Gastroenterology

Gastroenterology

Volume 141, Issue 6, December 2011, Pages 2166-2175.e7
Gastroenterology

Original Research
Basic and Translational—Alimentary Tract
Loss of Glucagon-Like Peptide-2–Induced Proliferation Following Intestinal Epithelial Insulin-Like Growth Factor-1–Receptor Deletion

https://doi.org/10.1053/j.gastro.2011.09.014Get rights and content

Background & Aims

Glucagon-like peptide-2 (GLP-2) is an intestinal hormone that promotes growth of the gastrointestinal tract. Although insulin-like growth factor (IGF)-1 and the IGF-1 receptor (IGF-1R) are required for GLP-2–induced proliferation of crypt cells, little is known about localization of the IGF-1R which mediates the intestinotropic actions of GLP-2.

Methods

We examined intestinal growth and proliferative responses in mice with conditional deletion of IGF-1R from intestinal epithelial cells (IE-igf1rKO) after acute administration (30–90 min) of GLP-2, in response to 24-hour fasting and re-feeding (to induce GLP-2–dependent adaptation), and after chronic exposure (10 days) to GLP-2.

Results

IE-igf1rKO mice had normal small intestinal weight, morphometric parameters, proliferative indices, and distribution of differentiated epithelial cell lineages. Acute administration of GLP-2 increased nuclear translocation of β-catenin in non-Paneth crypt cells and stimulated the crypt–cell proliferative marker c-Myc in control but not IE-igf1rKO mice. Small intestinal weight, crypt depth, villus height, and crypt-cell proliferation were decreased in control and IE-igf1rKO mice after 24-hour fasting. Although re-feeding control mice restored all of these parameters, re-fed IE-igf1rKO mice had reductions in adaptive regrowth of the villi and crypt-cell proliferation. Control mice that were given chronic GLP-2 had increases in small intestinal weight, mucosal cross-sectional area, crypt depth, villus height, and crypt-cell proliferation. However, the GLP-2–induced increase in crypt-cell proliferation was not observed in IE-igf1rKO mice, and growth of the crypt–villus axis was reduced.

Conclusions

The proliferative responses of the intestinal epithelium to exogenous GLP-2 administration and conditions of GLP-2–dependent adaptive re-growth require the intestinal epithelial IGF-1R.

Section snippets

Animals

IE-igf1rKO mice were generated by crossing villin-Cre-ERT2+/0 and Igf1rflox/flox mice,22, 23, 24 both on a C57BL/6 background (Figure 1A). The resultant villin-Cre-ERT2+/0; Igf1rflox/+ offspring were mated to Igf1rflox/flox mice to generate the villin-Cre-ERT2+/0; Igf1rflox/flox (IE-igf1rKO) animals. Female villin-Cre-ERT2 breeders were avoided in F1 and F2 generations because of potential Cre excision from the maternal genome.25 Age- and sex-matched littermate Igf1rflox/flox and villin-Cre-ER

IGF-1R Deletion in IE-igf1rKO Mice

Polymerase chain reaction (PCR) analysis of IECs collected by laser capture microdissection 1 day after tamoxifen treatment showed the presence of the excised gene fragment (204 bp) only in cells from IE-igf1rKO mice as compared with control (Igf1rflox/flox) animals (Figure 1B). Densitometric analysis of the PCR products from 2 mice indicated approximately 85% recombination in IECs from the IE-igf1rKO mice. Moreover, quantitative reverse-transcription (qRT)-PCR for IGF-1R mRNA in jejunal mucosa

Discussion

Based on the localization of both IGF-130, 31, 32, 33, 34 and the GLP-2R11 in the intestinal subepithelial myofibroblasts that subtend the crypt epithelium, as well as on expression of the IGF-1R on the basolateral membrane of crypt IECs,30, 35 we have proposed a role for the IEC IGF-1R in GLP-2–mediated SI proliferation.3, 4 Moreover, we recently showed that GLP-2 not only stimulates IGF-1 secretion from cultured fetal rat intestinal cells,13 but also increases IGF-1 mRNA transcript levels in

Acknowledgments

The authors thank Dr Sylvie Robine (Institut Curie-Centre National de la Recherche Scientique, Paris, France) for the kind gift of the villin-cre mice, and Mr Angelo Izzo for technical support. The authors also thank Mr Brent Steer and Dr Phil Marsden (University of Toronto) for assistance with laser capture microdissection procedures.

References (41)

  • P.K. Lund et al.

    Somatomedin-C/insulin-like growth factor-I and insulin-like growth factor-II mRNAs in rat fetal and adult tissues

    J Biol Chem

    (1986)
  • O. Muhlemann et al.

    Recognition and elimination of nonsense mRNA

    Biochim Biophys Acta

    (2008)
  • J.P. Liu et al.

    Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r)

    Cell

    (1993)
  • S. Dhanvantari et al.

    Role of prohormone convertases in the tissue-specific processing of proglucagon

    Mol Endocrinol

    (1996)
  • P.E. Dube et al.

    Frontiers in glucagon-like peptide-2: multiple actions, multiple mediators

    Am J Physiol Endocrinol Metab

    (2007)
  • D.W. Nelson et al.

    Insulin-like growth factor I and glucagon-like peptide-2 responses to fasting followed by controlled or ad libitum refeeding in rats

    Am J Physiol Regul Integr Comp Physiol

    (2008)
  • M. Bjerknes et al.

    Modulation of specific intestinal epithelial progenitors by enteric neurons

    Proc Natl Acad Sci U S A

    (2001)
  • A. Ramsanahie et al.

    Effect of GLP-2 on mucosal morphology and SGLT1 expression in tissue-engineered neointestine

    Am J Physiol Gastrointest Liver Physiol

    (2003)
  • P.E. Dube et al.

    Glucagon-like peptide-2 activates beta-catenin signaling in the mouse intestinal crypt: role of insulin-like growth factor-I

    Endocrinology

    (2008)
  • D.L. Sigalet et al.

    Enteric neural pathways mediate the anti-inflammatory actions of glucagon-like peptide 2

    Am J Physiol Gastrointest Liver Physiol

    (2007)
  • Cited by (70)

    • Glucagon-Like Peptide-2 Stimulates S-Phase Entry of Intestinal Lgr5+ Stem Cells

      2022, Cellular and Molecular Gastroenterology and Hepatology
    • Feeding colostrum or a 1:1 colostrum:milk mixture for 3 days postnatal increases small intestinal development and minimally influences plasma glucagon-like peptide-2 and serum insulin-like growth factor-1 concentrations in Holstein bull calves

      2020, Journal of Dairy Science
      Citation Excerpt :

      Of particular interest to the current study is the oral delivery of IGF-1 in colostrum (Pakkanen and Aalto, 1997; Blum and Hammon, 2000) to the neonatal calf by its small intestine-targeted mitogenic capacity to increase epithelial cell proliferation (MacDonald, 1999; Shen et al., 2004). Because the mitogenic action of IGF-1 can be enhanced by GLP-2 (Dubé et al., 2006; Rowland et al., 2011; Leen et al., 2011), these 2 hormones were promising targets to determine their relationship to neonatal intestinal development and postnatal nutrient consumption. Yet, study findings did not support the hypothesis that elevated plasma GLP-2 and serum IGF-1 concentrations would reflect and coincide with enhanced intestinal development by COL compared with MIX or WM feeding.

    View all citing articles on Scopus

    Conflicts of interest This author discloses the following: Patricia Brubaker has received consulting fees from Glaxo-Smith-Kline and Regeneron. The remaining authors disclose no conflicts.

    Funding Katherine Rowland was supported by a Doctoral Research Award from the Canadian Institutes of Health Research in partnership with the Canadian Digestive Health Foundation, and by a Banting and Best Diabetes Centre Graduate Studentship, University of Toronto; Shivangi Trivedi and Ken Wan were supported by Canadian Association of Gastroenterology Summer Studentships; and Patricia Brubaker was supported by the Canada Research Chairs Program. These studies were supported by an operating grant from the Canadian Institutes of Health Research (#MOP-9940 to PLB); and by the National Institutes of Health (RO1 #DK67536; to RNK).

    View full text