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.

  • Technical Report
  • Published:

Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture

Subjects

Abstract

The application of primary organoid cultures containing epithelial and mesenchymal elements to cancer modeling holds promise for combining the accurate multilineage differentiation and physiology of in vivo systems with the facile in vitro manipulation of transformed cell lines. Here we used a single air-liquid interface culture method without modification to engineer oncogenic mutations into primary epithelial and mesenchymal organoids from mouse colon, stomach and pancreas. Pancreatic and gastric organoids exhibited dysplasia as a result of expression of Kras carrying the G12D mutation (KrasG12D), p53 loss or both and readily generated adenocarcinoma after in vivo transplantation. In contrast, primary colon organoids required combinatorial Apc, p53, KrasG12D and Smad4 mutations for progressive transformation to invasive adenocarcinoma-like histology in vitro and tumorigenicity in vivo, recapitulating multi-hit models of colorectal cancer (CRC), as compared to the more promiscuous transformation of small intestinal organoids. Colon organoid culture functionally validated the microRNA miR-483 as a dominant driver oncogene at the IGF2 (insulin-like growth factor-2) 11p15.5 CRC amplicon, inducing dysplasia in vitro and tumorigenicity in vivo. These studies demonstrate the general utility of a highly tractable primary organoid system for cancer modeling and driver oncogene validation in diverse gastrointestinal tissues.

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: In vitro oncogenic transformation of primary pancreatic organoids and in vivo tumorigenesis.
Figure 2: In vitro oncogenic transformation of primary gastric organoids and in vivo tumorigenesis.
Figure 3: Systematic evaluation of oncogene modules of increasing complexity in primary colon organoids.
Figure 4: Serial passage and in vivo transplantation of colon four-gene AKPS organoids.
Figure 5: Comparative functional validation of miR-483 and Igf2 transforming activity in primary Apc-null colon organoid culture.

Similar content being viewed by others

References

  1. Hynds, R.E. & Giangreco, A. Concise review: the relevance of human stem cell–derived organoid models for epithelial translational medicine. Stem Cells 31, 417–422 (2013).

    Article  CAS  Google Scholar 

  2. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science 340, 1190–1194 (2013).

    Article  CAS  Google Scholar 

  3. Stelzner, M. et al. A nomenclature for intestinal in vitro cultures. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G1359–G1363 (2012).

    Article  CAS  Google Scholar 

  4. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  Google Scholar 

  5. Pylayeva-Gupta, Y., Lee, K.E. & Bar-Sagi, D. Microdissection and culture of murine pancreatic ductal epithelial cells. Methods Mol. Biol. 980, 267–279 (2013).

    Article  CAS  Google Scholar 

  6. Jin, L. et al. Colony-forming cells in the adult mouse pancreas are expandable in Matrigel and form endocrine/acinar colonies in laminin hydrogel. Proc. Natl. Acad. Sci. USA 110, 3907–3912 (2013).

    Article  CAS  Google Scholar 

  7. Smukler, S.R. et al. The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell 8, 281–293 (2011).

    Article  CAS  Google Scholar 

  8. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    Article  CAS  Google Scholar 

  9. Ootani, A. et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat. Med. 15, 701–706 (2009).

    Article  CAS  Google Scholar 

  10. Katano, T. et al. Establishment of a long-term three-dimensional primary culture of mouse glandular stomach epithelial cells within the stem cell niche. Biochem. Biophys. Res. Commun. 432, 558–563 (2013).

    Article  CAS  Google Scholar 

  11. Ghajar, C.M. & Bissell, M.J. Tumor engineering: the other face of tissue engineering. Tissue Eng. Part A 16, 2153–2156 (2010).

    Article  Google Scholar 

  12. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  13. Wescott, M.P. et al. Pancreatic ductal morphogenesis and the Pdx1 homeodomain transcription factor. Mol. Biol. Cell 20, 4838–4844 (2009).

    Article  CAS  Google Scholar 

  14. Rovira, M. et al. Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc. Natl. Acad. Sci. USA 107, 75–80 (2010).

    Article  CAS  Google Scholar 

  15. Hingorani, S.R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  17. Haigis, K.M. et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat. Genet. 40, 600–608 (2008).

    Article  CAS  Google Scholar 

  18. Sansom, O.J. et al. Loss of Apc allows phenotypic manifestation of the transforming properties of an endogenous K-ras oncogene in vivo. Proc. Natl. Acad. Sci. USA 103, 14122–14127 (2006).

    Article  CAS  Google Scholar 

  19. 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 

  20. Shibata, H. et al. Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene. Science 278, 120–123 (1997).

    Article  CAS  Google Scholar 

  21. Hung, K.E. et al. Development of a mouse model for sporadic and metastatic colon tumors and its use in assessing drug treatment. Proc. Natl. Acad. Sci. USA 107, 1565–1570 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  23. Tanaka, T., Soriano, M.A. & Grusby, M.J. SLIM is a nuclear ubiquitin E3 ligase that negatively regulates STAT signaling. Immunity 22, 729–736 (2005).

    Article  CAS  Google Scholar 

  24. Attali, M. et al. Control of beta-cell differentiation by the pancreatic mesenchyme. Diabetes 56, 1248–1258 (2007).

    Article  CAS  Google Scholar 

  25. Duvillié, B., Heinis, M. & Stetsyuk, V. In vivo and in vitro techniques to study pancreas development and islet cell function. Endocr. Dev. 12, 46–54 (2007).

    Article  Google Scholar 

  26. Lee, K.E. & Bar-Sagi, D. Oncogenic KRas suppresses inflammation-associated senescence of pancreatic ductal cells. Cancer Cell 18, 448–458 (2010).

    Article  CAS  Google Scholar 

  27. Onuma, K. et al. Genetic reconstitution of tumorigenesis in primary intestinal cells. Proc. Natl. Acad. Sci. USA 110, 11127–11132 (2013).

    Article  CAS  Google Scholar 

  28. Leung, C.T. & Brugge, J.S. Outgrowth of single oncogene-expressing cells from suppressive epithelial environments. Nature 482, 410–413 (2012).

    Article  CAS  Google Scholar 

  29. Kim, J. et al. An iPSC line from human pancreatic ductal adenocarcinoma undergoes early to invasive stages of pancreatic cancer progression. Cell Reports 3, 2088–2099 (2013).

    Article  CAS  Google Scholar 

  30. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  Google Scholar 

  31. March, H.N. et al. Insertional mutagenesis identifies multiple networks of cooperating genes driving intestinal tumorigenesis. Nat. Genet. 43, 1202–1209 (2011).

    Article  CAS  Google Scholar 

  32. Starr, T.K. et al. A transposon-based genetic screen in mice identifies genes altered in colorectal cancer. Science 323, 1747–1750 (2009).

    Article  CAS  Google Scholar 

  33. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  Google Scholar 

  34. Firestein, R. et al. CDK8 is a colorectal cancer oncogene that regulates β-catenin activity. Nature 455, 547–551 (2008).

    Article  CAS  Google Scholar 

  35. Veronese, A. et al. Oncogenic role of miR-483–3p at the IGF2/483 locus. Cancer Res. 70, 3140–3149 (2010).

    Article  CAS  Google Scholar 

  36. Harper, J. et al. Soluble IGF2 receptor rescues ApcMin/+ intestinal adenoma progression induced by Igf2 loss of imprinting. Cancer Res. 66, 1940–1948 (2006).

    Article  CAS  Google Scholar 

  37. Timp, W., Levchenko, A. & Feinberg, A.P. A new link between epigenetic progenitor lesions in cancer and the dynamics of signal transduction. Cell Cycle 8, 383–390 (2009).

    Article  CAS  Google Scholar 

  38. Hassan, A.B. & Howell, J.A. Insulin-like growth factor II supply modifies growth of intestinal adenoma in ApcMin/+ mice. Cancer Res. 60, 1070–1076 (2000).

    CAS  PubMed  Google Scholar 

  39. Qu, Z. et al. DNA methylation–dependent repression of PDZ-LIM domain–containing protein 2 in colon cancer and its role as a potential therapeutic target. Cancer Res. 70, 1766–1772 (2010).

    Article  CAS  Google Scholar 

  40. Spence, J.R. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109 (2011).

    Article  Google Scholar 

  41. Sasai, K. et al. Oncogene-mediated human lung epithelial cell transformation produces adenocarcinoma phenotypes in vivo. Cancer Res. 71, 2541–2549 (2011).

    Article  CAS  Google Scholar 

  42. Dickins, R.A. et al. Probing tumor phenotypes using stable and regulated synthetic microRNA precursors. Nat. Genet. 37, 1289–1295 (2005).

    Article  CAS  Google Scholar 

  43. Griffiths-Jones, S., Saini, H.K., van Dongen, S. & Enright, A.J. miRBase: tools for microRNA genomics. Nucleic Acids Res. 36, D154–D158 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to members of the Kuo laboratory, M. Amieva, R. Honaker, T. Jacks and C. Chartier for helpful discussion, P. Chu and S. Michie for assistance with histology, S. Lowe (Memorial Sloan Kettering Cancer Center) for LMP-based retroviruses, B. Williams (Van Andel Research Institute) for mouse strains, R. Mulligan and J.-S. Lee (Harvard University) and J. Chen (Stanford University) for lentiviral and retroviral reagents. X.L. was supported by the Dean's Fellowship at Stanford University, A.O. was supported by a Physician-Scientist Fellowship from the California Institute for Regenerative Medicine, J.T.N. was supported by a Postdoctoral Fellowship (124574-PF-13-296-01-TBG) from the American Cancer Society, C.W.-M.C. was supported by the Croucher Foundation, and T.Y. was supported by the Royal College of Surgeons. L.N. was supported by an American Society for Clinical Oncology Young Investigator Award and US National Institutes of Health grant K08CA166512. C.-Z.C. was supported by grants from the US National Institutes of Health (1R01AI073724 and Pioneer Award DP1CA174421) and the W.M. Keck Foundation. We thank the generous support from the Innovate Foundation and the Krishnan-Shah Family Foundation to C.J.K. This work was also supported by US National Institutes of Health grants K08DK078033 and U01CA084301 to K.E.H.; US National Cancer Institute Integrative Cancer Biology Program grant U01CA151920, US National Cancer Institute Cancer Target Discovery and Development grant 1U01CA168424 and a Fidelity Foundation grant to C.J.K. and H.P.J.; and the US National Institutes of Health Transformative R01DK085720 and US National Institute of Diabetes and Digestive and Kidney Diseases Intestinal Stem Cell Consortium grant U01DK085527 to C.J.K.

Author information

Authors and Affiliations

Authors

Contributions

X.L., L.N., A.O., D.C.C., M.A.C., P.G.R. and J.T.N. designed and executed organoid experiments. R.K.P. performed blinded histologic interpretation. O.G. and X.G. performed bioinformatic analyses. C.W.-M.C., T.Y., J.Y. and J.W. performed organoid experiments and produced viruses. S.R., L.D.A. and K.E.H. provided reagents. K.E.H., C.-Z.C., S.K.P., H.P.J. and C.J.K. designed experiments. X.L., L.N., D.C.C. and C.J.K. wrote the manuscript.

Corresponding author

Correspondence to Calvin J Kuo.

Ethics declarations

Competing interests

C.J.K., L.N. and A.O. have filed patents with the United States Patent and Trademark Office to cover the use of this organoid technique for cancer modeling.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Discussion (PDF 13480 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, X., Nadauld, L., Ootani, A. et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med 20, 769–777 (2014). https://doi.org/10.1038/nm.3585

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3585

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