Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nutrients ; 15(20)2023 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-37892464

RESUMO

Previously, it has been indicated that oat polar lipids included in a liquid meal may have the potential to beneficially modulate various cardiometabolic variables. The purpose of this study was to evaluate the effects of oat polar lipids in a solid food matrix on acute and second meal glucose tolerance, blood lipids, and concentrations of gut-derived hormones. The oat polar lipids were consumed at breakfast and effects on the biomarkers were investigated in the postprandial period and following a standardized lunch. Twenty young, healthy subjects consumed in total four different breakfast meals in a crossover study design. The breakfasts consisted of 1. White wheat bread (WWB) with an added 7.5 g of oat polar lipids (PLL); 2. WWB with an added 15 g of oat polar lipids (PLH); 3. WWB with and added 16.6 g of rapeseed oil (RSO) as a representative of commonly consumed oils; and 4. WWB consumed alone, included as a reference. All products with added lipids contained equivalent amounts of fat (16.6 g) and available carbohydrates (50 g). Rapeseed oil was added to the oat polar lipid meals to equal 16.6 g of total fat. The standardized lunch was composed of WWB and meatballs and was served 3.5 h after the breakfast. Test variables (blood glucose, serum insulin, triglyceride (TG), free fatty acids (FFA), ghrelin, GLP-1, PYY, and GIP) were measured at fasting and repeatedly during the 5.5 h after ingestion of the breakfast. After breakfast, PLH substantially lowered postprandial glucose and insulin responses (iAUC 0-120 min) compared with RSO and WWB (p < 0.05). Furthermore, a reduced glycaemic response to lunch (210-330 min) was observed following the PLH breakfast compared to all of the other breakfasts served (p < 0.05). Oat polar lipids (PLH) significantly reduced TG and ghrelin and increased circulating gut hormones GLP-1 and PYY compared to RSO (p < 0.05). The results show that exchanging part of the dietary lipids with oat polar lipids has the potential to improve postprandial blood glucose regulation and gut hormones and thus may have a preventive effect against type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Hormônios Gastrointestinais , Humanos , Grelina , Desjejum , Glicemia , Estudos Cross-Over , Avena , Voluntários Saudáveis , Óleo de Brassica napus , Fibras na Dieta , Refeições , Insulina , Peptídeo 1 Semelhante ao Glucagon , Lipídeos , Período Pós-Prandial
2.
Nutrients ; 14(7)2022 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-35405964

RESUMO

Diabetes is expected to increase up to 700 million people worldwide with type 2 diabetes being the most frequent. The use of nutritional interventions is one of the most natural approaches for managing the disease. Minerals are of paramount importance in order to preserve and obtain good health and among them molybdenum is an essential component. There are no studies about the consumption of biofortified food with molybdenum on glucose homeostasis but recent studies in humans suggest that molybdenum could exert hypoglycemic effects. The present study aims to assess if consumption of lettuce biofortified with molybdenum influences glucose homeostasis and whether the effects would be due to changes in gastrointestinal hormone levels and specifically Peptide YY (PYY), Glucagon-Like Peptide 1 (GLP-1), Glucagon-Like Peptide 2 (GLP-2), and Gastric Inhibitory Polypeptide (GIP). A cohort of 24 people was supplemented with biofortified lettuce for 12 days. Blood and urine samples were obtained at baseline (T0) and after 12 days (T2) of supplementation. Blood was analyzed for glucose, insulin, insulin resistance, ß-cell function, and insulin sensitivity, PYY, GLP-1, GLP-2 and GIP. Urine samples were tested for molybdenum concentration. The results showed that consumption of lettuce biofortified with molybdenum for 12 days did not affect beta cell function but significantly reduced fasting glucose, insulin, insulin resistance and increased insulin sensitivity in healthy people. Consumption of biofortified lettuce did not show any modification in urine concentration of molybdenum among the groups. These data suggest that consumption of lettuce biofortified with molybdenum improves glucose homeostasis and PYY and GIP are involved in the action mechanism.


Assuntos
Diabetes Mellitus Tipo 2 , Alimentos Fortificados , Resistência à Insulina , Molibdênio , Glicemia , Polipeptídeo Inibidor Gástrico , Peptídeo 1 Semelhante ao Glucagon , Peptídeo 2 Semelhante ao Glucagon , Glucose , Homeostase , Humanos , Insulina , Lactuca , Molibdênio/administração & dosagem , Peptídeo YY
3.
Front Nutr ; 9: 827630, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35198590

RESUMO

Exercise and diet are important factors for energy balance and appetite regulation. The aim of this study was to investigate the effect of 8 weeks High Intensity Interval Training (HIIT) and vitamin D3 supplementation in sedentary overweight men. Forty-eight participants were randomly assigned to one of the following four groups (n = 12): HIIT + VitD, HIIT + placebo (3 sessions per week, 10 × 1 min interval cycling at 90-100% VO2peak separated by 1 min active recovery at 15% VO2peakfor 8 weeks), Vit D and control groups. Participants received 2,000 IU/day 25 (OH) D3 or placebo. Measurements were taken pre and post training after 10 h overnight fasting. Insulin, weight, BMI and body fat percentage were significantly decreased, but PYY was significantly increased in the HIIT + Vit D and HIIT + placebo groups (p = 0.001 and p = 0.001, respectively) after 8 weeks of HIIT. Insulin (p = 0.009, p = 0.001), weight, BMI and body fat percentage (p = 0.001, p = 0.001) were significantly lower in the HIIT + Vit D and HIIT + placebo groups compared to the Vit D and control groups. However, PYY was significantly higher in the HIIT + Vit D group compared to the Vit D (p = 0.025) and control groups (p = 0.007) and also in the HIIT + placebo group compared to the Vit D (p = 0.037) and control groups (p = 0.032) after 8 weeks of HIIT. The combination of regular HIIT with vitamin D supplementation has a effect on appetite control and body composition.

4.
Mol Metab ; 55: 101392, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34781035

RESUMO

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.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Obesidade/metabolismo , Peptídeo YY/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Energia/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Derivação Gástrica , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Hipotálamo , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/fisiopatologia , Peptídeo YY/fisiologia , Redução de Peso
5.
Nutrients ; 14(1)2021 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-35010991

RESUMO

BACKGROUND: The hypothalamus is an important brain region for the regulation of energy balance. Roux-en-Y gastric bypass (RYGB) surgery and gut hormone-based treatments are known to reduce body weight, but their effects on hypothalamic gene expression and signaling pathways are poorly studied. METHODS: Diet-induced obese male Wistar rats were randomized into the following groups: RYGB, sham operation, sham + body weight-matched (BWM) to the RYGB group, osmotic minipump delivering PYY3-36 (0.1 mg/kg/day), liraglutide s.c. (0.4 mg/kg/day), PYY3-36 + liraglutide, and saline. All groups (except BWM) were kept on a free choice of high- and low-fat diets. Four weeks after interventions, hypothalami were collected for RNA sequencing. RESULTS: While rats in the RYGB, BWM, and PYY3-36 + liraglutide groups had comparable reductions in body weight, only RYGB and BWM treatment had a major impact on hypothalamic gene expression. In these groups, hypothalamic leptin receptor expression as well as the JAK-STAT, PI3K-Akt, and AMPK signaling pathways were upregulated. No significant changes could be detected in PYY3-36 + liraglutide-, liraglutide-, and PYY-treated groups. CONCLUSIONS: Despite causing similar body weight changes compared to RYGB and BWM, PYY3-36 + liraglutide treatment does not impact hypothalamic gene expression. Whether this striking difference is favorable or unfavorable to metabolic health in the long term requires further investigation.


Assuntos
Hormônios Gastrointestinais/farmacologia , Hipotálamo/metabolismo , Liraglutida/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Transcriptoma/efeitos dos fármacos , Animais , Peso Corporal , Restrição Calórica , Modelos Animais de Doenças , Metabolismo Energético , Derivação Gástrica , Expressão Gênica/efeitos dos fármacos , Masculino , Obesidade , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
6.
Biomolecules ; 10(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492958

RESUMO

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.


Assuntos
Colecistocinina/metabolismo , Extratos Vegetais/farmacologia , Proantocianidinas/farmacologia , Animais , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Peptídeo YY/metabolismo , Extratos Vegetais/química , Proantocianidinas/química , Sementes/química , Suínos , Vitis/química
7.
Nutrients ; 12(3)2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-32183423

RESUMO

The satiating effect of whey proteins depends upon their unique amino acid composition because there is no difference when comparing whey proteins or a mix of amino acids mimicking the amino acid composition of whey proteins. The specific amino acids underlying the satiating effect of whey proteins have not been investigated to date. AIMS AND METHODS: The aim of the present study was to evaluate the appetite-suppressant effect of an isocaloric drink containing whey proteins or maltodextrins on appetite (satiety/hunger measured by a visual analogue scale or VAS), anorexigenic gastrointestinal peptides (circulating levels of glucagon-like peptide 1 (GLP-1) and peptide tyrosine tyrosine (PYY)) and amino acids (circulating levels of single, total [TAA] and branched-chain amino acids [BCAA]) in a cohort of obese female subjects (n = 8; age: 18.4 ± 3.1 years; body mass index, BMI: 39.2 ± 4.6 kg/m2). RESULTS: Each drink significantly increased satiety and decreased hunger, the effects being more evident with whey proteins than maltodextrins. Similarly, circulating levels of GLP-1, PYY and amino acids (TAA, BCAA and alanine, arginine, asparagine, citrulline, glutamine, hydroxyproline, isoleucine, histidine, leucine, lysine, methionine, ornithine, phenylalanine, proline, serine, threonine, tyrosine, and valine) were significantly higher with whey proteins than maltodextrins. In subjects administered whey proteins (but not maltodextrins), isoleucine, leucine, lysine, methionine, phenylalanine, proline, tyrosine, and valine were significantly correlated with hunger (negatively), satiety, and GLP-1 (positively). CONCLUSIONS: Eight specific amino acids (isoleucine, leucine, lysine, methionine, phenylalanine, proline, tyrosine, and valine) were implicated in the appetite-suppressant and GLP-1-stimulating effects of whey proteins, which may be mediated by their binding with nutrient-sensing receptors expressed by L cells within the gastrointestinal wall. The long-term satiating effect of whey proteins and the effectiveness of a supplementation with these amino acids (i.e., as a nutraceutical intervention) administered during body weight reduction programs need to be further investigated.


Assuntos
Aminoácidos/sangue , Depressores do Apetite/administração & dosagem , Bebidas , Peptídeo 1 Semelhante ao Glucagon/efeitos dos fármacos , Obesidade/fisiopatologia , Proteínas do Soro do Leite/administração & dosagem , Adolescente , Apetite/efeitos dos fármacos , Estudos Cross-Over , Dipeptídeos/efeitos dos fármacos , Células Enteroendócrinas/metabolismo , Feminino , Humanos , Isoleucina/sangue , Leucina/sangue , Lisina/sangue , Metionina/sangue , Obesidade/terapia , Fenilalanina/sangue , Polissacarídeos/administração & dosagem , Prolina/sangue , Tirosina/sangue , Valina/sangue , Adulto Jovem
8.
J Nutr Sci ; 8: e15, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037218

RESUMO

Coffee consumption is inversely associated with the risk of non-alcoholic fatty liver disease (NAFLD). A gap in the literature still exists concerning the intestinal mechanisms that are involved in the protective effect of coffee consumption towards NAFLD. In this study, twenty-four C57BL/6J mice were divided into three groups each receiving a standard diet, a high-fat diet (HFD) or an HFD plus decaffeinated coffee (HFD+COFFEE) for 12 weeks. Coffee supplementation reduced HFD-induced liver macrovesicular steatosis (P < 0·01) and serum cholesterol (P < 0·001), alanine aminotransferase and glucose (P < 0·05). Accordingly, liver PPAR- α (P < 0·05) and acyl-CoA oxidase-1 (P < 0·05) as well as duodenal ATP-binding cassette (ABC) subfamily A1 (ABCA1) and subfamily G1 (ABCG1) (P < 0·05) mRNA expressions increased with coffee consumption. Compared with HFD animals, HFD+COFFEE mice had more undigested lipids in the caecal content and higher free fatty acid receptor-1 mRNA expression in the duodenum and colon. Furthermore, they showed an up-regulation of duodenal and colonic zonulin-1 (P < 0·05), duodenal claudin (P < 0·05) and duodenal peptide YY (P < 0·05) mRNA as well as a higher abundance of Alcaligenaceae in the faeces (P < 0·05). HFD+COFFEE mice had an energy intake comparable with HFD-fed mice but starting from the eighth intervention week they gained significantly less weight over time. Data altogether showed that coffee supplementation prevented HFD-induced NAFLD in mice by reducing hepatic fat deposition and metabolic derangement through modification of pathways underpinning liver fat oxidation, intestinal cholesterol efflux, energy metabolism and gut permeability. The hepatic and metabolic benefits induced by coffee were accompanied by changes in the gut microbiota.


Assuntos
Café/metabolismo , Dieta Hiperlipídica/efeitos adversos , Intestinos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Acil-CoA Oxidase/metabolismo , Alanina Transaminase/sangue , Alcaligenaceae , Animais , Glicemia , Colesterol/sangue , Claudinas/metabolismo , Suplementos Nutricionais , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Haptoglobinas/metabolismo , Fígado/patologia , Masculino , Síndrome Metabólica , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/metabolismo , Polifenóis/farmacologia , Precursores de Proteínas/metabolismo , RNA Mensageiro/metabolismo
9.
Neuropeptides ; 73: 89-95, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30471778

RESUMO

Peptide YY(3-36) ((PYY(3-36)) and glucagon like peptide 1 (GLP-1) in combination reduce food intake and body weight in an additive or synergistic manner in animal models and in humans. Nevertheless, the mechanisms behind are not completely understood. The present study aims to investigate the effect of combining PYY(3-36) and the GLP-1 receptor agonist exendin-4 (Ex4) by examining acute food intake and global neuronal activation as measured by c-fos in C57BL/6 J mice. An additive reduction in food intake was found 1.5 h after s.c dosing with the combination of PYY(3-36) (200 µg/kg) and Ex4 (2.5 µg/kg). This was associated with a synergistic enhancement of c-fos reactivity in central amygdalar nucleus (CeA), rostral part of the mediobasal arcuate nucleus (ARH), supratrigeminal nucleus (SUT), lateral parabrachial nucleus (PB), area postrema (AP) and nucleus tractus solitarius (NTS) compared to vehicle, PYY(3-36) and Ex4 individually dosed mice. The regions activated by Ex4 individually and PYY(3-36) and Ex4 in combination resembled each other, but the combination group had a significantly stronger c-fos response. Twenty-five brain areas were activated by PYY(3-36) and Ex4 in combination compared to vehicle versus nine brain areas by Ex4 individually. No significant increase in c-fos reactivity was found by PYY(3-36) compared to vehicle dosed mice. The neuronal activation of ARH and the AP/NTS to PB to CeA pathway is important for appetite regulation while SUT has not previously been reported in the regulation of energy balance. As PYY(3-36) and Ex4 act on different neurons leading to recruitment of different signalling pathways within and to the brain, an interaction of these pathways may contribute to their additive/synergistic action. Thus, PYY(3-36) boosts the effect of Ex4 possibly by inducing less inhibition of neuronal activity leading to an enhanced neuronal activity induced by Ex4.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Exenatida/farmacologia , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Tonsila do Cerebelo/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
10.
J Nutr ; 148(5): 693-701, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29897544

RESUMO

BACKGROUND: Higher-protein meals (>25 g protein/meal) have been associated with enhanced satiety but the role of amino acids is unclear. Leucine has been proposed to stimulate satiety in rodents but has not been assessed in humans. OBJECTIVE: We assessed the acute effects of lower-protein nutrition bars, enhanced with a leucine peptide (LP), on postprandial appetite sensations in combination with plasma leucine and peptide YY (PYY) in healthy women. METHODS: Utilizing a double-blind randomized crossover design, 40 healthy women [28 ± 7.5 y; body mass index (BMI, in kg/m2): 23.5 ± 2.4] consumed the following isocaloric (180 kcal) pre-loads on 3 separate visits: control bar [9 g protein with 0 g added LP (0-g LP)] or treatment bars [11 g protein with 2 g added LP (2-g LP) or 13 g protein with 3 g added LP (3-g LP)]. Pre- and postprandial hunger, desire to eat, prospective food consumption (PFC), fullness, and plasma leucine were assessed every 30 min for 240 min. Plasma PYY was assessed hourly for 240 min (n = 24). RESULTS: Main effects of time (P < 0.0001) and treatment (P < 0.03) were detected for postprandial hunger, desire to eat, PFC, and fullness. Post hoc analyses revealed that the 2-g and 3-g LP bars elicited greater increases in fullness and greater decreases in PFC compared with 0-g LP (all, P < 0.05) with no differences between the 2-g and 3-g LP bars. The 2-g bar elicited greater decreases in hunger and desire to eat compared with the 0-g LP bar (both, P ≤ 0.01), whereas 3-g LP did not. Appetite incremental areas under the curves (iAUCs) and PYY outcomes were not different between bars. A treatment × time interaction was detected for plasma leucine with increases occurring in a leucine-dose-dependent manner (P < 0.0001). CONCLUSION: Despite the dose-dependent increases in plasma leucine following the consumption of lower-protein bars enhanced with LP, only the 2-g LP bar elicited consistent postprandial changes in select appetite sensations compared with the 0-g LP bar. This study was registered on clinicaltrials.gov as NCT02091570.


Assuntos
Apetite/fisiologia , Proteínas Alimentares/administração & dosagem , Leucina/administração & dosagem , Período Pós-Prandial/fisiologia , Adolescente , Adulto , Estudos Cross-Over , Suplementos Nutricionais , Método Duplo-Cego , Ingestão de Alimentos/fisiologia , Feminino , Humanos , Leucina/sangue , Refeições , Pessoa de Meia-Idade , Peptídeo YY/sangue , Estudos Prospectivos , Saciação/fisiologia , Adulto Jovem
11.
Mol Neurobiol ; 55(9): 7555-7566, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29429048

RESUMO

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.


Assuntos
Apatia , Comportamento Animal , Imersão , Peptídeo YY/metabolismo , Receptores de Dopamina D2/metabolismo , Água , Animais , Apatia/efeitos dos fármacos , Peso Corporal , Corticosterona/sangue , Dopamina/metabolismo , Ingestão de Alimentos , Regulação da Expressão Gênica , Humanos , Hipotálamo/metabolismo , Interleucina-6/administração & dosagem , Interleucina-6/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Comportamento de Nidação , Tamanho do Órgão , Peptídeo YY/administração & dosagem , Peptídeo YY/farmacologia , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/metabolismo
12.
Peptides ; 100: 269-274, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29412828

RESUMO

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.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Obesidade/tratamento farmacológico , Peptídeo YY/uso terapêutico , Receptores de Neuropeptídeo Y/genética , Regulação do Apetite/efeitos dos fármacos , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Hipotálamo/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Fragmentos de Peptídeos/uso terapêutico , Peptídeo YY/metabolismo , Receptores de Neuropeptídeo Y/metabolismo
13.
Dig Dis Sci ; 62(12): 3350-3358, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29030744

RESUMO

BACKGROUND: Peptide tyrosine tyrosine 3-36 (peptide YY 3-36 or PYY 3-36) reduces food intake by unknown site(s). AIM: To test the hypothesis that the gastrointestinal tract contains sites of action regulating meal size (MS) and intermeal interval (IMI) length by PYY 3-36. METHODS: Peptide YY 3-36 (0, 1, 5, 10 and 20 nmol/kg) was injected in the aorta, the artery that supplies the gastrointestinal tract, prior to the onset of the dark cycle in free feeding male Sprague-Dawley rats and food intake was measured. Then, PYY 3-36 (25 nmol/kg) was injected intraperitoneally in these rats and Fos-like immunoreactivity (Fos-LI, a marker for neuronal activation) was quantified in the small intestinal enteric neurons, both myenteric and submucosal, and the dorsal vagal complex (DVC) of the hindbrain. RESULTS: PYY 3-36 reduced first MS, decreased IMI length, shortened duration of first meal and increased Fos-LI in enteric and DVC neurons. However, PYY 3-36 failed to change the size of the second meal, satiety ratio, latency to first meal, number of meals and 24 h intake relative to saline control. CONCLUSION: The gastrointestinal tract may contain sites of action regulating MS reduction by PYY 3-36.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Sistema Nervoso Entérico/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos , Masculino , Ratos Sprague-Dawley
14.
Nutrition ; 41: 14-23, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28760423

RESUMO

OBJECTIVE: Previous studies have shown that polyunsaturated fats (PUFAs) elicit a greater response in satiety after a single-meal challenge compared with other types of fats. The long-term effects of PUFAs on satiety, however, remain unknown. The aim of this study was to determine subjective and physiological hunger and satiety responses to high-fat (HF) meals before and after a 7-d PUFA-rich diet. METHODS: Twenty-six, healthy weight (body mass index 18-24.9 kg/m2), sedentary adults were randomly assigned to either a 7-d PUFA-rich diet (n = 8 men and n = 8 women) or a 7-d control diet (n = 5 men and n = 5 women). After a 3-d lead-in diet, participants reported for the baseline visit where anthropometrics, fasting visual analog scale (VAS) measurements, and a fasting blood sample were collected. Then, two HF meals (breakfast and lunch) were consumed. Postprandial blood draws and VAS measures were collected approximately every 30 min for 4 h after each meal, for a total of 8 h. RESULTS: From pre- to post-PUFA-rich diet, there was a decrease in fasting ghrelin (P < 0.05) and an increase in fasting peptide YY (PYY; P < 0.05); however, there were no changes in fasting insulin or leptin concentrations. The postprandial response for PYY was higher after the PUFA-rich diet visit compared to baseline (P < 0.01). However, there were no differences in the postprandial response for ghrelin, insulin, leptin, or VAS measures from pre- to post-diet in either the PUFA-rich diet or control (ns). CONCLUSION: A PUFA-rich diet consumed for 7 d favorably altered fasting and postprandial physiological markers of hunger and satiety; yet, did not alter subjective ratings of hunger or fullness.


Assuntos
Dieta Hiperlipídica/métodos , Gorduras na Dieta/farmacologia , Ácidos Graxos Insaturados/farmacologia , Fome/efeitos dos fármacos , Resposta de Saciedade/efeitos dos fármacos , Adolescente , Adulto , Gorduras na Dieta/sangue , Ácidos Graxos Insaturados/sangue , Feminino , Grelina/sangue , Grelina/efeitos dos fármacos , Humanos , Masculino , Peptídeo YY/sangue , Peptídeo YY/efeitos dos fármacos , Saciação/efeitos dos fármacos , Adulto Jovem
15.
Neuromodulation ; 20(8): 774-782, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28795473

RESUMO

OBJECTIVES: Gastric electrical stimulation (GES) is an alternative therapy to treat patients with intractable vomiting. A preclinical study has demonstrated the modulation of the gastrointestinal (GI) peptide ghrelin by GES but such mechanism has never been investigated in patients. The aim of this work was to assess the effect of GES on GI peptide levels in patients with intractable vomiting. MATERIALS AND METHODS: Twenty-one patients were randomized to receive either ON or OFF GES, 14 completed the study (10 ON, 4 OFF stimulation). Vomiting episodes, gastric emptying, and gastrointestinal quality of life index (GIQLI) were assessed. Gastric and blood samples were collected before and four months after the ON period of gastric stimulation. mRNA and/or peptide levels were assessed in gastric biopsies for ghrelin, leptin, and NUCB2/nesfatin-1 and in duodenal biopsies for glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) using RT-qPCR and multiplex technology. Ghrelin, leptin, GLP-1, PYY, gastric inhibitory peptide (GIP), and NUCB2/nesfatin-1 levels also were quantified in blood samples. RESULTS: Among clinical parameters, vomiting episodes were slightly reduced by GES (p = 0.09). In tissue, mRNA or protein levels were not modified following chronic GES. In blood, a significant reduction of postprandial PYY levels (p < 0.05) was observed at M4 and a reduction of NUCB2/nesfatin-1 levels in fasted patients (p < 0.05). Increased plasma leptin levels after GES were correlated with reduction of vomiting and improvement of GIQLI. CONCLUSIONS: GES reduces NUCB2/nesfatin-1 levels under fasting conditions and postprandial PYY levels in patients suffering from nausea and/or vomiting refractory to pharmacological therapies.


Assuntos
Terapia por Estimulação Elétrica/métodos , Hormônios Gastrointestinais/sangue , Trato Gastrointestinal/metabolismo , Vômito/sangue , Vômito/terapia , Adulto , Proteínas de Ligação ao Cálcio/sangue , Estudos Cross-Over , Proteínas de Ligação a DNA/sangue , Método Duplo-Cego , Jejum/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/sangue , Nucleobindinas , Peptídeo YY/sangue , Período Pós-Prandial/fisiologia , Receptores dos Hormônios Gastrointestinais/sangue
16.
Neurosci Lett ; 655: 54-60, 2017 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-28684238

RESUMO

Peripheral anorectic hormones, such as peptide YY (PYY) and oxytocin (OXT), suppress food intake. A newly identified anorectic neuropeptide, nesfatin-1, is synthesized in both peripheral tissue and the central nervous system, particularly by various nuclei in the hypothalamus and brainstem. Here, we examined the effects of intraperitoneal (ip) administration of PYY3-36, OXT, and OXT analog, on nesfatin-1-immunoreactive (ir) neurons in the rat hypothalamus and brainstem, using Fos double fluorescence-immunohistochemistry. The ip administration of OXT and OXT analog significantly increased the number of nesfatin-1-ir neurons expressing Fos-ir in the paraventricular nucleus, the arcuate nucleus, and the nucleus tractus solitarius, but not in the supraoptic nucleus, the lateral hypothalamic area, and the area postrema. No differences in the percentage of nesfatin-1-ir neurons expressing Fos in the nuclei of the hypothalamus and brainstem were observed, between rats treated with vehicle or those treated with PYY3-36. The decreased food intake, induced by OXT and OXT analog, was attenuated significantly by pretreatment with intracerebroventricular administration of antisense nesfatin-1. These results suggested that nesfatin-1-expressing neurons in the hypothalamus and brainstem may play a role in sensing the peripheral level of OXT and its suppression of feeding in rats.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ocitocina/farmacologia , Animais , Tronco Encefálico/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação a DNA/genética , Hipotálamo/metabolismo , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Proteínas do Tecido Nervoso/genética , Nucleobindinas , Oligonucleotídeos Antissenso/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar
17.
Mol Nutr Food Res ; 61(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28371338

RESUMO

SCOPE: Nasturtium plants contain the glucosinolate glucotropaeolin and its corresponding breakdown product benzyl isothiocyanate (BITC), the latter being intensively studied with regard to cancer chemoprevention and anti-inflammatory properties. In addition, recent research has shown that isothiocyanates are able to activate the release of several gut hormones in vitro and in rodent studies. Here, we tested the effects of a dietary nasturtium administration on circulating levels of gut hormones in humans. METHODS AND RESULTS: Metabolically healthy males (n = 15) received a single oral dose of 10 g freeze-dried nasturtium leaf material suspended in water or only water (control). Blood samples were taken every hour and serum concentrations of insulin, C-peptide, glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and peptide (PYY) were analyzed. Oral nasturtium intake resulted in an increased release of PYY over a time period of 6 h whereas circulating levels of other hormones were not changed. CONCLUSION: Given the finding that nasturtium consumption enhances secretion of PYY, a key hormone involved in energy regulation, special diets containing nasturtium, or supplementation with nasturtium or BITC might be considered in the treatment of obesity.


Assuntos
Suplementos Nutricionais , Nasturtium , Peptídeo YY/sangue , Administração Oral , Adulto , Glicemia/metabolismo , Peptídeo C/sangue , Polipeptídeo Inibidor Gástrico/sangue , Variação Genética , Peptídeo 1 Semelhante ao Glucagon/sangue , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Receptores Acoplados a Proteínas G/genética
18.
J Appl Toxicol ; 37(7): 784-791, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28138998

RESUMO

Deoxynivalenol (DON), one of trichothecene mycotoxins produced by the fungus Fusarium, is commonly detected in cereal foods and in secondary food production across the world. Lower concentrations of DON induce a dose-related feed refusal (anorexia), whereas it acts as a potent emetic agent at higher levels. DON-induced emesis in humans and livestock can be observed and recorded in both undeveloped and developed regions such as Lixian, Guide and Huangzhong in China and Illinois in the USA. Some studies with different animal models (pigs and minks) suggested that DON could change expressions of 5-hydroxytryptamine, peptide YY, neuropeptide Y2 receptor and nucleobindin-2/nesfatin-1 in plasma and different areas of the brain. Some selective antagonist of 5-hydroxytryptamine 3 receptors can inhibit DON-induced emetic response. Otherwise, the Ca2+ homeostasis and MAPK pathway could be potential directions in future studies. Dolasetron, dantrolene and JNJ-31020028 can be used in clinical treatment but they have potential toxic effects. (-)Epicatechin, ginger phytochemicals and isoflavone can be tested in in vitro and in vivo for their usage as food additives for reducing the emesis. The present review summarizes and discusses some information from previous and recent prominent publications with the aim to provide some comprehensive and helpful data for understanding the mechanism of DON-induced emesis. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Micotoxinas/farmacocinética , Micotoxinas/toxicidade , Tricotecenos/farmacocinética , Tricotecenos/toxicidade , Vômito/induzido quimicamente , Vômito/fisiopatologia , Animais , China , Humanos , Estudos Prospectivos
19.
Neuroimage Clin ; 8: 1-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26110109

RESUMO

Functional, molecular and genetic neuroimaging has highlighted the existence of brain anomalies and neural vulnerability factors related to obesity and eating disorders such as binge eating or anorexia nervosa. In particular, decreased basal metabolism in the prefrontal cortex and striatum as well as dopaminergic alterations have been described in obese subjects, in parallel with increased activation of reward brain areas in response to palatable food cues. Elevated reward region responsivity may trigger food craving and predict future weight gain. This opens the way to prevention studies using functional and molecular neuroimaging to perform early diagnostics and to phenotype subjects at risk by exploring different neurobehavioral dimensions of the food choices and motivation processes. In the first part of this review, advantages and limitations of neuroimaging techniques, such as functional magnetic resonance imaging (fMRI), positron emission tomography (PET), single photon emission computed tomography (SPECT), pharmacogenetic fMRI and functional near-infrared spectroscopy (fNIRS) will be discussed in the context of recent work dealing with eating behavior, with a particular focus on obesity. In the second part of the review, non-invasive strategies to modulate food-related brain processes and functions will be presented. At the leading edge of non-invasive brain-based technologies is real-time fMRI (rtfMRI) neurofeedback, which is a powerful tool to better understand the complexity of human brain-behavior relationships. rtfMRI, alone or when combined with other techniques and tools such as EEG and cognitive therapy, could be used to alter neural plasticity and learned behavior to optimize and/or restore healthy cognition and eating behavior. Other promising non-invasive neuromodulation approaches being explored are repetitive transcranial magnetic stimulation (rTMS) and transcranial direct-current stimulation (tDCS). Converging evidence points at the value of these non-invasive neuromodulation strategies to study basic mechanisms underlying eating behavior and to treat its disorders. Both of these approaches will be compared in light of recent work in this field, while addressing technical and practical questions. The third part of this review will be dedicated to invasive neuromodulation strategies, such as vagus nerve stimulation (VNS) and deep brain stimulation (DBS). In combination with neuroimaging approaches, these techniques are promising experimental tools to unravel the intricate relationships between homeostatic and hedonic brain circuits. Their potential as additional therapeutic tools to combat pharmacorefractory morbid obesity or acute eating disorders will be discussed, in terms of technical challenges, applicability and ethics. In a general discussion, we will put the brain at the core of fundamental research, prevention and therapy in the context of obesity and eating disorders. First, we will discuss the possibility to identify new biological markers of brain functions. Second, we will highlight the potential of neuroimaging and neuromodulation in individualized medicine. Third, we will introduce the ethical questions that are concomitant to the emergence of new neuromodulation therapies.


Assuntos
Encéfalo/fisiopatologia , Terapia por Estimulação Elétrica/métodos , Eletroencefalografia/métodos , Comportamento Alimentar/fisiologia , Transtornos da Alimentação e da Ingestão de Alimentos , Neurorretroalimentação/métodos , Neuroimagem/métodos , Obesidade , Estimulação Magnética Transcraniana/métodos , Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Transtornos da Alimentação e da Ingestão de Alimentos/prevenção & controle , Transtornos da Alimentação e da Ingestão de Alimentos/terapia , Humanos , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Obesidade/terapia
20.
Trends Endocrinol Metab ; 26(3): 125-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25662369

RESUMO

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.


Assuntos
Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Humanos , Peptídeo YY/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA