Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elaine de Heuvel is active.

Publication


Featured researches published by Elaine de Heuvel.


Journal of Pediatric Surgery | 2010

The influence of nutrients, biliary-pancreatic secretions, and systemic trophic hormones on intestinal adaptation in a Roux-en-Y bypass model.

Esmaeel Taqi; Laurie E. Wallace; Elaine de Heuvel; Prasanth K. Chelikani; Huiyuan Zheng; Hans-Rudolph Berthoud; Jens J. Holst; David L. Sigalet

PURPOSE The signals that govern the upregulation of nutrient absorption (adaptation) after intestinal resection are not well understood. A Gastric Roux-en-Y bypass (GRYB) model was used to isolate the relative contributions of direct mucosal stimulation by nutrients, biliary-pancreatic secretions, and systemic enteric hormones on intestinal adaptation in short bowel syndrome. METHODS Male rats (350-400 g; n = 8/group) underwent sham or GRYB with pair feeding and were observed for 14 days. Weight and serum hormonal levels (glucagon-like peptide-2 [GLP-2], PYY) were quantified. Adaptation was assessed by intestinal morphology and crypt cell kinetics in each intestinal limb of the bypass and the equivalent points in the sham intestine. Mucosal growth factors and expression of transporter proteins were measured in each limb of the model. RESULTS The GRYB animals lost weight compared to controls and exhibited significant adaptive changes with increased bowel width, villus height, crypt depth, and proliferation indices in the alimentary and common intestinal limbs. Although the biliary limb did not adapt at the mucosa, it did show an increased bowel width and crypt cell proliferation rate. The bypass animals had elevated levels of systemic PYY and GLP-2. At the mucosal level, insulin-like growth factor-1 (IGF-1) and basic fibroblast growth factor (bFGF) increased in all limbs of the bypass animals, whereas keratinocyte growth factor (KGF) and epidermal growth factor (EGF) had variable responses. The expression of the passive transporter of glucose, GLUT-2, expression was increased, whereas GLUT-5 was unchanged in all limbs of the bypass groups. Expression of the active mucosal transporter of glucose, SGLT-1 was decreased in the alimentary limb. CONCLUSIONS Adaptation occurred maximally in intestinal segments stimulated by nutrients. Partial adaptation in the biliary limb may reflect the effects of systemic hormones. Mucosal content of IGF-1, bFGF, and EGF appear to be stimulated by systemic hormones, potentially GLP-2, whereas KGF may be locally regulated. Further studies to examine the relationships between the factors controlling nutrient-induced adaptation are suggested. Direct contact with nutrients appears to be the most potent factor in inducing mucosal adaptation.


American Journal of Physiology-endocrinology and Metabolism | 2012

Glucagon-like peptide 2 induces vasoactive intestinal polypeptide expression in enteric neurons via phophatidylinositol 3-kinase-γ signaling.

Elaine de Heuvel; Laurie E. Wallace; Keith A. Sharkey; David L. Sigalet

Glucagon-like peptide 2 (GLP-2) is an enteroendocrine hormone trophic for intestinal mucosa; it has been shown to increase enteric neuronal expression of vasoactive intestinal polypeptide (VIP) in vivo. We hypothesized that GLP-2 would regulate VIP expression in enteric neurons via a phosphatidylinositol-3 kinase-γ (PI3Kγ) pathway. The mechanism of action of GLP-2 was investigated using primary cultures derived from the submucosal plexus (SMP) of the rat and mouse colon. GLP-2 (10(-8) M) stimulation for 24 h increased the proportion of enteric neurons expressing VIP (GLP-2: 40 ± 6% vs. control: 22 ± 5%). GLP-2 receptor expression was identified by immunohistochemistry on neurons (HuC/D+) and glial cells (GFAP+) but not on smooth muscle or fibroblasts in culture. Over 1-4 h, GLP-2 stimulation of SMP increased phosphorylated Akt/Akt ratios 6.1-fold, phosphorylated ERK/ERK 2.5-fold, and p70S6K 2.2-fold but did not affect intracellular cAMP. PI3Kγ gene deletion or pharmacological blockade of PI3Kγ, mammalian target of rapamycin (mTOR), and MEK/ERK pathways blocked the increase in VIP expression by GLP-2. GLP-2 increased the expression of growth factors and their receptors in SMP cells in culture [IGF-1r (3.2-fold increase), EGFr (5-fold), and ErbB-2-4r (6- to 7-fold)] and ligands [IGF-I (1.5-fold), amphiregulin (2.5-fold), epiregulin (3.2-fold), EGF (7.5-fold), heparin-bound EGF (2.0-fold), β-cellulin (50-fold increase), and neuregulins 2-4 (300-fold increase) (by qRT-PCR)]. We conclude that GLP-2 acts on enteric neurons and glial cells in culture via a PI3Kγ/Akt pathway, stimulating neuronal differentiation via mTOR and ERK pathways, and expression of receptors and ligands for the IGF-I and ErbB pathways.


Regulatory Peptides | 2014

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

David L. Sigalet; Elaine de Heuvel; Laurie E. Wallace; Estrella S. Bulloch; Justine M. Turner; Paul W. Wales; Patrick N. Nation; Pamela R. Wizzard; B. Hartmann; Meena Assad; Jens J. Holst

BACKGROUND The enteroendocrine hormone glucagon like peptide-2 (GLP-2) and its ligands are under development as therapeutic agents for a variety of intestinal pathologies. A number of these conditions occur in neonates and infants, and thus a detailed understanding of the effects of GLP-2 during the phase of rapid growth during infancy is required to guide the development of therapeutic applications. We studied the effects of GLP-2 in the neonatal pig to determine the potential effects of exogenous administration. METHODS Two day old newborn domestic piglets were treated with GLP-2 (1-33) at 40 μg/kg/day or control drug vehicle (saline), by subcutaneous injection, given in two doses per day, (n=6/group) for 42 days. Animals were weaned normally, over days 21-25. In the fifth week of life, they underwent neuro-developmental testing, and a pharmacokinetic study. On day 42, they were euthanized, and a complete necropsy performed, with histological assessment of tissues from all major organs. RESULTS GLP-2 treatment was well tolerated, one control animal died from unrelated causes. There were no effects of GLP-2 on weight gain, feed intake, or behavior. In the treated animals, GLP-2 levels were significantly elevated at 2400±600 pM while at necropsy, organ weights and histology were not affected except in the intestine, where the villus height in the small intestine and the crypt depth, throughout the small intestine and colon, were increased. Similarly, the rate of crypt cell proliferation (Ki-67 staining) was increased in the GLP-2 treated animals and the rate of apoptosis (Caspase-3) was decreased, the depth of the microvilli was increased and the expression of the mRNA for the GLP-2 receptor was decreased throughout the small and large intestine. CONCLUSIONS In these growing animals, exogenous GLP-2 at pharmacologic doses was well tolerated, with effects confined to the gastrointestinal tract.


Journal of Parenteral and Enteral Nutrition | 2017

A Safety and Dosing Study of Glucagon-Like Peptide 2 in Children With Intestinal Failure

David L. Sigalet; Mary Brindle; Dana Boctor; Linda Casey; Bryan Dicken; Sonia A. Butterworth; Viona Lam; Vikram Karnik; Elaine de Heuvel; Bolette Hartmann; Jens J. Holst

Background and Aims: A glucagon-like peptide 2 (GLP-2) analogue is approved for adults with intestinal failure, but no studies of GLP-2 have included children. This study examined the pharmacokinetics, safety, and nutritional effects of GLP-2 in children with intestinal failure. Methods: Native human GLP-2(1-33) was synthesized following good manufacturing practices. In an open-label trial, with parental consent, 7 parenteral nutrition–dependent pediatric patients were treated with subcutaneous GLP-2 (20 µg/kg/d) for 3 days (phase 1) and, if tolerated, continued for 42 days (phase 2). Nutritional treatment was directed by the primary caregivers. Patients were followed to 1 year. Results: Seven patients were enrolled (age: 4.0 ± 0.8 years; bowel length, mean ± SEM: 24% ± 4% of predicted). All were parenteral nutrition dependent since birth, receiving 44% ± 5% of calories by parenteral nutrition. GLP-2 treatment had no effect on vital signs (blood pressure, heart rate, and temperature) and caused no significant adverse events. Peak GLP-2 levels were 380 pM (day 3) and 295 pM (day 42), with no change in half-life or endogenous GLP-2 levels. Nutritional indices showed a numeric improvement in z scores and citrulline levels; the z score was maintained while citrulline levels returned to baseline once GLP-2 was discontinued. Conclusions: GLP-2 was well tolerated in children, with a pharmacokinetic profile similar to that of adults. There were no changes in endogenous GLP-2 release or metabolism. These results suggest that GLP-2 ligands may be safely used in pediatric patients; larger trials are suggested to investigate nutritional effects.


PLOS ONE | 2017

Effects of exogenous glucagon-like peptide-2 and distal bowel resection on intestinal and systemic adaptive responses in rats.

Sarah W. Lai; Elaine de Heuvel; Laurie E. Wallace; Bolette Hartmann; Jens J. Holst; Mary Brindle; Prasanth K. Chelikani; David L. Sigalet

Objective To determine the effects of exogenous glucagon-like peptide-2 (GLP-2), with or without massive distal bowel resection, on adaptation of jejunal mucosa, enteric neurons, gut hormones and tissue reserves in rats. Background GLP-2 is a gut hormone known to be trophic for small bowel mucosa, and to mimic intestinal adaptation in short bowel syndrome (SBS). However, the effects of exogenous GLP-2 and SBS on enteric neurons are unclear. Methods Sprague Dawley rats were randomized to four treatments: Transected Bowel (TB) (n = 8), TB + GLP-2 (2.5 nmol/kg/h, n = 8), SBS (n = 5), or SBS + GLP-2 (2.5 nmol/kg/h, n = 9). SBS groups underwent a 60% jejunoileal resection with cecectomy and jejunocolic anastomosis. All rats were maintained on parenteral nutrition for 7 d. Parameters measured included gut morphometry, qPCR for hexose transporter (SGLT-1, GLUT-2, GLUT-5) and GLP-2 receptor mRNA, whole mount immunohistochemistry for neurons (HuC/D, VIP, nNOS), plasma glucose, gut hormones, and body composition. Results Resection increased the proportion of nNOS immunopositive myenteric neurons, intestinal muscularis propria thickness and crypt cell proliferation, which were not recapitulated by GLP-2 therapy. Exogenous GLP-2 increased jejunal mucosal surface area without affecting enteric VIP or nNOS neuronal immunopositivity, attenuated resection-induced reductions in jejunal hexose transporter abundance (SGLT-1, GLUT-2), increased plasma amylin and decreased peptide YY concentrations. Exogenous GLP-2 attenuated resection-induced increases in blood glucose and body fat loss. Conclusions Exogenous GLP-2 stimulates jejunal adaptation independent of enteric neuronal VIP or nNOS changes, and has divergent effects on plasma amylin and peptide YY concentrations. The novel ability of exogenous GLP-2 to modulate resection-induced changes in peripheral glucose and lipid reserves may be important in understanding the whole-body response following intestinal resection, and is worthy of further study.


Journal of Pediatric Surgery | 2017

A safety and pharmacokinetic dosing study of glucagon-like peptide 2 in infants with intestinal failure

David L. Sigalet; Mary Brindle; Dana Boctor; Bryan Dicken; Viona Lam; Lily Sia Lu; Elaine de Heuvel; Bolette Hartmann; Jens J. Holst

BACKGROUND & AIMS Glucagon-like peptide 2 (GLP-2) analogues are approved for adults with intestinal failure (IF), but no studies have included infants. This study examined the pharmacokinetics (PK), safety, and nutritional effects of GLP-2 in infants with IF. METHODS With parental consent (Health Canada Protocol:150,979), parenteral nutrition (PN)-dependent infants were treated with 5-20-μg/kg/day GLP-2 for 3days (phase 1), and if tolerated continued for 42days (phase 2). Nutritional therapy was by primary caregivers, and follow-up was to one year. RESULTS Six patients were enrolled, age 5.4±3.2months, bowel length: 27±12% of predicted, PN dependent (67±18% of calories). GLP-2 did not affect vital signs, nor were there significant adverse events during the trial. Dosing 5μg/kg/day gave GLP-2 levels of 52-57pmol/L, with no change in half-life or endogenous GLP-2 levels. Enteral feeds, weight, Z scores, stooling frequency, and citrulline levels improved numerically. The trial was discontinued early because of a drop in potency. CONCLUSIONS GLP-2 was well tolerated in infants, and pK was similar to children with no changes in endogenous GLP-2 release. The findings suggest that GLP-2 ligands may be safely used in infants and may have beneficial effects on nutritional status. Further study is required. LEVEL OF EVIDENCE 2b Prospective Interventional Study.


Gastroenterology | 2014

Mo1788 Vasoactive Intestinal Polypeptide and Neuronal Nitric Oxide Synthase Act via Parallel Pathways As Mediators of GLP-2 Anti-Inflammatory Activity

Laurie E. Wallace; Elaine de Heuvel; James A. Waschek; David L. Sigalet

Glucagon-like peptide-2 (GLP-2) has potent anti-inflammatory effects in the intestinal mucosa. Vasoactive intestinal polypeptide (VIP) neurons in the enteric neuronal plexus have been shown to be important in these actions using pharmacologic blockers; but little is known about other specific subsets of neurotransmitters, such as nNOS. Aim: To investigate the role of VIP and nNOS signaling, individually and together, in mediating the antiinflammatory effects of GLP-2. Methods: With animal committee use approval, mice with selective knockouts of VIP-/-, nNOS-/-, and combined VIP-/-/ nNOS-/were used, with wild type (WT) controls; all animals were treated with trinitrobenzene sulfonic acid (TNBS) to induce colitis. Animals received (1-33) GLP-2 (50 μg/kg s.c. bid) or saline for 5 days. Mice were sacrificed and colonic tissue was taken for inflammation indices (myeloperoxidase [MPO] and histological damage scores), immunohistochemistry (BrdU:proliferation marker; Caspase-3: apoptosis marker), and mucosal cytokine levels (TNFα, IL1β, IFNγ, IL10). Results: GLP-2 treatment resulted in significant anti-inflammatory effects in VIP-/-and nNOS-/animals, but had no effects in the VIP-//nNOS-/treated animals. The response included improved weight and mucosal inflammation indices with reduced levels of proinflammatory cytokines (TNFα, IL1β, IFNγ), and reduced proliferation and apoptosis rates. In VIP-/nNOS-/animals treated with GLP-2 these cytokines and cell kinetic rates remained unchanged. (See table, n=8 per group, data: Mean±SEM, *p<0.05 by ANOVA) Conclusions: These findings suggest that the anti-inflammatory effects of GLP-2 can be mediated by either VIP or nNOS pathways, however when both pathways are abolished concomitantly, the anti-inflammatory effects of GLP-2 are lost. Further study is required to identify the enteric neuronal circuitry which mediates these potent anti-inflammatory effects


Gastroenterology | 2008

T1205 Glucagon-Like Peptide-2 Improves Healing of TNBS Colitis By Altering Immune Regulatory Cell Signaling in the Lamina Propria

Catherine Ivory; Laurie E. Wallace; Elaine de Heuvel; David L. Sigalet


Gastroenterology | 2014

Su2094 Safety and Dosing Study of Glucagon-Like Peptide 2 (GLP-2) in Children With Intestinal Failure

David L. Sigalet; Viona Lam; Vikram Karnik; Mary Brindle; Bolette Hartmann; Dana Boctor; Linda Casey; Bryan Dicken; Sharla Stoffman; Elaine de Heuvel; Gail Wrightwilson; Laurie E. Wallace; Jens J. Holst


Gastroenterology | 2011

Glucagon-Like Peptide 2 Induces Neuronal Proliferation via an Activation Dependent, but cAMP Independent Pathway

Elaine de Heuvel; Laurie E. Wallace; Keith A. Sharkey; David L. Sigalet

Collaboration


Dive into the Elaine de Heuvel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jens J. Holst

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar

Dana Boctor

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Mary Brindle

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Viona Lam

Alberta Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge