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:

A new tumor suppressor role for the Notch pathway in bladder cancer

Abstract

The Notch signaling pathway controls cell fates through interactions between neighboring cells by positively or negatively affecting the processes of proliferation, differentiation and apoptosis in a context-dependent manner1. This pathway has been implicated in human cancer as both an oncogene and a tumor suppressor2. Here we report new inactivating mutations in Notch pathway components in over 40% of human bladder cancers examined. Bladder cancer is the fourth most commonly diagnosed malignancy in the male population of the United States3. Thus far, driver mutations in fibroblast growth factor receptor 3 (FGFR3) and, less commonly, in RAS proteins have been identified4,5. We show that Notch activation in bladder cancer cells suppresses proliferation both in vitro and in vivo by directly upregulating dual-specificity phosphatases (DUSPs), thus reducing the phosphorylation of ERK1 and ERK2 (ERK1/2). In mouse models, genetic inactivation of Notch signaling leads to Erk1/2 phosphorylation, resulting in tumorigenesis in the urinary tract. Collectively our findings show that loss of Notch activity is a driving event in urothelial cancer.

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: Identification, molecular characterization and clinical impact of Notch pathway alterations in patients with TCC.
Figure 2: Patients with TCC harboring mutations in the Notch pathway alone or in combination with FGFR3 or RAS mutations show increased ERK1/2 phosphorylation.
Figure 3: NOTCH1 suppresses TCC cell growth and ERK1/2 phosphorylation through direct DUSP transcriptional upregulation.
Figure 4: Mouse models of urothelial cancer.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

NCBI Reference Sequence

References

  1. Artavanis-Tsakonas, S., Rand, M.D. & Lake, R.J. Notch signaling: cell fate control and signal integration in development. Science 284, 770–776 (1999).

    Article  CAS  Google Scholar 

  2. Lobry, C., Oh, P. & Aifantis, I. Oncogenic and tumor suppressor functions of Notch in cancer: it's NOTCH what you think. J. Exp. Med. 208, 1931–1935 (2011).

    Article  CAS  Google Scholar 

  3. Jemal, A., Siegel, R., Xu, J. & Ward, E. Cancer statistics, 2010. CA Cancer J. Clin. 60, 277–300 (2010).

    Article  Google Scholar 

  4. Cordon-Cardo, C. Molecular alterations associated with bladder cancer initiation and progression. Scand. J. Urol. Nephrol. Suppl. 154–165 (2008).

    Article  Google Scholar 

  5. Knowles, M.A. Molecular pathogenesis of bladder cancer. Int. J. Clin. Oncol. 13, 287–297 (2008).

    Article  CAS  Google Scholar 

  6. Nicolas, M. et al. Notch1 functions as a tumor suppressor in mouse skin. Nat. Genet. 33, 416–421 (2003).

    Article  CAS  Google Scholar 

  7. Lefort, K. et al. Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKα kinases. Genes Dev. 21, 562–577 (2007).

    Article  CAS  Google Scholar 

  8. Klinakis, A. et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature 473, 230–233 (2011).

    Article  CAS  Google Scholar 

  9. Wang, N.J. et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc. Natl. Acad. Sci. USA 108, 17761–17766 (2011).

    Article  CAS  Google Scholar 

  10. Agrawal, N. et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science 333, 1154–1157 (2011).

    Article  CAS  Google Scholar 

  11. Stransky, N. et al. The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157–1160 (2011).

    Article  CAS  Google Scholar 

  12. Jerez, A. et al. Topography, clinical, and genomic correlates of 5q myeloid malignancies revisited. J. Clin. Oncol. 30, 1343–1349 (2012).

    Article  Google Scholar 

  13. Kimura, F. et al. Destabilization of chromosome 9 in transitional cell carcinoma of the urinary bladder. Br. J. Cancer 85, 1887–1893 (2001).

    Article  CAS  Google Scholar 

  14. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  Google Scholar 

  15. Funahashi, Y. et al. A notch1 ectodomain construct inhibits endothelial notch signaling, tumor growth, and angiogenesis. Cancer Res. 68, 4727–4735 (2008).

    Article  CAS  Google Scholar 

  16. Cancer Genome Atlas Research. N. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 507, 315–322 (2014).

  17. Guo, G. et al. Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat. Genet. 45, 1459–1463 (2013).

    Article  CAS  Google Scholar 

  18. Iyer, G. et al. Prevalence and co-occurrence of actionable genomic alterations in high-grade bladder cancer. J. Clin. Oncol. 31, 3133–3140 (2013).

    Article  CAS  Google Scholar 

  19. Cazier, J.B. et al. Whole-genome sequencing of bladder cancers reveals somatic CDKN1A mutations and clinicopathological associations with mutation burden. Nat. Commun. 5, 3756 (2014).

    Article  CAS  Google Scholar 

  20. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

  21. Weng, A.P. et al. Growth suppression of pre-T acute lymphoblastic leukemia cells by inhibition of notch signaling. Mol. Cell. Biol. 23, 655–664 (2003).

    Article  CAS  Google Scholar 

  22. Maraver, A. et al. Therapeutic effect of γ-secretase inhibition in KrasG12V-driven non-small cell lung carcinoma by derepression of DUSP1 and inhibition of ERK. Cancer Cell 22, 222–234 (2012).

    Article  CAS  Google Scholar 

  23. Yu, H.M., Liu, B., Chiu, S.Y., Costantini, F. & Hsu, W. Development of a unique system for spatiotemporal and lineage-specific gene expression in mice. Proc. Natl. Acad. Sci. USA 102, 8615–8620 (2005).

    Article  CAS  Google Scholar 

  24. Perl, A.K., Wert, S.E., Nagy, A., Lobe, C.G. & Whitsett, J.A. Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc. Natl. Acad. Sci. USA 99, 10482–10487 (2002).

    Article  CAS  Google Scholar 

  25. Karni-Schmidt, O. et al. Distinct expression profiles of p63 variants during urothelial development and bladder cancer progression. Am. J. Pathol. 178, 1350–1360 (2011).

    Article  CAS  Google Scholar 

  26. Shin, K. et al. Cellular origin of bladder neoplasia and tissue dynamics of its progression to invasive carcinoma. Nat. Cell Biol. 16, 469–478 (2014).

    Article  CAS  Google Scholar 

  27. Choi, W. et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 25, 152–165 (2014).

    Article  CAS  Google Scholar 

  28. Ho, P.L., Kurtova, A. & Chan, K.S. Normal and neoplastic urothelial stem cells: getting to the root of the problem. Nat. Rev. Urol. 9, 583–594 (2012).

    Article  CAS  Google Scholar 

  29. Buonamici, S. et al. CCR7 signalling as an essential regulator of CNS infiltration in T-cell leukaemia. Nature 459, 1000–1004 (2009).

    Article  CAS  Google Scholar 

  30. Dotto, G.P. Crosstalk of Notch with p53 and p63 in cancer growth control. Nat. Rev. Cancer 9, 587–595 (2009).

    Article  CAS  Google Scholar 

  31. Han, H. et al. Inducible gene knockout of transcription factor recombination signal binding protein-J reveals its essential role in T versus B lineage decision. Int. Immunol. 14, 637–645 (2002).

    Article  CAS  Google Scholar 

  32. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    Article  CAS  Google Scholar 

  33. Ahmad, I. et al. K-Ras and β-catenin mutations cooperate with Fgfr3 mutations in mice to promote tumorigenesis in the skin and lung, but not in the bladder. Dis. Model. Mech. 4, 548–555 (2011).

    Article  CAS  Google Scholar 

  34. Zhou, H. et al. Urothelial tumor initiation requires deregulation of multiple signaling pathways: implications in target-based therapies. Carcinogenesis 33, 770–780 (2012).

    Article  CAS  Google Scholar 

  35. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔC(T) method. Methods 25, 402–408 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Kitajewski (Columbia University), P. Dotto (University of Lausanne), M. Post (Maastricht University), D. Stravopodis (University f Athens) and K. Zoi (Biomedical Research Foundation Academy of Athens) for donating reagents, S. Artavanis-Tsakonas (Harvard Medical School) for donating mice, Z. Kanaki for pronuclear injections for the generation of the UpkII-Cre-eGFP line, M. Roubelaki for help with the cell cycle analysis and A. Efstratiadis for critically reading the manuscript. This work was supported by a Marie Curie Reintegration grant (224821), a Greek General Secretariat for Research and Technology 'Excellence' grant (UTN_1466) and a Fondation Santé Grant in Biomedical Sciences to A.K.

Author information

Authors and Affiliations

Authors

Contributions

A.K. and T.R. conceived the study and designed all experiments. T.R., P.V. and M.A. performed all experiments. A.P. analyzed expression array data. C.V. performed the mouse histology analysis. A.S. and K.S. provided the library of human tumor samples. A.K. wrote the manuscript.

Corresponding author

Correspondence to Apostolos Klinakis.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1–10 (PDF 4201 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rampias, T., Vgenopoulou, P., Avgeris, M. et al. A new tumor suppressor role for the Notch pathway in bladder cancer. Nat Med 20, 1199–1205 (2014). https://doi.org/10.1038/nm.3678

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3678

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