Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nutr Rev ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926118

RESUMEN

CONTEXT: Pregnancy increases nutrient demand, but how nutrient uptake and its determinants adapt to facilitate this is unclear. OBJECTIVE: This review aimed to identify and characterize evidence and evidence gaps regarding changes in gastrointestinal nutrient absorption and its determinants during pregnancy in monogastric mammals. DATA SOURCES: A scoping review of peer-reviewed sources was conducted across PubMed, Scopus, Web of Science, Embase, and ProQuest (theses and dissertations) databases. DATA EXTRACTION: Data extracted included species, pregnancy stages and outcomes. Where sufficient data for a given outcome was available, relative values were summarized graphically or in tables, to allow comparison across pregnancy stages and/or small intestine regions. Searches identified 26 855 sources, of which only 159 were eligible. Mechanistic studies were largely restricted to rodents, and most compared non- and late-pregnant groups, with fewer studies including early- or mid-pregnant groups. DATA ANALYSIS: During pregnancy, there is some evidence for greater capacity for glucose uptake but unchanged amino acid uptake, and good evidence for increased uptake of calcium, iron, and zinc, and slower gastrointestinal passage of nutrients. The available evidence indicates that acute glucose uptake, gastric emptying, and the activities of sucrase, maltase, and lactase do not change during pregnancy. Gaps in the knowledge include the effects of pregnancy on uptake of specific amino acids, lipids, and most minerals and vitamins. CONCLUSION: The results indicate that the gastrointestinal tract adapts during pregnancy to facilitate increased nutrient absorption. Additional data is required in order to assess the underlying mechanisms for and impacts on the absorption of many nutrients, as well as to determine the timing of these adaptations.

2.
Peptides ; 172: 171114, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37926186

RESUMEN

Small intestinal satiation pathways involve nutrient-induced stimulation of chemoreceptors leading to release of satiety hormones from intestinal enteroendocrine cells (ECCs). Whether adaptations in these pathways contribute to increased maternal food intake during pregnancy is unknown. To determine the expression of intestinal nutrient-sensors and satiety hormone transcripts and proteins across pregnancy in mice. Female C57BL/6J mice (10-12 weeks old) were randomized to mating and then tissue collection at early- (6.5 d), mid- (12.5 d) or late-pregnancy (17.5 d), or to an unmated age matched control group. Relative transcript expression of intestinal fatty acid, peptide and amino acid and carbohydrate chemoreceptors, as well as gut hormones was determined across pregnancy. The density of G-protein coupled receptor 93 (GPR93), free fatty acid receptor (FFAR) 4, cholecystokinin (CCK) and glucagon-like peptide1 (GLP-1) immunopositive cells was then compared between non-pregnant and late-pregnant mice. Duodenal GPR93 expression was lower in late pregnant than non-pregnant mice (P < 0.05). Ileal FFAR1 expression was higher at mid- than at early- or late-pregnancy. Ileal FFAR2 expression was higher at mid-pregnancy than in early pregnancy. Although FFAR4 expression was consistently lower in late-pregnant than non-pregnant mice (P < 0.001), the density of FFAR4 immunopositive cells was higher in the jejunum of late-pregnant than non-pregnant mice. A subset of protein and fatty acid chemoreceptor transcripts undergo region-specific change during murine pregnancy, which could augment hormone release and contribute to increased food intake. Further investigations are needed to determine the functional relevance of these changes.


Asunto(s)
Hormonas Gastrointestinales , Saciedad , Animales , Femenino , Ratones , Embarazo , Colecistoquinina/metabolismo , Ácidos Grasos , Hormonas Gastrointestinales/metabolismo , Ratones Endogámicos C57BL , Nutrientes , Saciedad/fisiología
3.
Exp Physiol ; 108(6): 865-873, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37022128

RESUMEN

NEW FINDINGS: What is the central question of this study? Body mass and food intake change during the female ovarian cycle: does glucose transport by the small intestine also vary? What is the main finding and its importance? We have optimised Ussing chamber methodology to measure region-specific active glucose transport in the small intestine of adult C57BL/6 mice. Our study provides the first evidence that jejunal active glucose transport changes during the oestrous cycle in mice, and is higher at pro-oestrus than oestrus. These results demonstrate adaptation in active glucose uptake, concurrent with previously reported changes in food intake. ABSTRACT: Food intake changes across the ovarian cycle in rodents and humans, with a nadir during the pre-ovulatory phase and a peak during the luteal phase. However, it is unknown whether the rate of intestinal glucose absorption also changes. We therefore mounted small intestinal sections from C57BL/6 female mice (8-9 weeks old) in Ussing chambers and measured active ex vivo glucose transport via the change in short-circuit current (∆Isc ) induced by glucose. Tissue viability was confirmed by a positive ∆Isc response to 100 µM carbachol following each experiment. Active glucose transport, assessed after addition of 5, 10, 25 or 45 mM d-glucose to the mucosal chamber, was highest at 45 mM glucose in the distal jejunum compared to duodenum and ileum (P < 0.01). Incubation with the sodium-glucose cotransporter 1 (SGLT1) inhibitor phlorizin reduced active glucose transport in a dose-dependent manner in all regions (P < 0.01). Active glucose uptake induced by addition of 45 mM glucose to the mucosal chamber in the absence or presence of phlorizin was assessed in jejunum at each oestrous cycle stage (n = 9-10 mice per stage). Overall, active glucose uptake was lower at oestrus compared to pro-oestrus (P = 0.025). This study establishes an ex vivo method to measure region-specific glucose transport in the mouse small intestine. Our results provide the first direct evidence that SGLT1-mediated glucose transport in the jejunum changes across the ovarian cycle. The mechanisms underlying these adaptations in nutrient absorption remain to be elucidated.


Asunto(s)
Glucosa , Florizina , Humanos , Femenino , Animales , Ratones , Glucosa/metabolismo , Florizina/metabolismo , Ratones Endogámicos C57BL , Intestino Delgado/metabolismo , Yeyuno , Absorción Intestinal , Mucosa Intestinal/metabolismo
4.
Am J Clin Nutr ; 118(1): 68-76, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37061165

RESUMEN

BACKGROUND: Multiple studies have independently investigated the associations of the consumption of individual beverage types and specific plasma biomarkers with the risk of type 2 diabetes (T2D). However, as individuals do not consume single beverage types exclusively and plasma biomarkers do not act in isolation, it remains unclear how patterns of beverage consumption and plasma biomarker networks associate both with each other and T2D risk. OBJECTIVES: We aimed to elucidate potential dietary determinants of T2D risk by defining a model that describes habitual beverage consumption profiles in relation to identified networks of circulating plasma biomarkers. METHODS: This study included 1,461 case and 1,568 control participants from case-control studies of T2D nested within the Nurses' Health Study. Participants completed validated semiquantitative food frequency questionnaires that assessed habitual beverage consumption, and they provided blood samples from which 27 plasma biomarkers of cardiometabolic risk were identified. Common exploratory factor analysis (EFA) identified factors that separately described beverage consumption profiles and biomarker networks. Multivariable-adjusted regression elucidated the relationships between beverage and biomarker factors and T2D risk. RESULTS: EFA revealed five factors describing unique beverage consumption profiles and seven factors describing biomarker networks. The factor describing alcoholic beverage consumption was associated with a reduced risk of T2D (odds ratio [OR]: 0.50 [0.40, 0.64], P<0.001) mediated, in part, by the factor describing increased concentrations of adiponectin biomarkers (19.9% [12.0, 31.1] P = 0.004). The factor describing low-calorie sweetened beverage (LCSBs) consumption was associated with an increased risk of T2D (OR: 1.33 [1.03, 1.72], P = 0.021), and the factor describing lower concentrations of insulin-like growth factor binding proteins 1 and 2, and soluble leptin receptor, and increased leptin concentrations (P = 0.005). CONCLUSIONS: Moderate alcohol consumption was associated with reduced T2D risk, mediated in part by increased circulating adiponectin. LCSB consumption was associated with both increased T2D risk and perturbed insulin-like growth factor and leptin signaling.


Asunto(s)
Diabetes Mellitus Tipo 2 , Leptina , Humanos , Diabetes Mellitus Tipo 2/etiología , Adiponectina , Bebidas/efectos adversos , Biomarcadores , Factores de Riesgo
5.
Diabetes Obes Metab ; 24(6): 1132-1142, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35238131

RESUMEN

AIM: To determine the serum bile acid (BA) response to 75-g oral glucose in individuals without diabetes, and whether this is attenuated in patients with 'early' type 2 diabetes (T2D) and related to the glycaemic response at 2 hours in either group. METHODS: Forty newly diagnosed, treatment-naïve Han Chinese T2D subjects and 40 age-, gender-, and body mass index-matched controls without T2D ingested a 75-g glucose drink after an overnight fast. Plasma glucose and serum concentrations of total and individual BAs, fibroblast growth factor-19 (FGF-19), total glucagon-like peptide-1 (GLP-1), and insulin, were measured before and 2 hours after oral glucose. RESULTS: Fasting total BA levels were higher in T2D than control subjects (P < .05). At 2 hours, the BA profile exhibited a shift from baseline in both groups, with increases in conjugated BAs and/or decreases in unconjugated BAs. There were increases in total BA and FGF-19 levels in control (both P < .05), but not T2D, subjects. Plasma glucose concentrations at 2 hours related inversely to serum total BA levels in control subjects (r = -0.42, P = .006). Total GLP-1 and the insulin/glucose ratio were increased at 2 hours in both groups, and the magnitude of the increase was greater in control subjects. CONCLUSIONS: The serum BA response to a 75-g oral glucose load is attenuated in patients with 'early' T2D, as is the secretion of FGF-19 and GLP-1, while in individuals without T2D it correlates with 2-hour plasma glucose levels. These observations support a role for BAs in the regulation of postprandial glucose metabolism.


Asunto(s)
Glucemia , Diabetes Mellitus Tipo 2 , Ácidos y Sales Biliares , Glucemia/metabolismo , Factores de Crecimiento de Fibroblastos , Péptido 1 Similar al Glucagón , Glucosa/metabolismo , Humanos , Insulina
6.
JBI Evid Synth ; 20(2): 640-646, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35165214

RESUMEN

OBJECTIVE: The aim of this review is to characterize the current state of literature and knowledge regarding adaptations of gastrointestinal nutrient absorption, and the determinants of this absorption during pregnancy in monogastric mammals. INTRODUCTION: Energy demands increase significantly during pregnancy due to the metabolic demands associated with placental and fetal growth, and the deposition of fat stores that support postnatal lactation. Previous studies have examined anatomical changes within the small intestine, but have focused on specific pregnancy stages or specific regions of the small intestine. Importantly, little is known about changes in nutrient absorption during pregnancy, and the underlying mechanisms that lead to these changes. An understanding of these adaptations will inform research to improve pregnancy outcomes for both mothers and newborns in the future. INCLUSION CRITERIA: This review will include primary literature that describes gastrointestinal nutrient absorption and/or its determinants during pregnancy in monogastric mammals, including humans and rodents. Only data for normal pregnancies will be included, and models of pathology and illness will be excluded. Studies must include comparisons between pregnant animals at known stages of pregnancy, and non-pregnant controls, or compare animals at different stages of pregnancy. METHODS: The following databases will be searched for literature on this topic: PubMed, Scopus, Web of Science, Embase, MEDLINE, and ProQuest Dissertations and Theses. Evidence screening and selection will be carried out independently by two reviewers, and conflicts will be resolved through discussion with additional members of the review team. Data will be extracted and presented in tables and/or figures, together with a narrative summary.


Asunto(s)
Placenta , Resultado del Embarazo , Animales , Femenino , Desarrollo Fetal , Humanos , Recién Nacido , Mamíferos , Nutrientes , Embarazo , Literatura de Revisión como Asunto
7.
J Clin Endocrinol Metab ; 107(4): e1643-e1652, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-34791325

RESUMEN

CONTEXT: Both gastric emptying and the secretion of glucagon-like peptide-1 (GLP-1) are major determinants of postprandial glycemia in health and type 2 diabetes (T2D). GLP-1 secretion after a meal is dependent on the entry of nutrients into the small intestine, which, in turn, slows gastric emptying. OBJECTIVE: To define the relationship between gastric emptying and the GLP-1 response to both oral and small intestinal nutrients in subjects with and without T2D. METHODS: We evaluated: (i) the relationship between gastric emptying (breath test) and postprandial GLP-1 levels after a mashed potato meal in 73 individuals with T2D; (ii) inter-individual variations in GLP-1 response to (a) intraduodenal glucose (4 kcal/min) during euglycemia and hyperglycemia in 11 healthy and 12 T2D, subjects, (b) intraduodenal fat (2 kcal/min) in 15 T2D subjects, and (c) intraduodenal protein (3 kcal/min) in 10 healthy subjects; and (iii) the relationship between gastric emptying (breath test) of 75 g oral glucose and the GLP-1 response to intraduodenal glucose (4 kcal/min) in 21 subjects (9 healthy, 12 T2D). RESULTS: The GLP-1 response to the mashed potato meal was unrelated to the gastric half-emptying time (T50). The GLP-1 responses to intraduodenal glucose, fat, and protein varied substantially between individuals, but intra-individual variation to glucose was modest. The T50 of oral glucose was related directly to the GLP-1 response to intraduodenal glucose (r = 0.65, P = 0.002). CONCLUSION: In a given individual, gastric emptying is not a determinant of the postprandial GLP-1 response. However, the intrinsic gastric emptying rate is determined in part by the responsiveness of GLP-1 to intestinal nutrients.


Asunto(s)
Diabetes Mellitus Tipo 2 , Péptido 1 Similar al Glucagón , Glucemia/metabolismo , Vaciamiento Gástrico/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Glucosa , Humanos , Insulina/metabolismo , Nutrientes , Periodo Posprandial
8.
Peptides ; 146: 170673, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34627956

RESUMEN

AIMS: This study investigated the nutrient-mediated modulation of total ghrelin (TG) and acyl ghrelin (AG) secretion from the mouse gastric mucosa, and the role of long-chain fatty acid chemosensors, FFAR4 and CD36, in lipid-mediated modulation of TG and AG release. METHODS: Ex-vivo experiments were conducted using mouse gastric mucosa to examine the effects of nutrients (D-glucose, L-phenylalanine, peptone (mixture of oligopeptides & single amino acids), D-mannitol, α-linolenic acid and fat emulsion (intralipid)) on TG and AG secretion. Additionally, inhibition of FFAR4 and CD36 on α-linolenic acid and intralipid-mediated regulation of TG and AG secretion was assessed. RESULTS: TG and AG secretion were unaffected by glucose and D-mannitol. Peptone stimulated the release of TG and AG. In contrast, L-phenylalanine reduced AG secretion only. Intralipid reduced TG secretion and stimulated AG secretion, and α-linolenic acid reduced AG release, without affecting TG mobilisation. Modulation of ghrelin secretion by lipids occurred in an FFAR4 and CD36-independent manner. CONCLUSION: Ghrelin secretion is modulated in a nutrient-specific manner by proteins and lipids, with TG and AG displaying independent responses to the same stimuli. In addition, FFAR4 and CD36 do not participate in modulation of TG and AG secretion by α-linolenic acid and intralipid.


Asunto(s)
Antígenos CD36/metabolismo , Mucosa Gástrica/metabolismo , Ghrelina/análogos & derivados , Lípidos/análisis , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ghrelina/sangre , Ghrelina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Obesity (Silver Spring) ; 29(11): 1813-1824, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34623766

RESUMEN

A sufficient and balanced maternal diet is critical to meet the nutritional demands of the developing fetus and to facilitate deposition of fat reserves for lactation. Multiple adaptations occur to meet these energy requirements, including reductions in energy expenditure and increases in maternal food intake. The central nervous system plays a vital role in the regulation of food intake and energy homeostasis and responds to multiple metabolic and nutrient cues, including those arising from the gastrointestinal tract. This review describes the nutrient requirements of pregnancy and the impact of over- and undernutrition on the risk of pregnancy complications and adult disease in progeny. The central and peripheral regulation of food intake is then discussed, with particular emphasis on the adaptations that occur during pregnancy and the mechanisms that drive these changes, including the possible role of the pregnancy-associated hormones progesterone, estrogen, prolactin, and growth hormone. We identify the need for deeper mechanistic understanding of maternal adaptations, in particular, changes in gut-brain axis satiety signaling. Improved understanding of food intake regulation during pregnancy will provide a basis to inform strategies that prevent maternal under- or overnutrition, improve fetal health, and reduce the long-term health and economic burden for mothers and offspring.


Asunto(s)
Eje Cerebro-Intestino , Lactancia , Ingestión de Alimentos , Metabolismo Energético , Femenino , Humanos , Necesidades Nutricionales , Embarazo
10.
Nutrients ; 13(4)2021 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-33800566

RESUMEN

Bile acids are cholesterol-derived metabolites with a well-established role in the digestion and absorption of dietary fat. More recently, the discovery of bile acids as natural ligands for the nuclear farnesoid X receptor (FXR) and membrane Takeda G-protein-coupled receptor 5 (TGR5), and the recognition of the effects of FXR and TGR5 signaling have led to a paradigm shift in knowledge regarding bile acid physiology and metabolic health. Bile acids are now recognized as signaling molecules that orchestrate blood glucose, lipid and energy metabolism. Changes in FXR and/or TGR5 signaling modulates the secretion of gastrointestinal hormones including glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), hepatic gluconeogenesis, glycogen synthesis, energy expenditure, and the composition of the gut microbiome. These effects may contribute to the metabolic benefits of bile acid sequestrants, metformin, and bariatric surgery. This review focuses on the role of bile acids in energy intake and body weight, particularly their effects on gastrointestinal hormone secretion, the changes in obesity and T2D, and their potential relevance to the management of metabolic disorders.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Enfermedades Metabólicas/metabolismo , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Hormonas Gastrointestinales/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Gastroenterology ; 161(2): 536-547.e2, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33848536

RESUMEN

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.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Mucosa Intestinal/metabolismo , Péptido YY/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/metabolismo , Comunicación Autocrina , Glucemia/metabolismo , Estudios de Casos y Controles , Células Enteroendocrinas/efectos de los fármacos , Glucosa/administración & dosificación , Prueba de Tolerancia a la Glucosa , Humanos , Mucosa Intestinal/efectos de los fármacos , Mutación con Pérdida de Función , Comunicación Paracrina , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Vías Secretoras , Transducción de Señal , Factores de Tiempo , alfa-MSH/farmacología
12.
Endocrinology ; 162(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33534908

RESUMEN

Glucagon is secreted by pancreatic α cells in response to hypoglycemia and increases hepatic glucose output through hepatic glucagon receptors (GCGRs). There is evidence supporting the notion of extrapancreatic glucagon but its source and physiological functions remain elusive. Intestinal tissue samples were obtained from patients undergoing surgical resection of cancer. Mass spectrometry analysis was used to detect glucagon from mucosal lysate. Static incubations of mucosal tissue were performed to assess glucagon secretory response. Glucagon concentration was quantitated using a highly specific sandwich enzyme-linked immunosorbent assay. A cholesterol uptake assay and an isolated murine colonic motility assay were used to assess the physiological functions of intestinal GCGRs. Fully processed glucagon was detected by mass spectrometry in human intestinal mucosal lysate. High glucose evoked significant glucagon secretion from human ileal tissue independent of sodium glucose cotransporter and KATP channels, contrasting glucose-induced glucagon-like peptide 1 (GLP-1) secretion. The GLP-1 receptor agonist Exendin-4 attenuated glucose-induced glucagon secretion from the human ileum. GCGR blockade significantly increased cholesterol uptake in human ileal crypt culture and markedly slowed ex vivo colonic motility. Our findings describe the human gut as a potential source of extrapancreatic glucagon and demonstrate a novel enteric glucagon/GCGR circuit with important physiological functions beyond glycemic regulation.


Asunto(s)
Glucagón/metabolismo , Mucosa Intestinal/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Colesterol/metabolismo , Estudios de Cohortes , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Masculino , Persona de Mediana Edad
13.
Acta Physiol (Oxf) ; 231(3): e13588, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33249751

RESUMEN

Ghrelin is a gastric hormone with multiple physiological functions, including the stimulation of food intake and adiposity. It is well established that circulating ghrelin levels are closely associated with feeding patterns, rising strongly before a meal and lowering upon food intake. However, the mechanisms underlying the modulation of ghrelin secretion are not fully understood. The purpose of this review is to discuss current knowledge on the circadian oscillation of circulating ghrelin levels, the neural mechanisms stimulating fasting ghrelin levels and peripheral mechanisms modulating postprandial ghrelin levels. Furthermore, the therapeutic potential of targeting the ghrelin pathway is discussed in the context of the treatment of various metabolic disorders, including obesity, type 2 diabetes, diabetic gastroparesis and Prader-Willi syndrome. Moreover, eating disorders including anorexia nervosa, bulimia nervosa and binge-eating disorder are also discussed.


Asunto(s)
Diabetes Mellitus Tipo 2 , Síndrome de Prader-Willi , Ingestión de Alimentos , Ghrelina , Humanos , Obesidad , Periodo Posprandial
14.
Diabetes Res Clin Pract ; 171: 108610, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33301790

RESUMEN

AIM: Gastric emptying is a major determinant of the glycaemic response to carbohydrate and is frequently abnormal in type 2 diabetes (T2DM). There is little information about how chronic glycaemic control affects gastric emptying in T2DM. We evaluated gastric emptying of a 75 g glucose drink in community-based patients with T2DM of short duration with good or poor glycaemic control, and compared this to young and older controls. METHODS: T2DM patients managed by diet and/or metformin, either well-controlled or poorly-controlled, together with young and age-matched older controls without diabetes, consumed a 75 g oral glucose drink containing 150 mg 13C-acetate for evaluation of gastric emptying (breath test) and blood glucose over 180 min. RESULTS: The gastric half-emptying time (T50) was longer in the older than the young non-diabetic subjects (P = 0.041), but shorter in well-controlled T2DM patients than age-matched older controls (P = 0.043). The T50 in poorly-controlled T2DM patients was shorter than in older controls (P = 0.006), but similar to young non-diabetic subjects. CONCLUSIONS: Gastric emptying of a glucose drink is delayed with ageing, but more rapid in patients with T2DM of relatively short duration, regardless of their glycaemic status. These observations support interventions that slow gastric emptying to improve postprandial glycaemia in these patients with T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Vaciamiento Gástrico/fisiología , Prueba de Tolerancia a la Glucosa/métodos , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
15.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G183-G192, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33206550

RESUMEN

Gastric vagal afferents (GVAs) sense food-related mechanical stimuli and signal to the central nervous system, to integrate control of meal termination. Pregnancy is characterized by increased maternal food intake, which is essential for normal fetal growth and to maximize progeny survival and health. However, it is unknown whether GVA function is altered during pregnancy to promote food intake. This study aimed to determine the mechanosensitivity of GVAs and food intake during early, mid-, and late stages of pregnancy in mice. Pregnant mice consumed more food compared with nonpregnant mice, notably in the light phase during mid- and late pregnancy. The increased food intake was predominantly due to light-phase increases in meal size across all stages of pregnancy. The sensitivity of GVA tension receptors to gastric distension was significantly attenuated in mid- and late pregnancy, whereas the sensitivity of GVA mucosal receptors to mucosal stroking was unchanged during pregnancy. To determine whether pregnancy-associated hormonal changes drive these adaptations, the effects of estradiol, progesterone, prolactin, and growth hormone on GVA tension receptor mechanosensitivity were determined in nonpregnant female mice. The sensitivity of GVA tension receptors to gastric distension was augmented by estradiol, attenuated by growth hormone, and unaffected by progesterone or prolactin. Together, the data indicate that the sensitivity of GVA tension receptors to tension is reduced during pregnancy, which may attenuate the perception of gastric fullness and explain increased food intake. Further, these adaptations may be driven by increases in maternal circulating growth hormone levels during pregnancy.NEW & NOTEWORTHY This study provides first evidence that gastric vagal afferent signaling is attenuated during pregnancy and inversely associated with meal size. Growth hormone attenuated mechanosensitivity of gastric vagal afferents, adding support that increases in maternal growth hormone may mediate adaptations in gastric vagal afferent signaling during pregnancy. These findings have important implications for the peripheral control of food intake during pregnancy.


Asunto(s)
Vías Aferentes/fisiología , Plasticidad Neuronal/fisiología , Estómago/inervación , Nervio Vago/fisiología , Animales , Femenino , Ratones , Embarazo
16.
World J Gastroenterol ; 26(25): 3562-3576, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32742126

RESUMEN

The gastrointestinal tract is the key interface between the ingesta and the human body. There is wide recognition that the gastrointestinal response to nutrients or bioactive compounds, particularly the secretion of numerous hormones, is critical to the regulation of appetite, body weight and blood glucose. This concept has led to an increasing focus on "gut-based" strategies for the management of metabolic disorders, including type 2 diabetes and obesity. Understanding the underlying mechanisms and downstream effects of nutrient-gut interactions is fundamental to effective translation of this knowledge to clinical practice. To this end, an array of research tools and platforms have been developed to better understand the mechanisms of gut hormone secretion from enteroendocrine cells. This review discusses the evolution of in vitro and in vivo models and the integration of innovative techniques that will ultimately enable the development of novel therapies for metabolic diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hormonas Gastrointestinales , Microbioma Gastrointestinal , Células Enteroendocrinas , Tracto Gastrointestinal , Humanos , Nutrientes
17.
Neurogastroenterol Motil ; 32(12): e13944, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32666613

RESUMEN

BACKGROUND: The ability of the gut to detect nutrients is critical to the regulation of gut hormone secretion, food intake, and postprandial blood glucose control. Ingested nutrients are detected by specific gut chemosensors. However, knowledge of these chemosensors has primarily been derived from the intestine, while available information on gastric chemosensors is limited. This study aimed to investigate the nutrient-sensing repertoire of the mouse stomach with particular emphasis on ghrelin cells. METHODS: Quantitative RT-PCR was used to determine mRNA levels of nutrient chemosensors (protein: G protein-coupled receptor 93 [GPR93], calcium-sensing receptor [CaSR], metabotropic glutamate receptor type 4 [mGluR4]; fatty acids: CD36, FFAR2&4; sweet/umami taste: T1R3), taste transduction components (TRPM5, GNAT2&3), and ghrelin and ghrelin-processing enzymes (PC1/3, ghrelin O-acyltransferase [GOAT]) in the gastric corpus and antrum of adult male C57BL/6 mice. Immunohistochemistry was performed to assess protein expression of chemosensors (GPR93, T1R3, CD36, and FFAR4) and their co-localization with ghrelin. KEY RESULTS: Most nutrient chemosensors had higher mRNA levels in the antrum compared to the corpus, except for CD36, GNAT2, ghrelin, and GOAT. Similar regional distribution was observed at the protein level. At least 60% of ghrelin-positive cells expressed T1R3 and FFAR4, and over 80% expressed GPR93 and CD36. CONCLUSIONS AND INFERENCES: The cellular mechanisms for the detection of nutrients are expressed in a region-specific manner in the mouse stomach and gastric ghrelin cells. These gastric nutrient chemosensors may play a role modulating gastrointestinal responses, such as the inhibition of ghrelin secretion following food intake.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/metabolismo , Ghrelina/metabolismo , Nutrientes/metabolismo , Animales , Células Quimiorreceptoras/química , Mucosa Gástrica/química , Ghrelina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Nutrientes/genética , Antro Pilórico/química , Antro Pilórico/citología , Antro Pilórico/metabolismo , Estómago/química , Estómago/citología
18.
Nature ; 574(7776): 63-68, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31554967

RESUMEN

The gp130 receptor cytokines IL-6 and CNTF improve metabolic homeostasis but have limited therapeutic use for the treatment of type 2 diabetes. Accordingly, we engineered the gp130 ligand IC7Fc, in which one gp130-binding site is removed from IL-6 and replaced with the LIF-receptor-binding site from CNTF, fused with the Fc domain of immunoglobulin G, creating a cytokine with CNTF-like, but IL-6-receptor-dependent, signalling. Here we show that IC7Fc improves glucose tolerance and hyperglycaemia and prevents weight gain and liver steatosis in mice. In addition, IC7Fc either increases, or prevents the loss of, skeletal muscle mass by activation of the transcriptional regulator YAP1. In human-cell-based assays, and in non-human primates, IC7Fc treatment results in no signs of inflammation or immunogenicity. Thus, IC7Fc is a realistic next-generation biological agent for the treatment of type 2 diabetes and muscle atrophy, disorders that are currently pandemic.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Citocinas/síntesis química , Citocinas/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Unión Competitiva , Citocinas/química , Diabetes Mellitus Tipo 2/metabolismo , Diseño de Fármacos , Hígado Graso/prevención & control , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Incretinas/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Masculino , Ratones , Músculo Esquelético/efectos de los fármacos , Obesidad/metabolismo , Páncreas/metabolismo , Fosfoproteínas/metabolismo , Ingeniería de Proteínas , Receptores de Interleucina-6/metabolismo , Transducción de Señal , Factores de Transcripción , Aumento de Peso/efectos de los fármacos , Proteínas Señalizadoras YAP
19.
Eur J Nucl Med Mol Imaging ; 46(11): 2380-2391, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31338548

RESUMEN

PURPOSES: Whether low-calorie sweeteners (LCS), such as sucralose and acesulfame K, can alter glucose metabolism is uncertain, particularly given the inconsistent observations relating to insulin resistance in recent human trials. We hypothesized that these discrepancies are accounted for by the surrogate tools used to evaluate insulin resistance and that PET 18FDG, given its capacity to quantify insulin sensitivity in individual organs, would be more sensitive in identifying changes in glucose metabolism. Accordingly, we performed a comprehensive evaluation of the effects of LCS on whole-body and organ-specific glucose uptake and insulin sensitivity in a large animal model of morbid obesity. METHODS: Twenty mini-pigs with morbid obesity were fed an obesogenic diet enriched with LCS (sucralose 1 mg/kg/day and acesulfame K 0.5 mg/kg/day, LCS diet group), or without LCS (control group), for 3 months. Glucose uptake and insulin sensitivity were determined for the duodenum, liver, skeletal muscle, adipose tissue and brain using dynamic PET 18FDG scanning together with direct measurement of arterial input function. Body composition was also measured using CT imaging and energy metabolism quantified with indirect calorimetry. RESULTS: The LCS diet increased subcutaneous abdominal fat by ≈ 20% without causing weight gain, and reduced insulin clearance by ≈ 40%, while whole-body glucose uptake and insulin sensitivity were unchanged. In contrast, glucose uptake in the duodenum, liver and brain increased by 57, 66 and 29% relative to the control diet group (P < 0.05 for all), while insulin sensitivity increased by 53, 55 and 28% (P < 0.05 for all), respectively. In the brain, glucose uptake increased significantly only in the frontal cortex, associated with improved metabolic connectivity towards the hippocampus and the amygdala. CONCLUSIONS: In miniature pigs, the combination of sucralose and acesulfame K is biologically active. While not affecting whole-body insulin resistance, it increases insulin sensitivity and glucose uptake in specific tissues, mimicking the effects of obesity in the adipose tissue and in the brain.


Asunto(s)
Insulina/metabolismo , Obesidad/metabolismo , Obesidad/fisiopatología , Edulcorantes/farmacología , Tejido Adiposo/metabolismo , Amígdala del Cerebelo/diagnóstico por imagen , Alimentación Animal , Animales , Composición Corporal , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18 , Lóbulo Frontal/diagnóstico por imagen , Glucosa/metabolismo , Hipocampo/diagnóstico por imagen , Resistencia a la Insulina , Masculino , Sacarosa/análogos & derivados , Sacarosa/farmacología , Porcinos , Porcinos Enanos , Tiazinas/farmacología , Tomografía Computarizada por Rayos X
20.
J Clin Endocrinol Metab ; 104(7): 2668-2674, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30759215

RESUMEN

CONTEXT: The antidiabetic drug metformin causes weight loss, but the underlying mechanisms are unclear. Recent clinical studies show that metformin increases plasma levels of the anorectic gut hormone, peptide YY (PYY), but whether this is through a direct effect on the gut is unknown. OBJECTIVE: We hypothesized that exposure of human gut mucosal tissue to metformin would acutely trigger PYY secretion. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTIONS: Mucosal tissue was prepared from 46 human colonic and 9 ileal samples obtained after surgical resection and ex vivo secretion assays were performed. Tissue was exposed to metformin, as well as a series of other compounds as part of our mechanistic studies, in static incubations. Supernatant was sampled after 15 minutes. MAIN OUTCOME MEASURES: PYY levels in supernatant, measured using ELISA. RESULTS: Metformin increased PYY secretion from both ileal (P < 0.05) and colonic (P < 0.001) epithelia. Both basal and metformin-induced PYY secretion were unchanged across body mass index or in tissues obtained from individuals with type 2 diabetes. Metformin-dependent PYY secretion was blocked by inhibitors of the plasma membrane monoamine transporter (PMAT) and the serotonin reuptake transporter (SERT), as well as by an inhibitor of AMP kinase (AMPK). CONCLUSIONS: This is a report of a direct action of metformin on the gut epithelium to trigger PYY secretion in humans, occurring via cell internalization through PMAT and SERT and intracellular activation of AMPK. Our results provide further support that the role of metformin in the treatment of metabolic syndrome has a gut-based component.


Asunto(s)
Hipoglucemiantes/farmacología , Mucosa Intestinal/efectos de los fármacos , Metformina/farmacología , Péptido YY/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Anciano , Colon/citología , Colon/efectos de los fármacos , Colon/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Íleon/citología , Íleon/efectos de los fármacos , Íleon/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Masculino , Metformina/uso terapéutico , Persona de Mediana Edad , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Pérdida de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...