Network


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

Hotspot


Dive into the research topics where Laurie E. Wallace is active.

Publication


Featured researches published by Laurie E. Wallace.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2008

Interleukin-10-independent anti-inflammatory actions of glucagon-like peptide 2

Catherine Ivory; Laurie E. Wallace; Donna-Marie McCafferty; David L. Sigalet

Glucagon-like peptide 2 (GLP-2) is an important intestinal growth factor with anti-inflammatory activity. We hypothesized that GLP-2 decreases mucosal inflammation and the associated increased epithelial proliferation by downregulation of Th1 cytokines attributable to reprogramming of lamina propria immune regulatory cells via an interleukin-10 (IL-10)-independent pathway. The effects of GLP-2 treatment were studied using the IL-10-deficient (IL-10(-/-)) mouse model of colitis. Wild-type and IL-10(-/-) mice received saline or GLP-2 (50 microg/kg sc) treatment for 5 days. GLP-2 treatment resulted in significant amelioration of animal weight loss and reduced intestinal inflammation as assessed by histopathology and myeloperoxidase levels compared with saline-treated animals. In colitis animals, GLP-2 treatment also reduced crypt cell proliferation and crypt cell apoptosis. Proinflammatory (IL-1beta, TNF-alpha, IFN-gamma,) cytokine protein levels were significantly reduced after GLP-2 treatment, whereas IL-4 was significantly increased and IL-6 production was unchanged. Fluorescence-activated cell sorting analysis of lamina propria cells demonstrated a decrease in the CD4(+) T cell population following GLP-2 treatment in colitic mice and an increase in CD11b(+)/F4/80(+) macrophages but no change in CD25(+)FoxP3 T cells or CD11c(+) dendritic cells. In colitis animals, intracellular cytokine analysis demonstrated that GLP-2 decreased lamina propria macrophage TNF-alpha production but increased IGF-1 production, whereas transforming growth factor-beta was unchanged. GLP-2-mediated reduction of crypt cell proliferation was associated with an increase in intestinal epithelial cell suppressor of cytokine signaling (SOCS)-3 expression and reduced STAT-3 signaling. This study shows that the anti-inflammatory effects of GLP-2 are IL-10 independent and that GLP-2 alters the mucosal response of inflamed intestinal epithelial cells and macrophages. In addition, the suggested mechanism of the reduction in inflammation-induced proliferation is attributable to GLP-2 activation of the SOCS-3 pathway, which antagonizes the IL-6-mediated increase in STAT-3 signaling.


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.


Neurogastroenterology and Motility | 2010

The effects of glucagon-like peptide 2 on enteric neurons in intestinal inflammation.

David L. Sigalet; Laurie E. Wallace; E. De Heuval; Keith A. Sharkey

Background  Intestinal inflammation alters the structure and function of the enteric nervous system (ENS). Glucagon‐like peptide 2 (GLP‐2) reduces intestinal inflammation and has trophic effects on isolated neurons. This study examined the effects of GLP‐2 treatment on the submucosal plexus of rat colon in the trinitrobenzene sulfonic acid (TNBS) model of colitis.


Journal of Surgical Research | 2009

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

Tatsuru Kaji; Hiroaki Tanaka; Heather Redstone; Laurie E. Wallace; Jens J. Holst; David L. Sigalet

BACKGROUND We investigated the effects of variations in the postresection timing of glucagon-like peptide-2 (GLP-2) administration on intestinal morphology and activity. METHODS A rat model of 90% intestinal resection (SBR) with exclusively parenteral nutritional (TPN) was used. Early versus late postresection GLP-2 stimulation was compared between SBR + TPN alone, SBR + TPN + GLP-2 (first wk), and SBR + TPN + GLP-2 (second wk) (n = 8/group). On d 14, animals were sacrificed and remnant ileum analyzed for morphology, crypt cell proliferation index (CPI), apoptosis index (API), and nutrient transporter expression (SGLT-1, GLUT-2, GLUT-5). In a separate study, the resection-induced effect on acute GLP-2 responsiveness was studied at d 3 and 10, in control or SBR animals, both supported with TPN. (n = 6). RESULTS Bowel length, weight, and width were increased in SBR + TPN + GLP-2 (first wk) compared with the SBR + TPN alone and SBR + TPN + GLP-2 (second wk) groups. Animal weight, villus height, total mucosal surface area, and CPI increased in both GLP-2 treated groups compared with the SBR + TPN group. Villus height and crypt depth effects were most pronounced in the SBR + TPN + GLP-2 (second wk) group. Increased expression of mRNA for the GLP-2 receptor was noted at d 3, declining below baseline by d 10, however this was not correlated with GLP-2 activation of enteric neurons. Exogenous GLP-2 increased the activation of submucosal neurons at d 3 in controls; resected animals had a higher baseline activity, but exogenous GLP-2 did not activate this further at either d 3 or 10 postresection. CONCLUSIONS GLP-2 effects on intestinal growth are maximal in the early postresection period and are associated with an apparent increase in expression of the receptor but no increase in neuronal activation. This suggests that the intestinal adaptive and growth promoting actions of GLP-2 may be mediated by non-neuronal effector pathways. Although further studies are required, early treatment with GLP-2 following resection may maximize intestinal growth.


Pediatrics | 2008

Functional Ontogeny of the Proglucagon-Derived Peptide Axis in the Premature Human Neonate

Harish Amin; Jens J. Holst; Bolette Hartmann; Laurie E. Wallace; James G. Wright; David L. Sigalet

BACKGROUND. The regulation of intestinal growth and development in human neonates is incompletely understood, which hinders the provision of nutrients enterally. The “hindgut” hormones glucagon-like peptides 1 and 2 have been shown to play an important role in the regulation of nutrient assimilation, intestinal growth, and function. OBJECTIVE. Our goal was to investigate the production of glucagon-like peptides 1 and 2 in premature human infants and examine the effects of prematurity and feeding on hormone release. PATIENTS AND METHODS. With informed consent, premature infants who were admitted to a tertiary neonatal intensive care nursery (gestational age: 28–32 weeks) were monitored with weekly determinations of postprandial glucagon-like peptide 1 and 2 levels. Comparison studies with groups of normal infants and adults were performed. Hormone levels were obtained by using specific radioimmunoassay for glucagon-like peptide 1 (1–36) and glucagon-like peptide 2 (1–33), modified for small sample volumes; accurate monitoring of enteral intake was performed at all of the sampling time points. RESULTS. Forty-five infants with a mean gestational age of 29.6 ± 1.9 weeks were studied; fasting levels of both glucagon-like peptides 1 and 2 were elevated. There was no correlation between gestational age and glucagon-like peptide 2 output. However, both glucagon-like peptide 1 and 2 levels were correlated with the caloric value of feeds. CONCLUSIONS. The premature human neonate has significantly higher fasting levels of glucagon-like peptides 1 and 2 compared with adults; feeding increases these levels further. These findings suggest that the proglucagon-derived peptides may have a role in normal intestinal development and nutrient handling.


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 Pediatric Surgery | 2009

Nutritional effects of the serial transverse enteroplasty procedure in experimental short bowel syndrome

Tatsuru Kaji; Hiroaki Tanaka; Laurie E. Wallace; Dragan Kravarusic; Jens J. Holst; David L. Sigalet

BACKGROUND/PURPOSE The serial transverse enteroplasty (STEP) procedure appears beneficial clinically, but the mechanism(s) underlying these effects remains unclear. The present study evaluated the nutritional, hormonal, and morphologic effects of the STEP procedure in a rodent model of short bowel syndrome. METHODS With institutional animal care ethics approval, Sprague-Dawley rats underwent an 80% distal bowel resection, anastomosing the 30 cm remnant of jejunum to the ascending colon; at day 14, animals were randomly assigned to control or a STEP procedure (n = 8/group). Animals were pair-fed with normal chow; after a further 3 weeks, intestinal transit, hormonal and metabolic balance studies were done, and intestinal tissues were taken for analysis. RESULTS The STEP group had increased weight gain (resected: -0.34% +/- 2.9% vs STEP: 2.5% +/- 1.5%), increased bowel length (34.1 +/- 1.5 vs 36.9 +/- 2.2 cm), increased jejunal villus height (555 +/- 59 vs 635 +/- 65 microm), decreased rates of crypt cell apoptosis, increased expression of mRNA for the GLP-2 receptor, and increased postprandial production of glucagon-like peptide 2 (45 +/- 14 vs 65 +/- 12 pmol/L) (P < .05 by Student t test). There were no differences in intestinal transit; absorption of total calories, protein, fat, or carbohydrate; crypt cell proliferation rates; or the expression of intestinal transporter proteins (SGLT-1, GLUT-2, and GLUT-5). CONCLUSIONS The STEP procedure improves weight gain and augments gross and microscopic intestinal morphology in severe experimental short bowel syndrome. Postprandial GLP-2 levels are increased, as is the expression of the GLP-2 receptor; these mechanisms may contribute to these metabolic effects and may be useful in guiding the use of the STEP procedure clinically.


European Journal of Pharmacology | 2008

The effects of variations in dose and method of administration on glucagon like peptide-2 activity in the rat.

Tatsuru Kaji; Hiroaki Tanaka; Jens J. Holst; Heather Redstone; Laurie E. Wallace; Elaine de Heuval; David L. Sigalet

Glucagon-like peptide-2 (GLP-2) is a potent, intestinal-specific trophic hormone. However, the relationship between the dose and timing of GLP-2 administration and these trophic effects is not clear. We investigated the effects of variations in the dose and timing of GLP-2 administration on its intestinal trophic activity. A rodent model of total parenteral nutrition (TPN) mucosal atrophy was used, examining intestinal morphology in the adult male rat after 5 days. Groups were: controls, maintained with TPN alone and GLP-2 treated groups (high dose; 240 microg/kg/day, low dose; 24 microg/kg/day) given by continuous or intermittent (over 1 h, twice daily) intravenous infusion. Body weight and total small bowel length were significantly increased in the high dose, continuous infusion group. Both high dose infusion methods increased total small bowel weight, villus height, crypt depth, and total mucosal surface area. Both high dose infusion and low dose intermittent infusion routes increased crypt cell proliferation (P<0.05 for all comparisons). Both high dose routes gave nearly equivalent exposures; low dose continuous infusion gave higher exposure but intermittent low dose infusion resulted in an increase in crypt proliferation; neither low dose method resulted in morphologic changes. There were no differences in transporter protein expression or apoptosis rates. High dose continuous infusion appears to maximally induce intestinal growth, and also increases weight gain, while high dose GLP-2 intermittent infusion results in similar morphologic effects. A threshold level for the induction of proliferative and morphologic effects was seen in the low dose groups. These observations may be relevant for planning therapeutic trials.


Gastroenterology Research and Practice | 2010

The Effect of Glucagon-Like Peptide-2 Receptor Agonists on Colonic Anastomotic Wound Healing

Heather Redstone; William D. Buie; David A. Hart; Laurie E. Wallace; Pamela J. Hornby; Sarah Sague; Jen J. Holst; David L. Sigalet

Background. Glucagon-like peptide 2 (GLP-2) is an intestinal specific trophic hormone, with therapeutic potential; the effects on intestinal healing are unknown. We used a rat model of colonic healing, under normoxic, and stress (hypoxic) conditions to examine the effect of GLP-2 on intestinal healing. Methods. Following colonic transection and reanastomosis, animals were randomized to one of six groups (n = 8/group): controls, native GLP-2, long-acting GLP-2 (GLP-2- MIMETIBODY, GLP-2-MMB), animals were housed under normoxic or hypoxic (11%  O2) conditions. Animals were studied five days post-operation for anastomotic strength and wound characteristics. Results. Anastomotic bursting pressure was unchanged by GLP-2 or GLP-2-MMB in normoxic or hypoxic animals; both treatments increased crypt cell proliferation. Wound IL-1β increased with GLP-2; IFNγ with GLP-2 and GLP-2-MMB. IL-10 and TGF-β were decreased; Type I collagen mRNA expression increased in hypoxic animals while Type III collagen was reduced with both GLP-2 agonists. GLP-2 MMB, but not native GLP-2 increased TIMP 1-3 mRNA levels in hypoxia. Conclusions. The effects on CCP, cytokines and wound healing were similar for both GLP-2 agonists under normoxic and hypoxic conditions; anastomotic strength was not affected. This suggests that GLP-2 (or agonists) could be safely used peri-operatively; direct studies will be required.

Collaboration


Dive into the Laurie E. Wallace'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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
Researchain Logo
Decentralizing Knowledge