Pediatric/congenital/developmental
Temporal Changes in the Intestinal Growth Promoting Effects of Glucagon-Like Peptide 2 Following Intestinal Resection

https://doi.org/10.1016/j.jss.2008.05.007Get rights and content

Background

We investigated the effects of variations in the postresection timing of glucagon-like peptide-2 (GLP-2) administration on intestinal morphology and activity.

Methods

A rat model of 90% intestinal resection (SBR) with exclusively parenteral nutritional (TPN) was used. Early versus late postresection GLP-2 stimulation was compared between SBR + TPN alone, SBR + TPN + GLP-2 (first wk), and SBR + TPN + GLP-2 (second wk) (n = 8/group). On d 14, animals were sacrificed and remnant ileum analyzed for morphology, crypt cell proliferation index (CPI), apoptosis index (API), and nutrient transporter expression (SGLT-1, GLUT-2, GLUT-5). In a separate study, the resection-induced effect on acute GLP-2 responsiveness was studied at d 3 and 10, in control or SBR animals, both supported with TPN. (n = 6).

Results

Bowel length, weight, and width were increased in SBR + TPN + GLP-2 (first wk) compared with the SBR + TPN alone and SBR + TPN + GLP-2 (second wk) groups. Animal weight, villus height, total mucosal surface area, and CPI increased in both GLP-2 treated groups compared with the SBR + TPN group. Villus height and crypt depth effects were most pronounced in the SBR + TPN + GLP-2 (second wk) group. Increased expression of mRNA for the GLP-2 receptor was noted at d 3, declining below baseline by d 10, however this was not correlated with GLP-2 activation of enteric neurons. Exogenous GLP-2 increased the activation of submucosal neurons at d 3 in controls; resected animals had a higher baseline activity, but exogenous GLP-2 did not activate this further at either d 3 or 10 postresection.

Conclusions

GLP-2 effects on intestinal growth are maximal in the early postresection period and are associated with an apparent increase in expression of the receptor but no increase in neuronal activation. This suggests that the intestinal adaptive and growth promoting actions of GLP-2 may be mediated by non-neuronal effector pathways. Although further studies are required, early treatment with GLP-2 following resection may maximize intestinal growth.

Introduction

The regulation of nutrient absorptive capacity is an important component of normal intestinal physiology. In the natural state, individuals are subjected to widely varying types and quantities of nutrients [1]. Following states of intestinal disease or inflammation, additional compensation is required, so that proximal intestinal dysfunction may be offset by increased uptake of nutrients in the distal intestine. An extreme variation of this is the response to the reduction of nutrient absorptive capacity associated with the loss of surface area following massive intestinal resection, known as short bowel syndrome (SBS) [2]. After resection, the residual intestine increases both intestinal surface area and absorptive enzymatic capacity, primarily by increasing the rate of crypt cell proliferation. In turn, this augments the nutrient absorptive capacity of the remnant intestine, a process known as “adaptation” [3, 4]. After a 90% resection of the proximal small intestine in the rat, the residual bowel up-regulates function so that after 7 d, nutrient absorption is restored to preresection efficiency [5]. The factors that control this adaptation are poorly understood, but they appear to be related to signals induced by enteral nutrients. Clinically, adaptation appears to only occur if luminal nutrition is administered [2, 3]. Previous studies from our group have shown that endogenous glucagon-like peptide 2 (GLP-2) is highly associated with the adaptive process, and exogenously administered GLP-2 can induce adaptation without the requirement for enteral nutrition [6, 7, 8, 9].

GLP-2 is a 33-amino acid member of the pituitary adenylate cyclase-activating glucagon superfamily [6, 9, 10]. GLP-2 is secreted from ileal and colonic enteroendocrine L cells in response to both proximal enteric neuronal signaling and the presence of luminal nutrients [6, 10, 11]. Previous studies have demonstrated that GLP-2 administration results in intestinal hypertrophy by increasing the crypt cell proliferation rate, which then results in increased villus height, crypt depth, and an overall increase in small intestinal length and weight [8, 12, 13]. This mimics the features of spontaneous adaptation, but occurred in animals maintained with entirely parenteral nutrition [7]. In addition, GLP-2 increased the expression of glucose transport protein (SGLT-1) much more than in enterally fed animals [7]. The mechanism(s) of action of GLP-2 in stimulating these effects is not clear; the receptor is confined to cells within the enteric neuronal system, entero-endocrine cells, and myofibroblasts in the submucosa [5, 9, 13]. In previous experimental studies, it has been shown that anti-inflammatory effects of GLP-2 are associated with activation of submucosal VIP expressing neurons of the enteric neuronal system [14]. However, in studies using knockout models, it has been shown that the trophic effects of GLP-2 on the intestinal mucosa appear to be dependent on local production of IGF-1, likely from myofibroblasts adjacent to the dividing crypt cells [15]. In clinical studies, GLP-2 has been shown to improve energy absorption in adult patients with SBS [16]. From this background, it is hypothesized that GLP-2 acts as a general regulator of nutrient absorptive capacity. It may become a useful therapy for human patients with impaired nutrient absorption [2, 6, 10]; however the optimal timing of administration postoperation is not known. The present studies examined (1) the hypothesis that the timing of GLP-2 administration would affect the trophic effects on the residual intestine, with early administration increasing the effects, and (2) the hypothesis that there would be an increase in the sensitivity of the residual intestine to exogenous GLP-2 administration, due to an increase in the expression of the GLP-2 receptor and an associated increase in the activation of the submucosal plexus of the enteric neuronal system. Herein, we show that early GLP-2 administration does have greater effects on intestinal growth following resection, and that GLP-2 receptor mRNA expression is up-regulated in the early postresection period, but that GLP-2 stimulation does not cause an increase in enteric neuronal activation within the submucosa in this same time period.

Section snippets

Animals

Male Sprague Dawley (SD) rats weighing 250 to 300 g were housed in metabolic caging with free access to food and water, and acclimatized to their environment for 5 d before experiments. Animals were maintained under standardized temperature, humidity, and 12 h light-dark cycles. Animal studies were conducted under the guidelines established by the Canadian Council of Animal Care following the approval of the Animal Care Committee at the University of Calgary.

Study Design

The effects of variations in timing

Results

In general, the animals did well. Two animals died from complications related to the intravenous catheter; these animals were replaced. Two further animals, one each from the SBR + TPN + GLP– 2 (first wk) and SBR + TPN + GLP-2 (second wk) groups had catheter occlusions; these catheters were replaced under a brief general anesthetic on d 3 and 4 of study, respectively, and their results used in all data fields.

Examining the effects of GLP-2 given immediately or delayed following resection, there

Discussion

In this study, we have demonstrated that following intestinal resection an early increase in the effect of exogenous GLP-2 on intestinal growth occurs, with persistent effects following withdrawal of the hormone. Similar, but less profound morphological effects, with increased mucosal proliferative and nutrient transporter effects were seen with delayed GLP-2 treatment. Both early and late GLP-2 administration increased weight accretion in TPN maintained animals. An increase in expression of

Acknowledgments

This study were supported by the Alberta Children's Hospital Research Foundation, and the Department of Surgery Research Fund, University of Calgary. Infrastructure support of the Sigalet Laboratory is provided by the Crohn's and Colitis Foundation of Canada. TK was supported by a Research Fellowship from the Department of Pediatric Surgery, Kagoshima University. HT was supported by a Research Scholarship from the Department of Pediatric Surgery, Kurume University School of Medicine. DLS has

References (36)

  • R.C. Williamson

    Intestinal adaptation (second of two parts)Mechanisms of control

    N Engl J Med

    (1978)
  • J.J. Cisler et al.

    Intestinal adaptation in short bowel syndrome

    J Investig Med

    (2005)
  • G.R. Martin et al.

    Glucagon-like peptide-2 induces intestinal adaptation in parenterally fed rats with short bowel syndrome

    Am J Physiol Gastrointest Liver Physiol

    (2004)
  • G.R. Martin et al.

    Nutrient-stimulated GLP-2 release and crypt cell proliferation in experimental short bowel syndrome

    Am J Physiol Gastrointest Liver Physiol

    (2005)
  • G.R. Martin et al.

    Gut hormones and short bowel syndrome: The enigmatic role of glucagon-like peptide-2 in the regulation of intestinal adaptation

    World J Gastroenterol

    (2006)
  • D.L. Sigalet

    ALX-0600 (NPS Allelix Corp)

    Curr Opin Investig Drugs

    (2001)
  • J.N. Roberge et al.

    Secretion of proglucagon-derived peptides in response to intestinal luminal nutrients

    Endocrinology

    (1991)
  • D.J. Drucker et al.

    Induction of intestinal epithelial proliferation by glucagon-like peptide 2

    Proc Natl Acad Sci. USA

    (1996)
  • Cited by (27)

    • Teduglutide effects on gene regulation of fibrogenesis on an animal model of intestinal anastomosis

      2017, Journal of Surgical Research
      Citation Excerpt :

      In “Ted+” groups, teduglutide (American Peptide Company) was prepared following the manufacturer's recommendations and administered subcutaneously in the postoperative period (starting on the day of the operation), 200 μg/kg/d, dissolved in 0.25 mL phosphate-buffered saline pH 7.4 (Gibco; Life Technologies). This posology has already been associated with enterotrophic effects in previous studies.15-18 Most distal segment, with 4 cm length, was carefully removed from each ileal operative specimen.

    • Effects of chronic glucagon-like peptide-2 therapy during weaning in neonatal pigs

      2014, Regulatory Peptides
      Citation Excerpt :

      This is of significant importance if GLP-2 is to be used chronically in infants with intestinal failure, in whom a major goal of therapy is to increase the enteral intake of nutrients, and calories [38,39]. Examining the effects on the intestinal tissues, the results are similar to those seen in previous studies; GLP-2 increases the rate of crypt cell proliferation, and decreases the rate of apoptosis [16,28]. However, it is notable that the effects were somewhat different in different sections of the intestine; there were greater increases in the rate of proliferation in the jejunum and ileum, with no significant increase in the colon.

    • A dose-equivalent comparison of the effects of continuous subcutaneous glucagon-like peptide 2 (GLP-2) infusions versus meal related GLP-2 injections in the treatment of short bowel syndrome (SBS) patients

      2013, Regulatory Peptides
      Citation Excerpt :

      It is, however, possible that the optimal dose may be highly individual depending on remnant bowel function and ability to tolerate treatment. This raises the possibility that there is receptor downregulation or desensitization, as seen in animal models [22,23]. Given the adverse event profile and potential risks of treatment with GLP-2 and teduglutide, the use of the lowest effective dose is desirable [12,13].

    • Ghrelin and glucagon-like peptide-2 increase immediately following massive small bowel resection

      2013, Peptides
      Citation Excerpt :

      From our laboratory findings, we could imagine a more effective method of inducing residual intestinal adaptation after massive small bowel resection. In a previous study, GLP-2 receptor expression was shown to have significantly increased by postoperative day 3 in 90% SBR model rats [13]. Moreover, continued intravenous administration of GLP-2 during the first postoperative week was required to maximize the adaptation of the remaining bowel in rats [12,13].

    View all citing articles on Scopus
    View full text