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.

  • Letter
  • Published:

Linking the p53 tumour suppressor pathway to somatic cell reprogramming

Abstract

Reprogramming somatic cells to induced pluripotent stem (iPS) cells has been accomplished by expressing pluripotency factors and oncogenes1,2,3,4,5,6,7,8, but the low frequency and tendency to induce malignant transformation9 compromise the clinical utility of this powerful approach. We address both issues by investigating the mechanisms limiting reprogramming efficiency in somatic cells. Here we show that reprogramming factors can activate the p53 (also known as Trp53 in mice, TP53 in humans) pathway. Reducing signalling to p53 by expressing a mutated version of one of its negative regulators, by deleting or knocking down p53 or its target gene, p21 (also known as Cdkn1a), or by antagonizing reprogramming-induced apoptosis in mouse fibroblasts increases reprogramming efficiency. Notably, decreasing p53 protein levels enabled fibroblasts to give rise to iPS cells capable of generating germline-transmitting chimaeric mice using only Oct4 (also known as Pou5f1) and Sox2. Furthermore, silencing of p53 significantly increased the reprogramming efficiency of human somatic cells. These results provide insights into reprogramming mechanisms and suggest new routes to more efficient reprogramming while minimizing the use of oncogenes.

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: Increased generation of iPS cells by blocking p53 and p21.
Figure 2: Modulation of p53 activity alters reprogramming efficiency.
Figure 3: Generation and characterization of 2F-p53KD-iPS cells by p53 downregulation.
Figure 4: Downregulation of p53 activity increases reprogramming efficiency of human somatic cells.

Similar content being viewed by others

References

  1. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Park, I. H. et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141–146 (2008)

    Article  ADS  CAS  PubMed  Google Scholar 

  5. Lowry, W. E. et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc. Natl Acad. Sci. USA 105, 2883–2888 (2008)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. Wernig, M., Meissner, A., Cassady, J. P. & Jaenisch, R. c-Myc is dispensable for direct reprogramming of mouse fibroblasts. Cell Stem Cell 2, 10–12 (2008)

    Article  CAS  PubMed  Google Scholar 

  9. Okita, K., Ichisaka, T. & Yamanaka, S. Generation of germline-competent induced pluripotent stem cells. Nature 448, 313–317 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Rowland, B. D., Bernards, R. & Peeper, D. S. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nature Cell Biol. 7, 1074–1082 (2005)

    Article  CAS  PubMed  Google Scholar 

  11. Kanatsu-Shinohara, M. et al. Generation of pluripotent stem cells from neonatal mouse testis. Cell 119, 1001–1012 (2004)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Cleveland, J. L. & Sherr, C. J. Antagonism of Myc functions by Arf. Cancer Cell 6, 309–311 (2004)

    Article  CAS  PubMed  Google Scholar 

  14. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004)

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Miyashita, T. & Reed, J. C. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80, 293–299 (1995)

    Article  CAS  PubMed  Google Scholar 

  16. Kamijo, T. et al. Functional and physical interactions of the ARF tumor suppressor with p53 and Mdm2. Proc. Natl Acad. Sci. USA 95, 8292–8297 (1998)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Pomerantz, J. et al. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2’s inhibition of p53. Cell 92, 713–723 (1998)

    Article  CAS  PubMed  Google Scholar 

  18. Zhang, Y., Xiong, Y. & Yarbrough, W. G. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell 92, 725–734 (1998)

    Article  CAS  PubMed  Google Scholar 

  19. Knudsen, E. S. & Knudsen, K. E. Tailoring to RB: tumour suppressor status and therapeutic response. Nature Rev. Cancer 8, 714–724 (2008)

    Article  CAS  Google Scholar 

  20. Marine, J. C., Dyer, M. A. & Jochemsen, A. G. MDMX: from bench to bedside. J. Cell Sci. 120, 371–378 (2007)

    Article  CAS  PubMed  Google Scholar 

  21. Wang, Y. V. et al. Increased radioresistance and accelerated B cell lymphomas in mice with Mdmx mutations that prevent modifications by DNA-damage-activated kinases. Cancer Cell 16, 33–43 (2009)

    Article  PubMed  PubMed Central  Google Scholar 

  22. Foster, K. W. et al. Oncogene expression cloning by retroviral transduction of adenovirus E1A-immortalized rat kidney RK3E cells: transformation of a host with epithelial features by c-MYC and the zinc finger protein GKLF. Cell Growth Differ. 10, 423–434 (1999)

    CAS  PubMed  Google Scholar 

  23. Shaulian, E., Zauberman, A., Ginsberg, D. & Oren, M. Identification of a minimal transforming domain of p53: negative dominance through abrogation of sequence-specific DNA binding. Mol. Cell. Biol. 12, 5581–5592 (1992)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin, T. et al. p53 induces differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nature Cell Biol. 7, 165–171 (2005)

    Article  CAS  PubMed  Google Scholar 

  25. Jiang, J. et al. A core Klf circuitry regulates self-renewal of embryonic stem cells. Nature Cell Biol. 10, 353–360 (2008)

    Article  PubMed  Google Scholar 

  26. Komarov, P. G. et al. A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science 285, 1733–1737 (1999)

    Article  CAS  PubMed  Google Scholar 

  27. Shi, Y. et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell 3, 568–574 (2008)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Kim, D. et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 4, 472–476 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gonzalez, F. et al. Generation of mouse-induced pluripotent stem cells by transient expression of a single nonviral polycistronic vector. Proc. Natl Acad. Sci. USA 106, 8918–8922 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kitamura, T. et al. Retrovirus-mediated gene transfer and expression cloning: powerful tools in functional genomics. Exp. Hematol. 31, 1007–1014 (2003)

    Article  CAS  PubMed  Google Scholar 

  32. Miyoshi, H., Blömer, U., Takahashi, M., Gage, F. H. & Verma, I. M. Development of a self-inactivating lentivirus vector. J. Virol. 72, 8150–8157 (1998)

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Sherley, J. L. Guanine nucleotide biosynthesis is regulated by the cellular p53 concentration. J. Biol. Chem. 266, 24815–24828 (1991)

    CAS  PubMed  Google Scholar 

  34. Huppi, K. et al. Molecular cloning, sequencing, chromosomal localization and expression of mouse p21 (Waf1). Oncogene 9, 3017–3020 (1994)

    CAS  PubMed  Google Scholar 

  35. Takahashi, K., Okita, K., Nakagawa, M. & Yamanaka, S. Induction of pluripotent stem cells from fibroblast cultures. Nature Protocols 2, 3081–3089 (2007)

    Article  CAS  PubMed  Google Scholar 

  36. Wiznerowicz, M. & Trono, D. Conditional suppression of cellular genes: lentivirus vector-mediated drug-inducible RNA interference. J. Virol. 77, 8957–8961 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Blelloch, R., Venere, M., Yen, J. & Ramalho-Santos, M. Generation of induced pluripotent stem cells in the absence of drug selection. Cell Stem Cell 1, 245–247 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Blelloch, R. et al. Reprogramming efficiency following somatic cell nuclear transfer is influenced by the differentiation and methylation state of the donor nucleus. Stem Cells 24, 2007–2013 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to the CMRB Histology & Bioimaging and Cell culture Platforms for assistance, S. Boue for microarray analysis, S. Kim for help with maintenance of mouse colonies, Y. Richaud for technical assistance, M. Nagao for preparation of mouse neural stem cells, K. Brennand and F. Gage for preparation of human neural stem cells, I. Verma and A. Consiglio for advice and help with lentiviral transduction, Y. Dayn for chimaeric mouse production, and all members of the Gene Expression Laboratory and CMRB for discussions and M. Serrano for sharing unpublished results. J.S. was partially supported by Astellas Pharma Inc. T.K. was partially supported by Japan Society for the Promotion of Science. Work in the laboratory of G.M.W. was supported by NIH grants (5 R01 CA061449 and CA100845). Work in the laboratory of J.C.I.B. was supported by grants from the NIH, Tercel, Marato, G. Harold and Leila Y. Mathers Charitable Foundation and Fundacion Cellex.

Author Contributions T.K. and J.S. contributed to the experimental work, project planning, data analysis and writing the manuscript and contributed equally to this work. Y.V.W. contributed to the experimental work, data analysis, writing the manuscript and established Mdmx mutant mice. S.M., L.B.M. and A.R. contributed to the experimental work, data analysis and writing the manuscript. G.M.W. and J.C.I.B. contributed to project planning and writing the manuscript, and supervised all the work. G.M.W. and J.C.I.B. are co-contributing corresponding authors.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Geoffrey M. Wahl or Juan Carlos Izpisúa Belmonte.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-3, Supplementary References and Supplementary Figures 1-27 with Legends. (PDF 12910 kb)

Supplementary Data

This file contains microarray analysis on MEF, 2F-p53KD-iPS cell clones (#1 and #6) and mouse ES. (XLS 8680 kb)

Supplementary Movie 1

In this movie file beating cells were observed in embryoid bodies (d10) derived from 2F-iPS clone #1. (MOV 566 kb)

Supplementary Movie 2

In this movie file beating cells were observed in embryoid bodies (d14) derived from 2F-iPS clone #3. (MOV 567 kb)

Supplementary Movie 3

In this movie file beating cells were observed in embryoid bodies (d15) derived from 2F-iPS clone #6. (MOV 593 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kawamura, T., Suzuki, J., Wang, Y. et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144 (2009). https://doi.org/10.1038/nature08311

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature08311

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