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:

Notch inhibition allows oncogene-independent generation of iPS cells

An Erratum to this article was published on 13 November 2014

An Erratum to this article was published on 13 November 2014

This article has been updated

Abstract

The reprogramming of somatic cells to pluripotency using defined transcription factors holds great promise for biomedicine. However, human reprogramming remains inefficient and relies either on the use of the potentially dangerous oncogenes KLF4 and CMYC or the genetic inhibition of the tumor suppressor gene p53. We hypothesized that inhibition of signal transduction pathways that promote differentiation of the target somatic cells during development might relieve the requirement for non-core pluripotency factors during induced pluripotent stem cell (iPSC) reprogramming. Here, we show that inhibition of Notch greatly improves the efficiency of iPSC generation from mouse and human keratinocytes by suppressing p21 in a p53-independent manner and thereby enriching for undifferentiated cells capable of long-term self-renewal. Pharmacological inhibition of Notch enabled routine production of human iPSCs without KLF4 and CMYC while leaving p53 activity intact. Thus, restricting the development of somatic cells by altering intercellular communication enables the production of safer human iPSCs.

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: DAPT treatment promotes mouse and human keratinocyte reprogramming.
Figure 2: Îł-secretase inhibition promotes reprogramming by blocking Notch signaling.
Figure 3: Notch inhibition promotes keratinocyte reprogramming by suppressing p21.
Figure 4: Highly efficient reprogramming with NOTCH and DOT1L inhibition.
Figure 5: NOTCH inhibition suppresses p21 without reducing p53 activity.
Figure 6: Model of iPSC generation from human keratinocytes.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 29 July 2014

    In the version of this article initially published, Julia TCW's name was misspelled as Julia T C W. In addition, her initials in the author contribution statement should have read J.T. instead of J.T.C.W. The error has been corrected in the HTML and PDF versions of the article.

  • 14 August 2014

    In the version of this article initially published, a black bar was erroneously placed in the scrambled shRNA column in Figure 3g. The error has been corrected for the PDF and HTML versions of the article.

References

  1. Aoi, T. et al. Generation of pluripotent stem cells from adult mouse liver and stomach cells. Science 321, 699–702 (2008).

    Article  CAS  Google Scholar 

  2. Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 26, 101–106 (2008).

    Article  CAS  Google Scholar 

  3. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144 (2009).

    Article  CAS  Google Scholar 

  4. Okita, K. et al. A more efficient method to generate integration-free human iPS cells. Nat. Methods 8, 409–412 (2011).

    Article  CAS  Google Scholar 

  5. Son, M.J. et al. Nicotinamide overcomes pluripotency deficits and reprogramming barriers. Stem Cells 31, 1121–1135 (2013).

    Article  CAS  Google Scholar 

  6. Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature 460, 1132–1135 (2009).

    Article  CAS  Google Scholar 

  7. Utikal, J. et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460, 1145–1148 (2009).

    Article  CAS  Google Scholar 

  8. Marión, R.M. et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 460, 1149–1153 (2009).

    Article  Google Scholar 

  9. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    Article  CAS  Google Scholar 

  10. Li, Y. et al. The p53-PUMA axis suppresses iPSC generation. Nat Commun. 4, 2174 (2013).

    Article  Google Scholar 

  11. Lake, B.B. et al. Context-dependent enhancement of induced pluripotent stem cell reprogramming by silencing Puma. Stem Cells 30, 888–897 (2012).

    Article  CAS  Google Scholar 

  12. Guo, S. et al. Nonstochastic reprogramming from a privileged somatic cell state. Cell 156, 649–662 (2014).

    Article  CAS  Google Scholar 

  13. Lee, Y.L. et al. Sirtuin 1 facilitates generation of induced pluripotent stem cells from mouse embryonic fibroblasts through the miR-34a and p53 pathways. PLoS ONE 7, e45633 (2012).

    Article  CAS  Google Scholar 

  14. Brosh, R. et al. p53 counteracts reprogramming by inhibiting mesenchymal-to-epithelial transition. Cell Death Differ. 20, 312–320 (2013).

    Article  CAS  Google Scholar 

  15. Ye, D. et al. MiR-138 promotes induced pluripotent stem cell generation through the regulation of the p53 signaling. Stem Cells 30, 1645–1654 (2012).

    Article  CAS  Google Scholar 

  16. Wang, J. et al. p53-facilitated miR-199a-3p regulates somatic cell reprogramming. Stem Cells 30, 1405–1413 (2012).

    Article  CAS  Google Scholar 

  17. Zhu, S. et al. Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell 7, 651–655 (2010).

    Article  CAS  Google Scholar 

  18. Silva, J. et al. Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS Biol. 6, e253 (2008).

    Article  Google Scholar 

  19. Ichida, J.K. et al. A small-molecule inhibitor of Tgf-β signaling replaces Sox2 in reprogramming by inducing Nanog. Cell Stem Cell 5, 491–503 (2009).

    Article  CAS  Google Scholar 

  20. Hou, P. et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341, 651–654 (2013).

    Article  CAS  Google Scholar 

  21. Huangfu, D. et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795–797 (2008).

    Article  CAS  Google Scholar 

  22. Federation, A.J., Bradner, J.E. & Meissner, A. The use of small molecules in somatic-cell reprogramming. Trends Cell Biol. 24, 179–187 (2014).

    Article  CAS  Google Scholar 

  23. Amabile, G. & Meissner, A. Induced pluripotent stem cells: current progress and potential for regenerative medicine. Trends Mol. Med. 15, 59–68 (2009).

    Article  CAS  Google Scholar 

  24. Eminli, S. et al. Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells. Nat. Genet. 41, 968–976 (2009).

    Article  CAS  Google Scholar 

  25. Gurdon, J.B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 10, 622–640 (1962).

    CAS  Google Scholar 

  26. Li, J., Greco, V., Guasch, G., Fuchs, E. & Mombaerts, P. Mice cloned from skin cells. Proc. Natl. Acad. Sci. USA 104, 2738–2743 (2007).

    Article  CAS  Google Scholar 

  27. Artavanis-Tsakonas, S. & Muskavitch, M.A. Notch: the past, the present, and the future. Curr. Top. Dev. Biol. 92, 1–29 (2010).

    Article  CAS  Google Scholar 

  28. Bray, S.J. Notch signalling: a simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 7, 678–689 (2006).

    Article  CAS  Google Scholar 

  29. Topley, G.I., Okuyama, R., Gonzales, J.G., Conti, C. & Dotto, G.P. p21WAF1/Cip1 functions as a suppressor of malignant skin tumor formation and a determinant of keratinocyte stem-cell potential. Proc. Natl. Acad. Sci. USA 96, 9089–9094 (1999).

    Article  CAS  Google Scholar 

  30. Missero, C., Di Cunto, F., Kiyokawa, H., Koff, A. & Dotto, G.P. The absence of p21Cip1/WAF1 alters keratinocyte growth and differentiation and promotes ras-tumor progression. Genes Dev. 10, 3065–3075 (1996).

    Article  CAS  Google Scholar 

  31. Aasen, T. & Belmonte, J.C. Isolation and cultivation of human keratinocytes from skin or plucked hair for the generation of induced pluripotent stem cells. Nat. Protoc. 5, 371–382 (2010).

    Article  CAS  Google Scholar 

  32. Aasen, T. et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat. Biotechnol. 26, 1276–1284 (2008).

    Article  CAS  Google Scholar 

  33. Blanpain, C., Lowry, W.E., Pasolli, H.A. & Fuchs, E. Canonical notch signaling functions as a commitment switch in the epidermal lineage. Genes Dev. 20, 3022–3035 (2006).

    Article  CAS  Google Scholar 

  34. Bock, C. et al. Reference maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439–452 (2011).

    Article  CAS  Google Scholar 

  35. Fuwa, H. et al. Divergent synthesis of multifunctional molecular probes to elucidate the enzyme specificity of dipeptidic γ-secretase inhibitors. ACS Chem. Biol. 2, 408–418 (2007).

    Article  CAS  Google Scholar 

  36. Nam, Y., Sliz, P., Song, L., Aster, J.C. & Blacklow, S.C. Structural basis for cooperativity in recruitment of MAML coactivators to Notch transcription complexes. Cell 124, 973–983 (2006).

    Article  CAS  Google Scholar 

  37. Nam, Y., Weng, A.P., Aster, J.C. & Blacklow, S.C. Structural requirements for assembly of the CSL.Intracellular Notch1.Mastermind-like 1 transcriptional activation complex. J. Biol. Chem. 278, 21232–21239 (2003).

    Article  CAS  Google Scholar 

  38. Lefort, K. & Dotto, G.P. Notch signaling in the integrated control of keratinocyte growth/differentiation and tumor suppression. Semin. Cancer Biol. 14, 374–386 (2004).

    Article  CAS  Google Scholar 

  39. Jones, P.H. & Watt, F.M. Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression. Cell 73, 713–724 (1993).

    Article  CAS  Google Scholar 

  40. Kavian, N. et al. Targeting ADAM-17/notch signaling abrogates the development of systemic sclerosis in a murine model. Arthritis Rheum. 62, 3477–3487 (2010).

    Article  CAS  Google Scholar 

  41. Allen, A.S. et al. De novo mutations in epileptic encephalopathies. Nature 501, 217–221 (2013).

    Article  CAS  Google Scholar 

  42. Huangfu, D. et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 26, 1269–1275 (2008).

    Article  CAS  Google Scholar 

  43. Mikkelsen, T.S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008).

    Article  CAS  Google Scholar 

  44. Onder, T.T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598–602 (2012).

    Article  CAS  Google Scholar 

  45. Gore, A. et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 471, 63–67 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank E. Son for assistance with microarray data analysis, S. Sato for assistance with chimera experiments, E. Kiskinis for assistance with nanostring analysis and K. Koszka and M. Yamaki for assistance with teratoma experiments. The authors are grateful for the financial support that made this work possible. K.E. was supported by US National Institutes of Health (NIH) R01 grant 5R01GM096067, NIH P01 grant 5P01GM099117 and the Howard Hughes Medical Institute. A.M. was supported by NIH P01 grant 5P01GM099117. H.A. was supported by grants from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan, Grant-in-aid for Scientific Research (21390456) and Grant-in-Aid for challenging Exploratory Research (22659304) and a grant from JST-CREST. J.K.I. was supported by a Stan and Fiona Druckenmiller–New York Stem Cell Foundation postdoctoral fellowship, NIH K99 grant 1K99NS077435-01A1, NIH R00 grant 4R00NS077435-03 and the Novartis Institutes for BioMedical Research. C.B. was supported by a Feodor Lynen Fellowship from the Alexander von Humboldt Foundation.

Author information

Authors and Affiliations

Authors

Contributions

A.M. and J.E.B. hypothesized that Notch inhibition might aid reprogramming. J.K.I., J.T., A.M., A.U., L.L.R. and K.E. designed reprogramming and mechanistic experiments to test the hypothesis. J.K.I., J.T., A.C.C., L.A.W., Y.S., M.T.M., S.S., G.A. and H.A. performed reprogramming experiments and characterization of the iPSCs. C.B. and M.Z. performed bioinformatic analysis of transcriptional data characterizing the iPSCs. J.K.I., J.T., A.C.C. and Y.S. performed experiments to determine the mechanism of action of DAPT and Notch inhibition in reprogramming. K.E., J.K.I. and J.T. discovered and confirmed the mechanism of action of DAPT. K.E. and J.K.I. wrote the paper. All authors helped in paper revision.

Corresponding authors

Correspondence to Hidenori Akutsu, Alexander Meissner or Kevin Eggan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results and Supplementary Figures 1–7. (PDF 1148 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ichida, J., TCW, J., Williams, L. et al. Notch inhibition allows oncogene-independent generation of iPS cells. Nat Chem Biol 10, 632–639 (2014). https://doi.org/10.1038/nchembio.1552

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1552

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