Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Peptides ; 140: 170532, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33744371

RESUMEN

OBJECTIVES: To analyse the peptidomics of mouse enteroendocrine cells (EECs) and human gastrointestinal (GI) tissue and identify novel gut derived peptides. METHODS: High resolution nano-flow liquid chromatography mass spectrometry (LC-MS/MS) was performed on (i) flow-cytometry purified NeuroD1 positive cells from mouse and homogenised human intestinal biopsies, (ii) supernatants from primary murine intestinal cultures, (iii) intestinal homogenates from mice fed high fat diet. Candidate bioactive peptides were selected on the basis of species conservation, high expression/biosynthesis in EECs and evidence of regulated secretionin vitro. Candidate novel gut-derived peptides were chronically administered to mice to assess effects on food intake and glucose tolerance. RESULTS: A large number of peptide fragments were identified from human and mouse, including known full-length gut hormones and enzymatic degradation products. EEC-specific peptides were largely from vesicular proteins, particularly prohormones, granins and processing enzymes, of which several exhibited regulated secretion in vitro. No regulated peptides were identified from previously unknown genes. High fat feeding particularly affected the distal colon, resulting in reduced peptide levels from GCG, PYY and INSL5. Of the two candidate novel peptides tested in vivo, a peptide from Chromogranin A (ChgA 435-462a) had no measurable effect, but a progastrin-derived peptide (Gast p59-79), modestly improved glucose tolerance in lean mice. CONCLUSION: LC-MS/MS peptidomic analysis of murine EECs and human GI tissue identified the spectrum of peptides produced by EECs, including a potential novel gut hormone, Gast p59-79, with minor effects on glucose tolerance.


Asunto(s)
Células Enteroendocrinas/metabolismo , Gastrinas/farmacología , Tracto Gastrointestinal/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Péptidos/metabolismo , Precursores de Proteínas/farmacología , Proteoma/metabolismo , Delgadez/tratamiento farmacológico , Animales , Células Cultivadas , Glucosa/metabolismo , Humanos , Masculino , Ratones , Modelos Animales , Péptidos/química , Proteoma/análisis , Delgadez/metabolismo
2.
Nat Commun ; 11(1): 1078, 2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-32081871

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
FASEB J ; 34(1): 1846-1858, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914635

RESUMEN

P53 has been implicated in the pathogenesis of obesity and diabetes; however, the mechanisms and tissue sites of action are incompletely defined. Therefore, we investigated the role of hepatocyte p53 in metabolic homeostasis using a hepatocyte-specific p53 knockout mouse model. To gain further mechanistic insight, we studied mice under two complementary conditions of restricted weight gain: vertical sleeve gastrectomy (VSG) or food restriction. VSG or sham surgery was performed in high-fat diet-fed male hepatocyte-specific p53 wild-type and knockout littermates. Sham-operated mice were fed ad libitum or food restricted to match their body weight to VSG-operated mice. Hepatocyte-specific p53 ablation in sham-operated ad libitum-fed mice impaired glucose homeostasis, increased body weight, and decreased energy expenditure without changing food intake. The metabolic deficits induced by hepatocyte-specific p53 ablation were corrected, in part by food restriction, and completely by VSG. Unlike food restriction, VSG corrected the effect of hepatocyte p53 ablation to lower energy expenditure, resulting in a greater improvement in glucose homeostasis compared with food restricted mice. These data reveal an important new role for hepatocyte p53 in the regulation of energy expenditure and body weight and suggest that VSG can improve alterations in energetics associated with p53 dysregulation.


Asunto(s)
Hepatocitos/metabolismo , Enfermedades Metabólicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Restricción Calórica/métodos , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Alimentos , Gastrectomía/métodos , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Aumento de Peso/fisiología , Pérdida de Peso
4.
Nat Commun ; 10(1): 1546, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948720

RESUMEN

The insulin/IGF-1 signalling pathway is a key regulator of metabolism and the rate of ageing. We previously documented that systemic inactivation of phosphoinositide 3-kinase (PI3K) p110α, the principal PI3K isoform that positively regulates insulin signalling, results in a beneficial metabolic effect in aged mice. Here we demonstrate that deletion of p110α specifically in the adipose tissue leads to less fat accumulation over a significant part of adult life and allows the maintenance of normal glucose tolerance despite insulin resistance. This effect of p110α inactivation is due to a potentiating effect on ß-adrenergic signalling, which leads to increased catecholamine-induced energy expenditure in the adipose tissue. Our findings provide a paradigm of how partial inactivation of an essential component of the insulin signalling pathway can have an overall beneficial metabolic effect and suggest that PI3K inhibition could potentiate the effect of ß-adrenergic agonists in the treatment of obesity and its associated comorbidities.


Asunto(s)
Tejido Adiposo/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/fisiología , Factores de Edad , Animales , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Resistencia a la Insulina/genética , Ratones Transgénicos , Obesidad/metabolismo , Transducción de Señal
5.
Cell Rep ; 26(6): 1399-1408.e6, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30726726

RESUMEN

Bariatric surgery is widely used to treat obesity and improves type 2 diabetes beyond expectations from the degree of weight loss. Elevated post-prandial concentrations of glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and insulin are widely reported, but the importance of GLP-1 in post-bariatric physiology remains debated. Here, we show that GLP-1 is a major driver of insulin secretion after bariatric surgery, as demonstrated by blocking GLP-1 receptors (GLP1Rs) post-gastrectomy in lean humans using Exendin-9 or in mice using an anti-GLP1R antibody. Transcriptomics and peptidomics analyses revealed that human and mouse enteroendocrine cells were unaltered post-surgery; instead, we found that elevated plasma GLP-1 and PYY correlated with increased nutrient delivery to the distal gut in mice. We conclude that increased GLP-1 secretion after bariatric surgery arises from rapid nutrient delivery to the distal gut and is a key driver of enhanced insulin secretion.


Asunto(s)
Cirugía Bariátrica , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Homeostasis , Obesidad/metabolismo , Adulto , Animales , Células Enteroendocrinas/metabolismo , Femenino , Péptido 1 Similar al Glucagón/sangre , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/uso terapéutico , Secreción de Insulina , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/tratamiento farmacológico , Obesidad/cirugía , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/uso terapéutico , Péptido YY/metabolismo , Periodo Posoperatorio , Transcriptoma
6.
Dis Model Mech ; 10(3): 235-243, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28093508

RESUMEN

Bariatric surgery, such as vertical sleeve gastrectomy (VSG), causes remarkable improvements in cardiometabolic health, including hypertension remission. However, the mechanisms responsible remain undefined and poorly studied. Therefore, we developed and validated the first murine model of VSG that recapitulates the blood pressure-lowering effect of VSG using gold-standard radiotelemetry technology. We used this model to investigate several potential mechanisms, including body mass, brain endoplasmic reticulum (ER) stress signaling and brain inflammatory signaling, which are all critical contributors to the pathogenesis of obesity-associated hypertension. Mice fed on a high-fat diet underwent sham or VSG surgery and radiotelemeter implantation. Sham mice were fed ad libitum or were food restricted to match their body mass to VSG-operated mice to determine the role of body mass in the ability of VSG to lower blood pressure. Blood pressure was then measured in freely moving unstressed mice by radiotelemetry. VSG decreased energy intake, body mass and fat mass. Mean arterial blood pressure (MAP) was reduced in VSG-operated mice compared with both sham-operated groups. VSG-induced reductions in MAP were accompanied by a body mass-independent decrease in hypothalamic ER stress, hypothalamic inflammation and sympathetic nervous system tone. Assessment of gut microbial populations revealed VSG-induced increases in the relative abundance of Gammaproteobacteria and Enterococcus, and decreases in Adlercreutzia These results suggest that VSG reduces blood pressure, but this is only partly due to the reduction in body weight. VSG-induced reductions in blood pressure may be driven by a decrease in hypothalamic ER stress and inflammatory signaling, and shifts in gut microbial populations.


Asunto(s)
Presión Sanguínea , Estrés del Retículo Endoplásmico , Gastrectomía , Hipotálamo/patología , Animales , Peso Corporal , Ciego/microbiología , Ingestión de Energía , Ayuno/sangre , Microbioma Gastrointestinal , Ghrelina/sangre , Inflamación/patología , Leptina/sangre , Masculino , Ratones Endogámicos C57BL , Norepinefrina/sangre , Transducción de Señal
7.
Gut ; 66(2): 226-234, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-26511794

RESUMEN

OBJECTIVE: Vertical sleeve gastrectomy (VSG) produces high rates of type 2 diabetes remission; however, the mechanisms responsible remain incompletely defined. VSG increases circulating bile acid concentrations and bile acid signalling through TGR5 improves glucose homeostasis. Therefore, we investigated the role of TGR5 signalling in mediating the glucoregulatory benefits of VSG. DESIGN: VSG or sham surgery was performed in high-fat-fed male Tgr5+/+ (wild type) and Tgr5-/- (knockout) littermates. Sham-operated mice were fed ad libitum or food restricted to match their body weight to VSG-operated mice. Body weight, food intake, energy expenditure, insulin signalling and circulating bile acid profiles were measured and oral glucose tolerance testing, islet immunohistochemistry and gut microbial profiling were performed. RESULTS: VSG decreased food intake and body weight, increased energy expenditure and circulating bile acid concentrations, improved fasting glycaemia, glucose tolerance and glucose-stimulated insulin secretion, enhanced nutrient-stimulated glucagon-like peptide 1 secretion and produced favourable shifts in gut microbial populations in both genotypes. However, the body weight-independent improvements in fasting glycaemia, glucose tolerance, hepatic insulin signalling, hepatic inflammation and islet morphology after VSG were attenuated in Tgr5-/- relative to Tgr5+/+ mice. Furthermore, VSG produced metabolically favourable alterations in circulating bile acid profiles that were blunted in Tgr5-/- relative to Tgr5+/+ mice. TGR5-dependent regulation of hepatic Cyp8b1 expression may have contributed to TGR5-mediated shifts in the circulating bile acid pool after VSG. CONCLUSIONS: These results suggest that TGR5 contributes to the glucoregulatory benefits of VSG surgery by promoting metabolically favourable shifts in the circulating bile acid pool.


Asunto(s)
Ácidos y Sales Biliares/sangre , Glucemia/metabolismo , Gastrectomía , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Ayuno , Gastrectomía/métodos , Microbioma Gastrointestinal , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Islotes Pancreáticos/química , Islotes Pancreáticos/patología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Esteroide 12-alfa-Hidroxilasa/metabolismo
8.
Endocrinology ; 157(9): 3405-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27501183

RESUMEN

Vertical sleeve gastrectomy (VSG) produces high rates of type 2 diabetes remission; however, the mechanisms responsible for this remain incompletely defined. Glucagon-like peptide-1 (GLP-1) is a gut hormone that contributes to the maintenance of glucose homeostasis and is elevated after VSG. VSG-induced increases in postprandial GLP-1 secretion have been proposed to contribute to the glucoregulatory benefits of VSG; however, previous work has been equivocal. In order to test the contribution of enhanced ß-cell GLP-1 receptor (GLP-1R) signaling we used a ß-cell-specific tamoxifen-inducible GLP-1R knockout mouse model. Male ß-cell-specific Glp-1r(ß-cell+/+) wild type (WT) and Glp-1r(ß-cell-/-) knockout (KO) littermates were placed on a high-fat diet for 6 weeks and then switched to high-fat diet supplemented with tamoxifen for the rest of the study. Mice underwent sham or VSG surgery after 2 weeks of tamoxifen diet and were fed ad libitum postoperatively. Mice underwent oral glucose tolerance testing at 3 weeks and were euthanized at 6 weeks after surgery. VSG reduced body weight and food intake independent of genotype. However, glucose tolerance was only improved in VSG WT compared with sham WT, whereas VSG KO had impaired glucose tolerance relative to VSG WT. Augmentation of glucose-stimulated insulin secretion during the oral glucose tolerance test was blunted in VSG KO compared with VSG WT. Therefore, our data suggest that enhanced ß-cell GLP-1R signaling contributes to improved glucose regulation after VSG by promoting increased glucose-stimulated insulin secretion.


Asunto(s)
Gastrectomía , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Trastornos del Metabolismo de la Glucosa/cirugía , Células Secretoras de Insulina/metabolismo , Animales , Peso Corporal , Ingestión de Alimentos , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones Noqueados , Tamoxifeno
9.
Obesity (Silver Spring) ; 23(6): 1194-200, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25958858

RESUMEN

OBJECTIVE: The G-protein coupled receptor family C group 6 member A (GPRC6A) is activated by proteinogenic amino acids and may sense amino acids in the gastrointestinal tract and the brain. The study investigated whether GPRC6A was necessary for the effects of low- and high-protein diets on body weight and food intake in mice. METHODS: The role of GPRC6A in mediating the effects of a low-protein diet on body weight was investigated in GPRC6a knockout (GPRC6a-KO) and wild-type (WT) mice fed a control diet (18% protein) or a low-protein diet (6% protein) for 9 days. The role of GPRC6A in mediating the effects of a high-protein diet on body weight was investigated in GPRC6a-KO and WT mice fed a control diet (18% protein) or a high-protein diet (50% protein) for 5 weeks. RESULTS: A high-protein diet reduced body weight gain and food intake compared with a control diet in both WT and GPRC6a-KO mice. A low-protein diet decreased body weight gain in GPRC6a-KO mice. CONCLUSIONS: GPRC6A was not necessary for the effects of a low- or high-protein diet on body weight and likely does not play a role in protein-induced satiety.


Asunto(s)
Peso Corporal/efectos de los fármacos , Proteínas en la Dieta/administración & dosificación , Receptores Acoplados a Proteínas G/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Aumento de Peso/efectos de los fármacos
10.
Br J Clin Pharmacol ; 74(6): 911-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22452339

RESUMEN

Obesity is a major worldwide health problem. The treatment options are severely limited. The development of novel anti-obesity drugs is fraught with efficacy and safety issues. Consequently, several investigational anti-obesity drugs have failed to gain marketing approval in recent years. Anorectic gut hormones offer a potentially safe and viable option for the treatment of obesity. The prospective utility of gut hormones has improved drastically in recent years with the development of longer acting analogues. Additionally, specific combinations of gut hormones have been demonstrated to have additive anorectic effects. This article reviews the current stage of anti-obesity drugs in development, focusing on gut hormone-based therapies.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Hormonas Gastrointestinales/uso terapéutico , Obesidad/tratamiento farmacológico , Tracto Gastrointestinal/efectos de los fármacos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA