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.

  • Review Article
  • Published:

Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration

Key Points

  • Intestinal stem cells are responsible for the remarkable ability of the intestinal epithelium to efficiently renew and repair itself throughout life.

  • The intestinal stem cells reside within specialized instructive niches at the crypt base. WNT is an essential niche factor supplied by both Paneth cells and non-epithelial cells.

  • The identification of new intestinal stem cell markers has greatly improved our understanding of stem cell biology during homeostasis and disease.

  • Intestinal stem cells can now be purified and used to grow new epithelia ex vivo for regenerative medicine applications.

  • 'Reserve' stem cells are quickly recruited to drive epithelial regeneration following loss of the regular stem cell pool due to injury.

Abstract

Small populations of adult stem cells are responsible for the remarkable ability of the epithelial lining of the intestine to be efficiently renewed and repaired throughout life. The recent discovery of specific markers for these stem cells, together with the development of new technologies to track endogenous stem cell activity in vivo and to exploit their ability to generate new epithelia ex vivo, has greatly improved our understanding of stem cell-driven homeostasis, regeneration and cancer in the intestine. These exciting new insights into the biology of intestinal stem cells have the potential to accelerate the development of stem cell-based therapies and ameliorate cancer treatments.

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: Epithelial self-renewal in the intestinal epithelium.
Figure 2: Intestinal stem cells and their defining niche.
Figure 3: Models of epithelial regeneration in the small intestine.

Similar content being viewed by others

References

  1. Leblond, C. P. & Walker, B. E. Renewal of cell populations. Physiol. Rev. 36, 255–276 (1956).

    Article  CAS  PubMed  Google Scholar 

  2. Barker, N., van de Wetering, M. & Clevers, H. The intestinal stem cell. Genes Dev. 22, 1856–1864 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Potten, C. S., Gandara, R., Mahida, Y. R., Loeffler, M. & Wright, N. A. The stem cells of small intestinal crypts: where are they? Cell Prolif. 42, 731–750 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Blanpain, C. & Simons, B. D. Unravelling stem cell dynamics by lineage tracing. Nature Rev. Mol. Cell Biol. 14, 489–502 (2013).

    Article  CAS  Google Scholar 

  5. 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  PubMed  Google Scholar 

  6. Barker, N., Bartfeld, S. & Clevers, H. Tissue-resident adult stem cell populations of rapidly self-renewing organs. Cell Stem Cell 7, 656–670 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Buczacki, S. J. et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 495, 65–69 (2013). Identified LRCs as Paneth cell progenitors capable of reverting to stem cells after damage.

    Article  CAS  PubMed  Google Scholar 

  8. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011). Demonstrated that the intestinal epithelium is able to survive ablation of the LGR5+ stem cell compartment, indicating the existence of reserve stem cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yilmaz, O. H. et al. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 486, 490–495 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gerbe, F., Legraverend, C. & Jay, P. The intestinal epithelium tuft cells: specification and function. Cell. Mol. Life Sci. 69, 2907–2917 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Watson, A. J. & Hughes, K. R. TNF-α-induced intestinal epithelial cell shedding: implications for intestinal barrier function. Ann. NY Acad. Sci. 1258, 1–8 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Bjerknes, M. & Cheng, H. Gastrointestinal stem cells. II. Intestinal stem cells. Am. J. Physiol. Gastrointest. Liver Physiol. 289, G381–G387 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Ireland, H., Houghton, C., Howard, L. & Winton, D. J. Cellular inheritance of a Cre-activated reporter gene to determine Paneth cell longevity in the murine small intestine. Dev. Dyn. 233, 1332–1336 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Grosse, A. S. et al. Cell dynamics in fetal intestinal epithelium: implications for intestinal growth and morphogenesis. Development 138, 4423–4432 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gregorieff, A. & Clevers, H. Wnt signaling in the intestinal epithelium: from endoderm to cancer. Genes Dev. 19, 877–890 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Madison, B. B. et al. Epithelial hedgehog signals pattern the intestinal crypt-villus axis. Development 132, 279–289 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. 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  PubMed  Google Scholar 

  18. Cheng, H. & Bjerknes, M. Whole population cell kinetics and postnatal development of the mouse intestinal epithelium. Anat. Rec. 211, 420–426 (1985).

    Article  CAS  PubMed  Google Scholar 

  19. Dehmer, J. J. et al. Expansion of intestinal epithelial stem cells during murine development. PLoS ONE 6, e27070 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mustata, R. C. et al. Identification of Lgr5-independent spheroid-generating progenitors of the mouse fetal intestinal epithelium. Cell Rep. 5, 421–432 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Itzkovitz, S., Blat, I. C., Jacks, T., Clevers, H. & van Oudenaarden, A. Optimality in the development of intestinal crypts. Cell 148, 608–619 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fordham, R. P. et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell http://dx.doi.org/10.1016/j.stem.2013.09.015 (2013).

  23. Ponder, B. A., Festing, M. F. & Wilkinson, M. M. An allelic difference determines reciprocal patterns of expression of binding sites for Dolichos biflorus lectin in inbred strains of mice. J. Embryol. Exp. Morphol. 87, 229–239 (1985).

    CAS  PubMed  Google Scholar 

  24. Schmidt, G. H., Garbutt, D. J., Wilkinson, M. M. & Ponder, B. A. Clonal analysis of intestinal crypt populations in mouse aggregation chimaeras. J. Embryol. Exp. Morphol. 85, 121–130 (1985).

    CAS  PubMed  Google Scholar 

  25. Ponder, B. A. et al. Derivation of mouse intestinal crypts from single progenitor cells. Nature 313, 689–691 (1985). Showed that adult crypts harbour clonal stem cell populations derived from a single progenitor cell.

    Article  CAS  PubMed  Google Scholar 

  26. Hermiston, M. L., Green, R. P. & Gordon, J. I. Chimeric-transgenic mice represent a powerful tool for studying how the proliferation and differentiation programs of intestinal epithelial cell lineages are regulated. Proc. Natl Acad. Sci. USA 90, 8866–8870 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bjerknes, M. & Cheng, H. Clonal analysis of mouse intestinal epithelial progenitors. Gastroenterology 116, 7–14 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Gutierrez-Gonzalez, L. et al. Analysis of the clonal architecture of the human small intestinal epithelium establishes a common stem cell for all lineages and reveals a mechanism for the fixation and spread of mutations. J. Pathol. 217, 489–496 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Greaves, L. C. et al. Mitochondrial DNA mutations are established in human colonic stem cells, and mutated clones expand by crypt fission. Proc. Natl Acad. Sci. USA 103, 714–719 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Taylor, R. W. et al. Mitochondrial DNA mutations in human colonic crypt stem cells. J. Clin. Invest. 112, 1351–1360 (2003). Established the clonality of human crypt stem cell populations by examining mitochondrial mutation patterns.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Novelli, M. R. et al. Polyclonal origin of colonic adenomas in an XO/XY patient with FAP. Science 272, 1187–1190 (1996). Used chimeric human patient cells to show that human colonic crypts are maintained by a clonal population of adult stem cells.

    Article  CAS  PubMed  Google Scholar 

  32. Hendry, J. H., Roberts, S. A. & Potten, C. S. The clonogen content of murine intestinal crypts: dependence on radiation dose used in its determination. Radiat. Res. 132, 115–119 (1992).

    Article  CAS  PubMed  Google Scholar 

  33. Potten, C. S. & Loeffler, M. A comprehensive model of the crypts of the small intestine of the mouse provides insight into the mechanisms of cell migration and the proliferation hierarchy. J. Theor. Biol. 127, 381–391 (1987).

    Article  CAS  PubMed  Google Scholar 

  34. Bjerknes, M. & Cheng, H. The stem-cell zone of the small intestinal epithelium. III. Evidence from columnar, enteroendocrine, and mucous cells in the adult mouse. Am. J. Anat. 160, 77–91 (1981).

    Article  CAS  PubMed  Google Scholar 

  35. Kozar, S. et al. Continuous clonal labeling reveals small numbers of functional stem cells in intestinal crypts and adenomas. Cell Stem Cell 13, 626–633 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Winton, D. J., Blount, M. A. & Ponder, B. A. A clonal marker induced by mutation in mouse intestinal epithelium. Nature 333, 463–466 (1988). Reported the existence of multipotent, self-renewing stem cells in the adult small intestine, as shown by the use of a random somatic mutation approach.

    Article  CAS  PubMed  Google Scholar 

  37. Cairnie, A. B., Lamerton, L. F. & Steel, G. G. Cell proliferation studies in the intestinal epithelium of the rat. I. Determination of the kinetic parameters. Exp. Cell Res. 39, 528–538 (1965).

    Article  CAS  PubMed  Google Scholar 

  38. Qiu, J. M., Roberts, S. A. & Potten, C. S. Cell migration in the small and large bowel shows a strong circadian rhythm. Epithelial Cell Biol. 3, 137–148 (1994). Together with reference 37, this paper examined cell migration rates in the intestinal epithelium and proposed position +4 as the origin of cell migration.

    CAS  PubMed  Google Scholar 

  39. Cairns, J. Mutation selection and natural history of cancer. Nature 255, 197–200 (1975).

    Article  CAS  PubMed  Google Scholar 

  40. Potten, C. S., Booth, C. & Pritchard, D. M. The intestinal epithelial stem cell: the mucosal governor. Int. J. Exp. Pathol. 78, 219–243 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cheng, H. & Leblond, C. P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Unitarian Theory of the origin of the four epithelial cell types. Am. J. Anat. 141, 537–561 (1974). Part of a seminal series of papers that described a probable common CBC cell origin for the four main epithelial cell lineages.

    Article  CAS  PubMed  Google Scholar 

  42. Cheng, H. & Leblond, C. P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. I. Columnar cell. Am. J. Anat. 141, 461–479 (1974).

    Article  CAS  PubMed  Google Scholar 

  43. Cheng, H. & Leblond, C. P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. III. Entero-endocrine cells. Am. J. Anat. 141, 503–519 (1974).

    Article  CAS  PubMed  Google Scholar 

  44. Cheng, H. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. IV. Paneth cells. Am. J. Anat. 141, 521–535 (1974).

    Article  CAS  PubMed  Google Scholar 

  45. Cheng, H. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. II. Mucous cells. Am. J. Anat. 141, 481–501 (1974).

    Article  CAS  PubMed  Google Scholar 

  46. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007). Reported the validation of LGR5 as a marker of CBC stem cells in the small intestine and colon using in vivo lineage tracing.

    Article  CAS  PubMed  Google Scholar 

  47. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009). Described a novel ex vivo culture system capable of sustaining the long-term growth of near-physiological intestinal epithelia from isolated LGR5+ stem cells.

    Article  CAS  PubMed  Google Scholar 

  48. Potten, C. S. Extreme sensitivity of some intestinal crypt cells to X and y-irradiation. Nature 269, 518–521 (1977). Analysed the sensitivity of epithelial populations at various locations within the crypt to irradiation, showing that +4 cells are highly radiosensitive.

    Article  CAS  PubMed  Google Scholar 

  49. Lansdorp, P. M. Immortal strands? Give me a break. Cell 129, 1244–1247 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Jung, P. et al. Isolation and in vitro expansion of human colonic stem cells. Nature Med. 17, 1225–1227 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Munoz, J. et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent '+4' cell markers. EMBO J. 31, 3079–3091 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Van der Flier, L. G. et al. The intestinal Wnt/TCF signature. Gastroenterology 132, 628–632 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. 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  PubMed  Google Scholar 

  54. de Lau, W. et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 476, 293–297 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Carmon, K. S., Gong, X., Lin, Q., Thomas, A. & Liu, Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling. Proc. Natl Acad. Sci. USA 108, 11452–11457 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Carmon, K. S., Lin, Q., Gong, X., Thomas, A. & Liu, Q. LGR5 interacts and co-internalizes with Wnt receptors to modulate Wnt/β-catenin signaling. Mol Cell. Biol. 32, 2054–2064 http://dx.doi.org/10.1128/MCB.00272-12 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Glinka, A. et al. LGR4 and LGR5 are R-spondin receptors mediating Wnt/β-catenin and Wnt/PCP signalling. EMBO Rep. 12, 1055–1061 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Koo, B.-K. et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 488, 665–669 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Hao, H. X. et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 485, 195–200 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Fafilek, B. et al. Troy, a tumor necrosis factor receptor family member, interacts with lgr5 to inhibit wnt signaling in intestinal stem cells. Gastroenterology 144, 381–391 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Chen, P. H., Chen, X., Lin, Z., Fang, D. & He, X. The structural basis of R-spondin recognition by LGR5 and RNF43. Genes Dev. 27, 1345–1350 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Peng, W. C. et al. Structure of stem cell growth factor R-spondin 1 in complex with the ectodomain of its receptor LGR5. Cell Rep. 3, 1885–1892 (2013).

    Article  CAS  PubMed  Google Scholar 

  63. Wang, D. et al. Structural basis for R-spondin recognition by LGR4/5/6 receptors. Genes Dev. 27, 1339–1344 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Lopez-Garcia, C., Klein, A. M., Simons, B. D. & Winton, D. J. Intestinal stem cell replacement follows a pattern of neutral drift. Science 330, 822–825 (2010). References 64 and 65 report the use of multicolor tracing and computer modelling to examine the population dynamics of Lgr5+ stem cells in the small intestine

    Article  CAS  PubMed  Google Scholar 

  66. Quyn, A. J. et al. Spindle orientation bias in gut epithelial stem cell compartments is lost in precancerous tissue. Cell Stem Cell 6, 175–181 (2010).

    Article  CAS  PubMed  Google Scholar 

  67. Bellis, J. et al. The tumor suppressor Apc controls planar cell polarities central to gut homeostasis. J. Cell Biol. 198, 331–341 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Escobar, M. et al. Intestinal epithelial stem cells do not protect their genome by asymmetric chromosome segregation. Nature Commun. 2, 258 (2011).

    Article  CAS  Google Scholar 

  69. Schepers, A. G., Vries, R., van den Born, M., van de Wetering, M. & Clevers, H. Lgr5 intestinal stem cells have high telomerase activity and randomly segregate their chromosomes. EMBO J. 30, 1104–1109 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Steinhauser, M. L. et al. Multi-isotope imaging mass spectrometry quantifies stem cell division and metabolism. Nature 481, 516–519 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Buske, P. et al. On the biomechanics of stem cell niche formation in the gut — modelling growing organoids. FEBS J. 279, 3475–3487 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Buske, P. et al. A comprehensive model of the spatio-temporal stem cell and tissue organisation in the intestinal crypt. PLoS Comput. Biol. 7, e1001045 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kaaij, L. T. et al. DNA methylation dynamics during intestinal stem cell differentiation reveals enhancers driving gene expression in the villus. Genome Biol. 14, R50 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sakamori, R. et al. Cdc42 and Rab8a are critical for intestinal stem cell division, survival, and differentiation in mice. J. Clin. Invest. 122, 1052–1065 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Heijmans, J. et al. ER stress causes rapid loss of intestinal epithelial stemness through activation of the unfolded protein response. Cell Rep. 3, 1128–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  76. Sangiorgi, E. & Capecchi, M. R. Bmi1 is expressed in vivo in intestinal stem cells. Nature Genet. 40, 915–920 (2008). Reported the first validation of a +4 marker by in vivo lineage tracing.

    Article  CAS  PubMed  Google Scholar 

  77. Yan, K. S. et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl Acad. Sci. USA 109, 466–471 (2012).

    Article  PubMed  Google Scholar 

  78. Itzkovitz, S. et al. Single-molecule transcript counting of stem-cell markers in the mouse intestine. Nature Cell Biol. 14, 106–114 (2012).

    Article  CAS  Google Scholar 

  79. Montgomery, R. K. et al. Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc. Natl Acad. Sci. USA 108, 179–184 (2011).

    Article  PubMed  Google Scholar 

  80. Powell, A. E. et al. The pan-ErbB negative regulator Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 149, 146–158 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Takeda, N. et al. Interconversion between intestinal stem cell populations in distinct niches. Science 334, 1420–1424 (2011). Showed that +4 stem cells can interconvert with LGR5+ stem cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Jensen, K. B. et al. Lrig1 expression defines a distinct multipotent stem cell population in mammalian epidermis. Cell Stem Cell 4, 427–439 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Wong, V. W. et al. Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nature Cell Biol. 14, 401–408 (2012).

    Article  CAS  PubMed  Google Scholar 

  84. Breault, D. T. et al. Generation of mTert–GFP mice as a model to identify and study tissue progenitor cells. Proc. Natl Acad. Sci. USA 105, 10420–10425 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Roberts, S. A., Hendry, J. H. & Potten, C. S. Deduction of the clonogen content of intestinal crypts: a direct comparison of two-dose and multiple-dose methodologies. Radiat. Res. 141, 303–308 (1995).

    Article  CAS  PubMed  Google Scholar 

  86. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418 (2011).

    Article  CAS  PubMed  Google Scholar 

  87. Roth, S. et al. Paneth cells in intestinal homeostasis and tissue injury. PLoS ONE 7, e38965 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. van Es, J. H. et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nature Cell Biol. 14, 1099–1104 (2012). Showed that DLL1+ secretory progenitors are the reserve stem cell population activated in response to injury.

    Article  CAS  PubMed  Google Scholar 

  89. Kemper, K. et al. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells 30, 2378–2386 (2012).

    Article  CAS  PubMed  Google Scholar 

  90. Garabedian, E. M., Roberts, L. J., McNevin, M. S. & Gordon, J. I. Examining the role of Paneth cells in the small intestine by lineage ablation in transgenic mice. J. Biol. Chem. 272, 23729–23740 (1997).

    Article  CAS  PubMed  Google Scholar 

  91. Durand, A. et al. Functional intestinal stem cells after Paneth cell ablation induced by the loss of transcription factor Math1 (Atoh1). Proc. Natl Acad. Sci. USA 109, 8965–8970 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kim, T. H., Escudero, S. & Shivdasani, R. A. Intact function of Lgr5 receptor-expressing intestinal stem cells in the absence of Paneth cells. Proc. Natl Acad. Sci. USA 109, 3932–3937 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Farin, H. F., Van Es, J. H. & Clevers, H. Redundant sources of Wnt regulate intestinal stem cells and promote formation of Paneth cells. Gastroenterology 143, 1518–1529.e7 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Rothenberg, M. E. et al. Identification of a cKit+ colonic crypt base secretory cell that supports Lgr5+ stem cells in mice. Gastroenterology 142, 1195–1205.e6 (2012).

    Article  CAS  PubMed  Google Scholar 

  95. Blache, P. et al. SOX9 is an intestine crypt transcription factor, is regulated by the Wnt pathway, and represses the CDX2 and MUC2 genes. J. Cell Biol. 166, 37–47 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Bastide, P. et al. Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. J. Cell Biol. 178, 635–648 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Formeister, E. J. et al. Distinct SOX9 levels differentially mark stem/progenitor populations and enteroendocrine cells of the small intestine epithelium. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G1108–G1118 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Van Landeghem, L. et al. Activation of two distinct Sox9-EGFP-expressing intestinal stem cell populations during crypt regeneration after irradiation. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G1111–G1132 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Furuyama, K. et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nature Genet. 43, 34–41 (2011).

    Article  CAS  PubMed  Google Scholar 

  100. Gracz, A. D., Ramalingam, S. & Magness, S. T. Sox9 expression marks a subset of CD24-expressing small intestine epithelial stem cells that form organoids in vitro. Am. J. Physiol. Gastrointest Liver Physiol. 298, G590–G600 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Potten, C. S. et al. Identification of a putative intestinal stem cell and early lineage marker; musashi-1. Differentiation 71, 28–41 (2003).

    Article  CAS  PubMed  Google Scholar 

  102. Kayahara, T. et al. Candidate markers for stem and early progenitor cells, Musashi-1 and Hes1, are expressed in crypt base columnar cells of mouse small intestine. FEBS Lett. 535, 131–135 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Cambuli, F. M., Rezza, A., Nadjar, J. & Plateroti, M. Musashi1-eGFP mice, a new tool for differential isolation of the intestinal stem cell populations. Stem Cells 31, 2273–2278 (2013).

    Article  CAS  Google Scholar 

  104. Takeda, H., Koso, H., Tessarollo, L., Copeland, N. G. & Jenkins, N. A. Musashi1-CreERT2: a new cre line for conditional mutagenesis in neural stem cells. Genesis 51, 128–134 (2013).

    Article  CAS  PubMed  Google Scholar 

  105. 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  PubMed  Google Scholar 

  106. Shmelkov, S. V. et al. CD133 expression is not restricted to stem cells, and both CD133+ and CD133 metastatic colon cancer cells initiate tumors. J. Clin. Invest. 118, 2111–2120 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Yin, A. H. et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 90, 5002–5012 (1997).

    CAS  PubMed  Google Scholar 

  108. Zhu, L. et al. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature 457, 603–607 (2009).

    Article  CAS  PubMed  Google Scholar 

  109. Snippert, H. J. et al. Prominin-1/CD133 marks stem cells and early progenitors in mouse small intestine. Gastroenterology 136, 2187–2194 (2009).

    Article  CAS  PubMed  Google Scholar 

  110. Barker, N., van Oudenaarden, A. & Clevers, H. Identifying the stem cell of the intestinal crypt: strategies and pitfalls. Cell Stem Cell 11, 452–460 (2012).

    Article  CAS  PubMed  Google Scholar 

  111. He, X. C. et al. PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nature Genet. 39, 189–198 (2007).

    Article  CAS  PubMed  Google Scholar 

  112. He, X. C. et al. BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-β-catenin signaling. Nature Genet. 36, 1117–1121 (2004).

    Article  CAS  PubMed  Google Scholar 

  113. Bjerknes, M. & Cheng, H. Re-examination of P-PTEN staining patterns in the intestinal crypt. Nature Genet. 37, 1016–1017; author reply 1017–1018 (2005).

    Article  CAS  PubMed  Google Scholar 

  114. Demidov, O. N. et al. Wip1 phosphatase regulates p53-dependent apoptosis of stem cells and tumorigenesis in the mouse intestine. Cell Stem Cell 1, 180–190 (2007).

    Article  CAS  PubMed  Google Scholar 

  115. Giannakis, M. et al. Molecular properties of adult mouse gastric and intestinal epithelial progenitors in their niches. J. Biol. Chem. 281, 11292–11300 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. May, R. et al. Identification of a novel putative gastrointestinal stem cell and adenoma stem cell marker, doublecortin and CaM kinase-like-1, following radiation injury and in adenomatous polyposis coli/multiple intestinal neoplasia mice. Stem Cells 26, 630–637 (2008).

    Article  PubMed  Google Scholar 

  117. May, R. et al. Doublecortin and CaM kinase-like-1 and leucine-rich-repeat-containing G-protein-coupled receptor mark quiescent and cycling intestinal stem cells, respectively. Stem Cells 27, 2571–2579 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Gerbe, F., Brulin, B., Makrini, L., Legraverend, C. & Jay, P. DCAMKL-1 expression identifies Tuft cells rather than stem cells in the adult mouse intestinal epithelium. Gastroenterology 137, 2179–2180; author reply 2180–2181 (2009).

    Article  CAS  PubMed  Google Scholar 

  119. Nakanishi, Y. et al. Dclk1 distinguishes between tumor and normal stem cells in the intestine. Nature Genet. 45, 98–103 (2013).

    Article  CAS  PubMed  Google Scholar 

  120. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Sala, F. G. et al. A multicellular approach forms a significant amount of tissue-engineered small intestine in the mouse. Tissue Eng. Part A 17, 1841–1850 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Yui, S. et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nature Med. 18, 618–623 (2012). First demonstration that intestinal epithelia cultured ex vivo can be used to repair damaged colonic epithelia in live mice.

    Article  CAS  PubMed  Google Scholar 

  123. Magney, J. E., Erlandsen, S. L., Bjerknes, M. L. & Cheng, H. Morphology of the basal surface and evidence for paracrinelike cells. 177, 43–53 (1986).

Download references

Acknowledgements

The author thanks the members of the Barker group for critical input. N.B. is supported by the Agency for Science, Technology and Research (A*STAR).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nick Barker.

Ethics declarations

Competing interests

The author declares no competing financial interests.

Supplementary information

Supplementary information S1 (box)

Stem cells in intestinal cancer (PDF 128 kb)

PowerPoint slides

Glossary

Niches

Specialized instructive microenvironments in which stem cells reside. Niches provide all of the factors necessary to regulate stem cell survival and function.

Intestinal villus

A finger-like structure covered in simple columnar epithelium that projects into the intestinal lumen to maximize the surface area for digestion and absorption.

Duodenum

The proximal third of the small intestine, closest to the stomach. Characterized by the presence of long villi to ensure maximal nutrient digestion and absorption.

Intestinal crypt

Tubular invaginations of the epithelium harbouring the stem cells and their proliferating progeny; responsible for driving epithelial homeostasis and regeneration.

Chimeric mice

Mice that are comprised of two or more populations of genetically distinct cells.

Lgr5–EGFP–ires–CreERT2/R26R–lacZ mouse model

Generated by crossing Lgr5–EGFP–ires–CreERT2 (which encodes a tamoxifen-activatable Cre enzyme that catalyzes recombination across DNA sequences called loxP sites) and R26R (Rosa26 reporter construct)–lacZ mouse strains. Facilitates the conditional activation of the lacZ reporter gene in LGR5-expressing cells in living tissues to evaluate their stem cell identity via lineage tracing.

Bmi1–ires–CreERT2/R26R–lacZ mouse model

Generated by crossing Bmi1-ires-CreERT2 and R26R-lacZ mouse strains. Facilitates the conditional activation of the lacZ reporter gene in BMI1-expressing cells in living tissues to evaluate their stem cell identity via lineage tracing.

Ires

(Internal ribosome entry site). A ribosome-binding site present in the middle of an mRNA that facilitates internal translation initiation to generate an independent protein.

Pulse–chase

A method for detecting quiescent label-retaining cells (LRCs) in vivo. A nucleotide analogue (the label) is administered to the mouse for a short period (the pulse); this is followed by an extended period when no further nucleotide analogue is given (the chase). Actively dividing cells rapidly dilute out the label during DNA replication, and labelled cells are lost within 3–4 rounds of cell division. Non-dividing (quiescent) cells do not dilute out the label and thus remain detectable as LRCs.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Barker, N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol 15, 19–33 (2014). https://doi.org/10.1038/nrm3721

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm3721

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