Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Mol Sci ; 20(10)2019 May 23.
Article in English | MEDLINE | ID: mdl-31126070

ABSTRACT

A correlation between gastrointestinal (GI) inflammation and gut hormones has reported that inflammatory stimuli including bacterial endotoxins, lipopolysaccharides (LPS), TNFα, IL-1ß, and IL-6 induces high levels of incretin hormone leading to glucose dysregulation. Although incretin hormones are immediately secreted in response to environmental stimuli, such as nutrients, cytokines, and LPS, but studies of glucose-induced incretin secretion in an inflamed state are limited. We hypothesized that GI inflammatory conditions induce over-stimulated incretin secretion via an increase of glucose-sensing receptors. To confirm our hypothesis, we observed the alteration of glucose-induced incretin secretion and glucose-sensing receptors in a GI inflammatory mouse model, and we treated a conditioned media (Mϕ 30%) containing inflammatory cytokines in intestinal epithelium cells and enteroendocrine L-like NCI-H716 cells. In GI-inflamed mice, we observed that over-stimulated incretin secretion and insulin release in response to glucose and sodium glucose cotransporter (Sglt1) was increased. Incubation with Mϕ 30% increases Sglt1 and induces glucose-induced GLP-1 secretion with increasing intracellular calcium influx. Phloridzin, an sglt1 inhibitor, inhibits glucose-induced GLP-1 secretion, ERK activation, and calcium influx. These findings suggest that the abnormalities of incretin secretion leading to metabolic disturbances in GI inflammatory disease by an increase of Sglt1.


Subject(s)
Gastroenteritis/immunology , Glucose/immunology , Insulin/immunology , Sodium-Glucose Transporter 1/immunology , Animals , Cell Line , Cells, Cultured , Female , Gastric Inhibitory Polypeptide/immunology , Gastroenteritis/pathology , Glucagon-Like Peptide 1/immunology , Incretins/immunology , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice, Inbred C57BL
2.
Am J Physiol Endocrinol Metab ; 309(12): E1008-18, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26487006

ABSTRACT

Previous reports have suggested that the abrogation of gastric inhibitory polypeptide (GIP) signaling could be exploited to prevent and treat obesity and obesity-related disorders in humans. This study was designed to determine whether immunoneutralization of GIP, using a newly developed specific monoclonal antibody (mAb), would prevent the development of obesity. Specific mAb directed against the carboxy terminus of mouse GIP was identified, and its effects on the insulin response to oral and to intraperitoneal (ip) glucose and on weight gain were evaluated. Administration of mAb (30 mg/kg body wt, BW) to mice attenuated the insulin response to oral glucose by 70% and completely eliminated the response to ip glucose coadministered with human GIP. Nine-week-old C57BL/6 mice injected with GIP mAbs (60 mg·kg BW(-1)·wk(-1)) for 17 wk gained 46.5% less weight than control mice fed an identical high-fat diet (P < 0.001). No significant differences in the quantity of food consumed were detected between the two treatment groups. Furthermore, magnetic resonance imaging demonstrated that subcutaneous, omental, and hepatic fat were 1.97-, 3.46-, and 2.15-fold, respectively, lower in mAb-treated animals than in controls. Moreover, serum insulin, leptin, total cholesterol (TC), low-density lipoprotein (LDL), and triglycerides were significantly reduced, whereas the high-density lipoprotein (HDL)/TC ratio was 1.25-fold higher in treated animals than in controls. These studies support the hypothesis that a reduction in GIP signaling using a GIP-neutralizing mAb might provide a useful method for the treatment and prevention of obesity and related disorders.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Gastric Inhibitory Polypeptide/immunology , Obesity/immunology , Obesity/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Obesity/diagnosis , Signal Transduction/drug effects , Signal Transduction/immunology , Treatment Outcome
3.
Br J Nutr ; 108(1): 46-56, 2012 Jul 14.
Article in English | MEDLINE | ID: mdl-22260799

ABSTRACT

The effects of active immunisation with gastric inhibitory polypeptide (GIP) or (proline3)GIP-ovalbumin conjugates on insulin resistance, metabolic dysfunction, energy expenditure and cognition were examined in high-fat-fed mice. Normal mice were injected (subcutaneously) once every 14 d for 98 d with GIP-ovalbumin conjugates, with transfer to a high-fat diet on day 21. Active immunisation resulted in GIP antibody generation and significantly (P < 0·01 to P < 0·001) reduced circulating non-fasting plasma insulin concentrations compared to high-fat control mice from day 70 onwards. The glycaemic responses to intraperitoneal glucose or nutrient ingestion were significantly improved in all treated mice, with corresponding stimulated plasma insulin levels depressed compared to high-fat controls. These changes were associated with substantially (P < 0·001) improved glucose-lowering responses to exogenous insulin and decreases of muscle and fat TAG, pancreatic insulin, circulating total and LDL-cholesterol levels (P < 0·01 to P < 0·001). Treatment with GIP-ovalbumin conjugates was not associated with alterations in energy expenditure, indirect calorimetry or aspects of cognitive function. The observed changes were almost identical in GIP and (Pro3)GIP immunised mice and were independent of any effects on food intake or body weight. Further tests established that coupling of GIP peptides to ovalbumin abolished any intrinsic insulin-releasing or glucose-lowering activity. These results suggest that induction of GIP-neutralising antibodies with GIP-ovalbumin conjugates is an effective means of countering the metabolic abnormalities induced by high-fat feeding and does not adversely have an impact on a marker of cognition function or energy expenditure.


Subject(s)
Cognition/drug effects , Energy Metabolism/drug effects , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/immunology , Ovalbumin/chemistry , Ovalbumin/immunology , Animal Feed , Animals , Antibodies/blood , Blood Glucose , Diet , Dietary Fats/adverse effects , Homeostasis , Immunization , Insulin Resistance , Male , Mice , Time Factors
4.
Clin Chem ; 57(6): 849-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21515744

ABSTRACT

BACKGROUND: Glucose-dependent insulinotropic peptide (GIP) is an incretin peptide secreted by intestinal K cells that stimulates insulin secretion in a glucose-dependent manner. It is secreted as an active, intact 42-amino acid peptide GIP(1-42), which is rapidly degraded by dipeptidyl peptidase 4 to GIP(3-42), which is inactive. There is currently no described monoclonal antibody-based sandwich immunoassay to quantify concentrations of GIP(1-42), the active form of the peptide. METHODS: To create a sandwich ELISA for GIP(1-42), we generated a monoclonal antibody specific for the intact N-terminus of the peptide, which was further optimized to increase its affinity. We used this antibody as a conjugate antibody in a sandwich ELISA and paired it with an anti-total GIP capture monoclonal antibody to create a dual monoclonal sandwich ELISA for GIP(1-42). RESULTS: The sandwich ELISA was highly specific for GIP(1-42) and did not recognize GIP(3-42). The ELISA demonstrated a broad dynamic range and a lower limit of quantification of 5 ng/L. Using the ELISA, we were able to show that GIP(1-42) concentrations in healthy volunteers increased dramatically in the postprandial state compared to the fasting state. GIP(1-42) values were correlated with total GIP values overall; however, there was substantial interindividual variation. CONCLUSIONS: The use of an N-terminal-specific monoclonal antibody in a sandwich ELISA format provides a robust and convenient method for measuring concentrations of GIP(1-42), the active form of the incretin hormone. This ELISA should help to improve our understanding of the role of GIP(1-42) in regulating glucose-dependent insulin secretion.


Subject(s)
Antibodies, Monoclonal , Gastric Inhibitory Polypeptide/blood , Incretins/blood , Animals , Antibodies, Monoclonal/isolation & purification , Enzyme-Linked Immunosorbent Assay/methods , Gastric Inhibitory Polypeptide/immunology , Humans , Incretins/immunology , Mice
5.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G608-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20185691

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a hormone released from enteroendocrine K cells in response to meals. Posttranslational processing of the precursor protein pro-GIP at residue 65 by proprotein convertase subtilisin/kexin type 1 (PC1/3) in gut K cells gives rise to the established 42-amino-acid form of GIP (GIP(1-42)). However, the pro-GIP peptide sequence contains a consensus cleavage site for PC2 at residues 52-55 and we identified PC2 immunoreactivity in a subset of K cells, suggesting the potential existence of a COOH-terminal truncated GIP isoform, GIP(1-30). Indeed a subset of mouse and human K cells display GIP immunoreactivity with GIP antibodies directed to the mid portion of the peptide, but not with a COOH-terminal-directed GIP antibody, indicative of the presence of a truncated form of GIP. This population of cells represents approximately 5-15% of the total GIP-immunoreactive cells in mice, depending on the region of intestine, and is virtually absent in mice lacking PC2. Amidated GIP(1-30) and GIP(1-42) have comparable potency at stimulating somatostatin release in the perfused mouse stomach. Therefore, GIP(1-30) represents a naturally occurring, biologically active form of GIP.


Subject(s)
Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Peptide Fragments/metabolism , Proprotein Convertase 1/metabolism , Animals , Furin/biosynthesis , Gastric Inhibitory Polypeptide/immunology , Gastric Inhibitory Polypeptide/physiology , Gastric Mucosa/metabolism , Humans , Mice , Peptide Fragments/physiology , Proprotein Convertase 2/metabolism , Somatostatin/metabolism , Stomach/drug effects
6.
Scand J Immunol ; 72(1): 1-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20591069

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is involved in the aetiology of obesity induced by overnutrition, and blocking GIP activity may be valuable to anti-obesity treatment. However, GIP and GIP receptor are closely related to various brain functions which have caused very little data to be published concerning this cerebral functionality after blocking GIP activity. Here, we showed that active vaccination of mature rats with GIP immunoconjugates [GIP-keyhole limpet haemocyanin (KLH)] was associated with changes in body weight. Furthermore, we also observed significant changes in brain function and behaviour. Data indicated that GIP-KLH-immunized rats showed decreased spontaneous activity in the open field test, decreased cerebral glucose utilization assessed by 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (PET/CT), and increased apoptosis and proliferation of hippocampal granule cells marked by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) or proliferating cell nuclear antigen method. In conclusion, we have shown that vaccine-induced antibodies inhibited GIP activity in vivo and led to significant changes in brain function and behaviour, which underscore the need to address any potential problems GIP-targeted immunotherapy may involve in further research.


Subject(s)
Antibodies/blood , Apoptosis/immunology , Brain/immunology , Gastric Inhibitory Polypeptide/immunology , Glucose/immunology , Animals , Behavior, Animal , Body Weight/immunology , Eating/immunology , Immunization , Immunohistochemistry , In Situ Nick-End Labeling , Male , Maze Learning , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms , Statistics, Nonparametric
7.
Diabetes Obes Metab ; 12(9): 744-51, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20649625

ABSTRACT

AIM: Ablation of gastric inhibitory polypeptide (GIP) receptor signalling can prevent many of the metabolic abnormalities associated with dietary-induced obesity-diabetes. The present study was designed to assess the ability of active immunization against (Pro(3))GIP to counter metabolic dysfunction associated with diet-induced obesity in high-fat-fed mice. METHODS: Normal male Swiss NIH mice were injected (s.c.) once every 14 days for 98 days with complexed (Pro(3))GIP peptide, with transfer to a high-fat diet on day 21. RESULTS: Active immunization against (Pro(3))GIP resulted in circulating GIP antibody production and significantly (p < 0.05 p < 0.01) reduced circulating blood glucose concentrations compared to high-fat control mice from day 84 onwards. Glucose levels were not significantly different from lean controls. The glycaemic response to i.p. glucose was correspondingly improved (p < 0.01) in (Pro(3))GIP-immunized mice. Furthermore, circulating and glucose-stimulated plasma insulin levels were significantly (p < 0.01 to p < 0.001) depressed compared to high-fat control mice. Liver triglyceride, pancreatic insulin and circulating LDL-cholesterol levels were also significantly reduced in (Pro(3))GIP-immunized mice. These changes were independent of any effects on food intake or body weight. The glucose-lowering effect of native GIP was annulled in (Pro(3))GIP-immunized mice consistent with the induction of biologically effective GIP-specific neutralizing antibodies. CONCLUSION: These results suggest that immunoneutralization of GIP represents an effective means of countering the disruption of metabolic processes induced by high-fat feeding.


Subject(s)
Antibodies, Neutralizing/immunology , Blood Glucose/metabolism , Dietary Fats/administration & dosage , Gastric Inhibitory Polypeptide/immunology , Obesity/prevention & control , Animals , Body Weight/physiology , Gastric Inhibitory Polypeptide/administration & dosage , Gastric Inhibitory Polypeptide/blood , Insulin/blood , Insulin Resistance/physiology , Lipids/blood , Male , Mice , Obesity/drug therapy , Pancreas/metabolism
8.
Peptides ; 125: 170227, 2020 03.
Article in English | MEDLINE | ID: mdl-31805296

ABSTRACT

Gastric inhibitory polypeptide (GIP) is a regulatory peptide expressed in the mammalian upper small intestine, and both GIP and its receptor (GIPR) are expressed in the cortex and hippocampus regions of the brain as well. While learning and memory deficits have been observed in GIPR-/- mice, the effects of peripheral GIP immunoneutralization on motor-coordination, learning, and memory have not been examined. In the present study, adult GIPR-/- mice (KO) and age-matched wild-type C57BL/6 J mice (WT) received weekly vehicle PBS injections for 12 weeks, while a third group of wild-type mice were injected weekly for 12 weeks with 30 mg/kg body weight humanized GIP-mAb (AB) to assess the possibility of long-term effects of peripheral GIP antagonism on rodent memory and behavior. All mice groups then underwent a battery of tests that evaluated motor behavior, body coordination, and memory. Performance deficits in several memory studies after 12 weeks of treatment were demonstrated in KO, but not in AB or WT mice. Body coordination performance showed no significant differences among the 3 groups. A similar short-term study (3 injections over 9 days) was also conducted and the results were similar to those from the long-term study. Thus, short-term and long-term peripheral GIP antagonism by GIP-mAb did not appear to affect learning and memory in mice, consistent with the notion that the GIP-mAb does not cross the blood brain barrier. Furthermore, our studies indicate that GIP signaling in the brain appears to involve local neurocrine pathways.


Subject(s)
Antibodies, Monoclonal/pharmacology , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Learning/drug effects , Memory/drug effects , Motor Activity/drug effects , Receptors, Gastrointestinal Hormone/physiology , Animals , Disease Models, Animal , Gastric Inhibitory Polypeptide/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
9.
Diabetologia ; 52(9): 1724-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19533083

ABSTRACT

Gastric inhibitory polypeptide (GIP) is a physiological gut peptide secreted from the intestinal K-cells with well documented insulin-releasing actions. However, the GIP receptor is widely distributed in peripheral organs, including the pancreas, gut, adipose tissue, heart, adrenal cortex and brain, suggesting that it may have other functions. The presence of functional GIP receptors on adipocytes and the key role played by GIP in lipid metabolism and fat deposition suggest a possible beneficial effect of compromised GIP action in obesity and insulin resistance. Several key observations in animal models of obesity-related diabetes with chemically or genetically mediated biological GIP deficiency support this concept. Thus, obese diabetic animals with compromised GIP action due to peptide-based GIP receptor antagonists, small molecular weight GIP receptor antagonists, vaccination against GIP, genetic knockout of GIP receptor or targeted K-cell destruction are protected against obesity and associated metabolic disturbances. In addition, by causing preferential oxidation of fat, blockade of GIP signalling clears triacylglycerol deposits from liver and muscle, thereby restoring mechanisms for suppression of hepatic glucose output and improving insulin sensitivity. Emerging evidence also suggests that rapid cure of diabetes in grossly obese patients undergoing bypass surgery is mediated, in part, by surgical removal of GIP-secreting K-cells in the upper small intestine.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus/drug therapy , Gastric Inhibitory Polypeptide/therapeutic use , Obesity/drug therapy , Animals , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Female , Gastric Bypass , Gastric Inhibitory Polypeptide/immunology , Gastric Inhibitory Polypeptide/metabolism , Humans , Insulin/physiology , Insulin Resistance , Mice , Mice, Knockout , Obesity/prevention & control , Obesity/surgery , Ovariectomy , Receptors, Gastrointestinal Hormone/deficiency , Vaccines/therapeutic use
10.
Endocrinology ; 111(5): 1601-6, 1982 Nov.
Article in English | MEDLINE | ID: mdl-6751797

ABSTRACT

The action of gastric inhibitory polypeptide (GIP) antiserum on glucose tolerance and insulin secretion after an intraduodenal glucose load (600 mg/kg) was examined in anesthetized rats. In control experiments the insulin secretion was nearly doubled when glucose was administered intraduodenally, as compared to an iv glucose load to simulate the blood glucose curve after the intraduodenal glucose administration. After injection of GIP antiserum, the glucose curve resulting from the intraduodenal glucose load was slightly elevated and the insulin response was significantly reduced. No free GIP could be measured in the plasma of antibody-treated rats. However, the GIP antiserum did not offset the incretin effect of the intraduodenal glucose load completely. In control experiments the same amount of GIP antibody completely blocked the insulinotropic effect of exogenous porcine GIP (0.6 microgram/kg . h). In nonanesthetized rats serial oral glucose tolerance tests were performed for 14 days after injection of the GIP antiserum. Despite the blockage of endogenous GIP, the glucose tolerance did not change significantly in the antibody-treated group of rats as compared to a control group. These data indicate that GIP is not the exclusive incretin and that additional gut factors with insulinotropic activity exist.


Subject(s)
Gastric Inhibitory Polypeptide/deficiency , Gastrointestinal Hormones/deficiency , Glucose/pharmacology , Insulin/metabolism , Animals , Blood Glucose/metabolism , Duodenum , Gastric Inhibitory Polypeptide/immunology , Glucose/administration & dosage , Immune Sera , Insulin Secretion , Male , Rats , Rats, Inbred Strains
11.
Transplantation ; 70(5): 794-9, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11003360

ABSTRACT

BACKGROUND: The aim of this study was to investigate familial amyloidotic polyneuropathy, Portuguese type patients' endocrine cell content in the stomach and duodenum before and after liver transplantation, and to relate the findings to the patients' gastrointestinal disturbances. METHODS: Ten liver-transplanted familial amyloidotic polyneuropathy, Portuguese type patients and 10 healthy controls were seen. Endocrine cells were identified by immunohistochemistry and quantified with computerized image analysis. The activity of the cells was appraised by measurements of the cell secretory index and nuclear area. Clinical symptoms were obtained from the patients' medical records. RESULTS: After transplantation, a significant increase of several endocrine cell types were noted, and the pretransplant depletion of several types of endocrine cells disappeared. For no type of endocrine cell was any difference compared with controls noted after transplantation. There was no significant decrease of the amount of amyloid in the biopsies after liver transplantation. The patients' symptoms remained generally unchanged after transplantation, although a substantial time lapse between pretransplant evaluation and transplantation was present. CONCLUSIONS: Liver transplantation restores the endocrine cells in the upper part of the gastrointestinal tract. The restoration was not correlated with an improvement of the patients' symptoms. No decrease of the amyloid deposits was noted.


Subject(s)
Amyloid Neuropathies/surgery , Endocrine Glands/cytology , Enteroendocrine Cells/cytology , Liver Transplantation , Adult , Amyloid Neuropathies/pathology , Body Mass Index , Cell Count , Duodenum/chemistry , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/physiology , Female , Gastric Inhibitory Polypeptide/immunology , Humans , Immunohistochemistry , Male , Middle Aged , Pyloric Antrum/chemistry , Secretin/immunology , Serotonin/immunology , Somatostatin/immunology
12.
J Histochem Cytochem ; 31(6): 811-7, 1983 Jun.
Article in English | MEDLINE | ID: mdl-6188783

ABSTRACT

Although gastric inhibitory peptide (GIP) has never been detected outside the upper small intestine by immunochemical methods, GIP-like immunoreactivity has been demonstrated by immunocytochemistry in the glucagon/glicentin cells of pancreas, and gut. In the present study several GIP antisera (five polyclonal and one monoclonal) were tested on specimens from pancreas and intestines of several mammalian species, including man. Two of the polyclonal antisera and the monoclonal one stained cells in the upper small intestine only, while the other three also stained cells in the pancreas, ileum, and colon. Monoclonal anti-GIP did not stain GIP cells in man. The immunostaining produced could not be abolished by pretreatment of the antisera with glucagon or glicentin in excess, whereas small amounts of synthetic or natural porcine GIP prevented the immunostaining. Thus, three of the antisera are specific for GIP, while the other three recognize not only GIP but also GIP-like peptides. The results suggest that the glucagon/glicentin cells contain peptides distinct from GIP but sharing an immunodeterminant with GIP. The GIP-like immunoreactivity in the glucagon cells of the rat pancreas was not altered by infusion of GIP or by elimination of the bulk of endogenous GIP by resection of the upper small intestine, indicating that the GIP-like peptide is produced in the glucagon cells rather than accumulated from the circulation. The nature of this GIP-like peptide is unknown. Conceivably, it represents the cryptic portion of the glucagon precursor molecule. In some species a proportion of the GIP cells in the proximal small intestine displayed glicentin-like immunoreactivity as well, emphasizing the relationship between GIP cells on the one hand and glucagon/glicentin cells on the other.


Subject(s)
Gastric Inhibitory Polypeptide/immunology , Gastrointestinal Hormones/immunology , Glucagon/physiology , Protein Precursors/physiology , Animals , Antibodies/analysis , Antibody Specificity , Cats , Dogs , Guinea Pigs , Histocytochemistry , Humans , Immunochemistry , Intestine, Small/cytology , Pancreas/cytology , Proglucagon , Rabbits , Rats , Staining and Labeling , Swine/immunology
13.
Peptides ; 6 Suppl 3: 203-8, 1985.
Article in English | MEDLINE | ID: mdl-2870472

ABSTRACT

Methanol extracted skins from 84 species of amphibia were screened, measuring by RIAs: gastrin-CCK, VIP, calcitonin, GIP, PP and motilin. G-CCK-like immunoreactivity was found in 97.6%; VIP-like immunoreactivity in 41%; CT-like immunoreactivity in 34%; GIP-like immunoreactivity in 10%; PP-like immunoreactivity in 40% and MT-like immunoreactivity in 60% of the samples. The use of a sequence-specific radioimmunoassay and of gel-chromatography confirmed the caerulein-CCK-8-like nature of the immunoreactive material. Detected amounts of the other peptides (VIP, CT, GIP, PP, MT) were too low for bioassay or chromatographic studies, thus leaving the question open if they are due to some kind of unspecific interferences or, most likely, to species-specificity differences of the used antisera.


Subject(s)
APUD Cells/physiology , Anura/physiology , Peptides/physiology , Animals , Calcitonin/analysis , Calcitonin/immunology , Cholecystokinin/analysis , Cholecystokinin/immunology , Gastric Inhibitory Polypeptide/analysis , Gastric Inhibitory Polypeptide/immunology , Gastrins/analysis , Gastrins/immunology , Motilin/analysis , Motilin/immunology , Pancreatic Polypeptide/analysis , Pancreatic Polypeptide/immunology , Peptides/analysis , Peptides/immunology , Radioimmunoassay , Skin/analysis , Skin/immunology , Skin Physiological Phenomena , Species Specificity , Vasoactive Intestinal Peptide/analysis , Vasoactive Intestinal Peptide/immunology
14.
Regul Pept ; 9(1-2): 35-46, 1984 Sep.
Article in English | MEDLINE | ID: mdl-6505290

ABSTRACT

The Sephadex G-50 gel filtration profile of immunoreactive gastric inhibitory polypeptide (GIP) in porcine and human gastrointestinal mucosa was determined in assays with antisera obtained from five different groups working with GIP. Tissue was extracted in acid ethanol or using a boiling method. Three well-defined components were detected with three of the antisera: one component corresponding to natural porcine GIP (5 kDa GIP), one component corresponding to what has been called 8 kDa GIP, and one component somewhat larger than the latter. One antiserum did not measure 8 kDa GIP at all, while the fifth antiserum measured small amounts of 8 kDa GIP in porcine but not in human extracts. While the antisera measured the same amounts of GIP in porcine extracts, two of the antisera measured significantly more GIP than the remaining three in extracts of human mucosa. The elution position of human 5 kDa GIP differed significantly from that of porcine 5 kDa GIP. In addition to the identification of a new molecular form of GIP, and the demonstration of important specificity differences among GIP antisera currently in use, the present results indicate that human and porcine 5 kDa GIP differ in chemical composition.


Subject(s)
Gastric Inhibitory Polypeptide/isolation & purification , Gastric Mucosa/analysis , Gastrointestinal Hormones/isolation & purification , Intestinal Mucosa/analysis , Animals , Antibodies , Chromatography, Gel , Gastric Inhibitory Polypeptide/immunology , Humans , In Vitro Techniques , Species Specificity , Swine
15.
Ups J Med Sci ; 87(1): 81-5, 1982.
Article in English | MEDLINE | ID: mdl-6750891

ABSTRACT

The occurrence of gastrin and gastric inhibitory polypeptide (GIP) was investigated immunocytochemically in 17 foetal and neonatal human pancreata of gestational ages ranging from 12-41 weeks. GIP immunoreactive cells were observed in the pancreas of five foetuses with gestational ages of 18-20 weeks. These cells were located in islet-like cell clusters, at the base of tubular structures and among the exocrine-like acini. They were sometimes seen to emit a single long protrusion. The controls used, including preincubation of the antisera with anticomplement Clq, emphasized the specificity of the observed immunoreaction. No gastrin-immunoreactive cells were seen in any of the foetal or neonatal pancreata examined.


Subject(s)
Fetus/analysis , Gastric Inhibitory Polypeptide/analysis , Gastrointestinal Hormones/analysis , Islets of Langerhans/analysis , Animals , Gastric Inhibitory Polypeptide/immunology , Gastrins/analysis , Gastrins/immunology , Guinea Pigs , Humans , Immune Sera/immunology , In Vitro Techniques , Infant, Newborn , Islets of Langerhans/embryology , Rabbits
17.
Domest Anim Endocrinol ; 45(1): 49-54, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23751572

ABSTRACT

Glucose-dependent insulinotropic peptide (GIP), glucagon-like peptide (GLP)-1 and GLP-2 are hormones secreted from specialized K cells (GIP) and L cells (GLP-1, GLP-2) in the intestinal mucosa. These hormones play major roles in health and disease by modulating insulin secretion, satiety, and multiple intestinal functions. The aim of this study was to describe the distribution of K cells and L cells in the intestines of healthy cats. Samples of duodenum, mid-jejunum, ileum, cecum, and colon were collected from 5 cats that were euthanized for reasons unrelated to this study and had no gross or histologic evidence of gastrointestinal disease. Samples stained with rabbit-anti-porcine GIP, mouse-anti-(all mammals) GLP-1, or rabbit-anti-(all mammals) GLP-2 antibodies were used to determine the number of cells in 15 randomly selected 400× microscopic fields. In contrast to other mammals (eg, dogs) in which K cells are not present in the ileum and aborally, GIP-expressing cells are abundant throughout the intestines in cats (>6/high-power field in the ileum). Cells expressing GLP-1 or GLP-2 were most abundant in the ileum (>9/high-power field) as in other mammals, but, although GLP-1-expressing cells were abundant throughout the intestines, GLP-2-expressing cells were rarely found in the duodenum. In conclusion, the distribution of GIP-secreting K cells in cats is different from the distribution of K cells that is described in other mammals. The difference in distribution of GLP-2- and GLP-1-expressing cells suggests that more than 1 distinct population of L cells is present in cats.


Subject(s)
Cats/anatomy & histology , Glucagon-Like Peptide 1/analysis , Intestines/cytology , Neuroendocrine Cells/cytology , Animals , Antibodies , Cecum/cytology , Colon/cytology , Duodenum/cytology , Female , Gastric Inhibitory Polypeptide/analysis , Gastric Inhibitory Polypeptide/immunology , Glucagon-Like Peptide 1/immunology , Glucagon-Like Peptide 2/analysis , Glucagon-Like Peptide 2/immunology , Ileum/cytology , Immunohistochemistry , Intestines/chemistry , Jejunum/cytology , Male , Mice , Neuroendocrine Cells/chemistry , Neuroendocrine Cells/classification , Rabbits , Species Specificity
18.
Anat Histol Embryol ; 40(3): 163-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21133986

ABSTRACT

The presence and distribution of glucose-dependent insulinotropic polypeptide or gastric inhibitory polypeptide (GIP), gastric-releasing peptide (GRP) and glucagon immunoreactivity were studied in the small intestine of the New Hampshire chicken using immunohistochemistry. This is the first report of the presence of GIP-immunoreactive (ir) cells in avian small intestine. GIP, GRP and glucagon immunoreactivity was localized in the epithelium of the villi and crypts of the duodenum, jejunum and ileum. In particular, both in the duodenum and in the jejunum immunoreactive endocrine cells to GIP, GRP and glucagon were observed. In the ileum, we noticed GIP-ir and glucagon-ir cells. GRP-ir was found in nerve fibres of all three segments of the small intestine. The distribution of these bioactive agents in the intestinal tract of the chicken suggests that GIP and glucagon may play a role in the enteropancreatic axis in which intestinal peptides modulate pancreas secretion.


Subject(s)
Chickens , Gastric Inhibitory Polypeptide/analysis , Gastrin-Releasing Peptide/analysis , Glucagon/analysis , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Intestine, Small/chemistry , Animals , Duodenum/chemistry , Duodenum/cytology , Fluorescent Antibody Technique , Gastric Inhibitory Polypeptide/immunology , Gastric Mucosa/chemistry , Gastric Mucosa/cytology , Gastrin-Releasing Peptide/immunology , Glucagon/immunology , Ileum/chemistry , Ileum/cytology , Intestine, Small/cytology , Jejunum/chemistry , Jejunum/cytology , Male , Pancreas/metabolism
19.
Best Pract Res Clin Endocrinol Metab ; 23(4): 425-32, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19748060

ABSTRACT

The measurement of the incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), using immunologically based assays is made difficult by the fact that the processing of the precursor molecules gives rise to a number of different peptides which cross-react with antisera raised against the two hormones. For GLP-1, the picture is further complicated because of the necessity to differentiate between the intestinal and pancreatic proglucagon products. Finally, once secreted, both incretins are rapidly degraded by the enzyme dipeptidyl peptidase-4 (DPP-4) to generate metabolites which have lost their insulinotropic activities. These metabolites are the major circulating forms of the incretins, accounting for 60-80% of total immunoreactive GLP-1 and GIP in the peripheral plasma, while concentrations of the intact forms can be very low and, in some cases, barely detectable. The use of highly specific assays using well-characterised antisera and careful sample handling is therefore required for a reliable determination of incretin hormone concentrations.


Subject(s)
Gastric Inhibitory Polypeptide/analysis , Glucagon-Like Peptide 1/analysis , Radioimmunoassay/methods , Cross Reactions , Dipeptidyl Peptidase 4/metabolism , Gastric Inhibitory Polypeptide/immunology , Glucagon-Like Peptide 1/immunology , Humans , Proglucagon/metabolism
20.
Biol Chem ; 390(1): 75-80, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18937625

ABSTRACT

Recent research suggests that long-term ablation of gastric inhibitory polypeptide (GIP) receptor signalling can reverse or prevent many of the metabolic abnormalities associated with dietary and genetically induced obesity-diabetes. The present study was designed to assess the sub-chronic effects of passive or active immunisation against GIP in ob/ob mice. Initial acute administration of GIP antibody together with oral glucose in ob/ob mice significantly increased the glycaemic excursion compared to controls (p<0.05). This was associated with a significant reduction (p<0.05) in the overall glucose-mediated insulin response. However, sub-chronic passive GIP immunisation was not associated with any changes in body weight, food intake or metabolic control. In contrast, active immunisation against GIP for 56 days in young ob/ob mice resulted in significantly (p<0.05) reduced circulating plasma glucose concentrations on day 56 compared to controls. There was a tendency for decreased circulating insulin in GIP immunised mice. The glycaemic response to intraperitoneal glucose was correspondingly improved (p<0.05) in mice immunised against GIP. Glucose-stimulated insulin levels were not significantly different from controls. Furthermore, insulin sensitivity was similar in mice immunised against GIP and respective controls. Overall, the results reveal that active, as opposed to passive, immunisation against GIP improves blood glucose control ob/ob mice.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus/blood , Disease Models, Animal , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Gastric Inhibitory Polypeptide/immunology , Obesity/blood , Vaccination , Animals , Antibodies/immunology , Diabetes Mellitus/metabolism , Female , Gastric Inhibitory Polypeptide/blood , Homeostasis/immunology , Humans , Immunization, Passive , Insulin/metabolism , Male , Mice , Obesity/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL