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:

CKIα ablation highlights a critical role for p53 in invasiveness control

Abstract

The mature gut renews continuously and rapidly throughout adult life, often in a damage-inflicting micro-environment. The major driving force for self-renewal of the intestinal epithelium is the Wnt-mediated signalling pathway, and Wnt signalling is frequently hyperactivated in colorectal cancer1. Here we show that casein kinase Iα (CKIα), a component of the β-catenin-destruction complex1, is a critical regulator of the Wnt signalling pathway. Inducing the ablation of Csnk1a1 (the gene encoding CKIα) in the gut triggers massive Wnt activation, surprisingly without causing tumorigenesis. CKIα-deficient epithelium shows many of the features of human colorectal tumours in addition to Wnt activation, in particular the induction of the DNA damage response and cellular senescence, both of which are thought to provide a barrier against malignant transformation2. The epithelial DNA damage response in mice is accompanied by substantial activation of p53, suggesting that the p53 pathway may counteract the pro-tumorigenic effects of Wnt hyperactivation. Notably, the transition from benign adenomas to invasive colorectal cancer in humans is typically linked to p53 inactivation, underscoring the importance of p53 as a safeguard against malignant progression3; however, the mechanism of p53-mediated tumour suppression is unknown. We show that the maintenance of intestinal homeostasis in CKIα-deficient gut requires p53-mediated growth control, because the combined ablation of Csnk1a1 and either p53 or its target gene p21 (also known as Waf1, Cip1, Sdi1 and Cdkn1a) triggered high-grade dysplasia with extensive proliferation. Unexpectedly, these ablations also induced non-proliferating cells to invade the villous lamina propria rapidly, producing invasive carcinomas throughout the small bowel. Furthermore, in p53-deficient gut, loss of heterozygosity of the gene encoding CKIα caused a highly invasive carcinoma, indicating that CKIα functions as a tumour suppressor when p53 is inactivated. We identified a set of genes (the p53-suppressed invasiveness signature, PSIS) that is activated by the loss of both p53 and CKIα and which probably accounts for the brisk induction of invasiveness. PSIS transcription and tumour invasion were suppressed by p21, independently of cell cycle control. Restraining tissue invasion through suppressing PSIS expression is thus a novel tumour-suppressor function of wild-type p53.

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: Csnk1a1 ablation induces Wnt hyperinduction.
Figure 2: Persistent DDR and cellular senescence in Csnk1a1 Δgut mice.
Figure 3: Widespread invasive carcinomas in the Csnk1a1 Δgut p53 Δgut (double-knockout) bowel.
Figure 4: Invasiveness suppression by p53, through p21.

Similar content being viewed by others

References

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

    Article  CAS  Google Scholar 

  2. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Sansom, O. J. et al. Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration. Genes Dev. 18, 1385–1390 (2004)

    Article  CAS  Google Scholar 

  5. Collado, M. et al. Senescence in premalignant tumours. Nature 436, 642 (2005)

    Article  CAS  ADS  Google Scholar 

  6. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006)

    Article  CAS  ADS  Google Scholar 

  7. Junttila, M. R. & Evan, G. I. p53—a Jack of all trades but master of none. Nature Rev. Cancer 9, 821–829 (2009)

    Article  CAS  Google Scholar 

  8. Kamijo, T. et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19 ARF . Cell 91, 649–659 (1997)

    Article  CAS  Google Scholar 

  9. Damalas, A., Kahan, S., Shtutman, M., Ben-Ze'ev, A. & Oren, M. Deregulated β-catenin induces a p53- and ARF-dependent growth arrest and cooperates with Ras in transformation. EMBO J. 20, 4912–4922 (2001)

    Article  CAS  Google Scholar 

  10. Sherr, C. J. Divorcing ARF and p53: an unsettled case. Nature Rev. Cancer 6, 663–673 (2006)

    Article  CAS  Google Scholar 

  11. Valentin-Vega, Y. A., Box, N., Terzian, T. & Lozano, G. Mdm4 loss in the intestinal epithelium leads to compartmentalized cell death but no tissue abnormalities. Differentiation 77, 442–449 (2009)

    Article  CAS  Google Scholar 

  12. Wilson, J. W., Pritchard, D. M., Hickman, J. A. & Potten, C. S. Radiation-induced p53 and p21WAF-1/CIP1 expression in the murine intestinal epithelium: apoptosis and cell cycle arrest. Am. J. Pathol. 153, 899–909 (1998)

    Article  CAS  Google Scholar 

  13. Riley, T., Sontag, E., Chen, P. & Levine, A. Transcriptional control of human p53-regulated genes. Nature Rev. Mol. Cell Biol. 9, 402–412 (2008)

    Article  CAS  Google Scholar 

  14. Andreu, P. et al. Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine. Development 132, 1443–1451 (2005)

    Article  CAS  Google Scholar 

  15. Fodde, R. et al. A targeted chain-termination mutation in the mouse Apc gene results in multiple intestinal tumors. Proc. Natl Acad. Sci. USA 91, 8969–8973 (1994)

    Article  CAS  ADS  Google Scholar 

  16. Taketo, M. M. & Edelmann, W. Mouse models of colon cancer. Gastroenterology 136, 780–798 (2009)

    Article  CAS  Google Scholar 

  17. Donehower, L. A. e. t. a. l. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356, 215–221 (1992)

    Article  CAS  ADS  Google Scholar 

  18. Fazeli, A. et al. Effects of p53 mutations on apoptosis in mouse intestinal and human colonic adenomas. Proc. Natl Acad. Sci. USA 94, 10199–10204 (1997)

    Article  CAS  ADS  Google Scholar 

  19. Reed, K. R. et al. A limited role for p53 in modulating the immediate phenotype of Apc loss in the intestine. BMC Cancer 8, 162 (2008)

    Article  Google Scholar 

  20. Andreu, P. et al. Identification of the IFITM family as a new molecular marker in human colorectal tumors. Cancer Res. 66, 1949–1955 (2006)

    Article  CAS  Google Scholar 

  21. Hatano, H. et al. IFN-induced transmembrane protein 1 promotes invasion at early stage of head and neck cancer progression. Clin. Cancer Res. 14, 6097–6105 (2008)

    Article  CAS  Google Scholar 

  22. Petrova, T. V. et al. Transcription factor PROX1 induces colon cancer progression by promoting the transition from benign to highly dysplastic phenotype. Cancer Cell 13, 407–419 (2008)

    Article  CAS  Google Scholar 

  23. Cole, M. F., Johnstone, S. E., Newman, J. J., Kagey, M. H. & Young, R. A. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells. Genes Dev. 22, 746–755 (2008)

    Article  CAS  Google Scholar 

  24. Yin, J. et al. p53 point mutations in dysplastic and cancerous ulcerative colitis lesions. Gastroenterology 104, 1633–1639 (1993)

    Article  CAS  Google Scholar 

  25. Tabach, Y. et al. The promoters of human cell cycle genes integrate signals from two tumor suppressive pathways during cellular transformation. Mol. Syst. Biol. 1, 2005.0022 (2005)

    Article  Google Scholar 

  26. Abbas, T. & Dutta, A. p21 in cancer: intricate networks and multiple activities. Nature Rev. Cancer 9, 400–414 (2009)

    Article  CAS  Google Scholar 

  27. Wang, S. P. et al. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nature Cell Biol. 11, 694–704 (2009)

    Article  CAS  Google Scholar 

  28. Muller, P. A. et al. Mutant p53 drives invasion by promoting integrin recycling. Cell 139, 1327–1341 (2009)

    Article  Google Scholar 

  29. Bedenne, L. et al. Adenoma–carcinoma sequence or ‘de novo’ carcinogenesis? A study of adenomatous remnants in a population-based series of large bowel cancers. Cancer 69, 883–888 (1992)

    Article  CAS  Google Scholar 

  30. Coppe, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99–118 (2010)

    Article  CAS  Google Scholar 

  31. Lallemand, Y., Luria, V., Haffner-Krausz, R. & Lonai, P. Maternally expressed PGK-Cre transgene as a tool for early and uniform activation of the Cre site-specific recombinase. Transgenic Res. 7, 105–112 (1998)

    Article  CAS  Google Scholar 

  32. el Marjou, F. et al. Tissue-specific and inducible Cre-mediated recombination in the gut epithelium. Genesis 39, 186–193 (2004)

    Article  CAS  Google Scholar 

  33. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nature Genet. 29, 418–425 (2001)

    Article  CAS  Google Scholar 

  34. Brugarolas, J. et al. Radiation-induced cell cycle arrest compromised by p21 deficiency. Nature 377, 552–557 (1995)

    Article  CAS  ADS  Google Scholar 

  35. Greten, F. R. et al. IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 118, 285–296 (2004)

    Article  CAS  Google Scholar 

  36. Davalli, A. M. et al. A selective decrease in the β cell mass of human islets transplanted into diabetic nude mice. Transplantation 59, 817–820 (1995)

    Article  CAS  Google Scholar 

  37. Dimri, G. P. & Campisi, J. Molecular and cell biology of replicative senescence. Cold Spring Harb. Symp. Quant. Biol. 59, 67–73 (1994)

    Article  CAS  Google Scholar 

  38. Naldini, L., Blomer, U., Gage, F. H., Trono, D. & Verma, I. M. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc. Natl Acad. Sci. USA 93, 11382–11388 (1996)

    Article  CAS  ADS  Google Scholar 

  39. Naldini, L. et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 272, 263–267 (1996)

    Article  CAS  ADS  Google Scholar 

  40. Ko, C. H., Shen, S. C., Lee, T. J. & Chen, Y. C. Myricetin inhibits matrix metalloproteinase 2 protein expression and enzyme activity in colorectal carcinoma cells. Mol. Cancer Ther. 4, 281–290 (2005)

    CAS  PubMed  Google Scholar 

  41. Irizarry, R. A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003)

    Article  Google Scholar 

  42. Gotea, V. & Ovcharenko, I. DiRE: identifying distant regulatory elements of co-expressed genes. Nucleic Acids Res. 36, W133–W139 (2008)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Robine for the Vil1–Cre–ERT2 mice, O. Sansom and B. Romagnolo for intestinal sections of ApcΔgut mice, K. Rajewsky for the pGEM–loxP–Neo–loxP and pCA–NLS–Cre vectors; and E. Horwitz, M. Farago, D. Naor, N. Asherie and D. Knigin for providing expertise and reagents. We are grateful to A. Yaron for critical reading of the manuscript. This work was supported by the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (AMRF), the Israel Science Foundation, the RUBICON EC Network of Excellence, the Israel Cancer Research Fund and Deutsches Krebsforschungszentrum–Ministry of Science and Technology (DKFZ–MOST). Z.W. is supported by a Marie-Curie Intra-European Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Most experiments were performed by E.E., A.P. and R.E.G. Additional experimental work was carried out by Y.M., G.B., G.C., I.S.-A., I.B., R.H.-K. and Z.W. Experimental design and interpretation of data were conducted by all authors. S.J. supervised the gene targeting. The project was supervised by E.P. and Y.B.-N., and the paper was written by E.E., A.P., R.E.G., K.A., M.O., E.P. and Y.B.-N.

Corresponding authors

Correspondence to Eli Pikarsky or Yinon Ben-Neriah.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

The file contains Supplementary Figures 1-11 with legends, Supplementary Tables 1-3 and references for Supplementary Table 3. (PDF 6938 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Elyada, E., Pribluda, A., Goldstein, R. et al. CKIα ablation highlights a critical role for p53 in invasiveness control. Nature 470, 409–413 (2011). https://doi.org/10.1038/nature09673

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature09673

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: 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