Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.437
Filtrer
1.
J Clin Invest ; 134(14)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39007271

RÉSUMÉ

The melanocortin-3 receptor (MC3R) regulates GABA release from agouti-related protein (AgRP) nerve terminals and thus tonically suppresses multiple circuits involved in feeding behavior and energy homeostasis. Here, we examined the role of the MC3R and the melanocortin system in regulating the response to various anorexigenic agents. The genetic deletion or pharmacological inhibition of the MC3R, or subthreshold doses of an MC4R agonist, improved the dose responsiveness to glucagon-like peptide 1 (GLP1) agonists, as assayed by inhibition of food intake and weight loss. An enhanced anorectic response to the acute satiety factors peptide YY (PYY3-36) and cholecystokinin (CCK) and the long-term adipostatic factor leptin demonstrated that increased sensitivity to anorectic agents was a generalized result of MC3R antagonism. We observed enhanced neuronal activation in multiple hypothalamic nuclei using Fos IHC following low-dose liraglutide in MC3R-KO mice (Mc3r-/-), supporting the hypothesis that the MC3R is a negative regulator of circuits that control multiple aspects of feeding behavior. The enhanced anorectic response in Mc3r-/- mice after administration of GLP1 analogs was also independent of the incretin effects and malaise induced by GLP1 receptor (GLP1R) analogs, suggesting that MC3R antagonists or MC4R agonists may have value in enhancing the dose-response range of obesity therapeutics.


Sujet(s)
Liraglutide , Récepteur de la mélanocortine de type 3 , Récepteur de la mélanocortine de type 4 , Animaux , Mâle , Souris , Anorexigènes/pharmacologie , Cholécystokinine/métabolisme , Consommation alimentaire/effets des médicaments et des substances chimiques , Glucagon-like peptide 1/métabolisme , Hypothalamus/métabolisme , Leptine/métabolisme , Liraglutide/pharmacologie , Souris de lignée C57BL , Souris knockout , Peptide YY/métabolisme , Peptide YY/génétique , Récepteur de la mélanocortine de type 3/génétique , Récepteur de la mélanocortine de type 3/métabolisme , Récepteur de la mélanocortine de type 3/agonistes , Récepteur de la mélanocortine de type 4/métabolisme , Récepteur de la mélanocortine de type 4/génétique , Récepteur de la mélanocortine de type 4/agonistes
2.
Sci Rep ; 14(1): 14971, 2024 06 28.
Article de Anglais | MEDLINE | ID: mdl-38951515

RÉSUMÉ

Fetal alcohol spectrum disorders (FASD) are a severe developmental condition resulting from exposure to alcohol during pregnancy. The aim of this study was to examine the concentrations of hormones involved in appetite regulation-ghrelin, leptin, and putative peptide YY-3 (PYY)-in the serum of individuals with FASD. Additionally, we investigated the relationship between these hormone levels and clinical indicators. We conducted an enzyme-linked immunosorbent assay on samples collected from 62 FASD patients and 23 individuals without the condition. Our results revealed a significant decrease in leptin levels among FASD patients compared to the control group (5.124 vs. 6.838 ng/mL, p = 0.002). We revealed no statistically significant differences in the levels of other hormones studied (ghrelin and PYY). Comparisons of hormone levels were also conducted in three subgroups: FAS, neurobehavioral disorders associated with prenatal alcohol exposure and FASD risk, as well as by sex. Assignment to FASD subgroups indicated changes only for leptin. Sex had no effect on the levels of hormones. Moreover, the levels of leptin showed a negative correlation with cortisol levels and a positive correlation with BMI and proopiomelanocortin. Alterations in appetite regulation can contribute to the improper development of children with FASD, which might be another factor that should be taken into consideration in the proper treatment of patients.


Sujet(s)
Troubles du spectre de l'alcoolisation foetale , Ghréline , Leptine , Peptide YY , Humains , Leptine/sang , Troubles du spectre de l'alcoolisation foetale/sang , Femelle , Ghréline/sang , Mâle , Peptide YY/sang , Grossesse , Enfant , Adulte , Études cas-témoins , Enfant d'âge préscolaire
3.
Nutrients ; 16(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38999832

RÉSUMÉ

BACKGROUND: The differential effects of pecans versus other popular snack foods on appetite and blood markers of metabolism and satiety have not been well studied. This study investigated the effects of a single mid-morning snack of pecans or tortilla chips on subjective appetite, food intake, blood measures of hormones and metabolites, and resting energy expenditure. METHODS: Twenty participants with overweight and obesity were enrolled in a within-participants, randomized crossover trial. Participants had indwelling catheters placed for blood sampling and were fed a standardized breakfast, followed two hours later by a 250 kcal snack of either pecans or tortilla chips, and then by a self-selected lunch. Visual analog scale (VAS) appetite measures, blood markers, and energy expenditure were taken at intervals after food consumption. RESULTS: VAS ratings, energy, food intake and macronutrient composition did not differ between treatment conditions, but glucose and insulin were significantly more elevated after tortilla chips. Free fatty acids (FFA), triglycerides (TG), peptide YY (PYY), and glucagon-like peptide-1 (GLP-1) were higher after consuming pecans compared to tortilla chips. CONCLUSIONS: Pecan consumption improves postprandial glucose and insulin profiles which would be beneficial to individuals at risk of developing type 2 diabetes. Further studies are needed to investigate whether increased relative secretion of PYY and GLP-1 after eating pecans versus tortilla chips may affect subjective appetite and energy intake if consumed chronically.


Sujet(s)
Appétit , Marqueurs biologiques , Études croisées , Métabolisme énergétique , Insuline , Obésité , Surpoids , Casse-croute , Humains , Mâle , Femelle , Adulte , Obésité/sang , Marqueurs biologiques/sang , Surpoids/sang , Insuline/sang , Glycémie/métabolisme , Glucagon-like peptide 1/sang , Adulte d'âge moyen , Volontaires sains , Consommation alimentaire/physiologie , Ration calorique , Hydrates de carbone alimentaires/administration et posologie , Peptide YY/sang , Période post-prandiale , Jeune adulte
4.
Pharmacol Res Perspect ; 12(4): e1243, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39016695

RÉSUMÉ

Obesity, a global health challenge, necessitates innovative approaches for effective management. Targeting gut peptides in the development of anti-obesity pharmaceuticals has already demonstrated significant efficacy. Ghrelin, peptide YY (PYY), cholecystokinin (CCK), and amylin are crucial in appetite regulation offering promising targets for pharmacological interventions in obesity treatment using both peptide-based and small molecule-based pharmaceuticals. Ghrelin, a sole orexigenic gut peptide, has a potential for anti-obesity therapies through various approaches, including endogenous ghrelin neutralization, ghrelin receptor antagonists, ghrelin O-acyltransferase, and functional inhibitors. Anorexigenic gut peptides, peptide YY, cholecystokinin, and amylin, have exhibited appetite-reducing effects in animal models and humans. Overcoming substantial obstacles is imperative for translating these findings into clinically effective pharmaceuticals. Peptide YY and cholecystokinin analogues, characterized by prolonged half-life and resistance to proteolytic enzymes, present viable options. Positive allosteric modulators emerge as a novel approach for modulating the cholecystokinin pathway. Amylin is currently the most promising, with both amylin analogues and dual amylin and calcitonin receptor agonists (DACRAs) progressing to advanced stages of clinical trials. Despite persistent challenges, innovative pharmaceutical strategies provide a glimpse into the future of anti-obesity therapies.


Sujet(s)
Agents antiobésité , Régulation de l'appétit , Cholécystokinine , Obésité , Humains , Animaux , Obésité/traitement médicamenteux , Agents antiobésité/pharmacologie , Agents antiobésité/usage thérapeutique , Cholécystokinine/métabolisme , Cholécystokinine/pharmacologie , Régulation de l'appétit/effets des médicaments et des substances chimiques , Ghréline/pharmacologie , Ghréline/usage thérapeutique , Polypeptide amyloïde des ilots/métabolisme , Polypeptide amyloïde des ilots/pharmacologie , Peptide YY/pharmacologie , Peptide YY/usage thérapeutique , Anorexigènes/pharmacologie , Anorexigènes/usage thérapeutique
5.
Psychoneuroendocrinology ; 167: 107063, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38896990

RÉSUMÉ

Disruptions in appetite-regulating hormones may contribute to the development and/or maintenance of avoidant/restrictive food intake disorder (ARFID). No study has previously assessed fasting levels of orexigenic ghrelin or anorexigenic peptide YY (PYY), nor their trajectory in response to food intake among youth with ARFID across the weight spectrum. We measured fasting and postprandial (30, 60, 120 minutes post-meal) levels of ghrelin and PYY among 127 males and females with full and subthreshold ARFID (n = 95) and healthy controls (HC; n = 32). We used latent growth curve analyses to examine differences in the trajectories of ghrelin and PYY between ARFID and HC. Fasting levels of ghrelin did not differ in ARFID compared to HC. Among ARFID, ghrelin levels declined more gradually than among HC in the first hour post meal (p =.005), but continued to decline between 60 and 120 minutes post meal, whereas HC plateaued (p =.005). Fasting and PYY trajectory did not differ by group. Findings did not change after adjusting for BMI percentile (M(SD)ARFID = 37(35); M(SD)HC = 53(26); p =.006) or calories consumed during the test meal (M(SD)ARFID = 294(118); M(SD)HC = 384 (48); p <.001). These data highlight a distinct trajectory of ghrelin following a test meal in youth with ARFID. Future research should examine ghrelin dysfunction as an etiological or maintenance factor of ARFID.


Sujet(s)
Trouble de l'alimentation sélective et évitante , Consommation alimentaire , Jeûne , Ghréline , Peptide YY , Période post-prandiale , Humains , Ghréline/sang , Peptide YY/sang , Femelle , Mâle , Adolescent , Période post-prandiale/physiologie , Jeûne/physiologie , Consommation alimentaire/physiologie , Repas/physiologie , Enfant , Indice de masse corporelle , Jeune adulte , Appétit/physiologie
6.
Peptides ; 179: 171256, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38825012

RÉSUMÉ

The approval of the glucagon-like peptide 1 (GLP-1) mimetics semaglutide and liraglutide for management of obesity, independent of type 2 diabetes (T2DM), has initiated a resurgence of interest in gut-hormone derived peptide therapies for the management of metabolic diseases, but side-effect profile is a concern for these medicines. However, the recent approval of tirzepatide for obesity and T2DM, a glucose-dependent insulinotropic polypeptide (GIP), GLP-1 receptor co-agonist peptide therapy, may provide a somewhat more tolerable option. Despite this, an increasing number of non-incretin alternative peptides are in development for obesity, and it stands to reason that other hormones will take to the limelight in the coming years, such as peptides from the neuropeptide Y family. This narrative review outlines the therapeutic promise of the neuropeptide Y family of peptides, comprising of the 36 amino acid polypeptides neuropeptide Y (NPY), peptide tyrosine-tyrosine (PYY) and pancreatic polypeptide (PP), as well as their derivatives. This family of peptides exerts a number of metabolically relevant effects such as appetite regulation and can influence pancreatic beta-cell survival. Although some of these actions still require full translation to the human setting, potential therapeutic application in obesity and type 2 diabetes is conceivable. However, like GLP-1 and GIP, the endogenous NPY, PYY and PP peptide forms are subject to rapid in vivo degradation and inactivation by the serine peptidase, dipeptidyl-peptidase 4 (DPP-4), and hence require structural modification to prolong circulating half-life. Numerous protective modification strategies are discussed in this regard herein, alongside related impact on biological activity profile and therapeutic promise.


Sujet(s)
Diabète de type 2 , Neuropeptide Y , Obésité , Humains , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Obésité/traitement médicamenteux , Obésité/métabolisme , Neuropeptide Y/métabolisme , Animaux , Peptides glucagon-like/usage thérapeutique , Liraglutide/usage thérapeutique , Liraglutide/pharmacologie , Polypeptide pancréatique/métabolisme , Peptide YY/métabolisme , Peptide YY/usage thérapeutique , Peptide gastrointestinal/usage thérapeutique , Peptide gastrointestinal/métabolisme , Glucagon-like peptide 1/métabolisme , Glucagon-like peptide 1/usage thérapeutique , Récepteur du peptide-1 similaire au glucagon/agonistes , Récepteur du peptide-1 similaire au glucagon/métabolisme , Récepteur du peptide-2 similaire au glucagon
7.
Nutr Res ; 127: 27-39, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38843565

RÉSUMÉ

Gut peptides play a role in signaling appetite control in the hypothalamus. Limited knowledge exists regarding the release of these peptides in individuals with obesity before and during external stimuli. We hypothesize that the expression of gut peptides is different in the fasting and postprandial states in the scenario of obesity. PubMed/MEDLINE, Scopus, and Science Direct electronic databases were searched. The meta-analysis was performed using Review Manager Software. Randomized controlled trials that measured gut peptides in both obese and lean subjects were included in the analysis. A total of 552 subjects with obesity were enrolled in 25 trials. The gut peptide profile did not show any significant difference between obese and lean subjects for glucagon-like peptide 1 (95% confidence interval [CI], -1.21 to 0.38; P = .30), peptide YY (95% CI, -1.47 to 0.18; P = .13), and cholecystokinin (95% CI, -1.25 to 1.28; P = .98). Gut peptides are decreased by an increased high-fat, high-carbohydrate diet and by decreased chewing. There is no statistically significant difference in gut peptides between individuals with obesity and leanness in a fasting state. However, the release of gut peptides is affected in individuals with obesity following external stimuli, such as dietary interventions and chewing. Further studies are necessary to investigate the relationship between various stimuli and the release of gut peptides, as well as their impact on appetite regulation in subjects with obesity.


Sujet(s)
Cholécystokinine , Jeûne , Glucagon-like peptide 1 , Obésité , Peptide YY , Période post-prandiale , Humains , Obésité/métabolisme , Peptide YY/sang , Peptide YY/métabolisme , Glucagon-like peptide 1/sang , Glucagon-like peptide 1/métabolisme , Cholécystokinine/métabolisme , Cholécystokinine/sang , Hormones gastrointestinales/métabolisme , Hormones gastrointestinales/sang , Adulte , Essais contrôlés randomisés comme sujet
8.
Sci Transl Med ; 16(752): eadm8132, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896603

RÉSUMÉ

The human ileum contains a high density of enteroendocrine L-cells, which release the appetite-suppressing hormones glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in response to food intake. Recent evidence highlighted the potential role of food structures in PYY release, but the link between food structures, ileal metabolites, and appetite hormone release remains unclear owing to limited access to intact human ileum. In a randomized crossover trial (ISRCTN11327221; isrctn.com), we investigated the role of human ileum in GLP-1 and PYY release by giving healthy volunteers diets differing in fiber and food structure: high-fiber (intact or disrupted food structures) or low-fiber disrupted food structures. We used nasoenteric tubes to sample chyme from the intact distal ileum lumina of humans in the fasted state and every 60 min for 480 min postprandially. We demonstrate the highly dynamic, wide-ranging molecular environment of the ileum over time, with a substantial decrease in ileum bacterial numbers and bacterial metabolites after food intake. We also show that high-fiber diets, independent of food structure, increased PYY release compared with a low-fiber diet during 0 to 240 min postprandially. High-fiber diets also increased ileal stachyose, and a disrupted high-fiber diet increased certain ileal amino acids. Treatment of human ileal organoids with ileal fluids or an amino acid and stachyose mixture stimulated PYY expression in a similar profile to blood PYY concentrations, confirming the role of ileal metabolites in PYY release. Our study demonstrates the diet-induced changes over time in the metabolite environment of intact human ileum, which play a role in PYY release.


Sujet(s)
Régime alimentaire , Iléum , Peptide YY , Humains , Iléum/métabolisme , Peptide YY/métabolisme , Adulte , Mâle , Fibre alimentaire/métabolisme , Glucagon-like peptide 1/métabolisme , Femelle , Métabolome , Période post-prandiale , Études croisées , Jeune adulte
9.
Anat Histol Embryol ; 53(4): e13074, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38864153

RÉSUMÉ

In this study, we investigated amylin-like substance distribution in the pancreas of Japanese quail (Coturnix japonica) using a specific anti-rat amylin serum. We detected amylin-immunoreactive cells dispersed in the pancreatic extra-islet region but not in the islet region. The synthetic rat amylin-containing serum pre-absorption abolished the staining profile. Almost all amylin-immunoreactive cells were immuno-positive for peptide YY (PYY). In addition, certain amylin-immunoreactive cells stained immuno-positive for glucagon. Amylin and PYY co-secreted from the extra-islet cells might participate in the insulin and glucagon release regulation in the pancreas and food intake modulation through the central nervous system.


Sujet(s)
Coturnix , Glucagon , Polypeptide amyloïde des ilots , Pancréas , Peptide YY , Animaux , Peptide YY/métabolisme , Polypeptide amyloïde des ilots/métabolisme , Coturnix/métabolisme , Glucagon/métabolisme , Pancréas/métabolisme , Immunohistochimie/médecine vétérinaire , Ilots pancréatiques/métabolisme , Mâle , Rats
10.
Nutrients ; 16(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892651

RÉSUMÉ

The consumption of protein-rich foods stimulates satiety more than other macronutrient-rich foods; however, the underlying mechanisms-of-action are not well-characterized. The objective of this study was to identify the direct and indirect effects of postprandial amino acid (AA) responses on satiety. Seventeen women (mean ± SEM, age: 33 ± 1 year; BMI: 27.8 ± 0.1 kg/m2) consumed a eucaloric, plant-based diet containing two servings of lean beef/day (i.e., 7.5 oz (207 g)) for 7 days. During day 6, the participants completed a 12 h controlled-feeding, clinical testing day including repeated satiety questionnaires and blood sampling to assess pre- and postprandial plasma AAs, PYY, and GLP-1. Regression and mediation analyses were completed to assess AA predictors and hormonal mediators. Total plasma AAs explained 41.1% of the variance in perceived daily fullness (p < 0.001), 61.0% in PYY (p < 0.001), and 66.1% in GLP-1 (p < 0.001) concentrations, respectively. Several individual AAs significantly predicted fluctuations in daily fullness, PYY, and GLP-1. In completing mediation analyses, the effect of plasma leucine on daily fullness was fully mediated by circulating PYY concentrations (indirect effect = B: 0.09 [Boot 95% CI: 0.032, 0.17]) as no leucine-fullness direct effect was observed. No other mediators were identified. Although a number of circulating AAs predict satiety, leucine was found to do so through changes in PYY concentrations in middle-aged women.


Sujet(s)
Acides aminés , Surpoids , Peptide YY , Période post-prandiale , Viande rouge , Satiété , Humains , Femelle , Adulte , Acides aminés/sang , Peptide YY/sang , Satiété/effets des médicaments et des substances chimiques , Surpoids/sang , Glucagon-like peptide 1/sang , Marqueurs biologiques/sang , Repas , Animaux , Bovins , Sensation de satiété/effets des médicaments et des substances chimiques
11.
Appetite ; 200: 107509, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38795943

RÉSUMÉ

Glycomacropeptide (GMP) has a unique amino acid profile which may make less satiating than other dietary proteins. This study assessed the feasibility and likely acceptability of a leucine-enriched GMP drink and determined appetite response in older adults (OA). Thirteen OA (11f; 70 ± 4 years) were recruited for sensory assessments of a leucine-enriched GMP drink when mixed with water and with fruit smoothie, compared with whey protein isolate (WHEY). Participants also partook in a single focus group exploring acceptability to protein and supplementation. Separately, a counterbalanced, double-blind study with twelve OA (8f; 69 ± 3 years) was conducted to determine appetite and gut hormone responses. Fasting subjective appetite was recorded using visual analogue scales and a fasted venous blood sample was collected (to measures acyl-ghrelin, PYY, GLP-1, and CCK) before participants consumed either: GMP protein (27g + 3g leucine, 350 mL water), WHEY (30g, 350 mL water), or water. Participants rested for 240 min, with appetite measures and blood sampling throughout. An ad libitum pasta-based meal was then consumed. Sensory testing revealed low pleasantness rating for GMP in water vs. WHEY (16 ± 14 vs 31 ± 24, p = 0.016). GMP addition to smoothie reduced pleasantness (26 ± 21 vs. 61 ± 29, p = 0.009) and worsened the aroma (46 ± 15 vs. 69 ± 28, p = 0.014). The focus group revealed uncertainty of protein needs and a scepticism of supplements, with preference for food. Gut hormone response did not differ between GMP and WHEY (nAUC for all gut hormones p > 0.05). There was no difference between conditions for lunch ad libitum intake (549 ± 171 kcal, 512 ± 238 kcal, 460 ± 199 kcal for GMP, WHEY, and water, p = 0.175), or for subjective appetite response. Leucine-enriched GMP was not less satiating than WHEY, and low palatability and scepticism of supplements question the likely acceptability of GMP supplementation. Providing trusted nutritional advice and food enrichment/fortification may be preferred strategies for increasing protein intake in OA.


Sujet(s)
Appétit , Caséines , Études de faisabilité , Hormones gastrointestinales , Fragments peptidiques , Protéines de lactosérum , Humains , Femelle , Mâle , Appétit/effets des médicaments et des substances chimiques , Sujet âgé , Projets pilotes , Hormones gastrointestinales/sang , Méthode en double aveugle , Caséines/administration et posologie , Caséines/pharmacologie , Protéines de lactosérum/administration et posologie , Protéines de lactosérum/pharmacologie , Fragments peptidiques/sang , Leucine/administration et posologie , Leucine/pharmacologie , Ghréline/sang , Satiété/effets des médicaments et des substances chimiques , Consommation alimentaire , Compléments alimentaires , Adulte d'âge moyen , Peptide YY/sang , Glucagon-like peptide 1/sang , Protéines alimentaires/administration et posologie
12.
Article de Anglais | MEDLINE | ID: mdl-38703991

RÉSUMÉ

The pond loach (Misgurnus anguillicaudatus) is an important aquaculture freshwater species, used as an ornamental fish, food source for humans and angling bait. Pond loaches are resistant to fasting and extreme environmental conditions, including temperature and low oxygen levels. Little is known about how these factors affect the feeding physiology and the endocrine regulation of feeding of loaches. In this study, we examined the effects of fasting, as well as increased temperature and decreased oxygen levels on food intake and transcript levels of appetite regulators. Fasted fish had lower blood glucose levels, and lower expression levels of intestine CCK and PYY, and brain CART1, but had higher levels of brain orexin and ghrelin than fed fish. Fish held at 30 °C had higher food intake, glucose levels, and mRNA levels of intestine CCK and PYY, and brain CART2, but lower brain orexin levels than fish at 20 °C. Fish held at low oxygen levels had a lower food intake, higher intestine CCKa and ghrelin, and brain orexin, CART2 and ghrelin mRNA expression levels than fish held at high O2 levels. Our results suggest that fasting and high temperatures increase the expression of orexigenic and anorexigenic factors respectively, whereas the increase in expression of both orexigenic and anorexigenic factors in low O2 environments might not be related to their role in feeding, but possibly to protection from tissue damage. The results of our study might shed new light on how pond loaches are able to cope with extreme environmental conditions such as low food availability, extreme temperatures and hypoxia.


Sujet(s)
Cypriniformes , Jeûne , Ghréline , Animaux , Jeûne/physiologie , Cypriniformes/physiologie , Cypriniformes/génétique , Cypriniformes/métabolisme , Ghréline/métabolisme , Orexines/métabolisme , Encéphale/métabolisme , Encéphale/physiologie , Cholécystokinine/métabolisme , Régulation de l'appétit/physiologie , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Glycémie/métabolisme , Oxygène/métabolisme , Peptide YY/métabolisme , Peptide YY/sang , Consommation alimentaire/physiologie , Température , Comportement alimentaire/physiologie
13.
Neuropeptides ; 105: 102427, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38579490

RÉSUMÉ

Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.


Sujet(s)
Glucagon-like peptide 1 , Peptide YY , Humains , Animaux , Peptide YY/métabolisme , Peptide YY/pharmacologie , Glucagon-like peptide 1/métabolisme , Anxiété/traitement médicamenteux , Anxiété/métabolisme , Fragments peptidiques/pharmacologie , Comportement de recherche de substances/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Obésité/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme
14.
Nat Commun ; 15(1): 3514, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664401

RÉSUMÉ

Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.


Sujet(s)
Consommation alimentaire , Cellules entéroendocrines , Acide glutamique , Neuropeptides , Peptide YY , Animaux , Cellules entéroendocrines/métabolisme , Femelle , Neuropeptides/métabolisme , Neuropeptides/génétique , Consommation alimentaire/physiologie , Peptide YY/métabolisme , Acide glutamique/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Drosophila melanogaster/métabolisme , Comportement alimentaire/physiologie , Récepteurs métabotropes au glutamate/métabolisme , Neurones dopaminergiques/métabolisme , Régime alimentaire
15.
Environ Res ; 252(Pt 1): 118846, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38582428

RÉSUMÉ

BACKGROUND: Appetite hormones are considered a promising target in fighting obesity as impaired appetite hormone levels have already been associated with obesity. However, further insights in the drivers of appetite hormone levels are needed. OBJECTIVES: In this study, we investigated the associations of fasting appetite hormone levels with lifestyle and environmental exposures in children and adolescents. METHODS: A total of 534 fasting blood samples were collected from children and adolescents (4-16y,50% boys) and appetite hormone levels (glucagon-like peptide-1 (GLP-1), peptide YY (PYY), pancreatic polypeptide (PP), leptin and ghrelin) were measured. Exposures included dietary quality (fiber-rich food intake, sugar propensity, fat propensity), psychosocial stress (happiness, negative emotions, negative life events and emotional problems), sleep duration, physical activity and environmental quality (long term black carbon (BC), particulate matter <2.5 µM (PM2.5), nitrogen dioxide (NO2) exposure, and green space in a 100 m and 2000 m radius around the residence). A multi-exposure score was calculated to combine all the exposures at study in one measure. Associations of individual exposures and multi-exposure score with appetite hormone levels were evaluated using linear mixed regression models adjusting for sex, age, socioeconomic status, waist-to-height ratio and multiple testing. RESULTS: GLP-1 was associated with air pollution exposure (NO2 ß* = -0.13, BC ß* = -0.15, PM2.5 ß* = -0.16, all p < 0.001). Leptin was associated with green space in a 100 m radius around the residence (ß* = -0.11; p = 0.002). Ghrelin was associated with negative emotions (active ghrelin ß* = -0.16; p = 0.04, total ghrelin ß* = -0.23; p = 0.0051) and happiness (active ghrelin ß* = 0.25; p < 0.001, total ghrelin ß* = 0.26; p < 0.001). Furthermore, total ghrelin levels were associated with the multi-exposure score, reflecting unhealthy exposures and lifestyle (ß* = -0.22; p = 0.036). DISCUSSION: Our findings provide new insights into the associations of exposures with appetite hormone levels, which are of high interest for preventive obesity research. Further research is crucial to reveal the underlying mechanisms of the observed associations.


Sujet(s)
Exposition environnementale , Mode de vie , Humains , Enfant , Mâle , Femelle , Adolescent , Enfant d'âge préscolaire , Ghréline/sang , Glucagon-like peptide 1/sang , Appétit , Leptine/sang , Peptide YY/sang
16.
Appetite ; 198: 107362, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38636667

RÉSUMÉ

This was a preliminary study that examined whether appetite regulation is altered during the menstrual cycle or with oral contraceptives. Ten naturally cycling females (NON-USERS) and nine tri-phasic oral contraceptive using females (USERS) completed experimental sessions during each menstrual phase (follicular phase: FP; ovulatory phase: OP; luteal phase: LP). Appetite perceptions and blood samples were obtained fasted, 30, 60, and 90 min post-prandial to measure acylated ghrelin, active glucagon-like peptide-1 (GLP-1), and total peptide tyrosine tyrosine (PYY). Changes were considered important if p < 0.100 and the effect size was ≥medium. There appeared to be a three-way (group x phase x time) interaction for acylated ghrelin where concentrations appeared to be greater in USERS versus NON-USERS during the OP 90-min post-prandial and during the LP fasted, and 90-min post-prandial. In USERS, ghrelin appeared to be greater 90-min post-prandial in the OP versus the FP with no other apparent differences between phases. There were no apparent differences between phases in NON-USERS. There appeared to be a three-way interaction for PYY where concentrations appeared to be greater in USERS during the FP 60-min post-prandial and during the OP 30-min post-prandial. In USERS PYY appeared to be greater 60-min post-prandial during the OP versus the LP with no other apparent differences. There were no apparent differences between phases in NON-USERS. There appeared to be no effect of group or phase on GLP-1, or appetite perceptions. These data demonstrate small effects of menstrual cycle phase and oral contraceptive use on the acylated ghrelin and total PYY response to a standardized meal, with no effects on active GLP-1 or perceived appetite, though more work with a large sample size is necessary.


Sujet(s)
Ghréline , Glucagon-like peptide 1 , Cycle menstruel , Peptide YY , Période post-prandiale , Humains , Femelle , Ghréline/sang , Glucagon-like peptide 1/sang , Peptide YY/sang , Jeune adulte , Adulte , Contraceptifs oraux/administration et posologie , Contraceptifs oraux/pharmacologie , Appétit , Régulation de l'appétit/physiologie , Adolescent , Jeûne , Acylation
17.
Rev Assoc Med Bras (1992) ; 70(1): e20230263, 2024.
Article de Anglais | MEDLINE | ID: mdl-38511748

RÉSUMÉ

OBJECTIVE: Diet and exercise, which are the building blocks of obesity management, provide weight loss by creating a negative energy balance. However, the effect of energy deficit induced by long-term diet and exercise on appetite hormones remains unclear. The study was designed to determine the effect of a 12-week diet and exercise program applied to obese individuals on the levels of appetite hormones, namely, ghrelin, GLP-1, and PYY. METHODS: A total of 62 obese individuals (BMI≥30) and 48 healthy controls (BMI 18.50-29.99) participated in the study. Appropriate diet (1000-1500 kcal/day) and exercise (at least 5000 steps/day) programs were applied to obese individuals according to age, gender, and BMI. The ghrelin, GLP-1, and PYY values of the participants were analyzed by the ELISA method and commercial kit by taking venous blood samples before and after 12 weeks of treatment. RESULTS: While ghrelin levels of individuals decreased significantly after diet and exercise, PYY levels increased significantly. However, despite the treatment applied, the GLP-1 and PYY levels of the case group did not reach the levels of the control group. CONCLUSION: Long-term diet and exercise intervention had a positive effect on appetite regulation hormones. It reduced ghrelin levels after treatment. Associated weight loss was facilitated. In the case group, increased satiety hormones after combined treatment supported the maintenance of body weight by increasing satiety.


Sujet(s)
Ghréline , Glucagon-like peptide 1 , Humains , Peptide YY , Obésité/thérapie , Perte de poids/physiologie , Régime alimentaire
18.
Peptides ; 176: 171186, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38490484

RÉSUMÉ

Circulating insulin levels are known to be increased in people with higher body mass index (BMI) due to effects of adiposity on insulin resistance, whilst gut hormones have a more complex relationship, with fasting peptideYY (PYY) reported to be inversely related to BMI. This study aimed to further explore fasting and post prandial pancreatic and gut hormone concentrations in plasma samples from obese and non-obese participants. Participants with healthy BMI (n=15), overweight BMI (n=29) and obesity (n=161) had samples taken fasting and 30 min post mixed liquid meal for analysis of glucagon-like peptide-1 (GLP-1), PYY, glucose-dependent insulinotropic polypeptide (GIP), insulin and glucagon. Data visualiation used linear discriminant analysis for dimensionality reduction, to visualise the data and assess scaling of each hormone. Fasting levels of insulin, GIP and PYY were shown to be key classifiers between the 3 groups on ANCOVA analysis, with an observation of increased GIP levels in overweight, but not obese participants. In non-obese subjects, fasting GIP, PYY and insulin correlated with BMI, whereas in subjects with obesity only the pancreatic hormones glucagon and insulin correlated with BMI. Concentrations of total GLP-1 in the fasting state correlated strongly with glucagon levels, highlighting potential assay cross-reactivities. The study, which included a relatively large number of subjects with severe obesity, supported previous evidence of BMI correlating negatively with fasting PYY and positively with fasting insulin. The observation of increased fasting GIP levels in overweight but not obese participants deserves further validation and mechanistic investigation.


Sujet(s)
Indice de masse corporelle , Jeûne , Peptide gastrointestinal , Glucagon-like peptide 1 , Insuline , Obésité , Peptide YY , Humains , Obésité/sang , Mâle , Femelle , Adulte , Jeûne/sang , Peptide YY/sang , Adulte d'âge moyen , Glucagon-like peptide 1/sang , Peptide gastrointestinal/sang , Insuline/sang , Période post-prandiale , Glucagon/sang , Hormones gastrointestinales/sang
19.
Nutr Diabetes ; 14(1): 9, 2024 03 06.
Article de Anglais | MEDLINE | ID: mdl-38448413

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Large intestinal fermentation of dietary fiber may control meal-related glycemia and appetite via the production of short-chain fatty acids (SCFA) and the secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). We investigated whether this mechanism contributes to the efficacy of the Roux-en-Y gastric bypass (RYGB) by assessing the effect of oligofructose-enriched inulin (inulin) vs. maltodextrin (MDX) on breath hydrogen (a marker of intestinal fermentation), plasma SCFAs, gut hormones, insulin and blood glucose concentrations as well as appetite in RYGB patients. METHOD: Eight RYGB patients were studied on two occasions before and ~8 months after surgery using a cross-over design. Each patient received 300 ml orange juice containing 25 g inulin or an equicaloric load of 15.5 g MDX after an overnight fast followed by a fixed portion snack served 3 h postprandially. Blood samples were collected over 5 h and breath hydrogen measured as well as appetite assessed using visual analog scales. RESULTS: Surgery increased postprandial secretion of GLP-1 and PYY (P ≤ 0.05); lowered blood glucose and plasma insulin increments (P ≤ 0.05) and reduced appetite ratings in response to both inulin and MDX. The effect of inulin on breath hydrogen was accelerated after surgery with an increase that was earlier in onset (2.5 h vs. 3 h, P ≤ 0.05), but less pronounced in magnitude. There was, however, no effect of inulin on plasma SCFAs or plasma GLP-1 and PYY after the snack at 3 h, neither before nor after surgery. Interestingly, inulin appeared to further potentiate the early-phase glucose-lowering and second-meal (3-5 h) appetite-suppressive effect of surgery with the latter showing a strong correlation with early-phase breath hydrogen concentrations. CONCLUSION: RYGB surgery accelerates large intestinal fermentation of inulin, however, without measurable effects on plasma SCFAs or plasma GLP-1 and PYY. The glucose-lowering and appetite-suppressive effects of surgery appear to be potentiated with inulin.


Sujet(s)
Dérivation gastrique , Insulines , Humains , Inuline/pharmacologie , Appétit , Projets pilotes , Glycémie , Études croisées , Études prospectives , Peptide YY , Glucagon-like peptide 1 , Perception
20.
Mol Metab ; 83: 101918, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38499083

RÉSUMÉ

OBJECTIVE: In the small intestine, the products of digestion of dietary triacylglycerol (TAG), fatty acids (FA) and monoacylglycerol, are taken up by absorptive cells, enterocytes, for systemic energy delivery. These digestion products can also bind receptors on endocrine cells to stimulate the release of hormones capable of influencing systemic energy metabolism. The initial phase of intestinal FA absorption involves the acylation of FAs to acyl-CoA by the acyl-CoA long chain synthetase (ACSL) enzymes. ACSL5 is abundantly expressed in the small intestinal epithelium where it is the major ACSL isoform, contributing approximately 80% of total ACSL activity. In mice with whole body deficiency of ACSL5, the rate of dietary fat absorption is reduced and energy expenditure is increased. However, the mechanisms by which intestinal ACSL5 contributes to intestinal FA metabolism, enteroendocrine signaling, and regulation of energy expenditure remain undefined. Here, we test the hypothesis that intestinal ACSL5 regulates energy metabolism by influencing dietary fat absorption and enteroendocrine signaling. METHODS: To explore the role of intestinal ACSL5 in energy balance and intestinal dietary fat absorption, a novel mouse model of intestine specific ACSL5 deficiency (ACSL5IKO) was generated by breeding ACSL5 floxed (ACSL5loxP/loxP) to mice harboring the tamoxifen inducible, villin-Cre recombinase. ACSL5IKO and control, ACSL5loxP/loxP mice were fed chow (low in fat) or a 60% high fat diet (HFD), and metabolic phenotyping was performed including, body weight, body composition, insulin and glucose tolerance tests, energy expenditure, physical activity, and food intake studies. Pair-feeding studies were performed to determine the role of food intake in regulating development of obesity. Studies of dietary fat absorption, fecal lipid excretion, intestinal mucosal FA content, and circulating levels of glucagon like peptide 1 (GLP-1) and peptide YY (PYY) in response to a TAG challenge were performed. Treatment with a GLP-1 receptor antagonist was performed to determine the contribution of GLP-1 to acute regulation of food intake. RESULTS: We found that ACSL5IKO mice experienced rapid and sustained protection from body weight and fat mass accumulation during HFD feeding. While intestine specific deficiency of ACSL5 delayed gastric emptying and reduced dietary fat secretion, it did not result in increased excretion of dietary lipid in feces. Energy expenditure and physical activity were not increased in ACSL5IKO mice. Mice deficient in intestinal ACSL5 display significantly reduced energy intake during HFD, but not chow feeding. When HFD intake of control mice was matched to ACSL5IKO during pair-feeding studies, no differences in body weight or fat mass gain were observed between groups. Postprandial GLP-1 and PYY were significantly elevated in ACSL5IKO mice secondary to increased FA content in the distal small intestine. Blockade of GLP-1 signaling by administration of a long-acting GLP-1 receptor antagonist partially restored HFD intake of ACSL5IKO. CONCLUSIONS: These data indicate that intestinal ACSL5 serves as a critical regulator of energy balance, protecting mice from diet-induced obesity exclusively by increasing satiety and reducing food intake during HFD feeding. The reduction in food intake observed in ACSL5IKO mice is driven, in part, by increased postprandial GLP-1 and PYY secretion. These effects are only observed during HFD feeding, suggesting that altered processing of dietary fat following intestinal ACSL5 ablation contributes to GLP-1 and PYY mediated increases in satiety.


Sujet(s)
Coenzyme A ligases , Alimentation riche en graisse , Glucagon-like peptide 1 , Obésité , Peptide YY , Animaux , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Souris , Obésité/métabolisme , Mâle , Glucagon-like peptide 1/métabolisme , Peptide YY/métabolisme , Souris de lignée C57BL , Consommation alimentaire , Période post-prandiale , Métabolisme énergétique , Souris knockout
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE