Skip to main content

Advertisement

Log in

Constant hypoxia inhibits osteoclast differentiation and bone resorption by regulating phosphorylation of JNK and IκBα

  • Original Research Paper
  • Published:
Inflammation Research Aims and scope Submit manuscript

Abstract

Background

Osteoclasts are responsible for the bone loss in rheumatoid arthritis (RA). Hypoxia has been suggested to play key roles in pathological bone loss. However, the current understanding of the effects of hypoxia on osteoclastogenesis is controversial. Effects of hypoxia on both the formation and function of osteoclasts requires examination. In the current study, we aimed to explore the effect of hypoxia on osteoclast differentiation and the underlying mechanisms.

Methods

RAW264.7 cells and murine bone-marrow-derived monocytes were used to induce osteoclastogenesis in the presence of macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa B ligand (RANKL). Hypoxic conditions were maintained in a hypoxic chamber at 5% CO2 and 1% O2, balanced with N2. Osteoclasts were detected by tartrate-resistant acid phosphatase (TRAP) staining. A bone resorption assay was carried out in vitro using bone slices. RT-PCR was conducted to detect osteoclast markers and transcription factors. The phosphorylation of nuclear factor-κBα (IκBα), c-Jun N-terminal kinase (JNK), extracellular regulated protein kinase (ERK), and p38 was detected by western blotting. Mann–Whitney U test or Student’s t test was used to compare differences between the two groups.

Results

TRAP staining and the bone resorption assay revealed that hypoxia-restrained osteoclast differentiation and bone resorption. Expression of osteoclast markers including cathepsin K, RANK, and TRAP decreased during osteoclast differentiation under hypoxic conditions (all P < 0.05). Hypoxia at 1% O2 did not affect cell viability, whereas it dramatically abated RANKL-dependent phosphorylation of the JNK-mitogen-activated protein kinases (MAPK) and IκBα pathways. Moreover, the expression of nuclear factor of activated T-cell cytoplasmic 1 (NFATc1) was inhibited under hypoxic conditions (all P < 0.05).

Conclusions

These results suggest that constant hypoxia at 1% O2 significantly restrains osteoclast formation and resorbing function without affecting cell viability. Constant hypoxia might inhibit RANKL-induced osteoclastogenesis by regulating NFATc1 expression via interfering the phosphorylation of JNK and IκBα.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Smolen JS, Aletaha D, Barton A, Burmester GR, Emery P, Firestein GS, et al. Rheumatoid arthritis. Nat Rev Dis Primers. 2018;4:18001. https://doi.org/10.1038/nrdp.2018.1.

    Article  PubMed  Google Scholar 

  2. Li ZG. Facing the challenge of low recognition and high disability in rheumatoid arthritis in Chinese. Natl Med J Chin. 2009;89:1873–5.

    Google Scholar 

  3. Ji J, Zhang L, Zhang Q, Yin R, Fu T, Li L, et al. Functional disability associated with disease and quality-of-life parameters in Chinese patients with rheumatoid arthritis. Health Qual Life Outcomes. 2017;15:89. https://doi.org/10.1186/s12955-017-0659-z.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Tanaka Y, Ohira T. Mechanisms and therapeutic targets for bone damage in rheumatoid arthritis, in particular the RANK-RANKL system. Curr Opin Pharmacol. 2018;40:110–9. https://doi.org/10.1016/j.coph.2018.03.006.

    Article  CAS  PubMed  Google Scholar 

  5. Maurizi A, Rucci N. The osteoclast in bone metastasis: player and target. Cancers Basel. 2018. https://doi.org/10.3390/cancers10070218.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Ugay L, Kochetkova E, Nevzorova V, Maistrovskaia Y. Role of osteoprotegerin and receptor activator of nuclear factor-kappaB ligand in bone loss related to advanced chronic obstructive pulmonary disease. Chin Med J Engl. 2016;129:1696–703. https://doi.org/10.4103/0366-6999.185857.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Wang RY, Yang SH, Xu WH. Role of epithelium sodium channel in bone formation. Chin Med J. 2016;129:594–600. https://doi.org/10.4103/0366-6999.176994.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of osteoclast differentiation by cytokine networks. Immun Netw. 2018;18:e8. https://doi.org/10.4110/in.2018.18.e8.

    Article  Google Scholar 

  9. Nanke Y, Kobashigawa T, Yago T, Kawamoto M, Yamanaka H, Kotake S. RANK expression and osteoclastogenesis in human monocytes in peripheral blood from rheumatoid arthritis patients. Biomed Res Int. 2016;2016:4874195. https://doi.org/10.1155/2016/4874195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. An J, Hao D, Zhang Q, Chen B, Zhang R, Wang Y, et al. Natural products for treatment of bone erosive diseases: the effects and mechanisms on inhibiting osteoclastogenesis and bone resorption. Int Immunopharmacol. 2016;36:118–31. https://doi.org/10.1016/j.intimp.2016.04.024.

    Article  CAS  PubMed  Google Scholar 

  11. Ye H, Arron JR, Lamothe B, Cirilli M, Kobayashi T, Shevde NK, et al. Distinct molecular mechanism for initiating TRAF6 signalling. Nature. 2002;418:443–7. https://doi.org/10.1038/nature00888.

    Article  CAS  PubMed  Google Scholar 

  12. Li L, Sapkota M, Gao M, Choi H, Soh Y. Macrolactin F inhibits RANKL-mediated osteoclastogenesis by suppressing Akt, MAPK and NFATc1 pathways and promotes osteoblastogenesis through a BMP-2/smad /Akt/Runx2 signaling pathway. Eur J Pharmacol. 2017;815:202–9. https://doi.org/10.1016/j.ejphar.2017.09.015.

    Article  CAS  PubMed  Google Scholar 

  13. Oh JH, Lee NK. Up-regulation of RANK expression via ERK1/2 by insulin contributes to the enhancement of osteoclast differentiation. Mol Cells. 2017;40:371–7. https://doi.org/10.14348/molcells.2017.0025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cong Q, Jia H, Li P, Qiu S, Yeh J, Wang Y, et al. p38alpha MAPK regulates proliferation and differentiation of osteoclast progenitors and bone remodeling in an aging-dependent manner. Sci Rep. 2017;7:45964. https://doi.org/10.1038/srep45964.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond. Front Immunol 2014;5:511. https://doi.org/10.3389/fimmu.2014.00511.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Park JH, Lee NK, Lee SY. Current understanding of RANK signaling in osteoclast differentiation and maturation. Mol Cells. 2017;40:706–13. https://doi.org/10.14348/molcells.2017.0225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kim H, Choi HK, Shin JH, Kim KH, Huh JY, Lee SA, et al. Selective inhibition of RANK blocks osteoclast maturation and function and prevents bone loss in mice. J Clin Invest. 2009;119:813–25. https://doi.org/10.1172/JCI36809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shinohara M, Takayanagi H. Analysis of NFATc1-centered transcription factor regulatory networks in osteoclast formation. Methods Mol Biol. 2014;1164:171–6. https://doi.org/10.1007/978-1-4939-0805-9_14.

    Article  CAS  PubMed  Google Scholar 

  19. Oikawa T, Oyama M, Kozuka-Hata H, Uehara S, Udagawa N, Saya H, et al. Tks5-dependent formation of circumferential podosomes/invadopodia mediates cell-cell fusion. J Cell Biol. 2012;197:553–68. https://doi.org/10.1083/jcb.201111116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ono T, Nakashima T. Recent advances in osteoclast biology. Histochem Cell Biol. 2018;149:325–41. https://doi.org/10.1007/s00418-018-1636-2.

    Article  CAS  PubMed  Google Scholar 

  21. Matsumoto M, Kogawa M, Wada S, Takayanagi H, Tsujimoto M, Katayama S, et al. Essential role of p38 mitogen-activated protein kinase in cathepsin K gene expression during osteoclastogenesis through association of NFATc1 and PU.1. J Biol Chem. 2004;279:45969–79. https://doi.org/10.1074/jbc.M408795200.

    Article  CAS  PubMed  Google Scholar 

  22. Zhou F, Shen Y, Liu B, Chen X, Wan L, Peng D. Gastrodin inhibits osteoclastogenesis via down-regulating the NFATc1 signaling pathway and stimulates osseointegration in vitro. Biochem Biophys Res Commun. 2017;484:820–6. https://doi.org/10.1016/j.bbrc.2017.01.179.

    Article  CAS  PubMed  Google Scholar 

  23. Sivakumar B, Akhavani MA, Winlove CP, Taylor PC, Paleolog EM, Kang N. Synovial hypoxia as a cause of tendon rupture in rheumatoid arthritis. J Hand Surg Am. 2008;33:49–58. https://doi.org/10.1016/j.jhsa.2007.09.002.

    Article  PubMed  Google Scholar 

  24. Lund-Olesen K. Oxygen tension in synovial fluids. Arthritis Rheum. 1970;13:769–76. https://doi.org/10.1002/art.1780130606.

    Article  CAS  PubMed  Google Scholar 

  25. Treuhaft PS, DJ MC. Synovial fluid pH, lactate, oxygen and carbon dioxide partial pressure in various joint diseases. Arthritis Rheum. 1971;14:475–84. https://doi.org/10.1002/art.1780140407.

    Article  CAS  PubMed  Google Scholar 

  26. Knowles HJ. Hypoxic regulation of osteoclast differentiation and bone resorption activity. Hypoxia Auckl. 2015;3:73–82. https://doi.org/10.2147/HP.S95960.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Arnett TR, Gibbons DC, Utting JC, Orriss IR, Hoebertz A, Rosendaal M, et al. Hypoxia is a major stimulator of osteoclast formation and bone resorption. J Cell Physiol. 2003;196:2–8. https://doi.org/10.1002/jcp.10321.

    Article  CAS  PubMed  Google Scholar 

  28. Utting JC, Flanagan AM, Brandao-Burch A, Orriss IR, Arnett TR. Hypoxia stimulates osteoclast formation from human peripheral blood. Cell Biochem Funct. 2010;28:374–80. https://doi.org/10.1002/cbf.1660.

    Article  CAS  PubMed  Google Scholar 

  29. Muzylak M, Price JS, Horton MA. Hypoxia induces giant osteoclast formation and extensive bone resorption in the cat. Calcif Tissue Int. 2006;79:301–9. https://doi.org/10.1007/s00223-006-0082-7.

    Article  CAS  PubMed  Google Scholar 

  30. Leger AJ, Altobelli A, Mosquea LM, Belanger AJ, Song A, Cheng SH, et al. Inhibition of osteoclastogenesis by prolyl hydroxylase inhibitor dimethyloxallyl glycine. J Bone Miner Metab. 2010;28:510–9. https://doi.org/10.1007/s00774-010-0171-6.

    Article  CAS  PubMed  Google Scholar 

  31. Kang H, Yang K, Xiao L, Guo L, Guo C, Yan Y, et al. Osteoblast hypoxia-inducible factor-1alpha pathway activation restrains osteoclastogenesis via the interleukin-33-MicroRNA-34a-Notch1 pathway. Front Immunol. 2017;8:1312. https://doi.org/10.3389/fimmu.2017.01312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hulley PA, Bishop T, Vernet A, Schneider JE, Edwards JR, Athanasou NA, et al. Hypoxia-inducible factor 1-alpha does not regulate osteoclastogenesis but enhances bone resorption activity via prolyl-4-hydroxylase 2. J Pathol. 2017;242:322–33. https://doi.org/10.1002/path.4906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yu R, Li C, Sun L, Jian L, Ma Z, Zhao J, et al. Hypoxia induces production of citrullinated proteins in human fibroblast-like synoviocytes through regulating HIF1alpha. Scand J Immunol. 2018;87:e12654. https://doi.org/10.1111/sji.12654.

    Article  CAS  PubMed  Google Scholar 

  34. Knowles HJ, Athanasou NA. Acute hypoxia and osteoclast activity: a balance between enhanced resorption and increased apoptosis. J Pathol. 2009;218:256–64. https://doi.org/10.1002/path.2534.

    Article  CAS  PubMed  Google Scholar 

  35. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11:393–410. https://doi.org/10.1038/nrc3064.

    Article  CAS  PubMed  Google Scholar 

  36. Wouters BG, Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat Rev Cancer. 2008;8:851–64. https://doi.org/10.1038/nrc2501.

    Article  CAS  PubMed  Google Scholar 

  37. Shao J, Zhang Y, Yang T, Qi J, Zhang L, Deng L. HIF-1alpha disturbs osteoblasts and osteoclasts coupling in bone remodeling by up-regulating OPG expression. In Vitro Cell Dev Biol Anim. 2015;51:808–14. https://doi.org/10.1007/s11626-015-9895-x.

    Article  CAS  PubMed  Google Scholar 

  38. Saleh H, Eeles D, Hodge JM, Nicholson GC, Gu R, Pompolo S, et al. Interleukin-33, a target of parathyroid hormone and oncostatin m, increases osteoblastic matrix mineral deposition and inhibits osteoclast formation in vitro. Endocrinology. 2011;152:1911–22. https://doi.org/10.1210/en.2010-1268.

    Article  CAS  PubMed  Google Scholar 

  39. Schulze J, Bickert T, Beil FT, Zaiss MM, Albers J, Wintges K, et al. Interleukin-33 is expressed in differentiated osteoblasts and blocks osteoclast formation from bone marrow precursor cells. J Bone Miner Res. 2011;26:704–17. https://doi.org/10.1002/jbmr.269.

    Article  CAS  PubMed  Google Scholar 

  40. Wu C, Rankin EB, Castellini L, Alcudia JF, LaGory EL, Andersen R, et al. Oxygen-sensing PHDs regulate bone homeostasis through the modulation of osteoprotegerin. Genes Dev. 2015;29:817–31. https://doi.org/10.1101/gad.255.000.114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Song F, Zhou L, Zhao J, Liu Q, Yang M, Tan R, et al. Eriodictyol inhibits RANKL-induced osteoclast formation and function via inhibition of NFATC1 activity. J Cell Physiol. 2016;231(9):1983–93. https://doi.org/10.1002/jcp.25304.

    Article  CAS  PubMed  Google Scholar 

  42. Quiñonez-Flores CM, González-Chávez SA, Pacheco-Tena C. Hypoxia and its implications in rheumatoid arthritis. J Biomed Sci. 2016;23(1):62. https://doi.org/10.1186/s12929-016-0281-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We gratefully thank the Medical Research Center of Peking University Third Hospital for kindly providing the RAW264.7 cells. We thank the Medical Research Center of Peking University Third Hospital for providing experimental equipment and technical support.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81471599) and the National Young Scholars Foundation of China (No. 81501387), the Prairie Fire Program (LYJH-92).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Changhong Li or Xiangyuan Liu.

Ethics declarations

Conflict of interest

There are no conflicts of interest.

Additional information

Responsible Editor: Jason J. McDougall.

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 20 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, Z., Yu, R., Zhao, J. et al. Constant hypoxia inhibits osteoclast differentiation and bone resorption by regulating phosphorylation of JNK and IκBα. Inflamm. Res. 68, 157–166 (2019). https://doi.org/10.1007/s00011-018-1209-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00011-018-1209-9

Keywords

Navigation