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:

SCFβ-TRCP controls oncogenic transformation and neural differentiation through REST degradation

Abstract

The RE1-silencing transcription factor (REST, also known as NRSF) is a master repressor of neuronal gene expression and neuronal programmes in non-neuronal lineages1,2,3. Recently, REST was identified as a human tumour suppressor in epithelial tissues4, suggesting that its regulation may have important physiological and pathological consequences. However, the pathways controlling REST have yet to be elucidated. Here we show that REST is regulated by ubiquitin-mediated proteolysis, and use an RNA interference (RNAi) screen to identify a Skp1-Cul1-F-box protein complex containing the F-box protein β-TRCP (SCFβ-TRCP) as an E3 ubiquitin ligase responsible for REST degradation. β-TRCP binds and ubiquitinates REST and controls its stability through a conserved phospho-degron. During neural differentiation, REST is degraded in a β-TRCP-dependent manner. β-TRCP is required for proper neural differentiation only in the presence of REST, indicating that β-TRCP facilitates this process through degradation of REST. Conversely, failure to degrade REST attenuates differentiation. Furthermore, we find that β-TRCP overexpression, which is common in human epithelial cancers, causes oncogenic transformation of human mammary epithelial cells and that this pathogenic function requires REST degradation. Thus, REST is a key target in β-TRCP-driven transformation and the β-TRCP–REST axis is a new regulatory pathway controlling neurogenesis.

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: Identification of β-TRCP and FBW4 ubiquitin ligases as regulators of REST stability.
Figure 2: A conserved phospho-degron in REST is required for regulation by β-TRCP.
Figure 3: β-TRCP targets REST during oncogenic transformation.
Figure 4: The β-TRCP-REST pathway controls neural differentiation.

Similar content being viewed by others

References

  1. Chong, J. A. et al. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons. Cell 80, 949–957 (1995)

    Article  CAS  Google Scholar 

  2. Schoenherr, C. J. & Anderson, D. J. The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science 267, 1360–1363 (1995)

    Article  ADS  CAS  Google Scholar 

  3. Ballas, N. & Mandel, G. The many faces of REST oversee epigenetic programming of neuronal genes. Curr. Opin. Neurobiol. 15, 500–506 (2005)

    Article  CAS  Google Scholar 

  4. Westbrook, T. F. et al. A genetic screen for candidate tumor suppressors identifies REST. Cell 121, 837–848 (2005)

    Article  CAS  Google Scholar 

  5. Ballas, N., Grunseich, C., Lu, D. D., Speh, J. C. & Mandel, G. REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell 121, 645–657 (2005)

    Article  CAS  Google Scholar 

  6. Petroski, M. D. & Deshaies, R. J. Function and regulation of cullin-RING ubiquitin ligases. Nature Rev. Mol. Cell Biol. 6, 9–20 (2005)

    Article  CAS  Google Scholar 

  7. Bai, C. et al. SKP1 connects cell cycle regulators to the ubiquitin proteolysis machinery through a novel motif, the F-box. Cell 86, 263–274 (1996)

    Article  CAS  Google Scholar 

  8. Skowyra, D., Craig, K. L., Tyers, M., Elledge, S. J. & Harper, J. W. F-box proteins are receptors that recruit phosphorylated substrates to the SCF ubiquitin-ligase complex. Cell 91, 209–219 (1997)

    Article  CAS  Google Scholar 

  9. Guardavaccaro, D. et al. Control of meiotic and mitotic progression by the F box protein β-Trcp1 in vivo. Dev. Cell 4, 799–812 (2003)

    Article  CAS  Google Scholar 

  10. Shirogane, T., Jin, J., Ang, X. L. & Harper, J. W. SCFβ-TRCP controls clock-dependent transcription via casein kinase 1-dependent degradation of the mammalian period-1 (Per1) protein. J. Biol. Chem. 280, 26863–26872 (2005)

    Article  CAS  Google Scholar 

  11. Busino, L. et al. Degradation of Cdc25A by β-TrCP during S phase and in response to DNA damage. Nature 426, 87–91 (2003)

    Article  ADS  CAS  Google Scholar 

  12. Jin, J. et al. SCFβ-TRCP links Chk1 signaling to degradation of the Cdc25A protein phosphatase. Genes Dev. 17, 3062–3074 (2003)

    Article  CAS  Google Scholar 

  13. Winston, J. T. et al. The SCFβ-TRCP-ubiquitin ligase complex associates specifically with phosphorylated destruction motifs in IκBα- and β-catenin and stimulates IκBα- ubiquitination in vitro. Genes Dev. 13, 270–283 (1999)

    Article  CAS  Google Scholar 

  14. Fuchs, S. Y., Spiegelman, V. S. & Kumar, K. G. The many faces of β-TrCP E3 ubiquitin ligases: reflections in the magic mirror of cancer. Oncogene 23, 2028–2036 (2004)

    Article  CAS  Google Scholar 

  15. Wu, G. et al. Structure of a β-TrCP1-Skp1-β-catenin complex: destruction motif binding and lysine specificity of the SCF(β-TrCP1) ubiquitin ligase. Mol. Cell 11, 1445–1456 (2003)

    Article  CAS  Google Scholar 

  16. Zhao, J. J. et al. Human mammary epithelial cell transformation through the activation of phosphatidylinositol 3-kinase. Cancer Cell 3, 483–495 (2003)

    Article  CAS  Google Scholar 

  17. Kudo, Y. et al. Role of F-box protein β-Trcp1 in mammary gland development and tumorigenesis. Mol. Cell. Biol. 24, 8184–8194 (2004)

    Article  CAS  Google Scholar 

  18. Nishikawa, S., Jakt, L. M. & Era, T. Embryonic stem-cell culture as a tool for developmental cell biology. Nature Rev. Mol. Cell Biol. 8, 502–507 (2007)

    Article  CAS  Google Scholar 

  19. Ying, Q. L., Stavridis, M., Griffiths, D., Li, M. & Smith, A. Conversion of embryonic stem cells into neuroectodermal precursors in adherent monoculture. Nature Biotechnol. 21, 183–186 (2003)

    Article  CAS  Google Scholar 

  20. Aubert, J. et al. Screening for mammalian neural genes via fluorescence-activated cell sorter purification of neural precursors from Sox1-gfp knock-in mice. Proc. Natl Acad. Sci. USA 100 (Suppl 1). 11836–11841 (2003)

    Article  ADS  CAS  Google Scholar 

  21. Bain, G., Kitchens, D., Yao, M., Huettner, J. E. & Gottlieb, D. I. Embryonic stem cells express neuronal properties in vitro. Dev. Biol. 168, 342–357 (1995)

    Article  CAS  Google Scholar 

  22. Fuller, G. N. et al. Many human medulloblastoma tumors overexpress repressor element-1 silencing transcription (REST)/neuron-restrictive silencer factor, which can be functionally countered by REST-VP16. Mol. Cancer Ther. 4, 343–349 (2005)

    CAS  PubMed  Google Scholar 

  23. Su, X. et al. Abnormal expression of REST/NRSF and Myc in neural stem/progenitor cells causes cerebellar tumors by blocking neuronal differentiation. Mol. Cell. Biol. 26, 1666–1678 (2006)

    Article  CAS  Google Scholar 

  24. Saitoh, T. & Katoh, M. Expression profiles of β-TRCP1 and β-TRCP2, and mutation analysis of β-TRCP2 in gastric cancer. Int. J. Oncol. 18, 959–964 (2001)

    CAS  PubMed  Google Scholar 

  25. Noubissi, F. K. et al. CRD-BP mediates stabilization of β-TrCP1 and c-myc mRNA in response to β-catenin signalling. Nature 441, 898–901 (2006)

    Article  ADS  CAS  Google Scholar 

  26. Jin, J. et al. Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 18, 2573–2580 (2004)

    Article  CAS  Google Scholar 

  27. Ying, Q. L. & Smith, A. G. Defined conditions for neural commitment and differentiation. Methods Enzymol. 365, 327–341 (2003)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Mandel and R. Truant for providing REST-C antibodies and mRFP-REST cDNA, respectively. We thank J. Luo, D. Nguyen and D. Lee for suggestions and reading of the manuscript. We are grateful to R. Mulligan, A. Balazs, J. Jin, M. Sheng and V. Dixit for providing reagents, and to D. Guardavaccaro and M. Pagano for communicating results before publication. T.F.W. is a fellow of the Susan G. Komen for the Cure. G.H. is a Helen Hay Whitney post-doctoral fellow. X.L.A. is supported by a National Institutes of Health predoctoral fellowship. This work was supported by National Institutes of Health grants to Y.S., J.W.H., and a US Army Innovator Award to S.J.E. S.J.E. is an Investigator with the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to J. Wade Harper or Stephen J. Elledge.

Supplementary information

Supplementary Information

The file contains Supplementary Notes with additional references and Supplementary Figures S1-S11 with Legends. (PDF 1131 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Westbrook, T., Hu, G., Ang, X. et al. SCFβ-TRCP controls oncogenic transformation and neural differentiation through REST degradation. Nature 452, 370–374 (2008). https://doi.org/10.1038/nature06780

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature06780

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