Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
Annu Rev Physiol ; 76: 561-83, 2014.
Article in English | MEDLINE | ID: mdl-24161075

ABSTRACT

Glucagon-like peptide-2 (GLP-2) is a 33-amino-acid proglucagon-derived peptide secreted from enteroendocrine L cells. GLP-2 circulates at low basal levels in the fasting period, and plasma levels rise rapidly after food ingestion. Renal clearance and enzymatic inactivation control the elimination of bioactive GLP-2. GLP-2 increases mesenteric blood flow and activates proabsorptive pathways in the gut, facilitating nutrient absorption. GLP-2 also enhances gut barrier function and induces proliferative and cytoprotective pathways in the small bowel. The actions of GLP-2 are transduced via a single G protein-coupled receptor (GLP-2R), expressed predominantly within the gastrointestinal tract. Disruption of GLP-2R signaling increases susceptibility to gut injury and impairs the adaptive mucosal response to refeeding. Sustained augmentation of GLP-2R signaling reduces the requirement for parenteral nutrition in human subjects with short-bowel syndrome. Hence GLP-2 integrates nutrient-derived signals to optimize mucosal integrity and energy absorption.


Subject(s)
Glucagon-Like Peptide 2/pharmacology , Glucagon-Like Peptide 2/physiology , Animals , Gastrointestinal Diseases/physiopathology , Gastrointestinal Motility/physiology , Gastrointestinal Tract/blood supply , Gastrointestinal Tract/physiology , Glucagon-Like Peptide-2 Receptor , Humans , Intestinal Absorption , Ischemia/physiopathology , Radiation Injuries/physiopathology , Receptors, Glucagon/biosynthesis , Receptors, Glucagon/drug effects , Receptors, Glucagon/metabolism , Short Bowel Syndrome/physiopathology , Signal Transduction
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 19(3): 368-371, 2017 Mar.
Article in Zh | MEDLINE | ID: mdl-28302214

ABSTRACT

In recent years, more and more studies have noted the close association between gut microbiota and the development and progression of obesity. Gut microbiota may act on obesity by increasing energy intake, affecting the secretion of intestinal hormones, inducing chronic systemic inflammation, and producing insulin resistance. This article reviews the association between childhood obesity and gut microbiota, as well as possible mechanisms, in an attempt to provide a reference for the etiology, prevention and treatment of childhood obesity.


Subject(s)
Gastrointestinal Microbiome , Obesity/etiology , Animals , Energy Metabolism , Glucagon-Like Peptide 2/physiology , Humans , Insulin Resistance , Obesity/microbiology , Obesity/prevention & control
3.
Curr Opin Gastroenterol ; 32(6): 461-466, 2016 11.
Article in English | MEDLINE | ID: mdl-27653163

ABSTRACT

PURPOSE OF REVIEW: To summarize and illuminate the recent findings regarding gastroduodenal mucosal defense mechanisms and the specific biomolecules involved in regulating this process, such as glucagon-like peptides (GLPs). RECENT FINDINGS: There has been a growing interest in luminal nutrient chemosensing and its physiological effects throughout the digestive system. From the ingestion of food in the oral cavity to the processing and absorption of nutrients in the intestines, nutrient chemosensing receptors signal the production and release of numerous bioactive peptides from enteroendocrine cells, such as the proglucagon-derived peptides. There has been a major emphasis on two proglucagon-derived peptides, namely GLP-1 and GLP-2, due to their apparent beneficial effect on gut structure, function, and on metabolic processes. As an incretin, GLP-1 not only enhances the effect and release of insulin on pancreatic ßcells but also has been implicated in having trophic effects on the intestinal epithelium. In addition, GLP-2, the other major proglucagon-derived peptide, has potent intestinotrophic effects, such as increasing the rate of mucosal stem cell proliferation, mucosal blood flow, and fluid absorption, as well as augmenting the rate of duodenal bicarbonate secretion to improve gastric mucosal health and longevity. SUMMARY: Understanding the mechanisms underlying nutrient chemosensing and how it relates to GLP release can further elucidate how the gut functions in response to cellular changes and disturbances. Furthermore, a more in-depth comprehension of GLP release and its tissue-specific effects will help improve the utility of GLP-1 and GLP-2 receptor agonists in clinical settings. This, in turn, should help patients suffering from intestinal failure, malabsorption, and mucosal injury.


Subject(s)
Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Nutritional Physiological Phenomena/physiology , Clinical Trials as Topic , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptides/therapeutic use , Humans
4.
Curr Opin Gastroenterol ; 30(2): 182-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24406477

ABSTRACT

PURPOSE OF REVIEW: Teduglutide, a recombinant analog of human glucagon-like peptide 2, has recently been approved in the US and Europe (Gattex and Revestive, respectively) as the first targeted treatment of short bowel syndrome-associated intestinal failure (SBS-IF). Glucagon-like peptide 2 improves structural and functional intestinal adaptation following intestinal resection by decelerating a rapid gastric emptying, by decreasing gastric hypersecretion, by increasing intestinal blood flow and by promoting intestinal growth. This review summarizes the findings from phase 2 and 3 studies preceding the US Food and Drug Administration and the European Medicines Agency approval of subcutaneous teduglutide for this orphan condition. RECENT FINDINGS: In a 3-week, phase 2, metabolic balance study, teduglutide increased intestinal wet weight absorption by approximately 700 g/day and reduced fecal energy losses by approximately 0.8 MJ/day (∼200 kcal/day). In two subsequent 24-week, phase 3 studies, teduglutide reduced the need for parenteral support in the same magnitude. Teduglutide had an acceptable tolerability profile, where adverse events generally were of gastrointestinal origin consistent with the known mechanism of action. SUMMARY: Teduglutide will add incremental benefit to the limited medical treatment armamentarium in SBS patients by maximizing intestinal absorption, decreasing fecal losses, thereby decreasing or even eliminating the need for parenteral support. Future research should target and implement other key hormones with similar and possible additive or synergistic effects, thereby further promoting structural and functional adaptation and intestinal rehabilitation in these severely disabled SBS patients.


Subject(s)
Gastrointestinal Agents/therapeutic use , Peptides/therapeutic use , Short Bowel Syndrome/drug therapy , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Glucagon-Like Peptide 2/physiology , Humans , Intestinal Absorption/drug effects , Parenteral Nutrition/methods , Short Bowel Syndrome/physiopathology
5.
Nutrients ; 16(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339669

ABSTRACT

Food intake regulation is a complex mechanism involving the interaction between central and peripheral structures. Among the latter, the gastrointestinal tract represents one of the main sources of both nervous and hormonal signals, which reach the central nervous system that integrates them and sends the resulting information downstream to effector organs involved in energy homeostasis. Gut hormones released by nutrient-sensing enteroendocrine cells can send signals to central structures involved in the regulation of food intake through more than one mechanism. One of these is through the modulation of gastric motor phenomena known to be a source of peripheral satiety signals. In the present review, our attention will be focused on the ability of the glucagon-like peptide 2 (GLP-2) hormone to modulate gastrointestinal motor activity and discuss how its effects could be related to peripheral satiety signals generated in the stomach and involved in the regulation of food intake through the gut-brain axis. A better understanding of the possible role of GLP-2 in regulating food intake through the gut-brain axis could represent a starting point for the development of new strategies to treat some pathological conditions, such as obesity.


Subject(s)
Brain-Gut Axis , Eating , Glucagon-Like Peptide 2 , Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptide 2/metabolism , Humans , Eating/physiology , Animals , Brain-Gut Axis/physiology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/metabolism , Brain/physiology , Brain/metabolism , Gastrointestinal Motility/physiology
6.
Nat Rev Endocrinol ; 20(9): 553-564, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38858581

ABSTRACT

Bone resorption follows a circadian rhythm, with a marked reduction in circulating markers of resorption (such as carboxy-terminal telopeptide region of collagen type I in serum) in the postprandial period. Several gut hormones, including glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide 1 (GLP1) and GLP2, have been linked to this effect in humans and rodent models. These hormones are secreted from enteroendocrine cells in the gastrointestinal tract in response to a variety of stimuli and effect a wide range of physiological processes within and outside the gut. Single GLP1, dual GLP1-GIP or GLP1-glucagon and triple GLP1-GIP-glucagon receptor agonists have been developed for the treatment of type 2 diabetes mellitus and obesity. In addition, single GIP, GLP1 and GLP2 analogues have been investigated in preclinical studies as novel therapeutics to improve bone strength in bone fragility disorders. Dual GIP-GLP2 analogues have been developed that show therapeutic promise for bone fragility in preclinical studies and seem to exert considerable activity at the bone material level. This Review summarizes the evidence of the action of gut hormones on bone homeostasis and physiology.


Subject(s)
Bone and Bones , Gastric Inhibitory Polypeptide , Gastrointestinal Hormones , Homeostasis , Humans , Homeostasis/physiology , Bone and Bones/metabolism , Bone and Bones/drug effects , Bone and Bones/physiology , Animals , Gastrointestinal Hormones/physiology , Gastrointestinal Hormones/metabolism , Gastric Inhibitory Polypeptide/metabolism , Bone Resorption/drug therapy , Bone Resorption/metabolism , Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptide 2/metabolism , Glucagon-Like Peptide 1/metabolism
7.
Curr Osteoporos Rep ; 11(3): 194-202, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913000

ABSTRACT

Nutritional status is an essential component in determining whole body energy homeostasis. The balance between energy/food intake and metabolism is governed by a range of hormones secreted from various parts of the body. Their subsequent dissemination via the blood results in a wide range of biological responses including satiety, hunger, and glucose uptake. The roles of these systemic hormones also extend to bone regulation with animal and clinical studies establishing a relationship between these regulatory pathways. This review covers the gastrointestinal hormones, ghrelin, PYY, GIP, GLP-1, and GLP-2, and the adipokines, leptin, and adiponectin and their roles in regulating bone homeostasis. Their known actions are reviewed, with an emphasis upon recent advances in understanding. Taken together, this review outlines an expanding appreciation of the interactions between bone mass and the nutritional control of whole body energy balance by gut and adipose tissue.


Subject(s)
Appetite/physiology , Bone and Bones/metabolism , Energy Metabolism/physiology , Gastrointestinal Hormones/physiology , Homeostasis/physiology , Adiponectin/physiology , Animals , Gastric Inhibitory Polypeptide/physiology , Ghrelin/physiology , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide 2/physiology , Humans , Leptin/physiology , Peptide YY/physiology
8.
Diabetes ; 71(7): 1388-1399, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35476805

ABSTRACT

Postprandial dyslipidemia is a metabolic condition commonly associated with insulin-resistant states, such as obesity and type 2 diabetes. It is characterized by the overproduction of intestinal chylomicron particles and excess atherogenic chylomicron remnants in circulation. We have previously shown that glucagon-like peptide 2 (GLP-2) augments dietary fat uptake and chylomicron production in insulin-resistant states; however, the underlying mechanisms remain unclear. Previous studies have implicated nitric oxide (NO) in the absorptive actions of GLP-2. In this study, we report a novel role for neuronal NO synthase (nNOS)-mediated NO generation in lipid uptake and chylomicron formation based on studies in C57BL/6J mice, nNOS-/- mice, and Syrian golden hamsters after intraduodenal and oral fat administration. GLP-2 treatment in wild-type (WT) mice significantly increased postprandial lipid accumulation and circulating apolipoprotein B48 protein levels, while these effects were abolished in nNOS-/- mice. nNOS inhibition in Syrian golden hamsters and protein kinase G (PKG) inhibition in WT mice also abrogated the effect of GLP-2 on postprandial lipid accumulation. These studies demonstrate a novel mechanism in which nNOS-generated NO is crucial for GLP-2-mediated lipid absorption and chylomicron production in both mouse and hamster models. Overall, our data implicate an nNOS-PKG-mediated pathway in GLP-2-mediated stimulation of dietary fat absorption and intestinal chylomicron production.


Subject(s)
Chylomicrons , Diabetes Mellitus, Type 2 , Animals , Chylomicrons/metabolism , Cricetinae , Dietary Fats/pharmacology , Glucagon-Like Peptide 2/pharmacology , Glucagon-Like Peptide 2/physiology , Insulin/metabolism , Intestinal Absorption , Mesocricetus , Mice , Mice, Inbred C57BL , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism
9.
Mol Cell Biochem ; 350(1-2): 13-27, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21153865

ABSTRACT

Tumor necrosis factor-alpha (TNF-α) is a multifunctional cytokine, which has the ability to produce cytotoxicity via induction of cell death and cell cycle arrest. Blocking the synthesis of protective proteins through a transcriptional inhibitor such as actinomycin D (Act D) sensitizes many cell types to TNF-α toxicity. Teduglutide, h[Gly(2)]GLP-2, is a protease-resistant synthetic analog of glucagon-like peptide-2 (GLP-2) which is an intestinotrophic peptide. In this study, we evaluated this potential of GLP-2 on apoptosis, cell proliferation, and oxidant-antioxidant system on a mouse model of intestinal injury induced by TNF-α/Act D. The intestinal injury was induced by intraperitoneal administration of 15 µg/kg TNF-α and 800 µg/kg Act D per mouse. Animals were injected subcutaneously 200 µg/kg h[Gly(2)]GLP-2 every 12 h for 10 consecutive days prior to the administration of TNF-α and Act D. The model of intestinal injury induced by TNF-α/Act D, which is the new animal model for the intestinal disorders, was characterized by the degeneration of intestinal mucosa, an increase in apoptotic index, expression of active caspase-3, lipid peroxidation and glutathione (GSH) levels, glutathione peroxidase (GPx) and superoxide dismutase (SOD) activities; a decrease in cell proliferation and catalase (CAT) activity. h[Gly(2)]GLP-2 pretreatment prevented the TNF-α/Act D-induced oxidative injury by a significant reduction in the intestinal injury, apoptotic index, expression of active caspase-3, lipid peroxidation and GSH levels, GPx and SOD activities; a markedly increase in cell proliferation, and CAT activity. These results demonstrate that GLP-2 has a protective, antiapoptotic, proliferative, and antioxidant effects against to TNF-α/Act D-induced intestinal injury. It is suggested that GLP-2 may potentially be useful as a therapeutic agent in TNF-α-mediated intestinal disorders.


Subject(s)
Antioxidants/metabolism , Apoptosis , Cell Proliferation , Glucagon-Like Peptide 2/physiology , Intestinal Diseases/pathology , Oxidants/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Proliferation/drug effects , Dactinomycin , Disease Models, Animal , Drug Evaluation, Preclinical , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Glucagon-Like Peptide 2/pharmacology , Glucagon-Like Peptide 2/therapeutic use , Intestinal Diseases/chemically induced , Intestinal Diseases/drug therapy , Intestinal Diseases/metabolism , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha
10.
Am J Physiol Endocrinol Metab ; 298(2): E156-66, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19920220

ABSTRACT

Glucagon-like peptide-2 (GLP-2) is a neuropeptide secreted from endocrine cells in the gut and neurons in the brain. GLP-2 stimulates intestinal crypt cell proliferation and mucosal blood flow while decreasing gastric emptying and gut motility. However, a GLP-2-mediated signaling network has not been fully established in primary cells. Since the GLP-2 receptor mRNA and protein were highly expressed in the mouse hippocampus, we further characterized that human (125)I-labeled GLP-2(1-33) specifically bound to cultured hippocampal neurons with K(d) = 0.48 nM, and GLP-2 acutely induced subcellular translocalization of the early gene c-Fos. Using the whole cell patch clamp, we recorded barium currents (I(Ba)) flowing through voltage-gated Ca(2+) channels (VGCC) in those neurons in the presence of GLP-2 with and without inhibitors. We showed that GLP-2 (20 nM) enhanced the whole cell I(Ba) mediated by L-type VGCC that was defined using an L-type Ca(2+) channel blocker (nifedipine, 10 microM). Moreover, GLP-2-potentiation of L-type VGCC was abolished in neurons pretreated with a PKA inhibitor (PKI(14-22), 1 microM). Finally, using a fluorescent nonmetabolized glucose analog (6-NBDG) tracing imaging, we showed that glucose was taken up directly by cultured neurons. GLP-2 increased 2-deoxy-d-[(3)H]glucose uptake that was dependent upon dosage, activation of PKA, and potentiation of L-type VGCC. We conclude that GLP-2 potentiates L-type VGCC activity through activating PKA signaling, partially stimulating glucose uptake by primary cultured hippocampal neurons. The potentiation of L-type VGCC may be physiologically relevant to GLP-2-induced neuroendocrine modulation of neurotransmitter release and hormone secretion.


Subject(s)
Calcium Channels, L-Type/metabolism , Glucagon-Like Peptide 2/physiology , Glucose/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Glucagon-Like Peptide-2 Receptor , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/analysis , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
11.
Gastroenterology ; 137(3): 997-1005, 1005.e1-4, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19482026

ABSTRACT

BACKGROUND & AIMS: Excessive postprandial lipemia is a prevalent condition that results from intestinal oversecretion of apolipoprotein B48 (apoB48)-containing lipoproteins. Glucagon-like peptide-2 (GLP-2) is a gastrointestinal-derived intestinotropic hormone that links nutrient absorption to intestinal structure and function. We investigated the effects of GLP-2 on intestinal lipid absorption and lipoprotein production. METHODS: Intestinal lipid absorption and chylomicron production were quantified in hamsters, wild-type mice, and Cd36(-/-) mice infused with exogenous GLP-2. Newly synthesized apoB48 was metabolically labelled in primary hamster jejunal fragments. Fatty acid absorption was measured, and putative fatty acid transporters were assessed by immunoblotting. RESULTS: Human GLP-2 increased secretion of the triglyceride (TG)-rich lipoprotein (TRL)-apoB48 following oral administration of olive oil to hamsters; TRL and cholesterol mass each increased 3-fold. Fast protein liquid chromatography profiling indicated that GLP-2 stimulated secretion of chylomicron/very low-density lipoprotein-sized particles. Moreover, GLP-2 directly stimulated apoB48 secretion in jejunal fragments cultured ex vivo, increased expression of fully glycosylated cluster of differentiation 36/fatty acid translocase (CD36), and induced intestinal absorption of [(3)H]triolein. The ability of GLP-2 to increase intestinal lipoprotein production was lost in Cd36(-/-) mice. CONCLUSIONS: GLP-2 stimulates intestinal apoB48-containing lipoprotein secretion, possibly through increased lipid uptake, via a pathway that requires CD36. These findings suggest that GLP-2 represents a nutrient-dependent signal that regulates intestinal lipid absorption and the assembly and secretion of TRLs from intestinal enterocytes.


Subject(s)
Chylomicrons/metabolism , Glucagon-Like Peptide 2/physiology , Intestinal Absorption/physiology , Jejunum/metabolism , Lipid Metabolism , Lipoproteins/metabolism , Triglycerides/metabolism , Animals , Apolipoprotein B-48/blood , Apolipoprotein B-48/metabolism , CD36 Antigens/metabolism , Cricetinae , Dietary Fats, Unsaturated/administration & dosage , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Glucagon-Like Peptide 2/pharmacology , Intestinal Absorption/drug effects , Lipoproteins/chemistry , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/chemistry , Triolein/metabolism
12.
J Pharmacol Exp Ther ; 335(2): 332-41, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719938

ABSTRACT

The effects of glucagon-like peptide 2 (GLP-2) on expression and activity of jejunal multidrug resistance-associated protein 2 (Mrp2; Abcc2) and glutathione transferase (GST) were evaluated. After GLP-2 treatment (12 µg/100 g b.wt. s.c., every 12 h, for 5 consecutive days), Mrp2 and the α class of GST proteins and their corresponding mRNAs were increased, suggesting a transcriptional regulation. Mrp2 was localized at the apical membrane of the enterocyte in control and GLP-2 groups, as detected by confocal immunofluorescence microscopy. As a functional assay, everted intestinal sacs were incubated in the presence of 1-chloro-2,4-dinitrobenzene in the mucosal compartment, and the glutathione-conjugated derivative, dinitrophenyl-S-glutathione (DNP-SG; model Mrp2 substrate), was detected in the same compartment by high-performance liquid chromatography. A significant increase in apical secretion of DNP-SG was detected in the GLP-2 group, consistent with simultaneous up-regulation of Mrp2 and GST. GLP-2 also promoted an increase in cAMP levels as detected in homogenates of intestinal mucosa. Treatment of rats with 2',3'-dideoxyadenosine (DDA), a specific inhibitor of adenylyl cyclase, abolished the increase in cAMP levels and Mrp2 protein promoted by GLP-2, suggesting cAMP as a mediator of Mrp2 modulation. Increased expression of Mrp2 and cAMP levels in response to GLP-2 occurred not only at the tip but also at the middle region of the villus, where constitutive expression of Mrp2 is normally low. In conclusion, our study suggests a role for GLP-2 in the prevention of cell toxicity of the intestinal mucosa by increasing Mrp2 chemical barrier function.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Glucagon-Like Peptide 2/pharmacology , Intestinal Mucosa/drug effects , Jejunum/drug effects , Adenylyl Cyclase Inhibitors , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Cyclic AMP/metabolism , Dideoxyadenosine/pharmacology , Enterocytes/drug effects , Enterocytes/enzymology , Enterocytes/metabolism , Enterocytes/pathology , Female , Fluorescent Antibody Technique , Glucagon-Like Peptide 2/physiology , Glutathione Transferase/biosynthesis , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Jejunum/enzymology , Jejunum/metabolism , Jejunum/pathology , Lactation/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Gut ; 58(8): 1091-103, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19240062

ABSTRACT

BACKGROUND AND AIMS: Obese and diabetic mice display enhanced intestinal permeability and metabolic endotoxaemia that participate in the occurrence of metabolic disorders. Our recent data support the idea that a selective increase of Bifidobacterium spp. reduces the impact of high-fat diet-induced metabolic endotoxaemia and inflammatory disorders. Here, we hypothesised that prebiotic modulation of gut microbiota lowers intestinal permeability, by a mechanism involving glucagon-like peptide-2 (GLP-2) thereby improving inflammation and metabolic disorders during obesity and diabetes. METHODS: Study 1: ob/ob mice (Ob-CT) were treated with either prebiotic (Ob-Pre) or non-prebiotic carbohydrates as control (Ob-Cell). Study 2: Ob-CT and Ob-Pre mice were treated with GLP-2 antagonist or saline. Study 3: Ob-CT mice were treated with a GLP-2 agonist or saline. We assessed changes in the gut microbiota, intestinal permeability, gut peptides, intestinal epithelial tight-junction proteins ZO-1 and occludin (qPCR and immunohistochemistry), hepatic and systemic inflammation. RESULTS: Prebiotic-treated mice exhibited a lower plasma lipopolysaccharide (LPS) and cytokines, and a decreased hepatic expression of inflammatory and oxidative stress markers. This decreased inflammatory tone was associated with a lower intestinal permeability and improved tight-junction integrity compared to controls. Prebiotic increased the endogenous intestinotrophic proglucagon-derived peptide (GLP-2) production whereas the GLP-2 antagonist abolished most of the prebiotic effects. Finally, pharmacological GLP-2 treatment decreased gut permeability, systemic and hepatic inflammatory phenotype associated with obesity to a similar extent as that observed following prebiotic-induced changes in gut microbiota. CONCLUSION: We found that a selective gut microbiota change controls and increases endogenous GLP-2 production, and consequently improves gut barrier functions by a GLP-2-dependent mechanism, contributing to the improvement of gut barrier functions during obesity and diabetes.


Subject(s)
Cecum/microbiology , Glucagon-Like Peptide 2/physiology , Inflammation/prevention & control , Obesity/complications , Probiotics/therapeutic use , Adiposity/drug effects , Adiposity/physiology , Animals , Bacteria/isolation & purification , Cecum/physiopathology , Endotoxemia/etiology , Endotoxemia/prevention & control , Glucagon-Like Peptide 2/agonists , Glucagon-Like Peptide 2/antagonists & inhibitors , Hepatitis/etiology , Hepatitis/prevention & control , Inflammation/etiology , Inflammation/microbiology , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Membrane Proteins/metabolism , Mice , Mice, Obese , Obesity/microbiology , Obesity/physiopathology , Occludin , Oxidative Stress/drug effects , Oxidative Stress/physiology , Permeability , Phosphoproteins/metabolism , Proglucagon/genetics , RNA, Messenger/genetics , Tight Junctions/metabolism , Zonula Occludens-1 Protein
14.
Neuropeptides ; 81: 102031, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32143816

ABSTRACT

Glucagon-like peptide-2 (GLP-2) has been reported to indirectly relax gastric smooth muscle. In the present study we investigated, through a combined mechanical and immunohistochemical approach, whether GLP-2 interferes with the electrical field stimulation (EFS)-induced vipergic relaxant responses and the mechanism through which it occurs. For functional experiments, strips from the mouse gastric fundus were mounted in organ baths for isometric recording of the mechanical activity. Vasoactive intestinal peptide (VIP) immunoreactivity in GLP-2 exposed specimens was also evaluated by immunohistochemistry. In carbachol pre-contracted strips, GLP-2 (20 nM) evoked a tetrodotoxin (TTX)-sensitive relaxation, similar in shape to the TTX-insensitive of 100 nM VIP. In the presence of GLP-2, VIP had no longer effects and no more response to GLP-2 was observed following VIP receptor saturation. EFS (4-16 Hz) induced a fast relaxant response followed, at the higher stimulation frequencies (≥ 8 Hz), by a slow one. This latter was abolished either by GLP-2 or VIP receptor saturation as well as by the VIP receptor antagonist, VIP 6-28 (10 µM). A decrease of VIP-immunoreactive nerve structures in the GLP-2 exposed specimens was observed. These results suggest that, in the mouse gastric fundus, GLP-2 influences the EFS-induced slow relaxant response by promoting neuronal VIP release.


Subject(s)
Gastric Fundus/physiology , Glucagon-Like Peptide 2/physiology , Neurons/physiology , Vasoactive Intestinal Peptide/physiology , Animals , Female , Mice , Muscle Contraction/physiology , Muscle, Smooth/physiology
15.
Curr Opin Clin Nutr Metab Care ; 12(5): 526-32, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19474717

ABSTRACT

PURPOSE OF REVIEW: The medical management of short bowel syndrome frequently requires lifelong parenteral nutrition. Methods of increasing intestinal absorption and reducing parenteral nutrition dependence, by improving postresection intestinal adaptation, are increasingly being explored. Glucagon-like peptide-2 (GLP-2) is an important intestinotrophic growth factor and mediator of intestinal adaptation. This review summarizes our current understanding of GLP-2 physiology and provides an update on clinical trials in short bowel syndrome and related conditions. RECENT FINDINGS: There is growing understanding how the effects of GLP-2 are mediated by downstream effectors such as insulin-like growth factor-1. In the treatment of short bowel syndrome, GLP-2 and the long-acting GLP-2 analogue teduglutide (Gattex) are effective in improving fluid absorption. A recent multicentre, placebo-controlled study demonstrates that this can translate into meaningful reductions in parenteral nutrition requirements. Treatment dose and timing of treatment initiation might influence the mucosal growth response. Most of the small intestine has to be preserved to facilitate the previously documented benefits of GLP-2 on bone metabolism. Therapeutic uses of GLP-2 in other gastrointestinal conditions are being explored. GLP-2 treatment appears well tolerated, although concerns about the long-term use of this growth-promoting agent remain. SUMMARY: GLP-2 therapy holds promise as an adjuvant treatment modality for short bowel syndrome and other gastrointestinal disorders.


Subject(s)
Adaptation, Physiological/drug effects , Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptide 2/therapeutic use , Short Bowel Syndrome/drug therapy , Adaptation, Physiological/physiology , Humans , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/growth & development , Intestinal Mucosa/pathology , Nutritional Requirements , Parenteral Nutrition , Peptides , Short Bowel Syndrome/physiopathology , Treatment Outcome
16.
World J Pediatr ; 15(6): 565-571, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31598832

ABSTRACT

BACKGROUND: The interaction of adropin, glucagon-like peptide-2 (GLP2), angiopoietin-like protein 4 (ANGPTL4), and with childhood obesity and glucose metabolism is inconsistent. This study is to evaluate the association of the three cytokines and glucose homeostasis. METHODS: This was a cross-sectional study of children with obesity ranging from 5 to 14 years compared to age- and sex-matched children of normal weight. Fasting plasma glucose (FPG), oral glucose tolerance test 2-hour plasma glucose (OGTT2hPG), and insulin (INS) were measured, and serum adropin, GLP2, and ANGPTL4 levels were measured by enzyme-linked immunosorbent assay. The body mass index (BMI), BMI-Z scores, waist-to-hip ratio (WHR), and homeostasis model assessment of insulin resistance (HOMA-IR) were calculated. RESULTS: Thirty-nine children (9.70 ± 1.71 years, 18 females) with obesity and 29 normal weight children (8.98 ± 1.98 years, 16 females) were assessed. The levels of INS, HOMA-IR and GLP2 of the obesity group were significantly higher than the controls (P < 0.05). Pearson correlation analysis showed that serum GLP2 was positively associated with WHR, FPG, and OGTT2hPG, and adropin was negatively associated with BMI, BMI-Z, WHR, INS, and HOMA-IR (all P < 0.05). Furthermore, GLP2 were negatively associated with adropin and ANGPTL4 (both P < 0.05). By binary logistic regression, adropin and GLP2 were found to be independent markers of obesity. Multiple linear regression showed that GLP2 was associated with OGTT2hPG, and adropin was associated with INS and HOMA-IR (all P < 0.05). CONCLUSIONS: Obese children had elevated GLP2 concentrations, and adropin and GLP2 associated with both childhood obesity and glucose homeostasis. Furthermore, there may be a physiologic interplay between adropin and GLP2 in obese children.


Subject(s)
Angiopoietin-Like Protein 4/physiology , Glucagon-Like Peptide 2/physiology , Glucose/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Pediatric Obesity/metabolism , Child , Cross-Sectional Studies , Female , Glucose Tolerance Test , Homeostasis , Humans , Male
17.
Semin Pediatr Surg ; 27(4): 237-241, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30342598

ABSTRACT

The treatment paradigm for pediatric patients with short bowel syndrome (SBS) and intestinal failure (IF) has changed significantly over recent years; the development of dedicated IF teams, refinements in PN and surgical treatments have greatly improved survival. The majority of SBS patients undergo intestinal adaptation such that nutrient absorption from enteral feeds increases and the child can come off of PN. This "adaptation" or upregulation in nutrient absorptive capacity is still poorly understood; the enteric hormone Glucagon like peptide 2 (GLP-2) appears to be a key regulator in this process. The development of Teduglutide, a long acting GLP-2 ligand as a therapy to specifically enhance adaptation has been anticipated as a further shift in the paradigm. This article reviews the physiology of GLP-2 with an emphasis on the known or potential roles in infants and children with SBS and IF. The results and implications of the present studies and approved indications for GLP-2 and its ligands are discussed. Finally, the potential future uses of GLP-2 ligands in the pediatric population are considered.


Subject(s)
Adaptation, Physiological/physiology , Gastrointestinal Agents/therapeutic use , Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptide 2/therapeutic use , Short Bowel Syndrome/drug therapy , Short Bowel Syndrome/physiopathology , Child , Humans , Infant
18.
Obes Surg ; 28(2): 506-512, 2018 02.
Article in English | MEDLINE | ID: mdl-28808865

ABSTRACT

BACKGROUND: This paper aimed to evaluate the influence of modified biliopancreatic diversion (BPD) on the levels of GLP-1 and GLP-2 and correlate them with satiety regulation. METHODS: This is a pilot prospective cohort study that evaluated six mildly obese individuals with type 2 diabetes mellitus, which underwent modified BPD and were followed-up for 12 months. Levels of GLP-1 and GLP-2 after a standard meal tolerance test were determined and correlated with satiety scores obtained by means of a visual analogue scale (VAS). RESULTS: There were significant changes in BMI (33 ± 2.2 versus 26.3 ± 2.2 kg/m2; p < 0.001), HbA1c (7.9 ± 1.6 versus 5.8 ± 1.2%; p = 0.026), total cholesterol (172.3 ± 11.1 versus 134.7 ± 16.1 mg/dL; p < 0.001), LDL-c (103.3 ± 13 versus 64.6 ± 12.2 mg/dL; p < 0.001), and postprandial GLP-2 (972.7 ± 326.2 versus 1993.2 ± 1024.7; p = 0. 044). None of the scores obtained in the VAS significantly changed after surgery. After surgery, there were significant correlations of VAS scores and GLP-1 levels in question 01 ("how hungry do you feel?"; R = -0.928; p = .008) and GLP-2 levels in questions 02 ("how full do you feel?" R = 0.943; p = 0.005) and 04 ("how much do you think you can eat now? R = -0.829; p = 0.042). CONCLUSIONS: Modified BPD does not lead to significant changes in satiety evaluated by the VAS; different aspects of satiety regulation are correlated with the postprandial levels of GLP-1 (hunger feeling) and GLP-2 (satiation feeling and desire to eat) 1 year after modified BPD, signaling a specific postoperative gut hormone-related modulation of appetite.


Subject(s)
Biliopancreatic Diversion/methods , Diabetes Mellitus, Type 2/surgery , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 2/blood , Obesity, Morbid/surgery , Satiation/physiology , Adult , Appetite Regulation/physiology , Biliopancreatic Diversion/adverse effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Female , Follow-Up Studies , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide 2/physiology , Humans , Male , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/complications , Pilot Projects , Postoperative Period , Postprandial Period/physiology
19.
Equine Vet J ; 50(6): 842-847, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29502360

ABSTRACT

BACKGROUND: Equine insulin dysregulation (ID) is a common and poorly understood disorder that increases the risk of laminitis. Recent data show that the condition may be associated with alteration of the enteroinsular axis and enhanced glucose bioavailability. Upregulation of glucagon-like peptide-2 (GLP-2), an intestinotrophic peptide, leads to enhanced nutrient uptake and metabolic dysfunction in other species. OBJECTIVES: The study aimed to 1) determine whether GLP-2 is differentially expressed in insulin-dysregulated ponies, compared with healthy ponies, and 2) confirm intestinal expression of the GLP-2 receptor in horses (eGLP-2R). STUDY DESIGN: Cohort study. METHODS: Fasting and post-prandial GLP-2 concentrations were measured in archived plasma samples obtained from 25 mixed-breed ponies during two feeding studies. Measurements were undertaken with an ELISA that was validated for equine use as part of the current study. Ponies were designated as healthy or insulin-dysregulated based on an oral glucose test, and the results were compared between groups. The gene expression of the eGLP-2R was determined with polymerase chain reaction. RESULTS: Basal, fasted plasma GLP-2 concentrations were higher in ponies with ID, compared with healthy ponies. Grazing increased GLP-2 in healthy, but not in insulin-dysregulated, ponies. The eGLP-2R gene was expressed in the small intestine and pancreas. MAIN LIMITATIONS: The study was performed with a relatively small sample size. The specificity of the GLP-2 assay could not be determined due to the lack of equine-specific assay standards. CONCLUSIONS: This study has demonstrated that GLP-2 may be important in the pathogenesis of equine ID and suggests that the eGLP-2R may be a novel therapeutic target for the treatment of equine ID.


Subject(s)
Glucagon-Like Peptide 2/physiology , Glucagon-Like Peptide-2 Receptor/metabolism , Horses/metabolism , Insulin/metabolism , Intestine, Small/metabolism , Animals , Cohort Studies , Eating/physiology , Enzyme-Linked Immunosorbent Assay/standards , Enzyme-Linked Immunosorbent Assay/veterinary , Fasting/metabolism , Female , Glucagon-Like Peptide 2/blood , Glucagon-Like Peptide 2/immunology , Glucose Tolerance Test/veterinary , Horses/blood , Male , Up-Regulation
20.
Neuropeptides ; 69: 66-75, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29703428

ABSTRACT

The central administration of glucagon-like peptide-2 (GLP-2) decreases blood pressure in rats. In the present study, we investigated the hypotensive effects of GLP-2 using spontaneously hypertensive rats (SHRs), an animal model of hypertension. The central administration of GLP-2 (0.6 µg) decreased mean arterial pressure (MAP) in SHRs (-24.1 ±â€¯4.5%; P < 0.05), but not in normotensive Wistar-Kyoto (WKY) rats (-10.6 ±â€¯7.4%; P > 0.05), whereas GLP-2 (6 µg) decreased MAP in WKY rats (-23.5 ±â€¯4.2%; P < 0.05) and SHRs (-46.7 ±â€¯11.6%; P < 0.01) under anesthesia with urethane and α-chloralose. Histological analyses revealed that the central administration of GLP-2 (6 µg) induced Fos immunoreactivity (Fos-IR) in the hypothalamic and medullary areas in WKY rats and SHRs. However, the distribution of Fos-IR in GABAergic neurons in the rostral ventrolateral medulla (RVLM) differed between WKY rats and SHRs. GLP-2 directly modulated the excitability of RVLM neurons in brainstem slices from SHRs, but not WKY rats. These results suggest that neuronal activity through the activation of GLP-2 receptors in the RVLM contributes to lowering blood pressure in SHRs.


Subject(s)
Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Brain/drug effects , Glucagon-Like Peptide 2/administration & dosage , Hypertension/physiopathology , Pressoreceptors/drug effects , Animals , Brain/metabolism , Catecholamines/metabolism , GABAergic Neurons/metabolism , Glucagon-Like Peptide 2/physiology , Hypertension/metabolism , Hypotension/chemically induced , Injections, Intraventricular , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Medulla Oblongata/physiology , Pressoreceptors/physiology , Rats, Inbred SHR , Rats, Inbred WKY , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL