Skip to main content
Organogenesis logoLink to Organogenesis
. 2008 Apr-Jun;4(2):87–91. doi: 10.4161/org.4.2.5854

Wnt signaling in gut organogenesis

Michael P Verzi 1, Ramesh A Shivdasani 1,
PMCID: PMC2634253  PMID: 19279719

Abstract

Wnt signaling regulates some aspect of development of nearly all endoderm-derived organs and Wnts mediate both differentiation and proliferation at different steps during visceral organogenesis. Wnt2b induces liver formation in zebrafish 1 and may combine with other inducers, Fibroblast Growth Factors 1 & 4 and Bone Morphogenetic Protein 4, to specify the mammalian liver.25 Later in development, Wnts are critical for liver expansion and, finally, for terminal hepatocyte differentiation,612 as reviewed elsewhere in this issue (Monga). Likewise, in the pancreas, Wnts drive proliferation of exocrine and endocrine cells13,14 and promote acinar cell differentiation,13,15 as reviewed in the chapter by Murtaugh. Here we examine the intricate involvement of Wnt signaling in growth and differentiation of the digestive tract.

Key words: canonical Wnt pathway, intestine, stomach, progenitor cells, Barx1, intestinal crypts, intestinal villi


Palmitoylated Wnt glycoproteins bind to the ectodomains of a receptor complex between Low-density lipoprotein receptor-related LRP5/6 and seven-pass transmembrane Frizzled proteins.16,17 Ligand binding triggers an intracellular signaling cascade that inactivates the tumor suppressor APC, an essential component of a protein complex that, in the absence of Wnt signals, phosphorylates the cytoplasmic pool of β-catenin and targets it for destruction. Inactivation of the degradation complex results in accumulation of β-catenin, which enters the nucleus and trans-activates gene expression through sequence-specific DNA-binding transcription factors of the Tcf/LEF family. Simply put, canonical Wnt signaling thus alters the transcriptional state of Tcf/LEF target genes. Although this simplified view ignores the complex regulation of the canonical pathway and known roles for Wnt proteins in planar cell polarity and calcium-dependent signaling, it is a useful starting point to consider canonical Wnt functions in organogenesis. Other contributions in this volume describe molecular features of Wnt signaling in greater detail (Habas). We focus on the role of Wnt signaling in development of the intestine and stomach, with emphasis on recent advances.

The stomach and intestine arise from a contiguous tube, with the luminal lining (mucosa) derived from embryonic endoderm and sub-mucosal cells derived from the splanchnic mesoderm. Early in development, the endoderm and surrounding mesenchyme in the stomach and intestine are morphologically indistinguishable.18 Undifferentiated gut-specific endoderm is subsequently patterned to generate distinct segments along its anterior-posterior (A-P) axis, largely on the basis of positional information encoded in the underlying mesenchyme.18,19 The small intestine mucosa extends characteristic finger-like luminal projections or villi, structures that are maintained throughout life by continuous regeneration of stem and progenitor cells found in sub-mucosal invaginations known as the crypts of Lieberkühn. A repeating structure of villi and intervening crypts runs the full length of the small bowel; organization of the colon (large intestine) is substantially similar, except that villi are replaced by flat cuffs. The role of canonical Wnt signaling in adult crypt-villus homeostasis is well established20 and represents the most intensively studied function of canonical Wnt signaling in the digestive tract.

Colorectal cancer, a common disease, results from constitutive activation of the canonical Wnt signaling pathway, which drives epithelial proliferation and differentiation in normal intestinal crypts.2123 Understanding how Wnt signaling relationships are established during gut development is therefore especially important in the intestine and will advance appreciation of oncogenic mechanisms.

Intestine

Intestinal epithelium contains four (small intestine) or three (colon) cell lineages that serve distinct functions. At the base of each small intestine (but not colonic) crypt are a handful of Paneth cells, which secrete anti-microbial defensin peptides.24,25 Also at or near the crypt base are the intestinal stem cells, which give rise to all daughter epithelial lineages.26,27 The stem cells' immediate progeny, a transit amplifying population, reside above the Paneth and stem cells and proliferate rapidly within the crypt microenvironment before their progeny migrate upward to populate small bowel villi or colonic cuffs. As cells move upward they exit the cell cycle and differentiate into enterocytes, goblet cells and enteroendocrine cells, which function to absorb nutrients, produce mucus and secrete hormones, respectively. In less than one week, cells transition from proliferating progenitors to differentiated descendants and die as they complete their migration toward the villus tip. The crypt-villus border demarcates a boundary between proliferating cells that are in the course of lineage commitment and differentiated, post-mitotic cells. The balance between these diverse populations is critical for maintaining epithelial homeostasis and bowel function.

Crypt cell turnover.

Genetic studies in mice implicate the canonical Wnt pathway in virtually every known crypt function. Homozygous deletion of Tcf4, a key transcriptional effector of Wnt signaling, compromises fetal and neonatal intervillus cells, the developmental forerunners of the crypts of Lieberkühn. These cells proliferate poorly and fail to feed the villus compartment, leading to death of animals from presumed absorptive dysfunction; expression of transcripts that mark normal crypts and intervillus regions is markedly reduced.28 These results are commonly regarded as proof of an essential role for canonical Wnt signaling in establishing the proliferative intervillus niche but they may equally reflect other, Wnt-independent functions for Tcf4. Stomach development seems unaffected in Tcf4-/- mice, which is not surprising as other evidence for a Wnt requirement in stomach epithelium is sparse. Nor is the Tcf4 requirement manifested equally along the intestine; the distal colon is spared. Because Wnt signaling seems to be essential for all intestinal crypts, such sparing may be attributed to redundancy among Tcf/LEF proteins. However, double-mutant Tcf3-/-;Tcf4-/- and Tcf4-/-;Lef1-/- embryos die of other developmental defects before they are old enough to examine the question of redundancy in gut epithelium.

As an alternative to examining Wnt-pathway function by gene targeting, two groups expressed the extracellular Wnt antagonist Dickkopf1 (Dkk1) in adult transgenic mice. This manipulation caused rapid loss of crypts and demonstrates that Wnt signaling is required not only to form crypts but also to maintain them in adults.29,30 Similarly, acute genetic ablation of APC, a negative regulator of canonical Wnt signaling, in adult mice induces crypt properties in villous epithelial cells, including proliferation and expression of crypt markers.31,32 The secreted protein R-spondin1 potentiates canonical Wnt signaling, partially independent of the Lrp5/6 co-receptor; injection of recombinant R-spondin1 in adult mice results in rapid and reversible crypt hyperproliferation.33 In summary, loss of Wnt signals is associated with loss of crypt regenerative capacity and aberrant activation of the pathway leads to uncontrolled growth. The sum of these studies strongly implicates the Wnt pathway in gut epithelial progenitor cell properties, including a proliferative role that echoes Wnt function in the liver and pancreas, and reveals that Wnt signaling in intestinal crypts is tightly controlled.

Differentiation of secretory cell lineages.

In addition to supporting progenitor cell expansion, the canonical Wnt pathway is required for proper differentiation and localization of secretory cell lineages. From their normal location at the crypt base, one infers that Paneth cells encounter especially high Wnt concentrations; indeed, Wnt activity in adult intestine is most easily detected in Paneth cells, both by immunostaining for nuclear β-catenin and reporter gene activity in the TOP-GAL mouse line.3436 Inactivation of the Wnt pathway in each of the experimental mouse models summarized above results in loss of Paneth cell markers,3638 whereas pathway activation increases Paneth cell numbers dramatically.31,32 In mice with activating Wnt mutations, Paneth cells also lose crypt-restricted localization and frequently come to lie along small bowel villi. Similarly, disruption of the Wnt receptor gene Frizzled55 mis-localizes Paneth cells high along intestinal villi.36 These observations are explained in large part by Wnt regulation of the intercellular EphB/ephrin-B signaling system for attraction or repulsion between cells. Wnt-dependent EphB/Ephrin-B signaling seems to be responsible for partitioning differentiated epithelial cells and promoting cryptward migration of the Paneth lineage.39,40 The proximal promoters of several Paneth cell-specific α-defensin genes contain conserved Tcf/LEF binding motifs. Control of gene transcription through these cis-elements is another aspect of Wnt function36 and likely represents a portion of the specific Wnt-dependent transcriptional program in Paneth cells.

The role of Wnt in differentiation of enteroendocrine and goblet cells is appreciated with lesser clarity. Tcf4-/- mice carry very few intestinal goblet cells and no enteroendocrine cells; they die before Paneth cells can be recognized morphologically28 but lack Panethcell gene products that are readily detected in control littermates.36 Inactivation of Wnt signaling through a DKK1 transgene resulted in surprising loss of all intestinal secretory cell lineages but not of absorptive enterocytes; these results hint at an essential role for Wnt in a primitive secretory progenitor or separately in each mature secretory cell type.29 In Wnt-activating APC mutants, Paneth cell numbers are increased, however this may be at the expense of goblet and enteroendocrine cells, as a dramatic decrease of these cell lineages is observed.31,32 Taken together, these studies suggest that Wnt signaling may be necessary to establish the secretory lineages but sustained, high-level Wnt activity is especially relevant in Paneth cells. Other observations suggest that the role of the Wnt pathway in intestinal cell differentiation may be more complex: R-spondin, which activates Wnt signaling and induces crypt hyperproliferation, does not affect differentiation or localization of mature cells.33

While these studies highlight the many facets of Wnt function in intestinal homeostasis, they also point to our limited understanding of how input from a single signaling pathway is interpreted to mediate diverse outcomes. Defining the basis for the different outcomes of Wnt activity—progenitor cell proliferation, lineage decisions, differentiation and cell migration—thus remains a major unsolved problem. Illustrating one example, disruption of the Wnt target gene Sox9 affects Paneth and goblet cell differentiation but not localization or differentiation of enteroendocrine cells.37 Overlapping sets of critical Wnt target genes must similarly mediate distinct functions in gut epithelium and also respond to the other signaling pathways that influence mucosal function, including Notch.4143 In Math1-/- mice, for example, secretory cell differentiation is compromised in a fashion reminiscent of genetic models with Wnt inactivation;4446 these results suggest that interactions between the Wnt and Notch pathways could determine secretory cell fates. Although attention is now shifting toward investigation of the molecular basis of individual Wnt effects, few themes have emerged to allow general conclusions.

Source and identity of intestinal Wnts.

In light of the subjacent mesenchyme's requirement in gut epithelial viability and function,18 one might assume that this tissue is the source of Wnt signals for crypt progenitors. However, detailed investigation has failed to identify Wnt-expressing cells among the loosely arranged fibroblasts that make up most of the mesenchyme or the myofibroblast network apposed to the basement membrane surrounding the intestinal crypts. By contrast, several Wnt mRNAs are identified within crypt epithelial cells, including Wnt3, Wnt6 and Wnt9b,47 which hints at the possibility of autocrine or paracrine Wnt regulation of intestinal crypt functions.

In both mouse and chick, components of the canonical Wnt pathway are expressed in complex spatial and temporal patterns along the anterior-posterior axis of the embryonic gut, suggesting the possibility of roles in patterning and organogenesis;4850 such functions remain uncertain. These comprehensive in situ hybridization analyses over multiple developmental stages revealed that Wnts and other pathway components are often restricted in expression in correspondence with distinct morphologic regions in the gut. For example, Frizzleds 4, 7 and 8 all contain an expression boundary within the developing chick cecum, suggesting a role in development of this structure.49,50 Future studies identifying the functions of these and other Wnt pathway components during gut organogenesis may clarify the reasons for complex expression patterns but functional analysis in mice is likely to be confounded by genetic redundancies.

Wnt function in villus morphogenesis.

Before the fetal small intestine generates its hallmark villi, the prospective gut mucosa is a simple, pseudostratified epithelium. Following rapid morphological changes that occur around the 14th day of gestation in mice, the earliest villus epithelial cells and intervening crypt precursors are distinguished from each other by expression of maturation and progenitor markers, respectively. Most studies we have summarized thus far were conducted in adult mice, and from their results one might infer that Wnt signaling shapes the earliest intestinal crypts. Surprisingly, the evidence for canonical Wnt activity does not appear in the developing intestine until about E16.5, as determined in TOP-GAL reporter mice or by nuclear localization of β-catenin.35 An additional surprise is that early Wnt reporter activity does not localize in the intervillus space, where the proliferating forerunners of intestinal crypts reside, but rather in maturing epithelial cells lining the earliest villi.35 In mice, intervillus regions begin to invade the sub-mucosa to form crypts well after birth, and canonical Wnt activity shifts from the villi to the intervillus regions about three days after birth. Thus, Wnt signaling may play a crucial role in establishing the crypts of Lieberkühn but appears to be inactive during villus morphogenesis even though there is clear anatomic separation of proliferating (intervillus) and differentiated (villus) cells. Despite absence of Wnt reporter activity, the genetic requirement for Tcf4 is manifested at the level of intervillus cell proliferation and early villus morphogenesis is intact in Tcf4-/- intestines.28 One possibility is that fetal intervillus Wnt activity is below the limits of detection, but this does not explain its presence in maturing villus cells; alternatively, the phenotype of Tcf4-/- mice may reflect a function other than transduction of the canonical Wnt signal. Identification of genes that Wnts regulate during gut organogenesis may help resolve these questions.

Stomach

The role of Wnt signaling in stomach development seems to be very different from that in the intestine, and whereas adult intestinal crypts depend on Wnts for myriad functions, gastric gland units, which are operationally similar, do not display a similar dependence. Nor do gastric cancers share a link to the canonical Wnt pathway.51,52

Mouse embryonic stomach endoderm closely resembles its intestinal counterpart until approximately gestational day 12; modulation of Wnt signaling after this stage appears to be critical for stomach differentiation. An insight into how stomach mesenchyme specifies its overlying epithelium came through the study of mice and cells lacking the homeodomain transcription factor Barx1, which is specifically and highly expressed in embryonic stomach mesenchyme. Barx1-/- embryos develop a rudimentary stomach that is notable for ectopic presence of intestinal epithelium in the distal segment, indicating a homeotic transformation.53,54 In recombinant cultures of embryonic tissue, normal stomach mesenchyme induces stomach epithelial marker expression in overlying endoderm, whereas mesenchyme depleted of Barx1 induces intestinal epithelial genes instead. Findings in both cultured tissue and mice hence reveal Barx1 as a pivotal factor in mesenchymal specification of stomach epithelium; furthermore, Barx1 imparts proper morphology and size to the developing stomach. A search for the molecules that might mediate mesenchymal induction of stomach epithelium uncovered two prime candidates, Sfrp1 and Sfrp2, soluble extracellular Wnt antagonists. Expression of these factors requires Barx1 and exogenous Sfrp restores the ability of Barx1-deficient mesenchyme to induce stomach-specific epithelium in vitro.53

These observations strongly suggest an inhibitory role for Wnt signaling in stomach organogenesis and for Barx1 in suppressing that signal to enable stomach epithelial specification; Wnt function in stomach development is hence distinct from that in the intestine. In the TOP-GAL reporter mouse strain,34 which expresses β-galactosidase (LacZ) in sites of canonical Wnt activity, a strong signal appears in normal mouse stomach endoderm by the 9th gestational day, begins to decay soon thereafter, and is no longer detectable in stomach mucosa at E16.5, when the Wnt pathway is first activated in intestinal villi. The purpose for Wnt signaling in the prospective stomach is unclear, but it is confined to the endoderm in a domain that corresponds very closely to the restricted expression of Barx1 in the underlying mesenchyme. Consistent with the idea that Barx1 acts to suppress Wnt signaling in adjacent endoderm, Wnt activity is protracted in Barx1-null stomach, where it persists at least until birth.53,54 Ectopic, Barx1-independent activation of the Wnt pathway using a floxed, activating β-catenin allele55 produces a stomach phenotype that resembles the Barx1-null organ, including ectopic expression of intestinal markers.54 This result independently validates the inhibitory role of Wnt signaling in stomach development.

Reduction or loss of Sfrp expression in the Barx1-/- stomach implies that Barx1 antagonizes Wnt signaling at least in part through Sfrp genes. However, it is unclear if these are direct transcriptional targets for Barx1 regulation or which Wnt ligands are relevant in stomach organogenesis. As in the intestine, which expresses multiple Wnt mRNAs,47,49,50 Heller and colleagues demonstrated the presence of several Wnt transcripts in various zones in fetal foregut mesenchyme.56 They also forced expression of Wnt1 and Wnt5a under control of the Pdx1 promoter, which is active in the distal stomach and proximal duodenum.57 Wnt1 overexpression extended stomach-type differentiation into the proximal intestine, which represents a phenotype opposite to that of Barx1-/- mice, and in some cases the distal stomach was duplicated; the pyloric sphincter failed to develop.56 Wnt5a overexpression reduced stomach size without affecting shape or histology. Wnt5a is also found in chick embryonic stomach mesenchyme during mucosal differentiation and its overexpression seems to promote activation of stomach-specific epithelial genes.58 Because these studies rely on gene overexpression and occur outside of a physiologic context, their connection with normal development may be limited. The role and gene targets of Wnt activity in the stomach before epithelial specification are unknown.

Observations in various experimental models raise the intriguing possibility that repression of Wnt signaling is a general requirement for organogenesis in foregut derivatives. Forced Wnt activation induces intestinal differentiation in prospective mouse lung59 and stomach.54 In early Xenopus embryos, Wnt/β-catenin activity also needs to be repressed in anterior endoderm to permit liver and pancreas development. High endogenous β-catenin activity in posterior endoderm promotes intestinal differentiation and represses foregut fates; inhibition of β-catenin activity in this region induces premature expression of early liver and pancreas markers.60 One simplistic possibility, observed in different organs and multiple organisms, is that Wnt signaling in endodermal derivatives leads to intestinal differentiation as the default53,59,60 and that distinct mechanisms operate in different regions of the digestive tract to suppress Wnt activity and allow alternative tissue fates.

Footnotes

Previously published online as an Organogenesis E-publication: http://www.landesbioscience.com/journals/organogenesis/article/5854

References

  • 1.Ober EA, Verkade H, Field HA, Stainier DY. Mesodermal Wnt2b signalling positively regulates liver specification. Nature. 2006;442:688–691. doi: 10.1038/nature04888. [DOI] [PubMed] [Google Scholar]
  • 2.Jung J, Zheng M, Goldfarb M, Zaret KS. Initiation of mammalian liver development from endoderm by fibroblast growth factors. Science. 1999;284:1998–2003. doi: 10.1126/science.284.5422.1998. [DOI] [PubMed] [Google Scholar]
  • 3.Rossi JM, Dunn NR, Hogan BL, Zaret KS. Distinct mesodermal signals, including BMPs from the septum transversum mesenchyme, are required in combination for hepatogenesis from the endoderm. Genes Dev. 2001;15:1998–2009. doi: 10.1101/gad.904601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Shackel N. Zebrafish and the understanding of liver development: the emerging role of the Wnt pathway in liver biology. Hepatology. 2007;45:540–541. doi: 10.1002/hep.21543. [DOI] [PubMed] [Google Scholar]
  • 5.Shin D, Shin CH, Tucker J, Ober EA, Rentzsch F, Poss KD, Hammerschmidt M, Mullins MC, Stainier DY. Bmp and Fgf signaling are essential for liver specification in zebrafish. Development. 2007;134:2041–2050. doi: 10.1242/dev.000281. [DOI] [PubMed] [Google Scholar]
  • 6.Apte U, Zeng G, Thompson MD, Muller P, Micsenyi A, Cieply B, Kaestner KH, Monga SP. beta-Catenin is critical for early postnatal liver growth. Am J Physiol Gastrointest Liver Physiol. 2007;292:1578–1585. doi: 10.1152/ajpgi.00359.2006. [DOI] [PubMed] [Google Scholar]
  • 7.Hussain SZ, Sneddon T, Tan X, Micsenyi A, Michalopoulos GK, Monga SP. Wnt impacts growth and differentiation in ex vivo liver development. Exp Cell Res. 2004;292:157–169. doi: 10.1016/j.yexcr.2003.08.020. [DOI] [PubMed] [Google Scholar]
  • 8.Monga SP, Mars WM, Pediaditakis P, Bell A, Mule K, Bowen WC, Wang X, Zarnegar R, Michalopoulos GK. Hepatocyte growth factor induces Wnt-independent nuclear translocation of beta-catenin after Met-beta-catenin dissociation in hepatocytes. Cancer Res. 2002;62:2064–2071. [PubMed] [Google Scholar]
  • 9.Monga SP, Monga HK, Tan X, Mule K, Pediaditakis P, Michalopoulos GK. Beta-catenin antisense studies in embryonic liver cultures: role in proliferation, apoptosis, and lineage specification. Gastroenterology. 2003;124:202–216. doi: 10.1053/gast.2003.50000. [DOI] [PubMed] [Google Scholar]
  • 10.Suksaweang S, Lin CM, Jiang TX, Hughes MW, Widelitz RB, Chuong CM. Morphogenesis of chicken liver: identification of localized growth zones and the role of beta-catenin/Wnt in size regulation. Dev Biol. 2004;266:109–122. doi: 10.1016/j.ydbio.2003.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tan X, Apte U, Micsenyi A, Kotsagrelos E, Luo JH, Ranganathan S, Monga DK, Bell A, Michalopoulos GK, Monga SP. Epidermal growth factor receptor: a novel target of the Wnt/beta-catenin pathway in liver. Gastroenterology. 2005;129:285–302. doi: 10.1053/j.gastro.2005.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tan X, Behari J, Cieply B, Michalopoulos GK, Monga SP. Conditional deletion of beta-catenin reveals its role in liver growth and regeneration. Gastroenterology. 2006;131:1561–1572. doi: 10.1053/j.gastro.2006.08.042. [DOI] [PubMed] [Google Scholar]
  • 13.Dessimoz J, Bonnard C, Huelsken J, Grapin-Botton A. Pancreas-specific deletion of beta-catenin reveals Wnt-dependent and Wnt-independent functions during development. Curr Biol. 2005;15:1677–1683. doi: 10.1016/j.cub.2005.08.037. [DOI] [PubMed] [Google Scholar]
  • 14.Rulifson IC, Karnik SK, Heiser PW, ten Berge D, Chen H, Gu X, Taketo MM, Nusse R, Hebrok M, Kim SK. Wnt signaling regulates pancreatic beta cell proliferation. Proc Natl Acad Sci USA. 2007;104:6247–6252. doi: 10.1073/pnas.0701509104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Murtaugh LC, Law AC, Dor Y, Melton DA. Beta-catenin is essential for pancreatic acinar but not islet development. Development. 2005;132:4663–4674. doi: 10.1242/dev.02063. [DOI] [PubMed] [Google Scholar]
  • 16.Coudreuse D, Korswagen HC. The making of Wnt: new insights into Wnt maturation, sorting and secretion. Development. 2007;134:3–12. doi: 10.1242/dev.02699. [DOI] [PubMed] [Google Scholar]
  • 17.Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006;281:22429–22433. doi: 10.1074/jbc.R600015200. [DOI] [PubMed] [Google Scholar]
  • 18.Wells JM, Melton DA. Vertebrate endoderm development. Annu Rev Cell Dev Biol. 1999;15:393–410. doi: 10.1146/annurev.cellbio.15.1.393. [DOI] [PubMed] [Google Scholar]
  • 19.Stainier DY. No organ left behind: tales of gut development and evolution. Science. 2005;307:1902–1904. doi: 10.1126/science.1108709. [DOI] [PubMed] [Google Scholar]
  • 20.Sancho E, Batlle E, Clevers H. Live and let die in the intestinal epithelium. Curr Opin Cell Biol. 2003;15:763–770. doi: 10.1016/j.ceb.2003.10.012. [DOI] [PubMed] [Google Scholar]
  • 21.Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480. doi: 10.1016/j.cell.2006.10.018. [DOI] [PubMed] [Google Scholar]
  • 22.Schneikert J, Behrens J. The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut. 2007;56:417–425. doi: 10.1136/gut.2006.093310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Clarke AR. Wnt signalling in the mouse intestine. Oncogene. 2006;25:7512–7521. doi: 10.1038/sj.onc.1210065. [DOI] [PubMed] [Google Scholar]
  • 24.Elphick DA, Mahida YR. Paneth cells: their role in innate immunity and inflammatory disease. Gut. 2005;54:1802–1809. doi: 10.1136/gut.2005.068601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ganz T. Microbiology: Gut defence. Nature. 2003;422:478–479. doi: 10.1038/422478a. [DOI] [PubMed] [Google Scholar]
  • 26.Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, Clevers H. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–1007. doi: 10.1038/nature06196. [DOI] [PubMed] [Google Scholar]
  • 27.Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the intestine: stem cells, signals and combinatorial control. Nat Rev Genet. 2006;7:349–359. doi: 10.1038/nrg1840. [DOI] [PubMed] [Google Scholar]
  • 28.Korinek V, Barker N, Moerer P, van Donselaar E, Huls G, Peters PJ, Clevers H. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet. 1998;19:379–383. doi: 10.1038/1270. [DOI] [PubMed] [Google Scholar]
  • 29.Pinto D, Gregorieff A, Begthel H, Clevers H. Canonical Wnt signals are essential for homeostasis of the intestinal epithelium. Genes Dev. 2003;17:1709–1713. doi: 10.1101/gad.267103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kuhnert F, Davis CR, Wang HT, Chu P, Lee M, Yuan J, Nusse R, Kuo CJ. Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1. Proc Natl Acad Sci USA. 2004;101:266–271. doi: 10.1073/pnas.2536800100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Andreu P, Colnot S, Godard C, Gad S, Chafey P, Niwa-Kawakita M, Laurent-Puig P, Kahn A, Robine S, Perret C, Romagnolo B. Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine. Development. 2005;132:1443–1451. doi: 10.1242/dev.01700. [DOI] [PubMed] [Google Scholar]
  • 32.Sansom OJ, Reed KR, Hayes AJ, Ireland H, Brinkmann H, Newton IP, Batlle E, Simon-Assmann P, Clevers H, Nathke IS, Clarke AR, Winton DJ. Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration. Genes Dev. 2004;18:1385–1390. doi: 10.1101/gad.287404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kim KA, Kakitani M, Zhao J, Oshima T, Tang T, Binnerts M, Liu Y, Boyle B, Park E, Emtage P, Funk WD, Tomizuka K. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science. 2005;309:1256–1259. doi: 10.1126/science.1112521. [DOI] [PubMed] [Google Scholar]
  • 34.DasGupta R, Fuchs E. Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development. 1999;126:4557–4568. doi: 10.1242/dev.126.20.4557. [DOI] [PubMed] [Google Scholar]
  • 35.Kim BM, Mao J, Taketo MM, Shivdasani RA. Phases of canonical Wnt signaling during the development of mouse intestinal epithelium. Gastroenterology. 2007;133:529–538. doi: 10.1053/j.gastro.2007.04.072. [DOI] [PubMed] [Google Scholar]
  • 36.van Es JH, Jay P, Gregorieff A, van Gijn ME, Jonkheer S, Hatzis P, Thiele A, van den Born M, Begthel H, Brabletz T, Taketo MM, Clevers H. Wnt signalling induces maturation of Paneth cells in intestinal crypts. Nat Cell Biol. 2005;7:381–386. doi: 10.1038/ncb1240. [DOI] [PubMed] [Google Scholar]
  • 37.Bastide P, Darido C, Pannequin J, Kist R, Robine S, Marty-Double C, Bibeau F, Scherer G, Joubert D, Hollande F, Blache P, Jay P. Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. J Cell Biol. 2007;178:635–648. doi: 10.1083/jcb.200704152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mori Akiyama Y, van den Born M, van Es JH, Hamilton SR, Adams HP, Zhang J, Clevers H, de Crombrugghe B. SOX9 is required for the differentiation of paneth cells in the intestinal epithelium. Gastroenterology. 2007;133:539–546. doi: 10.1053/j.gastro.2007.05.020. [DOI] [PubMed] [Google Scholar]
  • 39.Batlle E, Henderson JT, Beghtel H, van den Born MM, Sancho E, Huls G, Meeldijk J, Robertson J, van de Wetering M, Pawson T, Clevers H. Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell. 2002;111:251–263. doi: 10.1016/s0092-8674(02)01015-2. [DOI] [PubMed] [Google Scholar]
  • 40.van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, van der Horn K, Batlle E, Coudreuse D, Haramis AP, Tjon-Pon-Fong M, Moerer P, van den Born M, Soete G, Pals S, Eilers M, Medema R, Clevers H. The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell. 2002;111:241–250. doi: 10.1016/s0092-8674(02)01014-0. [DOI] [PubMed] [Google Scholar]
  • 41.Milano J, McKay J, Dagenais C, Foster Brown L, Pognan F, Gadient R, Jacobs RT, Zacco A, Greenberg B, Ciaccio PJ. Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004;82:341–358. doi: 10.1093/toxsci/kfh254. [DOI] [PubMed] [Google Scholar]
  • 42.van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, Begthel H, Cozijnsen M, Robine S, Winton DJ, Radtke F, Clevers H. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435:959–963. doi: 10.1038/nature03659. [DOI] [PubMed] [Google Scholar]
  • 43.Zecchini V, Domaschenz R, Winton D, Jones P. Notch signaling regulates the differentiation of post-mitotic intestinal epithelial cells. Genes Dev. 2005;19:1686–1691. doi: 10.1101/gad.341705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shroyer NF, Wallis D, Venken KJ, Bellen HJ, Zoghbi HY. Gfi1 functions downstream of Math1 to control intestinal secretory cell subtype allocation and differentiation. Genes Dev. 2005;19:2412–2417. doi: 10.1101/gad.1353905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yang Q, Bermingham NA, Finegold MJ, Zoghbi HY. Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science. 2001;294:2155–2158. doi: 10.1126/science.1065718. [DOI] [PubMed] [Google Scholar]
  • 46.Shroyer NF, Helmrath MA, Wang VY, Antalffy B, Henning SJ, Zoghbi HY. Intestine-specific ablation of mouse atonal homolog 1 (Math1) reveals a role in cellular homeostasis. Gastroenterology. 2007;132:2478–2488. doi: 10.1053/j.gastro.2007.03.047. [DOI] [PubMed] [Google Scholar]
  • 47.Gregorieff A, Pinto D, Begthel H, Destree O, Kielman M, Clevers H. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology. 2005;129:626–638. doi: 10.1016/j.gastro.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 48.Lickert H, Kispert A, Kutsch S, Kemler R. Expression patterns of Wnt genes in mouse gut development. Mech Dev. 2001;105:181–184. doi: 10.1016/s0925-4773(01)00390-2. [DOI] [PubMed] [Google Scholar]
  • 49.McBride HJ, Fatke B, Fraser SE. Wnt signaling components in the chicken intestinal tract. Dev Biol. 2003;256:18–33. doi: 10.1016/s0012-1606(02)00118-5. [DOI] [PubMed] [Google Scholar]
  • 50.Theodosiou NA, Tabin CJ. Wnt signaling during development of the gastrointestinal tract. Dev Biol. 2003;259:258–271. doi: 10.1016/s0012-1606(03)00185-4. [DOI] [PubMed] [Google Scholar]
  • 51.Smith MG, Hold GL, Tahara E, El-Omar EM. Cellular and molecular aspects of gastric cancer. World J Gastroenterol. 2006;12:2979–2990. doi: 10.3748/wjg.v12.i19.2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vogiatzi P, Vindigni C, Roviello F, Renieri A, Giordano A. Deciphering the underlying genetic and epigenetic events leading to gastric carcinogenesis. J Cell Physiol. 2007;211:287–295. doi: 10.1002/jcp.20982. [DOI] [PubMed] [Google Scholar]
  • 53.Kim BM, Buchner G, Miletich I, Sharpe PT, Shivdasani RA. The stomach mesenchymal transcription factor Barx1 specifies gastric epithelial identity through inhibition of transient Wnt signaling. Dev Cell. 2005;8:611–622. doi: 10.1016/j.devcel.2005.01.015. [DOI] [PubMed] [Google Scholar]
  • 54.Kim BM, Miletich I, Mao J, McMahon AP, Sharpe PA, Shivdasani RA. Independent functions and mechanisms for homeobox gene Barx1 in patterning mouse stomach and spleen. Development. 2007;134:3603–3613. doi: 10.1242/dev.009308. [DOI] [PubMed] [Google Scholar]
  • 55.Harada N, Tamai Y, Ishikawa T, Sauer B, Takaku K, Oshima M, Taketo MM. Intestinal polyposis in mice with a dominant stable mutation of the beta-catenin gene. Embo J. 1999;18:5931–5942. doi: 10.1093/emboj/18.21.5931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Heller RS, Dichmann DS, Jensen J, Miller C, Wong G, Madsen OD, Serup P. Expression patterns of Wnts, Frizzleds, sFRPs and misexpression in transgenic mice suggesting a role for Wnts in pancreas and foregut pattern formation. Dev Dyn. 2002;225:260–270. doi: 10.1002/dvdy.10157. [DOI] [PubMed] [Google Scholar]
  • 57.Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, Hogan BL, Wright CV. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development. 1996;122:983–995. doi: 10.1242/dev.122.3.983. [DOI] [PubMed] [Google Scholar]
  • 58.Listyorini D, Yasugi S. Expression and function of Wnt5a in the development of the glandular stomach in the chicken embryo. Dev Growth Differ. 2006;48:243–252. doi: 10.1111/j.1440-169X.2006.00861.x. [DOI] [PubMed] [Google Scholar]
  • 59.Okubo T, Hogan BL. Hyperactive Wnt signaling changes the developmental potential of embryonic lung endoderm. J Biol. 2004;3:11. doi: 10.1186/jbiol3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.McLin VA, Rankin SA, Zorn AM. Repression of Wnt/beta-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development. 2007;134:2207–2217. doi: 10.1242/dev.001230. [DOI] [PubMed] [Google Scholar]

Articles from Organogenesis are provided here courtesy of Taylor & Francis

RESOURCES