Intestinal stem cell function in Drosophila and mice

https://doi.org/10.1016/j.gde.2012.04.002Get rights and content

Epithelial cells of the digestive tracts of most animals are short-lived, and are constantly replenished by the progeny of long-lived, resident intestinal stem cells. Proper regulation of intestinal stem cell maintenance, proliferation and differentiation is critical for maintaining gut homeostasis. Here we review recent genetic studies of stem cell-mediated homeostatic growth in the Drosophila midgut and the mouse small intestine, highlighting similarities and differences in the mechanisms that control stem cell proliferation and differentiation.

Introduction

The endodermal portion of the insect intestine, termed the midgut, and its mammalian counterpart, comprising the stomach, small intestine and colon, serve as the animal's principal organs for digestion and nutrient absorption. In the mammalian small intestine, absorptive enterocytes and secretory goblet and enteroendocrine cells reside in finger-like protrusions known as villi. These cells are short-lived, being constantly shed from the villi and replaced by new cells generated in neighboring invaginations called the Crypts of Lieberkühn. The intestinal epithelium is perhaps the most rapidly turned over tissue in mammals, with enterocyte lifespans averaging a week or less. Intestinal stem cells (ISCs) reside at the basal ends of the crypts, intermingled with long-lived Paneth cells of the secretory lineage (Figure 1a). ISCs proliferate to self-renew and also give rise to transient progeny that amplify through further divisions. As cells exit the crypts and move apically, they differentiate into either absorptive enterocytes or one of three types of secretory cells: Paneth, enteroendocrine, or goblet. The mammalian colon is similarly maintained by ISCs located in crypts, but villi are absent and replaced by a smooth epithelium. In addition to these endodermal cells produced by ISCs, the mammalian intestine has stromal cells of several types – mesenchymal fibroblasts, immune cells and others – and is surrounded by mesodermally derived visceral muscle.

The endodermal portion of the Drosophila intestine, termed the midgut, undergoes similar dynamic cell turnover, also mediated by long-lived intestinal stem cells [1•, 2•]. The fly midgut however lacks crypts and villi, instead comprising a cellular monolayer ensheathed by two orthogonal layers of visceral muscle. Intestinal stem cells reside at the basal side of this epithelium, sandwiched between enterocytes and basement membrane produced partly by visceral muscle (Figure 1b). They divide to self-renew and to give rise to committed progenitors (called enteroblasts), which directly differentiate, without cell division, into two functional cell lineages similar to those found in vertebrates: absorptive enterocytes and enteroendocrine cells. Differentiating enterocytes endoreplicate their genome 2–3 times to increase their size and develop a brush border similar to that in mammals. Drosophila lacks the Paneth, Goblet, Stromal, and Dentritic cells found in mammals, but some of their functions – such as immunity and barrier production – are fulfilled by enterocytes. Instead of the thick mucosa produced by mammalian goblet cells, insect intestines have a tough but relatively thin (∼200 μm) membrane called the peritrophic matrix. This matrix is comprised of the exoskeletal protein chitin and glycoproteins including mucins related to those found in vertebrate mucosa, and it provides an essential barrier against infection by enteric pathogens [3].

Section snippets

Intestinal stem cell niches

The location and number of intestinal stem cells in the crypts of the mammalian intestine have long been debated. Studies using improved stem cell markers and elegant cell lineage tracing techniques, however, suggest that there may be two inter-convertible stem cell types present in the crypts [4••, 5••]. One cell type, located at the ‘+4’ position in the crypt (see Figure 1a), is marked by Bmi, Tert and Hopx expression. These slow-cycling, label-retaining cells can produce entire intestinal

Stem cell proliferation, differentiation, and the regenerative response

Genetic analyses identified canonical Wnt signaling as a primary force in maintaining tissue homeostasis in the murine intestine. Mice lacking the Tcf4 transcription factor or β-catenin, positive effectors in Wnt signaling, have reduced proliferation in the intestinal epithelium that depletes transient amplifying cells, and consequent loss of crypts and villi [18, 19, 20]. Conversely, activating Wnt signaling drives hyperproliferation in the crypts, as do loss-of-function mutations in APC (

Symmetric vs. asymmetric stem cell division

To maintain tissue homeostasis, stem cells must balance self-renewal with differentiation. Early models proposed that stem cells are immortal and always divide asymmetrically, replacing themselves at each division, but quantitative lineage analysis in the murine intestine has ruled out this classical model for tissue maintenance [54]. Rather, lineage tracing showed that fast-cycling intestinal stem cells divide symmetrically and are lost stochastically, mostly to differentiation. At the

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

B.A.E. was supported by NIH R01 GM51186, the ERC, and the DKFZ. H.J. was supported by startup funds from University of Texas, Southwestern Medical Center. We thank Jeffery Rosa and Parthive Patel for comments on the manuscript, and Hanna Reuter and Julien Elric for help with figures.

References (57)

  • T. Kuraishi et al.

    Genetic evidence for a protective role of the peritrophic matrix against intestinal bacterial infection in drosophila melanogaster

    Proc Natl Acad Sci USA

    (2011)
  • N. Takeda et al.

    Interconversion between intestinal stem cell populations in distinct niches

    Science (New York, NY)

    (2011)
  • H. Tian et al.

    A reserve stem cell population in small intestine renders lgr5-positive cells dispensable

    Nature

    (2011)
  • R.K. Montgomery et al.

    Mouse telomerase reverse transcriptase (mtert) expression marks slowly cycling intestinal stem cells

    Proc Natl Acad Sci USA

    (2011)
  • E. Sangiorgi et al.

    Bmi1 is expressed in vivo in intestinal stem cells

    Nat Genet

    (2008)
  • N. Barker et al.

    Identification of stem cells in small intestine and colon by marker gene lgr5

    Nature

    (2007)
  • K. Furuyama et al.

    Continuous cell supply from a sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine

    Nat Genet

    (2011)
  • L. Zhu et al.

    Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation

    Nature

    (2009)
  • T. Sato et al.

    Paneth cells constitute the niche for lgr5 stem cells in intestinal crypts

    Nature

    (2011)
  • T. Sato et al.

    Single lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche

    Nature

    (2009)
  • L.E. O’Brien et al.

    Altered modes of stem cell division drive adaptive intestinal growth

    Cell

    (2011)
  • G. Lin et al.

    Paracrine wingless signalling controls self-renewal of drosophila intestinal stem cells

    Nature

    (2008)
  • N. Xu et al.

    Egfr, wingless and jak/stat signaling cooperatively maintain drosophila intestinal stem cells

    Dev Biol

    (2011)
  • D. Mathur et al.

    A transient niche regulates the specification of drosophila intestinal stem cells

    Science (New York, NY)

    (2010)
  • H. Jiang et al.

    Egfr signaling regulates the proliferation of drosophila adult midgut progenitors

    Development

    (2009)
  • H. Ireland et al.

    Inducible cre-mediated control of gene expression in the murine gastrointestinal tract: effect of loss of beta-catenin

    Gastroenterology

    (2004)
  • T. Fevr et al.

    Wnt/beta-catenin is essential for intestinal homeostasis and maintenance of intestinal stem cells

    Mol Cell Biol

    (2007)
  • O.J. Sansom et al.

    Loss of apc in vivo immediately perturbs wnt signaling, differentiation, and migration

    Genes Dev

    (2004)
  • Cited by (125)

    View all citing articles on Scopus
    View full text