You are on page 1of 4

Recent Advances in Nutritional Sciences

Glucagon-Like Peptide 2: A In the last five years, glucagon-like peptide 2 (GLP-2)5


has emerged as one of the most intriguing and potent
Nutrient-Responsive Gut Growth modulators of intestinal growth and function. GLP-2 is a
Factor1,2,3 member of a family of glucagon-like peptides, which have a
variety of important biological functions not only within
the gastrointestinal (GI) tract, where they are produced,
Douglas G. Burrin,4 Yvette Petersen,* Barbara Stoll but also in the body as a whole in terms of carbohydrate
and Per Sangild* metabolism and appetite regulation. Although the biologi-
U.S. Department of Agriculture/ARS Children’s Nutrition Research cal function of pancreatic glucagon is well established, the
Center, Department of Pediatrics, Baylor College of Medicine, precise structural identity and function of gut glucagon, or
“enteroglucagon,” has until recently remained obscure. An

Downloaded from https://academic.oup.com/jn/article-abstract/131/3/709/4687255 by guest on 05 June 2020


Houston, TX 77030 and *Division of Animal Nutrition, Royal
Veterinary and Agricultural University, DK-1870 Copenhagen, early clue that glucagon-like peptides have intestinal tro-
Denmark phic actions was the report of small bowel hyperplasia in
patients with glucagonomas (1). Drucker and others (2)
reproduced this phenomenon in nude mice implanted with
ABSTRACT Glucagon-like peptide 2 (GLP-2) is a 33-amino glucagonomas, and established that the effect was due to
acid peptide derived from the tissue-specific, post-transla-
GLP-2. Several subsequent studies (3–5) have suggested
that GLP-2 is a gut peptide with trophic actions that are
tional processing of the proglucagon gene expressed in the
highly specific for the GI tract. Moreover, GLP-2 is cur-
intestinal enteroendocrine L-cell. The primary stimulus for
rently being developed for therapeutic clinical use; it re-
GLP-2 secretion is nutrient intake, and involves direct lu- cently was approved for treatment of gastrointestinal dis-
minal stimulation of the L-cell as well as indirect enteroen- ease in adults. The aim of this review is to provide an
docrine and neural mechanisms. The biological activity of overview of the biological functions of GLP-2 and to high-
GLP-2 in circulation is regulated by the proteolytic cleavage light the gaps in our understanding of its physiologic rele-
of the N-terminus by dipeptidylpeptidase IV. Several stud- vance and mechanism of action, especially in the develop-
ies have shown that GLP-2 has specific trophic effects on ing neonate.
the small and large intestine, which are mediated by stim-
ulation of cell proliferation and inhibition of apoptosis and Gene Expression. Glucagon-like peptide 2, a product of the
proteolysis. GLP-2 also has been shown to suppress gas- proglucagon gene, and belongs to a superfamily of genes that
tric motility and acid secretion, increase hexose transport code for gastrointestinal regulatory peptides including secretin,
activity and suppress food intake, specifically when infused glucose-dependent insulinotrophic peptide (GIP) and vasoac-
centrally. The actions of GLP-2 are mediated by a G-pro-
tive intestinal peptide. In the intestine, GLP-2 is produced as
a 33-amino acid peptide derived from post-translational pro-
tein–linked, membrane receptor (GLP-2R) that is localized
cessing of the proglucagon polypeptide in enteroendocrine “L”
largely to the gastrointestinal tract, but also is found in the
cells, which are located predominantly in the distal small
brain. The secretion of GLP-2 and expression of the intestine and colon (5,6). There is relatively high homology
GLP-2R are present in the late gestation fetus. However, (87–97%) in the GLP-2 peptide sequence among the species
the developing intestine does not become responsive to reported, including humans, pigs, cows and rats. Other prod-
the trophic effect of GLP-2 until after birth. Based on its ucts of the proglucagon gene in the intestine and brain are
efficacy in preventing atrophy and stimulating growth in the glucagon-like peptide 1, glicentin and oxyntomodulin. In the
neonatal gut, GLP-2 may be a promising therapeutic adju- A-cells of the pancreatic islets, glucagon is the major post-
vant for treatment of infants with compromised gut func- translational product of the gene. It is noteworthy that GLP-2
tion. J. Nutr. 131: 709 –712, 2001. is produced from proglucagon expressed in regions of the brain
(4). The tissue-specific processing of the proglucagon-derived
peptides is explained largely by the differential expression of
KEY WORDS: ● cell proliferation ● apoptosis ● gut hormone specific prohormone convertases in the intestinal L-cells and
● enteral nutrition ● neonate pancreatic A-cells. Much more is known about the molecular
regulation of proglucagon expression in pancreatic A-cells
than in the enteroendocrine L-cells (7). However, studies in
1
Supported by The Danish Agricultural and Veterinary Research Council rodents have demonstrated the presence of intestinal proglu-
(Program 9702803), National Institutes of Health grant RO1-HD33920 (D.G.B.)
and by federal funds from the U.S. Department of Agriculture, Agricultural Re-
search Service under Cooperative Agreement Number 58 – 6250-6 – 001.
2 5
The contents of this publication do not necessarily reflect the views or policies Abbreviations used: BHK, baby hamster kidney; DPP-IV, dipeptidylpepti-
of the U.S. Department of Agriculture, nor does mention of trade names, commercial dase IV; GI, gastrointestinal; GIP, glucose-dependent insulinotrophic peptide;
products, or organizations imply endorsement by the U.S. Government. GLP-2, glucagon-like peptide 2; GLP-2R, GLP-2 receptor; GRP, gastrin-releasing
3
Manuscript received 13 December 2000. peptide; MAP kinase, mitogen-activated protein kinase; PI-3, phosphatidylinositol
4
To whom correspondence should be addressed. 3 kinase; PKA, protein kinase A; RT-PCR, reverse transcriptase-polymerase chain
E-mail: dburrin@bcm.tmc.edu. reaction; SCFA, short-chain fatty acids.

0022-3166/01 $3.00 © 2001 American Society for Nutritional Sciences.

709
710 BURRIN ET AL.

cagon expression during fetal and early neonatal development also be regulated developmentally, based on evidence that
(8). Other studies have identified specific transcription factors GLP-2 is present at low levels in fetal plasma and the fact that
and aspects of the promoter sequence that are important for the concentration increases progressively during early neona-
the early development and tissue-specific regulation of proglu- tal development (8,18). It is interesting to note that the rate
cagon gene expression (9,10). of intestinal growth and its trophic response to enteral nutri-
tion also increase during early postnatal development, raising
Secretion. Many of the factors that regulate the secretion of the possibility that GLP-2 secretion may be involved. The
GLP-2 can be inferred from our understanding of GLP-1 marked increases in the proportion of dietary carbohydrate and
secretion; both peptides appear to be secreted in parallel and fiber associated with weaning may upregulate GLP-2 secretion
are derived from the same gene, precursor peptide and cell and partially mediate the increased gut growth and cell pro-
type. The primary stimulus for GLP-2 secretion is enteral liferation observed during this developmental transition.
nutrient intake, especially carbohydrate and lipid (11) (Fig.
1). In adult humans after an oral feeding, there is a biphasic Biological Activity and Metabolism. An important regu-
increase in the circulating GLP-2 concentration; a rapid initial latory aspect of GLP-2 activity and metabolism is its rapid rate

Downloaded from https://academic.oup.com/jn/article-abstract/131/3/709/4687255 by guest on 05 June 2020


increase occurs within 15 min, followed by a second increase of degradation and renal clearance once secreted from the
after ⬃1 h. Our studies in neonatal animals have also shown intestinal L-cell (19). GLP-2 is secreted as a 33-amino acid
that the circulating GLP-2 concentration increases approxi- peptide, but is rapidly degraded at an N-terminus site to GLP-2
mately fourfold within 1 h after an oral feeding and is posi- [3–33] in circulation, in large part, by dipeptidylpeptidase IV
tively correlated with the level of enteral intake (12,13). It is (DPP-IV). This results in a relatively short half-life (7 min) for
remarkable that although intestinal secretions of GLP-1 and full-length GLP-2 [1–33] compared with the truncated GLP-2
GLP-2 are stimulated by a meal, the pancreatic secretion of [3–33] form (27 min) (5). The truncated GLP-2 [3–33] is
glucagon is effectively suppressed, a fact that highlights the believed to be largely biologically inactive (20). To prolong its
tissue-specific regulation of proglucagon processing and secre- half-life and enhance the biological activity of GLP-2, the
tion. Although direct exposure of the L-cell to luminal nutri- exquisite sensitivity to DPP-IV has been exploited experimen-
ents stimulates GLP-2 secretion, the rapid increase in the tally using a synthetic GLP-2 peptide analog h[Gly2]-GLP-2,
circulating concentration after feeding, coupled with the with an N-terminus substitution of glycine for alanine (3,4)
largely distal localization of these cells, suggests the involve- and specific inhibitors of DPP-IV activity, such as valine-
ment of other indirect mechanisms. Indeed, evidence suggests pyrrolidide (20), both of which increase the potency of GLP-2.
that nutrient-mediated secretion of GLP-2 involves both en- The potency of GLP-2 is also greater in mice than in rats due
docrine stimulation, mediated by GIP released from enteroen- to the species differences in DPP-IV activity. It is important to
docrine K-cells, and neural reflexes involving gastrin-releasing note that the DPP IV enzyme activity is relatively abundant in
peptide (GRP) (14). Moreover, there is evidence that short- the intestinal mucosa and thus may have an important influ-
chain fatty acids (SCFA) stimulate ileal proglucagon expres- ence on the biological activity of GLP-2 even before it reaches
sion and GLP-2 secretion, and thus provide a partial explana- the systemic circulation (5). Furthermore, the intestinal ac-
tion for the intestinal trophic effect of SCFA infusion and tivity of DPP IV is higher in neonatal than adult animals, and
dietary fiber consumption (15,16). The stimulation of SCFA higher in the distal than proximal gut; however, it is not
production arising from fermentation of malabsorbed dietary known whether this alters the biological effect of GLP-2 in
carbohydrate may be a triggering mechanism for GLP-2 secre- neonates.
tion, and hence may act as an important trophic signal in
intestinal adaptation after massive small bowel resection. Con- Physiologic and Metabolic Effects. The peptide sequence
sistent with this concept are study findings indicating that the of GLP-2 was deduced initially after the proglucagon gene
resection of the distal small bowel and colon significantly sequence was reported by Bell and others (21). However, the
diminishes GLP-2 secretion due to loss of GLP-2–producing biological function of GLP-2 was virtually unknown until the
tissue and leads to intestinal failure (17). GLP-2 secretion may seminal studies by Drucker and others (2) revealed the marked
intestinal trophic effects of GLP-2. Their initial report and a
series of subsequent studies showed that chronic treatment
with GLP-2 has potent and specific trophic effects on the GI
tract that are characterized grossly by increased tissue mass and
mucosal thickness and morphologically by increased villous
length and crypt depth. In vivo studies in humans and pigs
have demonstrated that GLP-2 infusion acutely inhibits gas-
tric secretion and motility, an effect that is common among
the other peptides secreted from the intestinal L-cell, namely
GLP-1 and PYY (5). As such, GLP-2 may be implicated
together with these peptides in the “ileal brake” phenomenon
(6).
There are conflicting reports concerning the effects of
GLP-2 treatment on intestinal digestive enzyme and nutrient
transport. In some cases, GLP-2 has been found to increase the
activities and expression of hydrolases, such as sucrase-isomal-
tase, maltase-glucoamylase, lactase and aminopeptidase N
(18,22,23). However, some studies in rodents and neonatal
FIGURE 1 Overview of factors involved in the regulation of GLP-2 pigs indicate that GLP-2 enhances intestinal hexose transport
secretion. Abbreviations: GLP-2, glucagon-like peptide 2; GIP, glu- by modulating the activity and localization of GLUT2 and
cose-dependent insulinotrophic peptide; GRP, gastrin-releasing pep- SGLT1 (24,25,26), whereas others found expression of these
tide; SCFA, short-chain fatty acids. transporters to be decreased by GLP-2 (22). Furthermore,
GLP-2 ACTION IN THE GUT 711

studies show that GLP-2 increases amino acid transport from cells that are predominantly localized in the distal regions
(25,27). Yet, in vivo kinetics studies suggest only modest of the GI tract, yet its effects occur throughout the GI tract.
increases in nutrient absorption in GLP-2–treated mice (22). Given the limited and, to some extent, conflicting recent
In addition to the reported effects of GLP-2 on intestinal findings concerning the localization of the GLP-2R, it is
absorptive function, the increased mucosal growth and villous perhaps not surprising that our knowledge of its intracellu-
surface area that occur with GLP-2 treatment may have an lar signaling mechanisms is also in its infancy. On the basis
important role in gut barrier function. Indeed, GLP-2 has been of the known sequence information and initial biochemical
shown to reduce the permeability of the intestine to macro- characterization, the GLP-2R is a G-protein–linked mem-
molecules, decrease bacteria translocation and suppress the brane receptor, which activates a cyclic AMP, protein ki-
local expression of proinflammatory cytokines (4,28,29). nase A (PKA)-dependent pathway when transfected into
GLP-2 may have limited effects on systemic or whole-body baby hamster kidney (BHK) fibroblasts (35). However, the
metabolism, given evidence that its actions are confined coupling of GLP-2R activation with downstream cellular
largely to the GI tract. Indeed, there is no reported evidence events that mediate increased proliferation and cell survival
that systemic GLP-2 administration affects food intake, has not been established. The studies with transfected BHK

Downloaded from https://academic.oup.com/jn/article-abstract/131/3/709/4687255 by guest on 05 June 2020


growth rate or metabolism. Yet, it is interesting that central fibroblasts suggest that GLP-2 actions are not mediated by
administration of GLP-2 into the lateral cerebral ventricle PKA, phosphatidylinostiol 3 kinase (PI-3 kinase) or mito-
suppresses food intake in rats and thus, like GLP-1, it may be gen-activated protein (MAP) kinase pathways. However,
implicated in appetite regulation (30). We have recently the GLP-2– dependent stimulation of thymidine uptake in
found that GLP-2 stimulates intestinal protein anabolism in Caco-2 cells was suppressed in a dose-dependent manner by
neonatal piglets receiving total parenteral nutrition (TPN) by inhibitors of both the PI 3-kinase and MAP-kinase path-
suppressing proteolysis, whereas protein synthesis was unaf- ways (33). Thus, it is uncertain whether these signaling
fected (31). However, whether GLP-2 affects any aspect of pathways that are apparently activated by GLP-2 in Caco-2
whole-body protein, carbohydrate or lipid metabolism remains cells, but not in transfected BHK fibroblasts, are indeed
to be determined. Thus, from a physiologic perspective, the present in normal intestinal epithelial cells. This again
general picture that has emerged suggests that GLP-2 acts to raises the critical question of the existence of a secondary
slow the ingestion and transit of food through the GI tract, signal that may act via a heterologous, paracrine cellular
while increasing the absorption of nutrients from the small mechanism between enteroendocrine and other intestinal
intestine. epithelial cells.
An aspect of GLP-2 function in which we have been
Cellular Actions of GLP-2 Receptor. The cellular actions especially interested is the ontogeny of GLP-2R expression
of GLP-2 are mediated initially by binding to the GLP-2 and the onset of GLP-2 responsiveness during early mam-
receptor (GLP-2R). The recent cloning of cDNAs encoding malian development. A recent report in rodents (8) and our
the human and rat GLP-2R indicate that it is a G-protein– studies in pigs (unpublished results) indicate that the
linked, seven- transmembrane-domain receptor with homol- GLP-2R is expressed during fetal and neonatal develop-
ogy to glucagon, GLP-1 and GIP receptors (4). The human ment. These findings are consistent with evidence of tro-
GLP-2R is encoded by a single gene localized to chromosome phic and functional effects of GLP-2 in suckling rat pups
17p13.3. The initial cloning of the GLP-2R was performed and in TPN-fed piglets delivered preterm and at term.
using hypothalamic cDNA libraries; however, efforts to quan- Interestingly, however, in fetuses given GLP-2 infusions in
tify expression based on a ribonuclease protection assay de- utero for 6 d, there was no stimulation of intestinal growth
tected mRNA transcripts primarily in the GI tract. A recent or enhanced development of digestive hydrolase expression,
report has confirmed the tissue-specific distribution of the despite the presence of the GLP-2R transcript in the GI
GLP-2R mRNA, using Northern analysis and reverse tran- tract (18). In our neonatal piglets studies (31), we found
scriptase-polymerase chain reaction (RT-PCR), demonstrat- that although GLP-2 potently blocked the mucosal villous
ing expression in the gut, brain and the lung (32). This report atrophy normally induced by TPN, this was mediated by
has shed light on the more critical issue of localization within suppression of apoptosis and proteolysis. In contrast, the
the GI tract using immunohistochemistry, and provides evi- intestinal trophic effect of enteral nutrition was mediated
dence of GLP-2R immunopositivity localized to a limited by a suppression of apoptosis and a stimulation of cell
number of enteroendocrine cells within the stomach and the proliferation and protein synthesis. Thus, although it is
large intestine. That finding is supported by the fact that clear that restoring the circulating GLP-2 concentration to
RT-PCR analysis failed to detect GLP-2R transcripts among supraphysiologic levels can maintain near-normal intestinal
several intestinal epithelial cells lines studied. In contrast, growth in the absence of any enteral nutrient stimulus, the
another recent report demonstrated that GLP-2 treatment trophic effect was mechanistically different than that of
stimulated proliferation of colonic epithelial (Caco-2) cells, enteral nutrition. This finding raises a critical, yet unre-
albeit at concentrations that were ⬃100-fold greater than the solved, question regarding the physiologic importance of
circulating concentration in plasma (33). Yet another group GLP-2, given that most of the studies reported have admin-
istered supraphysiologic doses of GLP-2. The development
found that injection of 125I-GLP-2 into rats resulted in specific
of experimental approaches to block endogenous GLP-2
binding of labeled GLP-2 localized diffusely along the villous
action via immunoneutralization, peptide antagonist and
epithelium (34). Thus, the definitive cellular and regional
targeted disruption of the GLP-2R gene should provide
localization of the GLP-2R within the GI tract remains to be
answers to this question. However, it appears that during
established. However, if indeed the GLP-2R is localized to fetal life, GLP-2 is not essential for intestinal development,
enteroendocrine cells, this implies that there is a secondary based on recent evidence from newborn Pax6 mutant mice
signal or signals that mediate the biological action on sur- with deficient intestinal endocrine cell development and
rounding epithelial cells via a paracrine mechanism). An proglucagon expression (10).
additional interpretation of these recent reports is that GLP-2
clearly acts by an endocrine mechanism because it is secreted Therapeutic Potential. Since the original report of the
712 BURRIN ET AL.

intestinotrophic actions of GLP-2 by Drucker and col- (1999) Diabetic intestinal growth adaptation and glucagon-like peptide 2 in the
rat: effects of dietary fibre. Gut 45: 672– 678.
leagues (2), a number of studies have demonstrated the 17. Jeppesen, P. B., Hartmann, H., Thulesen, J., Hansen, B. S., Poulsen,
therapeutic efficacy of GLP-2 and the protease-resistant S. S., Holst, J. J. & Mortensen, P. B. (2000) Elevated plasma glucagon-like
h[Gly2]-GLP-2 analog in several models of intestinal dys- peptide 1 and 2 concentrations in ileum-resected short bowel patients with a
preserved colon. Gut 47: 370 –376.
function, injury and insufficiency, including TPN (31,36), 18. Petersen, Y. M., Hartmann, B., Schmidt, M. H., Holst, J. J. & Sangild, P. T.
massive small bowel resection (37), colitis and nonsteroidal (2000) Exogenous glucagon-like peptide 2 has a limited effect on mucosal
anti-inflammatory drug–induced enteritis (4), inflammatory growth and enzyme activity in the fetus when compared to the neonate. Gastro-
bowel syndrome (29), ischemic bowel (38) and chemother- enterology 118: A561 (abs.).
19. Tavares, W., Drucker, D. J. & Brubaker, P. L. (2000) Enzymatic- and
apeutic injury (39). The suppression of intestinal cytokine renal-dependent catabolism of the intestinotropic hormone glucagon-like pep-
expression (3,29) and proteolytic activity (31) may be key tide-2 in rats. Am. J. Physiol. 278: E134 –EE139.
cellular mechanisms whereby GLP-2 ameliorates gut injury 20. Hartmann, B., Thulesen, J., Kissow, H., Thulesen, S., Orskov, C., Ropke,
C., Poulsen, S. S. & Holst, J. J. (2000) Dipeptidyl peptidase IV inhibition
and inflammation. The fact that GLP-2 effectively stimu- enhances the intestinotrophic effect of glucagon-like peptide-2 in rats and mice.
lates intestinal growth, while slowing proximal bowel mo- Endocrinology 141: 4013– 4020.
tility and secretion, makes it a promising therapeutic option 21. Bell, G. I., Sanchez-Pescador, R., Laybourn, P. J. & Najarian, R. C.

Downloaded from https://academic.oup.com/jn/article-abstract/131/3/709/4687255 by guest on 05 June 2020


(1983) Exon duplication and divergence in the human preproglucagon gene.
for patients with short-bowel syndrome. In fact, the pro- Nature (Lond.) 304: 368 –371.
tease-resistant GLP-2 analog, h[Gly2]-GLP-2, also referred 22. Brubaker, P. L., Izzo, A., Hill, M. & Drucker, D. J. (1997) Intestinal
to as ALX-0600, was recently granted orphan drug status by function in mice with small bowel growth induced by glucagon-like peptide-2.
Am. J. Physiol. 272: E1050 –E1058.
the Food and Drug Administration and is in Phase II 23. Kitchen, P. A., Fitzgerald, A. J., Goodlad, R. A., Barley, N. F., Ghatei,
clinical trial for treatment of short-bowel syndrome in M. A., Legon, S., Bloom, S. R., Price, A., Walters, J.R.F. & Forbes, A. (2000)
adults. Moreover, in a recent study in short-bowel patients, Glucagon-like peptide-2 increases sucrase-isomaltase but not caudal-related
homeobox protein-2 gene expression. Am. J. Physiol. 278: G425–G428.
GLP-2 treatment increased nutrient absorption and weight 24. Buddington, R. K., Elnif, J. & Sangild, P. T. (2000) Glucagon-like
gain (40). Thus, provided that GLP-2 treatment is safe, peptide 2 stimulates intestinal growth and nutrient absorption in newborn pigs on
other clinical applications may be anticipated, including parenteral nutrition. FASEB J. 14: A211 (abs.).
the treatment of infants with compromised intestinal 25. Kato, Y., Yu, D. & Schwartz, M. Z. (1999) Glucagon-like peptide-2
enhances small intestinal absorptive function and mucosal mass in vivo. J. Pe-
function. diatr. Surg. 34: 18 –21.
26. Cheeseman, C. I. (1997) Upregulation of SGLT-1 transport activity in
rat jejunum induced by GLP-2 infusion in vivo. Am. J. Physiol. 273: R1965–R1971.
ACKNOWLEDGMENTS 27. Sangild, P. T., Buddington, R. K., Elnif, J., Burrin, D. G., Schmidt,
M. H., Petersen, Y. M., Holst, J. J., Hartmann, B. & Malo, C. C. (2000) The
stimulating effect of glucagon-like peptide 2 (GLP-2) on intestinal nutrient
The authors would like to thank Jane Schoppe for assistance in absorption in neonates is reduced after premature birth. Gastroenterology
preparation of the manuscript and Leslie Loddeke for editorial assis- 118: A561 (abs.).
tance. 28. Benjamin, M. A., McKay, D. M., Yang, P.-C., Cameron, H. & Perdue,
M. H. (2000) Glucagon-like peptide-2 enhances intestinal epithelial barrier
function of both transcellular and paracellular pathways in the mouse. Gut 47:
112–119.
LITERATURE CITED 29. Alavi, K., Schwartz, M. Z., Palazzo, J. P. & Prasad, R. (2000) Treatment
of inflammatory bowel disease in a rodent model with the intestinal growth factor
1. Gleeson, M. H., Bloom, S. R., Polak, J. M., Henry, K. & Dowling, R. H. glucagon-like peptide-2. J. Pediatr. Surg. 35: 847– 851.
(1971) Endocrine tumor in kidney affecting small bowel structure, motility, and 30. Tang-Christensen, M., Larsen, P. J., Thulsen, J., Romer, J. & Vrang, N.
absorptive function. Gut. 12: 773–782. (2000) The proglucagon-derived peptide, glucagon-like peptide-2, is a
2. Drucker, D. J., Erlich, P., Asa, S. L. & Brubaker, P. L. (1996) Induction neurotransmitter involved in the regulation of food intake. Nat. Med. 6: 802– 807.
of intestinal epithelial proliferation by glucagon-like peptide 2. Proc. Natl. Acad. 31. Burrin, D. G., Stoll, B., Jiang, R., Petersen, Y., Elnif, J., Buddington, R. K.,
Sci. U.S.A. 93: 7911–7916. Schmidt, M., Holst, J. J., Hartmann, B. & Sangild, P. T. (2000) GLP-2 stimu-
3. Drucker, D. J. (1999) Glucagon-like peptide 2. Trends Endocrinol. lates intestinal growth by suppressing proteolysis and apoptosis in parenterally
Metab. 10: 153–156. fed premature piglets. Am. J. Physiol. 279: 1249 –1256.
4. Lovshin, J. & Drucker, D. J. (2000) New frontiers in the biology of 32. Yusta, G., Huang, L., Munroe, D., Wolff, G., Fantaske, R., Sharma, S.,
GLP-2. Regul. Pept. 90: 27–32. Demchyshyn, L., Asa, S. L. & Drucker, D. J. (2000) Enteroendocrine localiza-
5. Holst, J. J. (2000) Gut hormones as pharmaceuticals from enteroglu- tion of GLP-2 receptor expression in humans and rodents. Gastroenterology 119:
cagon to GLP-1 and GLP-2. Regul. Pept. 93: 45–51. 744 –755.
6. Holst, J. J. (1997) Enteroglucagon. Annu. Rev. Physiol. 59: 257–271. 33. Jasleen, J., Shimoda, N., Shen, R., Tavakkolizadeh, A., Whang, E. E.,
7. Drucker, D. J. (1998) Glucagon-like peptides. Diabetes 47: 159 –169. Jacobs, D. O., Zinner, M. J. & Ashley, S. W. (2000) Signaling mechanisms of
8. Lovshin, J., Yusta, B., Iliopoulos, I., Migirdicyan, A., Dableh, L., glucagon-like peptide 2-induced intestinal epithelial cell proliferation. J. Surg.
Brubaker, P. L. & Drucker, D. J. (2000) Ontogeny of the glucagon-like Res. 90: 13–18.
peptide-2 receptor axis in the developing rat intestine. Endocrinology 141: 34. Thulesen, J., Hartmann, B., Orskov, C., Jeppesen, P. B., Holst, J. J. &
4194 – 4201. Poulsen, S. S. (2000) Potential targets for glucagon-like peptide 2 (GLP-2) in the
9. Nian, M., Drucker D. J. & Irwin, D. (1999) Divergent regulation of human rat: distribution and binding of i.v. injected 125I-GLP-2. Peptides 21: 1511–1517.
and rat proglucagon gene promoters in vivo. Am. J. Physiol. 277: G829 –G837. 35. Yusta, B., Boushey, R. P. & Drucker, D. J. (2000) The glucagon-like
10. Hill, M. E., Asa, S. L. & Drucker, D. J. (1999) Essential requirement for peptide-2 receptor mediates direct inhibition of cellular apoptosis via a cAMP-
Pax6 in control of enteroendocrine proglucagon gene transcription. Mol. Endo- dependent protein kinase-independent pathway. J. Biol. Chem. 45:
crinol. 13: 1474 –1486. 35345–35352.
11. Xiao, Q., Boushey, R. P., Drucker, D. J. & Brubaker, P. L. (1999) 36. Chance, W. T., Foley-Nelson, T., Thomas, I. & Balasubramaniam, A.
Secretion of the intestinotrophic hormone glucagon-like peptide 2 is differentially (1997) Prevention of parenteral nutrition-induced gut hypoplasia by coinfusion
regulated by nutrients in humans. Gastroenterology 117: 99 –105. of glucagon-like peptide-2. Am. J. Physiol. 273: G559 –G563.
12. Van Goudoever, J. B., Stoll, B., Hartmann, B., Holst, J. J., Reeds, P. J. & 37. Scott, R. B., Kirk, D., MacNaughton, W. K. & Meddings, J. B. (1998)
Burrin, D. G. (2001) Secretion of trophic gut peptides is not different in bolus- GLP-2 augments the adaptive response to massive intestinal resection in rat.
and continuously fed piglets. J. Nutr. 131: 729 –732. Am. J. Physiol. 38: G911–G921.
13. Burrin, D. G., Stoll, B., Jiang, R., Chang, X., Hartmann, B., Holst, J. J., 38. Prasad, R., Alavi, K. & Schwartz, M. Z. (2000) Glucagon like peptide-2
Greeley, G. H., Jr. & Reeds, P. J. (2000) Minimal enteral nutrient requirements analogue enhances intestinal mucosal mass after ischemia and reperfusion.
for intestinal growth in neonatal piglets: how much is enough? Am. J. Clin. Nutr. J. Pediatr. Surg. 35: 357–359.
71: 1603–1610. 39. Tavakkolizadeh, A., Shen, R., Abraham, P., Kormi, N., Seifert, P., Edel-
14. Rocca, A. S. & Brubaker, P. L. (1999) Role of the vagus nerve in man, E. R., Jacobs, D. O., Zinner, M. J., Ashley, S. W. & Whang, E. E. (2000)
mediating proximal nutrient-induced glucagon-like peptide-1 secretion. Endocri- Glucagon-like peptide 2: a new treatment for chemotherapy-induced enteritis.
nology 140: 1687–1694. J. Surg. Res. 91: 77– 82.
15. Tappenden, K. A. & McBurney, M. I. (1998) Systemic short-chain fatty 40. Jeppesen, P. B., Hartmann, B., Thulesen, J., Graff, J., Lohmann, J., Hansen,
acids rapidly alter gastrointestinal structure, function, and expression of early B. S., Tofteng, F., Poulsen, S. S., Madsen, J. L., Holst, J. J. & Mortensen, P. B.
response genes. Dig. Dis. Sci. 43: 1526 –1536. (2001) Glucagon-like peptide 2 improves nutrient absorption and nutritional status
16. Thulesen, J., Hartmann, B., Nielsen, C., Holst, J. J. & Poulsen, S. S. in short-bowel patients with no colon. Gastroenterology 120: 806 – 815.

You might also like