Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 639
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 97(1): 411-463, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003328

RESUMO

The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.


Assuntos
Colecistocinina/metabolismo , Derivação Gástrica , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Glicemia/metabolismo , Ingestão de Alimentos/fisiologia , Humanos , Obesidade/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-38703991

RESUMO

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.


Assuntos
Cipriniformes , Jejum , Grelina , Animais , Jejum/fisiologia , Cipriniformes/fisiologia , Cipriniformes/genética , Cipriniformes/metabolismo , Grelina/metabolismo , Orexinas/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiologia , Colecistocinina/metabolismo , Regulação do Apetite/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Glicemia/metabolismo , Oxigênio/metabolismo , Peptídeo YY/metabolismo , Peptídeo YY/sangue , Ingestão de Alimentos/fisiologia , Temperatura , Comportamento Alimentar/fisiologia
3.
J Physiol ; 601(5): 979-1016, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36661095

RESUMO

The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) are subcortical structures involved in entrainment of the brain's circadian system to photic and non-photic (e.g. metabolic and arousal) cues. Both receive information about environmental light from photoreceptors, exhibit infra-slow oscillations (ISO) in vivo, and connect to the master circadian clock. Although current evidence demonstrates that the IGL/VLG communicate metabolic information and are crucial for entrainment of circadian rhythms to time-restricted feeding, their sensitivity to food intake-related peptides has not been investigated yet. We examined the effect of metabolically relevant peptides on the spontaneous activity of IGL/VLG neurons. Using ex vivo and in vivo electrophysiological recordings as well as in situ hybridisation, we tested potential sensitivity of the IGL/VLG to anorexigenic and orexigenic peptides, such as cholecystokinin, glucagon-like peptide 1, oxyntomodulin, peptide YY, orexin A and ghrelin. We explored neuronal responses to these drugs during day and night, and in standard vs. high-fat diet conditions. We found that IGL/VLG neurons responded to all the substances tested, except peptide YY. Moreover, more neurons responded to anorexigenic drugs at night, while a high-fat diet affected the IGL/VLG sensitivity to orexigenic peptides. Interestingly, ISO neurons responded to light and orexin A, but did not respond to the other food intake-related peptides. In contrast, non-ISO cells were activated by metabolic peptides, with only some being responsive to light. Our results show for the first time that peptides involved in the body's energy homeostasis stimulate the thalamus and suggest functional separation of the IGL/VLG cells. KEY POINTS: The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) of the rodent thalamus process various signals and participate in circadian entrainment. In both structures, cells exhibiting infra-slow oscillatory activity as well as non-rhythmically firing neurons being observed. Here, we reveal that only one of these two groups of cells responds to anorexigenic (cholecystokinin, glucagon-like peptide 1 and oxyntomodulin) and orexigenic (ghrelin and orexin A) peptides. Neuronal responses vary depending on the time of day (day vs. night) and on the diet (standard vs. high-fat diet). Additionally, we visualised receptors to the tested peptides in the IGL/VLG using in situ hybridisation. Our results suggest that two electrophysiologically different subpopulations of IGL/VLG neurons are involved in two separate functions: one related to the body's energy homeostasis and one associated with the subcortical visual system.


Assuntos
Corpos Geniculados , Grelina , Colecistocinina/metabolismo , Ritmo Circadiano/fisiologia , Sinais (Psicologia) , Dieta Hiperlipídica , Corpos Geniculados/fisiologia , Grelina/metabolismo , Orexinas/metabolismo , Oxintomodulina/metabolismo , Peptídeo YY/metabolismo , Núcleo Supraquiasmático/metabolismo
4.
J Physiol ; 601(16): 3461-3480, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37269207

RESUMO

An understanding of the metabolic determinants of postexercise appetite regulation would facilitate development of adjunctive therapeutics to suppress compensatory eating behaviours and improve the efficacy of exercise as a weight-loss treatment. Metabolic responses to acute exercise are, however, dependent on pre-exercise nutritional practices, including carbohydrate intake. We therefore aimed to determine the interactive effects of dietary carbohydrate and exercise on plasma hormonal and metabolite responses and explore mediators of exercise-induced changes in appetite regulation across nutritional states. In this randomized crossover study, participants completed four 120 min visits: (i) control (water) followed by rest; (ii) control followed by exercise (30 min at ∼75% of maximal oxygen uptake); (iii) carbohydrate (75 g maltodextrin) followed by rest; and (iv) carbohydrate followed by exercise. An ad libitum meal was provided at the end of each 120 min visit, with blood sample collection and appetite assessment performed at predefined intervals. We found that dietary carbohydrate and exercise exerted independent effects on the hormones glucagon-like peptide 1 (carbohydrate, 16.8 pmol/L; exercise, 7.4 pmol/L), ghrelin (carbohydrate, -48.8 pmol/L; exercise: -22.7 pmol/L) and glucagon (carbohydrate, 9.8 ng/L; exercise, 8.2 ng/L) that were linked to the generation of distinct plasma 1 H nuclear magnetic resonance metabolic phenotypes. These metabolic responses were associated with changes in appetite and energy intake, and plasma acetate and succinate were subsequently identified as potential novel mediators of exercise-induced appetite and energy intake responses. In summary, dietary carbohydrate and exercise independently influence gastrointestinal hormones associated with appetite regulation. Future work is warranted to probe the mechanistic importance of plasma acetate and succinate in postexercise appetite regulation. KEY POINTS: Carbohydrate and exercise independently influence key appetite-regulating hormones. Temporal changes in postexercise appetite are linked to acetate, lactate and peptide YY. Postexercise energy intake is associated with glucagon-like peptide 1 and succinate levels.


Assuntos
Regulação do Apetite , Carboidratos da Dieta , Masculino , Apetite/fisiologia , Regulação do Apetite/fisiologia , Estudos Cross-Over , Ingestão de Energia/fisiologia , Exercício Físico/fisiologia , Grelina/metabolismo , Grelina/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Insulina/farmacologia , Peptídeo YY/metabolismo , Peptídeo YY/farmacologia , Succinatos/farmacologia , Humanos
5.
Diabetes Obes Metab ; 25(6): 1731-1739, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36811311

RESUMO

AIMS: To investigate whether the elevation in postprandial concentrations of the gut hormones glucagon-like peptide-1 (GLP-1), oxyntomodulin (OXM) and peptide YY (PYY) accounts for the beneficial changes in food preferences, sweet taste function and eating behaviour after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS: This was a secondary analysis of a randomized single-blind study in which we infused GLP-1, OXM, PYY (GOP) or 0.9% saline subcutaneously for 4 weeks in 24 subjects with obesity and prediabetes/diabetes, to replicate their peak postprandial concentrations, as measured at 1 month in a matched RYGB cohort (ClinicalTrials.gov NCT01945840). A 4-day food diary and validated eating behaviour questionnaires were completed. Sweet taste detection was measured using the method of constant stimuli. Correct sucrose identification (corrected hit rates) was recorded, and sweet taste detection thresholds (EC50s: half maximum effective concencration values) were derived from concentration curves. The intensity and consummatory reward value of sweet taste were assessed using the generalized Labelled Magnitude Scale. RESULTS: Mean daily energy intake was reduced by 27% with GOP but no significant changes in food preferences were observed, whereas a reduction in fat and increase in protein intake were seen post-RYGB. There was no change in corrected hit rates or detection thresholds for sucrose detection following GOP infusion. Additionally, GOP did not alter the intensity or consummatory reward value of sweet taste. A significant reduction in restraint eating, comparable to the RYGB group was observed with GOP. CONCLUSION: The elevation in plasma GOP concentrations after RYGB is unlikely to mediate changes in food preferences and sweet taste function after surgery but may promote restraint eating.


Assuntos
Derivação Gástrica , Hormônios Gastrointestinais , Estado Pré-Diabético , Humanos , Paladar , Preferências Alimentares , Método Simples-Cego , Estado Pré-Diabético/complicações , Obesidade/complicações , Obesidade/cirurgia , Derivação Gástrica/efeitos adversos , Derivação Gástrica/métodos , Peptídeo YY/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Sacarose , Voluntários
6.
Appetite ; 184: 106520, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36868314

RESUMO

Energy intake in the post-exercise state is highly variable and compensatory eating - i.e., (over-) compensation of the expended energy via increased post-exercise energy intake - occurs in some individuals but not others. We aimed to identify predictors of post-exercise energy intake and compensation. In a randomized crossover design, 57 healthy participants (21.7 [SD = 2.5] years; 23.7 [SD = 2.3] kg/m2, 75% White, 54% female) completed two laboratory-based test-meals following (1) 45-min exercise and (2) 45-min rest (control). We assessed associations between biological (sex, body composition, appetite hormones) and behavioral (habitual exercise via prospective exercise log, eating behavior traits) characteristics at baseline and total energy intake, relative energy intake (intake - exercise expenditure), and the difference between post-exercise and post-rest intake. We found a differential impact of biological and behavioral characteristics on total post-exercise energy intake in men and women. In men, only fasting (baseline) concentrations of appetite-regulating hormones (peptide YY [PYY, ß = 0.88, P < 0.001] and adiponectin [ß = 0.66, P = 0.005] predicted total post-exercise energy intake, while in women, only habitual exercise (ß = -0.44, P = 0.017) predicted total post-exercise energy intake. Predictors of relative intake were almost identical to those of total intake. The difference in energy intake between exercise and rest was associated with VO2peak (ß = -0.45, P = 0.020), fasting PYY (ß = 0.53, P = 0.036), and fasting adiponectin (ß = 0.57, P = 0.021) in men but not women (all P > 0.51). Our results show that biological and behavioral characteristics differentially affect total and relative post-exercise energy intake in men and women. This may help identify individuals who are more likely to compensate for the energy expended in exercise. Targeted countermeasures to prevent compensatory energy intake after exercise should take the demonstrated sex differences into account.


Assuntos
Adiponectina , Ingestão de Energia , Humanos , Feminino , Masculino , Estudos Prospectivos , Ingestão de Energia/fisiologia , Apetite/fisiologia , Exercício Físico/fisiologia , Peptídeo YY/metabolismo , Metabolismo Energético/fisiologia , Grelina/metabolismo
7.
Appetite ; 182: 106426, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36539160

RESUMO

Appetite is a determinant of dietary intake and is impacted by sex hormones, exercise, and body composition among individuals without chronic conditions. Whether appetite is altered by exercise in the context of estrogen suppression and cancer survivorship is unknown. This randomized cross-over study compared appetite and ad libitum energy intake (EI) after acute resistance exercise (REx) versus sedentary (SED) conditions and in relation to body composition and resting metabolic rate (RMR) in breast cancer survivors (BCS). Physically inactive premenopausal females with previous stage I-III estrogen receptor-positive breast cancer completed a single bout of REx or SED 35 minutes after a standardized breakfast meal. Appetite visual analog scales and hormones (total ghrelin and peptide-YY [PYY]) were measured before and 30, 90, 120, 150, and 180 minutes post-meal and expressed as area under the curve (AUC). Participants were offered a buffet-type meal 180 minutes after breakfast to assess ad libitum EI. Body composition (dual X-ray absorptiometry) and RMR (indirect calorimetry) were measured during a separate visit. Sixteen BCS were included (age: 46 ± 2 y, BMI: 24.9 ± 1.0 kg/m2). There were no differences in appetite ratings or EI between conditions. There were no differences in appetite hormone AUC, but REx resulted in lower ghrelin 120 (-85 ± 39 pg/mL, p = 0.031) and 180 (-114 ± 43 pg/mL, p = 0.018) minutes post-breakfast and higher PYY 90 (21 ± 10 pg/mL, p = 0.028) and 120 (14 ± 7 pg/mL, p = 0.041) minutes post-breakfast. Fat-free mass and RMR negatively correlated with hunger and prospective food consumption AUC after SED, but not REx. In sum, a single REx bout temporarily reduces orexigenic and increases anorexic appetite hormones, but not acute subjective appetite sensations or EI.


Assuntos
Neoplasias da Mama , Sobreviventes de Câncer , Treinamento Resistido , Feminino , Humanos , Adulto , Pessoa de Meia-Idade , Apetite , Grelina/metabolismo , Ingestão de Energia , Peptídeo YY/metabolismo , Sensação , Estudos Cross-Over
8.
Appetite ; 186: 106557, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37044176

RESUMO

The interaction of exercise with appetite control and energy intake has been widely studied due to the ability of exercise-related energy expenditure to influence energy and substrate balance. Many empirical studies have explored appetite and energy intake responses to acute (single) exercise bouts involving a variety of protocols in diverse populations revealing several consistent trends. The balance of evidence suggests that acute moderate-to-vigorous intensity land-based exercise suppresses subjective appetite feelings and the orexigenic hormone acylated ghrelin and elevates the anorexigenic hormones peptide YY and glucagon-like peptide-1. These perturbations are transient and hormone concentrations usually return to resting values in the hours after exercise without evoking compensatory increases in appetite or energy intake on the same day. This evidence counters the popular assertion that exercise transiently increases appetite and may prompt greater energy intake at subsequent meals. The indifference of the appetite control system to acute exercise-induced energy deficits contrasts with the immediate increases in appetite and energy intake provoked by equivalent diet-induced energy deficits. There is, however, considerable inter-individual variability in subjective appetite and hormonal responses to acute exercise with some individuals experiencing greater exercise-induced appetite suppression than others. Current evidence supports the promotion of exercise as a strategy for inducing a short-term energy deficit but the relevance of this for long-term appetite regulation and the control of body mass remains uncertain.


Assuntos
Regulação do Apetite , Apetite , Humanos , Apetite/fisiologia , Regulação do Apetite/fisiologia , Grelina/metabolismo , Exercício Físico/fisiologia , Ingestão de Energia/fisiologia , Peptídeo YY/metabolismo , Metabolismo Energético/fisiologia
9.
Gastroenterology ; 161(2): 536-547.e2, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33848536

RESUMO

OBJECTIVE: Hypothalamic melanocortin 4 receptors (MC4R) are a key regulator of energy homeostasis. Brain-penetrant MC4R agonists have failed, as concentrations required to suppress food intake also increase blood pressure. However, peripherally located MC4R may also mediate metabolic benefits of MC4R activation. Mc4r transcript is enriched in mouse enteroendocrine L cells and peripheral administration of the endogenous MC4R agonist, α-melanocyte stimulating hormone (α-MSH), triggers the release of the anorectic hormones Glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in mice. This study aimed to determine whether pathways linking MC4R and L-cell secretion exist in humans. DESIGN: GLP-1 and PYY levels were assessed in body mass index-matched individuals with or without loss-of-function MC4R mutations following an oral glucose tolerance test. Immunohistochemistry was performed on human intestinal sections to characterize the mucosal MC4R system. Static incubations with MC4R agonists were carried out on human intestinal epithelia, GLP-1 and PYY contents of secretion supernatants were assayed. RESULTS: Fasting PYY levels and oral glucose-induced GLP-1 secretion were reduced in humans carrying a total loss-of-function MC4R mutation. MC4R was localized to L cells and regulates GLP-1 and PYY secretion from ex vivo human intestine. α-MSH immunoreactivity in the human intestinal epithelia was predominantly localized to L cells. Glucose-sensitive mucosal pro-opiomelanocortin cells provide a local source of α-MSH that is essential for glucose-induced GLP-1 secretion in small intestine. CONCLUSION: Our findings describe a previously unidentified signaling nexus in the human gastrointestinal tract involving α-MSH release and MC4R activation on L cells in an autocrine and paracrine fashion. Outcomes from this study have direct implications for targeting mucosal MC4R to treat human metabolic disorders.


Assuntos
Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/metabolismo , Peptídeo YY/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , alfa-MSH/metabolismo , Comunicação Autócrina , Glicemia/metabolismo , Estudos de Casos e Controles , Células Enteroendócrinas/efeitos dos fármacos , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mutação com Perda de Função , Comunicação Parácrina , Pró-Opiomelanocortina/genética , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/genética , Via Secretória , Transdução de Sinais , Fatores de Tempo , alfa-MSH/farmacologia
10.
FASEB J ; 35(4): e21435, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33749879

RESUMO

Peptide YY (PYY), produced by endocrine L cells in the gut, is known for its critical role in regulating gastrointestinal functions as well as satiety. However, how these processes are integrated with maintaining a healthy gut microbiome composition is unknown. Here, we show that lack of PYY in mice leads to distinct changes in gut microbiome composition that are diet-dependent. While under chow diet only slight differences in gut microbiome composition could be observed, high-fat diet (HFD) aggravated these differences. Specifically an increased abundance of the Bacteroidetes phylum with a corresponding decrease of the Firmicutes/Bacteroidetes ratio could be detected in Pyy-knockout (KO) mice in response to HFD. Detailed analysis of the Bacteroidetes phylum further revealed that the Alistipes genus belonging to the Rikenellaceae family, the Parabacteroides belonging to the Tannerellaceae family, as well as Muribaculum were increased in Pyy-KO mice. In order to investigate whether these changes are associated with changed markers of gut barrier and immunity, we analyzed the colonic expression of various pro-inflammatory cytokines, as well as tight junction proteins and mucin 2, and identified increased mRNA expression of the tight junction proteins Cldn2 and Ocel1 in Pyy-KO mice, while pro-inflammatory cytokine expression was not significantly altered. Together these results highlight a critical gene-environment interaction between diet and the gut microbiome and its impact on homeostasis of the intestinal epithelium under conditions of reduced PYY signaling which is commonly seen under obese conditions.


Assuntos
Bactérias/classificação , Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal , Peptídeo YY/metabolismo , Animais , Composição Corporal , Camundongos , Camundongos Knockout , Peptídeo YY/genética
11.
Diabetes Obes Metab ; 24(12): 2353-2363, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35848461

RESUMO

AIM: To examine whether sequential administration of (d-Arg35 )-sea lamprey peptide tyrosine tyrosine (1-36) (SL-PYY) and the glucagon-like peptide-1 (GLP-1) mimetic, liraglutide, has beneficial effects in diabetes. METHODS: SL-PYY is an enzymatically stable neuropeptide Y1 receptor (NPY1R) agonist known to induce pancreatic beta-cell rest and improve overall beta-cell health. We employed SL-PYY and liraglutide to induce appropriate recurrent periods of beta-cell rest and stimulation, to assess therapeutic benefits in high fat fed (HFF) mice with streptozotocin (STZ)-induced insulin deficiency, namely HFF-STZ mice. RESULTS: Previous studies confirm that, at a dose of 0.25 nmol/kg, liraglutide exerts bioactivity over an 8-12 hour period in mice. Initial pharmacokinetic analysis revealed that 75 nmol/kg SL-PYY yielded a similar plasma drug time profile. When SL-PYY (75 nmol/kg) and liraglutide (0.25 nmol/kg) were administered sequentially at 08:00 AM and 08:00 PM, respectively, to HFF-STZ mice for 28 days, reductions in energy intake, body weight, circulating glucose, insulin and glucagon were noted. Similarly positive, but slightly less striking, effects were also apparent with twice-daily liraglutide-only therapy. The sequential SL-PYY and liraglutide treatment also improved insulin sensitivity and glucose-induced insulin secretory responses, which was not apparent with liraglutide treatment, although benefits on glucose tolerance were mild. Interestingly, combined therapy also elevated pancreatic insulin, decreased pancreatic glucagon and enhanced the plasma insulin/glucagon ratio compared with liraglutide alone. This was not associated with an enhancement of beneficial changes in islet cell areas, proliferation or apoptosis compared with liraglutide alone, but the numbers of centrally stained glucagon-positive islet cells were reduced by sequential combination therapy. CONCLUSION: These data show that NPY1R-induced intervals of beta-cell rest, combined with GLP-1R-stimulated periods of beta-cell stimulation, should be further evaluated as an effective treatment option for obesity-driven forms of diabetes.


Assuntos
Diabetes Mellitus Experimental , Neuropeptídeos , Animais , Camundongos , Glicemia/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/uso terapêutico , Glucose/uso terapêutico , Insulina/uso terapêutico , Liraglutida/farmacologia , Liraglutida/uso terapêutico , Neuropeptídeos/uso terapêutico , Peptídeo YY/metabolismo , Estreptozocina/uso terapêutico , Tirosina/uso terapêutico , Neuropeptídeo Y/farmacologia
12.
Handb Exp Pharmacol ; 274: 487-513, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35419620

RESUMO

Mimetics of the anorexigenic gut hormone glucagon-like peptide 1 (GLP-1) were originally developed as insulinotropic anti-diabetic drugs but also evoke significant weight loss, leading to their recent approval as obesity therapeutics. Co-activation of receptors for GLP-1 and other gut hormones which reduce food intake - peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP) - is now being explored clinically to enhance efficacy. An alternative approach involves pharmacologically stimulating endogenous secretion of these hormones from enteroendocrine cells (EECs) to recapitulate the metabolic consequences of bariatric surgery, where highly elevated postprandial levels of GLP-1 and PYY3-36 are thought to contribute to improved glycaemia and weight loss.


Assuntos
Polipeptídeo Inibidor Gástrico , Hormônios Gastrointestinais , Polipeptídeo Inibidor Gástrico/metabolismo , Hormônios Gastrointestinais/metabolismo , Peptídeo 1 Semelhante ao Glucagon , Humanos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Peptídeo YY/metabolismo , Redução de Peso
13.
Biochem Biophys Res Commun ; 557: 48-54, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33862459

RESUMO

Olfactory receptor 78 (Olfr78), which is also known as a receptor for short-chain fatty acids (SCFAs) produced via gut microbial fermentation from indigestible polysaccharides such as dietary fibers, is expressed in the enteroendocrine cells of the colon. However, the role of Olfr78 in gut hormone secretion remains unknown. Here, we aimed to investigate the function and mechanism of action of Olfr78 in vivo and in vitro. Toward this, we assessed the expression of Olfr78 in several tissues, affinity of Olfr78 to various monocarboxylates, and the secretion of anorexigenic gut hormone peptide YY (PYY) via Olfr78 using various molecular and biochemical techniques. Olfr78 was abundantly expressed in the colon and mouse enteroendocrine cell line STC-1 and showed specific affinity to SCFAs such as acetate and propionate, but not butyrate, in a monocarboxylate ligand screening assay using a heterologous expression system. Acetate promoted PYY secretion in STC-1 cells via Olfr78-protein kinase A signaling, whereas the effects were abolished by Olfr78 RNA interference. Colonic SCFAs production via oral administration of fructo-oligosaccharide significantly increased plasma PYY levels, whereas this effect was abolished in Olfr78-deficient and germ-free mice. These results suggested that the SCFA receptor Olfr78 is important for anti-obesity and anorexigenic effects of the gut microbiota and dietary fibers.


Assuntos
Anorexia/metabolismo , Anorexia/microbiologia , Ácidos Graxos Voláteis/farmacologia , Microbioma Gastrointestinal , Mucosa Intestinal/microbiologia , Obesidade/microbiologia , Peptídeo YY/metabolismo , Receptores Odorantes/metabolismo , Animais , Anorexia/patologia , Células Cultivadas , Modelos Animais de Doenças , Células Enteroendócrinas/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia
14.
Horm Behav ; 136: 105056, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34509673

RESUMO

Mammalian taste bud cells express receptors for numerous peptides implicated elsewhere in the body in the regulation of metabolism, nutrient assimilation, and satiety. The perturbation of several peptide signaling pathways in the gustatory periphery results in changes in behavioral and/or physiological responsiveness to subsets of taste stimuli. We previously showed that Peptide YY (PYY) - which is present in both saliva and in subsets of taste cells - can affect behavioral taste responsiveness and reduce food intake and body weight. Here, we investigated the contributions of taste bud-localized receptors for PYY and the related Neuropeptide Y (NPY) on behavioral taste responsiveness. Y1R, but not Y2R, null mice show reduced responsiveness to sweet, bitter, and salty taste stimuli in brief-access taste tests; similar results were seen when wildtype mice were exposed to Y receptor antagonists in the taste stimuli. Finally, mice in which the gene encoding the NPY propeptide was deleted also showed reduced taste responsiveness to sweet and bitter taste stimuli. Collectively, these results suggest that Y1R signaling, likely through its interactions with NPY, can modulate peripheral taste responsiveness in mice.


Assuntos
Papilas Gustativas , Paladar , Animais , Masculino , Mamíferos/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Peptídeo YY/metabolismo , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Papilas Gustativas/metabolismo
15.
J Surg Res ; 260: 399-408, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33261855

RESUMO

BACKGROUND: We have optimized a technique for cannulation of mesenteric lymph duct (MLD) in mice. Mice have low rates of intestinal lymph production; the MLDs are smaller and associated with fragile vasculature. Previous protocols for lymph collection based on the open lymph fistula model were associated with low success rates in mice. Bariatric surgery procedures worsen success rates due to postoperative adhesions and GI rearrangement. We have used this procedure to collect mesenteric lymph from mice undergoing bile diversion from gall bladder to ileum (GB-IL). HYPOTHESIS: We hypothesize that peptide YY (PYY) levels in mesenteric lymph will increase following nutrient delivery in mice undergoing bile diversion from gall bladder to ileum (GB-IL). METHODS AND RESULTS: We observe that cannulation of the MLD using a needled-catheter maintains lymph vessel integrity, prevents excessive lymph leakage, and is less traumatic, leading to high success rates (>95%). PYY levels in mesenteric lymph after GB-IL were significantly higher post nutrient infusion. The procedure takes approximately 20 min; small rodent surgical experience and practice are required for success. CONCLUSIONS: Intestinal lymph can be collected from mice, including those undergoing bariatric surgical procedures with high success rates by cannulation of the mesenteric lymph duct.


Assuntos
Cirurgia Bariátrica , Procedimentos Cirúrgicos do Sistema Biliar , Cateterismo/métodos , Linfa/metabolismo , Vasos Linfáticos/cirurgia , Mesentério/cirurgia , Peptídeo YY/metabolismo , Animais , Bile , Biomarcadores/metabolismo , Feminino , Vesícula Biliar/cirurgia , Íleo/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
16.
Arch Toxicol ; 95(8): 2797-2813, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34148100

RESUMO

The consumption of contaminated shellfish with okadaic acid (OA) group of toxins leads to diarrhoeic shellfish poisoning (DSP) characterized by a set of symptoms including nausea, vomiting and diarrhoea. These phycotoxins are Ser/Thr phosphatase inhibitors, which produce hyperphosphorylation in cellular proteins. However, this inhibition does not fully explain the symptomatology reported and other targets could be relevant to the toxicity. Previous studies have indicated a feasible involvement of the nervous system. We performed a set of in vivo approaches to elucidate whether neuropeptide Y (NPY), Peptide YY (PYY) or serotonin (5-HT) was implicated in the early OA-induced diarrhoea. Fasted Swiss female mice were administered NPY, PYY(3-36) or cyproheptadine intraperitoneal prior to oral OA treatment (250 µg/kg). A non-significant delay in diarrhoea onset was observed for NPY (107 µg/kg) and PYY(3-36) (1 mg/kg) pre-treatment. On the contrary, the serotonin antagonist cyproheptadine was able to block (10 mg/kg) or delay (0.1 and 1 mg/kg) diarrhoea onset suggesting a role of 5-HT. This is the first report of the possible involvement of serotonin in OA-induced poisoning.


Assuntos
Diarreia/etiologia , Ácido Okadáico/toxicidade , Serotonina/metabolismo , Animais , Ciproeptadina/farmacologia , Inibidores Enzimáticos/toxicidade , Feminino , Camundongos , Neuropeptídeo Y/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Antagonistas da Serotonina/farmacologia , Intoxicação por Frutos do Mar/fisiopatologia , Fatores de Tempo
17.
Appetite ; 164: 105260, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33848592

RESUMO

Nicotine has been shown to decrease appetite, food intake (FI) and body weight, but the mechanisms are unclear. The purpose of this review was to examine research on the effects of nicotine on energy balance by exploring physiological mechanisms and hormone regulation related to FI, subjective appetite and energy expenditure (EE). We searched PubMed and MEDLINE, and included articles investigating the effects of nicotine on central appetite regulation, FI, leptin, peptide-YY (PYY), ghrelin, glucagon-like peptide-1 (GLP-1), adiponectin, cholecystokinin (CCK), orexin, and EE. A total of 65 studies were included in the qualitative synthesis and review. Our findings suggest that the decrease in appetite and FI may be attributed to nicotinic alterations of neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) but the effect of nicotine on FI remains unclear. Furthermore, nicotine increases resting EE (REE) and physical activity EE (PAEE) in both smokers and non-smokers; and these increases may be a result of the catecholaminergic effect of nicotine. Decreases in body weight and appetite experienced by nicotine users results from increased EE and changes in the central hypothalamic regulation of appetite. There is not enough evidence to implicate a relationship between peripheral hormones and changes in appetite or FI after nicotine use. Although nicotine increases REE and PAEE, the effect of nicotine on other components of EE warrants further research. We conclude that further research evaluating the effect of nicotine on appetite hormones, FI and EE in humans is warranted.


Assuntos
Apetite , Metabolismo Energético , Nicotina , Regulação do Apetite , Ingestão de Energia , Grelina/metabolismo , Humanos , não Fumantes , Peptídeo YY/metabolismo , Fumantes
18.
Diabetologia ; 63(7): 1396-1407, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32342115

RESUMO

AIMS/HYPOTHESIS: Insulin-like peptide-5 (INSL5) is found only in distal colonic L cells, which co-express glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). GLP-1 is a well-known insulin secretagogue, and GLP-1 and PYY are anorexigenic, whereas INSL5 is considered orexigenic. We aimed to clarify the metabolic impact of selective stimulation of distal colonic L cells in mice. METHODS: Insl5 promoter-driven expression of Gq-coupled Designer Receptor Exclusively Activated by Designer Drugs (DREADD) was employed to activate distal colonic L cells (LdistalDq). IPGTT and food intake were assessed with and without DREADD activation. RESULTS: LdistalDq cell stimulation with clozapine N-oxide (CNO; 0.3 mg/kg i.p.) increased plasma GLP-1 and PYY (2.67- and 3.31-fold, respectively); INSL5 was not measurable in plasma but was co-secreted with GLP-1 and PYY in vitro. IPGTT (2 g/kg body weight) revealed significantly improved glucose tolerance following CNO injection. CNO-treated mice also exhibited reduced food intake and body weight after 24 h, and increased defecation, the latter being sensitive to 5-hydroxytryptamine (5-HT) receptor 3 inhibition. Pre-treatment with a GLP1 receptor-blocking antibody neutralised the CNO-dependent improvement in glucose tolerance but did not affect the reduction in food intake, and an independent group of animals pair-fed to the CNO-treatment group demonstrated attenuated weight loss. Pre-treatment with JNJ-31020028, a neuropeptide Y receptor type 2 antagonist, abolished the CNO-dependent effect on food intake. Assessment of whole body physiology in metabolic cages revealed LdistalDq cell stimulation increased energy expenditure and increased activity. Acute CNO-induced food intake and glucose homeostasis outcomes were maintained after 2 weeks on a high-fat diet. CONCLUSIONS/INTERPRETATION: This proof-of-concept study demonstrates that selective distal colonic L cell stimulation has beneficial metabolic outcomes. Graphical abstract.


Assuntos
Colo/metabolismo , Células L/metabolismo , Animais , Colo/citologia , Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Insulina/metabolismo , Masculino , Camundongos , Peptídeo YY/metabolismo , Proteínas/metabolismo
19.
Diabetologia ; 63(10): 2194-2204, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32728891

RESUMO

AIMS/HYPOTHESIS: Metformin is the only approved oral agent for youth with type 2 diabetes but its mechanism of action remains controversial. Recent data in adults suggest a primary role for the enteroinsular pathway, but there are no data in youth, in whom metformin efficacy is only ~50%. Our objectives were to compare incretin concentrations and rates of glucose production and gluconeogenesis in youth with type 2 diabetes before and after short-term metformin therapy compared with peers with normal glucose tolerance (NGT). METHODS: This is a case-control observational study in youth with type 2 diabetes who were not on metformin (n = 18) compared with youth with NGT (n = 10) who were evaluated with a 2 day protocol. A 75 g OGTT was administered to measure intact glucagon-like 1 peptide (iGLP-1), gastric inhibitory polypeptide (GIP) and peptide YY (PYY). Insulinogenic index (IGI) and whole-body insulin sensitivity were calculated using glucose and insulin levels from the OGTT. Basal rates of gluconeogenesis (2H2O), glucose production ([6,6-2H2]glucose) and whole-body lipolysis ([2H5]glycerol) were measured after an overnight fast on study day 2. Youth with type 2 diabetes (n = 9) were subsequently evaluated with an identical 2 day protocol after 3 months on the metformin study. RESULTS: Compared with individuals with NGT, those with type 2 diabetes had higher fasting (7.8 ± 2.5 vs 5.1 ± 0.3 mmol/l, mean ± SD p = 0.002) and 2 h glucose concentrations (13.8 ± 4.5 vs 5.9 ± 0.9 mmol/l, p = 0.001), higher rates of absolute gluconeogenesis (10.0 ± 1.7 vs 7.2 ± 1.1 µmol [kg fat-free mass (FFM)]-1 min-1, p < 0.001) and whole-body lipolysis (5.2 ± 0.9 vs 4.0 ± 1.4 µmol kgFFM-1 min-1, p < 0.01), but lower fasting iGLP-1 concentrations (0.5 ± 0.5 vs 1.3 ± 0.7 pmol/l, p < 0.01). Metformin decreased 2 h glucose (pre metformin 11.4 ± 2.8 vs post metformin 9.9 ± 1.9 mmol/l, p = 0.04) and was associated with ~20-50% increase in IGI (median [25th-75th percentile] pre 1.39 [0.89-1.47] vs post 1.43 [0.88-2.70], p = 0.04), fasting iGLP-1 (pre 0.3 ± 0.2 vs post 1.0 ± 0.7 pmol/l, p = 0.02), 2 h iGLP (pre 0.4 ± 0.2 vs post 1.2 ± 0.9 pmol/l, p = 0.06), fasting PYY (pre 6.3 ± 2.2 vs post 10.5 ± 4.3 pmol/l, p < 0.01) and 2 h PYY (pre 6.6 ± 2.9 vs post 9.0 ± 4.0 pmol/l, p < 0.01). There was no change in BMI, insulin sensitivity or GIP concentrations pre vs post metformin. There were no differences pre vs post metformin in rates of glucose production (15.0 ± 3.9 vs 14.9 ± 2.2 µmol kgFFM-1 min-1, p = 0.84), absolute gluconeogenesis (9.9 ± 1.8 vs 9.7 ± 1.7 µmol kgFFM-1 min-1, p = 0.76) or whole-body lipolysis (5.0 ± 0.7 vs 5.3 ± 1.3 µmol kgFFM-1 min-1, p = 0.20). Post metformin iGLP-1 and PYY concentrations in youth with type 2 diabetes were comparable to levels in youth with NGT. CONCLUSIONS/INTERPRETATION: Overall, the improved postprandial blood glucose levels and increase in incretins observed in the absence of changes in insulin sensitivity and gluconeogenesis, support an enteroinsular mechanistic pathway in youth with type 2 diabetes treated with short-term metformin.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Gluconeogênese , Hipoglicemiantes/uso terapêutico , Incretinas/metabolismo , Metformina/uso terapêutico , Adolescente , Estudos de Casos e Controles , Criança , Óxido de Deutério , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/biossíntese , Humanos , Secreção de Insulina , Masculino , Peptídeo YY/metabolismo
20.
Ann Surg ; 272(1): 72-80, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31592891

RESUMO

OBJECTIVE: To perform a structured systematic review and meta-analysis to evaluate changes in ghrelin, glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and gastric inhibitory peptide (GIP) gut hormone levels in patients after sleeve gastrectomy. BACKGROUND: Despite sleeve gastrectomy becoming the most common surgical weight loss procedure, weight loss mechanisms remain less clear. METHODS: Searches of PubMed, EMBASE, Web of Science, and Cochrane Library databases were performed through April 1, 2019, in accordance with PRISMA and MOOSE guidelines. Randomized controlled trials and prospective observational studies evaluating pre and post-procedure hormones fasting ghrelin, postprandial GLP-1, postprandial PYY, and fasting GIP levels were included. Hedge g with random-effects models were used to determine pooled effect size and corresponding 95% confidence intervals (CIs). RESULTS: A total of 28 studies (n = 653; 29.56% male) were included. Mean age was 42.00 ±â€Š5.48 years, with average follow-up of 11.70 ±â€Š11.38 months. Pre-procedure body mass index (BMI) was 46.01 ±â€Š4.07 kg/m with a postsleeve gastrectomy BMI of 34.07 ±â€Š3.73 kg/m, representing total body weight loss of 25.13 ±â€Š4.44% and excess weight loss of 57.48 ±â€Š9.64% (P < 0.001). Ghrelin decreased (Hedge g -1.486, 95% CI -1.884 to -1.089, I = 91.95%), whereas GLP-1 and PYY increased post-procedure (Hedge g 1.095, 95% CI 0.509 to 1.642, I = 84.38%; and Hedge g 1.396, 95% CI 0.781 to 2.011, I = 84.02%, respectively). GIP did not significantly change (Hedge g -0.213, 95% CI -1.019 to 0.592, I = 79.65%). CONCLUSIONS: Fasting ghrelin levels decreased, whereas postprandial GLP-1 and PYY increased after sleeve gastrectomy. Fasting GIP levels remained unchanged. Future studies are needed to assess the role of these gut hormones and relationship to weight loss and metabolic outcomes.


Assuntos
Biomarcadores/metabolismo , Gastrectomia/métodos , Obesidade Mórbida/cirurgia , Hormônios Gastrointestinais/metabolismo , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Peptídeo YY/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA