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.

  • Article
  • Published:

Wnt activity defines colon cancer stem cells and is regulated by the microenvironment

Abstract

Despite the presence of mutations in APC or β-catenin, which are believed to activate the Wnt signalling cascade constitutively, most colorectal cancers show cellular heterogeneity when β-catenin localization is analysed, indicating a more complex regulation of Wnt signalling. We explored this heterogeneity with a Wnt reporter construct and observed that high Wnt activity functionally designates the colon cancer stem cell (CSC) population. In adenocarcinomas, high activity of the Wnt pathway is observed preferentially in tumour cells located close to stromal myofibroblasts, indicating that Wnt activity and cancer stemness may be regulated by extrinsic cues. In agreement with this notion, myofibroblast-secreted factors, specifically hepatocyte growth factor, activate β-catenin-dependent transcription and subsequently CSC clonogenicity. More significantly, myofibroblast-secreted factors also restore the CSC phenotype in more differentiated tumour cells both in vitro and in vivo. We therefore propose that stemness of colon cancer cells is in part orchestrated by the microenvironment and is a much more dynamic quality than previously expected that can be defined by high Wnt activity.

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: Wnt heterogeneity in primary tumours and in CSC culture.
Figure 2: Cells with high Wnt signalling show CSC properties in vitro.
Figure 3: Cells with high Wnt signalling show CSC properties in vivo.
Figure 4: Ex vivo tumorigenic assay and single-cell-derived tumours.
Figure 5: Myofibroblasts support stem-cell properties and regulate Wnt signalling.
Figure 6: Myofibroblasts restore Wnt activity and clonogenic potential in TOP–GFPlow cells.
Figure 7: Myofibroblasts restore tumorigenicity in TOP–GFPlow Wnt-active cells and co-localize with highly Wnt-active cells in vivo.
Figure 8: Schematic representation of the proposed model.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Fearon, E. R. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61, 759–767 (1990).

    Article  CAS  Google Scholar 

  2. Clevers, H. Wnt/β-catenin signaling in development and disease. Cell 127, 469–480 (2006).

    Article  CAS  Google Scholar 

  3. Bienz, M. & Clevers, H. Linking colorectal cancer to Wnt signaling. Cell 103, 311–320 (2000).

    Article  CAS  Google Scholar 

  4. Vogelstein, B. et al. Genetic alterations during colorectal-tumor development. N. Engl. J. Med. 319, 525–532 (1988).

    Article  CAS  Google Scholar 

  5. Tetsu, O. & McCormick, F. β-Catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398, 422–426 (1999).

    Article  CAS  Google Scholar 

  6. He, T. C. et al. Identification of c-MYC as a target of the APC pathway. Science 281, 1509–1512 (1998).

    Article  CAS  Google Scholar 

  7. Korinek, V. et al. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nature Genet. 19, 379–383 (1998).

    Article  CAS  Google Scholar 

  8. Groden, J. et al. Response of colon cancer cell lines to the introduction of APC, a colon-specific tumor suppressor gene. Cancer Res. 55, 1531–1539 (1995).

    CAS  PubMed  Google Scholar 

  9. van de Wetering, M. et al. The β-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell 111, 241–250 (2002).

    Article  CAS  Google Scholar 

  10. Brabletz, T. et al. Variable β-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl Acad. Sci. USA 98, 10356–10361 (2001).

    Article  CAS  Google Scholar 

  11. Fodde, R. & Brabletz, T. Wnt/β-catenin signaling in cancer stemness and malignant behavior. Curr. Opin. Cell Biol. 19, 150–158 (2007).

    Article  CAS  Google Scholar 

  12. Vermeulen, L., Sprick, M. R., Kemper, K., Stassi, G. & Medema, J. P. Cancer stem cells—old concepts, new insights. Cell Death. Differ. 15, 947–958 (2008).

    Article  CAS  Google Scholar 

  13. O'Brien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445, 106–110 (2007).

    Article  CAS  Google Scholar 

  14. Todaro, M. et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 1, 389–402 (2007).

    Article  CAS  Google Scholar 

  15. Dalerba, P. et al. Phenotypic characterization of human colorectal cancer stem cells. Proc. Natl Acad. Sci. USA 104, 10158–10163 (2007).

    Article  CAS  Google Scholar 

  16. Ricci-Vitiani, L. et al. Identification and expansion of human colon-cancer-initiating cells. Nature 445, 111–115 (2007).

    Article  CAS  Google Scholar 

  17. Vermeulen, L. et al. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc. Natl Acad. Sci. USA 105, 13427–13432 (2008).

    Article  CAS  Google Scholar 

  18. Reya, T. et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 423, 409–414 (2003).

    Article  CAS  Google Scholar 

  19. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  Google Scholar 

  20. van der Flier, L. G. et al. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell 136, 903–912 (2009).

    Article  CAS  Google Scholar 

  21. Yeung, T. M., Gandhi, S. C., Wilding, J. L., Muschel, R. & Bodmer, W. F. Cancer stem cells from colorectal cancer-derived cell lines. Proc. Natl Acad. Sci. USA 107, 3722–3727 (2010).

    Article  CAS  Google Scholar 

  22. Kai, K. et al. Maintenance of HCT116 colon cancer cell line conforms to a stochastic model but not a cancer stem cell model. Cancer Sci. 100, 2275–2282 (2009).

    Article  CAS  Google Scholar 

  23. Clarke, M. F. et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66, 9339–9344 (2006).

    Article  CAS  Google Scholar 

  24. Karnoub, A. E. et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449, 557–563 (2007).

    Article  CAS  Google Scholar 

  25. Sato, T. et al. Single Lgr5 stem cells build crypt–villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  Google Scholar 

  26. Yen, T. H. & Wright, N. A. The gastrointestinal tract stem cell niche. Stem Cell Rev. 2, 203–212 (2006).

    Article  CAS  Google Scholar 

  27. Boon, E. M., van der, N. R., van de, W. M., Clevers, H. & Pals, S. T. Wnt signaling regulates expression of the receptor tyrosine kinase met in colorectal cancer. Cancer Res. 62, 5126–5128 (2002).

    CAS  PubMed  Google Scholar 

  28. Rasola, A. et al. A positive feedback loop between hepatocyte growth factor receptor and beta-catenin sustains colorectal cancer cell invasive growth. Oncogene 26, 1078–1087 (2007).

    Article  CAS  Google Scholar 

  29. Brembeck, F. H. et al. Essential role of BCL9-2 in the switch between β-catenin's adhesive and transcriptional functions. Genes Dev. 18, 2225–2230 (2004).

    Article  CAS  Google Scholar 

  30. Di Renzo, M. F. et al. Overexpression and amplification of the Met/HGF receptor gene during the progression of colorectal cancer. Clin. Cancer Res. 1, 147–154 (1995).

    CAS  PubMed  Google Scholar 

  31. Ng, S. S. et al. Phosphatidylinositol 3-kinase signaling does not activate the Wnt cascade. J. Biol. Chem. 284, 35308–35313 (2009).

    Article  CAS  Google Scholar 

  32. Fang, D. et al. Phosphorylation of β-catenin by AKT promotes β-catenin transcriptional activity. J. Biol. Chem. 282, 11221–11229 (2007).

    Article  CAS  Google Scholar 

  33. Borovski, T. et al. Tumor microvasculature supports proliferation and expansion of glioma-propagating cells. Int. J. Cancer 125, 1222–1230 (2009).

    Article  CAS  Google Scholar 

  34. Calabrese, C. et al. A perivascular niche for brain tumor stem cells. Cancer Cell 11, 69–82 (2007).

    Article  CAS  Google Scholar 

  35. Shackleton, M., Quintana, E., Fearon, E. R. & Morrison, S. J. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell 138, 822–829 (2009).

    Article  CAS  Google Scholar 

  36. Kelly, P. N., Dakic, A., Adams, J. M., Nutt, S. L. & Strasser, A. Tumor growth need not be driven by rare cancer stem cells. Science 317, 337 (2007).

    Article  CAS  Google Scholar 

  37. Quintana, E. et al. Efficient tumour formation by single human melanoma cells. Nature 456, 593–598 (2008).

    Article  CAS  Google Scholar 

  38. Guan, Y., Gerhard, B. & Hogge, D. E. Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 101, 3142–3149 (2003).

    Article  CAS  Google Scholar 

  39. Guzman, M. L. et al. Preferential induction of apoptosis for primary human leukemic stem cells. Proc. Natl Acad. Sci. USA 99, 16220–16225 (2002).

    Article  CAS  Google Scholar 

  40. Bao, S. et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756–760 (2006).

    Article  CAS  Google Scholar 

  41. Brabletz, T., Jung, A., Spaderna, S., Hlubek, F. & Kirchner, T. Opinion: migrating cancer stem cells—an integrated concept of malignant tumour progression. Nature Rev. Cancer 5, 744–749 (2005).

    Article  CAS  Google Scholar 

  42. Kammula, U. S. et al. Molecular co-expression of the c-Met oncogene and hepatocyte growth factor in primary colon cancer predicts tumor stage and clinical outcome. Cancer Lett. 248, 219–228 (2007).

    Article  CAS  Google Scholar 

  43. Corso, S. et al. Silencing the MET oncogene leads to regression of experimental tumors and metastases. Oncogene 27, 684–693 (2008).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  45. Stoker, M. & Perryman, M. An epithelial scatter factor released by embryo fibroblasts. J. Cell Sci. 77, 209–223 (1985).

    CAS  PubMed  Google Scholar 

  46. Grotegut, S., von, S. D., Christofori, G. & Lehembre, F. Hepatocyte growth factor induces cell scattering through MAPK/Egr-1-mediated upregulation of Snail. EMBO J. 25, 3534–3545 (2006).

    Article  CAS  Google Scholar 

  47. Moustakas, A. & Heldin, C. H. Signaling networks guiding epithelial–mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 98, 1512–1520 (2007).

    Article  CAS  Google Scholar 

  48. Tuynman, J. B. et al. Cyclooxygenase-2 inhibition inhibits c-Met kinase activity and Wnt activity in colon cancer. Cancer Res. 68, 1213–1220 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors are indebted to the work performed by Willem Bemelman and Thomas van Gulik, surgeons at the AMC without whom the studies described here would not have been possible. In addition, we thank Riccardo Fodde for constructive discussions on CSCs and Wnt, and the animal care takers for their work for this project. Finally, we thank Berend Hooibrink and Toni van Capel for assistance with fluorescence-activated cell sorting experiments. This work was supported by a VICI grant from the Netherlands Organisation for Scientific Research and a Dutch Cancer Society (KWF Kankerbestrijding) grant (2009-4416) (to J.P.M.), an Academisch Medisch Centrum (AMC) fellowship (to L.V. and F.d.S.M.), the AMC Graduate School (to K.K.) and by the Associazione Italiana per la Ricerca sul Cancro (AIRC) (to G.S. and M.T.).

Author information

Authors and Affiliations

Authors

Contributions

L.V., F.d.S.M., M.v.d.H., K.C., J.H.d.J., T.B., J.B.T., H.R., M.R.S., K.K. designed and conducted experiments. M.T. and C.M. isolated and cultured CSC lines, D.J.R., G.S. and J.P.M. planned and supervised the experiments. L.V., F.d.S.M and J.P.M wrote the manuscript.

Corresponding author

Correspondence to Jan Paul Medema.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1173 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vermeulen, L., De Sousa E Melo, F., van der Heijden, M. et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol 12, 468–476 (2010). https://doi.org/10.1038/ncb2048

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2048

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer