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.

miR-380-5p represses p53 to control cellular survival and is associated with poor outcome in MYCN-amplified neuroblastoma

Abstract

Inactivation of the p53 tumor suppressor pathway allows cell survival in times of stress and occurs in many human cancers; however, normal embryonic stem cells and some cancers such as neuroblastoma maintain wild-type human TP53 and mouse Trp53 (referred to collectively as p53 herein). Here we describe a miRNA, miR-380-5p, that represses p53 expression via a conserved sequence in the p53 3′ untranslated region (UTR). miR-380-5p is highly expressed in mouse embryonic stem cells and neuroblastomas, and high expression correlates with poor outcome in neuroblastomas with neuroblastoma derived v-myc myelocytomatosis viral-related oncogene (MYCN) amplification. miR-380 overexpression cooperates with activated HRAS oncoprotein to transform primary cells, block oncogene-induced senescence and form tumors in mice. Conversely, inhibition of endogenous miR-380-5p in embryonic stem or neuroblastoma cells results in induction of p53, and extensive apoptotic cell death. In vivo delivery of a miR-380-5p antagonist decreases tumor size in an orthotopic mouse model of neuroblastoma. We demonstrate a new mechanism of p53 regulation in cancer and stem cells and uncover a potential therapeutic target for neuroblastoma.

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: The p53 3′ UTR contains binding sites for miR-380-5p, a developmentally restricted miRNA.
Figure 2: miR-380-5p is required for ES cell survival.
Figure 3: miR-380-5p targets p53 and decreases cell death after genotoxic stress.
Figure 4: miR-380 prevents oncogene-induced senescence and increases tumor incidence in a mouse mammary cancer model.
Figure 5: miR-380-5p is expressed in mouse and human neuroblastoma and is associated with poor outcome in subjects with MYCN amplification.
Figure 6: Treatment with miR-380-5p antagonist induces p53-dependent cell death in neuroblastoma cells and decreases tumor growth in vivo.

Similar content being viewed by others

References

  1. Horn, H.F. & Vousden, K.H. Coping with stress: multiple ways to activate p53. Oncogene 26, 1306–1316 (2007).

    Article  CAS  Google Scholar 

  2. Levine, A.J., Hu, W. & Feng, Z. The P53 pathway: what questions remain to be explored? Cell Death Differ. 13, 1027–1036 (2006).

    Article  CAS  Google Scholar 

  3. Vogelstein, B. & Kinzler, K.W. Cancer genes and the pathways they control. Nat. Med. 10, 789–799 (2004).

    Article  CAS  Google Scholar 

  4. Petitjean, A., Achatz, M.I., Borresen-Dale, A.L., Hainaut, P. & Olivier, M. TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes. Oncogene 26, 2157–2165 (2007).

    Article  CAS  Google Scholar 

  5. Vazquez, A., Bond, E.E., Levine, A.J. & Bond, G.L. The genetics of the p53 pathway, apoptosis and cancer therapy. Nat. Rev. Drug Discov. 7, 979–987 (2008).

    Article  CAS  Google Scholar 

  6. Martins, C.P., Brown-Swigart, L. & Evan, G.I. Modeling the therapeutic efficacy of p53 restoration in tumors. Cell 127, 1323–1334 (2006).

    Article  CAS  Google Scholar 

  7. Ventura, A. et al. Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661–665 (2007).

    Article  CAS  Google Scholar 

  8. Xue, W. et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 (2007).

    Article  CAS  Google Scholar 

  9. Tweddle, D.A. et al. The p53 pathway and its inactivation in neuroblastoma. Cancer Lett. 197, 93–98 (2003).

    Article  CAS  Google Scholar 

  10. Yoon, H. et al. Gene expression profiling of isogenic cells with different TP53 gene dosage reveals numerous genes that are affected by TP53 dosage and identifies CSPG2 as a direct target of p53. Proc. Natl. Acad. Sci. USA 99, 15632–15637 (2002).

    Article  CAS  Google Scholar 

  11. Zhao, R. et al. Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev. 14, 981–993 (2000).

    Article  CAS  Google Scholar 

  12. Clarke, A.R., Gledhill, S., Hooper, M.L., Bird, C.C. & Wyllie, A.H. p53 dependence of early apoptotic and proliferative responses within the mouse intestinal epithelium following γ-irradiation. Oncogene 9, 1767–1773 (1994).

    CAS  PubMed  Google Scholar 

  13. Clarke, A.R. et al. Thymocyte apoptosis induced by p53-dependent and independent pathways. Nature 362, 849–852 (1993).

    Article  CAS  Google Scholar 

  14. Lowe, S.W., Schmitt, E.M., Smith, S.W., Osborne, B.A. & Jacks, T. p53 is required for radiation-induced apoptosis in mouse thymocytes. Nature 362, 847–849 (1993).

    Article  CAS  Google Scholar 

  15. Gottlieb, E. et al. Transgenic mouse model for studying the transcriptional activity of the p53 protein: age- and tissue-dependent changes in radiation-induced activation during embryogenesis. EMBO J. 16, 1381–1390 (1997).

    Article  CAS  Google Scholar 

  16. Venkatachalam, S. et al. Retention of wild-type p53 in tumors from p53 heterozygous mice: reduction of p53 dosage can promote cancer formation. EMBO J. 17, 4657–4667 (1998).

    Article  CAS  Google Scholar 

  17. Halaby, M.J. & Yang, D.Q. p53 translational control: a new facet of p53 regulation and its implication for tumorigenesis and cancer therapeutics. Gene 395, 1–7 (2007).

    Article  CAS  Google Scholar 

  18. Flynt, A.S. & Lai, E.C. Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat. Rev. Genet. 9, 831–842 (2008).

    Article  CAS  Google Scholar 

  19. Ventura, A. & Jacks, T. MicroRNAs and cancer: short RNAs go a long way. Cell 136, 586–591 (2009).

    Article  CAS  Google Scholar 

  20. Esquela-Kerscher, A. & Slack, F.J. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 6, 259–269 (2006).

    Article  CAS  Google Scholar 

  21. Enright, A.J. et al. MicroRNA targets in Drosophila. Genome Biol. 5, R1 (2003).

    Article  Google Scholar 

  22. Saetrom, P. et al. Distance constraints between microRNA target sites dictate efficacy and cooperativity. Nucleic Acids Res. 35, 2333–2342 (2007).

    Article  CAS  Google Scholar 

  23. Zhao, Y., Samal, E. & Srivastava, D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature 436, 214–220 (2005).

    Article  CAS  Google Scholar 

  24. Kuhn, D.E. et al. Experimental validation of miRNA targets. Methods 44, 47–54 (2008).

    Article  CAS  Google Scholar 

  25. Seitz, H. et al. A large imprinted microRNA gene cluster at the mouse Dlk1-Gtl2 domain. Genome Res. 14, 1741–1748 (2004).

    Article  CAS  Google Scholar 

  26. Tedeschi, A. & Di Giovanni, S. The non-apoptotic role of p53 in neuronal biology: enlightening the dark side of the moon. EMBO Rep. 10, 576–583 (2009).

    Article  CAS  Google Scholar 

  27. Le, M.T. et al. MicroRNA-125b is a novel negative regulator of p53. Genes Dev. 23, 862–876 (2009).

    Article  CAS  Google Scholar 

  28. Fu, L., Ma, W. & Benchimol, S. A translation repressor element resides in the 3′ untranslated region of human p53 mRNA. Oncogene 18, 6419–6424 (1999).

    Article  CAS  Google Scholar 

  29. Mazan-Mamczarz, K. et al. RNA-binding protein HuR enhances p53 translation in response to ultraviolet light irradiation. Proc. Natl. Acad. Sci. USA 100, 8354–8359 (2003).

    Article  CAS  Google Scholar 

  30. Sarkisian, C.J. et al. Dose-dependent oncogene-induced senescence in vivo and its evasion during mammary tumorigenesis. Nat. Cell Biol. 9, 493–505 (2007).

    Article  CAS  Google Scholar 

  31. Swarbrick, A., Roy, E., Allen, T. & Bishop, J.M. Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response. Proc. Natl. Acad. Sci. USA 105, 5402–5407 (2008).

    Article  CAS  Google Scholar 

  32. Welm, A.L., Kim, S., Welm, B.E. & Bishop, J.M. MET and MYC cooperate in mammary tumorigenesis. Proc. Natl. Acad. Sci. USA 102, 4324–4329 (2005).

    Article  CAS  Google Scholar 

  33. Hemann, M.T. et al. An epi-allelic series of p53 hypomorphs created by stable RNAi produces distinct tumor phenotypes in vivo. Nat. Genet. 33, 396–400 (2003).

    Article  CAS  Google Scholar 

  34. Scott, G.K. et al. Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b. J. Biol. Chem. 282, 1479–1486 (2007).

    Article  CAS  Google Scholar 

  35. Tweddle, D.A., Malcolm, A.J., Bown, N., Pearson, A.D. & Lunec, J. Evidence for the development of p53 mutations after cytotoxic therapy in a neuroblastoma cell line. Cancer Res. 61, 8–13 (2001).

    CAS  PubMed  Google Scholar 

  36. Weiss, W.A., Aldape, K., Mohapatra, G., Feuerstein, B.G. & Bishop, J.M. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 16, 2985–2995 (1997).

    Article  CAS  Google Scholar 

  37. Chesler, L. et al. Chemotherapy-induced apoptosis in a transgenic model of neuroblastoma proceeds through p53 induction. Neoplasia 10, 1268–1274 (2008).

    Article  CAS  Google Scholar 

  38. Liu, T. et al. Activation of tissue transglutaminase transcription by histone deacetylase inhibition as a therapeutic approach for Myc oncogenesis. Proc. Natl. Acad. Sci. USA 104, 18682–18687 (2007).

    Article  CAS  Google Scholar 

  39. Chesler, L. et al. Inhibition of phosphatidylinositol 3-kinase destabilizes Mycn protein and blocks malignant progression in neuroblastoma. Cancer Res. 66, 8139–8146 (2006).

    Article  CAS  Google Scholar 

  40. Hogarty, M.D. et al. ODC1 is a critical determinant of MYCN oncogenesis and a therapeutic target in neuroblastoma. Cancer Res. 68, 9735–9745 (2008).

    Article  CAS  Google Scholar 

  41. Rounbehler, R.J. et al. Targeting ornithine decarboxylase impairs development of MYCN-amplified neuroblastoma. Cancer Res. 69, 547–553 (2009).

    Article  CAS  Google Scholar 

  42. Davis, S. et al. Potent inhibition of microRNA in vivo without degradation. Nucleic Acids Res. 37, 70–77 (2009).

    Article  CAS  Google Scholar 

  43. Stadtfeld, M. et al. Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature 465, 175–181 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  45. Kota, J. et al. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell 137, 1005–1017 (2009).

    Article  CAS  Google Scholar 

  46. Medina, P.P., Nolde, M. & Slack, F.J. OncomiR addiction in an in vivo model of microRNA-21–induced pre-B-cell lymphoma. Nature advance online publication, doi:10.1038/nature09284 (8 August 2010).

  47. Ma, L. et al. Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model. Nat. Biotechnol. 28, 341–347 (2010).

    Article  CAS  Google Scholar 

  48. Zuker, M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 31, 3406–3415 (2003).

    Article  CAS  Google Scholar 

  49. Haber, M. et al. Association of high-level MRP1 expression with poor clinical outcome in a large prospective study of primary neuroblastoma. J. Clin. Oncol. 24, 1546–1553 (2006).

    Article  CAS  Google Scholar 

  50. Grundhoff, A., Sullivan, C.S. & Ganem, D. A combined computational and microarray-based approach identifies novel microRNAs encoded by human gamma-herpesviruses. RNA 12, 733–750 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We gratefully thank J.M. Bishop, N.K. Hayward and R.L. Sutherland for their support of this project, the Children's Oncology Group and M. Grimmer (University of California–San Francisco) for providing tumor samples, D. Lynch and J. Brugge (Harvard Medical School) for MCF10A cells expressing the ecotropic receptor, R. Jaenisch (Whitehead Institute) for Trp53−/− ES cells and S. Lowe (Cold Spring Harbor) for the p53 shRNA retrovirus. TH-MYCN transgenic mice were from W. Weiss (University of California–San Francisco). This work was supported by grants from the US National Institutes of Health: P50-CA58207, K08-CA104032 and 1R01CA136717 (to A.G.), 5R01DC005991 (to N.L.), R01CA102321, R01NS055750 and P01CA081403 (to W.W.) and K08NS48118 (to R.B.); the Susan G. Komen Foundation; the University of California–San Francisco Program for Breakthrough Biomedical Research (to A.G.); the G.W. Hooper Foundation; the Australian National Health and Medical Research Council (to T.P., M.H., M.D.N. and A. Swarbrick) and the Cancer Institute New South Wales (M.H. and M.D.N.). A. Swarbrick is a recipient of a Cancer Institute New South Wales early career development fellowship, and R.L.J. received a US National Science Foundation fellowship. A.G. is a V-Foundation Scholar, A. Shaw is a Cancer Institute New South Wales Research Scholar and Y.P. is supported by an Australian Postgraduate Award from the Australian National Health and Medical Research Council.

Author information

Authors and Affiliations

Authors

Contributions

S.L.W., A. Swarbrick and A.G. conceived and designed the experiments, discussed the results and wrote the manuscript. A. Swarbrick, S.L.W., A. Shaw, Y.P., A.N., A.G., R.L.J., C.S.S., C.S.H., P.L., A.B., N.H., Y.C. and L.L. performed experiments. L.J.A. and M.D.N. performed statistical analysis of the human neuroblastoma data set. A.C. provided human samples and clinical data, and E.G.M. provided anti-miRs for in vivo studies. M.H., T.P., W.W., N.L. and C.S.S. supervised experiments or experimental design.

Corresponding authors

Correspondence to Alexander Swarbrick or Andrei Goga.

Ethics declarations

Competing interests

E.G.M. is an employee and shareholder of Regulus Therapeutics.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Table 1 and Supplementary Methods (PDF 1169 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Swarbrick, A., Woods, S., Shaw, A. et al. miR-380-5p represses p53 to control cellular survival and is associated with poor outcome in MYCN-amplified neuroblastoma. Nat Med 16, 1134–1140 (2010). https://doi.org/10.1038/nm.2227

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2227

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research