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:

The oncogene c-Jun impedes somatic cell reprogramming

A Corrigendum to this article was published on 28 August 2015

This article has been updated

Abstract

Oncogenic transcription factors are known to mediate the conversion of somatic cells to tumour or induced pluripotent stem cells (iPSCs). Here we report c-Jun as a barrier for iPSC formation. c-Jun is expressed by and required for the proliferation of mouse embryonic fibroblasts (MEFs), but not mouse embryonic stem cells (mESCs). Consistently, c-Jun is induced during mESC differentiation, drives mESCs towards the endoderm lineage and completely blocks the generation of iPSCs from MEFs. Mechanistically, c-Jun activates mesenchymal-related genes, broadly suppresses the pluripotent ones, and derails the obligatory mesenchymal to epithelial transition during reprogramming. Furthermore, inhibition of c-Jun by shRNA, dominant-negative c-Jun or Jdp2 enhances reprogramming and replaces Oct4 among the Yamanaka factors. Finally, Jdp2 anchors 5 non-Yamanaka factors (Id1, Jhdm1b, Lrh1, Sall4 and Glis1) to reprogram MEFs into iPSCs. Our studies reveal c-Jun as a guardian of somatic cell fate and its suppression opens the gate to pluripotency.

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: c-Jun is induced during ESC differentiation and required for proliferation of MEFs, but not ESCs.
Figure 2: c-Jun induces ESC differentiation and inhibits iPSC generation.
Figure 3: c-Jun suppresses pluripotent genes but induces EMT genes.
Figure 4: c-Jun regulates MET genes directly.
Figure 5: c-Jun dominant-negative mutants c-JunDN, Jdp2 and c-Jun shRNA can enhance the efficiency of iPSC generation.
Figure 6: c-Jun dominant-negative mutants c-JunDN, Jdp2 and c-Jun shRNA can substitute Oct4 partially.
Figure 7: c-JunDN and Jdp2 target similar genes as Oct4 during reprogramming.
Figure 8: Induction of pluripotent stem cells from MEFs by non-Yamanaka factors.

Similar content being viewed by others

Change history

  • 10 August 2015

    In the version of this Article originally published, panels from Fig. 8e were mistakenly reproduced as Fig. 6g. The correct panels for Fig. 6g (KS/Jdp2-iPSC) have been amended in all online versions of the Article.

  • 28 August 2015

    Nat. Cell Biol. 17, 856–867 (2015); published online 22 June 2015; corrected after print 10 August 2015 In the version of this Article originally published, panels from Fig. 8e were mistakenly reproduced as Fig. 6g. The correct panels for Fig. 6g (KS/Jdp2-iPSC) are shown here and have been amended in all online versions of the Article.

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).

    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  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  CAS  PubMed  Google Scholar 

  5. Yamanaka, S. Pluripotency and nuclear reprogramming. Phil. Trans. R. Soc. Lond. B 363, 2079–2087 (2008).

    Article  CAS  Google Scholar 

  6. 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. Nat. Cell Biol. 7, 1074–1082 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Adhikary, S. & Eilers, M. Transcriptional regulation and transformation by Myc proteins. Nat. Rev. Mol. Cell Biol. 6, 635–645 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Avilion, A. A. et al. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 17, 126–140 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nichols, J. et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 95, 379–391 (1998).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Halazonetis, T. D., Georgopoulos, K., Greenberg, M. E. & Leder, P. c-Jun dimerizes with itself and with c-Fos, forming complexes of different DNA binding affinities. Cell 55, 917–924 (1988).

    Article  CAS  PubMed  Google Scholar 

  16. Glover, J. N. & Harrison, S. C. Crystal structure of the heterodimeric bZIP transcription factor c-Fos-c-Jun bound to DNA. Nature 373, 257–261 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Chiu, R. et al. The c-Fos protein interacts with c-Jun/AP-1 to stimulate transcription of AP-1 responsive genes. Cell 54, 541–552 (1988).

    Article  CAS  PubMed  Google Scholar 

  18. Schutte, J. et al. jun-B inhibits and c-fos stimulates the transforming and trans-activating activities of c-jun. Cell 59, 987–997 (1989).

    Article  CAS  PubMed  Google Scholar 

  19. Suzuki, T. et al. Difference in transcriptional regulatory function between c-Fos and Fra-2. Nucleic Acids Res. 19, 5537–5542 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Angel, P. & Karin, M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochim. Biophys. Acta 1072, 129–157 (1991).

    CAS  PubMed  Google Scholar 

  21. Shaulian, E. & Karin, M. AP-1 as a regulator of cell life and death. Nat. Cell Biol. 4, E131–E136 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Shaulian, E. & Karin, M. AP-1 in cell proliferation and survival. Oncogene 20, 2390–2400 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Johnson, R., Spiegelman, B., Hanahan, D. & Wisdom, R. Cellular transformation and malignancy induced by ras require c-jun. Mol. Cell. Biol. 16, 4504–4511 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hilberg, F., Aguzzi, A., Howells, N. & Wagner, E. F. c-jun is essential for normal mouse development and hepatogenesis. Nature 365, 179–181 (1993).

    Article  CAS  PubMed  Google Scholar 

  25. Johnson, R. S., van Lingen, B., Papaioannou, V. E. & Spiegelman, B. M. A null mutation at the c-jun locus causes embryonic lethality and retarded cell growth in culture. Genes Dev. 7, 1309–1317 (1993).

    Article  CAS  PubMed  Google Scholar 

  26. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Hockemeyer, D. et al. Genetic engineering of human pluripotent cells using TALE nucleases. Nat. Biotechnol. 29, 731–734 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schreiber, M. et al. Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev. 13, 607–619 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 26, 101–106 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Chen, J. et al. Rational optimization of reprogramming culture conditions for the generation of induced pluripotent stem cells with ultra-high efficiency and fast kinetics. Cell Res. 21, 884–894 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Hirai, S., Bourachot, B. & Yaniv, M. Both Jun and Fos contribute to transcription activation by the heterodimer. Oncogene 5, 39–46 (1990).

    CAS  PubMed  Google Scholar 

  33. Bohmann, D. et al. Human proto-oncogene c-jun encodes a DNA binding protein with structural and functional properties of transcription factor AP-1. Science 238, 1386–1392 (1987).

    Article  CAS  PubMed  Google Scholar 

  34. Angel, P. et al. Oncogene jun encodes a sequence-specific trans-activator similar to AP-1. Nature 332, 166–171 (1988).

    Article  CAS  PubMed  Google Scholar 

  35. Smeal, T., Binetruy, B., Mercola, D. A., Birrer, M. & Karin, M. Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73. Nature 354, 494–496 (1991).

    Article  CAS  PubMed  Google Scholar 

  36. Lloyd, A., Yancheva, N. & Wasylyk, B. Transformation suppressor activity of a Jun transcription factor lacking its activation domain. Nature 352, 635–638 (1991).

    Article  CAS  PubMed  Google Scholar 

  37. Alani, R. et al. The transactivating domain of the c-Jun proto-oncoprotein is required for cotransformation of rat embryo cells. Mol. Cell. Biol. 11, 6286–6295 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pulverer, B. J., Kyriakis, J. M., Avruch, J., Nikolakaki, E. & Woodgett, J. R. Phosphorylation of C-Jun mediated by map kinases. Nature 353, 670–674 (1991).

    Article  CAS  PubMed  Google Scholar 

  39. Brown, P. H., Chen, T. K. & Birrer, M. J. Mechanism of action of a dominant-negative mutant of c-Jun. Oncogene 9, 791–799 (1994).

    CAS  PubMed  Google Scholar 

  40. Behrens, A., Sibilia, M. & Wagner, E. F. Amino-terminal phosphorylation of c-Jun regulates stress-induced apoptosis and cellular proliferation. Nat. Genet. 21, 326–329 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Aronheim, A., Zandi, E., Hennemann, H., Elledge, S. J. & Karin, M. Isolation of an AP-1 repressor by a novel method for detecting protein–protein interactions. Mol. Cell. Biol. 17, 3094–3102 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jin, C. et al. JDP2, a repressor of AP-1, recruits a histone deacetylase 3 complex to inhibit the retinoic acid-induced differentiation of F9 cells. Mol. Cell. Biol. 22, 4815–4826 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Piu, F., Aronheim, A., Katz, S. & Karin, M. AP-1 repressor protein JDP-2: inhibition of UV-mediated apoptosis through p53 down-regulation. Mol. Cell. Biol. 21, 3012–3024 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Wang, T. et al. The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. Cell Stem Cell 9, 575–587 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Hochedlinger, K., Yamada, Y., Beard, C. & Jaenisch, R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell 121, 465–477 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Heng, J. C. et al. The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell 6, 167–174 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Chen, J. et al. BMPs functionally replace Klf4 and support efficient reprogramming of mouse fibroblasts by Oct4 alone. Cell Res. 21, 205–212 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Maekawa, M. et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature 474, 225–229 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Breitenlechner, C. et al. Protein kinase A in complex with Rho-kinase inhibitors Y-27632, Fasudil, and H-1152P: structural basis of selectivity. Structure 11, 1595–1607 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Pan, G. Identification of a nuclear localization signal in OCT4 and generation of a dominant negative mutant by its ablation. J. Biol. Chem. 279, 37013–37020 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Pesce, M. & Scholer, H. R. Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells 19, 271–278 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Sridharan, R. et al. Role of the murine reprogramming factors in the induction of pluripotency. Cell 136, 364–377 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Boyer, L. A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kim, J. B. et al. Oct4-induced pluripotency in adult neural stem cells. Cell 136, 411–419 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Esch, D. et al. A unique Oct4 interface is crucial for reprogramming to pluripotency. Nat. Cell Biol. 15, 295–301 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Wagner, E. F. AP-1–Introductory remarks. Oncogene 20, 2334–2335 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Eferl, R. & Wagner, E. F. AP-1: a double-edged sword in tumorigenesis. Nat. Rev. Cancer 3, 859–868 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Jochum, W., Passegue, E. & Wagner, E. F. AP-1 in mouse development and tumorigenesis. Oncogene 20, 2401–2412 (2001).

    Article  CAS  PubMed  Google Scholar 

  61. Ozanne, B. W., Spence, H. J., McGarry, L. C. & Hennigan, R. F. Transcription factors control invasion: AP-1 the first among equals. Oncogene 26, 1–10 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Esteban, M. A. et al. Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell 6, 71–79 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Shu, J. et al. Induction of pluripotency in mouse somatic cells with lineage specifiers. Cell 153, 963–975 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Qin, D., Li, W., Zhang, J. & Pei, D. Direct generation of ES-like cells from unmodified mouse embryonic fibroblasts by Oct4/Sox2/Myc/Klf4. Cell Res. 17, 959–962 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Chen, J. et al. Towards an optimized culture medium for the generation of mouse induced pluripotent stem cells. J. Biol. Chem. 285, 31066–31072 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jing, L., Jiekai, c. & Duanqing, P. Reprogramming mouse embryonic fibroblasts using different reprogramming factors. Nat. Protoc. Exch. http://dx.doi.org/10.1038/protex.2015.063 (2015).

  67. Chen, J. et al. H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat. Genet. 45, 34–42 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Huang, K. et al. Ribosomal RNA gene transcription mediated by the master genome regulator protein CCCTC-binding factor (CTCF) is negatively regulated by the condensin complex. J. Biol. Chem. 288, 26067–26077 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Marson, A. et al. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134, 521–533 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Zheng, M. Esteban, B. Qin and X. Shu for their critical comments. We appreciate the contributions of K. Mo, S. Chu, J. Xie, X. Zhao, H. Pang, S. Huang, R. Luo, Y. Li and K. Lai for technical assistance. We are grateful to other members in our laboratory for their supporting work. This work was supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (grant XDA01020401), the National Basic Research Program of China (grants 2014CB965200, 2012CB966802, 2015CB964800), the National Natural Science Foundation of China (grants 31421004, 31401264, 91419310, 31271357, 91413110, 31471210), Guangdong Natural Science Funds for Distinguished Young Scholars (grant S2013050014040), and Bureau of Science and Technology of Guangzhou Municipality (grants 2014J2200077, 2012J2200070).

Author information

Authors and Affiliations

Authors

Contributions

J.L. designed and performed the experiments, and analysed the data. Q.H. initiated the study and performed reprogramming experiments with M.P., S.Y. and S.C. T.P. D.L. and K.H. performed ChIP experiments. B.W. and D.L. performed ESC differentiation. J.Y., H.L. and W.H. performed qPCR analyses and RNA-seq. X.W. and A.P.H. analysed the RNA-seq and ChIP-seq data. J.K., Q.H. and D.L. constructed plasmids. Haoyu W., Hongling W. and B.L. performed the gene editing. Z.Z. performed the zebrafish experiment. Yongqiang C. and H.S. performed blastocyst injection. L.G. and You C. cultured the cells and identified the iPSC lines. Jing C. and X.L. screened the compounds. H.Y. supervised the ChIP-seq experiment. G.P. supervised the gene editing and sequencing. J.L., Jiekai C. and D.P. wrote the manuscript. Jiekai C. and D.P. conceived and supervised the whole study. D.P. approved the final version.

Corresponding authors

Correspondence to Jiekai Chen or Duanqing Pei.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Relative expression of AP-1 family genes on ESCs and somatic cells.

(a) Immunostaining of c-JUN and NANOG in MEFs and ESCs. ESCs were cultured in feeder free (up panel) or feeder (middle panel) condition, respectively. Scale bars, 100 μm. (b) Relative expression of AP-1 family members in MEFs, iPSCs and ESCs (Mean ± s.e.m., Two-tailed, unpaired t-test; n = 3 independent experiments, Junb P = 0024, Fra1 P = 0.0483, Fra1 P = 0.021). (c) Time course analysis of AP-1 family members during RA induced ESCs differentiation by RT–qPCR. Mouse ESCs were cultured in mES+RA (1 μM). (Mean ± s.d., n = 3 independent experiments). (d) Time course analysis of the pluripotent genes and AP-1 family members during EB formation by RT–qPCR (Mean ± s.d., n = 3 independent experiments). (e,f) Time course analysis of c-Jun expression during embryo development of Zebra fish (e) and EB formation of human ESCs (f) (e,f, n = 2 independent experiments). (g) Relative expression of the pluripotent genes in WT and Jun-deficient ESCs. Cells were culture in 2i+LIF medium (n = 2 independent experiments). (h) Relative expression of germ layer markers during the EB generation in WT and Jun-deficient ESCs (Mean ± s.d., n = 3 independent experiments). For source data, see Supplementary Table 3.

Supplementary Figure 2 The function of AP-1 family members on somatic reprogramming.

(a) Phase and GFP images of iPSC colonies generated with OKS+Ctrl, OKS+c-Jun, OKSM+Ctrl, and OKSM+c-Jun. Cells were culture in iCD1 medium. Scale bars, 250 μm. (b) c-Jun inhibit OKSM induced reprogramming (Mean ± s.d., Two-tailed, unpaired t-test, n = 9 wells from 3 independent experiments, P < 0.001). (c) The effect of AP-1 family members to OKS induced reprogramming in mES+Vc medium (Mean ± s.d., Two-tailed, unpaired t-test; n = 7 wells (Mock, Vector, Fos, Fra1, Fra2) or 6 wells (JunB, ATF2, ATF3) from 3 independent experiment, Vector-Fos p < 0.001, Vector-Fra2 P < 0.001, Vector-Fra1 P < 0.001). (d) The effect of c-Jun on reprogramming at different time windows. OKS, Retrovirus; c-JunTetOn, lenti-virus carrying an inducible c-Jun code sequence under the control of TetOn promoter. Reprogramming cells were cultured in iCD1 medium, Doxycycline (2 μg ml−1) was administrated at different time windows as indicated (n = 4 wells from 2 independent experiments). The initial cell number for reprogramming is 2 × 104/well (p12) for OKS protocol, or 1.5 × 104/well (p12) for OKSM protocol. For source data, see Supplementary Table 6.

Supplementary Figure 3 c-Jun suppresses pluripotent genes and actives EMT relative genes.

(a) Time course analysis the expression of EMT relative genes Ets1, Smad3 and Tgfb2 by RT–qPCR (mean ± s.d., Two-tailed, unpaired t-test; n = 3 independent experiments, Ets1 P = 0.004, Smad3 P = 0.006, Tgfb2 P = 0.002). (b) Time course analysis the expression of pluripotent related genes Oct4, Rex1 and Sall4 by RT–qPCR. c-JunTetOn ESCs were cultured in 2i+LIF medium with Dox (2 μg ml−1) treatment (mean ± s.d., One-way ANOVA, Dunnett’s multiple comparisons test; n = 4 independent experiments, Oct4 q = 4.922, Rex1 q = 6.844, Sall4 q = 7.263). For source data, see Supplementary Table 6.

Supplementary Figure 4 c-Jun actives EMT relative genes directly.

(a) The bindings of c-Jun on the Oct4, Rex1 and Sall4 loci were determined by ChIP-PCR. Arrows indicate the predicted binding site containing the conserved AP binding motif (mean ± s.d., n = 3 independent experiments). (b) ChIP-seq result of c-Jun at EMT related genes Zeb2, Dusp5, Smad3, Thy1, Ets1 and Tgfb2. For source data, see Supplementary Table 6.

Supplementary Figure 5 c-JunDN and Jdp2 enhance reprogramming efficiency.

(a) The construction of different c-Jun mutants. Basic motif, Leucine-Zipper and Transactivation domain were shown as yellow rectangle, light green rectangle and Red rectangle, respectively. (b) Effect of various c-Jun mutants on reprogramming (mean ± s.d., One-way ANOVA, Dunnett’s multiple comparisons test; n = 6 wells pooled from 3 independent experiments, a.a1-334 q = 20.35, a.a170-334 q = 17.03, a.a254-334 q = 13.19). (c) The construction of different Jdp2 mutants. (d) Effect of various Jdp2 mutants on reprogramming (mean ± s.d., One-way ANOVA, Dunnett’s multiple comparisons test; n = 6 wells pooled from 3 independent experiments, a.a1-163 q = 7.349, a.a50-163 q = 10.29, a.a50-163 q = 8.639). For iPSC generation, the initial cell number was 2 × 104/well (p12), Oct4-GFP+ colonies were quantified at post-infection Day12 in mES+Vc medium For source data, see Supplementary Table 6.

Supplementary Figure 6 c-Jun antagonists can replace Oct4 in somatic reprogramming.

(a) Normal karyotypes of representative KSM/c-JunDN, KS/c-JunDN, KSM/Jdp2, KSM/Jdp2 iPSCs colonies. (b) Immunofluorescence analysis the expression of pluripotent markers of indicated iPSCs colonies. Scale bars, 100 μm. (c) Bisulfite genomic sequencing of the Nanog and Oct4 promoter regions in indicated iPSCs colonies. (d) PCR analysis show the KSM-Jdp2 iPSC colonies contributed to all three germ layer in chimeric mice. (e) Normal karyotypes of representative KSM/c-Jun shRNA iPSCs colonies.

Supplementary Figure 7 c-Jun antagonists regulated different genes with Oct4 in MEFs.

(a) Heatmap for RNA-seq data from MEFs infected with control, Oct4, c-JunDN and Jdp2. MEFs, cultured in 10%FBS for 72 h after virus infection, were harvested for analysis. (b) c-JunWT promotes, but c-JunDN, Jdp2 and Oct4 inhibit, the proliferation of MEFs. Cells were cultured in 10%FBS (mean ± s.d., n = 3 wells, this experiment were repeated twice, one of the represented experiment was showed). For source data, see Supplementary Table 5 (a) and Supplementary Table 6 (b).

Supplementary Figure 8 non-Yamanaka factors reprogramming.

(a) Id1 and Id3 enhance OS induced reprogramming in iCD1 or iCD1+BMP medium. The initial cell number was 2 × 104 per well (p12), cells were cultured in iCD1 or iCD1+BMP4 (10ng/ml) medium. Oct4-GFP+ colonies were quantitated at post-infection Day16 (mean ± s.d., n = 4 wells pooled from 2 independent experiments). (b) Phase and GFP images of iPSC colonies generated with Oct4/Sox2/Id1 and Oct4/Jhdm1b/Id1. Scale bars, 250 μm. (c) The replacement of Oct4 by indicated genes. The initial cell number was 2 × 104 per well (p12). Cells were cultured in iCD1 medium. Oct4-GFP+ colonies were quantitated at post-infection Day16 (mean ± s.d., n = 6 wells pooled from 2 independent experiments). (d) Retroviral integrations of 6F-iPSCs analysed by PCR with genomic DNA. (e) Normal karyotypes of representative 6F iPSCs. (f) Retroviral integrations of 6F-iPSCs analysed by PCR with genomic DNA. For source data, see Supplementary Table 3.

Supplementary Figure 9 Uncropped western blot images.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1253 kb)

Supplementary Table 1

Supplementary Information (XLSX 35 kb)

Supplementary Table 2

Supplementary Information (XLSX 27 kb)

Supplementary Table 3

Supplementary Information (XLSX 202 kb)

Supplementary Table 4

Supplementary Information (XLSX 231 kb)

Supplementary Table 5

Supplementary Information (XLSX 188 kb)

Supplementary Table 6

Supplementary Information (XLSX 106 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Han, Q., Peng, T. et al. The oncogene c-Jun impedes somatic cell reprogramming. Nat Cell Biol 17, 856–867 (2015). https://doi.org/10.1038/ncb3193

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3193

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer