Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
JCI Insight ; 8(10)2023 05 22.
Article in English | MEDLINE | ID: mdl-37212283

ABSTRACT

Central glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) signaling is critical in GIP-based therapeutics' ability to lower body weight, but pathways leveraged by GIPR pharmacology in the brain remain incompletely understood. We explored the role of Gipr neurons in the hypothalamus and dorsal vagal complex (DVC) - brain regions critical to the control of energy balance. Hypothalamic Gipr expression was not necessary for the synergistic effect of GIPR/GLP-1R coagonism on body weight. While chemogenetic stimulation of both hypothalamic and DVC Gipr neurons suppressed food intake, activation of DVC Gipr neurons reduced ambulatory activity and induced conditioned taste avoidance, while there was no effect of a short-acting GIPR agonist (GIPRA). Within the DVC, Gipr neurons of the nucleus tractus solitarius (NTS), but not the area postrema (AP), projected to distal brain regions and were transcriptomically distinct. Peripherally dosed fluorescent GIPRAs revealed that access was restricted to circumventricular organs in the CNS. These data demonstrate that Gipr neurons in the hypothalamus, AP, and NTS differ in their connectivity, transcriptomic profile, peripheral accessibility, and appetite-controlling mechanisms. These results highlight the heterogeneity of the central GIPR signaling axis and suggest that studies into the effects of GIP pharmacology on feeding behavior should consider the interplay of multiple regulatory pathways.


Subject(s)
Hypothalamus , Receptors, Gastrointestinal Hormone , Body Weight , Brain Stem/metabolism , Gastric Inhibitory Polypeptide/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Feeding Behavior , Animals
2.
Cell Metab ; 33(4): 833-844.e5, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33571454

ABSTRACT

Uncertainty exists as to whether the glucose-dependent insulinotropic polypeptide receptor (GIPR) should be activated or inhibited for the treatment of obesity. Gipr was recently demonstrated in hypothalamic feeding centers, but the physiological relevance of CNS Gipr remains unknown. Here we show that HFD-fed CNS-Gipr KO mice and humanized (h)GIPR knockin mice with CNS-hGIPR deletion show decreased body weight and improved glucose metabolism. In DIO mice, acute central and peripheral administration of acyl-GIP increases cFos neuronal activity in hypothalamic feeding centers, and this coincides with decreased body weight and food intake and improved glucose handling. Chronic central and peripheral administration of acyl-GIP lowers body weight and food intake in wild-type mice, but shows blunted/absent efficacy in CNS-Gipr KO mice. Also, the superior metabolic effect of GLP-1/GIP co-agonism relative to GLP-1 is extinguished in CNS-Gipr KO mice. Our data hence establish a key role of CNS Gipr for control of energy metabolism.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Gastric Inhibitory Polypeptide/pharmacology , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction/drug effects , Animals , Central Nervous System/metabolism , Diet, High-Fat , Gastric Inhibitory Polypeptide/chemistry , Glucagon-Like Peptide 1/pharmacology , Humans , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/metabolism , Obesity/pathology , Obesity/prevention & control , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics
3.
J Clin Invest ; 129(9): 3786-3791, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31403469

ABSTRACT

Nutrient excess, a major driver of obesity, diminishes hypothalamic responses to exogenously administered leptin, a critical hormone of energy balance. Here, we aimed to identify a physiological signal that arises from excess caloric intake and negatively controls hypothalamic leptin action. We found that deficiency of the gastric inhibitory polypeptide receptor (Gipr) for the gut-derived incretin hormone GIP protected against diet-induced neural leptin resistance. Furthermore, a centrally administered antibody that neutralizes GIPR had remarkable antiobesity effects in diet-induced obese mice, including reduced body weight and adiposity, and a decreased hypothalamic level of SOCS3, an inhibitor of leptin actions. In contrast, centrally administered GIP diminished hypothalamic sensitivity to leptin and increased hypothalamic levels of Socs3. Finally, we show that GIP increased the active form of the small GTPase Rap1 in the brain and that its activation was required for the central actions of GIP. Altogether, our results identify GIPR/Rap1 signaling in the brain as a molecular pathway linking overnutrition to the control of neural leptin actions.


Subject(s)
Hypothalamus/metabolism , Incretins/metabolism , Leptin/metabolism , Obesity/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , Adiposity/genetics , Animals , Incretins/genetics , Leptin/genetics , Mice , Obesity/genetics , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , rap1 GTP-Binding Proteins/genetics
4.
Cell Metab ; 30(5): 987-996.e6, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31447324

ABSTRACT

Ambiguity regarding the role of glucose-dependent insulinotropic polypeptide (GIP) in obesity arises from conflicting reports asserting that both GIP receptor (GIPR) agonism and antagonism are effective strategies for inhibiting weight gain. To enable identification and manipulation of Gipr-expressing (Gipr) cells, we created Gipr-Cre knockin mice. As GIPR-agonists have recently been reported to suppress food intake, we aimed to identify central mediators of this effect. Gipr cells were identified in the arcuate, dorsomedial, and paraventricular nuclei of the hypothalamus, as confirmed by RNAscope in mouse and human. Single-cell RNA-seq identified clusters of hypothalamic Gipr cells exhibiting transcriptomic signatures for vascular, glial, and neuronal cells, the latter expressing somatostatin but little pro-opiomelanocortin or agouti-related peptide. Activation of Gq-DREADDs in hypothalamic Gipr cells suppressed food intake in vivo, which was not obviously additive with concomitant GLP1R activation. These data identify hypothalamic GIPR as a target for the regulation of energy balance.


Subject(s)
Eating/physiology , Hypothalamus/cytology , Neurons/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Aged, 80 and over , Animals , Eating/drug effects , Female , Gastric Inhibitory Polypeptide/metabolism , Gene Knock-In Techniques , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/genetics
5.
FASEB J ; 33(4): 5389-5398, 2019 04.
Article in English | MEDLINE | ID: mdl-30702925

ABSTRACT

With an increasing body of evidence regarding GPCR oligomerization and its clinical implications over the last decade, the modulation and dynamics of GPCR homo- and hetero-oligomers has more recently become an area of intense research focus. Previously, our lab showed in vitro heteromer formation between angiotensin II receptor type 1 subtype a (AT1aR) and secretin receptor (SCTR), which is involved in in vivo control of hyperosmolality-induced water drinking behavior. Because the secretin (SCT)/SCTR axis is crucial to the central actions of angiotensin II (ANGII) and both SCT and ANGII are capable of triggering vasopressin (Vp) release from hypothalamus, we investigated here the in vivo role of SCTR-AT1aR heteromer in regulating Vp release in hypothalamus using transmembrane peptides as tools. We showed that SCTR-AT1aR heteromer mediates stimulatory actions of both SCT and ANGII in hypothalamic Vp expression and release as well as neuronal activities via the immediate early gene cFos. The results from this study not only are consistent with our hypothesis that SCT and ANGII interact at the receptor level to mediate their water homeostatic activities but also provide evidence for in vivo functions of cross-class GPCR heteromers.-Mak, S. O. K., Zhang, L., Chow, B. K. C. In vivo actions of SCTR/AT1aR heteromer in controlling Vp expression and release via cFos/cAMP/CREB pathway in magnocellular neurons of PVN.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Angiotensin II/metabolism , Animals , Genes, fos/genetics , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Secretin/metabolism , Vasopressins/metabolism
6.
Eur J Pharmacol ; 834: 126-135, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30025814

ABSTRACT

Enteroendocrine derived hormones such as glucagon-like-peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), gastrin and xenin are known to exert complementary beneficial metabolic effects in diabetes. This study has assessed the biological activity and therapeutic utility of a novel GLP-1/gastrin/xenin hybrid peptide, namely exendin-4/gastrin/xenin-8-Gln hybrid, both alone and in combination with the stable GIP mimetic, (DAla2)GIP. Exendin-4/gastrin/xenin-8-Gln increased in vitro insulin secretion to a similar or superior extent, as the parent peptides. Insulinotropic effects were mainly linked to modulation of GLP-1 and neurotensin receptors. Exendin-4/gastrin/xenin-8-Gln also augmented the insulinotropic actions of (DAla2)GIP. Acute administration of exendin-4/gastrin/xenin-8-Gln in mice induced significant appetite suppressive, glucose lowering and insulin secretory effects, with a duration of biological action beyond 8 h. Twice daily administration of exendin-4, exendin-4/gastrin/xenin-8-Gln, either alone or in combination with (DAla2)GIP, reduced circulating glucose, increased plasma insulin as well as improving glucose tolerance, insulin sensitivity and metabolic response to GIP in high fat fed mice. Body weight, food intake, circulating glucagon and amylase activity were unaltered. All hybrid peptide treated high fat mice exhibited marked reductions in LDL-cholesterol and body fat mass. Energy expenditure and locomotor activity were increased in mice treated with exendin-4/gastrin/xenin-8-Gln in combination with (DAla2)GIP. Interestingly, exendin-4 and exendin-4/gastrin/xenin-8-Gln treatment, but not exendin-4/gastrin/xenin-8-Gln in combination with (DAla2)GIP, reduced pancreatic islet and beta-cell area when compared to high fat controls. These studies confirm that unimolecular multi-agonist peptide hormones exert beneficial metabolic effects in diabetes, highlighting their potential as novel treatment strategies.


Subject(s)
Exenatide/chemistry , Gastrins/chemistry , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Peptide Fragments/chemistry , Amylases/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Eating/drug effects , Fasting/blood , Glucagon/blood , Hypoglycemic Agents/therapeutic use , Insulin/blood , Insulin Resistance , Lipids/blood , Male , Mice , Pancrelipase/drug effects , Pancrelipase/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction/drug effects
7.
Diabetes ; 67(2): 309-320, 2018 02.
Article in English | MEDLINE | ID: mdl-29203510

ABSTRACT

Therapeutic engineering of glucagon-like peptide 1 (GLP-1) has enabled development of new medicines to treat type 2 diabetes. These injectable analogs achieve robust glycemic control by increasing concentrations of "GLP-1 equivalents" (∼50 pmol/L). Similar levels of endogenous GLP-1 occur after gastric bypass surgery, and mechanistic studies indicate glucose lowering by these procedures is driven by GLP-1. Therefore, because of the remarkable signaling and secretory capacity of the GLP-1 system, we sought to discover mechanisms that increase GLP-1 pharmacologically. To study active GLP-1, glucose-dependent insulinotropic polypeptide receptor (Gipr)-deficient mice receiving background dipeptidyl peptidase 4 (DPP4) inhibitor treatment were characterized as a model for evaluating oral agents that increase circulating GLP-1. A somatostatin receptor 5 antagonist, which blunts inhibition of GLP-1 release, and agonists for TGR5 and GPR40, which stimulate GLP-1 secretion, were investigated alone and in combination with the DPP4 inhibitor sitagliptin; these only modestly increased GLP-1 (∼5-30 pmol/L). However, combining molecules to simultaneously intervene at multiple regulatory nodes synergistically elevated active GLP-1 to unprecedented concentrations (∼300-400 pmol/L), drastically reducing glucose in Gipr null and Leprdb/db mice in a GLP-1 receptor-dependent manner. Our studies demonstrate that complementary pathways can be engaged to robustly increase GLP-1 without invasive surgical or injection regimens.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drugs, Investigational/therapeutic use , Glucagon-Like Peptide 1/analogs & derivatives , Models, Biological , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Drug Design , Drug Evaluation, Preclinical , Drug Resistance , Drug Synergism , Drug Therapy, Combination , Drugs, Investigational/administration & dosage , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/therapeutic use , Hyperglycemia/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Proof of Concept Study , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Sitagliptin Phosphate/therapeutic use
8.
Neuroreport ; 27(1): 23-32, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26555034

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonists have been shown to be neuroprotective in previous studies in animal models of Alzheimer's or Parkinson's disease. Recently, novel dual-GLP-1/GIP receptor agonists that activate both receptors (DA) were developed to treat diabetes. We tested the protective effects of a novel potent DA against middle cerebral artery occlusion injury in rats and compared it with a potent GLP-1 analog, Val(8)-GLP-1(glu-PAL). Animals were evaluated for neurologic deficit score, infarct volume, and immunohistochemical analyses of the brain at several time points after ischemia. The Val(8)-GLP-1(glu-PAL)-treated and DA-treated groups showed significantly reduced scores of neurological dysfunction, cerebral infarction size, and percentage of TUNEL-positive apoptotic neurons. Furthermore, the expression of the apoptosis marker Bax, the inflammation marker iNOS, and the survival marker Bcl-2 was significantly increased. The DA-treated group was better protected against neurodegeneration than the Val(8)-GLP-1(glu-PAL) group, and the scores of neurological dysfunction, cerebral infarction size, and expression of Bcl-2 were higher, whereas the percentage of TUNEL-positive neurons and the levels of Bax and iNOS were lower in the DA group. DA treatment reduced the infarct volume and improved the functional deficit. It also suppressed the inflammatory response and cell apoptosis after reperfusion. In conclusion, the novel GIP and GLP-1 dual-receptor agonist is more neuroprotective than a GLP-1 receptor agonist in key biomarkers of neuronal degeneration.


Subject(s)
Brain/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide-1 Receptor/agonists , Ischemic Attack, Transient/drug therapy , Lipopeptides/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Infarction, Middle Cerebral Artery , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Motor Activity/drug effects , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Random Allocation , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/metabolism , bcl-2-Associated X Protein/metabolism
9.
World J Gastroenterol ; 21(48): 13480-9, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26730159

ABSTRACT

AIM: To study the neural mechanism by which electroacupuncture (EA) at RN12 (Zhongwan) and BL21 (Weishu) regulates gastric motility. METHODS: One hundred and forty-four adult Sprague Dawley rats were studied in four separate experiments. Intragastric pressure was measured using custom-made rubber balloons, and extracellular neuron firing activity, which is sensitive to gastric distention in the dorsal vagal complex (DVC), was recorded by an electrophysiological technique. The expression levels of c-fos, motilin (MTL) and gastrin (GAS) in the paraventricular hypothalamic nucleus (PVN) were assayed by immunohistochemistry, and the expression levels of motilin receptor (MTL-R) and gastrin receptor (GAS-R) in both the PVN and the gastric antrum were assayed by western blotting. RESULTS: EA at RN12 + BL21 (gastric Shu and Mu points), BL21 (gastric Back-Shu point), RN12 (gastric Front-Mu point), resulted in increased neuron-activating frequency in the DVC (2.08 ± 0.050, 1.17 ± 0.023, 1.55 ± 0.079 vs 0.75 ± 0.046, P < 0.001) compared with a model group. The expression of c-fos (36.24 ± 1.67, 29.41 ± 2.55, 31.79 ± 3.00 vs 5.73 ± 2.18, P < 0.001), MTL (22.48 ± 2.66, 20.76 ± 2.41, 19.17 ± 1.71 vs 11.68 ± 2.52, P < 0.001), GAS (24.99 ± 2.95, 21.69 ± 3.24, 23.03 ± 3.09 vs 12.53 ± 2.15, P < 0.001), MTL-R (1.39 ± 0.05, 1.22 ± 0.05, 1.17 ± 0.12 vs 0.84 ± 0.06, P < 0.001), and GAS-R (1.07 ± 0.07, 0.91 ± 0.06, 0.78 ± 0.05 vs 0.45 ± 0.04, P < 0.001) increased in the PVN after EA compared with the model group. The expression of MTL-R (1.46 ± 0.14, 1.26 ± 0.11, 0.99 ± 0.07 vs 0.65 ± 0.03, P < 0.001), and GAS-R (1.63 ± 0.11, 1.26 ± 0.16, 1.13 ± 0.02 vs 0.80 ± 0.11, P < 0.001) increased in the gastric antrum after EA compared with the model group. Damaging the PVN resulted in reduced intragastric pressure (13.67 ± 3.72 vs 4.27 ± 1.48, P < 0.001). These data demonstrate that the signals induced by EA stimulation of acupoints RN12 and BL21 are detectable in the DVC and the PVN, and increase the levels of gastrointestinal hormones and their receptors in the PVN and gastric antrum to regulate gastric motility. CONCLUSION: EA at RN12 and BL21 regulates gastric motility, which may be achieved through the PVN-DVC-vagus-gastric neural pathway.


Subject(s)
Acupuncture Points , Electroacupuncture/methods , Gastric Emptying , Paraventricular Hypothalamic Nucleus/physiology , Solitary Nucleus/physiology , Stomach/innervation , Vagus Nerve/physiology , Action Potentials , Animals , Gastrins/metabolism , Male , Mechanotransduction, Cellular , Motilin/metabolism , Neural Pathways/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Pressure , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/metabolism
10.
Int J Obes (Lond) ; 39(3): 447-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25219528

ABSTRACT

BACKGROUND: High-protein diets promote weight loss and subsequent weight maintenance, but are difficult to adhere to. The mechanisms by which protein exerts these effects remain unclear. However, the amino acids produced by protein digestion may have a role in driving protein-induced satiety. METHODS: We tested the effects of a range of amino acids on food intake in rodents and identified l-cysteine as the most anorexigenic. Using rodents we further studied the effect of l-cysteine on food intake, behaviour and energy expenditure. We proceeded to investigate its effect on neuronal activation in the hypothalamus and brainstem before investigating its effect on gastric emptying and gut hormone release. The effect of l-cysteine on appetite scores and gut hormone release was then investigated in humans. RESULTS: l-Cysteine dose-dependently decreased food intake in both rats and mice following oral gavage and intraperitoneal administration. This effect did not appear to be secondary to behavioural or aversive side effects. l-Cysteine increased neuronal activation in the area postrema and delayed gastric emptying. It suppressed plasma acyl ghrelin levels and did not reduce food intake in transgenic ghrelin-overexpressing mice. Repeated l-cysteine administration decreased food intake in rats and obese mice. l-Cysteine reduced hunger and plasma acyl ghrelin levels in humans. CONCLUSIONS: Further work is required to determine the chronic effect of l-cysteine in rodents and humans on appetite and body weight, and whether l-cysteine contributes towards protein-induced satiety.


Subject(s)
Appetite Depressants/pharmacology , Appetite/drug effects , Cysteine/pharmacology , Eating/drug effects , Ghrelin/antagonists & inhibitors , Adult , Animals , Appetite Depressants/administration & dosage , Cysteine/administration & dosage , Dose-Response Relationship, Drug , Female , Gastrointestinal Hormones/metabolism , Ghrelin/metabolism , Humans , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger , Rats , Rats, Wistar , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Satiation
11.
Toxicol Appl Pharmacol ; 279(3): 419-427, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24997324

ABSTRACT

The incidence of micronucleated-cells is considered to be a marker of a genotoxic event and can be caused by direct- or indirect-DNA reactive mechanisms. In particular, small increases in the incidence of micronuclei, which are not associated with toxicity in the target tissue or any structurally altering properties of the compound, trigger the suspicion that an indirect mechanism could be at play. In a bone marrow micronucleus test of a synthetic peptide (a dual agonist of the GLP-1 and GIP receptors) that had been integrated into a regulatory 13-week repeat-dose toxicity study in the rat, small increases in the incidence of micronuclei had been observed, together with pronounced reductions in food intake and body weight gain. Because it is well established that folate plays a crucial role in maintaining genomic integrity and pronounced reductions in food intake and body weight gain were observed, folate levels were determined from plasma samples initially collected for toxicokinetic analytics. A dose-dependent decrease in plasma folate levels was evident after 4 weeks of treatment at the mid and high dose levels, persisted until the end of the treatment duration of 13-weeks and returned to baseline levels during the recovery period of 4 weeks. Based on these properties, and the fact that the compound tested (peptide) per se is not expected to reach the nucleus and cause DNA damage, the rationale is supported that the elevated incidence of micronucleated polychromatic erythrocytes is directly linked to the exaggerated pharmacology of the compound resulting in a decreased folate level.


Subject(s)
Folic Acid Deficiency/chemically induced , Mutagenicity Tests/methods , Mutagens , Peptides/toxicity , Animals , Body Temperature/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/ultrastructure , Cell Line, Tumor , Chromosome Aberrations/drug effects , Eating/drug effects , Erythropoiesis/drug effects , Folic Acid Deficiency/genetics , Humans , Lymphocytes/drug effects , Lymphocytes/ultrastructure , Malnutrition/chemically induced , Mice , Mice, Knockout , Micronucleus Tests , Rats , Receptors, Gastrointestinal Hormone/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
12.
Endocrinology ; 154(9): 3089-98, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23798598

ABSTRACT

We have previously demonstrated that ileal administration of the dietary protein hydrolysate prepared from corn zein (ZeinH) stimulated glucagon-like peptide-1 (GLP-1) secretion and attenuated hyperglycemia in rats. In this study, to examine whether oral administration of ZeinH improves glucose tolerance by stimulating GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) secretion, glucose tolerance tests were performed in normal Sprague-Dawley male rats and diabetic Goto-Kakizaki (GK) male rats. The test solution was gavaged before ip glucose injection in normal rats or gavaged together with glucose in GK rats. Blood samples were collected from the tail vein or by using the jugular catheter to measure glucose, insulin, GLP-1, and GIP levels. In the ip glucose tolerance test, oral administration of ZeinH (2 g/kg) significantly suppressed the glycemic response accompanied by an immediate increase in plasma GLP-1 and GIP levels in normal rats. In contrast, oral administration of another dietary peptide, meat hydrolysate, did not elicit a similar effect. The glucose-lowering effect of ZeinH was attenuated by a GLP-1 receptor antagonist or by a GIP receptor antagonist. Furthermore, oral ZeinH induced GLP-1 secretion and reduced glycemic response in GK rats under the oral glucose tolerance test. These results indicate that the oral administration of the dietary peptide ZeinH improves glucose tolerance in normal and diabetic rats by its incretin-releasing activity, namely, the incretinotropic effect.


Subject(s)
Diabetes Mellitus, Type 2/diet therapy , Enterocytes/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/therapeutic use , Protein Hydrolysates/therapeutic use , Zein/therapeutic use , Animals , Cell Line , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Dietary Supplements , Digestion , Enterocytes/drug effects , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/antagonists & inhibitors , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/antagonists & inhibitors , Hypoglycemic Agents/metabolism , Male , Mice , Protein Hydrolysates/administration & dosage , Protein Hydrolysates/antagonists & inhibitors , Protein Hydrolysates/metabolism , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/metabolism , Up-Regulation/drug effects , Zein/administration & dosage , Zein/antagonists & inhibitors , Zein/metabolism
13.
J Ethnopharmacol ; 148(2): 459-65, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23639361

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Poncirus fructus (PF), also known as a dried immature fruit of Poncirus trifoliata (L.) Raf. (Rutaceae), has long been traditionally used for the various gastrointestinal disorders in Eastern Asia. AIM OF STUDY: The aqueous extract of PF (PF-W) has the strong prokinetic effect, yet the underlying mechanism is still elusive. The present study investigated whether PF-W has any effect on motilin receptor or ghrelin receptor, since these receptors enhance intestinal motility when activated. MATERIALS AND METHODS: The effect of PF-W and its components on motilin or ghrelin receptor was determined by calcium imaging and whole-cell patch clamp methods. RESULTS: PF-W activates the ghrelin receptor, but not the motilin receptor, resulting in a transient increase of intracellular calcium levels. Furthermore, among various constituents of PF, only naringin and naringenin evoked the intracellular calcium augmentation via the ghrelin receptor. Moreover, cortistatin-8 - a ghrelin receptor inhibitor - specifically blocked naringin- and naringenin-induced calcium increases. In addition, naringin and naringenin induced inward currents in ghrelin receptor-expressing cells under whole-cell patch clamp configuration. CONCLUSION: PF-W activates the ghrelin receptor, and naringin and naringenin are key constituents responsible for the activation of ghrelin receptor. Therefore, the present study suggests that the ghrelin receptor is a molecular entity responsible for the strong prokinetic activity of PF-W.


Subject(s)
Flavanones/pharmacology , Gastrointestinal Agents/chemistry , Gastrointestinal Agents/pharmacology , Poncirus/chemistry , Receptors, Ghrelin/metabolism , Rutaceae/chemistry , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Fruit/chemistry , Gastrointestinal Motility/drug effects , HEK293 Cells , Humans , Neuropeptides/pharmacology , Plant Extracts/pharmacology , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Water/chemistry
14.
Physiol Res ; 60(6): 941-50, 2011.
Article in English | MEDLINE | ID: mdl-21995902

ABSTRACT

GIP (glucose dependent insulinotrophic polypeptide), originally identified as an incretin peptide synthesized in the gut, has recently been identified, along with its receptors (GIPR), in the brain. Our objective was to investigate the role of GIP in hypothalamic gene expression of biomarkers linked to regulating energy balance and feeding behavior related neurocircuitry. Rats with lateral cerebroventricular cannulas were administered 10 µg GIP or 10 microl artificial cerebrospinal fluid (aCSF) daily for 4 days, after which whole hypothalami were collected. Real time Taqman™ RT-PCR was used to quantitatively compare the mRNA expression levels of a set of genes in the hypothalamus. Administration of GIP resulted in up-regulation of hypothalamic mRNA levels of AVP (46.9±4.5 %), CART (25.9±2.7 %), CREB1 (38.5±4.5 %), GABRD (67.1±11 %), JAK2 (22.1±3.6 %), MAPK1 (33.8±7.8 %), NPY (25.3±5.3 %), OXT (49.1±5.1 %), STAT3 (21.6±3.8 %), and TH (33.9±8.5 %). In a second experiment the same set of genes was evaluated in GIPR(-/-) and GIPR(+/?) mice to determine the effect of lack of GIP stimulation on gene expression. In GIPR(-/-) mice expressions of the following genes were down-regulated: AVP (27.1±7.5 %), CART (28.3±3.7 %), OXT (25.2±5.8 %), PTGES (23.9±4.5 %), and STAT3 (8.8±2.3 %). These results suggest that AVP, CART, OXT and STAT3 may be involved in energy balance-related hypothalamic circuits affected by GIP.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Gene Expression , Hypothalamus/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Animals , Biomarkers/metabolism , Energy Metabolism/genetics , Feeding Behavior , Gastric Inhibitory Polypeptide/pharmacology , Hypothalamus/drug effects , Male , Mice , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/genetics
15.
FASEB J ; 24(12): 5024-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20739612

ABSTRACT

Fluid balance is critical to life and hence is tightly controlled in the body. Angiotensin II (ANGII), one of the most important components of this regulatory system, is recognized as a dipsogenic hormone that stimulates vasopressin (VP) expression and release. However, detailed mechanisms regarding how ANGII brings about these changes are not fully understood. In the present study, we show initially that the osmoregulatory functions of secretin (SCT) in the brain are similar to those of ANGII in mice and, more important, we discovered the role of SCT as the link between ANGII and its downstream effects. This was substantiated by the use of two knockout mice, SCTR(-/-) and SCT(-/-), in which we show the absence of an intact SCT/secretin receptor (SCTR) axis resulted in an abolishment or much reduced ANGII osmoregulatory functions. By immunohistochemical staining and in situ hybridization, the proteins and transcripts of SCT and its receptor are found in the paraventricular nucleus (PVN) and lamina terminalis. We propose that SCT produced in the circumventricular organs is transported and released in the PVN to stimulate vasopressin expression and release. In summary, our findings identify SCT and SCTR as novel elements of the ANGII osmoregulatory pathway in maintaining fluid balance in the body.


Subject(s)
Angiotensin II/pharmacology , Secretin/metabolism , Secretin/pharmacology , Animals , Drinking/drug effects , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Secretin/genetics , Vasopressins/metabolism , Water-Electrolyte Balance/drug effects
16.
Mol Pharmacol ; 76(2): 264-74, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19429716

ABSTRACT

Oligomerization of G protein-coupled receptors has been described, but its structural basis and functional importance have been inconsistent. Here, we demonstrate that the agonist occupied wild-type secretin receptor is predominantly in a guanine nucleotide-sensitive high-affinity state and exhibits negative cooperativity, whereas the monomeric receptor is primarily in a guanine nucleotide-insensitive lower affinity state. We previously demonstrated constitutive homodimerization of this receptor through the lipid-exposed face of transmembrane (TM) IV. We now use cysteine-scanning mutagenesis of 14 TM IV residues, bioluminescence resonance energy transfer (BRET), and functional analysis to map spatial approximations and functional importance of specific residues in this complex. All, except for three helix-facing mutants, trafficked to the cell surface, where secretin was shown to bind and elicit cAMP production. Cells expressing complementary-tagged receptors were treated with cuprous phenanthroline to establish disulfide bonds between spatially approximated cysteines. BRET was measured as an indication of receptor oligomerization and was repeated after competitive disruption of oligomers with TM IV peptide to distinguish covalent from noncovalent associations. Although all constructs generated a significant BRET signal, this was disrupted by peptide in all except for single-site mutants replacing five residues with cysteine. Of these, covalent stabilization of receptor homodimers through positions of Gly(243), Ile(247), and Ala(250) resulted in a GTP-sensitive high-affinity state of the receptor, whereas the same procedure with Ala(246) and Phe(240) mutants resulted in a GTP-insensitive lower affinity state. We propose the existence of a functionally important, structurally specific high-affinity dimeric state of the secretin receptor, which may be typical of family B G protein-coupled receptors.


Subject(s)
Protein Multimerization , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Secretin/metabolism , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , COS Cells , Cell Membrane/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Cysteine/metabolism , Dimerization , Fluorescence Resonance Energy Transfer , Inhibitory Concentration 50 , Membrane Lipids/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Protein Binding/genetics , Protein Structure, Secondary/genetics , Radioligand Assay , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics , Transfection
17.
Rev. clín. esp. (Ed. impr.) ; 207(10): 501-504, nov. 2007. ilus, tab
Article in Es | IBECS | ID: ibc-057842

ABSTRACT

Las incretinas son sustancias que se producen en el intestino y se liberan en respuesta a la ingestión oral de nutrientes, sobre todo hidratos de carbono, siendo poderosas secretagogas que aumentan la liberación de insulina. Las 2 hormonas incretinas más importantes son el polipéptido inhibidor gástrico (GIP) y el péptido-1 similar al glucagón (GLP-1). Además de estimular la secreción de insulina, el GLP-1 suprime la liberación de glucagón, enlentece el vaciamiento gástrico, mejora la sensibilidad a la insulina y reduce el consumo de alimentos. Otros nutrientes pueden estimular también la secreción de insulina, como son el ácido oleico y la proteína de suero. Hoy día se está desarrollando un nuevo arsenal terapéutico centrado en el papel de las incretinas para un mejor abordaje de la diabetes mellitus tipo 2 (DM 2) (AU)


Incretins are hormones produced in the intestine that are released in response to oral intake of nutrients, above all carbohydrates. They are powerful secretors that increase insulin release. The two most important incretin hormones are GIP (glucose-dependent insulinotropic peptide; also known as gastric inhibitory peptide) and GLP-1 (glucagon-like peptide-1). GLP-1 not only stimulates insulin secretion but also reduces glucagon release, slows gastric emptying, improves insulin sensitivity and increases satiety. Other nutrients may also stimulate insulin secretion: oleic acid and serum protein. Currently a new therapeutic armamentarium focused on the role of incretins is being developed to improve the treatment of type 2 diabetes mellitus (DM 2) (AU)


Subject(s)
Humans , Glucagon/analogs & derivatives , Glucagon/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Receptors, Gastrointestinal Hormone/metabolism , Protein Precursors , Insulin
18.
J Clin Endocrinol Metab ; 92(11): 4394-402, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17711922

ABSTRACT

CONTEXT/OBJECTIVES: The diagnosis of Zollinger-Ellison syndrome requires secretin testing in 60% of patients. Even with secretin, the diagnosis may be difficult because variable responses occur, and 6-30% have negative testing. The basis for variability or negative responses is unclear. It is unknown whether the tumor density of secretin receptors or the presence of a secretin-receptor-variant, which can act as a dominant negative, is important. The aim of this study was to investigate these possibilities. PATIENTS/METHODS: Secretin-receptor and variant mRNA expression was determined in gastrinomas using real-time PCR from 54 Zollinger-Ellison syndrome patients. Results were correlated with Western blotting, secretin-receptor immunohistochemistry, with gastrin-provocative test results and tumoral/clinical/laboratory features. RESULTS: Secretin-receptor mRNA was detectible in all gastrinomas but varied 132-fold with a mean of 0.89 +/- 0.12 molecules per beta-actin. Secretin-receptor PCR results correlated closely with Western blotting (r = 0.95; P < 0.0001) and receptor immunohistochemistry (P = 0.0015; r = 0.71). The variant was detected in all gastrinomas, but levels varied 102-fold and were 72-fold lower than the total. Secretin-receptor levels correlated with variant levels, Deltasecretin, but not Deltacalcium and with tumor location, but not growth, extent, or clinical responses. Variant levels did not correlate with the Deltasecretin. Detailed analysis provides no evidence that variant expression modified the secretin-receptor response or accounted for negative tests. CONCLUSIONS: Secretin-receptor and secretin-receptor-variant expressions occur in all gastrinomas. Because the expression of the total, but not variant, correlated with the secretin results and no evidence for dominant negative activity of the variant was found, our results suggest that the total secretin-receptor density is an important determinant of the secretin test response.


Subject(s)
Calcium/metabolism , Gastrinoma/genetics , Gene Expression Regulation/physiology , Pancreatic Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Zollinger-Ellison Syndrome/genetics , Zollinger-Ellison Syndrome/metabolism , Blotting, Western , DNA, Complementary/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Ann N Y Acad Sci ; 1070: 196-200, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888165

ABSTRACT

Previous studies demonstrated that secretin could be released from the cerebellum, where it exerts a facilitatory action on the GABAergic inputs into the Purkinje neurons. In the present article, we provide evidence of the endogenous release of secretin in the hypothalamus and the mechanisms underlying this release. Incubation of the hypothalamic explants with KCl induces the release of secretin to 4.35 +/- 0.45-fold of the basal level. This K+-induced release was tetrodotoxin and cadmium sensitive, suggesting the involvement of voltage-gated sodium and calcium channels. The use of specific blockers further revealed the involvement of L-, N-, and P-type high voltage-activated (HVA) calcium channels. Results present in the current article provide further and more solid evidence of the role of secretin as a neuropeptide in the mammalian central nervous system.


Subject(s)
Hypothalamus/metabolism , Secretin/metabolism , Animals , Male , Potassium Chloride/pharmacology , Rats , Rats, Inbred WF , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism
20.
J Biol Chem ; 279(51): 53806-17, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15383539

ABSTRACT

Three members of the ghrelin receptor family were characterized in parallel: the ghrelin receptor, the neurotensin receptor 2 and the orphan receptor GPR39. In transiently transfected COS-7 and human embryonic kidney 293 cells, all three receptors displayed a high degree of ligand-independent signaling activity. The structurally homologous motilin receptor served as a constitutively silent control; upon agonist stimulation, however, it signaled with a similar efficacy to the three related receptors. The constitutive activity of the ghrelin receptor and of neurotensin receptor 2 through the G(q), phospholipase C pathway was approximately 50% of their maximal capacity as determined through inositol phosphate accumulation. These two receptors also showed very high constitutive activity in activation of cAMP response element-driven transcription. GPR39 displayed a clear but lower degree of constitutive activity through the inositol phosphate and cAMP response element pathways. In contrast, GPR39 signaled with the highest constitutive activity in respect of activation of serum response element-dependent transcription, in part, possibly, through G(12/13) and Rho kinase. Antibody feeding experiments demonstrated that the epitope-tagged ghrelin receptor was constitutively internalized but could be trapped at the cell surface by an inverse agonist, whereas GPR39 remained at the cell surface. Mutational analysis showed that the constitutive activity of both the ghrelin receptor and GPR39 could systematically be tuned up and down depending on the size and hydrophobicity of the side chain in position VI:16 in the context of an aromatic residue at VII:09 and a large hydrophobic residue at VII:06. It is concluded that the three ghrelin-like receptors display an unusually high degree of constitutive activity, the structural basis for which is determined by an aromatic cluster on the inner face of the extracellular ends of TMs VI and VII.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/physiology , Receptors, Neurotensin/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cyclic AMP/metabolism , DNA Mutational Analysis , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Humans , Inositol Phosphates/metabolism , Ligands , MAP Kinase Signaling System , Microscopy , Models, Molecular , Molecular Sequence Data , Phosphatidylinositols/chemistry , Phylogeny , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Ghrelin , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Signal Transduction , Transcription, Genetic , Transfection , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL