Review Open Access
Copyright ©2006 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jul 14, 2006; 12(26): 4117-4129
Published online Jul 14, 2006. doi: 10.3748/wjg.v12.i26.4117
Gut hormones, and short bowel syndrome: The enigmatic role of glucagon-like peptide-2 in the regulation of intestinal adaptation
GR Martin, PL Beck, DL Sigalet, University of Calgary, Gastrointestinal Research Group, Calgary, Alberta T2N 4N1, Canada
Author contributions: All authors contributed equally to the work.
Supported by the Alberta Children’s Hospital Research Foundation
Correspondence to: Gary Martin, Department of Gastrointestinal Sciences, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW., Calgary, Alberta T2N 4N1, Canada. marting@ucalgary.ca
Telephone: +1-403-2203189 Fax: +1-403-2833840
Received: November 10, 2005
Revised: January 5, 2006
Accepted: January 14, 2006
Published online: July 14, 2006

Abstract

Short bowel syndrome (SBS) refers to the malabsorption of nutrients, water, and essential vitamins as a result of disease or surgical removal of parts of the small intestine. The most common reasons for removing part of the small intestine are due to surgical intervention for the treatment of either Crohn's disease or necrotizing enterocolitis. Intestinal adaptation following resection may take weeks to months to be achieved, thus nutritional support requires a variety of therapeutic measures, which include parenteral nutrition. Improper nutrition management can leave the SBS patient malnourished and/or dehydrated, which can be life threatening. The development of therapeutic strategies that reduce both the complications and medical costs associated with SBS/long-term parenteral nutrition while enhancing the intestinal adaptive response would be valuable.

Currently, therapeutic options available for the treatment of SBS are limited. There are many potential stimulators of intestinal adaptation including peptide hormones, growth factors, and neuronally-derived components. Glucagon-like peptide-2 (GLP-2) is one potential treatment for gastrointestinal disorders associated with insufficient mucosal function. A significant body of evidence demonstrates that GLP-2 is a trophic hormone that plays an important role in controlling intestinal adaptation. Recent data from clinical trials demonstrate that GLP-2 is safe, well-tolerated, and promotes intestinal growth in SBS patients. However, the mechanism of action and the localization of the glucagon-like peptide-2 receptor (GLP-2R) remains an enigma. This review summarizes the role of a number of mucosal-derived factors that might be involved with intestinal adaptation processes; however, this discussion primarily examines the physiology, mechanism of action, and utility of GLP-2 in the regulation of intestinal mucosal growth.

Key Words: Short bowel syndrome, Glucagon-like peptide-2, Epidermal growth factor, Insulin-like growth factor-I, Parenteral nutrition, Total parenteral nutrition, Intestinal adaptation, Intestinal mucosa, Gut hormones, Enteric nervous system



INTRODUCTION

Advances in surgical and medical care have made it possible for those with massive small bowel resections to survive. Unfortunately, a loss of greater than 50% of the small intestine often results in short bowel syndrome (SBS)[1-4]. SBS is characterized by the inefficient absorption of nutrients and fluids resulting in severe diarrhea, dehydration, electrolyte imbalances, nutrient deficiencies, weight loss, and frequently, a long-term dependence on parenteral nutrition[4-6]. In the pediatric population, total parenteral nutrition (TPN) maintenance-associated complications frequently result in sepsis, secondary liver failure, and high morbidity[5,7,8]. The development of therapeutic strategies that could reduce the consequences of SBS and long-term parenteral nutrition while enhancing the intestinal adaptive response would be valuable.

Numerous non-nutritional factors are potential stimulators of intestinal adaptation including peptide hormones, growth factors, and neuronally-derived components[9]. Some of the growth factors that have been shown to stimulate intestinal growth include glucagon-like peptide-2 (GLP-2), epidermal growth factor (EGF), growth hormone (GH), insulin-like growth factor-1 (IGF-1), neurotensin (NT), intestinal trefoil factor (ITF), transforming growth factor (TGF-α; TGF-β), and fibroblast growth factor (FGF)[10]. Table 1 lists some of the mucosa-derived humoral factors that have been shown to promote intestinal adaptation following intestinal resection.

Table 1 Growth factors as potential stimulators of intestinal adaptation.
Mucosal growth factors and SBS
Growth factorAmino acids /MWMain sourceGI receptor localization
GLP-233 aa/3.8 kDaIleal and colonic EE L-cells, pancreatic α cellsGLP-2R; EE cells, myenteric/submucosal neurons of ENS
EGF53 aa/6.4 kDaSubmandibular gland, duodenal Brunner’s gland, epithelial and Paneth cellsEGFR/HER/ErbB, HER2/Neu/ErbB2, HER3/ErbB3, HER4/ErbB4
KGF (FGF-7)194 aa/2.5 kDaFibroblast-like stromal cells in epithelial tissueKGFR; a/bFGFR epithelial cells
Hb-EGF86 aa/22 kDaSmooth muscle cellsActivate all as listed for the EGFR
GH191 aa/22 kDaLiverCRF type-1, PRL-R
TGF-α50-53 aa/6.2-6.4 kDaEpithelial cellsActivate all as listed for the EGFR
NT13 aa/?Ileum/colon >> proximal SB, EE N-cellsNTR1
PYY36 aa/42 kDaIleal and colonic EE L-cellsY1-R; EE cells, myenteric/submucosal neurons of ENS
GRP14 aa/?Neuroendocrine cells in the ENS, fibroblastsGRPR antrum, upper GI tract
CCK8, 33 aa/39 kDaDuodenal EE I-cellsCCKA >> CCKB upper GI tissue
myenteric/submucosal neurons of ENS
Gastrin17 aa/21 kDaGastric antrum G-cellsCCKB >> CCKA
34 aa/40 kDaPancreatic D-cellsupper GI tissue
IGF-I/III-70 aa/7.6 kDa,Proximal SB >> distal, liver,IGF-IR and IGF-IIR
II-67 aa/7.4 kDaFibroblastsepithelial cells, glial cells, fibroblasts

GLP-2 is a potential therapy currently under consideration for the treatment of gastrointestinal disorders that are associated with insufficient mucosal function. Evidence from both animal studies and clinical trials demonstrate that GLP-2 is a trophic hormone that plays an important role in the regulation of intestinal adaptation[11]. Under normal conditions, GLP-2 is secreted into the circulation in response to the ingestion of a mixed meal[12-15], particularly following one that contains carbohydrates, fatty acids, and fiber[16-18]. These studies have shown that following nutrient-induced release or exogenous administration, GLP-2 is involved in the regulation of cell proliferation, apoptosis, nutrient absorption, motility, as well as epithelial and intestinal permeability[15,19-25].

Initial pilot Phase II trials in SBS patients treated with teduglutide, a GLP-2 analog, observed that there were modest increases in fluid and nutrient absorption. Based upon the positive results of this trial, Phase III studies in adults with SBS were initiated. Subsequently, a recent article in GUT shows that teduglutide, using various dosing modalities over a 21 d experimental period, was safe, well tolerated, and promoted intestinal growth in SBS patients having either an end jejunostomy or a colon in continuity[26]. Moreover, teduglutide therapy in these SBS patients resulted in a significant increase in small intestinal villus height, crypt depth, and mitotic index. The authors state that the most significant clinical benefits of teduglutide in treating SBS patients were the reduction of intestinal wet weight excretion and in improving wet weight absorption[26]. There are several other clinical trials underway that are examining potential therapeutics for the treatment of SBS, unfortunately, Zorbitive® (hGH, glutamine) remains the only option presently available for prescriptive use.

This review summarizes the current understanding of a number of growth factors and their role in the treatment of SBS. As it is unlikely that there is just one specific growth factor responsible for maintaining mucosal growth, an important consideration is that there is likely some degree of regulation, transactivation, and crosstalk, between established stimulators of mucosal growth (i.e. epidermal growth factor, insulin-like growth factor-I, polyamines, GLP-2). Thus an understanding of the collective influence of these promoters of intestinal adaptation should not be dismissed. However, the primary focus of this discussion is to examine the physiology, mechanism of action, and utility of GLP-2 in the regulation of intestinal mucosal growth.

Discovery of GLP-2

The first indication that GLP-2 was trophic to the bowel was the observation that patients with glucagonomas had significantly elevated circulating enteroglucagon levels that was secondarily associated with intestinal mucosal hyperplasia[27,28]. The correlations between serum enteroglucagon levels and mucosal growth of the small intestine provided further evidence for the trophic effects of GLP-2[27]. A major breakthrough came in 1996 when it was shown that injection of proglucagon-producing tumors into nude mice resulted in significant mucosal hyperplasia[29]. Isolation of the various components of these proglucagon-producing tumors revealed that GLP-2 was the trophic factor responsible for the increased mucosal growth.

Additional evidence for intestinotrophic properties of GLP-2 came from studies in rats and mice in which GLP-2 administration induced intestinal hypertrophy i.e., significant increases in protein and DNA content, villus height, and bowel weight[21,25,30,31]. Moreover, GLP-2 treatment reduced mucosal atrophy associated with TPN administration and augmented the adaptive growth response following massive small bowel resection[19-21,32]. Our studies showed that in parenterally maintained animals (no luminal nutrition), infusion with GLP-2 stimulated intestinal adaptation following small bowel resection[18]. Without the infusion of GLP-2, intestinal adaptation did not occur. Similarly, the immunoneutralization of endogenous circulatory GLP-2 reduced the intestinal post-resection adaptive response in rats and rabbits[33].

What is GLP-2

GLP-2 is a member of the PACAP (Pituitary adenylate cyclase activating peptide)/Glucagon superfamily that includes the enteroglucagon/proglucagon-derived peptides (PGDP) that are produced in the L-cells of the small intestinal and colonic mucosa. The L-cells have been identified as the cellular origin of enteroglucagon (Figure 1)[24,34,35]. In 1983, the full sequence of hamster proglucagon, and later that year, the human sequence, were published revealing the sequences of GLP-1 and GLP-2[36,37]. GLP-2 is described as glucagon-like as it shares approximately 50% sequence homology with glucagon[37]. Sequencing of GLP-2 from human and pig intestine has confirmed the structure of GLP-2 as a 33 amino acid peptide that contains alanine at position 2 of the N-terminus (making it easily degraded by exopeptidases) and ends with a carboxy-terminal Asp residue[45]. GLP-2 and the other PGDPs are derived following the proteolytic cleavage and other enzymatic modifications of proglucagon in the organelles of the pancreatic α-cells and intestinal L-cells[38,39]. Interestingly, GLP-2 and the other products of post-translational processing (glicentin, GLP-1, oxyntomodulin) are produced in the intestinal L-cells, while glucagon and the major proglucagon fragment (MPGF) are produced in the α-cells of the pancreas[40]. They are formed from the same precursor molecule and therefore must be secreted in parallel. Differential expression of prohormone convertase (PC), the enzyme responsible for cleaving proglucagon into various hormonal products in the different cells, accounts for the different products. The PC2 enzyme, which has a greater abundance in the pancreatic α-cells, preferentially produces glucagon as its end product[41-43]. On the other hand, the PC1/3 enzyme, which is found in greater abundance in the intestinal L-cell, predominantly generates the glucagon-like peptides (GLP-1/2)[41]. Therefore, the differential processing of proglucagon in pancreatic α-cells by PC2, and by PC1/3 in the intestinal L-cell, might generate two unique hormonal products from the same precursor with each having a different physiological effect[43].

Figure 1
Figure 1 Potential pathways involved in the production, release, activity, and sites of action of GLP-2. GLP-2 produced by the L-cell is induced following the ingestion of a meal and can be induced either by direct stimulation of the L-cell, or potentially following the release of upstream neuronal /hormonal agonists. The GLP-2R has been localized to components of the enteric nervous system (ENS) and to regions of the brain, thus the growth-promoting effects associated with GLP-2 may involve interactions with the brain-gut axis. Potentially the ENS / vagal interface might be responsible for the early release of GLP-2.

Initially, the GLP-2 sequence was identified in all mammalian cDNA, but not in other species. This led researchers to theorize that GLP-2 was a late evolutionary addition. Subsequent studies have demonstrated that fish, chickens, and lizards generate GLP-2 in the gut as a result of tissue-specific alternative RNA splicing of proglucagon RNA transcript[44].

Site of GLP-2 production

The distal regions of the small bowel and/or the large intestine are necessary for GLP-2 production. This was demonstrated in a study in which patients with SBS, without a colon in continuity, had significant reductions in their post-prandial circulatory GLP-2. Subjects that had 140 cm or less of small bowel, with colon in continuity, exhibited a significant increase in their serum GLP-2 levels[46,47]. Conversely, fasting and post-prandial serum GLP-2 levels were elevated in SBS patients (colon intact, no ileum) compared with sex- and age-matched controls[46]. The locale of the GLP-2-containing tissue in the human intestine is debatable, though it was recently reported that the number of immunoreactive GLP-2 cells increase in a proximal to distal distribution in normal colonic tissue[48].

However, there is debate as to the contribution of different regions of the bowel to total GLP-2 production. A 90% intestinal resection, leaving only 10 cm of distal ileum, results in a significant elevation in serum GLP-2 levels in the rat[15]. These elevations in circulatory GLP-2 correlate with several measurable parameters of intestinal adaptation[18]. As the remainder of the small bowel had been removed, this suggests that the distal ileum and/or the colon are the major sites of the L-cells, the cells responsible for the production of GLP-2 (Figure 1). Others describe that in the rat, TNBS (trinitrobenzene sulphonic acid) -induced ileitis resulted in no change in the number of GLP-2 immunoreactive cells in the ileum, yet colonic GLP-2 immunoreactivity was increased (personal communication, Jennifer O’Hara and Dr. K.A. Sharkey).

Interestingly, in a rat SBS model in which only 20 cm of jejunum was anastomosed directly to the ascending colon[49] (i.e., removal of 80% of the distal small bowel including the ileum and cecum), circulatory GLP-2 levels were significantly increased in comparison to both controls and the 90% resection animals with ileum intact (Figure 2). This strongly suggests that the colon is an important source of GLP-2. These findings emphasize the importance of determining if the increase in serum GLP-2 levels are the result of either an increase in the L-cell population, or alternatively, as a consequence of augmented GLP-2 production by the L-cells. Our ongoing experiments suggest, at least in this rat model of SBS, that increased GLP-2 production is the result of an increase in the colonic L-cell population (unpublished observations).

Figure 2
Figure 2 An experimental rat model of SBS-induces changes in circulatory GLP-2 levels. The distal regions of the small bowel and/or the large intestine are necessary for GLP-2 production. In these experiments, the entire proximal small bowel was removed; therefore the distal ileum and/or the colon are likely the cells responsible for the production of GLP-2 as all other bowel tissue had been removed. Resected = 90% intestinal resection leaving a 10 cm ileal remnant. CEC = cecetomy + removal of all but 20 cm of jejunum. The remnant jejunum was anastomosed to the ascending colon. aP < 0.05 vs cecetomized animals; cP < 0.05 vs 90% proximal resected animals that were enterally-fed (10 cm remnant ileum).
GLP-2 receptor and activity

Currently, the localization of the glucagon-like-2 receptor (GLP-2R) remains an enigma. Conflicting data in the literature report the GLP-2R to be located on enteroendocrine cells[50], on non-epithelial elements such as neuronal cells[51], and subepithelial myofibroblasts (Figure 1)[52]. Moreover, the localization of the GLP-2R in the bowel might be region-specific. In a recent study by Orskov et al, GLP-2 receptors were found throughout the small and large bowel in humans, mice, marmoset, and rat[52]. Furthermore, the intestinal region that had the greatest GLP-2R immunoreactivity was the proximal small bowel. These data were confirmed by both immunohistochemistry and in situ hybridization that showed that the GLP-2R was localized to cells lying beneath the basal membrane of enterocytes[52].

The specific GLP-2 receptor has been isolated, sequenced, and found to be expressed in both the gut and in the hypothalamus (Figure 1)[53]. GLP-2 binds to a 7 transmembrane (7-TM) G protein-coupled receptor that is comprised of 550 amino acids. In humans, the expression of the GLP-2R is localized to specific enteroendocrine cells in the stomach as well as the small and large bowel[53-55]. Currently there have been only two human cervical carcinoma cell lines, including HeLa cells, in which GLP-2R mRNA transcripts have been detected[56]: the majority of the signal pathway determinations have been carried out in cells transfected with the GLP-2R[55]. The enteroendocrine cells that do express the GLP-2R have also been found to express other gut hormones such as glucagon-like peptide-1, glucose dependent insulinotropic peptide, peptide YY, and serotonin[50]. PACAP and VIP share the same VPAC1 receptor with equal affinity[34], thus it might be possible that other members of this family also share receptors.

There are a number of unanswered questions regarding the signaling mechanism involved in how GLP-2 stimulates mucosal growth. The initial studies on intracellular trafficking was done entirely in cell lines (BHK /COS) that had been transfected with the characterized GLP-2R[55,57]. These data showed that GLP-2/GLP-2R signaling involved classic G protein-coupled Gαs activation of adenylyl cyclase (AC), increased cAMP and PKA accumulation, and eventually ELK-1/c-fos/c-jun gene activation and increased cell proliferation (Figure 3A). Conversely, in epithelial cell lines devoid of the GLP-2R (as confirmed by both Western blot analysis and by the absence of mRNA transcripts), GLP-2 administration was still capable of increasing cell proliferation[56,58,59]. Thymidine uptake as a measure of an increased proliferation was significantly inhibited following pre-incubation with pertussis toxin (GαI inhibitor) or MAPK blockade, implying that GαI or EGFR/TyrK trafficking pathways are involved (Figure 3B). These studies also revealed that, in comparison with the transfected cell lines in which increases in cAMP correlated with augmented growth, there was a significant correlation between increased cell proliferation and decreased cAMP accumulation.

Figure 3
Figure 3 A: GLP-2 stimulation of the Gs-Coupled Receptor induced cell proliferation. The proposed GLP-2 /GLP-2R signaling pathway as characterized in cells that were transfected with the GLP-2R. See section on GLP-2 receptor-activity for description (pages 11-12); B: GLP-2 stimulation of the Gαi-Coupled Receptor induced cell proliferation. Possible trafficking pathways triggered by GLP-2 in cells that tested negative for the characterized GLP-2R. Potentially, GLP-2 induces divergent signaling pathways which results in an increase in the rate of cell proliferation. See section on GLP-2 receptor-activity for description (pages 11-12).

Furthermore, the possibility that there are multiple signaling mechanisms for GLP-2 is suggested by recent observations regarding the divergent effects of GLP-2 administration on epithelial cell proliferation. These effects were dependent on both the cell line and the dose of GLP-2 administered. Two independent studies demonstrated that administration of GLP-2 results in the inhibition of proliferation of non-transformed small intestinal epithelial cell lines (IEC-6, IEC-18), while increasing proliferation of cancer-derived human colonic epithelial cell lines (Caco-2, Colo 320)[59,60]. Moreover, there can be divergent effects of GLP-2 on epithelial cell migration, as GLP-2 significantly increased migration in the small intestine-derived IEC-6/IEC-18 cells, but had no effect on epithelial cell migration in the colonic Caco-2 or the Colo 320 cells[60].

An article recently published in Science may have uncovered another important component of the GLP-2 signaling pathway[61]. It is established that the intestinal epithelium undergoes constant renewal along the crypt-villus axis and that the β-catenin/T cell factor (TCF) transduction pathway is involved in these proliferation/differentiation events[62-64]. These data reveal that GLP-2 treatment of mice increases β-catenin levels suggesting that β-catenin might be a downstream mediator of GLP-2-induced crypt cell proliferation. In addition, the expression of a cDNA encoding human R-spondin1 (hRSpo1) in KI chimeras led to a substantial increase in the diameter, length, and weight of the small intestine, diffuse thickening of the mucosa, and crypt epithelial hyperplasia[61]. Their data showed that the administration of hRSpo1 potently affects proliferation of the intestinal epithelium through activation of β-catenin; the activation of this pathway by hRSpo1 indicates that the effect might be directly stimulated by receptor-mediated binding. It is interesting that the expression of hRSpo1 in human intestinal enteroendocrine cells, consistent with the role of this protein as a crypt cell mitogen, is similarly localized to the expression of GLP-2[61]. This is certainly an exciting find in regards to mapping out potential signaling pathways involved in GLP-2-induced intestinal growth.

Collectively, this dichotomy suggests that there are divergent signaling pathways involved in how GLP-2 stimulates cell proliferation. Secondly, it begs the question: are other uncharacterized GLP-2R’s or transactivation signaling mechanisms involved in the intestinotrophic effect of GLP-2 other than that initially described by Munroe et al[53]

GLP-2 interactions with the ENS/CNS

There is evidence that the GLP-2R is expressed on both murine enteric neurons and cerebral cortex astrocytes[51,65]. Bjerknes and Cheng showed by in situ hybridization that the GLP-2R is located on nonepithelial elements of the enteric nervous system. These results were further verified by reverse-transcriptase polymerase chain reaction (RT-PCR) for the GLP-2R mRNA[51]. Moreover, the GLP-2R was located on neurons and not on glial cells. These experiments demonstrate that GLP-2 is capable of inducing c-Fos activity in enteric neurons in as little as 7 min, and achieved maximal expression by 15 min. After 90 min, the increase in GLP-2-induced neuronal c-Fos activity was followed by increased c-Fos activity in the crypt cells of the jejunum and colon. These observations suggest that GLP-2 stimulates the GLP-2R leading to an increased c-Fos expression in myenteric neurons (Figure 4), which is secondarily followed by both an increase in c-Fos expression in the crypt cells and the stimulation of columnar crypt cell growth. Tetrodotoxin (TTX) pre-treatment abolished c-Fos expression in the crypt cells revealing that GLP-2 signaling in the bowel might be dependent on a transduction pathway requiring enteric neurons[51]. Potentially the intestinotrophic property associated with GLP-2 involves a second downstream mediator that is released upon GLP-2’s initial stimulation of the ENS (Figure 4).

Figure 4
Figure 4 GLP-2 stimulation of the enteric nervous system. GLP-2 binds to 7-TM G protein-coupled receptors localized to components of the ENS. This results in a conformational change in the GLP-2R that eventually activates c-fos expression via the Gαs, AC, c-AMP pathway. This suggests that a rise in circulatory GLP-2 levelsmight stimulate the GLP-2R leading to an increased stimulation of the myenteric and/or submucosal neurons. Potentially the trophic effects on the gut mucosa associated with GLP-2 are the result of a second downstream mediator, either a neurotransmitter or direct innervation that is activated upon GLP-2’s initial stimulation of the ENS localized GLP-2R.

The GLP-2R is also localized to regions of the hindbrain[66]. The dorsal medial hypothalamus (DMH) contains a dense plexus of GLP-2 immunoreactive fibers that express GLP-2 receptor mRNA (Figure 1). Though research regarding GLP-2’s involvement in neuronal activation is still in its infancy, it is interesting that GLP-2’s effects on enteric /vagal pathways are remarkably similar to those characterized for cholecystokinin[67,68]. This possibility has led to discussions regarding GLP-2 as a factor involved in the regulation of feeding. There is evidence that GLP-2 administration can inhibit feed intake[66,69,70] however, other investigators report no effect on appetite or energy intake at physiological concentrations of GLP-2[71]. This area of GLP-2 physiology requires further investigation.

These observations suggest a novel pathway of GLP-2 action as perhaps either a meal-stimulated endogenous release of GLP-2 from the L-cell, or potentially, the therapeutic administration of GLP-2, could activate enteric neuronal activity, resulting secondarily in the release of downstream mediators capable of inducing crypt cell proliferation (Figure 4). Currently, no studies have been published in which the expression of the GLP-2R in the human enteric nervous system was examined.

GLP-2 receptor antagonists

An obstacle when examining mechanistic pathways involved in GLP-2-induced small bowel adaptation is the lack of effective GLP-2R antagonists. One possibility would be to use the primary degradation product (GLP-23-33) to see if the interaction with the GLP-2R could reduce the adaptive response to intestinal resection. These results might be difficult to interpret as the treatment of mice with either GLP-21-33 or GLP-23-33 induces significant growth in both the small and large bowel, though the growth response induced by the degradation product is reduced[72]. Furthermore, functional studies and binding data indicate that GLP-23-33 acts as both a partial agonist and as a partial antagonist on the GLP-2R[72]. However, there is recent data that demonstrates GLP-23-33 antagonizes the growth-promoting activities of GLP-21-33in vitro and ex vivo[73]. Shin et al suggest that endogenous GLP-21-33 regulates the intestinal growth-promoting response via modulation of crypt-cell proliferation and villus apoptosis[73]. Collectively, these findings do not support the use of GLP-23-33 as a full antagonist of the GLP-2 receptor.

GLP-2 inactivation and metabolism

As mentioned earlier in this review, GLP-21-33 contains the amino acid alanine at position 2 thereby making it susceptible to degradation by the exopeptidase dipeptidyl peptidase IV (DPP-IV)[74]. For excellent reviews on the structural and functional properties of DPP-IV, see Mentlein or Lambeir[75,76]. The primary metabolism of GLP-21-33 by DPP-IV, which cleaves off two N-terminal amino acids, results in the formation of GLP-23-33, a potential receptor antagonist /agonist[72,73]. The elimination half-life of intact GLP-21-33 in humans is 7 min, whereas that of cleaved GLP-23-33 is 27 min[14], thus the cleavage of the NH2 terminal may potentially disrupt the signal transduction pathway and reduce GLP-2’s biological effectiveness. The administration of GLP-2 to DPP-IV deficient rats results in higher GLP-21-33 serum levels which correlate with a significant increase in small intestinal weight[74,77]. Similar increases in intestinal hypertrophy also occur in rats and mice administered a synthetic DPP-IV resistant GLP-2 analog (ALX-0600)[31,74] suggesting that the inactivation of GLP-21-33 by DPP-IV is a crucial limiting factor regarding GLP-2’s trophic effect on the gut mucosa. In humans, the replacement of alanine in position 2 of the GLP-2 peptide with glycine, has been shown to extend the half life from approximately 7 min, as for GLP-2, to 0.9-2.3 h.[26] Thus, DPP-IV resistant GLP-2 analogues might be useful therapeutically to help improve mucosal regeneration.

In contrast, there is evidence that DPP-IV may not be the only factor important in the degradation of GLP-2[78]. Geier et al did not detect an observable role for DPP-IV in the regulation of GLP-2 in DPP-IV-/- mice, as there were no measurable differences in GLP-2 levels or plasma DPP-IV-like activity between DPP-IV+/+ and DPP-IV-/- mice. Although DPP-IV-/- mice lack the DPP-IV gene, they do possess the genes for FAP, DP8, and DP9, and might have sufficient dipeptidyl peptidase levels to regulate GLP-2 cleavage. Moreover, the mice lacking the DPP-IV gene do not have either intrinsic resistance, or an enhanced rate of repair, in DSS-induced colitis. The authors concluded that residual dipeptidyl peptidase levels in the DPP-IV-/- mice results in insufficient GLP-21-33 bioavailability to protect these mice from DSS (dextran sodium sulfate) -induced damage The association between GLP-2 and DPP-IV family members will require further study.

Plasma concentrations of GLP-2 are not only influenced by the rate of secretion, but by the elimination or clearance rate. The kidney is important in the clearance of GLP-2 as there are increased circulatory levels in patients with renal failure[79,80]. In addition, experimental nephrectomy results in delayed clearance and increased circulating levels of GLP-2 in rats[81]. The increase in circulatory GLP-2 levels following nephrectomy is not surprising as it has been shown that the kidney is the site of greatest DPP-IV activity[75].

GLP-2 and intestinal inflammation

The treatment of animals with GLP-2 reduces harmful symptoms of inflammation associated with colitis as well as the mortality and the severity of both indomethacin and TNBS-induced enteritis[35,82,83]. Moreover, studies within our lab reveal that GLP-2 ameliorates inflammatory parameters associated with TNBS-induced ileitis (Gastroenterology abstract, T1530, 2005). These reductions in inflammation (decreased myeloperoxidase activity, interleukin-1β, and inducible nitric oxide synthase protein in mucosal tissue) 5 d post-TNBS occurred whether the GLP-2 was given simultaneously with TNBS, or 2 d post-TNBS injection. Others have observed that GLP-2 treatment initiated following chemotherapy enhances intestinal recovery[84]. A second report also showed the benefits of GLP-2 treatment as a preventative against cancer therapy-induced mucosal damage[85]. Teduglutide, a GLP-2 analog, given daily prior to whole body gamma-irradiation significantly increased crypt stem cell survival in mice when compared with vehicle-treated controls, though the protective effect was only observed when teduglutide was given prior to irradiation[85]. Collectively, these reports indicate that GLP-2 might be a useful therapeutic strategy in the treatment of diseases associated with intestinal inflammation.

There are conflicting reports regarding the effect of intestinal inflammation on GLP-2 production, particularly on the site of GLP-2 production. Intuitively, any situation that depletes the GLP-2-producing L-cells could potentially reduce the ability of GLP-2 to repair the mucosa. This could be the case in Crohn’s/colitis patients as inflammation of the intestine likely disrupts the L-cells. In a mouse-model of T-cell-induced inflammatory bowel disease (IBD), the amount of GLP-2 was significantly reduced in the colon[86], however, others report that TNBS-induced ileitis results in an increase in colonic GLP-2 immunoreactivity. (O’Hara and Sharkey, personal communications). Regarding the latter, perhaps an increase in local and circulatory GLP-2 is the result of IBD-induced diarrhea, thus increasing the nutrient load in the distal bowel, a well-described potent stimulus for inducing GLP-2 production[12,15,17]. Though speculative, perhaps increased growth and nutrient absorption in the proximal small intestine as a result of increased GLP-2 production would be successful in ameliorating inflammatory processes. Conversely, Schmidt et al found that both the fasting and meal-induced plasma levels of GLP-2 were not different between healthy controls and IBD patients[87]. This report suggests that L-cell secretion of GLP-2 is not altered in IBD. The discrepancies between these observations could be the result of model/species difference, or site-specific effects of inflammation in the bowel.

While GLP-2 is the most prominent enterotrophic peptide, there are a number of hormones that might be involved in the regulation of mucosal growth. These are briefly discussed as it is likely that processes involved in intestinal adaptation are regulated by a complex array of hormones, neuropeptides, and cytokines. A number of mucosal growth factors that might be useful for the treatment of short bowel syndrome are listed in Table 1.

Insulin-like growth factor-I

Another growth factor that might have a potential role in enhancing mucosal growth is insulin-like growth factor-I (IGF-I). IGF-I is produced mainly in the liver, but it is also synthesized locally in the gut[88]. There is evidence that IGF-I stimulates intestinal growth under experimental conditions of TPN therapy, intestinal resection, and radiation therapy[89]. In addition, the over-expression of the gene encoding for IGF-I is associated with increased small bowel length, small bowel weight, and crypt cell proliferation[90]. Conversely, others report that ileal IGF-I levels remain unchanged following intestinal resection[90,91].

Most of the circulating IGF-I is bound to IGF-binding proteins (IGFBPs) that potentially protect IGF-I from degradation thereby modulating the activity of IGF-I[92,93]. IGFBPs are capable of controlling the availability of circulating IGF-I to target tissues and are thus key components in the GH-IGF-I somatotropic axis. In humans, six IGFBPs have been identified and shown to modulate IGF-I actions differently[94]. Serum levels, jejunal tissue protein, and mRNA levels of IGF-I are reduced following small bowel resection[95]. One possibility is that there is a significant decrease in IGFBP-3 mRNA following intestinal resection, thus this rapid decrease in the IGFBP-3 could increase IGF-I bioavailability resulting in an enhanced adaptive response[96].

In SBS rats maintained on parenteral nutrition, IGF-I infusions induced jejunal hyperplasia and normalized ion transport[97]. Thus acute IGF-I treatments might prove to be beneficial in easing the transition from parental to enteral feeds thereby avoiding some of the long-term dilemmas associated with TPN therapy[98]. However, there is no significant evidence of serum or tissue concentration fluctuation of IGF-I following intestinal resection, therefore its utility as a treatment for SBS remains unclear.

Epidermal growth factor

Most of the EGF family of peptides (transforming growth factor-α, amphiregulin, heparin-binding EGF, epiregulin, betacellulin, neuregulin, neuregulin-2) are trophic to the gastrointestinal tract as they both stimulate crypt cell proliferation and suppress apoptosis[99]. There is evidence that EGF administration at the time of small bowel resection enhances the adaptive response. This appears to be accomplished by inducing mucosal hyperplasia and increasing nutrient absorption, while reducing intestinal permeability, weight loss, and apoptosis[49,100,101]. It is important to note that EGF therapy is most effective for intestinal adaptation when it is administered early following resection[102]. Very few studies have examined serum EGF levels following small bowel resection. Shin et al showed that small bowel resection does not change serum EGF levels, yet there was an increase in salivary and a decrease in urinary excretion of EGF[103]. Ileal epidermal growth factor receptor (EGFR) levels were significantly increased suggesting that the increased salivary EGF (significant endogenous source of EGF), enhanced EGF-R expression, and reduced urinary excretion of EGF, leads to an increase in ileal utilization of EGF following resection. Compared with control mice, the removal of the submandibular glands significantly attenuated the increase in villus height, total protein, and DNA content of the small bowel following massive small bowel resection[104].

It has also been suggested that intestinal adaptation following resection requires a functional EGFR[105]. Selective inhibition of the EGFR with an orogastric EGFR inhibitor (ZD1839, 50 mg/kg per day) results in impaired intestinal adaptation after small bowel resection[106,107]. There is also an impaired adaptive response to intestinal resection in waved-2 mice-mice that are predisposed to a naturally occurring mutation that results in a reduction in EGFR protein tyrosine kinase activity. In vitro studies demonstrate that serum from intestinal resection mice or rats is capable of stimulating a proliferative response in rat intestinal epithelial cells (IEC)[108]. This uncharacterized factor released into the serum of 7 d resected rats significantly increased EGF-R protein expression. Additionally, the proliferative response in IEC’s was abolished following the addition of a specific EGFR inhibitor. It would be intriguing to explore if there is cross talk between GLP-2, which is significantly increased following intestinal resection, and the EGFR signaling pathway. The basis for this speculation was discussed earlier and is depicted in Figure 3B.

PACAP and VIP

All but one of the members of the PACAP/glucagon superfamily, GIP, have been found as protein and/or mRNA in the brain and are thus classified as neuropeptides[34]. For an excellent review of this family of peptides, see Sherwood et al[34]. All of these peptides are found in the gut and signal through 7-TM G-protein coupled receptors. In humans, the present members include PACAP, glucagon, glucagon-like peptide-1, growth hormone releasing hormone, vasoactive intestinal peptide (VIP), secretin, glucose-dependent insulinotropic peptide (GIP), and GLP-2[34]. Both VIP and PACAP have a widespread distribution and are known to affect neural, circulatory, gastrointestinal, endocrine, and immune systems. There is evidence that VIP and PACAP administration reduces apoptosis and promotes the survival of neural cells by inducing bcl-2 activity. Increased bcl-2 activity is associated with an inhibition of caspase-3 activity and decreased cytoplasmic cytochrome C release[34]. Moreover, the BH4 domain of the pro-survival protein bcl-2 both binds to the C-terminus of Apaf-1 on caspase-9, thus inhibiting caspase-induced apoptosis, as well as directly or indirectly preventing the release of cytochrome C from the mitochondria[109,110]. Similar results were attained following the activation of GLP-2R signaling by GLP-2 administration in transfected cells treated with cyclohexamide or irinotecan. These data show that there was an inhibition of apoptosis, reduced caspase activation, and decreased mitochondrial cytochrome C release[57,84]. An additional parallel of these peptides is revealed following recent studies suggesting that GLP-2 is a vasodilator[111,112]. PACAP and VIP are both established as potent vasodilators[34].

At physiological concentrations, VIP and GLP-2 are both ineffective in stimulating cell growth directly in cell culture. Perhaps the intact ENS might need to be activated first as VIP receptors, and likely GLP-2 receptors, are located on enteric neurons[34,51]. It would be interesting to examine the growth-promoting potential of the ENS on epithelial cells in vivo.

There are conflicting results regarding plasma and tissue levels of VIP following small bowel resection and in studies examining SBS. These differences may be species-specific as following intestinal resection, dogs and pigs have a significant reduction in VIP levels, whereas in humans, there is either an elevation or no change in plasma VIP levels[113-116]. There is no evidence that VIP and PACAP administration is directly trophic for the small bowel thus a role for VIP in small bowel resection in the rat has not been described.

Peptide YY and Cholecystokinin

Further study is required when looking at the relationship of other co-localized mediators that are released from the L-cell. Peptide tyrosine tyrosine (PYY) is a 36 amino acid gastrointestinal peptide present in endocrine L-cells of the ileal, colonic, and rectal mucosa[117-121]. PYY is co-localized to some degree with GLP-1/GLP-2 in the L-cells[122,123], moreover, it generates many of the physiological actions that are also attributed to GLP-2. There are data that both GLP-2 and PYY: slow gastric emptying and intestinal transit; production rates in rats and humans are increased following intestinal resection, production rates are influenced by luminal nutrients in the hindgut; release might be influenced by a proximal gut hormone or perhaps a neuroendocrine mechanism[51,124-133]. In support of the latter, our results examining postprandial GLP-2 release[15], together with reports of the meal-stimulated PYY response[133], demonstrate that there are significant increases in the serum levels of both peptides in less than 15 min. Intestinal transit times are not that rapid, thus these early increases probably are not attributable to luminal nutrients in the distal intestine. Neuroendocrine factor induction of the early release of PYY was described in a study by Greeley et al[121]. They administered a meal to dogs and then diverted the entire meal out of the proximal intestine via the creation of a stoma. Thus, there was no possibility of nutrient stimulation of PYY containing L-cells in the distal gut. Interestingly, there was still significant increase in plasma PYY levels. In these same dogs, the removal of the ileum, colon, and rectum, resulted in no increase in plasma PYY levels in response to a meal. These findings replicate a human study previously discussed in this paper, but instead of PYY, GLP-2 levels in patients that received a colectomy were significantly reduced[47]. This is certainly supportive of either a blood-borne hormone or a neural pathway in the generation of the early release of PYY and most assuredly, GLP-2. Furthermore, we have observed that rats with 20 cm of jejunum diverted out an abdominal stoma (i.e. no possibility of meal contents directly contacting the distal bowel) still had a significant increase in post-prandial GLP-2 levels-unpublished data- (Figure 5).

Figure 5
Figure 5 Neural or humoral mechanism involvement in the early release of GLP-2. The delivery of a meal significantly increases serum GLP-2 levels in rats with discontinuous small bowel (ANOVA, P < 0.007). The disruption of the continuity of the bowel (stoma creation) did not inhibit the initial nutrient-stimulated GLP-2 response. Thus, GLP-2 production is increased at least in part by neural and/or hormonal mechanisms. Serum GLP-2 levels reported as ng/mL.

Several proximal gut hormones that are rapidly secreted following the ingestion of a meal have been tested (i.e., gastrin, glucose dependent insulinotropic peptide, secretin, neurotensin) and found to be ineffective in stimulating an increase in PYY[121]. Currently, no published literature has shown that these can stimulate GLP-2 release. One potential agonist, cholecystokinin (CCK), induces a dose-dependent release of PYY, and this CCK-induced PYY release, can be blocked by a CCK antagonist[121]. As CCK induces PYY production from L-cells, and having established that PYY and GLP-2 are co-localized, potentially, CCK might also be a foregut hormone responsible for stimulating the early release of GLP-2. It would be worthwhile to examine whether CCK also triggers GLP-2 secretion.

PYY administration has not shown that it is trophic to the gastrointestinal tract directly and it is unlikely that it is involved in the adaptive response following intestinal resection, though a reduction in intestinal transit may secondarily generate increased intestinal growth. These areas have not been explored. Both GLP-2 and PYY are inactivated by dipeptyl peptidases[75,76], thus additional studies of SBS should explore the relationship between serum and intestinal tissue levels of GLP-2, PYY, and DPP-IV.

Polyamines

Polyamines are present in prokaryotes, plants and animals. The polyamines putrescine and its derivatives spermidine and spermine are found in nearly all cells of higher eukaryotes[134,135]. The major role of polyamines in most cell types is to stimulate cell proliferation and thus is considered essential for life[135,136]. The inhibition of polyamine biosynthesis blocks cell growth[136,137]. The increase in intracellular polyamine levels and in the activity of ornithine decarboxylase (ODC), one of the rate-limiting enzymes in the strictly controlled polyamine biosynthetic pathway[138], are associated with rapid growth rates[139,140]. ODC activity in the rat small intestinal mucosa is increased following small bowel resection[134,138], parasitic or enteropathogenic bacteria-induced small intestinal inflammation or colitis[141,142], ischemia reperfusion[143], and following partial obstruction of the lumen. All of these instances are associated with an increase in mucosal growth. Of clinical relevance, mucosal ODC activity in the colon and rectum has been reported to be significantly higher in both ulcerative colitis and Crohn’s disease patients[144]. In addition, evidence shows that polyamines are protective of DNA during the S-phase of the growth cycle[135,145] and in part may explain the protective effects of GLP-2 following the administration of chemotherapeutic agents.

There is a rise in plasma enteroglucagon levels that precedes the activation of ODC[143]. Thus it could be speculated that an increase in GLP-2 might initiate the activation of ODC leading to the repair of injured intestinal mucosa. If so, it would not be unreasonable for the growth promoting and protective role of GLP-2 in the small bowel to act through an ODC-dependent formation of polyamines. Considering that the trophic effects of many growth-promoting gut hormones are blocked following ODC or polyamine synthesis inhibition[134,138], it would not be surprising if this pathway is involved as a downstream effector of GLP-2 induced intestinal growth.

Intestinal carcinogenesis

A great deal of attention has focused on the potential for cancerous growth attributable as a side effect following the application of growth factors. Recent results cautioning restraint regarding the utility of GLP-2 was indicated as GLP-2, and to a greater degree, a GLP-2 analog, promoted an increase in the growth of mucosal neoplasms in mice pre-induced to form colonic tumours[146]. Furthermore, there was a significant increase in tumour load in the mice treated with either GLP-2, or the synthetic Gly2-GLP-2 analogue. There is also evidence that GLP-2R mRNA transcripts can be detected in two human cervical carcinoma cell lines[56]. But does the application of growth factors such as GLP-2 or EGF always trigger neoplastic growth Could the application of growth factors prevent the development of intestinal neoplasms Perhaps the primary factor involved in neoplastic growth is not necessarily the addition of the growth factor, but rather a dysfunction in downstream receptor (i.e., mutation) or signal transduction pathways. For example, there is evidence that mutations in cytoplasmic signaling elements involved in the EGFR signaling cascade result in an alteration in signal transduction events that lead to changes in the downregulation/internalization of the EGFR[147]. Perhaps what occurs in established oncogenic cells is that a mutation in an element of the signaling cascade prevents GLP-2- or EGF-induced receptor downregulation. This theory is supported by recent work that shows that an oncogenic form of c-Cbl/c-Cbl, which stabilizes and ubiquitinates the cytoplasmic region of the EGFR, is able to prevent EGF-induced EGFR down regulation thus bypassing degradative pathways[148]. The consequences of this mutation are a bypass of receptor internalization and desensitization processes, an induction in the over expression of the EGFR, and finally, aberrant cell growth. A downstream defect in trafficking could potentially be the primary cause of neoplastic, abnormal cell growth, not necessarily the application of the growth factor.

In the case of the promotion of tumour size in mice following GLP-2 treatment as noted by Thulesen et al, or potentially as in the CaCO-2, T84, HeLa cells[56,58,59], what has occurred is that dysfunctional receptors and/or signal transduction pathways are already present within/on these cell types. As with any neoplastic condition, the application of growth factors is always a risk. Potentially the application of growth factors prior to the development of gastrointestinal neoplasms, such as during the early stages of Crohn’s disease or ulcerative colitis, would be beneficial in halting the progress of prolonged inflammation. It is well established that prolonged intestinal inflammation often secondarily leads to the development of mutations, cell dysfunction, and carcinogenic growth.

CONCLUSIONS

The trophic effect of GLP-2 on the intestine demonstrates that it is possible to modify or accelerate the process of intestinal adaptation, which would potentially make GLP-2 useful for the treatment of short bowel syndrome. Evidence shows that increases in GLP-2 production following intestinal resection are significantly correlated with intestinal adaptation. The association between an increase in nutrient malabsorption and GLP-2 production following intestinal resection has led to speculations that GLP-2 may act to control the nutrient absorptive capacity within the bowel. The increased availability of circulatory GLP-2 might then stimulate proximal mucosal growth resulting in enhanced nutrient absorptive capacity in the remaining intestinal remnant. Moreover, GLP-2 administration maintains epithelial barrier function and increases both crypt cell proliferation and weight gain in the absence of enteral nutrients. This supports the further development of GLP-2 as a therapeutic strategy that could enhance intestinal adaptation and reduce the consequences of parenteral feeding.

Although significant progress has been made towards elucidating the mechanisms mediating the trophic effects of GLP-2, several formidable challenges lie ahead. The biological and signaling role of the GLP-2R has not been fully defined, thus the mechanistic pathway (s) by which GLP-2 induces adaptive change in the small bowel presently remain elusive. Data showing that GLP-2 administration significantly increases cell proliferation in the absence of the characterized GLP-2R strongly suggest that GLP-2’s trophic effect might also include activation of either an uncharacterized GLP-2R or the transactivation of other receptor/signaling paradigms. Another key goal should be in determining the mechanism involved in the early release of GLP-2 following a meal or in malabsorptive states. Lastly, an emergent area of interest is the potential of GLP-2 to induce stimulation of the ENS as an intermediate in the regulation of intestinal growth. Regardless, there is a very significant and biologically important effect of GLP-2 on intestinal function and whole animal physiology that strongly supports further research into the use of GLP-2 as a therapy. The future use of GLP-2 therapy for the treatment of intestinal disorders is an enticing prospect; however, the promise of these initial results must be tempered as we await confirmatory data from clinical trials.

ACKNOWLEDGMENTS

In addition to the contribution by the co-authors, GR Martin is very grateful to Joseph Davison, Wallace MacNaughton, Keith Sharkey, and John Wallace for all their enthusiasm, guidance, and wisdom passed down over the past several years.

Footnotes

S- Editor Wang J L- Editor Karam SM E- Editor Bai SH

References
1.  Dowling RH, Booth CC. Structural and functional changes following small intestinal resection in the rat. Clin Sci. 1967;32:139-149.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Vanderhoof JA. Short bowel syndrome in children and small intestinal transplantation. Pediatr Clin North Am. 1996;43:533-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 82]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
3.  Williamson RC. Intestinal adaptation (first of two parts). Structural, functional and cytokinetic changes. N Engl J Med. 1978;298:1393-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 407]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
4.  Sigalet DL. Short bowel syndrome in infants and children: an overview. Semin Pediatr Surg. 2001;10:49-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 108]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
5.  Vanderhoof JA, Langnas AN, Pinch LW, Thompson JS, Kaufman SS. Short bowel syndrome. J Pediatr Gastroenterol Nutr. 1992;14:359-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 85]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
6.  Vanderhoof JA, Langnas AN. Short-bowel syndrome in children and adults. Gastroenterology. 1997;113:1767-1778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 249]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
7.  Meehan JJ, Georgeson KE. Prevention of liver failure in parenteral nutrition-dependent children with short bowel syndrome. J Pediatr Surg. 1997;32:473-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 61]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
8.  Briones ER, Iber FL. Liver and biliary tract changes and injury associated with total parenteral nutrition: pathogenesis and prevention. J Am Coll Nutr. 1995;14:219-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
9.  Baksheev L, Fuller PJ. Humoral factors in intestinal adaptation. Trends Endocrinol Metab. 2000;11:401-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
10.  Podolsky DK. Regulation of intestinal epithelial proliferation: a few answers, many questions. Am J Physiol. 1993;264:G179-G186.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Lovshin J, Drucker DJ. New frontiers in the biology of GLP-2. Regul Pept. 2000;90:27-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
12.  Xiao Q, Boushey RP, Drucker DJ, Brubaker PL. Secretion of the intestinotropic hormone glucagon-like peptide 2 is differentially regulated by nutrients in humans. Gastroenterology. 1999;117:99-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 155]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
13.  Brubaker PL, Crivici A, Izzo A, Ehrlich P, Tsai CH, Drucker DJ. Circulating and tissue forms of the intestinal growth factor, glucagon-like peptide-2. Endocrinology. 1997;138:4837-4843.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Hartmann B, Harr MB, Jeppesen PB, Wojdemann M, Deacon CF, Mortensen PB, Holst JJ. In vivo and in vitro degradation of glucagon-like peptide-2 in humans. J Clin Endocrinol Metab. 2000;85:2884-2888.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Martin GR, Wallace LE, Hartmann B, Holst JJ, Demchyshyn L, Toney K, Sigalet DL. Nutrient-stimulated GLP-2 release and crypt cell proliferation in experimental short bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2005;288:G431-G438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 99]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
16.  van Goudoever JB, Stoll B, Hartmann B, Holst JJ, Reeds PJ, Burrin DG. Secretion of trophic gut peptides is not different in bolus- and continuously fed piglets. J Nutr. 2001;131:729-732.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Burrin DG, Stoll B. Key nutrients and growth factors for the neonatal gastrointestinal tract. Clin Perinatol. 2002;29:65-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 76]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
18.  Martin GR, Wallace LE, Sigalet DL. Glucagon-like peptide-2 induces intestinal adaptation in parenterally fed rats with short bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2004;286:G964-G972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 109]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
19.  Burrin DG, Stoll B, Jiang R, Petersen Y, Elnif J, Buddington RK, Schmidt M, Holst JJ, Hartmann B, Sangild PT. GLP-2 stimulates intestinal growth in premature TPN-fed pigs by suppressing proteolysis and apoptosis. Am J Physiol Gastrointest Liver Physiol. 2000;279:G1249-G1256.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Burrin DG, Stoll B, Guan X, Cui L, Chang X, Holst JJ. Glucagon-like peptide 2 dose-dependently activates intestinal cell survival and proliferation in neonatal piglets. Endocrinology. 2005;146:22-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 116]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
21.  Sigalet DL, Martin GR. Hormonal therapy for short bowel syndrome. J Pediatr Surg. 2000;35:360-363; discussion 364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 33]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
22.  Scott RB, Kirk D, MacNaughton WK, Meddings JB. GLP-2 augments the adaptive response to massive intestinal resection in rat. Am J Physiol. 1998;275:G911-G921.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Warner BW. GLP-2 as therapy for the short-bowel syndrome. Gastroenterology. 2001;120:1041-1043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
24.  Drucker DJ. Biological actions and therapeutic potential of the glucagon-like peptides. Gastroenterology. 2002;122:531-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 387]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
25.  Tsai CH, Hill M, Asa SL, Brubaker PL, Drucker DJ. Intestinal growth-promoting properties of glucagon-like peptide-2 in mice. Am J Physiol. 1997;273:E77-E84.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Jeppesen PB, Sanguinetti EL, Buchman A, Howard L, Scolapio JS, Ziegler TR, Gregory J, Tappenden KA, Holst J, Mortensen PB. Teduglutide (ALX-0600), a dipeptidyl peptidase IV resistant glucagon-like peptide 2 analogue, improves intestinal function in short bowel syndrome patients. Gut. 2005;54:1224-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 326]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
27.  Bloom SR. An enteroglucagon tumour. Gut. 1972;13:520-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 166]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
28.  Gleeson MH, Bloom SR, Polak JM, Henry K, Dowling RH. Endocrine tumour in kidney affecting small bowel structure, motility, and absorptive function. Gut. 1971;12:773-782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 284]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
29.  Drucker DJ, Erlich P, Asa SL, Brubaker PL. Induction of intestinal epithelial proliferation by glucagon-like peptide 2. Proc Natl Acad Sci USA. 1996;93:7911-7916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 643]  [Cited by in F6Publishing: 670]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
30.  Brubaker PL, Izzo A, Hill M, Drucker DJ. Intestinal function in mice with small bowel growth induced by glucagon-like peptide-2. Am J Physiol. 1997;272:E1050-E1058.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Drucker DJ, DeForest L, Brubaker PL. Intestinal response to growth factors administered alone or in combination with human [Gly2]glucagon-like peptide 2. Am J Physiol. 1997;273:G1252-G1262.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Chance WT, Foley-Nelson T, Thomas I, Balasubramaniam A. Prevention of parenteral nutrition-induced gut hypoplasia by coinfusion of glucagon-like peptide-2. Am J Physiol. 1997;273:G559-G563.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Hartmann B, Thulesen J, Hare KJ, Kissow H, Orskov C, Poulsen SS, Holst JJ. Immunoneutralization of endogenous glucagon-like peptide-2 reduces adaptive intestinal growth in diabetic rats. Regul Pept. 2002;105:173-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 53]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
34.  Sherwood NM, Krueckl SL, McRory JE. The origin and function of the pituitary adenylate cyclase-activating polypeptide (PACAP)/glucagon superfamily. Endocr Rev. 2000;21:619-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 220]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
35.  Alavi K, Schwartz MZ, Palazzo JP, Prasad R. Treatment of inflammatory bowel disease in a rodent model with the intestinal growth factor glucagon-like peptide-2. J Pediatr Surg. 2000;35:847-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 78]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
36.  Bell GI, Sanchez-Pescador R, Laybourn PJ, Najarian RC. Exon duplication and divergence in the human preproglucagon gene. Nature. 1983;304:368-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 395]  [Cited by in F6Publishing: 415]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
37.  Bell GI, Santerre RF, Mullenbach GT. Hamster preproglucagon contains the sequence of glucagon and two related peptides. Nature. 1983;302:716-718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 411]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
38.  Orskov C, Holst JJ, Knuhtsen S, Baldissera FG, Poulsen SS, Nielsen OV. Glucagon-like peptides GLP-1 and GLP-2, predicted products of the glucagon gene, are secreted separately from pig small intestine but not pancreas. Endocrinology. 1986;119:1467-1475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 341]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
39.  Furuta M, Yano H, Zhou A, Rouillé Y, Holst JJ, Carroll R, Ravazzola M, Orci L, Furuta H, Steiner DF. Defective prohormone processing and altered pancreatic islet morphology in mice lacking active SPC2. Proc Natl Acad Sci USA. 1997;94:6646-6651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 348]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
40.  Holst JJ. Gut hormones as pharmaceuticals. From enteroglucagon to GLP-1 and GLP-2. Regul Pept. 2000;93:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Damholt AB, Buchan AM, Holst JJ, Kofod H. Proglucagon processing profile in canine L cells expressing endogenous prohormone convertase 1/3 and prohormone convertase 2. Endocrinology. 1999;140:4800-4808.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Dey A, Lipkind GM, Rouillé Y, Norrbom C, Stein J, Zhang C, Carroll R, Steiner DF. Significance of prohormone convertase 2, PC2, mediated initial cleavage at the proglucagon interdomain site, Lys70-Arg71, to generate glucagon. Endocrinology. 2005;146:713-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
43.  Webb GC, Dey A, Wang J, Stein J, Milewski M, Steiner DF. Altered proglucagon processing in an alpha-cell line derived from prohormone convertase 2 null mouse islets. J Biol Chem. 2004;279:31068-31075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
44.  Irwin DM, Wong J. Trout and chicken proglucagon: alternative splicing generates mRNA transcripts encoding glucagon-like peptide 2. Mol Endocrinol. 1995;9:267-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 36]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
45.  Irwin DM. Molecular evolution of proglucagon. Regul Pept. 2001;98:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
46.  Jeppesen PB, Hartmann B, Thulesen J, Hansen BS, Holst JJ, Poulsen SS, Mortensen PB. Elevated plasma glucagon-like peptide 1 and 2 concentrations in ileum resected short bowel patients with a preserved colon. Gut. 2000;47:370-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 155]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
47.  Jeppesen PB, Hartmann B, Hansen BS, Thulesen J, Holst JJ, Mortensen PB. Impaired meal stimulated glucagon-like peptide 2 response in ileal resected short bowel patients with intestinal failure. Gut. 1999;45:559-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 131]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
48.  Caddy G, Johnston C, Ardill J, Pogue K, Collins J, Gardiner K, Watson P. Distribution of glucagon-like peptide-2 in normal colonic tissue. Scand J Gastroenterol. 2003;38:798-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
49.  Sham J, Martin G, Meddings JB, Sigalet DL. Epidermal growth factor improves nutritional outcome in a rat model of short bowel syndrome. J Pediatr Surg. 2002;37:765-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
50.  Yusta B, Huang L, Munroe D, Wolff G, Fantaske R, Sharma S, Demchyshyn L, Asa SL, Drucker DJ. Enteroendocrine localization of GLP-2 receptor expression in humans and rodents. Gastroenterology. 2000;119:744-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 255]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
51.  Bjerknes M, Cheng H. Modulation of specific intestinal epithelial progenitors by enteric neurons. Proc Natl Acad Sci USA. 2001;98:12497-12502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 261]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
52.  Ørskov C, Hartmann B, Poulsen SS, Thulesen J, Hare KJ, Holst JJ. GLP-2 stimulates colonic growth via KGF, released by subepithelial myofibroblasts with GLP-2 receptors. Regul Pept. 2005;124:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 161]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
53.  Munroe DG, Gupta AK, Kooshesh F, Vyas TB, Rizkalla G, Wang H, Demchyshyn L, Yang ZJ, Kamboj RK, Chen H. Prototypic G protein-coupled receptor for the intestinotrophic factor glucagon-like peptide 2. Proc Natl Acad Sci USA. 1999;96:1569-1573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 257]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
54.  Lovshin J, Estall J, Yusta B, Brown TJ, Drucker DJ. Glucagon-like peptide (GLP)-2 action in the murine central nervous system is enhanced by elimination of GLP-1 receptor signaling. J Biol Chem. 2001;276:21489-21499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 91]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
55.  Yusta B, Somwar R, Wang F, Munroe D, Grinstein S, Klip A, Drucker DJ. Identification of glucagon-like peptide-2 (GLP-2)-activated signaling pathways in baby hamster kidney fibroblasts expressing the rat GLP-2 receptor. J Biol Chem. 1999;274:30459-30467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 80]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
56.  Koehler JA, Yusta B, Drucker DJ. The HeLa cell glucagon-like peptide-2 receptor is coupled to regulation of apoptosis and ERK1/2 activation through divergent signaling pathways. Mol Endocrinol. 2005;19:459-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
57.  Boushey RP, Yusta B, Drucker DJ. Glucagon-like peptide (GLP)-2 reduces chemotherapy-associated mortality and enhances cell survival in cells expressing a transfected GLP-2 receptor. Cancer Res. 2001;61:687-693.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Rocha FG, Shen KR, Jasleen J, Tavakkolizadeh A, Zinner MJ, Whang EE, Ashley SW. Glucagon-like peptide-2: divergent signaling pathways. J Surg Res. 2004;121:5-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
59.  Jasleen J, Ashley SW, Shimoda N, Zinner MJ, Whang EE. Glucagon-like peptide 2 stimulates intestinal epithelial proliferation in vitro. Dig Dis Sci. 2002;47:1135-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
60.  Bulut K, Meier JJ, Ansorge N, Felderbauer P, Schmitz F, Hoffmann P, Schmidt WE, Gallwitz B. Glucagon-like peptide 2 improves intestinal wound healing through induction of epithelial cell migration in vitro-evidence for a TGF--beta-mediated effect. Regul Pept. 2004;121:137-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
61.  Kim KA, Kakitani M, Zhao J, Oshima T, Tang T, Binnerts M, Liu Y, Boyle B, Park E, Emtage P. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science. 2005;309:1256-1259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 425]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
62.  Batlle E, Henderson JT, Beghtel H, van den Born MM, Sancho E, Huls G, Meeldijk J, Robertson J, van de Wetering M, Pawson T. Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell. 2002;111:251-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 857]  [Cited by in F6Publishing: 836]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
63.  Potten CS, Loeffler M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development. 1990;110:1001-1020.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Potten CS, Owen G, Roberts SA. The temporal and spatial changes in cell proliferation within the irradiated crypts of the murine small intestine. Int J Radiat Biol. 1990;57:185-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 99]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
65.  Velázquez E, Ruiz-Albusac JM, Blázquez E. Glucagon-like peptide-2 stimulates the proliferation of cultured rat astrocytes. Eur J Biochem. 2003;270:3001-3009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
66.  Tang-Christensen M, Larsen PJ, Thulesen J, Rømer J, Vrang N. The proglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitter involved in the regulation of food intake. Nat Med. 2000;6:802-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 198]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
67.  Sayegh AI, Ritter RC. Vagus nerve participates in CCK-induced Fos expression in hindbrain but not myenteric plexus. Brain Res. 2000;878:155-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
68.  Sayegh AI, Ritter RC. CCK-A receptor activation induces fos expression in myenteric neurons of rat small intestine. Regul Pept. 2000;88:75-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
69.  Larsen PJ, Vrang N, Orskov C, Hartmann B, Holst JJ, Tang-Christensen M. The central glucagon-like peptide-2 (GLP-2) containing pathway is involved in regulation of food intake in rats. Obesity Research. 2001;9:55S.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Tang-Christensen M, Vrang N, Hartmann B, Larsen LK, Orskov C, Holst JJ, Larsen PJ. The correlation between feeding status and the amount of proglucagon, GLP-1 and GLP-2 in the central nervous system of the rat. Diabetes. 2001;50:A373.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Sørensen LB, Flint A, Raben A, Hartmann B, Holst JJ, Astrup A. No effect of physiological concentrations of glucagon-like peptide-2 on appetite and energy intake in normal weight subjects. Int J Obes Relat Metab Disord. 2003;27:450-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
72.  Thulesen J, Knudsen LB, Hartmann B, Hastrup S, Kissow H, Jeppesen PB, Ørskov C, Holst JJ, Poulsen SS. The truncated metabolite GLP-2 (3-33) interacts with the GLP-2 receptor as a partial agonist. Regul Pept. 2002;103:9-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
73.  Shin ED, Estall JL, Izzo A, Drucker DJ, Brubaker PL. Mucosal adaptation to enteral nutrients is dependent on the physiologic actions of glucagon-like peptide-2 in mice. Gastroenterology. 2005;128:1340-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 105]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
74.  Drucker DJ, Shi Q, Crivici A, Sumner-Smith M, Tavares W, Hill M, DeForest L, Cooper S, Brubaker PL. Regulation of the biological activity of glucagon-like peptide 2 in vivo by dipeptidyl peptidase IV. Nat Biotechnol. 1997;15:673-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 199]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
75.  Mentlein R. Dipeptidyl-peptidase IV (CD26)--role in the inactivation of regulatory peptides. Regul Pept. 1999;85:9-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 994]  [Cited by in F6Publishing: 952]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
76.  Lambeir AM, Durinx C, Scharpé S, De Meester I. Dipeptidyl-peptidase IV from bench to bedside: an update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci. 2003;40:209-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 683]  [Cited by in F6Publishing: 685]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
77.  Drucker DJ, Boushey RP, Wang F, Hill ME, Brubaker PL, Yusta B. Biologic properties and therapeutic potential of glucagon-like peptide-2. JPEN J Parenter Enteral Nutr. 1999;23:S98-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
78.  Geier MS, Tenikoff D, Yazbeck R, McCaughan GW, Abbott CA, Howarth GS. Development and resolution of experimental colitis in mice with targeted deletion of dipeptidyl peptidase IV. J Cell Physiol. 2005;204:687-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
79.  Orskov C, Andreasen J, Holst JJ. All products of proglucagon are elevated in plasma from uremic patients. J Clin Endocrinol Metab. 1992;74:379-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
80.  Ruiz-Grande C, Pintado J, Alarcón C, Castilla C, Valverde I, López-Novoa JM. Renal catabolism of human glucagon-like peptides 1 and 2. Can J Physiol Pharmacol. 1990;68:1568-1573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 58]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
81.  Tavares W, Drucker DJ, Brubaker PL. Enzymatic- and renal-dependent catabolism of the intestinotropic hormone glucagon-like peptide-2 in rats. Am J Physiol Endocrinol Metab. 2000;278:E134-E139.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Boushey RP, Yusta B, Drucker DJ. Glucagon-like peptide 2 decreases mortality and reduces the severity of indomethacin-induced murine enteritis. Am J Physiol. 1999;277:E937-E947.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Drucker DJ, Yusta B, Boushey RP, DeForest L, Brubaker PL. Human [Gly2]GLP-2 reduces the severity of colonic injury in a murine model of experimental colitis. Am J Physiol. 1999;276:G79-G91.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Tavakkolizadeh A, Shen R, Abraham P, Kormi N, Seifert P, Edelman ER, Jacobs DO, Zinner MJ, Ashley SW, Whang EE. Glucagon-like peptide 2: a new treatment for chemotherapy-induced enteritis. J Surg Res. 2000;91:77-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
85.  Booth C, Booth D, Williamson S, Demchyshyn LL, Potten CS. Teduglutide ([Gly2]GLP-2) protects small intestinal stem cells from radiation damage. Cell Prolif. 2004;37:385-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
86.  Schmidt PT, Hartmann B, Bregenholt S, Hoist JJ, Claesson MH. Deficiency of the intestinal growth factor, glucagon-like peptide 2, in the colon of SCID mice with inflammatory bowel disease induced by transplantation of CD4+ T cells. Scand J Gastroenterol. 2000;35:522-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
87.  Schmidt PT, Ljung T, Hartmann B, Hare KJ, Holst JJ, Hellström PM. Tissue levels and post-prandial secretion of the intestinal growth factor, glucagon-like peptide-2, in controls and inflammatory bowel disease: comparison with peptide YY. Eur J Gastroenterol Hepatol. 2005;17:207-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
88.  Chen K, Nezu R, Wasa M, Sando K, Kamata S, Takagi Y, Okada A. Insulin-like growth factor-1 modulation of intestinal epithelial cell restitution. JPEN J Parenter Enteral Nutr. 1999;23:S89-S92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
89.  Lund PK. Molecular basis of intestinal adaptation: the role of the insulin-like growth factor system. Ann N Y Acad Sci. 1998;859:18-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
90.  Wang J, Niu W, Nikiforov Y, Naito S, Chernausek S, Witte D, LeRoith D, Strauch A, Fagin JA. Targeted overexpression of IGF-I evokes distinct patterns of organ remodeling in smooth muscle cell tissue beds of transgenic mice. J Clin Invest. 1997;100:1425-1439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 159]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
91.  Vanderhoof JA, McCusker RH, Clark R, Mohammadpour H, Blackwood DJ, Harty RF, Park JH. Truncated and native insulinlike growth factor I enhance mucosal adaptation after jejunoileal resection. Gastroenterology. 1992;102:1949-1956.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Lund PK. Insulin-like growth factor I: molecular biology and relevance to tissue-specific expression and action. Recent Prog Horm Res. 1994;49:125-148.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Ney DM, Huss DJ, Gillingham MB, Kritsch KR, Dahly EM, Talamantez JL, Adamo ML. Investigation of insulin-like growth factor (IGF)-I and insulin receptor binding and expression in jejunum of parenterally fed rats treated with IGF-I or growth hormone. Endocrinology. 1999;140:4850-4860.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Rosenzweig SA. What's new in the IGF-binding proteins. Growth Horm IGF Res. 2004;14:329-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 61]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
95.  Albiston AL, Herington AC. Tissue distribution and regulation of insulin-like growth factor (IGF)-binding protein-3 messenger ribonucleic acid (mRNA) in the rat: comparison with IGF-I mRNA expression. Endocrinology. 1992;130:497-502.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Albiston AL, Taylor RG, Herington AC, Beveridge DJ, Fuller PJ. Divergent ileal IGF-I and IGFBP-3 gene expression after small bowel resection: a novel mechanism to amplify IGF action. Mol Cell Endocrinol. 1992;83:R17-R20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
97.  Ney DM. Effects of insulin-like growth factor-I and growth hormone in models of parenteral nutrition. JPEN J Parenter Enteral Nutr. 1999;23:S184-S189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
98.  Gillingham MB, Dahly EM, Murali SG, Ney DM. IGF-I treatment facilitates transition from parenteral to enteral nutrition in rats with short bowel syndrome. Am J Physiol Regul Integr Comp Physiol. 2003;284:R363-R371.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Barnard JA, Beauchamp RD, Russell WE, Dubois RN, Coffey RJ. Epidermal growth factor-related peptides and their relevance to gastrointestinal pathophysiology. Gastroenterology. 1995;108:564-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 229]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
100.  Thompson JS. Epidermal growth factor and the short bowel syndrome. JPEN J Parenter Enteral Nutr. 1999;23:S113-S116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
101.  Helmrath MA, Shin CE, Erwin CR, Warner BW. The EGFEGF-receptor axis modulates enterocyte apoptosis during intestinal adaptation. J Surg Res. 1998;77:17-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
102.  Shin CE, Helmrath MA, Falcone RA Jr, Fox JW, Duane KR, Erwin CR, Warner BW. Epidermal growth factor augments adaptation following small bowel resection: optimal dosage, route, and timing of administration. J Surg Res. 1998;77:11-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 57]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
103.  Shin CE, Falcone RA Jr, Duane KR, Erwin CR, Warner BW. The distribution of endogenous epidermal growth factor after small bowel resection suggests increased intestinal utilization during adaptation. J Pediatr Surg. 1999;34:22-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
104.  Helmrath MA, Shin CE, Fox JW, Erwin CR, Warner BW. Adaptation after small bowel resection is attenuated by sialoadenectomy: the role for endogenous epidermal growth factor. Surgery. 1998;124:848-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
105.  Stern LE, Erwin CR, O'Brien DP, Huang FS, Warner BW. Serum from mice after small bowel resection enhances intestinal epithelial cell growth. J Pediatr Surg. 2001;36:184-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
106.  O'Brien DP, Nelson LA, Williams JL, Kemp CJ, Erwin CR, Warner BW. Selective inhibition of the epidermal growth factor receptor impairs intestinal adaptation after small bowel resection. J Surg Res. 2002;105:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
107.  Helmrath MA, Erwin CR, Warner BW. A defective EGF-receptor in waved-2 mice attenuates intestinal adaptation. J Surg Res. 1997;69:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 85]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
108.  Juno RJ, Williams JL, Knott AW, Erwin CR, O'brien DP, Warner BW. A serum factor after intestinal resection stimulates epidermal growth factor receptor signaling and proliferation in intestinal epithelial cells. Surgery. 2002;132:377-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
109.  Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP, Wang X. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science. 1997;275:1129-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3677]  [Cited by in F6Publishing: 3636]  [Article Influence: 134.7]  [Reference Citation Analysis (0)]
110.  Hu Y, Benedict MA, Wu D, Inohara N, Núñez G. Bcl-XL interacts with Apaf-1 and inhibits Apaf-1-dependent caspase-9 activation. Proc Natl Acad Sci USA. 1998;95:4386-4391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 396]  [Cited by in F6Publishing: 418]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
111.  Stephens J, Stoll B, Guan XF, Helmrath M, Burrin D. Relative stimulation of superior mesenteric arterial (SMA) blood flow by enteral nutrition and glucagon-like peptide 2 (GLP-2) in piglets. Gastroenterology. 2004;126:A142.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Guan X, Stoll B, Lu X, Tappenden KA, Holst JJ, Hartmann B, Burrin DG. GLP-2-mediated up-regulation of intestinal blood flow and glucose uptake is nitric oxide-dependent in TPN-fed piglets 1. Gastroenterology. 2003;125:136-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 150]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
113.  Adrian TE, Thompson JS, Quigley EM. Time course of adaptive regulatory peptide changes following massive small bowel resection in the dog. Dig Dis Sci. 1996;41:1194-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
114.  Martin Del Olmo JC, Carbajo Caballero MA, Blanco Alvarez JI, Audivert Mena L, Vaquero Puerta C. Decrease in plasma levels of vasoactive intestinal polypeptide in short bowel syndrome: experimental study. Int Surg. 1998;83:150-153.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Lezoche E, Carlei F, Vagni V, Mora GV, Speranza V. Elevated plasma levels of vasoactive intestinal polypeptide in short bowel syndrome. Am J Surg. 1983;145:369-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
116.  Andrews NJ, Irving MH. Human gut hormone profiles in patients with short bowel syndrome. Dig Dis Sci. 1992;37:729-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
117.  Tatemoto K. Isolation and characterization of peptide YY (PYY), a candidate gut hormone that inhibits pancreatic exocrine secretion. Proc Natl Acad Sci USA. 1982;79:2514-2518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 359]  [Cited by in F6Publishing: 320]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
118.  Tatemoto K, Mutt V. Isolation of two novel candidate hormones using a chemical method for finding naturally occurring polypeptides. Nature. 1980;285:417-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 507]  [Cited by in F6Publishing: 525]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
119.  El-Salhy M, Grimelius L, Wilander E, Ryberg B, Terenius L, Lundberg JM, Tatemoto K. Immunocytochemical identification of polypeptide YY (PYY) cells in the human gastrointestinal tract. Histochemistry. 1983;77:15-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 126]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
120.  El-Salhy M, Wilander E, Grimelius L, Terenius L, Lundberg JM, Tatemoto K. The distribution of polypeptide YY (PYY) - and pancreatic polypeptide (PP) - immunoreactive cells in the domestic fowl. Histochemistry. 1982;75:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
121.  Greeley GH Jr, Jeng YJ, Gomez G, Hashimoto T, Hill FL, Kern K, Kurosky T, Chuo HF, Thompson JC. Evidence for regulation of peptide-YY release by the proximal gut. Endocrinology. 1989;124:1438-1443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 100]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
122.  Ali-Rachedi A, Varndell IM, Adrian TE, Gapp DA, Van Noorden S, Bloom SR, Polak JM. Peptide YY (PYY) immunoreactivity is co-stored with glucagon-related immunoreactants in endocrine cells of the gut and pancreas. Histochemistry. 1984;80:487-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 174]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
123.  Böttcher G, Alumets J, Håkanson R, Sundler F. Co-existence of glicentin and peptide YY in colorectal L-cells in cat and man. An electron microscopic study. Regul Pept. 1986;13:283-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 88]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
124.  Savage AP, Gornacz GE, Adrian TE, Ghatei MA, Goodlad RA, Wright NA, Bloom SR. Is raised plasma peptide YY after intestinal resection in the rat responsible for the trophic response. Gut. 1985;26:1353-1358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
125.  Suzuki T, Nakaya M, Itoh Z, Tatemoto K, Mutt V. Inhibition of interdigestive contractile activity in the stomach by peptide YY in Heidenhain pouch dogs. Gastroenterology. 1983;85:114-121.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Allen JM, Fitzpatrick ML, Yeats JC, Darcy K, Adrian TE, Bloom SR. Effects of peptide YY and neuropeptide Y on gastric emptying in man. Digestion. 1984;30:255-262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 128]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
127.  Adrian TE, Savage AP, Fuessl HS, Wolfe K, Besterman HS, Bloom SR. Release of peptide YY (PYY) after resection of small bowel, colon, or pancreas in man. Surgery. 1987;101:715-719.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Wøjdemann M, Wettergren A, Hartmann B, Holst JJ. Glucagon-like peptide-2 inhibits centrally induced antral motility in pigs. Scand J Gastroenterol. 1998;33:828-832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 158]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
129.  Wøjdemann M, Wettergren A, Hartmann B, Hilsted L, Holst JJ. Inhibition of sham feeding-stimulated human gastric acid secretion by glucagon-like peptide-2. J Clin Endocrinol Metab. 1999;84:2513-2517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 32]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
130.  Shibata C, Naito H, Jin XL, Ueno T, Funayama Y, Fukushima K, Hashimoto A, Matsuno S, Sasaki I. Effect of glucagon, glicentin, glucagon-like peptide-1 and -2 on interdigestive gastroduodenal motility in dogs with a vagally denervated gastric pouch. Scand J Gastroenterol. 2001;36:1049-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
131.  Ljungmann K, Hartmann B, Kissmeyer-Nielsen P, Flyvbjerg A, Holst JJ, Laurberg S. Time-dependent intestinal adaptation and GLP-2 alterations after small bowel resection in rats. Am J Physiol Gastrointest Liver Physiol. 2001;281:G779-G785.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Sigalet DL, Martin GR. Mechanisms underlying intestinal adaptation after massive intestinal resection in the rat. J Pediatr Surg. 1998;33:889-892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 31]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
133.  Hill FL, Zhang T, Gomez G, Greeley GH Jr. Peptide YY, a new gut hormone (a mini-review). Steroids. 1991;56:77-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
134.  McCormack SA, Johnson LR. Role of polyamines in gastrointestinal mucosal growth. Am J Physiol. 1991;260:G795-G806.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Thomas T, Thomas TJ. Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications. Cell Mol Life Sci. 2001;58:244-258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 643]  [Cited by in F6Publishing: 621]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
136.  Morgan DM. Polyamines. An overview. Mol Biotechnol. 1999;11:229-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 84]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
137.  Thomas T, Kiang DT. Additive growth-inhibitory effects of DL-alpha-difluoromethylornithine and antiestrogens on MCF-7 breast cancer cell line. Biochem Biophys Res Commun. 1987;148:1338-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
138.  Johnson LR, McCormack SA. Healing of Gastrointestinal Mucosa: Involvement of Polyamines. News Physiol Sci. 1999;14:12-17.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Han X, Kazarinoff MN, Seiler N, Stanley BA. Rat colon ornithine and arginine metabolism: coordinated effects after proliferative stimuli. Am J Physiol Gastrointest Liver Physiol. 2001;280:G389-G399.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Seidel ER, Scemama JL. Gastrointestinal polyamines and regulation of mucosal growth and function. J Nutr Biochem. 1997;8:104-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
141.  Wang JY, Johnson LR, Tsai YH, Castro GA. Mucosal ornithine decarboxylase, polyamines, and hyperplasia in infected intestine. Am J Physiol. 1991;260:G45-G51.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Gobert AP, Cheng Y, Akhtar M, Mersey BD, Blumberg DR, Cross RK, Chaturvedi R, Drachenberg CB, Boucher JL, Hacker A. Protective role of arginase in a mouse model of colitis. J Immunol. 2004;173:2109-2117.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Meleagros L, Ghatei MA, Bloom SR. Release of vasodilator, but not vasoconstrictor, neuropeptides and of enteroglucagon by intestinal ischaemia/reperfusion in the rat. Gut. 1994;35:1701-1706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
144.  Pillai RB, Tolia V, Rabah R, Simpson PM, Vijesurier R, Lin CH. Increased colonic ornithine decarboxylase activity in inflammatory bowel disease in children. Dig Dis Sci. 1999;44:1565-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
145.  Antony T, Thomas T, Shirahata A, Thomas TJ. Selectivity of polyamines on the stability of RNA-DNA hybrids containing phosphodiester and phosphorothioate oligodeoxyribonucleotides. Biochemistry. 1999;38:10775-10784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
146.  Thulesen J, Hartmann B, Hare KJ, Kissow H, Ørskov C, Holst JJ, Poulsen SS. Glucagon-like peptide 2 (GLP-2) accelerates the growth of colonic neoplasms in mice. Gut. 2004;53:1145-1150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 90]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
147.  Hamm-Alvarez SF. Focus on "EGF receptor downregulation depends on a trafficking motif in the distal tyrosine kinase domain". Am J Physiol Cell Physiol. 2002;282:C417-C419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
148.  Levkowitz G, Waterman H, Zamir E, Kam Z, Oved S, Langdon WY, Beguinot L, Geiger B, Yarden Y. c-Cbl/Sli-1 regulates endocytic sorting and ubiquitination of the epidermal growth factor receptor. Genes Dev. 1998;12:3663-3674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 676]  [Cited by in F6Publishing: 667]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]