Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 619(7968): 143-150, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380764

RESUMEN

Caloric restriction that promotes weight loss is an effective strategy for treating non-alcoholic fatty liver disease and improving insulin sensitivity in people with type 2 diabetes1. Despite its effectiveness, in most individuals, weight loss is usually not maintained partly due to physiological adaptations that suppress energy expenditure, a process known as adaptive thermogenesis, the mechanistic underpinnings of which are unclear2,3. Treatment of rodents fed a high-fat diet with recombinant growth differentiating factor 15 (GDF15) reduces obesity and improves glycaemic control through glial-cell-derived neurotrophic factor family receptor α-like (GFRAL)-dependent suppression of food intake4-7. Here we find that, in addition to suppressing appetite, GDF15 counteracts compensatory reductions in energy expenditure, eliciting greater weight loss and reductions in non-alcoholic fatty liver disease (NAFLD) compared to caloric restriction alone. This effect of GDF15 to maintain energy expenditure during calorie restriction requires a GFRAL-ß-adrenergic-dependent signalling axis that increases fatty acid oxidation and calcium futile cycling in the skeletal muscle of mice. These data indicate that therapeutic targeting of the GDF15-GFRAL pathway may be useful for maintaining energy expenditure in skeletal muscle during caloric restriction.


Asunto(s)
Metabolismo Energético , Factor 15 de Diferenciación de Crecimiento , Músculo Esquelético , Pérdida de Peso , Animales , Humanos , Ratones , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Depresores del Apetito/uso terapéutico , Restricción Calórica , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Factor 15 de Diferenciación de Crecimiento/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/terapia , Receptores Adrenérgicos beta/metabolismo , Pérdida de Peso/efectos de los fármacos
2.
PLoS Biol ; 18(2): e3000629, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32097406

RESUMEN

Human biology has evolved to keep body fat within a range that supports survival. During the last 25 years, obesity biologists have uncovered key aspects of physiology that prevent fat mass from becoming too low. In contrast, the mechanisms that counteract excessive adipose expansion are largely unknown. Evidence dating back to the 1950s suggests the existence of a blood-borne molecule that defends against weight gain. In this article, we discuss the research supporting an "unidentified factor of overfeeding" and models that explain its role in body weight control. If it exists, revealing the identity of this factor could end a long-lasting enigma of energy balance regulation and facilitate a much-needed breakthrough in the pharmacological treatment of obesity.


Asunto(s)
Depresores del Apetito/metabolismo , Peso Corporal/fisiología , Hormonas/metabolismo , Tejido Adiposo/metabolismo , Animales , Depresores del Apetito/sangre , Hormonas/sangre , Humanos , Hiperfagia/genética , Hiperfagia/metabolismo , Obesidad/genética , Obesidad/metabolismo , Parabiosis , Aumento de Peso/fisiología
3.
Gut ; 71(5): 928-937, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34083384

RESUMEN

OBJECTIVE: Colonic enteroendocrine cells (EECs) store and release potent anorectic hormones that are key regulators of satiety. EECs express multiple nutrient sensing receptors, particularly for medium-chain fatty acids (MCFAs): GPR84 and FFAR4. Here we show a non-surgical approach with targeted colonic delivery of MCFA, which induces EEC and neuronal activation leading to anorectic effects. DESIGN: A randomised, double-blind, placebo-controlled, cross-over study was performed in obese adults given combined GPR84 and FFAR4 agonists in colonic release capsules before meals. We measured serum hormones, energy intake and appetite perception. Cell type, activation by agonists and hormone/serotonin release were determined in human colonic explants. Mouse colonic afferent nerve responses to nutrients/mediators were recorded electrophysiologically. RESULTS: Subjects receiving GPR84 and FFAR4 agonists had reduced overall calorific intake and increased postprandial levels of PYY versus placebo. Receptors including GPR84 and FFAR4 were coexpressed on human colonic EEC. Activation of GPR84 exclusively induced intracellular pERK, whereas FFAR4 selectively activated pCaMKII. Coactivation of GPR84 and FFAR4 induced both phosphoproteins, and superadditive release of GLP-1 and PYY. Nutrients and hormones convergently activated murine colonic afterent nerves via GLP-1, Y2 and 5-HT3 receptors. CONCLUSIONS: Colonic GPR84 and FFAR4 agonists reduce energy intake and increase postprandial PYY in obese adults. Human colonic EECs coexpress these receptors, which activate cells via parallel intracellular pathways and synergistically evoke hormone release. Further synergism occurs in sensory nerve responses to MCFA and EEC mediators. Thus, synergistic activation of colonic endocrine cells via nutrient receptors is an important target for metabolic regulation. TRAIL REGISTRATION NUMBER: NCT04292236.


Asunto(s)
Depresores del Apetito , Animales , Apetito , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Estudios Cruzados , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Ratones , Nutrientes , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-34534676

RESUMEN

Amylin is a 37-amino acid polypeptide that has been found to be involved in feeding regulation in some mammals, birds, and goldfish. We cloned amylin of Siberian sturgeon and detected its distribution pattern in 15 tissues. The expression levels in the periprandial period (pre-and post-feeding), the changes in the food intake, and the expression levels of related appetite factors after the intraperitoneal injection of amylin were detected. The expression of amylin was found to be the highest in the hypothalamus. Compared with 1 h pre-feeding, the expression levels of amylin in the hypothalamus and duodenum were increased significantly 1 h post-feeding. Compared with the control group (saline), intraperitoneal injection of 50 ng/g, 100 ng/g, and 200 ng/g of amylin significantly inhibited food intake at 1 h post injection, but not at 3 h and 6 h. The injection of 50 ng/g, 100 ng/g, and 200 ng/g amylin significantly inhibited the cumulative feed. After 1 h of 50 ng/g amylin injection, the levels of MC4R and somatostatin in the hypothalamus increased significantly, while the levels of amylin and NPY decreased significantly. The levels of CCK in the valvular intestine were increased significantly. Insulin in the duodenum was also increased significantly, but there was no significant change in ghrelin in the duodenum. These results show that amylin inhibits feeding in Siberian sturgeon by down-regulating the appetite-stimulating factor NPY and up-regulating the appetite-suppressing factors somatostatin, MC4R, CCK, and insulin. This study provides a theoretical basis for studying the feeding function and action mechanisms of amylin in Siberian sturgeon.


Asunto(s)
Proteínas de Peces/metabolismo , Peces/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Secuencia de Aminoácidos , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/metabolismo , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/genética , Regulación del Apetito/fisiología , Secuencia de Bases , Clonación Molecular , Duodeno/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Proteínas de Peces/administración & dosificación , Proteínas de Peces/genética , Peces/genética , Peces/fisiología , Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraperitoneales , Polipéptido Amiloide de los Islotes Pancreáticos/administración & dosificación , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Filogenia , Homología de Secuencia de Aminoácido , Distribución Tisular
5.
J Am Coll Nutr ; 39(4): 345-351, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31369353

RESUMEN

Nesfatin-1 is a peptide derived from nucleobindin-2 and involved in the regulation of food intake and hyperglycemia. Nesfatin-1 is a recently described anorexigenic peptide, which may be involved in weight loss, malnutrition, and the regulation of appetite. Nesfatin-1 has an effect on the regulation of glucose homeostasis as well as that of food intake. The aim of this article is to bring a different perspective to the readers on the effects of nesfatin-1 on food intake and hyperglycemia. The central injection of nesfatin-1 may produce anorexigenic effects. The circulating level of nesfatin-1 is thought to be regulated by nutritional status. Long-term changes in body weight can affect nesfatin-1 levels. In overweight and obese individuals, nesfatin-1 levels may increase. Nesfatin-1 is synthesized in the hypothalamic appetite control regions. Nesfatin-1 levels may decrease in individuals with diabetes but may increase in those with impaired glucose tolerance. Nesfatin-1 may have a reducing effect on glucose levels. In addition, an increase in glucose levels may lead to an increase in the release of nesfatin-1 from pancreatic cells. Injection of nesfatin-1 can prevent hepatic glucose formation and stimulate glucose uptake. Reduction of hypothalamic nesfatin-1 levels increases hepatic glucose flow and decreases glucose uptake from peripheral tissues. In the light of all this information, nesfatin-1 may be considered to be an important regulator in the metabolic process. Nesfatin-1 appears to be able to contribute to the treatment of obesity and diabetes because of its anorexigenic and antihyperglycemic effects. Key teaching pointsNesfatin-1 is a anorexigenic peptide.Nesfatin-1 is derived from Nucleobindin-2.Nucleobindin-2 mRNA is produced in different areas of the brain.Nesfatin-1 is an inhibitory factor on appetite and a regulator of energy balance that reduces the increase in body weight.


Asunto(s)
Depresores del Apetito/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Hiperglucemia/metabolismo , Nucleobindinas/metabolismo , Animales , Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Hipotálamo/metabolismo , Nucleobindinas/biosíntesis , Estado Nutricional/efectos de los fármacos
6.
J Nutr ; 149(3): 362-365, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30722047

RESUMEN

Oleoylethanolamide is a well-recognized anorectic compound which also has noteworthy effects on food-reward, influencing the acetylcholine (ACh)/dopamine (DA) balance in the cholinergic system. After its administration, oleoylethanolamide is quickly degraded into oleic acid and ethanolamine. The effect of oleic acid on the gut-brain axis has been extensively investigated, whereas ethanolamine has received scarce attention. However, there is scattered evidence from old and recent research that has underlined the influence of ethanolamine on the cholinergic system. In the present article, we propose a model by which the released ethanolamine contributes to the overall balance between DA and ACh after oleoylethanolamide administration.


Asunto(s)
Acetilcolina/metabolismo , Dopamina/metabolismo , Endocannabinoides/farmacología , Etanolamina/metabolismo , Conducta Alimentaria/efectos de los fármacos , Ácidos Oléicos/farmacología , Animales , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Endocannabinoides/metabolismo , Humanos , Ratones , Modelos Biológicos , Ácidos Oléicos/metabolismo , Ratas
7.
Diabetes Obes Metab ; 20(8): 1817-1828, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29687585

RESUMEN

Type 2 diabetes mellitus is now a worldwide health problem with increasing prevalence. Mounting efforts have been made to treat, prevent and predict this chronic disease. In recent years, increasing evidence from mice and clinical studies suggests that bone-derived molecules modulate glucose metabolism. This review aims to summarize our current understanding of the interplay between bone and glucose metabolism and to highlight potential new means of therapeutic intervention. The first molecule recognized as a link between bone and glucose metabolism is osteocalcin (OCN), which functions in its active form, that is, undercarboxylated OCN (ucOC). ucOC acts in promoting insulin expression and secretion, facilitating insulin sensitivity, and favouring glucose and fatty acid uptake and utilization. A second bone-derived molecule, lipocalin2, functions in suppressing appetite in mice through its action on the hypothalamus. Osteocytes, the most abundant cells in bone matrix, are suggested to act on the browning of white adipose tissue and energy expenditure through secretion of bone morphogenetic protein 7 and sclerostin. The involvement of bone resorption in glucose homeostasis has also been examined. However, there is evidence indicating the implication of the receptor activator of nuclear factor κ-B ligand, neuropeptide Y, and other known and unidentified bone-derived factors that function in glucose homeostasis. We summarize recent advances and the rationale for treating, preventing and predicting diabetes by skeleton intervention.


Asunto(s)
Huesos/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Modelos Biológicos , Estado Prediabético/tratamiento farmacológico , Animales , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Depresores del Apetito/uso terapéutico , Regulación del Apetito/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/prevención & control , Metabolismo Energético/efectos de los fármacos , Humanos , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Secreción de Insulina/efectos de los fármacos , Lipocalina 2/genética , Lipocalina 2/metabolismo , Lipocalina 2/farmacología , Lipocalina 2/uso terapéutico , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Neuropéptido Y/uso terapéutico , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocalcina/genética , Osteocalcina/metabolismo , Osteocalcina/farmacología , Osteocalcina/uso terapéutico , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/patología , Estado Prediabético/metabolismo , Estado Prediabético/patología , Estado Prediabético/prevención & control , Ligando RANK/genética , Ligando RANK/metabolismo , Ligando RANK/farmacología , Ligando RANK/uso terapéutico , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Secretagogos/metabolismo , Secretagogos/farmacología , Secretagogos/uso terapéutico
8.
Eur J Nutr ; 57(3): 965-979, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28161724

RESUMEN

PURPOSE: Increasing secretion and production of glucagon-like peptide-1 (GLP-1) by continuous ingestion of certain food components has been expected to prevent glucose intolerance and obesity. In this study, we examined whether a physiological dose (5% weight in diet) of digestion-resistant maltodextrin (RMD) has a GLP-1-promoting effect in rats fed a high-fat and high-sucrose (HFS) diet. METHODS: Rats were fed a control diet or the HFS (30% fat, 40% sucrose wt/wt) diet supplemented with 5% RMD or fructooligosaccharides (FOS) for 8 weeks or for 8 days in separated experiments. Glucose tolerance, energy intake, plasma and tissue GLP-1 concentrations, and cecal short-chain fatty acids concentrations were assessed. RESULTS: After 4 weeks of feeding, HFS-fed rats had significantly higher glycemic response to oral glucose than control rats, but rats fed HFS + RMD/FOS did not (approx. 50% reduction vs HFS rats). HFS + RMD/FOS-fed rats had higher GLP-1 responses (~twofold) to oral glucose, than control rats. After 8 weeks, visceral adipose tissue weight was significantly higher in HFS-fed rats than control rats, while HFS + RMD/FOS rats had a trend of reduced gain (~50%) of the tissue weight. GLP-1 contents and luminal propionate concentrations in the large intestine increased (>twofold) by adding RMD/FOS to HFS. Eight days feeding of RMD/FOS-supplemented diets reduced energy intake (~10%) and enhanced cecal GLP-1 production (~twofold), compared to HFS diet. CONCLUSIONS: The physiological dose of a prebiotic fiber promptly (within 8 days) promotes GLP-1 production in rats fed an obesogenic diet, which would help to prevent excess energy intake and fat accumulation.


Asunto(s)
Depresores del Apetito/uso terapéutico , Disbiosis/prevención & control , Péptido 1 Similar al Glucagón/agonistas , Obesidad/prevención & control , Oligosacáridos/uso terapéutico , Polisacáridos/uso terapéutico , Prebióticos , Adiposidad , Animales , Depresores del Apetito/metabolismo , Ciego/metabolismo , Ciego/microbiología , Ciego/patología , Dieta Occidental/efectos adversos , Digestión , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/patología , Ingestión de Energía , Ácidos Grasos Volátiles/metabolismo , Fermentación , Contenido Digestivo/química , Contenido Digestivo/microbiología , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Grasa Intraabdominal/patología , Masculino , Obesidad/metabolismo , Obesidad/microbiología , Obesidad/patología , Oligosacáridos/metabolismo , Tamaño de los Órganos , Polisacáridos/metabolismo , Ratas Sprague-Dawley
9.
Biochem Biophys Res Commun ; 494(1-2): 152-157, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29037812

RESUMEN

Apolipoprotein A-IV (apoA-IV) is a satiation factor that acts in the hypothalamus, however, the intracellular mechanisms responsible for this action are still largely unknown. Here we report that apoA-IV treatment elicited a rapid activation of the phosphatidylinositol-3-kinase (PI3K) signaling pathway in cultured primary hypothalamic neurons, and this effect was significantly attenuated by pretreatment with LY294002, an inhibitor of the PI3K pathway. To determine if the activation of PI3K is required for apoA-IV's inhibitory effect on food intake, apoA-IV was administered intracerebroventricularly. We found that apoA-IV significantly reduced food intake and activated PI3K signaling in the hypothalamus, and these effects were abolished by icv pre-treatment with LY294002. To identify the distinct brain sites where apoA-IV exerts its anorectic action, apoA-IV was administered into the ventromedial hypothalamus (VMH) through implanted bilateral cannula. At a low dose (0.5 µg), apoA-IV significantly inhibited food intake and activated PI3K signaling pathway in the VMH of lean rats, but not in high-fat diet-induced obese (DIO) rats. These results collectively demonstrate a critical role of the PI3K/Akt pathway in apoA-IV's anorectic action in lean rats and suggest a defective PI3K pathway in the VMH is responsible for the impaired apoA-IV's anorectic action in the DIO animals.


Asunto(s)
Apolipoproteínas A/metabolismo , Depresores del Apetito/metabolismo , Hipotálamo/metabolismo , Animales , Apolipoproteínas A/administración & dosificación , Depresores del Apetito/administración & dosificación , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos
10.
Horm Behav ; 93: 62-71, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28506816

RESUMEN

Ghrelin is an important gut-derived hormone with an appetite stimulatory role, while most of the intestinal hormones, including cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 (GLP-1), are appetite-inhibitors. Whether these important peptides with opposing roles on food intake interact to regulate energy balance in fish is currently unknown. The aim of this study was to characterize the putative crosstalk between ghrelin and CCK, PYY and GLP-1 in goldfish (Carassius auratus). We first determined the localization of CCK, PYY and GLP-1 in relation to ghrelin and its main receptor GHS-R1a (growth hormone secretagogue 1a) in the goldfish intestine by immunohistochemistry. Colocalization of ghrelin/GHS-R1a and CCK/PYY/GLP-1 was found primarily in the luminal border of the intestinal mucosa. In an intestinal explant culture, a significant decrease in prepro-cck, prepro-pyy and proglucagon transcript levels was observed after 60min of incubation with ghrelin, which was abolished by preincubation with the GHS-R1a ghrelin receptor antagonist [D-Lys3]-GHRP-6 (except for proglucagon). The protein expression of PYY and GLP-1 was also downregulated by ghrelin. Finally, intraperitoneal co-administration of CCK, PYY or GLP-1 with ghrelin results in no modification of food intake in goldfish. Overall, results of the present study show for the first time in fish that ghrelin exerts repressive effects on enteric anorexigens. It is likely that these interactions mediate the stimulatory effects of ghrelin on feeding and metabolism in fish.


Asunto(s)
Anorexia/metabolismo , Colecistoquinina/metabolismo , Ghrelina/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Intestinos/efectos de los fármacos , Péptido YY/metabolismo , Animales , Apetito/efectos de los fármacos , Depresores del Apetito/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Femenino , Ghrelina/metabolismo , Carpa Dorada , Mucosa Intestinal/metabolismo , Masculino , Oligopéptidos/metabolismo , Precursores de Proteínas/metabolismo , Receptores de Ghrelina/metabolismo
11.
Crit Rev Food Sci Nutr ; 57(10): 2218-2244, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-26252442

RESUMEN

The increasing prevalence of overweight and obesity requires new, effective prevention and treatment strategies. One approach to reduce energy intake is by developing novel foods with increased satiating properties, which may be accomplished by slowing down lipolysis to deliver substrates to the ileum, thereby enhancing natural gut-brain signaling pathways of satiety that are normally induced by meal intake. To develop slow release food additives, their processing in the gastrointestinal tract has to be understood; therefore, we start from a general description of the digestive system and relate that to in vitro modeling, satiety, and lipolytic mechanisms. The effects of physicochemical lipid composition, encapsulation matrix, and interfacial structure on lipolysis are emphasized. We give an overview of techniques and materials used, and discuss partitioning, which may be a key factor for encapsulation performance. Targeted release capsules that delay lipolysis form a real challenge because of the high efficiency of the digestive system; hardly any proof was found that intact orally ingested lipids can be released in the ileum and thereby induce satiety. We expect that this challenge could be tackled with structured o/w-emulsion-based systems that have some protection against lipase, e.g., by hindering bile salt adsorption and/or delaying lipase diffusion.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Digestión , Regulación hacia Abajo , Alimentos Especializados , Lipólisis , Modelos Biológicos , Sobrepeso/dietoterapia , Animales , Depresores del Apetito/administración & dosificación , Depresores del Apetito/metabolismo , Depresores del Apetito/uso terapéutico , Grasas de la Dieta/metabolismo , Grasas de la Dieta/uso terapéutico , Emulsiones , Ingestión de Energía , Aditivos Alimentarios/metabolismo , Aditivos Alimentarios/uso terapéutico , Tecnología de Alimentos/tendencias , Humanos , Absorción Intestinal , Sobrepeso/metabolismo , Respuesta de Saciedad
12.
Bioorg Med Chem Lett ; 27(20): 4626-4629, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28935264

RESUMEN

Neuromedin U (NMU) mediates various physiological functions via NMUR1 and NMUR2 receptors. NMUR2 has been considered a promising treatment option for diabetes and obesity. Although NMU-8, a shorter peptide, has potent agonist activity for both receptors, it is metabolically unstable. Therefore, NMU-8 analogs modified with long-chain alkyl moieties via a linker were synthesized. An octadecanoyl analog (17) with amino acid substitutions [αMePhe19, Nle21, and Arg(Me)24] and a linker [Tra-γGlu-PEG(2)] dramatically increased NMUR2 selectivity, with retention of high agonist activity. Subcutaneous administration of 17 induced anorectic activity in C57BL/6J mice. Owing to its high metabolic stability, 17 would be useful in clarifying the physiological role and therapeutic application of NMU.


Asunto(s)
Depresores del Apetito/metabolismo , Péptidos/metabolismo , Receptores de Neurotransmisores/metabolismo , Alquilación , Secuencia de Aminoácidos , Animales , Depresores del Apetito/química , Depresores del Apetito/farmacología , Ingestión de Alimentos/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/agonistas , Receptores de Neurotransmisores/antagonistas & inhibidores , Relación Estructura-Actividad
13.
Diabetes Obes Metab ; 15(2): 99-111, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22862822

RESUMEN

Twenty years of research established amylin as an important control of energy homeostasis. Amylin controls nutrient and energy fluxes by reducing energy intake, by modulating nutrient utilization via an inhibition of postprandial glucagon secretion and by increasing energy disposal via a prevention of compensatory decreases of energy expenditure in weight reduced individuals. Like many other gastrointestinal hormones, amylin is secreted in response to meals and it reduces eating by promoting meal-ending satiation. Not surprisingly, amylin interacts with many of these hormones to control eating. These interactions seem to occur at different levels because amylin seems to mediate the eating inhibitory effect of some of these gastrointestinal hormones, and the combination of some of these hormones seems to lead to a stronger reduction in eating than single hormones alone. Amylin's effect on eating is thought to be mediated by a stimulation of specific amylin receptors in the area postrema. Secondary brain sites that were defined to mediate amylin action - and hence potential additional sites of interaction with other hormones - include the nucleus of the solitary tract, the lateral parabrachial nucleus, the lateral hypothalamic area and other hypothalamic nuclei. The focus of this review is to summarize the current knowledge of amylin interactions in the control of eating. In most cases, these interactions have only been studied at a descriptive rather than a mechanistic level and despite the clear knowledge on primary sites of amylin action, the interaction sites between amylin and other hormones are often unknown.


Asunto(s)
Depresores del Apetito/metabolismo , Área Postrema/metabolismo , Colecistoquinina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Péptido YY/metabolismo , Adiposidad , Interacciones Farmacológicas , Metabolismo Energético , Estradiol/metabolismo , Conducta Alimentaria , Femenino , Homeostasis , Humanos , Hipotálamo/metabolismo , Masculino , Fragmentos de Péptidos , Saciedad
14.
Br J Nutr ; 109(7): 1338-48, 2013 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-22850280

RESUMEN

Galacto-oligosaccharides (GOS) are carbohydrates that are fermented by colonic microbiota. The present study examined effects of a 3-week dietary enrichment with 6 % (w/w) GOS on parameters of energy balance in forty-three male Wistar rats. GOS was tested with two doses of calcium phosphate (30 and 100 mmol/kg), known to differently affect colonic fermentation. After 17 d, isoenergetic test meals were presented and plasma responses of ghrelin, glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) were measured. On day 21 (study termination) epididymal fat pads and caecum were weighed. Additionally, gastrointestinal mucosal samples and proximal colonic contents were analysed for gene expression (ghrelin, proglucagon and PYY) and fermentation metabolites (SCFA and lactate), respectively. GOS reduced energy intake most prominently during the first week, without provoking compensatory overeating later on (average intake reduction: 14 %). The GOS-fed rats showed increased caecal and reduced fat-pad weight and increased gene expression of the satiety-related peptides, PYY (1.7-fold) and proglucagon (3.5-fold). Pre-meal baseline and post-meal plasma levels of PYY, but not of ghrelin or GLP-1, were higher in GOS-fed rats than in control rats. Ca enrichment resulted in higher energy intake (average 4.5 %). GOS diets increased lactic acid levels and slightly reduced butyric acid in proximal colonic contents. Ca abolished the GOS-related elevation of lactic acid, while increasing propionic acid levels, but did not inhibit GOS-related effects on energy intake, fat-pad weight or gene expression. These results indicate that dietary GOS stimulate a number of physiological mechanisms that can reduce energy intake, regardless of the calcium phosphate content of the diet.


Asunto(s)
Calcio de la Dieta/metabolismo , Fibras de la Dieta/metabolismo , Mucosa Gástrica/metabolismo , Hormonas Gastrointestinales/metabolismo , Mucosa Intestinal/metabolismo , Oligosacáridos/metabolismo , Respuesta de Saciedad , Animales , Depresores del Apetito/química , Depresores del Apetito/metabolismo , Fosfatos de Calcio/administración & dosificación , Calcio de la Dieta/administración & dosificación , Fibras de la Dieta/análisis , Ingestión de Energía , Fermentación , Galactosa/química , Galactosa/metabolismo , Hormonas Gastrointestinales/sangre , Hormonas Gastrointestinales/genética , Regulación de la Expresión Génica , Mucosa Intestinal/microbiología , Masculino , Oligosacáridos/química , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Organismos Libres de Patógenos Específicos , Aumento de Peso
15.
Br J Nutr ; 109(7): 1330-7, 2013 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-22850326

RESUMEN

The objective was to determine the effects of dietary fibre with bulking, viscous and gel-forming properties on satiation, and to identify the underlying mechanisms. We conducted a randomised crossover study with 121 men and women. Subjects were healthy, non-restrained eaters, aged 18-50 years and with normal BMI (18.5-25 kg/m²). Test products were cookies containing either: no added fibre (control), cellulose (bulking, 5 g/100 g), guar gum (viscous, 1.25 g/100 g and 2.5 g/100 g) or alginate (gel forming, 2.5 g/100 g and 5 g/100 g). Physico-chemical properties of the test products were confirmed in simulated upper gastrointestinal conditions. In a cinema setting, ad libitum intake of the test products was measured concurrently with oral exposure time per cookie by video recording. In a separate study with ten subjects, 4 h gastric emptying rate of a fixed amount of test products was assessed by ¹³C breath tests. Ad libitum energy intake was 22 % lower for the product with 5 g/100 g alginate (3.1 (sd 1.6) MJ) compared to control (4.0 (sd 2.2) MJ, P< 0.001). Intake of the other four products did not differ from control. Oral exposure time for the product with 5 g/100 g alginate (2.3 (sd 1.9) min) was 48 % longer than for control (1.6 (sd 0.9) min, P= 0.01). Gastric emptying of the 5 g/100 g alginate product was faster compared to control (P< 0.05). We concluded that the addition of 5 g/100 g alginate (i.e. gel-forming fibre) to a low-fibre cookie results in earlier satiation. This effect might be due to an increased oral exposure time.


Asunto(s)
Depresores del Apetito/metabolismo , Fibras de la Dieta/metabolismo , Alimentos Fortificados , Tracto Gastrointestinal/metabolismo , Respuesta de Saciedad , Adolescente , Adulto , Alginatos/química , Alginatos/metabolismo , Depresores del Apetito/química , Celulosa/química , Celulosa/metabolismo , Estudios Cruzados , Fibras de la Dieta/análisis , Femenino , Alimentos Fortificados/análisis , Galactanos/química , Galactanos/metabolismo , Vaciamiento Gástrico , Geles , Ácido Glucurónico/química , Ácido Glucurónico/metabolismo , Ácidos Hexurónicos/química , Ácidos Hexurónicos/metabolismo , Humanos , Masculino , Mananos/química , Mananos/metabolismo , Persona de Mediana Edad , Países Bajos , Gomas de Plantas/química , Gomas de Plantas/metabolismo , Método Simple Ciego , Encuestas y Cuestionarios , Viscosidad , Adulto Joven
16.
Xenobiotica ; 43(2): 211-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22830954

RESUMEN

Plasma concentrations of sibutramine and its two active metabolites after single oral dose of sibutramine were determined in Korean healthy male subjects with different CYP2B6 genotypes (CYP2B6*1/*1, *1/*6 and *6/*6), either alone or after four-day pretreatment with clopidogrel or clarithromycin. The pretreatment with clopidogrel and clarithromycin raised the mean area under the concentration-time curve (AUC) of sibutramine by 163% and 255%, respectively. Co-administration of clarithromycin, combined with CYP2B6*6/*6 genotype, led to highest concentration of sibutramine. The molar sum AUC (M1 + M2) was raised by 35% in the clopidogrel phase but not significantly affected by clarithromycin or CYP2B6 genotype. The CYP2B6*6/*6 subjects in the clopidogrel phase showed the highest molar AUC (M1 + M2) among three genotype groups throughout the three phases. The exposure of sibutramine and its metabolites seemed to be associated with the CYP2B6 genotype. The treatment of clopidogrel significantly altered the disposition of active metabolites as well as sibutramine, but clarithromycin only affects the disposition of sibutramine. These results suggest that the perturbation of CYP2B6 activity may contribute to the inter-individual variation of sibutramine drug responses although the clinical relevance is remained to be established.


Asunto(s)
Depresores del Apetito/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Claritromicina/farmacología , Ciclobutanos/metabolismo , Oxidorreductasas N-Desmetilantes/genética , Ticlopidina/análogos & derivados , Antibacterianos/farmacología , Depresores del Apetito/farmacocinética , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/metabolismo , Pueblo Asiatico , Clopidogrel , Ciclobutanos/farmacocinética , Citocromo P-450 CYP2B6 , Humanos , Masculino , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , Oxidorreductasas N-Desmetilantes/metabolismo , Inhibidores de Agregación Plaquetaria/farmacología , Polimorfismo Genético , República de Corea , Ticlopidina/farmacología
17.
Biochim Biophys Acta ; 1811(9): 508-12, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21723414

RESUMEN

N-acyl-phosphatidylethanolamine is a precursor phospholipid for anandamide, oleoylethanolamide, and other N-acylethanolamines, and it may in itself have biological functions in cell membranes. Recently, N-palmitoyl-phosphatidylethanolamine (NAPE) has been reported to function as an anorectic hormone secreted from the gut and acting on the brain (Gillum et al., [5]). In the current study, two of our laboratories independently investigated whether NAPE metabolites may be involved in mediating the anorectic action of NAPE i.p. injected in mice. Thus, the anorectic activity of a non-hydrolysable NAPE analogue, having ether bonds instead of ester bonds at sn1 and sn2 was compared with that of NAPE in molar equivalent doses. Furthermore, the anorectic effect of NAPE in NAPE-hydrolysing phospholipase D knockout animals was investigated. As negative controls, the NAPE precursor phosphatidylethanolamine and the related phospholipids phosphatidylcholine and phosphatidic acid were also tested. All compounds except one were found to inhibit food intake, raising the possibility that the effect of NAPE is non-specific.


Asunto(s)
Depresores del Apetito/farmacología , Ingestión de Alimentos/efectos de los fármacos , Fosfatidiletanolaminas/farmacología , Animales , Depresores del Apetito/química , Depresores del Apetito/metabolismo , Conducta Animal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Actividad Motora/efectos de los fármacos , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Fosfolipasa D/genética , Fosfolipasa D/metabolismo
18.
Am J Physiol Endocrinol Metab ; 303(12): E1408-18, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23074241

RESUMEN

Many peptides and other compounds that influence metabolism also influence food intake, and numerous hypotheses explaining the observed effects in terms of energy homeostasis have been suggested over the years. For example, cholecystokinin (CCK), a duodenal peptide secreted during meals that aids in digestion, also reduces ongoing food intake, thereby contributing to satiation; and insulin and leptin, hormones secreted in direct proportion to body fat, act in the brain to help control adiposity by reducing energy intake. These behavioral actions are often considered to be hard-wired, such that negative experiments, in which an administered compound fails to have its purported effect, are generally disregarded. In point of fact, failures to replicate the effects of compounds on food intake are commonplace, and this occurs both between and within laboratories. Failures to replicate have historically fueled heated debate about the efficacy and/or normal function of one or another compound, leading to confusion and ambiguity in the literature. We review these phenomena and their implications and argue that, rather than eliciting hard-wired behavioral responses in the maintenance of homeostasis, compounds that alter food intake are subjected to numerous influences that can render them completely ineffective at times and that a major reason for this variance is that food intake is not under stringent homeostatic control.


Asunto(s)
Dieta , Ingestión de Energía , Homeostasis , Respuesta de Saciedad , Animales , Depresores del Apetito/metabolismo , Depresores del Apetito/uso terapéutico , Estimulantes del Apetito/metabolismo , Estimulantes del Apetito/uso terapéutico , Dieta/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Reproducibilidad de los Resultados , Respuesta de Saciedad/efectos de los fármacos
19.
Br J Nutr ; 108(4): 755-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22313587

RESUMEN

Long-chain fatty acids have been shown to suppress appetite and reduce energy intake (EI) by stimulating the release of gastrointestinal hormones such as cholecystokinin (CCK). The effect of NEFA acyl chain length on these parameters is not comprehensively understood. An in vitro screen tested the capacity of individual NEFA (C12 to C22) to trigger CCK release. There was a gradient in CCK release with increasing chain length. DHA (C22) stimulated significantly (P < 0.01) more CCK release than all other NEFA tested. Subsequently, we conducted a randomised, controlled, crossover intervention study using healthy males (n 18). The effects of no treatment (NT) and oral doses of emulsified DHA-rich (DHA) and oleic acid (OA)-rich oils were compared using 24 h EI as the primary endpoint. Participants reported significantly (P = 0.039) lower total daily EI (29 % reduction) with DHA compared to NT. There were no differences between DHA compared to OA and OA compared to NT. There was no between-treatment difference in the time to, or EI of, the first post-intervention eating occasion. It is concluded that NEFA stimulate CCK release in a chain length-dependent manner up to C22. These effects may be extended to the in vivo setting, as a DHA-based emulsion significantly reduced short-term EI.


Asunto(s)
Depresores del Apetito/metabolismo , Colecistoquinina/metabolismo , Suplementos Dietéticos , Ácidos Docosahexaenoicos/metabolismo , Ingestión de Energía , Células Enteroendocrinas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Adulto , Depresores del Apetito/química , Línea Celular Tumoral , Estudios Cruzados , Ácidos Docosahexaenoicos/química , Emulsiones , Ácidos Grasos no Esterificados/química , Humanos , Masculino , Persona de Mediana Edad , Peso Molecular , Método Simple Ciego , Adulto Joven
20.
Anal Bioanal Chem ; 403(5): 1385-94, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22460079

RESUMEN

Owing to the tight control of methamphetamine, it is presumed that phentermine, an amphetamine-type anorectic, has recently been considered a supplement for methamphetamine abusers in Korea. In addition, the abuse of other anorectics obtained by inappropriate means has become a social issue. Hair is a useful specimen to prove chronic drug use. Therefore, an analytical method for the simultaneous detection of phentermine, phendimetrazine, amfepramone, fenfluramine, mazindol, methamphetamine, and 3,4-methylenedioxymethamphetamine (MDMA), as well as their metabolites, which covers the major amphetamines and anorectic agents in Korea, in hair was established and validated using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The drugs and their metabolites in hair were extracted using 1 % HCl in methanol and then filtered and analyzed by LC-MS/MS with electrospray ionization in positive mode. The validation results for selectivity, linearity, matrix effect, recovery, process efficiency, intra- and interassay precision and accuracy, and processed sample stability were satisfactory. The limits of detection ranged from 0.025 to 1 ng/10 mg hair and the limits of quantification were 0.25 ng/10 mg hair for every analyte except mazindol and phentermine, for which they were 10 ng/10 mg hair. The method was successfully applied for the segmental determination of selected anorectics, methamphetamine, MDMA, and their metabolites in hair from 39 drug suspects. Among the anorectics, phentermine and/or phendimetrazine were identified with or without methamphetamine in the hair samples. Closer supervision of the inappropriate use of anorectics is necessary. Also, hair analysis is useful for monitoring the abuse potential of unnoticed drugs.


Asunto(s)
Anfetaminas/análisis , Depresores del Apetito/análisis , Cromatografía Liquida/métodos , Cabello/química , Metanfetamina/análisis , N-Metil-3,4-metilenodioxianfetamina/análisis , Detección de Abuso de Sustancias/métodos , Espectrometría de Masas en Tándem/métodos , Anfetaminas/metabolismo , Depresores del Apetito/metabolismo , Cabello/metabolismo , Humanos , Metanfetamina/metabolismo , N-Metil-3,4-metilenodioxianfetamina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA