt signaling. J. Bone Miner. Res. 22, 1913–1923 (2007).

    CAS  PubMed  Google Scholar 

  107. Huelsken, J. & Birchmeier, W. New aspects of Wnt signaling pathways in higher vertebrates. Curr. Opin. Genet. Dev. 11, 547–553 (2001).

    CAS  PubMed  Google Scholar 

  108. Williams, B. O. & Insogna, K. L. Where Wnts went: the exploding field of Lrp5 and Lrp6 signaling in bone. J. Bone Miner. Res. 24, 171–178 (2009).

    CAS  PubMed  Google Scholar 

  109. Joiner, D. M., Ke, J., Zhong, Z., Xu, H. E. & Williams, B. O. LRP5 and LRP6 in development and disease. Trends Endocrinol. Metab. 24, 31–39 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Baron, R. & Kneissel, M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat. Med. 19, 179–192 (2013).

    CAS  PubMed  Google Scholar 

  111. Boyden, L. M. et al. High bone density due to a mutation in LDL-receptor-related protein 5. N. Engl. J. Med. 346, 1513–1521 (2002).

    CAS  PubMed  Google Scholar 

  112. Little, R. D. et al. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am. J. Hum. Genet. 70, 11–19 (2002).

    CAS  PubMed  Google Scholar 

  113. Houschyar, K. S. et al. Wnt pathway in bone repair and regeneration – what do we know so far. Front. Cell Dev. Biol. 6, 170 (2019).

    PubMed  PubMed Central  Google Scholar 

  114. Minear, S. et al. Wnt proteins promote bone regeneration. Sci. Transl Med. 2, 29ra30 (2010).

    PubMed  Google Scholar 

  115. Poole, K. E. S. et al. Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation. FASEB J. 19, 1842–1844 (2005).

    CAS  PubMed  Google Scholar 

  116. Ai, M., Holmen, S. L., Van Hul, W., Williams, B. O. & Warman, M. L. Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone mass-associated missense mutations in LRP5 affect canonical Wnt signaling. Mol. Cell Biol. 25, 4946–4955 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Brunkow, M. E. et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am. J. Hum. Genet. 68, 577–589 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Holmen, S. L. et al. Decreased BMD and limb deformities in mice carrying mutations in both Lrp5 and Lrp6. J. Bone Miner. Res. 19, 2033–2040 (2004).

    CAS  PubMed  Google Scholar 

  119. Kubota, T. et al. Lrp6 hypomorphic mutation affects bone mass through bone resorption in mice and impairs interaction with Mesd. J. Bone Miner. Res. 23, 1661–1671 (2008).

    CAS  PubMed  Google Scholar 

  120. Lin, G. L. & Hankenson, K. D. Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J. Cell Biochem. 112, 3491–3501 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Wu, M., Chen, G. & Li, Y. P. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 4, 16009 (2016).

    PubMed  PubMed Central  Google Scholar 

  122. Itasaki, N. & Hoppler, S. Crosstalk between Wnt and bone morphogenic protein signaling: a turbulent relationship. Dev. Dyn. 239, 16–33 (2010).

    CAS  PubMed  Google Scholar 

  123. Luo, Q. et al. Connective tissue growth factor (CTGF) is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells. J. Biol. Chem. 279, 55958–55968 (2004).

    CAS  PubMed  Google Scholar 

  124. Si, W. et al. CCN1/Cyr61 is regulated by the canonical Wnt signal and plays an important role in Wnt3A-induced osteoblast differentiation of mesenchymal stem cells. Mol. Cell Biol. 26, 2955–2964 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Boland, G. M., Perkins, G., Hall, D. J. & Tuan, R. S. Wnt 3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells. J. Cell Biochem. 93, 1210–1230 (2004).

    CAS  PubMed  Google Scholar 

  126. Chen, Y. et al. β-Catenin signaling pathway is crucial for bone morphogenetic protein 2 to induce new bone formation. J. Biol. Chem. 282, 526–533 (2007).

    CAS  PubMed  Google Scholar 

  127. Zhang, M. et al. BMP-2 modulates β-catenin signaling through stimulation of Lrp5 expression and inhibition of β-TrCP expression in osteoblasts. J. Cell Biochem. 108, 896–905 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Wrana, J. L. et al. TGFβ signals through a heteromeric protein kinase receptor complex. Cell 71, 1003–1014 (1992).

    CAS  PubMed  Google Scholar 

  129. Schmierer, B. & Hill, C. S. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat. Rev. Mol. Cell Biol. 8, 970–982 (2007).

    CAS  PubMed  Google Scholar 

  130. Salazar, V. S., Gamer, L. W. & Rosen, V. BMP signalling in skeletal development, disease and repair. Nat. Rev. Endocrinol. 12, 203–221 (2016).

    CAS  PubMed  Google Scholar 

  131. Katagiri, T. & Watabe, T. Bone morphogenetic proteins. Cold Spring Harb Perspect. Biol. https://doi.org/10.1101/cshperspect.a021899 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Salazar, V. S. et al. Reactivation of a developmental Bmp2 signaling center is required for therapeutic control of the murine periosteal niche. eLife https://doi.org/10.7554/eLife.42386 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Bandyopadhyay, A. et al. Genetic analysis of the roles of BMP2, BMP4, and BMP7 in limb patterning and skeletogenesis. PLoS Genet. 2, 2116–2130 (2006).

    CAS  Google Scholar 

  134. Tsuji, K. et al. BMP2 activity, although dispensable for bone formation, is required for the initiation of fracture healing. Nat. Genet. 38, 1424–1429 (2006). This work demonstrates the role of reoccurring BMP signalling for limb development and fracture healing of the limb.

    CAS  PubMed  Google Scholar 

  135. Lim, J. et al. Dual function of Bmpr1a signaling in restricting preosteoblast proliferation and stimulating osteoblast activity in mouse. Development 143, 339–347 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Fujii, M. et al. Roles of bone morphogenetic protein type I receptors and Smad proteins in osteoblast and chondroblast differentiation. Mol. Biol. Cell 10, 3801–3813 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Singhatanadgit, W. & Olsen, I. Endogenous BMPR-IB signaling is required for early osteoblast differentiation of human bone cells. Vitr. Cell Dev. Biol. Anim. 47, 251–259 (2011).

    CAS  Google Scholar 

  138. Yoshida, Y. et al. Negative regulation of BMP/Smad signaling by Tob in osteoblasts. Cell 103, 1085–1097 (2000).

    CAS  PubMed  Google Scholar 

  139. Zhang, Y. et al. Loss of BMP signaling through BMPR1A in osteoblasts leads to greater collagen cross-link maturation and material-level mechanical properties in mouse femoral trabecular compartments. Bone 88, 74–84 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Johnson, D. E. & Williams, L. T. Structural and functional diversity in the FGF receptor multigene family. Adv. Cancer Res. 60, 1–41 (1992).

    Google Scholar 

  141. Ornitz, D. M. et al. Receptor specificity of the fibroblast growth factor family. J. Biol. Chem. 271, 15292–15297 (1996).

    CAS  PubMed  Google Scholar 

  142. Ornitz, D. M. FGF signaling in the developing endochondral skeleton. Cytokine Growth Factor. Rev. 16, 205–213 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Montero, A. et al. Disruption of the fibroblast growth factor-2 gene results in decreased bone mass and bone formation. J. Clin. Invest. 105, 1085–1093 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Zhou, M. et al. Fibroblast growth factor 2 control of vascular tone. Nat. Med. 4, 201–207 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Crossley, P. H., Minowada, G., MacArthur, C. A. & Martin, G. R. Roles for FGF8 in the induction, initiation, and maintenance of chick limb development. Cell 84, 127–136 (1996).

    CAS  PubMed  Google Scholar 

  146. Lewandoski, M., Sun, X. & Martin, G. R. Fgf8 signalling from the AER is essential for normal limb development. Nat. Genet. 26, 460–463 (2000).

    CAS  PubMed  Google Scholar 

  147. Martin, G. R. The roles of FGFs in the early development of vertebrate limbs. Genes Dev. 12, 1571–1586 (1998).

    CAS  PubMed  Google Scholar 

  148. Min, H. et al. Fgf-10 is required for both limb and lung development and exhibits striking functional similarity to Drosophila branchless. Genes Dev. 12, 3156–3161 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Ohuchi, H. et al. The mesenchymal factor, FGF10, initiates and maintains the outgrowth of the chick limb bud through interaction with FGF8, an apical ectodermal factor. Development 124, 2235–2244 (1997).

    CAS  PubMed  Google Scholar 

  150. Mahmood, R. et al. A role for FGF-8 in the initiation and maintenance of vertebrate limb bud outgrowth. Curr. Biol. 5, 797–806 (1995).

    CAS  PubMed  Google Scholar 

  151. Heikinheimo, M., Lawshé, A., Shackleford, G. M., Wilson, D. B. & MacArthur, C. A. Fgf-8 expression in the post-gastrulation mouse suggests roles in the development of the face, limbs and central nervous system. Mech. Dev. 48, 129–138 (1994).

    CAS  PubMed  Google Scholar 

  152. Lin, J. M. et al. Actions of fibroblast growth factor-8 in bone cells in vitro. Am. J. Physiol. Endocrinol. Metab. 297, E142–E150 (2009).

    CAS  PubMed  Google Scholar 

  153. Yamaguchi, T. P., Conlon, R. A. & Rossant, J. Expression of the fibroblast growth factor receptor FGFR-1/flg during gastrulation and segmentation in the mouse embryo. Dev. Biol. 152, 75–88 (1992).

    CAS  PubMed  Google Scholar 

  154. Deng, C. et al. Fibroblast growth factor receptor-1 (FGFR-1) is essential for normal neural tube and limb development. Dev. Biol. 185, 42–54 (1997).

    CAS  PubMed  Google Scholar 

  155. Jacob, A. L., Smith, C., Partanen, J. & Ornitz, D. M. Fibroblast growth factor receptor 1 signaling in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev. Biol. 296, 315–328 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Verheyden, J. M., Lewandoski, M., Deng, C., Harfe, B. D. & Sun, X. Conditional inactivation of Fgfr1 in mouse defines its role in limb bud establishment, outgrowth and digit patterning. Development 132, 4235–4245 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Orr-Urtreger, A. et al. Developmental localization of the splicing alternatives of fibroblast growth factor receptor-2 (FGFR2). Dev. Biol. 158, 475–486 (1993).

    CAS  PubMed  Google Scholar 

  158. Li, X. et al. Fibroblast growth factor signaling and basement membrane assembly are connected during epithelial morphogenesis of the embryoid body. J. Cell Biol. 153, 811–822 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Arman, E., Haffner-Krausz, R., Chen, Y., Heath, J. K. & Lonai, P. Targeted disruption of fibroblast growth factor (FGF) receptor 2 suggests a role for FGF signaling in pregastrulation mammalian development. Proc. Natl Acad. Sci. USA 95, 5082–5087 (1998).

    CAS  PubMed  Google Scholar 

  160. Xu, X. et al. Fibroblast growth factor receptor 2 (FGFR2)-mediated reciprocal regulation loop between FGF8 and FGF10 is essential for limb induction. Development 125, 753–765 (1998).

    CAS  PubMed  Google Scholar 

  161. Wang, Y. et al. Abnormalities in cartilage and bone development in the Apert syndrome FGFR2(+/S252W) mouse. Development 132, 3537–3548 (2005).

    CAS  PubMed  Google Scholar 

  162. Claes, L., Recknagel, S. & Ignatius, A. Fracture healing under healthy and inflammatory conditions. Nat. Rev. Rheumatol. 8, 133–143 (2012).

    CAS  PubMed  Google Scholar 

  163. Glynne, A. J., Andrew, S. M., Freemont, A. J. & Marsh, D. R. Inflammatory cells in normal human fracture healing. Acta Orthop. 65, 462–466 (1994).

    Google Scholar 

  164. Bolander, M. E. Regulation of fracture repair by growth factors. Exp. Biol. Med. 200, 165–170 (1992).

    CAS  Google Scholar 

  165. Croes, M. et al. Proinflammatory mediators enhance the osteogenesis of human mesenchymal stem cells after lineage commitment. PLoS ONE 10, e0132781 (2015).

    PubMed  PubMed Central  Google Scholar 

  166. Lu, L. Y. et al. Pro-inflammatory M1 macrophages promote Osteogenesis by mesenchymal stem cells via the COX-2-prostaglandin E2 pathway. J. Orthop. Res. 35, 2378–2385 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Bernhardsson, M. & Aspenberg, P. Osteoblast precursors and inflammatory cells arrive simultaneously to sites of a trabecular-bone injury. Acta Orthop. 89, 457–461 (2018).

    PubMed  PubMed Central  Google Scholar 

  168. Ono, T. et al. IL-17-producing γδT cells enhance bone regeneration. Nat. Commun. 7, 10928 (2016). This work shows that the presence of the proinflammatory cytokine IL-17, from the niche, aided in bone regrowth after injury.

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Goerke, S. M., Obermeyer, J., Plaha, J., Stark, G. B. & Finkenzeller, G. Endothelial progenitor cells from peripheral blood support bone regeneration by provoking an angiogenic response. Microvasc. Res. 98, 40–47 (2015).

    CAS  PubMed  Google Scholar 

  170. Langen, U. H. et al. Cell-matrix signals specify bone endothelial cells during developmental osteogenesis. Nat. Cell Biol. 19, 189–201 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Kusumbe, A. P., Ramasamy, S. K. & Adams, R. H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature. 507, 323–328 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Ramasamy, S. K., Kusumbe, A. P., Wang, L. & Adams, R. H. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature. 507, 376–380 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Cao, J. et al. Sensory nerves affect bone regeneration in rabbit mandibular distraction osteogenesis. Int. J. Med. Sci. 16, 831–837 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Jones, R. E. et al. Skeletal stem cell-Schwann cell circuitry in Mandibular repair. Cell Rep. 28, 2757–2766.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Park, B. W., Kim, J. R., Lee, J. H. & Byun, J. H. Expression of nerve growth factor and vascular endothelial growth factor in the inferior alveolar nerve after distraction osteogenesis. Int. J. Oral Maxillofac. Surg. 35, 624–630 (2006).

    PubMed  Google Scholar 

  176. Wang, L. et al. Locally applied nerve growth factor enhances bone consolidation in a rabbit model of mandibular distraction osteogenesis. J. Orthop. Res. 24, 2238–2245 (2006).

    CAS  PubMed  Google Scholar 

  177. Emara, K. M., Diab, R. A. & Emara, A. K. Recent biological trends in management of fracture non-union. World J. Orthop. 6, 623–628 (2015).

    PubMed  PubMed Central  Google Scholar 

  178. Panteli, M., Pountos, I., Jones, E. & Giannoudis, P. V. Biological and molecular profile of fracture non-union tissue: current insights. J. Cell Mol. Med. 19, 685–713 (2015).

    PubMed  PubMed Central  Google Scholar 

  179. Jones, A. L. et al. Recombinant human BMP-2 and allograft compared with autogenous bone graft for reconstruction of diaphyseal tibial fractures with cortical defects: a randomized, controlled trial. J. Bone Joint Surg. Ser. Am. 88, 1431–1441 (2006).

    Google Scholar 

  180. Kawaguchi, H. et al. Local application of recombinant human fibroblast growth factor-2 on bone repair: a dose-escalation prospective trial on patients with osteotomy. J. Orthop. Res. 25, 480–487 (2007).

    CAS  PubMed  Google Scholar 

  181. Babcock, S. & Kellam, J. F. Hip fracture nonunions: diagnosis, treatment, and special considerations in elderly patients. Adv. Orthop. https://doi.org/10.1155/2018/1912762 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  182. Atanelov, Z. & Bentley, T. P. Greenstick fracture. StatPearls (2018).

  183. Kraft, C. T. et al. Trauma-induced heterotopic bone formation and the role of the immune system: a review. J. Trauma. Acute Care Surg. 80, 156–165 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Huang, H. et al. Relationship between heterotopic ossification and traumatic brain injury: Why severe traumatic brain injury increases the risk of heterotopic ossification. J. Orthop. Transl 12, 16–25 (2018).

    Google Scholar 

  185. Sorkin, M. et al. Regulation of heterotopic ossification by monocytes in a mouse model of aberrant wound healing. Nat Commun. https://doi.org/10.1038/s41467-019-14172-4 (2020).This work determines CD47 activation as a therapeutic approach for heterotopic ossification formation during wound healing.

  186. Agarwal, S. et al. Disruption of neutrophil extracellular traps (NETs) links mechanical strain to post-traumatic inflammation. Front. Immunol. 10, 2148 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Torossian, F. et al. Macrophage-derived oncostatin M contributes to human and mouse neurogenic heterotopic ossifications. JCI Insight 2, e96034 (2017).

    PubMed Central  Google Scholar 

  188. Hwang, C. et al. Mesenchymal VEGFA induces aberrant differentiation in heterotopic ossification. Bone Res. 7, 36 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Hsieh, H. H. S. et al. Coordinating tissue regeneration through transforming growth factor-β activated kinase 1 inactivation and reactivation. Stem Cells 37, 766–778 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Raggatt, L. J. et al. Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification. Am. J. Pathol. 184, 3192–3204 (2014).

    CAS  PubMed  Google Scholar 

  191. Agarwal, S. et al. Inhibition of Hif1α prevents both trauma-induced and genetic heterotopic ossification. Proc. Natl Acad. Sci. USA 113, E338–E347 (2016).

    CAS  PubMed  Google Scholar 

  192. Agarwal, S. et al. Scleraxis-lineage cells contribute to ectopic bone formation in muscle and tendon. Stem Cells 35, 705–710 (2017).

    CAS  PubMed  Google Scholar 

  193. Loder, S. J. et al. Characterizing the circulating cell populations in traumatic heterotopic ossification. Am. J. Pathol. 188, 2464–2473 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Dey, D. et al. Two tissue-resident progenitor lineages drive distinct phenotypes of heterotopic ossification. Sci. Transl Med. https://doi.org/10.1126/scitranslmed.aaf1090 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  195. Kan, C. et al. Gli1-labeled adult mesenchymal stem/progenitor cells and hedgehog signaling contribute to endochondral heterotopic ossification. Bone 109, 71–79 (2018).

    CAS  PubMed  Google Scholar 

  196. Eisner, C. et al. Murine tissue-resident PDGFRα+ fibro-adipogenic progenitors spontaneously acquire osteogenic phenotype in an altered inflammatory environment. J. Bone Miner. Res. (2020).

  197. Agarwal, S. et al. Analysis of bone-cartilage-stromal progenitor populations in trauma induced and genetic models of heterotopic ossification. Stem Cells 34, 1692–1701 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Agarwal, S. et al. Strategic targeting of multiple BMP receptors prevents trauma-induced heterotopic ossification. Mol. Ther. 25, 1974–1987 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Huber, A. K. et al. Immobilization after injury alters extracellular matrix and stem cell fate. J. Clin. Invest. https://doi.org/10.1172/JCI136142 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  200. Stepien, D. M. et al. Tuning macrophage phenotype to mitigate skeletal muscle fibrosis. J. Immunol. 204, 2203–2215 (2020).

    CAS  PubMed  Google Scholar 

  201. Peterson, J. R. et al. Effects of aging on osteogenic response and heterotopic ossification following burn injury in mice. Stem Cell Dev. 24, 205–213 (2015).

    CAS  Google Scholar 

  202. Ranganathan, K. et al. Role of gender in burn-induced heterotopic ossification and mesenchymal cell osteogenic differentiation. Plast. Reconstr. Surg. 135, 1631–1641 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Akiyama, H. et al. Osteo-chondroprogenitor cells are derived from Sox9 expressing precursors. Proc. Natl Acad. Sci. USA 102, 14665–14670 (2005).

    CAS  PubMed  Google Scholar 

  204. Maes, C. et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev. Cell 19, 329–344 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Greenbaum, A. et al. CXCL12 in early mesenchymal progenitors is required for haematopoietic stem-cell maintenance. Nature. 495, 227–230 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Xiong, J. et al. Osteocytes, not osteoblasts or lining cells, are the main source of the RANKL required for osteoclast formation in remodeling bone. PLoS ONE https://doi.org/10.1371/journal.pone.0138189 (2015).

  207. Pineault, K. M. et al. Hox11 genes regulate postnatal longitudinal bone growth and growth plate proliferation. Biol. Open 4, 1538–1548 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Yu, V. W. C. et al. FIAT represses ATF4-mediated transcription to regulate bone mass in transgenic mice. J. Cell Biol. 169, 591–601 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Ambrogini, E. et al. FoxO-mediated defense against oxidative stress in osteoblasts is indispensable for skeletal homeostasis in mice. Cell Metab. 11, 136–146 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Shimoyama, A. et al. Ihh/Gli2 signaling promotes osteoblast differentiation by regulating Runx2 expression and function. Mol. Biol. Cell 18, 2411–2418 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Li, J. et al. Suppressor of fused restraint of hedgehog activity level is critical for osteogenic proliferation and differentiation during calvarial bone development. J. Biol. Chem. 292, 15814–15825 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Funato, N. et al. Hand2 controls osteoblast differentiation in the branchial arch by inhibiting DNA binding of Runx2. Development 136, 615–625 (2009).

    CAS  PubMed  Google Scholar 

  213. Kanzler, B., Kuschert, S. J., Liu, Y. H. & Mallo, M. Hoxa-2 restricts the chondrogenic domain and inhibits bone formation during development of the branchial area. Development 125, 2587–2597 (1998).

    CAS  PubMed  Google Scholar 

  214. Komori, T. Regulation of osteoblast differentiation by runx2. Adv. Exp. Med. Biol. 658, 43–49 (2010).

    CAS  PubMed  Google Scholar 

  215. Hong, J. H. et al. TAZ, a transcriptional modulator of mesenchymal stem cell differentiation. Science 309, 1074–1078 (2005).

    CAS  PubMed  Google Scholar 

  216. Bialek, P. et al. A twist code determines the onset of osteoblast differentiation. Dev. Cell 6, 423–435 (2004).

    CAS  PubMed  Google Scholar 

  217. Cancela, L., Hsieh, C. L. & Francke, U. P. P. Molecular structure, chromosome assignment, and promoter organization of the human matrix Gla protein gene. J. Biol. Chem. 265, 15040–15048 (1990).

    CAS  PubMed  Google Scholar 

  218. Karsenty, G. & Park, R. W. Regulation of type I collagen genes expression. Int. Rev. Immunol. 12, 177–185 (1995).

    CAS  PubMed  Google Scholar 

  219. Pinzone, J. J. et al. The role of Dickkopf-1 in bone development, homeostasis, and disease. Blood 113, 517–525 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Kim, J. B. et al. Reconciling the roles of FAK in osteoblast differentiation, osteoclast remodeling, and bone regeneration. Bone 41, 39–51 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Li, X. et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J. Biol. Chem. 280, 19883–19887 (2005).

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Benjamin Levi or Michael T. Longaker.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks Noriaki Ono and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Osteon

A cylindrical structure consisting of a mineralized matrix and osteocytes that transports blood through connected canaliculi.

Long bone growth plate

An area of differentiating tissue located near the ends of long bones that enables physiological lengthening of the bones.

Axial skeleton

The portion of the skeleton consisting of the bones of the head and vertebrae.

Appendicular skeleton

The portion of the skeleton consisting of the bones of the appendages.

Cancellous bone

Mature adult bone consisting of spongy tissue meshwork typically found in the cores of vertebral bones and the ends of long bones.

Unicortical defect

A fracture involving only the outer and/or inner cortices on one side of the bone shaft.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Salhotra, A., Shah, H.N., Levi, B. et al. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 21, 696–711 (2020). https://doi.org/10.1038/s41580-020-00279-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-020-00279-w

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing