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.

  • Letter
  • Published:

Myostatin is a direct regulator of osteoclast differentiation and its inhibition reduces inflammatory joint destruction in mice

Abstract

Myostatin (also known as growth and differentiation factor 8) is a secreted member of the transforming growth factor-β (TGF-β) family that is mainly expressed in skeletal muscle, which is also its primary target tissue. Deletion of the myostatin gene (Mstn) in mice leads to muscle hypertrophy, and animal studies support the concept that myostatin is a negative regulator of muscle growth and regeneration1,2,3,4,5. However, myostatin deficiency also increases bone formation, mainly through loading-associated effects on bone6,7,8,9,10,11. Here we report a previously unknown direct role for myostatin in osteoclastogenesis and in the progressive loss of articular bone in rheumatoid arthritis (RA). We demonstrate that myostatin is highly expressed in the synovial tissues of RA subjects and of human tumor necrosis factor (TNF)-α transgenic (hTNFtg) mice, a model for human RA12. Myostatin strongly accelerates receptor activator of nuclear factor κB ligand (RANKL)-mediated osteoclast formation in vitro through transcription factor SMAD2-dependent regulation of nuclear factor of activated T-cells (NFATC1). Myostatin deficiency or antibody-mediated inhibition leads to an amelioration of arthritis severity in hTNFtg mice, chiefly reflected by less bone destruction. Consistent with these effects in hTNFtg mice, the lack of myostatin leads to increased grip strength and less bone erosion in the K/BxN serum-induced arthritis model in mice. The results strongly suggest that myostatin is a potent therapeutic target for interfering with osteoclast formation and joint destruction in RA.

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

Figure 1: High levels of myostatin in synovial tissues of arthritic individuals and mice.
Figure 2: Myostatin directly enhances osteoclast differentiation.
Figure 3: Deficiency or inhibition of myostatin ameliorates joint destruction in hTNFtg mice.
Figure 4: Myostatin enhances expression of key differentiation genes via SMAD2-dependent nuclear translocation of NFATC1.

Similar content being viewed by others

References

  1. McPherron, A.C., Lawler, A.M. & Lee, S.J. Regulation of skeletal muscle mass in mice by a new TGF-β superfamily member. Nature 387, 83–90 (1997).

    CAS  PubMed  Google Scholar 

  2. Huang, Z., Chen, X. & Chen, D. Myostatin: a novel insight into its role in metabolism, signal pathways, and expression regulation. Cell. Signal. 23, 1441–1446 (2011).

    CAS  PubMed  Google Scholar 

  3. Lin, J. et al. Myostatin knockout in mice increases myogenesis and decreases adipogenesis. Biochem. Biophys. Res. Commun. 291, 701–706 (2002).

    CAS  PubMed  Google Scholar 

  4. Wagner, K.R., Liu, X., Chang, X. & Allen, R.E. Muscle regeneration in the prolonged absence of myostatin. Proc. Natl. Acad. Sci. USA 102, 2519–2524 (2005).

    CAS  PubMed  Google Scholar 

  5. McCroskery, S. et al. Improved muscle healing through enhanced regeneration and reduced fibrosis in myostatin-null mice. J. Cell Sci. 118, 3531–3541 (2005).

    CAS  PubMed  Google Scholar 

  6. Kellum, E. et al. Myostatin (GDF-8) deficiency increases fracture callus size, Sox-5 expression, and callus bone volume. Bone 44, 17–23 (2009).

    CAS  PubMed  Google Scholar 

  7. Hamrick, M.W. et al. Loss of myostatin (GDF8) function increases osteogenic differentiation of bone marrow-derived mesenchymal stem cells but the osteogenic effect is ablated with unloading. Bone 40, 1544–1553 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hamrick, M.W. Increased bone mineral density in the femora of GDF8 knockout mice. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 272, 388–391 (2003).

    PubMed  Google Scholar 

  9. Hamrick, M.W., Pennington, C. & Byron, C.D. Bone architecture and disc degeneration in the lumbar spine of mice lacking GDF-8 (myostatin). J. Orthop. Res. 21, 1025–1032 (2003).

    CAS  PubMed  Google Scholar 

  10. Nicholson, E.K., Stock, S.R., Hamrick, M.W. & Ravosa, M.J. Biomineralization and adaptive plasticity of the temporomandibular joint in myostatin knockout mice. Arch. Oral Biol. 51, 37–49 (2006).

    CAS  PubMed  Google Scholar 

  11. Bialek, P. et al. A myostatin and activin decoy receptor enhances bone formation in mice. Bone 60, 162–171 (2014).

    CAS  PubMed  Google Scholar 

  12. Keffer, J. et al. Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis. EMBO J. 10, 4025–4031 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Goldring, S.R. Inflammatory mediators as essential elements in bone remodeling. Calcif. Tissue Int. 73, 97–100 (2003).

    CAS  PubMed  Google Scholar 

  14. Polzer, K. et al. Interleukin-1 is essential for systemic inflammatory bone loss. Ann. Rheum. Dis. 69, 284–290 (2010).

    CAS  PubMed  Google Scholar 

  15. Scott, D.L. et al. The links between joint damage and disability in rheumatoid arthritis. Rheumatology 39, 122–132 (2000).

    CAS  PubMed  Google Scholar 

  16. Goldring, S.R. Pathogenesis of bone erosions in rheumatoid arthritis. Curr. Opin. Rheumatol. 14, 406–410 (2002).

    PubMed  Google Scholar 

  17. Tsuboi, H. et al. Tartrate resistant acid phosphatase (TRAP) positive cells in rheumatoid synovium may induce the destruction of articular cartilage. Ann. Rheum. Dis. 62, 196–203 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Redlich, K. et al. Osteoclasts are essential for TNF-α-mediated joint destruction. J. Clin. Invest. 110, 1419–1427 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Schett, G. Cells of the synovium in rheumatoid arthritis. Osteoclasts. Arthritis Res. Ther. 9, 203 (2007).

    PubMed  PubMed Central  Google Scholar 

  20. Suzuki, Y., Nishikaku, F., Nakatuka, M. & Koga, Y. Osteoclast-like cells in murine collagen induced arthritis. J. Rheumatol. 25, 1154–1160 (1998).

    CAS  PubMed  Google Scholar 

  21. Walsh, N.C., Crotti, T.N., Goldring, S.R. & Gravallese, E.M. Rheumatic diseases: the effects of inflammation on bone. Immunol. Rev. 208, 228–251 (2005).

    CAS  PubMed  Google Scholar 

  22. Li, P. et al. RANK signalling is not required for TNF-α-mediated increase in CD11hi osteoclast precursors but is essential for mature osteoclast formation in TNF-α-mediated inflammatory arthritis. J. Bone Miner. Res. 19, 207–213 (2004).

    CAS  PubMed  Google Scholar 

  23. Lam, J. et al. TNF-α induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. J. Clin. Invest. 106, 1481–1488 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ritchlin, C.T., Haas-Smith, S.A., Li, P., Hicks, D.G. & Schwarz, E.M. Mechanisms of TNF-α- and RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis. J. Clin. Invest. 111, 821–831 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bertolini, D.R., Nedwin, G.E., Bringman, T.S., Smith, D.D. & Mundy, G.R. Stimulation of bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature 319, 516–518 (1986).

    CAS  PubMed  Google Scholar 

  26. Bradley, L., Yaworsky, P.J. & Walsh, F.S. Myostatin as a therapeutic target for musculoskeletal disease. Cell. Mol. Life Sci. 65, 2119–2124 (2008).

    CAS  PubMed  Google Scholar 

  27. Fuller, K., Bayley, K.E. & Chambers, T.J. Activin A is an essential cofactor for osteoclast induction. Biochem. Biophys. Res. Commun. 268, 2–7 (2000).

    CAS  PubMed  Google Scholar 

  28. Sugatani, T., Alvarez, U.M. & Hruska, K.A. Activin A stimulates IκB-α/NFκB and RANK expression for osteoclast differentiation, but not AKT survival pathway in osteoclast precursors. J. Cell. Biochem. 90, 59–67 (2003).

    CAS  PubMed  Google Scholar 

  29. Tani-Ishii, N., Tsunoda, A., Teranaka, T. & Umemoto, T. Autocrine regulation of osteoclast formation and bone resorption by IL-1 alpha and TNF alpha. J. Dent. Res. 78, 1617–1623 (1999).

    CAS  PubMed  Google Scholar 

  30. Korb-Pap, A. et al. Early structural changes in cartilage and bone are required for the attachment and invasion of inflamed synovial tissue during destructive inflammatory arthritis. Ann. Rheum. Dis. 71, 1004–1011 (2012).

    CAS  PubMed  Google Scholar 

  31. Lee, Z.H. & Kim, H.H. Signal transduction by receptor activator of nuclear factor kappa B in osteoclasts. Biochem. Biophys. Res. Commun. 305, 211–214 (2003).

    CAS  PubMed  Google Scholar 

  32. Lee, S.E. et al. The phosphatidylinositol 3-kinase, p38, and extracellular signal-regulated kinase pathways are involved in osteoclast differentiation. Bone 30, 71–77 (2002).

    CAS  PubMed  Google Scholar 

  33. Li, X. et al. p38 MAPK-mediated signals are required for inducing osteoclast differentiation but not for osteoclast function. Endocrinology 143, 3105–3113 (2002).

    CAS  PubMed  Google Scholar 

  34. Omata, Y. et al. Genome-wide comprehensive analysis reveals critical cooperation between Smad and c-Fos in RANKL-induced osteoclastogenesis. J. Bone Miner. Res. 30, 869–877 (2015).

    CAS  PubMed  Google Scholar 

  35. Li, Z.B., Kollias, H.D. & Wagner, K.R. Myostatin directly regulates skeletal muscle fibrosis. J. Biol. Chem. 283, 19371–19378 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Machida, H., Ogawa, K., Funaba, M., Mizutani, T. & Tsujimoto, M. mRNA expression of type I and type II receptors for activin, transforming growth factor-beta, and bone morphogenetic protein in the murine erythroleukemic cell line, F5-5.fl. Eur. J. Endocrinol. 143, 705–710 (2000).

    CAS  PubMed  Google Scholar 

  37. Kaneto, H. et al. Increased expression of TGF-β1 but not its receptor contributes to human obstructive nephropathy. Kidney Int. 56, 2137–2146 (1999).

    CAS  PubMed  Google Scholar 

  38. Inman, G.J. et al. SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 62, 65–74 (2002).

    CAS  PubMed  Google Scholar 

  39. Holzbaur, E.L. et al. Myostatin inhibition slows muscle atrophy in rodent models of amyotrophic lateral sclerosis. Neurobiol. Dis. 23, 697–707 (2006).

    CAS  PubMed  Google Scholar 

  40. Monach, P.A., Mathis, D. & Benoist, C. The K/BxN arthritis model. Curr. Protoc. Immunol. 81, 15.22.1–15.22.12 (2008).

    Google Scholar 

  41. Martinez, G.J. et al. Smad2 positively regulates the generation of Th17 cells. J. Biol. Chem. 285, 29039–29043 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S.-J. Lee (Johns Hopkins University School of Medicine) for providing us with the Mstn−/− mice, and G. Kollias (Alexander Fleming Biomedical Sciences Research Center) for the hTNF transgenic mice. We also thank Pfizer, Inc. for providing us with the myostatin-specific antibody. We thank X. Lin (Department of Surgery, Baylor College of Medicine) for providing us with the SMAD2 constructs. We thank V. Eckervogt and B. Truckenbrod for excellent technical support. This work was supported by the German Research Foundation (Deutsche Forschungsgemeinschaft; DA 1143/4-1 and DA 1143/4-2 to B.D. and T.P. as part of the Priority Programme SPP 1468, IMMUNOBONE) and by the Collaborative Research Center SFB 1009 granted to T.P.

Author information

Authors and Affiliations

Authors

Contributions

B.D. designed and performed all experiments and wrote the manuscript; C.K.-W. and C.W. participated in the histomorphometric analyses; A.S., A.K-P. and P.P. contributed to experimental design and prepared mice and tissues and participated in the antibody experiments; D. Brunert performed the ELISA experiments and prepared mice; S.H. and K.R. conducted the studies with the K/BxN mouse model; S.F. participated in the transfection studies and performed the luciferase assays; M.F., J.B. and D. Beckmann performed the qPCR and the co-culture experiments; and T.P. participated in data analysis, directed the project and wrote the manuscript.

Corresponding author

Correspondence to Thomas Pap.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1 and 2 (PDF 15337 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dankbar, B., Fennen, M., Brunert, D. et al. Myostatin is a direct regulator of osteoclast differentiation and its inhibition reduces inflammatory joint destruction in mice. Nat Med 21, 1085–1090 (2015). https://doi.org/10.1038/nm.3917

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3917

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research