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.

  • Original Article
  • Published:

SPSB3 targets SNAIL for degradation in GSK-3β phosphorylation-dependent manner and regulates metastasis

Abstract

Epithelial-mesenchymal transition (EMT) is a process during which normal epithelial cells acquire mesenchymal characteristics. EMT has a critical role in various human diseases especially in cancer. EMT facilitates tumor initiation and progression by mediating cancer cell stemness and motility. Zinc finger transcription factor SNAIL is one of the most important initiators of EMT. Therefore, it is of great significance to understand the regulating mechanism of SNAIL. In this study, we carried out a luciferase-based genome-wide screening using small interfering RNA library against ~200 of E3 ligases and ubiquitin-related genes and identified SOCS box protein SPSB3 as a novel E3 ligase component that targets SNAIL into polyubiquitination and degradation in response to GSK-3β phosphorylation of SNAIL. Functionally, we observed that SPSB3 overexpression greatly inhibits tumor metastasis by regulating SNAIL degradation both in vitro and in vivo. The expression of SPSB3 and SNAIL are negatively correlated in human esophageal squamous cell carcinoma tissues, and low SPSB3 expression indicates lymph node metastasis. Moreover, high SPSB3 expression indicates good survivals in various kinds of cancer. Collectively, these findings suggest that SPSB3-mediated SNAIL degradation has a vital role in regulating EMT and cancer progression.

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
Figure 2
Figure 3
Figure 4
Figure 5

Similar content being viewed by others

References

  1. Wan L, Pantel K, Kang Y . Tumor metastasis: moving new biological insights into the clinic. Nat Med 2013; 19: 1450–1464.

    Article  CAS  PubMed  Google Scholar 

  2. Marino N, Nakayama J, Collins JW, Steeg PS . Insights into the biology and prevention of tumor metastasis provided by the Nm23 metastasis suppressor gene. Cancer Metastasis Rev 2012; 31: 593–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pantel K, Brakenhoff RH . Dissecting the metastatic cascade. Nat Rev Cancer 2004; 4: 448–456.

    Article  CAS  PubMed  Google Scholar 

  4. Steeg PS . Targeting metastasis. Nat Rev Cancer 2016; 16: 201–218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Felipe Lima J, Nofech-Mozes S, Bayani J, Bartlett JM . EMT in breast carcinoma-a review. J Clin Med 2016; 5: pii: E65.

    Article  Google Scholar 

  6. Barrallo-Gimeno A, Nieto MA . The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development 2005; 132: 3151–3161.

    Article  CAS  PubMed  Google Scholar 

  7. Peinado H, Olmeda D, Cano A . Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 2007; 7: 415–428.

    Article  CAS  PubMed  Google Scholar 

  8. Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Sterner CJ et al. The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell 2005; 8: 197–209.

    Article  CAS  PubMed  Google Scholar 

  9. Diaz VM, Vinas-Castells R, Garcia de Herreros A . Regulation of the protein stability of EMT transcription factors. Cell Adh Migr 2014; 8: 418–428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Vinas-Castells R, Beltran M, Valls G, Gomez I, Garcia JM, Montserrat-Sentis B et al. The hypoxia-controlled FBXL14 ubiquitin ligase targets SNAIL1 for proteasome degradation. J Biol Chem 2010; 285: 3794–3805.

    Article  CAS  PubMed  Google Scholar 

  11. Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M et al. Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol 2004; 6: 931–940.

    Article  CAS  PubMed  Google Scholar 

  12. Vinas-Castells R, Frias A, Robles-Lanuza E, Zhang K, Longmore GD, Garcia de Herreros A et al. Nuclear ubiquitination by FBXL5 modulates Snail1 DNA binding and stability. Nucleic Acids Res 2014; 42: 1079–1094.

    Article  CAS  PubMed  Google Scholar 

  13. Jin Y, Shenoy AK, Doernberg S, Chen H, Luo H, Shen H et al. FBXO11 promotes ubiquitination of the Snail family of transcription factors in cancer progression and epidermal development. Cancer Lett 2015; 362: 70–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zheng H, Shen M, Zha YL, Li W, Wei Y, Blanco MA et al. PKD1 phosphorylation-dependent degradation of SNAIL by SCF-FBXO11 regulates epithelial-mesenchymal transition and metastasis. Cancer Cell 2014; 26: 358–373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sarikas A, Hartmann T, Pan ZQ . The cullin protein family. Genome Biol 2011; 12: 220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Okumura F, Joo-Okumura A, Nakatsukasa K, Kamura T . The role of cullin 5-containing ubiquitin ligases. Cell Div 2016; 11: 1.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008; 133: 704–715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cardozo T, Pagano M . The SCF ubiquitin ligase: insights into a molecular machine. Nat Rev Mol Cell Biol 2004; 5: 739–751.

    Article  CAS  PubMed  Google Scholar 

  19. Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat 2010; 123: 725–731.

    Article  PubMed  Google Scholar 

  20. Wang Y, Shi J, Chai K, Ying X, Zhou BP . The role of snail in EMT and tumorigenesis. Curr Cancer Drug Targets 2013; 13: 963–972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD, Doiphode RY et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells 2009; 27: 2059–2068.

    Article  CAS  PubMed  Google Scholar 

  22. Bruyere F, Namdarian B, Corcoran NM, Pedersen J, Ockrim J, Voelzke BB et al. Snail expression is an independent predictor of tumor recurrence in superficial bladder cancers. Urol Oncol 2010; 28: 591–596.

    Article  PubMed  Google Scholar 

  23. De Craene B, Berx G . Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer 2013; 13: 97–110.

    Article  CAS  PubMed  Google Scholar 

  24. Thiery JP, Acloque H, Huang RY, Nieto MA . Epithelial-mesenchymal transitions in development and disease. Cell 2009; 139: 871–890.

    Article  CAS  PubMed  Google Scholar 

  25. Wu Y, Deng J, Rychahou PG, Qiu S, Evers BM, Zhou BP . Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell 2009; 15: 416–428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen Y, Zhou C, Ji W, Mei Z, Hu B, Zhang W et al. ELL targets c-Myc for proteasomal degradation and suppresses tumour growth. Nat Commun 2016; 7: 11057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Paul I, Ahmed SF, Bhowmik A, Deb S, Ghosh MK . The ubiquitin ligase CHIP regulates c-Myc stability and transcriptional activity. Oncogene 2013; 32: 1284–1295.

    Article  CAS  PubMed  Google Scholar 

  28. Mei Z, Zhang D, Hu B, Wang J, Shen X, Xiao W . FBXO32 targets c-Myc for proteasomal degradation and inhibits c-Myc activity. J Biol Chem 2015; 290: 16202–16214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Welcker M, Orian A, Jin J, Grim JE, Harper JW, Eisenman RN et al. The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci USA 2004; 101: 9085–9090.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Popov N, Schulein C, Jaenicke LA, Eilers M . Ubiquitylation of the amino terminus of Myc by SCF(beta-TrCP) antagonizes SCF(Fbw7)-mediated turnover. Nat Cell Biol 2010; 12: 973–981.

    Article  CAS  PubMed  Google Scholar 

  31. Nishiya T, Matsumoto K, Maekawa S, Kajita E, Horinouchi T, Fujimuro M et al. Regulation of inducible nitric-oxide synthase by the SPRY domain- and SOCS box-containing proteins. J Biol Chem 2011; 286: 9009–9019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bastea LI, Doppler H, Balogun B, Storz P . Protein kinase D1 maintains the epithelial phenotype by inducing a DNA-bound, inactive SNAI1 transcriptional repressor complex. PLoS ONE 2012; 7: e30459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Du C, Zhang C, Hassan S, Biswas MH, Balaji KC . Protein kinase D1 suppresses epithelial-to-mesenchymal transition through phosphorylation of snail. Cancer Res 2010; 70: 7810–7819.

    Article  CAS  PubMed  Google Scholar 

  34. Pon YL, Zhou HY, Cheung AN, Ngan HY, Wong AS . p70 S6 kinase promotes epithelial to mesenchymal transition through snail induction in ovarian cancer cells. Cancer Res 2008; 68: 6524–6532.

    Article  CAS  PubMed  Google Scholar 

  35. Yang Z, Rayala S, Nguyen D, Vadlamudi RK, Chen S, Kumar R . Pak1 phosphorylation of snail, a master regulator of epithelial-to-mesenchyme transition, modulates snail's subcellular localization and functions. Cancer Res 2005; 65: 3179–3184.

    Article  CAS  PubMed  Google Scholar 

  36. Zhang K, Rodriguez-Aznar E, Yabuta N, Owen RJ, Mingot JM, Nojima H et al. Lats2 kinase potentiates Snail1 activity by promoting nuclear retention upon phosphorylation. EMBO J 2012; 31: 29–43.

    Article  PubMed  Google Scholar 

  37. Shimada Y, Imamura M, Wagata T, Yamaguchi N, Tobe T . Characterization of 21 newly established esophageal cancer cell lines. Cancer 1992; 69: 277–284.

    Article  CAS  PubMed  Google Scholar 

  38. Santner SJ, Dawson PJ, Tait L, Soule HD, Eliason J, Mohamed AN et al. Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cells. Breast Cancer Res Treat 2001; 65: 101–110.

    Article  CAS  PubMed  Google Scholar 

  39. Zhou H, Liu Y, Zhu R, Ding F, Wan Y, Li Y et al. FBXO32 suppresses breast cancer tumorigenesis through targeting KLF4 to proteasomal degradation. Oncogene 2017; 36: 3312–3321.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tian Y, Luo A, Cai Y, Su Q, Ding F, Chen H et al. MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. J Biol Chem 2010; 285: 7986–7994.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Professor Binhua P Zhou (University of Kentucky College of Medicine, Lexington, KY, USA) for kindly providing the plasmids of WT-GSK-3β and KD-GSK-3β. This work was supported by grants from the National Key R&D program of China (2016YFC1302100, 2013CB911004), the National Science Foundation of China (81420108025) and CAMS Initiative for Innovative Medicine (2016-I2M-1-001).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Z Liu.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Zhou, H., Zhu, R. et al. SPSB3 targets SNAIL for degradation in GSK-3β phosphorylation-dependent manner and regulates metastasis. Oncogene 37, 768–776 (2018). https://doi.org/10.1038/onc.2017.370

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2017.370

This article is cited by

Search

Quick links