Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Mol Metab ; 55: 101392, 2022 01.
Article in English | MEDLINE | ID: mdl-34781035

ABSTRACT

OBJECTIVE: Obesity-linked type 2 diabetes (T2D) is a worldwide health concern and many novel approaches are being considered for its treatment and subsequent prevention of serious comorbidities. Co-administration of glucagon like peptide 1 (GLP-1) and peptide YY3-36 (PYY3-36) renders a synergistic decrease in energy intake in obese men. However, mechanistic details of the synergy between these peptide agonists and their effects on metabolic homeostasis remain relatively scarce. METHODS: In this study, we utilized long-acting analogues of GLP-1 and PYY3-36 (via Fc-peptide conjugation) to better characterize the synergistic pharmacological benefits of their co-administration on body weight and glycaemic regulation in obese and diabetic mouse models. Hyperinsulinemic-euglycemic clamps were used to measure weight-independent effects of Fc-PYY3-36 + Fc-GLP-1 on insulin action. Fluorescent light sheet microscopy analysis of whole brain was performed to assess activation of brain regions. RESULTS: Co-administration of long-acting Fc-IgG/peptide conjugates of Fc-GLP-1 and Fc-PYY3-36 (specific for PYY receptor-2 (Y2R)) resulted in profound weight loss, restored glucose homeostasis, and recovered endogenous ß-cell function in two mouse models of obese T2D. Hyperinsulinemic-euglycemic clamps in C57BLKS/J db/db and diet-induced obese Y2R-deficient (Y2RKO) mice indicated Y2R is required for a weight-independent improvement in peripheral insulin sensitivity and enhanced hepatic glycogenesis. Brain cFos staining demonstrated distinct temporal activation of regions of the hypothalamus and hindbrain following Fc-PYY3-36 + Fc-GLP-1R agonist administration. CONCLUSIONS: These results reveal a therapeutic approach for obesity/T2D that improved insulin sensitivity and restored endogenous ß-cell function. These data also highlight the potential association between the gut-brain axis in control of metabolic homeostasis.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Peptide YY/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Eating/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Gastric Bypass , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/physiopathology , Peptide YY/physiology , Weight Loss
2.
Mol Nutr Food Res ; 65(17): e2100136, 2021 09.
Article in English | MEDLINE | ID: mdl-34272917

ABSTRACT

SCOPE: Obesity is a common disease worldwide and there is an urgent need for strategies to preventing obesity. METHODS AND RESULTS: The anti-obesity effect and mechanism of Ligilactobacillus salivarius LCK11 (LCK11) is studied using a C57BL/6J male mouse model in which obesity is induced by a high-fat diet (HFD). Results show that LCK11 can prevent HFD-induced obesity, reflected as inhibited body weight gain, abdominal and liver fat accumulation and dyslipidemia. Analysis of its mechanism shows that on the one hand, LCK11 can inhibit food intake through significantly improving the transcriptional and translational levels of peptide YY (PYY) in the rectum, in addition to the eventual serum PYY level; this is attributed to the activation of the toll-like receptor 2/nuclear factor-κB signaling pathway in enteroendocrine L cells by the peptidoglycan of LCK11. On the other hand, LCK11 supplementation effectively reduces the Firmicutes/Bacteroidetes ratio and shifts the overall structure of the HFD-disrupted gut microbiota toward that of mice fed on a low-fat diet; this also contributes to preventing obesity. CONCLUSION: LCK11 shows the potential to be used as a novel probiotic for preventing obesity by both promoting PYY secretion to inhibit food intake and regulating gut microbiota.


Subject(s)
Gastrointestinal Microbiome/physiology , Lactobacillaceae , Obesity/prevention & control , Peptide YY/metabolism , Adipose Tissue/physiology , Animals , Anti-Obesity Agents/pharmacology , Diet, High-Fat/adverse effects , Dyslipidemias/microbiology , Dyslipidemias/therapy , Eating , Enteroendocrine Cells/metabolism , Intestines/microbiology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/microbiology , Probiotics/pharmacology , Weight Gain
3.
Mol Nutr Food Res ; 64(16): e2000303, 2020 08.
Article in English | MEDLINE | ID: mdl-32613679

ABSTRACT

SCOPE: A grape-seed proanthocyanidin extract (GSPE) interacts at the intestinal level, enhancing glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) release, which modulate appetite and glucose homeostasis. Thus, enhancing L-cell numbers could be a strategy to promote hormone production, providing a potential strategy for obesity and type-2 diabetes mellitus (T2DM) treatment. METHODS AND RESULTS: Mice ileum organoids are used to evaluate the long-term effects of GSPE and two of its main components, epicatechin (EC) and gallic acid (GA), on intestinal differentiation. Hormone levels are determined using RIA and ELISA kits, and gene expression of transcription factors involved in intestinal cell differentiation, as well as markers of different cell types, are assessed by real-time qPCR. GSPE upregulates enterohormone gene expression and content, as well as the pan-endocrine marker chromogranin A. GSPE also modulates the temporal gene expression profile of early and late transcription factors involved in L-cell differentiation. Furthermore, GSPE upregulates goblet cell (Muc2) and enterocyte (sucraseisomaltase) markers, while downregulating stem cell markers (Lgr5+). Although EC and GA modified enterohormone release, they do not reproduce GSPE effects on transcription factor's profile. CONCLUSIONS: This study shows the potential role of GSPE in promoting enteroendocrine differentiation, effect that is not mediated by EC or GA.


Subject(s)
Gastrointestinal Hormones/metabolism , Grape Seed Extract/pharmacology , Ileum/cytology , Ileum/drug effects , Ileum/metabolism , Proanthocyanidins/pharmacology , Animals , Catechin/pharmacology , Cell Differentiation/drug effects , Enterocytes/cytology , Enterocytes/drug effects , Gallic Acid/pharmacology , Glucagon-Like Peptide 1/metabolism , Grape Seed Extract/chemistry , Mice, Inbred C57BL , Mucin-2/metabolism , Organoids , Peptide YY/metabolism , Proanthocyanidins/chemistry , Receptors, G-Protein-Coupled/metabolism
4.
Biomolecules ; 10(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32492958

ABSTRACT

Some beneficial effects of grape seed proanthocyanidin extract (GSPE) can be explained by the modulation of enterohormone secretion. As GSPE comprises a combination of different molecules, the pure compounds that cause these effects need to be elucidated. The enterohormones and chemoreceptors present in the gastrointestinal tract differ between species, so if humans are to gain beneficial effects, species closer to humans-and humans themselves-must be used. We demonstrate that 100 mg/L of GSPE stimulates peptide YY (PYY) release, but not glucagon-like peptide 1 (GLP-1) release in the human colon. We used a pig ex vivo system that differentiates between apical and basolateral intestinal sides to analyse how apical stimulation with GSPE and its pure compounds affects the gastrointestinal tract. In pigs, apical GSPE treatment stimulates the basolateral release of PYY in the duodenum and colon and that of GLP-1 in the ascending, but not the descending colon. In the duodenum, luminal stimulation with procyanidin dimer B2 increased PYY secretion, but not CCK secretion, while catechin monomers (catechin/epicatechin) significantly increased CCK release, but not PYY release. The differential effects of GSPE and its pure compounds on enterohormone release at the same intestinal segment suggest that they act through chemosensors located apically and unevenly distributed along the gastrointestinal tract.


Subject(s)
Cholecystokinin/metabolism , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Animals , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Peptide YY/metabolism , Plant Extracts/chemistry , Proanthocyanidins/chemistry , Seeds/chemistry , Swine , Vitis/chemistry
5.
Curr Opin Endocrinol Diabetes Obes ; 27(1): 11-15, 2020 02.
Article in English | MEDLINE | ID: mdl-31815784

ABSTRACT

PURPOSE OF REVIEW: In this review, authors have selected from literature the most recent and suggestive studies on therapy of nonalcoholic fatty liver disease (NAFLD). The selected interventions regulate the action of gastrointestinal peptides, such as gastric inhibitory polypeptide (GIP), nesfatin, peptide YY, cholecystokinin, and glucagon-like peptide 1 (GLP-1). These hormones have been found frequently modified in obesity and/or type 2 diabetes mellitus, morbidities mostly associated with NAFLD. This disease has a very high prevalence worldwide and could evolve in a more severe form, that is, nonalcoholic steatohepatitis, characterized by inflammation and fibrosis. The findings shown by this article describe the metabolic effects of new drugs, mainly but not only, as well of some old substances. RECENT FINDINGS: Recent approaches, in animal models or in humans, use synthetic GLP-1 receptor agonists, a centrally administered antibody neutralizing GIP receptor, curcumin, compound being active on nesfatin, resveratrol (antiinflammatory agent), and Ginseg, both of them acting on nesfatin, a cholecystokinin receptor analogue, and finally coffee functioning on YY peptide. SUMMARY: The implications of the presented findings, if they are confirmed in larger clinical trials, likely open the door to future application in clinical practice. In fact, nowadays, patients have only diet and article (incl abstract and keywords) exercise as well accepted recommendations. Thus, there are unmet needs to find substances that could really improve the progression of nonalcoholic steatohepatitis toward liver cirrhosis and hepatocellular carcinoma.


Subject(s)
Gastrointestinal Tract/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Peptide Hormones/metabolism , Phytochemicals/therapeutic use , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Drugs, Investigational/therapeutic use , Gastric Inhibitory Polypeptide/metabolism , Gastrointestinal Tract/drug effects , Glucagon-Like Peptide 1/metabolism , Humans , Liver/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/etiology , Obesity/complications , Obesity/epidemiology , Obesity/metabolism , Obesity/therapy , Peptide YY/metabolism , Phytotherapy/methods , Phytotherapy/trends
6.
Animal ; 12(12): 2499-2504, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29508681

ABSTRACT

Non-starch polysaccharides (NSP) present in wheat and barley can act as anti-nutrients leading to an increase in digesta viscosity and a reduction in nutrient digestibility. Xylanase, an NSP-degrading enzyme, has been shown to increase nutrient digestibility in pigs. The objectives of this study were: (1) to identify the optimum inclusion level of xylanase in grower pig diets by measuring the effect of increasing enzyme levels on growth performance, the concentration of volatile fatty acids (VFA) and peptide YY concentration in portal and peripheral blood of grower pigs and (2) to increase our understanding of the interrelationships between xylanase inclusion, VFA production and peptide YY secretion. A total of 512 grower pigs ((Large White×Landrace)×MAXGRO) were allocated to pens creating 32 replicates of four pigs per pen per treatment. Pigs were allocated to trial weighing 14.2±0.31 kg and remained on trial until ~41.5±3.31 kg. The experiment was a dose response design with four inclusion levels (0, 8000, 16 000 or 32 000 BXU/kg) of xylanase (Econase XT). Diets were cereal-based wheat, barley mix formulated to meet or exceed the nutrient requirements of grower pigs. Body weight and feed intake were recorded to calculate growth performance. Pen faecal samples were collected to estimate DM, organic matter (OM) and crude fibre (CF) apparent total-tract digestibility. At the end of the trial 16 pigs per treatment were euthanised by schedule 1 procedures. Peripheral and portal blood samples were collected for peptide YY and VFA analysis. The addition of xylanase to the diet had no effect on growth performance, DM, OM or CF total-tract digestibility; however, xylanase tended to have a quadratic effect on ileum pH with higher pH values recorded for pigs fed a diet supplemented with 8000 and 16 000 BXU/kg xylanase (P<0.1). Xylanase had no effect on peptide YY levels or VFA concentration. Total VFA concentration was higher in portal compared with peripheral blood (P<0.05). In conclusion, the addition of xylanase had no effect on grower pig performance, nutrient digestibility, VFA concentration or peptide YY concentration when fed up to 32 000 BXU/kg over a 35-day period. Pig performance was good for all treatments throughout the trial suggesting that diet quality was sufficient thus there were no beneficial effects of adding xylanase.


Subject(s)
Animal Feed/analysis , Dietary Supplements , Endo-1,4-beta Xylanases/administration & dosage , Fatty Acids, Volatile/blood , Peptide YY/blood , Swine/physiology , Animals , Body Weight/drug effects , Diet/veterinary , Digestion/drug effects , Edible Grain , Fatty Acids, Volatile/metabolism , Feces/chemistry , Female , Gastrointestinal Tract/metabolism , Hordeum , Male , Peptide YY/metabolism , Random Allocation , Swine/growth & development , Triticum
7.
Mol Neurobiol ; 55(9): 7555-7566, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29429048

ABSTRACT

Apathy is observed across several neurological and psychiatric conditions; however, its pathogenesis remains unclear. We clarified the involvement of brain-gut signaling in the disruption of goal-directed behavior. Male C57BL/6J mice were exposed to water immersion (WI) stress for 3 days. Food intake and nesting behavior were measured as indexes of motivation. Repeated WI caused decrease in food intake and nesting behavior. Plasma levels of peptide YY (PYY), IL-6, and ratio of dopamine metabolites in the striatum were significantly elevated after WI. PYY and IL-6 administration significantly decreased nesting behavior. The reductions in feeding and nesting behavior were blocked by PYY receptor (Y2R) antagonist or dopamine agonist. The ameliorative effect of the Y2R antagonist was diminished by the dopamine D2 receptor (D2R) antagonist. The reduction in goal-directed behavior is associated with dysfunction of D2R signaling via increased peripheral PYY, suggesting that PYY antagonism is a novel candidate for decline of motivation in several depressive diseases.


Subject(s)
Apathy , Behavior, Animal , Immersion , Peptide YY/metabolism , Receptors, Dopamine D2/metabolism , Water , Animals , Apathy/drug effects , Body Weight , Corticosterone/blood , Dopamine/metabolism , Eating , Gene Expression Regulation , Humans , Hypothalamus/metabolism , Interleukin-6/administration & dosage , Interleukin-6/pharmacology , Male , Mice, Inbred C57BL , Models, Biological , Nesting Behavior , Organ Size , Peptide YY/administration & dosage , Peptide YY/pharmacology , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism
8.
Peptides ; 100: 269-274, 2018 02.
Article in English | MEDLINE | ID: mdl-29412828

ABSTRACT

The vast majority of research to date on the gut hormone Peptide YY (PYY) has focused on appetite suppression and body weight regulation effects. These biological actions are believed to occur through interaction of PYY with hypothalamic Y2 receptors. However, more recent studies have added additional knowledge to understanding of the physiological, and potential therapeutic, roles of PYY beyond obesity alone. Thus, PYY has now been shown to impart improvements in pancreatic beta-cell survival and function, with obvious benefits for diabetes. This effect has been linked mainly to binding and activation of Y1 receptors by PYY, but more evidence is still required in this regard. Given the potential therapeutic promise of PYY-derived compounds, and complexity of receptor interactions, it is important to fully understand the complete biological action profile of PYY. Therefore, the current review aims to compile, evaluate and summarise current knowledge on PYY, with particular emphasis on obesity and diabetes treatment, and the importance of specific Y receptor interactions for this.


Subject(s)
Diabetes Mellitus/drug therapy , Obesity/drug therapy , Peptide YY/therapeutic use , Receptors, Neuropeptide Y/genetics , Appetite Regulation/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Humans , Hypothalamus/metabolism , Obesity/metabolism , Obesity/pathology , Peptide Fragments/therapeutic use , Peptide YY/metabolism , Receptors, Neuropeptide Y/metabolism
9.
Adv Neurobiol ; 19: 213-236, 2017.
Article in English | MEDLINE | ID: mdl-28933067

ABSTRACT

In moderately or morbidly obese patients, bariatric surgery has been proven to be an effective therapeutic approach to control body weight and comorbidities. Surgery-mediated modulation of brain function via modified postoperative secretion of gut peptides and vagal nerve stimulation was identified as an underlying mechanism in weight loss and improvement of weight-related diseases. Increased basal and postprandial plasma levels of gastrointestinal hormones like glucagon-like peptide 1 and peptide YY that act on specific areas of the hypothalamus to reduce food intake, either directly or mediated by the vagus nerve, are observed after surgery while suppression of meal-induced ghrelin release is increased. Hormones released from the adipose tissue like leptin and adiponectin are also affected and leptin plasma levels are reduced in treated patients. Besides homeostatic control of body weight, surgery also changes hedonistic behavior in regard to food intake and cognitive performance involving the limbic system and prefrontal areas.


Subject(s)
Bariatric Surgery , Brain/metabolism , Cognition , Energy Metabolism , Obesity/surgery , Adiponectin/metabolism , Brain/physiopathology , Eating , Feeding Behavior , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Homeostasis , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Leptin/metabolism , Limbic System/metabolism , Limbic System/physiopathology , Obesity/metabolism , Obesity/physiopathology , Obesity/psychology , Peptide YY/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Vagus Nerve/physiopathology
10.
Mol Metab ; 6(1): 48-60, 2017 01.
Article in English | MEDLINE | ID: mdl-28123937

ABSTRACT

OBJECTIVE: Dietary supplementation with fermentable carbohydrate protects against body weight gain. Fermentation by the resident gut microbiota produces short-chain fatty acids, which act at free fatty acid receptor 2 (FFAR2). Our aim was to test the hypothesis that FFAR2 is important in regulating the beneficial effects of fermentable carbohydrate on body weight and to understand the role of gut hormones PYY and GLP-1. METHODS: Wild-type or Ffar2-/- mice were fed an inulin supplemented or control diet. Mice were metabolically characterized and gut hormone concentrations, enteroendocrine cell density measurements were carried out. Intestinal organoids and colonic cultures were utilized to substantiate the in vivo findings. RESULTS: We provide new mechanistic insight into how fermentable carbohydrate regulates metabolism. Using mice that lack FFAR2, we demonstrate that the fermentable carbohydrate inulin acts via this receptor to drive an 87% increase in the density of cells that produce the appetite-suppressing hormone peptide YY (PYY), reduce food intake, and prevent diet-induced obesity. CONCLUSION: Our results demonstrate that FFAR2 is predominantly involved in regulating the effects of fermentable carbohydrate on metabolism and does so, in part, by enhancing PYY cell density and release. This highlights the potential for targeting enteroendocrine cell differentiation to treat obesity.


Subject(s)
Dietary Carbohydrates/metabolism , Peptide YY/metabolism , Receptors, Cell Surface/metabolism , Animals , Body Weight , Colon/cytology , Dietary Supplements , Eating , Fatty Acids, Volatile/metabolism , Fermentation , Fermented Foods , Gastrointestinal Hormones/metabolism , Gastrointestinal Microbiome/physiology , Glucagon-Like Peptide 1/metabolism , Inulin/metabolism , Male , Mice , Mice, Knockout , Obesity/metabolism , Receptors, Cell Surface/physiology , Weight Gain
11.
Peptides ; 88: 97-105, 2017 02.
Article in English | MEDLINE | ID: mdl-27988351

ABSTRACT

In vertebrates, the neuropeptide Y (NPY) family peptides have been recognized as key players in food intake regulation. NPY centrally promotes feeding, while peptide YY (PYY) and pancreatic polypeptide (PP) mediate satiety. The teleost tetraploidization is well-known to generate duplicates of both NPY and PYY; however, the functional diversification between the duplicate genes, especially in the regulation of food intake, remains unknown. In this study, we identified the two duplicates of NPY and PYY in Nile tilapia (Oreochromis niloticus). Both NPYa and NPYb were primarily expressed in the central nervous system (CNS), but the mRNA levels of NPYb were markedly lower than those of NPYa. Hypothalamic mRNA expression of NPYa, but not NPYb, decreased after feeding and increased after 7-days of fasting. However, both NPYa and NPYb caused a significant increase in food intake after an intracranial injection of 50ng/g body weight dose. PYYb, one of the duplicates of PYY, had an extremely high expression in the foregut and midgut, whereas another form of duplicate PYYa showed only moderate expression in the CNS. Both hypothalamic PYYa and foregut PYYb mRNA expression increased after feeding and decreased after 7-days of fasting. Furthermore, the intracranial injection of PYYb decreased food intake, but PYYa had no significant effect. Our results suggested that although the mature peptides of NPYa and NPYb can both stimulate food intake, NPYa is the main endogenous functional NPY for feeding regulation. A functional division has been identified in the duplicates of PYY, which deems PYYb as a gut-derived anorexigenic peptide and PYYa as a CNS-specific PYY in Nile tilapia.


Subject(s)
Eating/genetics , Neuropeptide Y/metabolism , Pancreatic Polypeptide/metabolism , Peptide YY/metabolism , Amino Acid Sequence , Animals , Appetite Regulation/genetics , Cichlids/metabolism , Hypothalamus/metabolism , Neuropeptide Y/genetics , Pancreatic Polypeptide/genetics , Peptide YY/genetics , RNA, Messenger/genetics , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
12.
Gen Comp Endocrinol ; 242: 11-17, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27118705

ABSTRACT

Peptide YY (PYY) functions as a postprandial satiety signal in mammals. However, the genomic information and physiological roles of chicken PYY have not yet been clarified, although PYY peptide was isolated from chicken intestines in 1992. In this study, we identified a full-length complementary DNA (cDNA) sequence encoding the chicken PYY precursor. The deduced amino acid sequence of chicken PYY was completely consistent with the previously identified peptide sequence. PYY mRNA was abundantly expressed in the small intestine compared with the large intestine. PYY mRNA levels in the jejunum were significantly higher during ad libitum feeding compared with fasting, suggesting that intestinal PYY expression is altered in response to nutritional status in chicks. Intravenous administration of PYY significantly suppressed food intake in chicks. Furthermore, neuropeptide Y receptor Y2, a possible target of PYY, was expressed in various brain regions including the appetite-regulating centers in chicks. This is the first evidence that the intestinal hormone PYY may function as an anorexigenic hormone in chicks.


Subject(s)
Chickens/metabolism , Eating/drug effects , Peptide YY/metabolism , Peptide YY/pharmacology , Administration, Intravenous , Amino Acid Sequence , Animals , Appetite/physiology , Brain/metabolism , DNA, Complementary , Gene Expression Regulation/physiology , Jejunum/metabolism , Peptide YY/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Satiety Response
13.
Diabetes Obes Metab ; 18(5): 508-18, 2016 May.
Article in English | MEDLINE | ID: mdl-26863991

ABSTRACT

AIMS: To investigate the anorectic effect of L-arginine (L-Arg) in rodents. METHODS: We investigated the effects of L-Arg on food intake, and the role of the anorectic gut hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), the G-protein-coupled receptor family C group 6 member A (GPRC6A) and the vagus nerve in mediating these effects in rodents. RESULTS: Oral gavage of L-Arg reduced food intake in rodents, and chronically reduced cumulative food intake in diet-induced obese mice. Lack of the GPRC6A in mice and subdiaphragmatic vagal deafferentation in rats did not influence these anorectic effects. L-Arg stimulated GLP-1 and PYY release in vitro and in vivo. Pharmacological blockade of GLP-1 and PYY receptors did not influence the anorectic effect of L-Arg. L-Arg-mediated PYY release modulated net ion transport across the gut mucosa. Intracerebroventricular (i.c.v.) and intraperitoneal (i.p.) administration of L-Arg suppressed food intake in rats. CONCLUSIONS: L-Arg reduced food intake and stimulated gut hormone release in rodents. The anorectic effect of L-Arg is unlikely to be mediated by GLP-1 and PYY, does not require GPRC6A signalling and is not mediated via the vagus. I.c.v. and i.p. administration of L-Arg suppressed food intake in rats, suggesting that L-Arg may act on the brain to influence food intake. Further work is required to determine the mechanisms by which L-Arg suppresses food intake and its utility in the treatment of obesity.


Subject(s)
Appetite Depressants/therapeutic use , Arginine/therapeutic use , Dietary Supplements , Gastrointestinal Agents/therapeutic use , Glucagon-Like Peptide 1/agonists , Obesity/diet therapy , Peptide YY/agonists , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/adverse effects , Appetite Depressants/pharmacology , Arginine/administration & dosage , Arginine/adverse effects , Cells, Cultured , Dietary Supplements/adverse effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/pharmacology , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , In Vitro Techniques , Injections, Intraperitoneal , Injections, Intraventricular , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Peptide YY/blood , Peptide YY/metabolism , Random Allocation , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Weight Loss/drug effects
14.
Am J Clin Nutr ; 103(2): 305-13, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718419

ABSTRACT

BACKGROUND: Capsaicin, which is the major pungent principle in chili peppers, is able to induce satiety and reduce caloric intake. The exact mechanism behind this satiating effect is still unknown. We hypothesized that capsaicin induces satiety through the release of gastrointestinal peptides, such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), from enteroendocrine cells in the small intestine. OBJECTIVE: We investigate the effects of an intraduodenal capsaicin infusion (1.5 mg pure capsaicin) in healthy volunteers on hunger, satiety, and gastrointestinal symptoms and the release of GLP-1 and PYY. DESIGN: Thirteen participants (7 women) [mean ± SEM age: 21.5 ± 0.6 y; body mass index (in kg/m(2)): 22.8 ± 0.6] participated in this single-blind, randomized, placebo-controlled crossover study with 2 different treatments. During test days, an intraduodenal infusion of either capsaicin or a placebo (physiologic saline) was performed with the use of a nasoduodenal catheter over a period of 30 min. Visual analog scale scores were used to measure hunger, satiety, and gastrointestinal symptoms. Blood samples were drawn at regular intervals for GLP-1 and PYY. Gallbladder volumes were measured with the use of real-time ultrasonography. RESULTS: The intraduodenal capsaicin infusion significantly increased satiety (P-treatment effect < 0.05) but also resulted in an increase in the gastrointestinal symptoms pain (P-treatment × time interaction < 0.0005), burning sensation (P-treatment × time interaction < 0.0001), nausea (P-treatment × time interaction < 0.05), and bloating (P-treatment × time interaction < 0.001) compared with the effects of the placebo infusion. Satiety scores had a positive correlation with all gastrointestinal symptoms. No differences in GLP-1 and PYY concentrations and gallbladder volumes were observed after the capsaicin infusion compared with after the placebo infusion. CONCLUSIONS: An intraduodenal infusion of capsaicin significantly increases satiety but does not affect plasma concentrations of GLP-1 and PYY. Rather, the effect on satiety seems related to gastrointestinal stress as shown by the associations with pain, burning sensation, nausea, and bloating scores. This trial was registered at clinicaltrials.gov as NCT01667523.


Subject(s)
Appetite Depressants/adverse effects , Capsaicin/adverse effects , Dietary Supplements/adverse effects , Enteritis/etiology , Glucagon-Like Peptide 1/metabolism , Peptide YY/metabolism , Satiety Response , Abdominal Pain/etiology , Adult , Appetite Depressants/administration & dosage , Biomarkers , Capsaicin/administration & dosage , Cross-Over Studies , Enteritis/metabolism , Enteritis/pathology , Enteritis/physiopathology , Female , Gallbladder/diagnostic imaging , Gallbladder/metabolism , Gallbladder/pathology , Glucagon-Like Peptide 1/blood , Humans , Intubation, Gastrointestinal , Nausea/etiology , Organ Size , Pain Measurement , Peptide YY/blood , Single-Blind Method , Ultrasonography , Young Adult
15.
Gastrointest Endosc ; 83(1): 48-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26271184

ABSTRACT

BACKGROUND AND AIMS: The aims of this article were to review the discrepancy between numbers of people requiring weight loss treatment and results and to assess the potential effects of pharmacologic treatments (recently approved for obesity) and endoscopically deployed devices on quantitative GI traits in development for obesity treatment. METHODS: We conducted a review of relevant literature to achieve our objectives. RESULTS: The 2013 guidelines increased the number of adults recommended for weight loss treatment by 20.9% (116.0 million to 140.2 million). There is an imbalance between efficacy and costs of commercial weight loss programs and drug therapy (average weight loss about 5 kg). The number of bariatric procedures performed in the United States has doubled in the past decade. The efficacy of bariatric surgery is attributed to reduction in the volume of the stomach, nutrient malabsorption with some types of surgery, increased postprandial incretin responses, and activation of farnesoid X receptor mechanisms. These GI and behavioral traits identify sub-phenotypes of obesity, based on recent research. CONCLUSIONS: The mechanisms or traits targeted by drug and device treatments include centrally mediated alterations of appetite or satiation, diversion of nutrients, and alteration of stomach capacity, gastric emptying, or incretin hormones. Future treatment may be individualized based on quantitative GI and behavioral traits measured in obese patients.


Subject(s)
Anti-Obesity Agents/therapeutic use , Bariatric Surgery/methods , Equipment and Supplies , Obesity/therapy , Combined Modality Therapy , Depression/psychology , Endoscopy, Gastrointestinal/methods , Feeding Behavior , Gastric Balloon , Gastric Bypass/methods , Gastric Emptying , Gastroplasty/methods , Glucagon-Like Peptide 1/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Obesity/metabolism , Obesity/physiopathology , Obesity/psychology , Organ Size , Peptide YY/metabolism , Precision Medicine , Principal Component Analysis , Satiation , Stomach/pathology , Treatment Outcome , Weight Loss
16.
Appetite ; 98: 142-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26706043

ABSTRACT

Activation of free fatty acid receptor (FFAR)2 and FFAR3 via colonic short-chain fatty acids, particularly propionate, are postulated to explain observed inverse associations between dietary fiber intake and body weight. Propionate is reported as the predominant colonic fermentation product from l-rhamnose, a natural monosaccharide that resists digestion and absorption reaching the colon intact, while effects of long-chain inulin on appetite have not been extensively investigated. In this single-blind randomized crossover study, healthy unrestrained eaters (n = 13) ingested 25.5 g/d l-rhamnose, 22.4 g/d inulin or no supplement (control) alongside a standardized breakfast and lunch, following a 6-d run-in to investigate if appetite was inhibited. Postprandial qualitative appetite, breath hydrogen, and plasma glucose, insulin, triglycerides and non-esterified fatty acids were assessed for 420 min, then an ad libitum meal was provided. Significant treatment x time effects were found for postprandial insulin (P = 0.009) and non-esterified fatty acids (P = 0.046) with a significantly lower insulin response for l-rhamnose (P = 0.023) than control. No differences between treatments were found for quantitative and qualitative appetite measures, although significant treatment x time effects for meal desire (P = 0.008) and desire to eat sweet (P = 0.036) were found. Breath hydrogen was significantly higher with inulin (P = 0.001) and l-rhamnose (P = 0.009) than control, indicating colonic fermentation. These findings suggest l-rhamnose may inhibit postprandial insulin secretion, however neither l-rhamnose or inulin influenced appetite.


Subject(s)
Appetite/drug effects , Colon/drug effects , Energy Intake , Insulin/metabolism , Propionates/blood , Rhamnose/administration & dosage , Adolescent , Adult , Blood Glucose/metabolism , Body Mass Index , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Colon/metabolism , Cross-Over Studies , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Dietary Proteins/administration & dosage , Dietary Supplements , Fatty Acids, Nonesterified/blood , Female , Glucagon-Like Peptide 1/metabolism , Humans , Insulin Resistance , Insulin Secretion , Inulin/administration & dosage , Male , Middle Aged , Peptide YY/metabolism , Postprandial Period/drug effects , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Single-Blind Method , Waist Circumference , Young Adult
17.
Cell Metab ; 23(2): 324-34, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26621107

ABSTRACT

The composition of gut microbiota has been associated with host metabolic phenotypes, but it is not known if gut bacteria may influence host appetite. Here we show that regular nutrient provision stabilizes exponential growth of E. coli, with the stationary phase occurring 20 min after nutrient supply accompanied by bacterial proteome changes, suggesting involvement of bacterial proteins in host satiety. Indeed, intestinal infusions of E. coli stationary phase proteins increased plasma PYY and their intraperitoneal injections suppressed acutely food intake and activated c-Fos in hypothalamic POMC neurons, while their repeated administrations reduced meal size. ClpB, a bacterial protein mimetic of α-MSH, was upregulated in the E. coli stationary phase, was detected in plasma proportional to ClpB DNA in feces, and stimulated firing rate of hypothalamic POMC neurons. Thus, these data show that bacterial proteins produced after nutrient-induced E. coli growth may signal meal termination. Furthermore, continuous exposure to E. coli proteins may influence long-term meal pattern.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/growth & development , Gastrointestinal Tract/microbiology , Satiety Response , Adenosine Triphosphate/biosynthesis , Amygdala/metabolism , Animals , Electrophysiological Phenomena , Endopeptidase Clp , Escherichia coli/metabolism , Feeding Behavior , Female , Glucagon-Like Peptide 1/metabolism , Heat-Shock Proteins/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Peptide YY/metabolism , Pro-Opiomelanocortin/metabolism , Proteomics , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Rats, Wistar
18.
PLoS One ; 10(8): e0136873, 2015.
Article in English | MEDLINE | ID: mdl-26312995

ABSTRACT

Activation of TGR5 via bile acids or bile acid analogs leads to the release of glucagon-like peptide-1 (GLP-1) from intestine, increases energy expenditure in brown adipose tissue, and increases gallbladder filling with bile. Here, we present compound 18, a non-bile acid agonist of TGR5 that demonstrates robust GLP-1 secretion in a mouse enteroendocrine cell line yet weak GLP-1 secretion in a human enteroendocrine cell line. Acute administration of compound 18 to mice increased GLP-1 and peptide YY (PYY) secretion, leading to a lowering of the glucose excursion in an oral glucose tolerance test (OGTT), while chronic administration led to weight loss. In addition, compound 18 showed a dose-dependent increase in gallbladder filling. Lastly, compound 18 failed to show similar pharmacological effects on GLP-1, PYY, and gallbladder filling in Tgr5 knockout mice. Together, these results demonstrate that compound 18 is a mouse-selective TGR5 agonist that induces GLP-1 and PYY secretion, and lowers the glucose excursion in an OGTT, but only at doses that simultaneously induce gallbladder filling. Overall, these data highlight the benefits and potential risks of using TGR5 agonists to treat diabetes and metabolic diseases.


Subject(s)
Gallbladder/drug effects , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Drug Evaluation, Preclinical/methods , Gallbladder/physiopathology , Gene Expression Regulation , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Peptide YY/metabolism , Receptors, G-Protein-Coupled/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Weight Loss/drug effects
19.
Nutr Res ; 35(5): 393-400, 2015 May.
Article in English | MEDLINE | ID: mdl-25823991

ABSTRACT

We hypothesized that a digestion-resistant maltodextrin, Fibersol-2 (Archer Daniels Midland/Matsutani LLC, Decatur, IL, USA) may impact satiety by decreasing hunger, prolonging satiation, and/or increasing peripheral satiety signals. In a randomized, double-blind, placebo-controlled crossover study, healthy subjects (9 men and 10 women) underwent 3 treatments in which they consumed a standardized meal with a tea containing 0, 5, or 10 g of Fibersol-2. A visual analog scale questionnaire was given in 30-minute intervals to measure subjective appetite and satiety. Blood was drawn just before the meal (time 0) and at 30, 60, 90, 120, 180, and 240 minutes after meal for measurements of plasma ghrelin, cholecystokinin, gastrin, peptide YY, gastric inhibitory polypeptide, and glucagon-like peptide-1, all by enzyme-linked immunosorbent assay. There were significant delays in hunger and increased satiety for 1.5 to 2 hours after treatment with 10 g of Fibersol-2. These delays did not occur after ingesting 0 or 5 g Fibersol-2 at any time. Control and 5 g Fibersol-2 treatments did not suppress increases in hunger postmeal; hunger scores increased and satiety scores decreased significantly (P < .05) at all time points relative to the first postmeal assessment. Plasma peptide YY and glucagon-like peptide-1 were significantly increased by the ingestion of meal with tea containing 10 g Fibersol-2 compared with 0 or 5 g Fibersol-2 (P < .05). This study demonstrated that 10 g Fibersol-2 with a meal stimulated production of satiety hormones and enhanced satiety.


Subject(s)
Appetite Depressants/therapeutic use , Glucagon-Like Peptide 1/agonists , Overweight/diet therapy , Peptide YY/agonists , Polysaccharides/therapeutic use , Prebiotics , Satiety Response , Adult , Appetite Depressants/administration & dosage , Appetite Depressants/chemistry , Body Mass Index , Cross-Over Studies , Double-Blind Method , Enteroendocrine Cells/metabolism , Female , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Humans , Lunch , Male , Middle Aged , Overweight/blood , Overweight/metabolism , Peptide YY/blood , Peptide YY/metabolism , Polysaccharides/administration & dosage , Polysaccharides/chemistry , Prebiotics/administration & dosage , Solubility , Tea , Viscosity , Young Adult
20.
Trends Endocrinol Metab ; 26(3): 125-35, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662369

ABSTRACT

Obesity develops when energy intake exceeds energy expenditure over time. Numerous neurotransmitters, hormones, and factors have been implicated to coordinately control energy homeostasis, centrally and peripherally. However, the neuropeptide Y (NPY) system has emerged as the one with the most critical functions in this process. While NPY centrally promotes feeding and reduces energy expenditure, peptide YY (PYY) and pancreatic polypeptide (PP), the other family members, mediate satiety. Importantly, recent research has uncovered additional functions for these peptides that go beyond the simple feeding/satiety circuits and indicate a more extensive function in controlling energy homeostasis. In this review, we will discuss the actions of the NPY system in the regulation of energy balance, with a particular focus on energy expenditure.


Subject(s)
Energy Intake/physiology , Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/metabolism , Neuropeptide Y/metabolism , Obesity/metabolism , Humans , Peptide YY/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL