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.

p53 regulates epithelial–mesenchymal transition and stem cell properties through modulating miRNAs

A Corrigendum to this article was published on 01 December 2011

An Addendum to this article was published on 01 December 2011

This article has been updated

Abstract

The epithelial–mesenchymal transition (EMT) has recently been linked to stem cell phenotype1,2. However, the molecular mechanism underlying EMT and regulation of stemness remains elusive. Here, using genomic approaches, we show that tumour suppressor p53 has a role in regulating both EMT and EMT-associated stem cell properties through transcriptional activation of the microRNA miR-200c. p53 transactivates miR-200c through direct binding to the miR-200c promoter. Loss of p53 in mammary epithelial cells leads to decreased expression of miR-200c and activates the EMT programme, accompanied by an increased mammary stem cell population. Re-expressing miR-200c suppresses genes that mediate EMT and stemness properties3,4 and thereby reverts the mesenchymal and stem-cell-like phenotype caused by loss of p53 to a differentiated epithelial cell phenotype. Furthermore, loss of p53 correlates with a decrease in the level of miR-200c, but an increase in the expression of EMT and stemness markers, and development of a high tumour grade in a cohort of breast tumours. This study elucidates a role for p53 in regulating EMT–MET (mesenchymal–epithelial transition) and stemness or differentiation plasticity, and reveals a potential therapeutic implication to suppress EMT-associated cancer stem cells through activation of the p53–miR-200c pathway.

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: p53 transcriptionally activates miR-183 and miR-200c.
Figure 2: p53 suppresses EMT phenotype and stem cell properties.
Figure 3: p53 suppresses the EMT phenotype and stem cell properties through upregulation of miR-200c.
Figure 4: Mutated p53 competes with wild-type p53 in the regulation of EMT and stemness markers.
Figure 5: Loss of p53 correlates with reduced miR-200c and enhanced ZEB1 and BMI1 expression levels.

Similar content being viewed by others

Change history

  • 15 November 2011

    In the version of this article initially published online and in print, Figs 2b and 4a contained panels that did not match those in Supplementary Fig. S5. These errors have been corrected in the HTML and PDF versions of the article.

References

  1. Mani, S. A. et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Polyak, K. & Weinberg, R. A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat. Rev. Cancer 9, 265–273 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Shimono, Y. et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 138, 592–603 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wellner, U. et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat. Cell Biol. 11, 1487–1495 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Thiery, J. P. & Sleeman, J. P. Complex networks orchestrate epithelial–mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 7, 131–142 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Junk, D. J. et al. Different mutant/wild-type p53 combinations cause a spectrum of increased invasive potential in nonmalignant immortalized human mammary epithelial cells. Neoplasia 10, 450–461 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Willis, A., Jung, E. J., Wakefield, T. & Chen, X. Mutant p53 exerts a dominant negative effect by preventing wild-type p53 from binding to the promoter of its target genes. Oncogene 23, 2330–2338 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Cadwell, C. & Zambetti, G. P. The effects of wild-type p53 tumor suppressor activity and mutant p53 gain-of-function on cell growth. Gene 277, 15–30 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Milner, J., Medcalf, E. A. & Cook, A. C. Tumor suppressor p53: analysis of wild-type and mutant p53 complexes. Mol. Cell Biol. 11, 12–19 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nielsen, L. L. et al. Efficacy of p53 adenovirus-mediated gene therapy against human breast cancer xenografts. Cancer Gene Ther. 4, 129–138 (1997).

    CAS  PubMed  Google Scholar 

  11. Araki, S. et al. TGF-β1-induced expression of human Mdm2 correlates with late-stage metastatic breast cancer. J. Clin. Invest. 120, 290–302 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Al-Hajj, M. & Clarke, M. F. Self-renewal and solid tumor stem cells. Oncogene 23, 7274–7282 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Kim, J. S., Lee, C., Bonifant, C. L., Ressom, H. & Waldman, T. Activation of p53-dependent growth suppression in human cells by mutations in PTEN or PIK3CA. Mol. Cell Biol. 27, 662–677 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Dull, T. et al. A third-generation lentivirus vector with a conditional packaging system. J. Virol. 72, 8463–8471 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Burk, U. et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 9, 582–589 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jacks, T. et al. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1–7 (1994).

    Article  CAS  PubMed  Google Scholar 

  17. Xia, W. et al. Phosphorylation/cytoplasmic localization of p21Cip1/WAF1 is associated with HER2/neu overexpression and provides a novel combination predictor for poor prognosis in breast cancer patients. Clin. Cancer Res. 10, 3815–3824 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Kramata, P. et al. Patches of mutant p53-immunoreactive epidermal cells induced by chronic UVB irradiation harbor the same p53 mutations as squamous cell carcinomas in the skin of hairless SKH-1 mice. Cancer Res. 65, 3577–3585 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Muret, J. et al. p53 status correlates with histopathological response in patients with soft tissue sarcomas treated using isolated limb perfusion with TNF-α and melphalan. Ann. Oncol. 19, 793–800 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Xirodimas, D. P. & Lane, D. P. Molecular evolution of the thermosensitive PAb1620 epitope of human p53 by DNA shuffling. J. Biol. Chem. 274, 28042–28049 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. A. Weinberg and S. Mani for providing HMEC and HMEC-Snail cells, E. Flores for providing the p53 knockout mice and J. Lang for technical assistance. This study was supported by RO1 CA109311, PO1 CA099031, Breast SPORE P50 CA116199, Cancer Center Support Grant CA16672, the National Breast Cancer Foundation, Inc., the Breast Cancer Research Foundation, the Sister Institution Fund of China Medical University Hospital and MD Anderson Cancer Center, the National Science Council of Taiwan (grants NSC-96-3111-B-039, NSC97-2917-I-564-107 and NSC-2632-B-039) and the Cancer Research Center for Excellence, Taiwan Department of Health (grant DOH99-TD-C-111-005 to M-C. H.). In memoriam, Ms. Serena Lin-Guo for her courageous fight against cancer.

Author information

Authors and Affiliations

Authors

Contributions

C-J.C., C-H.C. and M-C.H. designed and conceived the study. C-J.C. and C-H.C. performed the experiments and wrote the manuscript. W.X. performed and analysed the immunohistochemistry experiments. Y.X., S.K.R. and D.Y. performed the miRNA in situ hybridization experiments. J-Y.Y., C-W.L., W-H.Y., H-H.L. and M.L. helped with the biochemical experiments. C-T.C. and H.J.L. helped with the animal experiments. H.J.L. edited the manuscript.

Corresponding author

Correspondence to Mien-Chie Hung.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 570 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chang, CJ., Chao, CH., Xia, W. et al. p53 regulates epithelial–mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol 13, 317–323 (2011). https://doi.org/10.1038/ncb2173

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2173

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