Gastroenterology

Gastroenterology

Volume 140, Issue 4, April 2011, Pages 1219-1229.e2
Gastroenterology

Basic—Alimentary Tract
GATA Factors Regulate Proliferation, Differentiation, and Gene Expression in Small Intestine of Mature Mice

https://doi.org/10.1053/j.gastro.2011.01.033Get rights and content

Background & Aims

GATA transcription factors regulate proliferation, differentiation, and gene expression in multiple organs. GATA4 is expressed in the proximal 85% of the small intestine and regulates the jejunal-ileal gradient in absorptive enterocyte gene expression. GATA6 is co-expressed with GATA4 but also is expressed in the ileum; its function in the mature small intestine is unknown.

Methods

We investigated the function of GATA6 in small intestine using adult mice with conditional, inducible deletion of Gata6, or Gata6 and Gata4, specifically in the intestine.

Results

In ileum, deletion of Gata6 caused a decrease in crypt cell proliferation and numbers of enteroendocrine and Paneth cells, an increase in numbers of goblet-like cells in crypts, and altered expression of genes specific to absorptive enterocytes. In contrast to ileum, deletion of Gata6 caused an increase in numbers of Paneth cells in jejunum and ileum. Deletion of Gata6 and Gata4 resulted in a jejunal and duodenal phenotype that was nearly identical to that in the ileum after deletion of Gata6 alone, revealing common functions for GATA6 and GATA4.

Conclusions

GATA transcription factors are required for crypt cell proliferation, secretory cell differentiation, and absorptive enterocyte gene expression in the small intestinal epithelium.

Section snippets

Mice

Previously established and confirmed Gata6loxP/loxP, Gata4flap/flap, and transgenic VillinCreERT2 mice16, 20, 22, 23 were used in this study to produce conditional, inducible deletion of Gata6 or both Gata6 and Gata4 in the intestinal epithelium. Gata6loxP/loxP, VillinCreERT2-positive (G6del), Gata6loxP/loxP, Gata4flap/flap, VillinCreERT2-positive (G6G4del), and Gata6loxP/loxP, VillinCreERT2-negative or Gata6Wt/Wt, VillinCreERT2-positive (control) mice, 6–8 weeks of age, were treated with

Intestinal Gata6 Deletion Results in a Reduction in Villus Length, Villus Epithelial Cell Number, and Crypt Proliferation in Ileum

GATA6 was expressed in all differentiated and proliferating cells in the mature mouse small intestinal epithelium, with the highest staining intensity in the proliferative crypt compartment (Supplementary Figure 2). To determine the function of GATA6 in this tissue, a tamoxifen-inducible, intestine-specific Gata6 deletion model (G6del) was established (Supplementary Figure 3). Mice killed up to 4 weeks after Gata6 deletion showed normal growth and activity; in a cohort of male mice, body

Discussion

We previously showed that GATA4 specifically regulates jejuno–ileal differences in absorptive enterocyte gene expression and function.16, 22 Here, we show that in ileum, where Gata6 is expressed but Gata4 is not, conditional deletion of Gata6 results in a decrease in cellular proliferation in crypts, a decrease in enteroendocrine cell allocation, a conversion of Paneth cells into goblet-like cells at the base of crypts, and an alteration in the expression of specific absorptive enterocyte genes

Acknowledgments

The authors thank Ms M. P. Flanagan for technical support, Dr S. Hagan and A. Calhoun for technical assistance and insight with electron microscopy, Dr H. Clevers for samples from the Spdef knockout mice, Dr P. A. Dawson and Ms J. Haywood for valuable insight and technical support, Dr A. J. Ouellette for the alpha-defensin related sequence and alpha-defensin 1 antibodies, Dr D. K. Podolsky for the trefoil factor 3 antibody, Dr J. Whitsett for the SAM pointed domain containing ets transcription

References (41)

  • S. Ezoe et al.

    GATA-2/estrogen receptor chimera regulates cytokine-dependent growth of hematopoietic cells through accumulation of p21(WAF1) and p27(Kip1) proteins

    Blood

    (2002)
  • H. Perlman et al.

    GATA-6 induces p21(Cip1) expression and G1 cell cycle arrest

    J Biol Chem

    (1998)
  • P. Andreu et al.

    A genetic study of the role of the Wnt/beta-catenin signalling in Paneth cell differentiation

    Dev Biol

    (2008)
  • V. Korinek et al.

    Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4

    Nat Genet

    (1998)
  • F. Kuhnert et al.

    Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1

    Proc Natl Acad Sci U S A

    (2004)
  • D. Pinto et al.

    Canonical Wnt signals are essential for homeostasis of the intestinal epithelium

    Genes Dev

    (2003)
  • P. Andreu et al.

    Crypt-restricted proliferation and commitment to the Paneth cell lineage following Apc loss in the mouse intestine

    Development

    (2005)
  • C. Crosnier et al.

    Delta-Notch signalling controls commitment to a secretory fate in the zebrafish intestine

    Development

    (2005)
  • O. Riccio et al.

    Loss of intestinal crypt progenitor cells owing to inactivation of both Notch1 and Notch2 is accompanied by derepression of CDK inhibitors p27Kip1 and p57Kip2

    EMBO Rep

    (2008)
  • J.H. van Es et al.

    Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells

    Nature

    (2005)
  • Cited by (91)

    • Super-enhancers: Implications in gastric cancer

      2024, Mutation Research - Reviews in Mutation Research
    • Regulatory domains controlling high intestinal vitamin D receptor gene expression are conserved in mouse and human

      2022, Journal of Biological Chemistry
      Citation Excerpt :

      Among these, CDX2 is viewed as a master regulator of intestinal epithelial cell identity (24) that maintains a permissive chromatin environment that favorably influences the DNA binding of other TFs, for example, the intestine restricted TF HNF4a (25). Similarly, SMAD4 and HNF4 activate each other's expression and cobind to regulatory elements of intestinal differentiation genes to stabilize enterocyte identity (26), whereas other evidence shows that GATA4 is critical for gene expression in the proximal small intestine (27). However, the established role for these TFs neither precludes a role for other TFs in the small intestine nor does explain the regulation in the colon, where GATA4 is absent (28).

    View all citing articles on Scopus

    Transcript profiling: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE22416).

    Conflicts of interest The authors disclose no conflicts.

    Funding This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases grants RO3-DK-84167 (N.F.S.), RO1-DK-055743 (S.A.D.), RO1-DK-066226 (S.A.D.), RO1-DK-054111 (J.C.F.), and RO1-DK-061382 (S.D.K.); Harvard Digestive Disease Center grant 5P30-DK-34854; National Cancer Institute grant RO1-CA-142826 (N.F.S.); the Nutricia Research Foundation (E.B.); the Foundation De Drie Lichten (E.B.); and the Foundation Doctor Catharine van Tussenbroek (E.B.) in The Netherlands.

    View full text