Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 324(2): G115-G130, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36511517

ABSTRACT

Proteolipid protein 1 (Plp1) is highly expressed in enteric glia, labeling cells throughout the mucosa, muscularis, and the extrinsic innervation. Plp1 is a major constituent of myelin in the central and peripheral nervous systems, but the absence of myelin in the enteric nervous system (ENS) suggests another role for Plp1 in the gut. Although the functions of enteric glia are still being established, there is strong evidence that they regulate intestinal motility and permeability. To interrogate the role of Plp1 in enteric glia, we investigated gut motility, secretomotor function and permeability, and evaluated the ENS in mice lacking Plp1. We studied two time points: ∼3 mo (young) and >1 yr (old). Old Plp1 null mice exhibited increased fecal output, decreased fecal water content, faster whole gut transit times, reduced intestinal permeability, and faster colonic migrating motor complexes. Interestingly, in both young and old mice, the ENS exhibited normal glial and neuronal numbers as well as glial arborization density in the absence of Plp1. As Plp1-associated functions involve mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 (Mapk/Erk1/2) signaling and Mapk/Erk1/2 are reported to have a regulatory role in intestinal motility, we measured protein expression of Erk1/2 and its active form in the small intestine. Old Plp1 null mice had reduced levels of phosphorylated-Erk1/2. Although Plp1 is not required for the normal appearance of enteric glial cells, it has a regulatory role in intestinal motility and barrier function. Our results suggest that functional changes mediated by Plp1-expressing enteric glia may involve Erk1/2 activation.NEW & NOTEWORTHY Here, we describe that Plp1 regulates gut motility and barrier function. The functional effects of Plp1 eradication are only seen in old mice, not young. The effects of Plp1 appear to be mediated through the Erk1/2 pathway.


Subject(s)
Gastrointestinal Motility , Intestinal Mucosa , Myelin Proteolipid Protein , Animals , Mice , Enteric Nervous System/physiology , Gastrointestinal Motility/physiology , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Proteolipids/metabolism , Proteolipids/pharmacology , Myelin Proteolipid Protein/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology
2.
Adv Exp Med Biol ; 1383: 55-69, 2022.
Article in English | MEDLINE | ID: mdl-36587146

ABSTRACT

Propulsive gastrointestinal (GI) motility is critical for digestive physiology and host defense. GI motility is finely regulated by the intramural reflex pathways of the enteric nervous system (ENS). The ENS is in turn regulated by luminal factors: diet and the gut microbiota. The gut microbiota is a vast ecosystem of commensal bacteria, fungi, viruses, and other microbes. The gut microbiota not only regulates the motor programs of the ENS but also is critical for the normal structure and function of the ENS. In this chapter, we highlight recent research that has shed light on the microbial mechanisms of interaction with the ENS involved in the control of motility. Toll-like receptor signaling mechanisms have been shown to maintain the structural integrity of the ENS and the neurochemical phenotypes of enteric neurons, in part through the production of trophic factors including glia-derived neurotrophic factor. Microbiota-derived short-chain fatty acids and/or single-stranded RNA regulates the synthesis of serotonin in enterochromaffin cells, which are involved in the initiation of enteric reflexes, among other functions. Further evidence suggests a crucial role for microbial modulation of serotonin in maintaining the integrity of the ENS through enteric neurogenesis. Understanding the microbial pathways of enteric neural control sheds new light on digestive health and provides novel treatment strategies for GI motility disorders.


Subject(s)
Enteric Nervous System , Gastrointestinal Microbiome , Microbiota , Gastrointestinal Microbiome/physiology , Serotonin/metabolism , Enteric Nervous System/metabolism , Neurons/physiology , Gastrointestinal Motility/physiology
3.
Sci Rep ; 12(1): 20169, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418891

ABSTRACT

Gastrointestinal tract (gut) inflammation increases stress and threat-coping behaviors, which are associated with altered activity in fear-related neural circuits, such as the basolateral amygdala and hippocampus. It remains to be determined whether inflammation from the gut affects neural activity by altering dendritic spines. We hypothesized that acute inflammation alters dendritic spines in a brain region-specific manner. Here we show that acute gut inflammation (colitis) evoked by dextran sodium sulfate (DSS) did not affect the overall spine density in the CA1 region of hippocampus, but increased the relative proportion of immature spines to mature spines on basal dendrites of pyramidal neurons. In contrast, in animals with colitis, no changes in spine density or composition on dendrites of pyramidal cells was observed in the basolateral amygdala. Rather, we observed decreased spine density on dendrites of stellate neurons, but not the relative proportions of mature vs immature spines. We used cFos expression evoked by the forced swim task as a measure of neural activity during stress and found no effect of DSS on the density of cFos immunoreactive neurons in basolateral amygdala. In contrast, fewer CA1 neurons expressed cFos in mice with colitis, relative to controls. Furthermore, CA1 cFos expression negatively correlated with active stress-coping in the swim task and was negatively correlated with gut inflammation. These data reveal that the effects of acute gut inflammation on synaptic remodeling depend on brain region, neuronal phenotype, and dendrite location. In the hippocampus, a shift to immature spines and hypoactivity are more strongly related to colitis-evoked behavioral changes than is remodeling in basolateral amygdala.


Subject(s)
Basolateral Nuclear Complex , Colitis , Animals , Mice , Hippocampus , Pyramidal Cells , Inflammation , Colitis/chemically induced
4.
Am J Physiol Gastrointest Liver Physiol ; 323(3): G219-G238, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35787179

ABSTRACT

The endocannabinoid system of the gastrointestinal tract is involved in the control of intestinal barrier function. Whether the cannabinoid 1 (CB1) receptor is expressed on the intestinal epithelium and acutely regulates barrier function has not been determined. Here, we tested the hypothesis that ligands of the CB1 receptor acutely modulate small intestinal permeability and that this is associated with altered distribution of tight junction proteins. We examined the acute effects of CB1 receptor ligands on small intestinal permeability both in chow-fed and 2-wk high-fat diet (HFD)-fed mice using Ussing chambers. We assessed the distribution of CB1 receptor and tight junction proteins using immunofluorescence and the expression of CB1 receptor using PCR. A low level of CB1 expression was found on the intestinal epithelium. CB1 receptor was highly expressed on enteric nerves in the lamina propria. Neither the CB1/CB2 agonist CP55,940 nor the CB1 neutral antagonist AM6545 altered the flux of 4kDa FITC dextran (FD4) across the jejunum or ileum of chow-fed mice. Remarkably, both CP55,940 and AM6545 reduced FD4 flux across the jejunum and ileum in HFD-fed mice that have elevated baseline intestinal permeability. These effects were absent in CB1 knockout mice. CP55,940 reduced the expression of claudin-2, whereas AM6545 had little effect on claudin-2 expression. Neither ligand altered the expression of ZO-1. Our data suggest that CB1 receptor on the intestinal epithelium regulates tight junction protein expression and restores barrier function when it is increased following exposure to a HFD for 2 wk.NEW & NOTEWORTHY The endocannabinoid system of the gastrointestinal tract regulates homeostasis by acting as brake on motility and secretion. Here we show that when exposed to a high fat diet, intestinal permeability is increased and activation of the CB1 receptor on the intestinal epithelium restores barrier function. This work further highlights the role of the endocannabinoid system in regulating intestinal homeostasis when it is perturbed.


Subject(s)
Diet, High-Fat , Intestinal Mucosa , Receptor, Cannabinoid, CB1 , Animals , Claudin-2/metabolism , Diet, High-Fat/adverse effects , Endocannabinoids/physiology , Intestinal Mucosa/physiology , Mice , Permeability , Receptor, Cannabinoid, CB1/physiology
5.
Microbiome ; 9(1): 210, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702353

ABSTRACT

BACKGROUND: The intestinal microbiota plays an important role in regulating gastrointestinal (GI) physiology in part through interactions with the enteric nervous system (ENS). Alterations in the gut microbiome frequently occur together with disturbances in enteric neural control in pathophysiological conditions. However, the mechanisms by which the microbiota regulates GI function and the structure of the ENS are incompletely understood. Using a mouse model of antibiotic (Abx)-induced bacterial depletion, we sought to determine the molecular mechanisms of microbial regulation of intestinal function and the integrity of the ENS. Spontaneous reconstitution of the Abx-depleted microbiota was used to assess the plasticity of structure and function of the GI tract and ENS. Microbiota-dependent molecular mechanisms of ENS neuronal survival and neurogenesis were also assessed. RESULTS: Adult male and female Abx-treated mice exhibited alterations in GI structure and function, including a longer small intestine, slower transit time, increased carbachol-stimulated ion secretion, and increased intestinal permeability. These alterations were accompanied by the loss of enteric neurons in the ileum and proximal colon in both submucosal and myenteric plexuses. A reduction in the number of enteric glia was only observed in the ileal myenteric plexus. Recovery of the microbiota restored intestinal function and stimulated enteric neurogenesis leading to increases in the number of enteric glia and neurons. Lipopolysaccharide (LPS) supplementation enhanced neuronal survival alongside bacterial depletion, but had no effect on neuronal recovery once the Abx-induced neuronal loss was established. In contrast, short-chain fatty acids (SCFA) were able to restore neuronal numbers after Abx-induced neuronal loss, demonstrating that SCFA stimulate enteric neurogenesis in vivo. CONCLUSIONS: Our results demonstrate a role for the gut microbiota in regulating the structure and function of the GI tract in a sex-independent manner. Moreover, the microbiota is essential for the maintenance of ENS integrity, by regulating enteric neuronal survival and promoting neurogenesis. Molecular determinants of the microbiota, LPS and SCFA, regulate enteric neuronal survival, while SCFA also stimulates neurogenesis. Our data reveal new insights into the role of the gut microbiota that could lead to therapeutic developments for the treatment of enteric neuropathies. Video abstract.


Subject(s)
Enteric Nervous System , Gastrointestinal Microbiome , Animals , Enteric Nervous System/physiology , Female , Gastrointestinal Microbiome/physiology , Intestine, Small , Male , Mice , Neuroglia , Neurons/physiology
6.
PLoS One ; 12(7): e0181453, 2017.
Article in English | MEDLINE | ID: mdl-28738080

ABSTRACT

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.


Subject(s)
Adaptation, Physiological/drug effects , Glucagon-Like Peptide 2/pharmacokinetics , Intestinal Mucosa/drug effects , Animals , Digestive System Surgical Procedures/methods , Disease Models, Animal , Glucagon-Like Peptide 2/metabolism , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/metabolism , Male , Parenteral Nutrition/methods , Parenteral Nutrition, Total/methods , Rats , Rats, Sprague-Dawley , Short Bowel Syndrome/metabolism
7.
J Pediatr Surg ; 45(5): 987-95, 2010 May.
Article in English | MEDLINE | ID: mdl-20438940

ABSTRACT

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.


Subject(s)
Adaptation, Physiological , Gastric Bypass/rehabilitation , Intestinal Mucosa/metabolism , Short Bowel Syndrome/rehabilitation , Animals , Cell Proliferation , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 7/metabolism , Gastric Bypass/adverse effects , Glucagon-Like Peptide 2/metabolism , Insulin-Like Growth Factor I/metabolism , Intestine, Small/cytology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Short Bowel Syndrome/etiology , Short Bowel Syndrome/physiopathology , Sodium-Glucose Transporter 1/metabolism
8.
J Pediatr Surg ; 44(8): 1552-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19635304

ABSTRACT

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.


Subject(s)
Digestive System Surgical Procedures/methods , Short Bowel Syndrome/surgery , Weight Gain/physiology , Adaptation, Physiological , Anastomosis, Surgical , Animals , Apoptosis/physiology , Disease Models, Animal , Glucagon-Like Peptide 2/metabolism , Glucagon-Like Peptide-2 Receptor , Intestinal Absorption/physiology , Postprandial Period , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/metabolism , Short Bowel Syndrome/physiopathology
9.
J Surg Res ; 152(2): 271-80, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19062041

ABSTRACT

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.


Subject(s)
Glucagon-Like Peptide 2/pharmacology , Intestine, Small/surgery , Parenteral Nutrition, Total , Animals , Cell Division/drug effects , Intestine, Small/cytology , Intestine, Small/pathology , Intestine, Small/physiology , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Time Factors
10.
Am J Physiol Gastrointest Liver Physiol ; 295(6): G1202-10, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18845573

ABSTRACT

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.


Subject(s)
Glucagon-Like Peptide 2/pharmacology , Interleukin-10/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects , Colitis/drug therapy , Cytokines/metabolism , Disease Models, Animal , Interleukin-10/deficiency , Male , Mice , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/drug effects
11.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G211-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17395898

ABSTRACT

Glucagon-like peptide-2 (GLP-2) is an important regulator of nutritional absorptive capacity with anti-inflammatory actions. We hypothesized that GLP-2 reduces intestinal mucosal inflammation by activation of vasoactive intestinal polypeptide (VIP) neurons of the submucosal plexus. Ileitis or colitis was induced in rats by injection of trinitrobenzene sulfonic acid (TNBS), or colitis was induced by administration of dextran sodium sulfate (DSS) in drinking water. Subsets of animals received (1-33)-GLP-2 (50 mug/kg sc bid) either immediately or 2 days after the establishment of inflammation and were followed for 3-5 days. The involvement of VIP neurons was assessed by concomitant administration of GLP-2 and the VIP antagonist [Lys(1)-Pro(2,5)-Arg(3,4)-Tyr(6)]VIP and by immunohistochemical labeling of GLP-2-activated neurons. In all models, GLP-2 treatment, whether given immediately or delayed until inflammation was established, resulted in significant improvements in animal weights, mucosal inflammation indices (myeloperoxidase levels, histological mucosal scores), and reduced levels of inflammatory cytokines (IFN-gamma, TNF-alpha, IL-1beta) and inducible nitric oxide synthase, with increased levels of IL-10 in TNBS ileitis and DSS colitis. Reduced rates of crypt cell proliferation and of apoptosis within crypts in inflamed tissues were also noted with GLP-2 treatment. These effects were abolished with coadministration of GLP-2 and the VIP antagonist. GLP-2 was shown to activate neurons and to increase the number of cells expressing VIP in the submucosal plexus of the ileum. These findings suggest that GLP-2 acts as an anti-inflammatory agent through activation of enteric VIP neurons, independent of proliferative effects. They support further studies to examine the role of neural signaling in the regulation of intestinal inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Enteric Nervous System/physiology , Glucagon-Like Peptide 2/physiology , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Dextran Sulfate , Disease Models, Animal , Glucagon-Like Peptide 2/therapeutic use , Ileitis/chemically induced , Ileitis/pathology , Male , Neurotensin/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/pharmacology , Trinitrobenzenesulfonic Acid , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/pharmacology , Vasoactive Intestinal Peptide/physiology , Vasoactive Intestinal Peptide/therapeutic use
12.
Am J Physiol Gastrointest Liver Physiol ; 286(6): G964-72, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14962847

ABSTRACT

Glucagon-like peptide-2 (GLP-2) is an intestinal trophic enteroendocrine peptide that is associated with intestinal adaptation following resection. Herein, we investigate the effects of GLP-2 in a total parenteral nutrition (TPN)-supported model of experimental short bowel syndrome. Juvenile Sprague-Dawley rats underwent a 90% small intestinal resection and jugular catheter insertion. Rats were randomized to three groups: enteral diet and intravenous saline infusion, TPN only, or TPN + 10 microg.kg(-1).h(-1) GLP-2. Nutritional maintenance was isocaloric and isonitrogenous. After 7 days, intestinal permeability was assessed by quantifying the urinary recovery of gavaged carbohydrate probes. The following day, animals were euthanized, and intestinal tissue was processed for morphological and crypt cell proliferation (CCP) analysis, apoptosis (caspase-3), and expression of SGLT-1 and GLUT-5 transport proteins. TPN plus GLP-2 treatment resulted in increased bowel and body weight, villus height, intestinal mucosal surface area, CCP, and reduced intestinal permeability compared with the TPN alone animals (P < 0.05). GLP-2 treatment induced increases in serum GLP-2 levels and intestinal SGLT-1 expression (P < 0.01) compared with either TPN or enteral groups. No differences were seen in the villus apoptotic index between resection groups. Enterally fed resected animals had a significant decrease in crypt apoptotic indexes compared with nontreated animals. This study demonstrates that GLP-2 alone, without enteral feeding, stimulates indexes of intestinal adaptation. Secondly, villus hypertrophy associated with adaptation was predominantly due to an increase in CCP and not to changes in apoptotic rates. Further studies are warranted to establish the mechanisms of action and therapeutic potential of GLP-2.


Subject(s)
Adaptation, Physiological , Intestines/drug effects , Intestines/physiopathology , Parenteral Nutrition, Total , Peptides/pharmacology , Short Bowel Syndrome/physiopathology , Short Bowel Syndrome/therapy , Animals , Caspase 3 , Caspases/metabolism , Cell Division/drug effects , Enzyme Activation , Glucagon-Like Peptide 2 , Glucagon-Like Peptides , Humans , Ileum/pathology , Intestinal Absorption , Intestinal Mucosa/pathology , Male , Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/metabolism , Permeability , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Short Bowel Syndrome/pathology , Sodium-Glucose Transporter 1
13.
Pediatr Res ; 55(1): 19-26, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14561780

ABSTRACT

Small bowel resection decreases brush border membrane (BBM) glucose uptake kinetics. Oral epidermal growth factor (EGF) returns net glucose transport across intact tissue to control levels despite persistence of a defect in BBM glucose uptake. The purpose of this study was to examine the effects of resection and EGF treatment on sodium-dependent glucose cotransporter 1 (SGLT-1) expression in distal remnant tissue. New Zealand White rabbits (1 kg) underwent 70% small bowel resection (R). One group of resected animals (R-EGF) received oral EGF (40 microg/kg, days 3-8). Distal remnant tissue was harvested 10 d after surgery, and compared with controls (C). Mucosal SGLT-1 mRNA was measured by Northern blot, BBM SGLT-1 content by Western blot, and villus distribution of SGLT-1 protein and mRNA by immunofluorescence and in situ hybridization. Western blot indicated BBM from both resected and EGF-treated tissue had decreased SGLT-1 content (C, 0.55 +/- 0.04; R, 0.35 +/- 0.04; R-EGF, 0.35 +/- 0.03 trace OD; n = 5; p < 0.05). Northern blot revealed no alterations in mucosal SGLT-1 mRNA content in any group. SGLT-1 protein and mRNA localization in control tissues was characterized by a gradual increase in stain intensity from the base of the villus to the villus tip. Resection altered SGLT-1 protein and mRNA expression along the villus axis with intensity being strongest in the mid-villus region and little expression at the tip of the villus. Oral EGF normalized SGLT-1 protein and mRNA expression to control patterns. Resection alters SGLT-1 protein and mRNA expression along the villus axis, despite no change in total mucosal SGLT-1 mRNA content. EGF normalized villus SGLT-1 protein and mRNA distribution, without altering overall BBM SGLT-1 content or mucosal mRNA levels.


Subject(s)
Epidermal Growth Factor/pharmacology , Intestine, Small/physiology , Intestine, Small/surgery , Membrane Glycoproteins/genetics , Monosaccharide Transport Proteins/genetics , Administration, Oral , Animals , Basement Membrane/physiology , Blotting, Northern , Blotting, Western , Gene Expression , Immunohistochemistry , In Situ Hybridization , Membrane Glycoproteins/metabolism , Monosaccharide Transport Proteins/metabolism , RNA, Messenger/analysis , Rabbits , Sodium-Glucose Transporter 1
SELECTION OF CITATIONS
SEARCH DETAIL
...