Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Sci Rep ; 9(1): 4200, 2019 03 12.
Article En | MEDLINE | ID: mdl-30862908

Mutations in the HNF4A gene cause MODY1 and are associated with an increased risk of Type 2 diabetes mellitus. On the other hand, incretins are hormones that potentiate reductions in blood glucose levels. Given the established role of incretin-based therapy to treat diabetes and metabolic disorders, we investigated a possible regulatory link between intestinal epithelial HNF4α and glucose-dependent insulinotropic polypeptide (GIP), an incretin that is specifically produced by gut enteroendocrine cells. Conditional deletion of HNF4α in the whole intestinal epithelium was achieved by crossing Villin-Cre and Hnf4αloxP/loxP C57BL/6 mouse models. GIP expression was measured by qPCR, immunofluorescence and ELISA. Gene transcription was assessed by luciferase and electrophoretic mobility shift assays. Metabolic parameters were analyzed by indirect calorimetry and dual-energy X-ray absorptiometry. HNF4α specific deletion in the intestine led to a reduction in GIP. HNF4α was able to positively control Gip transcriptional activity in collaboration with GATA-4 transcription factor. Glucose homeostasis and glucose-stimulated insulin secretion remained unchanged in HNF4α deficient mice. Changes in GIP production in these mice did not impact nutrition or energy metabolism under normal physiology but led to a reduction of bone area and mineral content, a well described physiological consequence of GIP deficiency. Our findings point to a novel regulatory role between intestinal HNF4α and GIP with possible functional impact on bone density.


Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Hepatocyte Nuclear Factor 4/metabolism , Intestinal Mucosa/metabolism , Transcription, Genetic , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/genetics , Gene Deletion , Hepatocyte Nuclear Factor 4/genetics , Mice , Mice, Transgenic
2.
Peptides ; 78: 51-8, 2016 Apr.
Article En | MEDLINE | ID: mdl-26820940

Selective GLP-1 secretagogues represent a novel potential therapy for type 2 diabetes mellitus. This study examined the GLP-1 secretory activity of the ethnomedicinal plant, Fagonia cretica, which is postulated to possess anti-diabetic activity. After extraction and fractionation extracts and purified compounds were tested for GLP-1 and GIP secretory activity in pGIP/neo STC-1 cells. Intracellular levels of incretin hormones and their gene expression were also determined. Crude F. cretica extracts stimulated both GLP-1 and GIP secretion, increased cellular hormone content, and upregulated gene expression of proglucagon, GIP and prohormone convertase. However, ethyl acetate partitioning significantly enriched GLP-1 secretory activity and this fraction underwent bioactivity-guided fractionation. Three isolated compounds were potent and selective GLP-1 secretagogues: quinovic acid (QA) and two QA derivatives, QA-3ß-O-ß-D-glycopyranoside and QA-3ß-O-ß-D-glucopyranosyl-(28→1)-ß-D-glucopyranosyl ester. All QA compounds activated the TGR5 receptor and increased intracellular incretin levels and gene expression. QA derivatives were more potent GLP-1 secretagogues than QA. This is the first time that QA and its naturally-occurring derivatives have been shown to activate TGR5 and stimulate GLP-1 secretion. These data provide a plausible mechanism for the ethnomedicinal use of F. cretica and may assist in the ongoing development of selective GLP-1 agonists.


Enteroendocrine Cells/drug effects , Gastric Inhibitory Polypeptide/agonists , Glucagon-Like Peptide 1/agonists , Hypoglycemic Agents/pharmacology , Proglucagon/agonists , Zygophyllaceae/chemistry , Cell Line , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glycosides/isolation & purification , Glycosides/pharmacology , Humans , Hypoglycemic Agents/isolation & purification , Incretins/agonists , Incretins/genetics , Incretins/metabolism , Plant Components, Aerial/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Proglucagon/biosynthesis , Proglucagon/genetics , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Triterpenes/isolation & purification , Triterpenes/pharmacology
3.
J Endocrinol ; 228(1): 39-48, 2016 Jan.
Article En | MEDLINE | ID: mdl-26483393

The incretin hormones glucagon-like peptide-1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted from intestinal endocrine cells, the so-called L- and K-cells. The cells are derived from a common precursor and are highly related, and co-expression of the two hormones in so-called L/K-cells has been reported. To investigate the relationship between the GLP1- and GIP-producing cells more closely, we generated a transgenic mouse model expressing a fluorescent marker in GIP-positive cells. In combination with a mouse strain with fluorescent GLP1 cells, we were able to estimate the overlap between the two cell types. Furthermore, we used primary cultured intestinal cells and isolated perfused mouse intestine to measure the secretion of GIP and GLP1 in response to different stimuli. Overlapping GLP1 and GIP cells were rare (∼5%). KCl, glucose and forskolin+IBMX increased the secretion of both GLP1 and GIP, whereas bombesin/neuromedin C only stimulated GLP1 secretion. Expression analysis showed high expression of the bombesin 2 receptor in GLP1 positive cells, but no expression in GIP-positive cells. These data indicate both expressional and functional differences between the GLP1-producing 'L-cell' and the GIP-producing 'K-cell'.


Enteroendocrine Cells/classification , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Receptors, Bombesin/analysis , Animals , Calcium/analysis , Cell Separation , Cells, Cultured , Enteroendocrine Cells/chemistry , Female , Flow Cytometry , Fluorescent Dyes , Gastric Inhibitory Polypeptide/analysis , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/analysis , Glucagon-Like Peptide 1/metabolism , Integrases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Bombesin/genetics
4.
PLoS One ; 10(8): e0134728, 2015.
Article En | MEDLINE | ID: mdl-26266950

Gastric inhibitory polypeptide (GIP, glucose-dependent insulinotropic polypeptide) is expressed by intestinal K cells to regulate glucose-induced insulin secretion. The impact of Roux-en Y bypass (RYGB) surgery on blood GIP is highly contraversial. This study was conducted to address the mechanism of controversy. GIP mRNA was examined in the intestine, and serum GIP was determined using Luminex and ELISA in diet-induced obese (DIO) mice. The assays were conducted in RYGB mice in fasting and fed conditions. Food preference, weight loss and insulin sensitivity were monitored in RYGB mice. In DIO mice, GIP mRNA was increased by 80% in all sections of the small intestine over the lean control. The increase was observed in both fasting and fed conditions. After RYGB surgery, the food-induced GIP expression was selectively reduced in the Roux-limb, but not in the biliopancreatic and common limbs of intestine in fed condition. Lack of stimulation by glucose or cholesterol contributed to the reduction. Jejunal mucosa of Roux-limb exhibited hypertrophy, but villous surface was decreased by the undigested food. Serum GIP (total) was significantly higher in the fasting condition, but not in the fed condition due to attenuated GIP response to food intake in RYGB mice. The GIP alteration was associated with chow diet preference, sustained weight loss and insulin sensitization in RYGB mice. RYGB increased serum GIP in the fasting, but not in the fed conditions. The loss of food-induced GIP response in Roux-limb of intestine likely contributes to the attenuated serum GIP response to feeding.


Gastric Bypass/adverse effects , Gastric Inhibitory Polypeptide/blood , Insulin/metabolism , Intestinal Mucosa/metabolism , Obesity/blood , Animals , Blood Glucose , Cholesterol/metabolism , Diet , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/genetics , Gene Expression Regulation , Humans , Insulin Resistance/genetics , Mice , Mice, Obese , Obesity/genetics , Obesity/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/blood
5.
Diabetes Metab Res Rev ; 30(5): 354-71, 2014 Jul.
Article En | MEDLINE | ID: mdl-24989141

Therapies targeting the action of incretin hormones have been under close scrutiny in recent years. The incretin effect has been defined as postprandial enhancement of insulin secretion by gut-derived factors. Likewise, incretin mimetics and incretin effect amplifiers are the two different incretin-based treatment strategies developed for the treatment of diabetes. Although, incretin mimetics produce effects very similar to those of natural incretin hormones, incretin effect amplifiers act by inhibiting dipeptidyl peptidase-4 (DPP-4) enzyme to increase plasma concentration of incretins and their biologic effects. Because glucagon-like peptide-1 (GLP-1) is an incretin hormone with various anti-diabetic actions including stimulation of glucose-induced insulin secretion, inhibition of glucagon secretion, hepatic glucose production and gastric emptying, it has been evaluated as a novel therapeutic agent for the treatment of type 2 diabetes mellitus (T2DM). GLP-1 also manifests trophic effects on pancreas such as pancreatic beta cell growth and differentiation. Because DPP-4 is the enzyme responsible for the inactivation of GLP-1, DPP-4 inhibition represents another potential strategy to increase plasma concentration of GLP-1 to enhance the incretin effect. Thus, anti-diabetic properties of these two classes of drugs have stimulated substantial clinical interest in the potential of incretin-based therapeutic agents as a means to control glucose homeostasis in T2DM patients. Despite this fact, clinical use of GLP-1 mimetics and DPP-4 inhibitors have raised substantial concerns owing to possible side effects of the treatments involving increased risk for pancreatitis, and C-cell adenoma/carcinoma. Thus, controversial issues in incretin-based therapies under development are reviewed and discussed in this manuscript.


Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Incretins/therapeutic use , Animals , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Exenatide , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin/metabolism , Humans , Incretins/adverse effects , Insulin/metabolism , Insulin Glargine , Insulin Secretion , Insulin, Long-Acting/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Peptides/therapeutic use , Receptors, Glucagon/agonists , Receptors, Glucagon/drug effects , Venoms/therapeutic use
6.
Diabetes ; 62(2): 510-8, 2013 Feb.
Article En | MEDLINE | ID: mdl-23099862

Glucagon and glucagon-like peptide-1 (GLP-1) are produced in pancreatic α-cells and enteroendocrine L-cells, respectively, in a tissue-specific manner from the same precursor, proglucagon, that is encoded by glucagon gene (Gcg), and play critical roles in glucose homeostasis. Here, we studied glucose homeostasis and ß-cell function of Gcg-deficient mice that are homozygous for a Gcg-GFP knock-in allele (Gcg(gfp/gfp)). The Gcg(gfp/gfp) mice displayed improved glucose tolerance and enhanced insulin secretion, as assessed by both oral glucose tolerance test (OGTT) and intraperitoneal glucose tolerance test (IPGTT). Responses of glucose-dependent insulinotropic polypeptide (GIP) to both oral and intraperitoneal glucose loads were unexpectedly enhanced in Gcg(gfp/gfp) mice, and immunohistochemistry localized GIP to pancreatic ß-cells of Gcg(gfp/gfp) mice. Furthermore, secretion of GIP in response to glucose was detected in isolated islets of Gcg(gfp/gfp) mice. Blockade of GIP action in vitro and in vivo by cAMP antagonism and genetic deletion of the GIP receptor, respectively, almost completely abrogated enhanced insulin secretion in Gcg(gfp/gfp) mice. These results indicate that ectopic GIP expression in ß-cells maintains insulin secretion in the absence of proglucagon-derived peptides (PGDPs), revealing a novel compensatory mechanism for sustaining incretin hormone action in islets.


Gastric Inhibitory Polypeptide/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Proglucagon/metabolism , Animals , Cyclic AMP/antagonists & inhibitors , Gastric Inhibitory Polypeptide/genetics , Gene Deletion , Gene Knock-In Techniques , Glucagon-Like Peptide-1 Receptor , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Incretins/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Mice , Proglucagon/analysis , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/metabolism
7.
Dev Dyn ; 241(12): 1986-92, 2012 Dec.
Article En | MEDLINE | ID: mdl-23027401

BACKGROUND: Glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) are hormones secreted by L and K cells, respectively, and by LK cells. To characterize L and K cells during development, we examined ileum from embryonic (e)- 12 to e-17. RESULTS: GLP-1 cells were first seen at e-15 and their number increased at e-17. At e-17, most GLP-1 cells co-expressed GIP. The transcription factors Pax6 and Pdx-1 are required for GIP expression, while Pax6 activates the expression of GLP-1. At e-17, the mucosa has GIP+ Pax6+, GIP+ Pdx-1+, GLP-1+ Pax6+, and GLP-1+ Pdx-1+ cells. Unlike ileal L cells of postnatal and adult mice, a subset of ileal L cells of e-17 embryos co-expressed GLP-1 and glucagon (Glu). Glu-positive cells contain proprotein-convertase 2 (PC2) and PC3/1, the enzymes responsible for Glu and GLP-1 synthesis, respectively. CONCLUSIONS: Our findings indicate that most GLP-1+ cells of ileum of e-17 embryos co-express GIP and, therefore, are LK cells. In addition, a subset of GLP-1+ cells of embryos but not of neonates co-express glucagon, indicating that the expression of Glu in GLP-1+ cells disappears after birth.


Embryo, Mammalian/metabolism , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Gene Expression Regulation, Developmental/physiology , Glucagon-Like Peptide 1/biosynthesis , Ileum/embryology , Animals , Embryo, Mammalian/cytology , Enteroendocrine Cells/cytology , Gastric Inhibitory Polypeptide/genetics , Glucagon-Like Peptide 1/genetics , Ileum/cytology , Mice , Proprotein Convertases/biosynthesis , Proprotein Convertases/genetics
8.
Endocrinology ; 153(12): 5782-95, 2012 Dec.
Article En | MEDLINE | ID: mdl-23064014

Enteroendocrine cells such as duodenal cholecystokinin (CCK cells) are generally thought to be confined to certain segments of the gastrointestinal (GI) tract and to store and release peptides derived from only a single peptide precursor. In the current study, however, transgenic mice expressing enhanced green fluorescent protein (eGFP) under the control of the CCK promoter demonstrated a distribution pattern of CCK-eGFP positive cells that extended throughout the intestine. Quantitative PCR and liquid chromatography-mass spectrometry proteomic analyses of isolated, FACS-purified CCK-eGFP-positive cells demonstrated expression of not only CCK but also glucagon-like peptide 1 (GLP-1), gastric inhibitory peptide (GIP), peptide YY (PYY), neurotensin, and secretin, but not somatostatin. Immunohistochemistry confirmed this expression pattern. The broad coexpression phenomenon was observed both in crypts and villi as demonstrated by immunohistochemistry and FACS analysis of separated cell populations. Single-cell quantitative PCR indicated that approximately half of the duodenal CCK-eGFP cells express one peptide precursor in addition to CCK, whereas an additional smaller fraction expresses two peptide precursors in addition to CCK. The coexpression pattern was further confirmed through a cell ablation study based on expression of the human diphtheria toxin receptor under the control of the proglucagon promoter, in which activation of the receptor resulted in a marked reduction not only in GLP-1 cells, but also PYY, neurotensin, GIP, CCK, and secretin cells, whereas somatostatin cells were spared. Key elements of the coexpression pattern were confirmed by immunohistochemical double staining in human small intestine. It is concluded that a lineage of mature enteroendocrine cells have the ability to coexpress members of a group of functionally related peptides: CCK, secretin, GIP, GLP-1, PYY, and neurotensin, suggesting a potential therapeutic target for the treatment and prevention of diabetes and obesity.


Cholecystokinin/biosynthesis , Enteroendocrine Cells/cytology , Gastric Inhibitory Polypeptide/biosynthesis , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Neurotensin/biosynthesis , Peptide YY/metabolism , Animals , Cell Lineage , Cell Separation , Diabetes Mellitus/prevention & control , Enteroendocrine Cells/metabolism , Flow Cytometry , Ghrelin/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry/methods , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Obesity/prevention & control , Promoter Regions, Genetic
9.
PLoS One ; 7(7): e40156, 2012.
Article En | MEDLINE | ID: mdl-22802954

Glucose-dependent insulinotropic polypeptide (GIP) is a gastrointestinal hormone that potentiates glucose-stimulated insulin secretion during a meal. Since GIP has also been shown to exert ß-cell prosurvival and adipocyte lipogenic effects in rodents, both GIP receptor agonists and antagonists have been considered as potential therapeutics in type 2 diabetes (T2DM). In the present study, we tested the hypothesis that chronically elevating GIP levels in a transgenic (Tg) mouse model would increase adipose tissue expansion and exert beneficial effects on glucose homeostasis. In contrast, although GIP Tg mice demonstrated enhanced ß-cell function, resulting in improved glucose tolerance and insulin sensitivity, they exhibited reduced diet-induced obesity. Adipose tissue macrophage infiltration and hepatic steatosis were both greatly reduced, and a number of genes involved in lipid metabolism/inflammatory signaling pathways were found to be down-regulated. Reduced adiposity in GIP Tg mice was associated with decreased energy intake, involving overexpression of hypothalamic GIP. Together, these studies suggest that, in the context of over-nutrition, transgenic GIP overexpression has the potential to improve hepatic and adipocyte function as well as glucose homeostasis.


Fatty Liver/prevention & control , Gastric Inhibitory Polypeptide/biosynthesis , Glucose/metabolism , Homeostasis , Obesity/etiology , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin Resistance/physiology , Male , Mice , Mice, Transgenic , Obesity/metabolism
10.
Orv Hetil ; 152(48): 1922-30, 2011 Nov 27.
Article Hu | MEDLINE | ID: mdl-22167829

The discovery of incretins-glucagon-like peptide (GLP)-1 and glucose-dependent insulinotrop peptide (GIP)-, clarification of their physiological properties as well as therapeutic application of incretin-based blood glucose lowering drugs opened new perspectives in the medical management of type 2 diabetes. New results of basic research investigations led to revaluation of the role of GIP in metabolic processes and a more established use of GLP-1 action. The article overviews the most relevant data of production and effects of incretins, as well as future possibilities of their therapeutic use.


Diabetes Mellitus/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptides/metabolism , Incretins/metabolism , Animals , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptides/biosynthesis , Glucagon-Like Peptides/genetics , Glucagon-Like Peptides/physiology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/biosynthesis , Incretins/genetics , Incretins/physiology
11.
Am J Physiol Endocrinol Metab ; 300(5): E923-32, 2011 May.
Article En | MEDLINE | ID: mdl-21304061

Dramatic improvement of type 2 diabetes is commonly observed after bariatric surgery. However, the mechanisms behind the alterations in glucose homeostasis are still elusive. We examined the effect of duodenal-jejunal bypass (DJB), which maintains the gastric volume intact while bypassing the entire duodenum and the proximal jejunum, on glycemic control, ß-cell mass, islet morphology, and changes in enteroendocrine cell populations in nonobese diabetic Goto-Kakizaki (GK) rats and nondiabetic control Wistar rats. We performed DJB or sham surgery in GK and Wistar rats. Blood glucose levels and glucose tolerance were monitored, and the plasma insulin, glucagon-like peptide-1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) levels were measured. ß-Cell area, islet fibrosis, intestinal morphology, and the density of enteroendocrine cells expressing GLP-1 and/or GIP were quantified. Improved postprandial glycemia was observed from 3 mo after DJB in diabetic GK rats, persisting until 12 mo after surgery. Compared with the sham-GK rats, the DJB-GK rats had an increased ß-cell area and a decreased islet fibrosis, increased insulin secretion with increased GLP-1 secretion in response to a mixed meal, and an increased population of cells coexpressing GIP and GLP-1 in the jejunum anastomosed to the stomach. In contrast, DJB impaired glucose tolerance in nondiabetic Wistar rats. In conclusion, although DJB worsens glucose homeostasis in normal nondiabetic Wistar rats, it can prevent long-term aggravation of glucose homeostasis in diabetic GK rats in association with changes in intestinal enteroendocrine cell populations, increased GLP-1 production, and reduced ß-cell deterioration.


Bariatric Surgery , Diabetes Mellitus, Type 2/surgery , Duodenum/physiology , Endocrine System/cytology , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Hyperglycemia/blood , Insulin-Secreting Cells/drug effects , Jejunum/physiology , Animals , Blood Glucose/metabolism , Body Composition/physiology , Body Weight/physiology , Diabetes Mellitus, Type 2/pathology , Endocrine System/drug effects , Endocrine System/metabolism , Enzyme-Linked Immunosorbent Assay , Fibrosis , Glucose Tolerance Test , Immunohistochemistry , Incretins/blood , Islets of Langerhans/pathology , Male , Rats , Rats, Wistar
12.
Regul Pept ; 167(1): 91-6, 2011 Feb 25.
Article En | MEDLINE | ID: mdl-21146561

Cold acclimation is initially associated with shivering thermogenesis in skeletal muscle followed by adaptive non-shivering thermogenesis, particularly in brown adipose tissue (BAT). In response, hyperphagia occurs to meet increased metabolic demand and thermoregulation. The present study investigates the effects of cold (4 ± 1 °C) acclimation and hyperphagia on circulating and intestinal levels of gastric inhibitory polypeptide (GIP) in rats. Pair fed animals were used as additional controls in some experiments. Cold acclimation for 42 days significantly (p<0.01) increased daily food intake. There was no corresponding change in body weight. However, body weights of pair fed cold exposed rats were significantly (p<0.01) reduced compared to controls and ad libitum fed cold exposed rats. By day 42, non-fasting plasma glucose was increased (p<0.05) by chronic cold exposure regardless of food intake. Corresponding plasma insulin concentrations were significantly (p<0.01) lower in pair fed cold exposed rats. Circulating GIP levels were elevated (p<0.05) in ad libitum fed cold acclimated rats on days 18 and 24, but returned to normal levels by the end of the study. The glycaemic response to oral glucose was improved (p<0.01) in all cold exposed rats, with significantly (p<0.05) elevated GIP responses in ad libitum fed rats and significantly (p<0.05) reduced insulin responses in pair fed rats. In keeping with this, insulin sensitivity was enhanced (p<0.05) in cold exposed rats compared to controls. By the end of the study, cold acclimated rats had significantly (p<0.01) increased BAT mass and intestinal concentrations of GIP and GLP-1 compared to controls, independent of food intake. These data indicate that changes in the secretion and actions of GIP may be involved in the metabolic adaptations to cold acclimation in rats.


Acclimatization/physiology , Cold Temperature , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Adipose Tissue, Brown/metabolism , Animals , Blood Glucose/metabolism , Body Temperature Regulation/physiology , Body Weight , Eating/physiology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glycemic Index , Hyperphagia/metabolism , Insulin/blood , Insulin Resistance , Intestinal Mucosa/metabolism , Male , Organ Size , Rats , Rats, Wistar
13.
Trends Endocrinol Metab ; 20(6): 280-6, 2009 Aug.
Article En | MEDLINE | ID: mdl-19595611

The incretin hormones, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), are produced predominantly by enteroendocrine cells and have multiple blood glucose-lowering effects. Recent years have seen a surge of interest in understanding the basic physiology and pathophysiology of incretins and in applying this knowledge to the treatment of diabetes and obesity. Considerable gains have been made in elucidating the mechanisms controlling incretin secretion, and there is growing evidence to suggest that incretins might be involved in the rapid reversal of diabetes observed in gastric bypass patients. Here, we review these recent advances and outline the multiple strategies being pursued to exploit the potential therapeutic benefits of GIP and GLP-1.


Incretins/physiology , Animals , Bariatric Surgery , Diabetes Mellitus/therapy , Dipeptidyl-Peptidase IV Inhibitors , Drug Discovery , Eating/physiology , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/physiology , Humans , Hypoglycemic Agents , Incretins/biosynthesis , Incretins/metabolism , Islets of Langerhans/metabolism , Molecular Mimicry , Obesity/therapy
14.
Oncol Rep ; 21(1): 107-12, 2009 Jan.
Article En | MEDLINE | ID: mdl-19082450

We have previously demonstrated that gastric and intestinal endocrine cell (End-cell) marker expression is important for assessment of the histogenesis of endocrine cell tumors. However, the End-cell phenotypes of carcinoid tumors in the rectum remain largely unclear. We therefore examined marker expression of rectal carcinoid tumors. We evaluated 20 rectal carcinoid tumors (as well as 8 from the stomach for comparison) phenotypically, using gastrin, gastric inhibitory polypeptide (GIP) and glucagons-like peptide-1 (GLP-1) as End-cell markers. Rectal carcinoid tumors were divided into 3 endocrine-gastric (e-G), 16 endocrine-gastric-and-intestinal mixed (e-GI), 1 endocrine-intestinal (e-I), and 0 endocrine-null (e-N) types, thus 19 (e-G+ e-GI types, 95%) had gastric phenotypic expression, while 17 (e-GI+ e-I types, 85%) harbored intestinal elements. Stomach carcinoid tumors were classified as 6 e-G and 2 e-N types, respectively. In conclusion, most rectal carcinoid tumors exhibited the e-GI type, suggesting the importance of gastric End-cell marker expression for histogenesis of the rectal carcinoid tumors. Further studies of pathological and biological analyses are needed to clarify the histogenesis of the carcinoid tumors.


Biomarkers, Tumor/analysis , Carcinoid Tumor/metabolism , Rectal Neoplasms/metabolism , Animals , Carcinoid Tumor/classification , Endocrine Cells/metabolism , Female , Gastric Inhibitory Polypeptide/biosynthesis , Gastrins/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Humans , Immunohistochemistry , Male , Middle Aged , Phenotype , Rectal Neoplasms/classification
15.
J Microsc ; 232(3): 493-7, 2008 Dec.
Article En | MEDLINE | ID: mdl-19094027

Some studies indicate that diabetes mellitus exerts an influence on the gastrointestinal tract and its diffuse neuroendocrine system (DNES) in regard to cellular density and neuroendocrine content. Since there is no data about relationship between experimentally induced non-insulin-dependent (type 2) diabetes mellitus (NIDDM) on the gut K cells, the aim of our study was to investigate immunohistochemical, stereological and ultrastructural changes of rat K cells after 12 days of dexamethasone treatment. Twenty male Wistar rats aged 30 days were given daily intraperitoneally 2 mg kg(-1) dexamethasone (group DEX, 10 rats) or saline (group C, 10 rats) for 12 days. Tissue specimens were obtained from each antrum with corpus and different parts of the small (SI) and large intestine (LI) of all animals. Immunohistochemistry was carried out using antisera against the GIP and insulin. Transmission electron microscopy was also used. Although, according to the literature data, rat K cells are present in the duodenum and jejunum and, to a lesser extent, in the ileum, in the present study we observed that those cells were abundant also in all parts of the LI. We observed generally that GIP-producing K cells were augmented in all parts of SI and decreased in the LI of DEX rats. Insulin immunoreactivity (ir) coexpressed with GIP-ir in K cells and was stronger in the SI of DEX rats as compared with C rats. We also found by electron microscopy that small intestinal K cells have features not only of GIP-secreted but also of insulin-secreted cells. We concluded that dexamethasone treatment caused proliferation of K cells in the rat SI, and simultaneously transformation of GIP-producing K cells to insulin-synthesizing cells.


Dexamethasone/administration & dosage , Diabetes Mellitus, Type 2/chemically induced , Gastric Inhibitory Polypeptide/biosynthesis , Intestine, Large/cytology , Intestine, Small/cytology , Stomach/cytology , Animals , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Rats , Rats, Wistar
16.
Am J Physiol Endocrinol Metab ; 295(3): E648-57, 2008 Sep.
Article En | MEDLINE | ID: mdl-18593849

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are incretin hormones that play important roles in maintaining glucose homeostasis and are being actively pursued as novel therapeutic agents for diabetes. GIP is produced by dispersed enteroendocrine cells and interestingly at times is coexpressed with GLP-1. We sought to determine the factors that selectively define GIP- vs. GLP-1-expressing cells. We performed comparative immunostaining of Pax6 and Pdx1 in GIP- and GLP-1-secreting cells. We investigated whether Pax6 and Pdx1 activate the human GIP promoter in control IEC-6 cells and GIP-expressing STC-1 cells. EMSA was performed to assess the binding of these transcription factors to the GIP promoter. Pax6 and Pdx1 consistently colocalized in GIP-immunoreactive cells. Cells that coexpress GIP and GLP-1 were Pax6 and Pdx1 positive, whereas cells expressing only GLP-1 were Pax6 positive but did not express Pdx1. GIP promoter activity was enhanced in IEC-6 cells by exogenous Pax6 or Pdx1 and diminished in STC-1 cells by inhibition of endogenous Pax6 or Pdx1 by dominant-negative forms. Promoter truncation analysis revealed a major loss of promoter activity when the sequence between -184 to -145 bp was deleted. EMSA studies indicated that Pax6 and Pdx1 bind to this proximal sequence of the human GIP promoter. Our findings indicate that concomitant expression of Pax6 and Pdx1 is important for GIP expression. Our results also suggest that the presence of Pdx1 defines whether GLP-1-expressing gastrointestinal L cells also coexpress GIP.


Eye Proteins/genetics , Gastric Inhibitory Polypeptide/biosynthesis , Homeodomain Proteins/genetics , Paired Box Transcription Factors/genetics , Proglucagon/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Adenoviridae/genetics , Amino Acid Sequence , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Genetic Vectors , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/genetics , Humans , Immunohistochemistry , Incretins/metabolism , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , PAX6 Transcription Factor , Plasmids , Rats , Rats, Wistar
17.
Am J Physiol Endocrinol Metab ; 295(4): E779-84, 2008 Oct.
Article En | MEDLINE | ID: mdl-18612044

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) regulate islet function after carbohydrate ingestion. Whether incretin hormones are of importance for islet function after ingestion of noncarbohydrate macronutrients is not known. This study therefore examined integrated incretin and islet hormone responses to ingestion of pure fat (oleic acid; 0.88 g/kg) or protein (milk and egg protein; 2 g/kg) over 5 h in healthy men, aged 20-25 yr (n=12); plain water ingestion served as control. Both intact (active) and total GLP-1 and GIP levels were determined as was plasma activity of dipeptidyl peptidase-4 (DPP-4). Following water ingestion, glucose, insulin, glucagon, GLP-1, and GIP levels and DPP-4 activity were stable during the 5-h study period. Both fat and protein ingestion increased insulin, glucagon, GIP, and GLP-1 levels without affecting glucose levels or DPP-4 activity. The GLP-1 responses were similar after protein and fat, whereas the early (30 min) GIP response was higher after protein than after fat ingestion (P<0.001). This was associated with sevenfold higher insulin and glucagon responses compared with fat ingestion (both P<0.001). After protein, the early GIP, but not GLP-1, responses correlated to insulin (r(2)=0.86; P=0.0001) but not glucagon responses. In contrast, after fat ingestion, GLP-1 and GIP did not correlate to islet hormones. We conclude that, whereas protein and fat release both incretin and islet hormones, the early GIP secretion after protein ingestion may be of primary importance to islet hormone secretion.


Dietary Fats/pharmacology , Dietary Proteins/pharmacology , Incretins/biosynthesis , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Pancreatic Hormones/metabolism , Adult , Blood Glucose/metabolism , Body Water/metabolism , Diet , Dipeptidyl Peptidase 4/blood , Fatty Acids, Nonesterified/blood , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon/blood , Glucagon-Like Peptide 1/biosynthesis , Humans , Insulin/blood , Male , Triglycerides/blood
18.
J Clin Endocrinol Metab ; 91(9): 3611-8, 2006 Sep.
Article En | MEDLINE | ID: mdl-16772347

CONTEXT: The mechanisms responsible for the ectopic adrenal expression of glucose-dependent insulinotropic peptide (GIP) receptor (GIPR) in GIP-dependent Cushing's syndrome (CS) are unknown. Chronic adrenal stimulation by ACTH in Cushing's disease or GIP in GIP-dependent ACTH-independent macronodular adrenal hyperplasia both lead to the induction of genes implicated in adrenal proliferation and steroidogenesis. OBJECTIVE: The objective of the study was to identify genes differentially expressed specifically in GIP-dependent CS that could be implicated in the ectopic expression of GIPR. METHODS: We used the Affymetrix U133 plus 2.0 microarray oligochips to compare the whole genome expression profile of adrenal tissues from five cases of GIP-dependent bilateral ACTH-independent macronodular adrenal hyperplasia with CS, one case of GIP-dependent unilateral adenoma with CS, five cases of ACTH-dependent hyperplasias, and a pool of adrenals from 62 normal individuals. RESULTS: After data normalization and statistical filtering, 723 genes with differential expression were identified, including 461 genes or sequences with a known functional implication, classified in eight dominant functional classes. Specific findings include repression of perilipin, the overexpression of 13 G protein-coupled receptors, and the potential involvement of Rho-GTPases. We also isolated 94 probe sets potentially linked to the formation of GIP-dependent nodules adjacent to the diffuse hyperplasia. These included probe sets related to the linker histone H1 and repression of RXRa and CCND2. The expression profiles for eight genes were confirmed by real-time RT-PCR. CONCLUSION: This study identified an extensive series of potentially novel target candidate genes that could be implicated in the molecular mechanisms of ectopic expression of the GIPR as well as in the multistep progression of GIP-dependent CS.


Adrenal Glands/pathology , Cushing Syndrome/genetics , Gastric Inhibitory Polypeptide/genetics , Receptors, Gastrointestinal Hormone/genetics , Adolescent , Adrenal Glands/metabolism , Adult , Cushing Syndrome/metabolism , Gastric Inhibitory Polypeptide/biosynthesis , Gene Expression Profiling , Humans , Hyperplasia , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA/chemistry , RNA/genetics , Receptors, Gastrointestinal Hormone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
Cell Metab ; 3(3): 153-65, 2006 Mar.
Article En | MEDLINE | ID: mdl-16517403

Gut peptides, exemplified by glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted in a nutrient-dependent manner and stimulate glucose-dependent insulin secretion. Both GIP and GLP-1 also promote beta cell proliferation and inhibit apoptosis, leading to expansion of beta cell mass. GLP-1, but not GIP, controls glycemia via additional actions on glucose sensors, inhibition of gastric emptying, food intake and glucagon secretion. Furthermore, GLP-1, unlike GIP, potently stimulates insulin secretion and reduces blood glucose in human subjects with type 2 diabetes. This article summarizes current concepts of incretin action and highlights the potential therapeutic utility of GLP-1 receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors for the treatment of type 2 diabetes.


Gastric Inhibitory Polypeptide/metabolism , Gastrointestinal Hormones/metabolism , Glucagon-Like Peptide 1/metabolism , Animals , Diabetes Mellitus, Type 2/therapy , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Pancreas/metabolism
20.
J Neurosci ; 25(7): 1816-25, 2005 Feb 16.
Article En | MEDLINE | ID: mdl-15716418

The hippocampal dentate gyrus (DG) is an area of active proliferation and neurogenesis within the adult brain. The molecular events controlling adult cell genesis in the hippocampus essentially remain unknown. It has been reported previously that adult male and female rats from the strains Sprague Dawley (SD) and spontaneously hypertensive (SHR) have a marked difference in proliferation rates of cells in the hippocampal DG. To exploit this natural variability and identify potential regulators of cell genesis in the hippocampus, hippocampal gene expression from male SHR as well as male and female SD rats was analyzed using a cDNA array strategy. Hippocampal expression of the gene-encoding glucose-dependent insulinotropic polypeptide (GIP) varied strongly in parallel with cell-proliferation rates in the adult rat DG. Moreover, robust GIP immunoreactivity could be detected in the DG. The GIP receptor is expressed by cultured adult hippocampal progenitors and throughout the granule cell layer of the DG, including progenitor cells. Thus, these cells have the ability to respond to GIP. Indeed, exogenously delivered GIP induced proliferation of adult-derived hippocampal progenitors in vivo as well as in vitro, and adult GIP receptor knock-out mice exhibit a significantly lower number of newborn cells in the hippocampal DG compared with wild-type mice. This investigation demonstrates the presence of GIP in the brain for the first time and provides evidence for a regulatory function for GIP in progenitor cell proliferation.


Dentate Gyrus/metabolism , Gastric Inhibitory Polypeptide/physiology , Stem Cells/cytology , Animals , Cell Division/drug effects , Dentate Gyrus/cytology , Female , Gastric Inhibitory Polypeptide/biosynthesis , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Profiling , Hypertension/genetics , Hypertension/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology
...