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:

miR-34 miRNAs provide a barrier for somatic cell reprogramming

Abstract

Somatic reprogramming induced by defined transcription factors is a low-efficiency process that is enhanced by p53 deficiency1,2,3,4,5. So far, p21 is the only p53 target shown to contribute to p53 repression of iPSC (induced pluripotent stem cell) generation1,3, indicating that additional p53 targets may regulate this process. Here, we demonstrate that miR-34 microRNAs (miRNAs), particularly miR-34a, exhibit p53-dependent induction during reprogramming. Mir34a deficiency in mice significantly increased reprogramming efficiency and kinetics, with miR-34a and p21 cooperatively regulating somatic reprogramming downstream of p53. Unlike p53 deficiency, which enhances reprogramming at the expense of iPSC pluripotency, genetic ablation of Mir34a promoted iPSC generation without compromising self-renewal or differentiation. Suppression of reprogramming by miR-34a was due, at least in part, to repression of pluripotency genes, including Nanog, Sox2 and Mycn (also known as N-Myc). This post-transcriptional gene repression by miR-34a also regulated iPSC differentiation kinetics. miR-34b and c similarly repressed reprogramming; and all three miR-34 miRNAs acted cooperatively in this process. Taken together, our findings identified miR-34 miRNAs as p53 targets that play an essential role in restraining somatic reprogramming.

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: Generation of Mir34a and Mir34b/c knockout MEFs.
Figure 2: Deficiency of miR-34 miRNAs increases reprogramming efficiency.
Figure 3: miR-34a and p21 cooperate to repress iPSC generation.
Figure 4: M i r 34 a−/− iPSCs functionally resemble wild-type iPSCs.
Figure 5: Mir34a represses Nanog, Sox2 and N-Myc expression post-transcriptionally.

Similar content being viewed by others

References

  1. 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 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  6. Yamanaka, S. & Takahashi, K. Induction of pluripotent stem cells from mouse fibroblast cultures. Tanpakushitsu Kakusan Koso 51, 2346–2351 (2006).

    CAS  PubMed  Google Scholar 

  7. 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 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 24, 2239–2263 (2010).

    Article  CAS  Google Scholar 

  11. Krizhanovsky, V. & Lowe, S. W. Stem cells: the promises and perils of p53. Nature 460, 1085–1086 (2009).

    Article  CAS  Google Scholar 

  12. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    Article  CAS  Google Scholar 

  13. 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 

  14. Riley, T., Sontag, E., Chen, P. & Levine, A. Transcriptional control of human p53-regulated genes. Nat. Rev. Mol. Cell Biol. 9, 402–412 (2008).

    Article  CAS  Google Scholar 

  15. Banito, A. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 23, 2134–2139 (2009).

    Article  CAS  Google Scholar 

  16. He, L. et al. A microRNA component of the p53 tumour suppressor network. Nature 447, 1130–1134 (2007).

    Article  CAS  Google Scholar 

  17. Raver-Shapira, N. et al. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol. Cell 26, 731–743 (2007).

    Article  CAS  Google Scholar 

  18. Chang, T. C. et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell 26, 745–752 (2007).

    Article  CAS  Google Scholar 

  19. Ambros, V. The functions of animal microRNAs. Nature 431, 350–355 (2004).

    Article  CAS  Google Scholar 

  20. Bartel, D. P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297 (2004).

    Article  CAS  Google Scholar 

  21. Hermeking, H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 17, 193–199 (2009).

    Article  Google Scholar 

  22. Lengner, C. J. et al. Oct4 expression is not required for mouse somatic stem cell self-renewal. Cell Stem Cell 1, 403–415 (2007).

    Article  CAS  Google Scholar 

  23. Miranda, K. C. et al. A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell 126, 1203–1217 (2006).

    Article  CAS  Google Scholar 

  24. Yamanaka, S. & Blau, H. M. Nuclear reprogramming to a pluripotent state by three approaches. Nature 465, 704–712 (2010).

    Article  CAS  Google Scholar 

  25. Nakagawa, M., Takizawa, N., Narita, M., Ichisaka, T. & Yamanaka, S. Promotion of direct reprogramming by transformation-deficient Myc. Proc. Natl Acad. Sci. USA 107, 14152–14157 (2010).

    Article  CAS  Google Scholar 

  26. Wei, J. S. et al. The MYCN oncogene is a direct target of miR-34a. Oncogene 27, 5204–5213 (2008).

    Article  CAS  Google Scholar 

  27. Cole, K. A. et al. A functional screen identifies miR-34a as a candidate neuroblastoma tumor suppressor gene. Mol. Cancer Res. 6, 735–742 (2008).

    Article  CAS  Google Scholar 

  28. Kuijk, E. W. et al. PTEN and TRP53 independently suppress Nanog expression in spermatogonial stem cells. Stem Cells Dev. 19, 979–988 (2009).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Tarantino, C. et al. miRNA 34a, 100, and 137 modulate differentiation of mouse embryonic stem cells. FASEB J. 24, 3255–3263 (2010).

    Article  CAS  Google Scholar 

  31. Filipowicz, W., Bhattacharyya, S. N. & Sonenberg, N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat. Rev. Genet. 9, 102–114 (2008).

    Article  CAS  Google Scholar 

  32. Melton, C., Judson, R. L. & Blelloch, R. Opposing microRNA families regulate self-renewal in mouse embryonic stem cells. Nature 463, 621–626 (2010).

    Article  CAS  Google Scholar 

  33. Wang, Y. et al. Embryonic stem cell-specific microRNAs regulate the G1-S transition and promote rapid proliferation. Nat. Genet. 40, 1478–1483 (2008).

    Article  CAS  Google Scholar 

  34. Wang, Y., Medvid, R., Melton, C., Jaenisch, R. & Blelloch, R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat. Genet. 39, 380–385 (2007).

    Article  CAS  Google Scholar 

  35. Heo, I. et al. Lin28 mediates the terminal uridylation of let-7 precursor MicroRNA. Mol. Cell 32, 276–284 (2008).

    Article  CAS  Google Scholar 

  36. Newman, M. A., Thomson, J. M. & Hammond, S. M. Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. RNA 14, 1539–1549 (2008).

    Article  CAS  Google Scholar 

  37. Viswanathan, S. R., Daley, G. Q. & Gregory, R. I. Selective blockade of microRNA processing by Lin28. Science 320, 97–100 (2008).

    Article  CAS  Google Scholar 

  38. Anokye-Danso, F. et al. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8, 376–388 (2011).

    Article  CAS  Google Scholar 

  39. Miyoshi, N. et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 8, 633–638 (2011).

    Article  CAS  Google Scholar 

  40. Samavarchi-Tehrani, P. et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell 7, 64–77 (2010).

    Article  CAS  Google Scholar 

  41. Schaniel, C. et al. Smarcc1/Baf155 couples self-renewal gene repression with changes in chromatin structure in mouse embryonic stem cells. Stem Cells 27, 2979–2991 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Cummins, J. M. et al. The colorectal microRNAome. Proc. Natl Acad. Sci. USA 103, 3687–3692 (2006).

    Article  CAS  Google Scholar 

  43. 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  Google Scholar 

  44. Judson, R. L., Babiarz, J. E., Venere, M. & Blelloch, R. Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat. Biotechnol. 27, 459–461 (2009).

    Article  CAS  Google Scholar 

  45. Brambrink, T. et al. Sequential expression of pluripotency markers during direct reprogramming of mouse somatic cells. Cell Stem Cell 2, 151–159 (2008).

    Article  CAS  Google Scholar 

  46. Debacq-Chainiaux, F., Erusalimsky, J. D., Campisi, J. & Toussaint, O. Protocols to detect senescence-associated β-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 4, 1798–1806 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Zaghi, A. Basila, A. Perez, G. Lai, M. Foth, A. Kemp, A. Fresnoza, A. Valeros, A. Fritz, D. Schichnes and H. Noller for technical assistance and A. Economides for discussions and input. We also thank J. M. Halbleib, D. Schaffer and R. M. Harland for reading the manuscripts, and I. Lemischka, Q. Li, M. Hemann and B. Vogelstein for sharing reagents. In particular, we are grateful for the pathological expertise R. Van Andel offered us. L.H. is a Searle Scholar, and is supported by a new faculty award by California Institute for Regenerative Medicine (RN2-00923-1) and an R01 (R01 CA139067) and an R00 grant (R00 CA126186) from the National Cancer Institute (NCI). G.J.H. is a Howard Hughes Medical Institute investigator, and is supported by a program project grant from the NCI. C-P.L. is supported by a Siebel postdoctoral fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Y.J.C., C-P.L. and L.H. designed all experiments and carried out the majority of the experiments shown in all figures and supplementary figures. J.J.H. and Y.Z. carried out immunofluorescence analyses and teratoma analyses to characterize the pluripotency of the iPSCs. L.H., X.H., P.B. and G.J.H. generated knockout constructs for Mir34a and Mir34b/c, and identified the correctly targeted ESC clones for Mir34a. N.O. and P.B. identified the correctly targeted ESC clone for Mir34b/c, validated Mir34a and Mir34b/c targeting in mice by Southern blotting and generated M i r 34 a−/−, M i r 34 b/c−/− and Mir34 triple knockout MEFs. S.Y.K. and G.J.H. carried out the blastocyst injection for M i r 34 a+/− and M i r 34 b/c+/− ESC clones. A.O., S.Y.K. and G.G.H. characterized the pluripotency of iPSCs using chimaera assays. M.J.B. and C.C. contributed to the identification of miR-34a targets.

Corresponding author

Correspondence to Lin He.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1265 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Choi, Y., Lin, CP., Ho, J. et al. miR-34 miRNAs provide a barrier for somatic cell reprogramming. Nat Cell Biol 13, 1353–1360 (2011). https://doi.org/10.1038/ncb2366

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2366

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