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.

  • Article
  • Published:

Wnt-modified materials mediate asymmetric stem cell division to direct human osteogenic tissue formation for bone repair

This article has been updated

Abstract

The maintenance of human skeletal stem cells (hSSCs) and their progeny in bone defects is a major challenge. Here, we report on a transplantable bandage containing a three-dimensional Wnt-induced osteogenic tissue model (WIOTM). This bandage facilitates the long-term viability of hSSCs (8 weeks) and their progeny, and enables bone repair in an in vivo mouse model of critical-sized calvarial defects. The newly forming bone is structurally comparable to mature cortical bone and consists of human and murine cells. Furthermore, we show that the mechanism of WIOTM formation is governed by Wnt-mediated asymmetric cell division of hSSCs. Covalently immobilizing Wnts onto synthetic materials can polarize single dividing hSSCs, orient the spindle and simultaneously generate a Wnt-proximal hSSC and a differentiation-prone Wnt-distal cell. Our results provide insight into the regulation of human osteogenesis and represent a promising approach to deliver human osteogenic constructs that can survive in vivo and contribute to bone repair.

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: Localized Wnt3a orients division in hSSCs and induces asymmetric distribution of Wnt/β-catenin pathway components.
Fig. 2: Localized Wnt3a induces asymmetric distribution of cell fate markers in dividing cells and the WIOTM.
Fig. 3: In vitro functional characterization of the Wnt3a-bandage.
Fig. 4: In vivo functional evaluation of Wnt3a- and WIOTM-bandages in the repair of critical-sized bone defects.
Fig. 5: In vivo characterization of bone formation driven by Wnt3a- and WIOTM-bandages.
Fig. 6: In vivo characterization of connective-like tissue of bone defects treated with the bandages.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

Data availability

All numerical data included in the production of figures, plots and statistical analysis can be found in the figures, main text and Source Data. Additional statistical analysis values (F values, t values, degrees of freedom and P values) are presented in the Source Data documents. Source data are provided with this paper.

Change history

  • 16 October 2020

    In the version of this Article originally published, the links to the Supplementary Information and Source Data files were jumbled; these have now been corrected.

References

  1. Fernandez de Grado, G. et al. Bone substitutes: a review of their characteristics, clinical use, and perspectives for large bone defects management. J. Tissue Eng. 9, 2041731418776819 (2018).

    Google Scholar 

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

    CAS  Google Scholar 

  3. Zeitouni, S. et al. Human mesenchymal stem cell-derived matrices for enhanced osteoregeneration. Sci. Transl. Med. 4, 132ra55 (2012).

    Google Scholar 

  4. Tsai, T.-L. & Li, W.-J. Identification of bone marrow-derived soluble factors regulating human mesenchymal stem cells for bone regeneration. Stem Cell Reports 8, 387–400 (2017).

    CAS  Google Scholar 

  5. Krause, U. et al. Pharmaceutical modulation of canonical Wnt signaling in multipotent stromal cells for improved osteoinductive therapy. Proc. Natl Acad. Sci. USA 107, 4147–4152 (2010).

    CAS  Google Scholar 

  6. Im, J.-Y. et al. Bone regeneration of mouse critical-sized calvarial defects with human mesenchymal stem cells in scaffold. Lab. Anim. Res. 29, 196–203 (2013).

    Google Scholar 

  7. Brennan, M. Á. et al. Pre-clinical studies of bone regeneration with human bone marrow stromal cells and biphasic calcium phosphate. Stem Cell Res. Ther. 5, 114–115 (2014).

    Google Scholar 

  8. Nakahara, H. et al. Bone repair by transplantation of hTERT-immortalized human mesenchymal stem cells in mice. Transplantation 88, 346–353 (2009).

    Google Scholar 

  9. Lowndes, M., Junyent, S. & Habib, S. J. Constructing cellular niche properties by localized presentation of Wnt proteins on synthetic surfaces. Nat. Protoc. 12, 1498–1512 (2017).

    CAS  Google Scholar 

  10. Garcin, C. L. & Habib, S. J. A comparative perspective on Wnt/β-catenin signalling in cell fate determination. Results Probl. Cell Differ. 61, 323–350 (2017).

    CAS  Google Scholar 

  11. Mills, K. M., Szczerkowski, J. L. A. & Habib, S. J. Wnt ligand presentation and reception: from the stem cell niche to tissue engineering. Open Biol. 7, 170140 (2017).

    Google Scholar 

  12. Junyent, S. et al. Specialized cytonemes induce self-organization of stem cells. Proc. Natl Acad. Sci. USA 117, 7236–7244 (2020).

    CAS  Google Scholar 

  13. Habib, S. J. et al. A localized Wnt signal orients asymmetric stem cell division in vitro. Science 339, 1445–1448 (2013).

    CAS  Google Scholar 

  14. Lowndes, M., Rotherham, M., Price, J. C., Haj El, A. J. & Habib, S. J. Immobilized WNT proteins act as a stem cell niche for tissue engineering. Stem Cell Reports 7, 126–137 (2016).

    CAS  Google Scholar 

  15. Schlessinger, K., McManus, E. J. & Hall, A. Cdc42 and noncanonical Wnt signal transduction pathways cooperate to promote cell polarity. J. Cell Biol. 178, 355–361 (2007).

    CAS  Google Scholar 

  16. Inaba, M. & Yamashita, Y. M. Asymmetric stem cell division: precision for robustness. Cell Stem Cell 11, 461–469 (2012).

    CAS  Google Scholar 

  17. Holley, R. J. et al. Comparative quantification of the surfaceome of human multipotent mesenchymal progenitor cells. Stem Cell Reports 4, 473–488 (2015).

    CAS  Google Scholar 

  18. Zohar, R., Cheifetz, S., McCulloch, C. A. & Sodek, J. Analysis of intracellular osteopontin as a marker of osteoblastic cell differentiation and mesenchymal cell migration. Eur. J. Oral Sci. 106 (Suppl. 1), 401–407 (1998).

    CAS  Google Scholar 

  19. Junaid, A., Moon, M. C., Harding, G. E. J. & Zahradka, P. Osteopontin localizes to the nucleus of 293 cells and associates with polo-like kinase-1. Am. J. Physiol. Cell Physiol. 292, C919–C926 (2007).

    CAS  Google Scholar 

  20. Hutmacher, D. W. & Sittinger, M. Periosteal cells in bone tissue engineering. Tissue Eng. 9 (Suppl. 1), S45–S64 (2003).

    CAS  Google Scholar 

  21. Malikmammadov, E., Tanir, T. E., Kiziltay, A., Hasirci, V. & Hasirci, N. PCL and PCL-based materials in biomedical applications. J. Biomater. Sci. Polym. Ed. 29, 863–893 (2018).

    CAS  Google Scholar 

  22. Zhu, Y., Gao, C., Liu, X. & Shen, J. Surface modification of polycaprolactone membrane via aminolysis and biomacromolecule immobilization for promoting cytocompatibility of human endothelial cells. Biomacromolecules 3, 1312–1319 (2002).

    CAS  Google Scholar 

  23. Doro, D. H., Grigoriadis, A. E. & Liu, K. J. Calvarial suture-derived stem cells and their contribution to cranial bone repair. Front. Physiol. 8, 956 (2017).

    Google Scholar 

  24. Zhao, H. et al. The suture provides a niche for mesenchymal stem cells of craniofacial bones. Nat. Cell Biol. 17, 386–396 (2015).

    CAS  Google Scholar 

  25. Maruyama, T., Jeong, J., Sheu, T.-J. & Hsu, W. Stem cells of the suture mesenchyme in craniofacial bone development, repair and regeneration. Nat. Commun. 7, 10526 (2016).

    CAS  Google Scholar 

  26. Wilk, K. et al. Postnatal calvarial skeletal stem cells expressing PRX1 reside exclusively in the calvarial sutures and are required for bone regeneration. Stem Cell Reports 8, 933–946 (2017).

    CAS  Google Scholar 

  27. Morinobu, M. et al. Osteopontin expression in osteoblasts and osteocytes during bone formation under mechanical stress in the calvarial suture in vivo. J. Bone Miner. Res. 18, 1706–1715 (2003).

    CAS  Google Scholar 

  28. Bonewald, L. F. The amazing osteocyte. J. Bone Miner. Res. 26, 229–238 (2011).

    CAS  Google Scholar 

  29. 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  Google Scholar 

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

    CAS  Google Scholar 

  31. Goldstein, B., Takeshita, H., Mizumoto, K. & Sawa, H. Wnt signals can function as positional cues in establishing cell polarity. Dev. Cell 10, 391–396 (2006).

    CAS  Google Scholar 

  32. Corish, P. & Tyler-Smith, C. Attenuation of green fluorescent protein half-life in mammalian cells. Protein Eng. 12, 1035–1040 (1999).

    CAS  Google Scholar 

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

    Google Scholar 

  34. Murphy, M. P., Quarto, N., Longaker, M. T. & Wan, D. C. * Calvarial defects: cell-based reconstructive strategies in the murine model. Tissue Eng. C 23, 971–981 (2017).

    Google Scholar 

  35. Epstein, N. E. Pros, cons, and costs of INFUSE in spinal surgery. Surg. Neurol. Int 2, 10 (2011).

    Google Scholar 

  36. Baht, G. S., Vi, L. & Alman, B. A. The role of the immune cells in fracture healing. Curr. Osteoporos. Rep. 16, 138–145 (2018).

    Google Scholar 

  37. Keeney, M. et al. Scaffold-mediated BMP-2 minicircle DNA delivery accelerated bone repair in a mouse critical-size calvarial defect model. J. Biomed. Mater. Res. A 104, 2099–2107 (2016).

    CAS  Google Scholar 

  38. Inoue, S., Otsuka, H., Takito, J. & Nakamura, M. Decisive differences in the bone repair processes of the metaphysis and diaphysis in young mice. Bone Rep. 8, 1–8 (2018).

    Google Scholar 

  39. Lu, C. et al. Cellular basis for age-related changes in fracture repair. J. Orthop. Res. 23, 1300–1307 (2005).

    CAS  Google Scholar 

  40. Levi, B. et al. In vivo directed differentiation of pluripotent stem cells for skeletal regeneration. Proc. Natl Acad. Sci. USA 109, 20379–20384 (2012).

    CAS  Google Scholar 

  41. Levi, B. et al. Human adipose derived stromal cells heal critical size mouse calvarial defects. PLoS ONE 5, e11177 (2010).

    Google Scholar 

  42. Hyun, J. et al. Enhancing in vivo survival of adipose-derived stromal cells through Bcl-2 overexpression using a minicircle vector. Stem Cells Transl. Med. 2, 690–702 (2013).

    CAS  Google Scholar 

  43. Lough, D. et al. Regeneration of vascularized corticocancellous bone and diploic space using muscle-derived stem cells: a translational biologic alternative for healing critical bone defects. Plast. Reconstr. Surg. 139, 893–905 (2017).

    CAS  Google Scholar 

  44. Fan, J. et al. Delivery of phenamil enhances BMP-2-induced osteogenic differentiation of adipose-derived stem cells and bone formation in calvarial defects. Tissue Eng. A 21, 2053–2065 (2015).

    CAS  Google Scholar 

  45. Zhang, F. & King, M. W. Biodegradable polymers as the pivotal player in the design of tissue engineering scaffolds. Adv. Healthc. Mater. 264, e1901358 (2020).

    Google Scholar 

  46. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

    Google Scholar 

  47. Penfornis, P. & Pochampally, R. Isolation and expansion of mesenchymal stem cells/multipotential stromal cells from human bone marrow. Methods Mol. Biol. 698, 11–21 (2011).

    CAS  Google Scholar 

  48. Willert, K. K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 (2003).

    CAS  Google Scholar 

  49. Mikels, A. J. & Nusse, R. Purified Wnt5a protein activates or inhibits β-catenin–TCF signaling depending on receptor context. PLoS Biol. 4, e115 (2006).

    Google Scholar 

  50. Zen Blue Edition 2.5 software (Zeiss, 2020).

  51. NIS-Elements Viewer 5.2 software (Nikon, 2020).

  52. Leica Application Suite X 3.7 software (Leica, 2020).

  53. ImageJ (Fiji, 2020).

  54. Volocity 6.5 (Quorum Technologies, 2020).

  55. Imaris 9.5 (Oxford Instruments, 2020).

  56. Operetta High-Content Imaging System (PerkinElmer, 2020).

  57. Image Lab 6.1 (Bio-Rad, 2020).

  58. Parallax Microview software (Parallax Innovations, 2020).

  59. NDP.view 2.5 (Hamamatsu Photonics, 2020).

  60. Prism 8 (GraphPad Software, 2020).

Download references

Acknowledgements

We thank A. Streit, E. Gentleman, A. Grants and H. Drukmann for discussions about the project. We thank P. Sharpe, T. Desai, N. Ali and M. Tewary for critical reading of the manuscript. We thank C. Healy for his help with µCT imaging. We acknowledge financial support from the Department of Health through the National Institute for Health Research (NIHR) Comprehensive Biomedical Research Centre award to the Guy’s & St Thomas’ National Health Service Foundation Trust in partnership with King’s College London and the King’s College Hospital NHS Foundation Trust. K.J.L. and D.H.D. received funding from CAPES, Dental Institute Seed Funding and the BBSRC. This project was supported mainly by a Sir Henry Dale Fellowship (102513/Z/13/Z, S.J.H.). Additional financial support from the UK Regenerative Medicine Platform (MR/R015635/1, S.J.H.) and a London Advanced Therapy Award (S.J.H.) is also acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

S.J.H. conceived the project and designed the experiments. J.R., Y.O., S.S.N., S.J. and S.J.H. performed and analysed the in vitro experiments. Y.O. and D.H.D. performed the in vivo experiments. Y.O. and J.R. analysed the µCT data. J.R., S.S.N. and S.J. performed the histological analysis. J.R. and S.J.H. interpreted the data. A.J.E.H. and K.J.L. commented on the in vivo experiments. J.R., S.J. and S.J.H. prepared the figures. S.J.H. wrote the manuscript. All authors commented on the manuscript. S.J.H. led the project and provided financial support for the project.

Corresponding author

Correspondence to Shukry J. Habib.

Ethics declarations

Competing interests

King’s College London has filed a patent application (P6125GB00) on Tissue Regeneration Patch that includes the Wnt-bandage and WIOTM-bandage.

Additional information

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

Supplementary information

Source data

Source Data Fig. 1

Numerical and statistical source data for Fig. 1.

Source Data Fig. 2

Numerical and statistical source data for Fig. 2.

Source Data Fig. 3

Numerical and statistical source data for Fig. 3.

Source Data Fig. 3 images

Numerical and statistical source data for Fig. 3 images.

Source Data Fig. 4

Numerical and statistical source data for Fig. 4.

Source Data Fig. 5

Numerical and statistical source data for Fig. 5.

Source Data Fig. 6

Numerical and statistical source data for Fig. 6.

Source Data Supplementary Fig. 1

Numerical and statistical source data for Supplementary Fig. 1.

Source Data Supplementary Fig. 2

Numerical and statistical source data for Supplementary Fig. 3.

Source Data Supplementary Fig. 3

Numerical and statistical source data for Supplementary Fig. 4

Source Data Supplementary Fig. 4

Numerical and statistical source data for Supplementary Fig. 5.

Source Data Supplementary Fig. 5

Numerical and statistical source data for Supplementary Fig. 6.

Source Data Supplementary Fig. 6

Numerical and statistical source data for Supplementary Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Okuchi, Y., Reeves, J., Ng, S.S. et al. Wnt-modified materials mediate asymmetric stem cell division to direct human osteogenic tissue formation for bone repair. Nat. Mater. 20, 108–118 (2021). https://doi.org/10.1038/s41563-020-0786-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-020-0786-5

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