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.

  • Protocol
  • Published:

Isolation and functional assessment of mouse skeletal stem cell lineage

Abstract

There are limited methods available to study skeletal stem, progenitor, and progeny cell activity in normal and diseased contexts. Most protocols for skeletal stem cell isolation are based on the extent to which cells adhere to plastic or whether they express a limited repertoire of surface markers. Here, we describe a flow cytometry–based approach that does not require in vitro selection and that uses eight surface markers to distinguish and isolate mouse skeletal stem cells (mSSCs); bone, cartilage, and stromal progenitors (mBCSPs); and five downstream differentiated subtypes, including chondroprogenitors, two types of osteoprogenitors, and two types of hematopoiesis-supportive stroma. We provide instructions for the optimal mechanical and chemical digestion of bone and bone marrow, as well as the subsequent flow-cytometry-activated cell sorting (FACS) gating schemes required to maximally yield viable skeletal-lineage cells. We also describe a methodology for renal subcapsular transplantation and in vitro colony-formation assays on the isolated mSSCs. The isolation of mSSCs can be completed in 9 h, with at least 1 h more required for transplantation. Experience with flow cytometry and mouse surgical procedures is recommended before attempting the protocol. Our system has wide applications and has already been used to study skeletal response to fracture, diabetes, and osteoarthritis, as well as hematopoietic stem cell–niche interactions in the bone marrow.

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: Overview of the protocol for skeletal stem cell lineage isolation and functional assessment.
Figure 2: Schema for mechanical digestion of skeletal tissue.
Figure 3: Skeletal stem cell lineage hierarchy and flow cytometry gating scheme.
Figure 4: Fluorescence-minus-one (FMO) controls for mouse skeletal lineage gating strategy.
Figure 5: Schema for renal subcapsular transplantation of skeletal-lineage cells.
Figure 6: Movat pentachrome staining of renal subcapsular grafts.
Figure 7: In vitro colony-formation assays with p-mSSCs.

Similar content being viewed by others

References

  1. Reya, T., Morrison, S., Clarke, M. & Weissman, I. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Fuchs, E. & Segre, J. Stem cells: a new lease on life. Cell 100, 143–155 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Zhu, H. et al. A protocol for isolation and culture of mesenchymal stem cells from mouse compact bone. Nat. Protoc. 5, 550–560 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Soleimani, M. & Nadri, S. A protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. Nat. Protoc. 4, 102–106 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Caplan, A. Mesenchymal stem cells. J. Orthop. Res. 9, 641–650 (1991).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Zhou, B., Yue, R., Murphy, M., Peyer, J. & Morrison, S. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell 15, 154–168 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Méndez-Ferrer, S. et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466, 829–834 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Pinho, S. et al. PDGFRα and CD51 mark human Nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J. Exp. Med. 210, 1351–1367 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Park, D. et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell 10, 259–272 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Phinney, D. Functional heterogeneity of mesenchymal stem cells: implications for cell therapy. J. Cell. Biochem. 113, 2806–2812 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Chan, C. et al. Identification and specification of the mouse skeletal stem cell. Cell 160, 285–298 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chan, C. et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature 457, 490–494 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ambrosi, T. et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell 20, 771–784.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Morikawa, S. et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J. Exp. Med. 206, 2483–2496 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Szade, K. et al. Where hematopoietic stem cells live: the bone marrow niche. Antioxid. Redox Signal. http://dx.doi.org/10.1089/ars.2017.7419 (2018).

  20. Reinisch, A. et al. A humanized bone marrow ossicle xenotransplantation model enables improved engraftment of healthy and leukemic human hematopoietic cells. Nat. Med. 22, 812–821 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Reinisch, A., Hernandez, D., Schallmoser, K. & Majeti, R. Generation and use of a humanized bone-marrow-ossicle niche for hematopoietic xenotransplantation into mice. Nat. Protoc. 12, 2169–2188 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Morrison, S. & Scadden, D. The bone marrow niche for haematopoietic stem cells. Nature 505, 327–334 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Calvi, L. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Sugiyama, T., Kohara, H., Noda, M. & Nagasawa, T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 25, 977–988 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Omatsu, Y. et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 33, 387–399 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Shiozawa, Y. et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cogle, C. et al. Bone marrow niche in the myelodysplastic syndromes. Leuk. Res. 39, 1020–1027 (2015).

    Article  PubMed  Google Scholar 

  29. Schepers, K., Campbell, T. & Passegué, E. Normal and leukemic stem cell niches: insights and therapeutic opportunities. Cell Stem Cell 16, 254–267 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Raaijmakers, M. et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 464, 852–857 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, N. et al. Endocrine regulation of energy metabolism by the skeleton. Cell 130, 456–469 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  35. Lee, R., Hannig, J., Matthews, K., Myerov, A. & Chen, C. Pharmaceutical therapies for sealing of permeabilized cell membranes in electrical injuries. Ann. N.Y. Acad. Sci. 888, 266–273 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Garvey, W., Fathi, A., Bigelow, F., Carpenter, B. & Jimenez, C. Improved Movat pentachrome stain. Stain Technol. 61, 60–62 (1986).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. McCarty and C. Wang for mouse-colony management; P. Pereira, T. Storm, E. Seo, and T. Naik for laboratory management; and P. Lovelace, J. Ho, and S. Weber for FACS management. This study was supported by the National Institutes of Health (NIH; grants R56 DE025597, R01 DE021683, R21 DE024230, R01 DE019434, RC2 DE020771, U01 HL099776, and R21 DE019274 to M.T.L.; grants U01HL099999, 5 R01 CA86065, and 5 R01 L058770 to I.L.W.); a Siebel Fellowship from the Thomas and Stacey Siebel Foundation, a Prostate Cancer Foundation Young Investigator Award, and a National Institute on Aging Research Career Development Award (grant 1K99AG049958-01A1) to C.K.F.C.; the California Institute for Regenerative Medicine (CIRM; grant TR1-01249), the Oak Foundation, the Hagey Laboratory for Pediatric Regenerative Medicine, and the Gunn/Olivier Research Fund to M.T.L.; a Howard Hughes Medical Institute Medical Student Research Fellowship to G.S.G.; and The Plastic Surgery Research Foundation National Endowment for Plastic Surgery to M.P.M.

Author information

Authors and Affiliations

Authors

Contributions

C.K.F.C., G.S.G., M.T.L., and I.L.W. conceived the isolation strategy and functional assays. M.T.L. and I.L.W. supervised the project. G.S.G., M.P.M., O.M., and M.L. developed the protocol, performed the experiments, and analyzed the data. G.S.G. wrote the manuscript. R.E.B., L.S.K., R.C.R., A.S., and A.M. assisted with flow cytometry, in vitro assays, and manuscript preparation.

Corresponding author

Correspondence to Charles K F Chan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Effect of successive chemical digests on total cell yield.

Total cell yield from crushed bones increases with each successive digest but saturates after three digests [Steps 11-15]. Briefly, crushed 8-week mouse skeletal tissue was separated from whole bone marrow and digested in digest buffer containing 3000 U ml−1 type II collagenase. Total cell number before and after each successive digest was counted by hemocytometer and mean and standard deviation for n = 4 was calculated. All animal experiments in this figure are in accordance with the Stanford's Administrative Panel on Laboratory Animal Care (APLAC) and received approval from the Institutional Review Board (IRB).

Supplementary information

Supplementary Text and Figures

Supplementary Figure 1. (PDF 199 kb)

Supplementary Data

Estimated total number of cells obtained from individual 8-week-old C57BL/6 mice. The data in this file were used to obtain the averages ± standard deviation stated in Table 1. (XLSX 9 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gulati, G., Murphy, M., Marecic, O. et al. Isolation and functional assessment of mouse skeletal stem cell lineage. Nat Protoc 13, 1294–1309 (2018). https://doi.org/10.1038/nprot.2018.041

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2018.041

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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