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:

Adhesion strength–based, label-free isolation of human pluripotent stem cells

Abstract

We demonstrate substantial differences in 'adhesive signature' between human pluripotent stem cells (hPSCs), partially reprogrammed cells, somatic cells and hPSC-derived differentiated progeny. We exploited these differential adhesion strengths to rapidly (over 10 min) and efficiently isolate fully reprogrammed induced hPSCs (hiPSCs) as intact colonies from heterogeneous reprogramming cultures and from differentiated progeny using microfluidics. hiPSCs were isolated label free, enriched to 95%–99% purity with >80% survival, and had normal transcriptional profiles, differentiation potential and karyotypes. We also applied this strategy to isolate hPSCs (hiPSCs and human embryonic stem cells) during routine culture and show that it may be extended to isolate hPSC-derived lineage-specific stem cells or differentiated cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Adhesion of hiPSCs undergoing reprogramming and differentiation.
Figure 2: Adhesion strength–based isolation of pluripotent stem cells in microfluidic devices.
Figure 3: Adhesion strength–based isolation of hiPSCs from a heterogeneous reprogramming culture.
Figure 4: Adhesion strength–based enrichment of hiPSCs from differentiating cultures.

Similar content being viewed by others

References

  1. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  Google Scholar 

  2. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    Article  CAS  Google Scholar 

  3. Robinton, D.A. & Daley, G.Q. The promise of induced pluripotent stem cells in research and therapy. Nature 481, 295–305 (2012).

    Article  CAS  Google Scholar 

  4. Zhao, Y. et al. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell 3, 475–479 (2008).

    Article  CAS  Google Scholar 

  5. Enver, T. et al. Cellular differentiation hierarchies in normal and culture-adapted human embryonic stem cells. Hum. Mol. Genet. 14, 3129–3140 (2005).

    Article  CAS  Google Scholar 

  6. Rodin, S. et al. Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nat. Biotechnol. 28, 611–615 (2010).

    Article  CAS  Google Scholar 

  7. Boheler, K.R. et al. Differentiation of pluripotent embryonic stem cells into cardiomyocytes. Circ. Res. 91, 189–201 (2002).

    Article  CAS  Google Scholar 

  8. Burridge, P.W. et al. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS ONE 6, e18293 (2011).

    Article  CAS  Google Scholar 

  9. Cho, M.S. et al. Highly efficient and large-scale generation of functional dopamine neurons from human embryonic stem cells. Proc. Natl. Acad. Sci. USA 105, 3392–3397 (2008).

    Article  CAS  Google Scholar 

  10. Lee, G. et al. Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells. Nat. Biotechnol. 25, 1468–1475 (2007).

    Article  CAS  Google Scholar 

  11. Tang, C. et al. An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells. Nat. Biotechnol. 29, 829–834 (2011).

    Article  CAS  Google Scholar 

  12. Ohgushi, M. et al. Molecular pathway and cell state responsible for dissociation-induced apoptosis in human pluripotent stem cells. Cell Stem Cell 7, 225–239 (2010).

    Article  CAS  Google Scholar 

  13. Nandivada, H. et al. Fabrication of synthetic polymer coatings and their use in feeder-free culture of human embryonic stem cells. Nat. Protoc. 6, 1037–1043 (2011).

    Article  CAS  Google Scholar 

  14. Baker, D.E. et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat. Biotechnol. 25, 207–215 (2007).

    Article  CAS  Google Scholar 

  15. Mitalipova, M.M. et al. Preserving the genetic integrity of human embryonic stem cells. Nat. Biotechnol. 23, 19–20 (2005).

    Article  CAS  Google Scholar 

  16. Hartung, O., Huo, H., Daley, G.Q. & Schlaeger, T.M. Clump passaging and expansion of human embryonic and induced pluripotent stem cells on mouse embryonic fibroblast feeder cells. Curr. Protoc. Stem Cell Biol. 14, 1C.10 (2010).

    Google Scholar 

  17. McCracken, K.W., Howell, J.C., Wells, J.M. & Spence, J.R. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat. Protoc. 6, 1920–1928 (2011).

    Article  CAS  Google Scholar 

  18. Bajpai, R., Lesperance, J., Kim, M. & Terskikh, A.V. Efficient propagation of single cells Accutase-dissociated human embryonic stem cells. Mol. Reprod. Dev. 75, 818–827 (2008).

    Article  CAS  Google Scholar 

  19. Maitra, A. et al. Genomic alterations in cultured human embryonic stem cells. Nat. Genet. 37, 1099–1103 (2005).

    Article  CAS  Google Scholar 

  20. Ellerström, C., Strehl, R., Noaksson, K., Hyllner, J. & Semb, H. Facilitated expansion of human embryonic stem cells by single-cell enzymatic dissociation. Stem Cells 25, 1690–1696 (2007).

    Article  Google Scholar 

  21. Meng, G., Liu, S. & Rancourt, D.E. Rapid isolation of undifferentiated human pluripotent stem cells from extremely differentiated colonies. Stem Cells Dev. 20, 583–591 (2011).

    Article  Google Scholar 

  22. Chan, E.M. et al. Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells. Nat. Biotechnol. 27, 1033–1037 (2009).

    Article  CAS  Google Scholar 

  23. Gallant, N.D., Michael, K.E. & García, A.J. Cell adhesion strengthening: contributions of adhesive area, integrin binding, and focal adhesion assembly. Mol. Biol. Cell 16, 4329–4340 (2005).

    Article  CAS  Google Scholar 

  24. Meng, Y. et al. Characterization of integrin engagement during defined human embryonic stem cell culture. FASEB J. 24, 1056–1065 (2010).

    Article  CAS  Google Scholar 

  25. Staerk, J. et al. Reprogramming of human peripheral blood cells to induced pluripotent stem cells. Cell Stem Cell 7, 20–24 (2010).

    Article  CAS  Google Scholar 

  26. Wojdacz, T.K., Dobrovic, A. & Algar, E.M. Rapid detection of methylation change at H19 in human imprinting disorders using methylation-sensitive high-resolution melting. Hum. Mutat. 29, 1255–1260 (2008).

    Article  CAS  Google Scholar 

  27. Wojdacz, T.K., Dobrovic, A. & Hansen, L.L. Methylation-sensitive high-resolution melting. Nat. Protoc. 3, 1903–1908 (2008).

    Article  CAS  Google Scholar 

  28. Nagaoka, M., Si-Tayeb, K., Akaike, T. & Duncan, S.A. Culture of human pluripotent stem cells using completely defined conditions on a recombinant E-cadherin substratum. BMC Dev. Biol. 10, 60 (2010).

    Article  Google Scholar 

  29. Blagovic, K., Kim, L.Y. & Voldman, J. Microfluidic perfusion for regulating diffusible signaling in stem cells. PLoS ONE 6, e22892 (2011).

    Article  CAS  Google Scholar 

  30. Gómez-Sjöberg, R., Leyrat, A.A., Pirone, D.M., Chen, C.S. & Quake, S.R. Versatile, fully automated, microfluidic cell culture system. Anal. Chem. 79, 8557–8563 (2007).

    Article  Google Scholar 

  31. Kim, J.Y. et al. A cell culturing system that integrates the cell loading function on a single platform and evaluation of the pulsatile pumping effect on cells. Biomed. Microdevices 10, 11–20 (2008).

    Article  CAS  Google Scholar 

  32. Lu, H. et al. Microfluidic shear devices for quantitative analysis of cell adhesion. Anal. Chem. 76, 5257–5264 (2004).

    Article  CAS  Google Scholar 

  33. Dhara, S.K. et al. Human neural progenitor cells derived from embryonic stem cells in feeder-free cultures. Differentiation 76, 454–464 (2008).

    Article  CAS  Google Scholar 

  34. Laflamme, M.A. et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25, 1015–1024 (2007).

    Article  CAS  Google Scholar 

  35. Fu, J. et al. Mechanical regulation of cell function with geometrically modulated elastomeric substrates. Nat. Methods 7, 733–736 (2010).

    Article  CAS  Google Scholar 

  36. McDonald, J.C. et al. Fabrication of microfluidic systems in poly(dimethylsiloxane). Electrophoresis 21, 27–40 (2000).

    Article  CAS  Google Scholar 

  37. Bratt-Leal, A.M., Carpenedo, R.L., Ungrin, M.D., Zandstra, P.W. & McDevitt, T.C. Incorporation of biomaterials in multicellular aggregates modulates pluripotent stem cell differentiation. Biomaterials 32, 48–56 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by an American Recovery and Reinvestment Act (ARRA) supplement to the US National Institutes of Health (NIH) grant R01 GM065918 (A.J.G.), NIH R43 NS080407 (J.M.C. and A.J.G.), the Stem Cell Engineering Center at Georgia Institute of Technology (T.C.M.), a Sloan Foundation Fellowship (H.L.), the National Science Foundation (NSF) DBI-0649833 (H.L.) and an ARRA sub-award under RC1CA144825 (H.L.), NSF CMMI-1129611 (J.F.), the Georgia Tech Emory Center for Regenerative Medicine and the Parker H. Petit Institute for Bioengineering and Bioscience at Georgia Institute of Technology. We thank J. Wu (Stanford Univ.) for providing blood cell–derived hiPSCs, C. Xu (Emory Univ.) for providing hiPSC-derived cardiomyocytes, T. Hookway for cardiomyocyte culture, A. Ortiz for teratoma studies, and A. Cheng for her help with focal adhesion assays. We thank J. Phillips, D. Dumbauld, Y. Wang and A. Bratt-Leal for insightful discussions and suggestions.

Author information

Authors and Affiliations

Authors

Contributions

A.S. and S.S. conducted all adhesion and microfluidic studies, collected data and performed data analysis. S.S., A.S. and H.L. developed microfluidic methods. J.M.C. and S.L.S. established and provided the IMR90-derived hiPSC cells and neural stem cells. T.L., W.C. and J.F. developed and provided micropatterns. M.T.C. and A.S. conducted microarray and epigenetic analysis. A.J.G. and T.C.M. developed the concept, and together with A.S. contributed to the planning and design of the project. A.S., S.S., T.C.M., and A.J.G. wrote the manuscript, and all authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Andrés J García.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–20 and Supplementary Table 1 (PDF 22257 kb)

Real-time detachment of hiPSCs cocultured with IMR90 cells by μSHEAR

A shear stress of 125 dynes cm–2 was applied to achieve selective detachment. White arrows denote contaminating fibroblasts. (MOV 31906 kb)

Real-time detachment of blood-derived hiPSCs cocultured with blood cells by μSHEAR

A shear stress of 10 dynes cm–2 was applied for 1 min to deplete blood cells, and then the shear stress was increased to 100 dynes cm–2 to achieve selective hiPSC detachment. (MOV 1111 kb)

Beating cardiomyocytes pre-μSHEAR

Cultured hiPSC-derived cardiomyocytes demonstrate spontaneous contractile activity. (MOV 7992 kb)

Beating cardiomyocytes post-μSHEAR

After μSHEAR, residual hiPSC-derived cardiomyocytes in the devices that were were trypsinized and cultured demonstrate spontaneous contractile activity. (MOV 11720 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Singh, A., Suri, S., Lee, T. et al. Adhesion strength–based, label-free isolation of human pluripotent stem cells. Nat Methods 10, 438–444 (2013). https://doi.org/10.1038/nmeth.2437

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.2437

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