Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Diabetologia ; 59(2): 363-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26537124

RESUMEN

AIMS/HYPOTHESIS: Normal glucose metabolism depends on pancreatic secretion of insulin and glucagon. The bihormonal hypothesis states that while lack of insulin leads to glucose underutilisation, glucagon excess is the principal factor in diabetic glucose overproduction. A recent study reported that streptozotocin-treated glucagon receptor knockout mice have normal glucose tolerance. We investigated the impact of acute disruption of glucagon secretin or action in a mouse model of severe diabetes by three different approaches: (1) alpha cell elimination; (2) glucagon immunoneutralisation; and (3) glucagon receptor antagonism, in order to evaluate the effect of these on glucose tolerance. METHODS: Severe diabetes was induced in transgenic and wild-type mice by streptozotocin. Glucose metabolism was investigated using OGTT in transgenic mice with the human diphtheria toxin receptor expressed in proglucagon producing cells allowing for diphtheria toxin (DT)-induced alpha cell ablation and in mice treated with either a specific high affinity glucagon antibody or a specific glucagon receptor antagonist. RESULTS: Near-total alpha cell elimination was induced in transgenic mice upon DT administration and resulted in a massive decrease in pancreatic glucagon content. Oral glucose tolerance in diabetic mice was neither affected by glucagon immunoneutralisation, glucagon receptor antagonism, nor alpha cell removal, but did not deteriorate further compared with mice with intact alpha cell mass. CONCLUSIONS/INTERPRETATION: Disruption of glucagon action/secretion did not improve glucose tolerance in diabetic mice. Near-total alpha cell elimination may have prevented further deterioration. Our findings support insulin lack as the major factor underlying hyperglycaemia in beta cell-deficient diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Glucagón , Intolerancia a la Glucosa , Insulina/deficiencia , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Toxina Diftérica , Glucagón/antagonistas & inhibidores , Glucagón/metabolismo , Glucagón/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/patología , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/genética , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/genética , Estreptozocina
2.
PLoS Biol ; 11(4): e1001542, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630454

RESUMEN

Secretory vesicles in endocrine cells store hormones such as growth hormone (GH) and insulin before their release into the bloodstream. The molecular mechanisms governing budding of immature secretory vesicles from the trans-Golgi network (TGN) and their subsequent maturation remain unclear. Here, we identify the lipid binding BAR (Bin/amphiphysin/Rvs) domain protein PICK1 (protein interacting with C kinase 1) as a key component early in the biogenesis of secretory vesicles in GH-producing cells. Both PICK1-deficient Drosophila and mice displayed somatic growth retardation. Growth retardation was rescued in flies by reintroducing PICK1 in neurosecretory cells producing somatotropic peptides. PICK1-deficient mice were characterized by decreased body weight and length, increased fat accumulation, impaired GH secretion, and decreased storage of GH in the pituitary. Decreased GH storage was supported by electron microscopy showing prominent reduction in secretory vesicle number. Evidence was also obtained for impaired insulin secretion associated with decreased glucose tolerance. PICK1 localized in cells to immature secretory vesicles, and the PICK1 BAR domain was shown by live imaging to associate with vesicles budding from the TGN and to possess membrane-sculpting properties in vitro. In mouse pituitary, PICK1 co-localized with the BAR domain protein ICA69, and PICK1 deficiency abolished ICA69 protein expression. In the Drosophila brain, PICK1 and ICA69 co-immunoprecipitated and showed mutually dependent expression. Finally, both in a Drosophila model of type 2 diabetes and in high-fat-diet-induced obese mice, we observed up-regulation of PICK1 mRNA expression. Our findings suggest that PICK1, together with ICA69, is critical during budding of immature secretory vesicles from the TGN and thus for vesicular storage of GH and possibly other hormones. The data link two BAR domain proteins to membrane remodeling processes in the secretory pathway of peptidergic endocrine cells and support an important role of PICK1/ICA69 in maintenance of metabolic homeostasis.


Asunto(s)
Intolerancia a la Glucosa/metabolismo , Trastornos del Crecimiento/metabolismo , Proteínas Nucleares/deficiencia , Vesículas Secretoras/metabolismo , Animales , Autoantígenos/fisiología , Células COS , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Línea Celular , Chlorocebus aethiops , Drosophila melanogaster , Femenino , Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Trastornos del Crecimiento/genética , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/metabolismo , Homeostasis , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Hipófisis/metabolismo , Unión Proteica , Transporte de Proteínas , Ratas , Imagen de Lapso de Tiempo , Red trans-Golgi/metabolismo
3.
Br J Nutr ; 115(4): 629-36, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824730

RESUMEN

Dietary advanced glycation end products (AGE) formed during heating of food have gained interest as potential nutritional toxins with adverse effects on inflammation and glucose metabolism. In the present study, we investigated the short-term effects of high and low molecular weight (HMW and LMW) dietary AGE on insulin sensitivity, expression of the receptor for AGE (RAGE), the AGE receptor 1 (AGER1) and TNF-α, F2-isoprostaglandins, body composition and food intake. For 2 weeks, thirty-six Sprague-Dawley rats were fed a diet containing 20% milk powder with different proportions of this being given as heated milk powder (0, 40 or 100%), either native (HMW) or hydrolysed (LMW). Gene expression of RAGE and AGER1 in whole blood increased in the group receiving a high AGE LMW diet, which also had the highest urinary excretion of the AGE, methylglyoxal-derived hydroimidazolone 1 (MG-H1). Urinary excretion of N ε-carboxymethyl-lysine increased with increasing proportion of heat-treated milk powder in the HMW and LMW diets but was unrelated to gene expression. There was no difference in insulin sensitivity, F2-isoprostaglandins, food intake, water intake, body weight or body composition between the groups. In conclusion, RAGE and AGER1 expression can be influenced by a high AGE diet after only 2 weeks in proportion to MG-H1 excretion. No other short-term effects were observed.


Asunto(s)
Dieta/efectos adversos , Productos Finales de Glicación Avanzada/efectos adversos , Hexosiltransferasas/metabolismo , Receptor para Productos Finales de Glicación Avanzada/agonistas , Regulación hacia Arriba , Animales , Biomarcadores/sangre , Biomarcadores/orina , Ingestión de Energía , Productos Finales de Glicación Avanzada/administración & dosificación , Productos Finales de Glicación Avanzada/química , Productos Finales de Glicación Avanzada/orina , Hexosiltransferasas/sangre , Hexosiltransferasas/química , Hexosiltransferasas/genética , Calor/efectos adversos , Imidazoles/orina , Imidazolinas/orina , Lisina/análogos & derivados , Lisina/orina , Masculino , Proteínas de la Leche/administración & dosificación , Proteínas de la Leche/efectos adversos , Proteínas de la Leche/química , Peso Molecular , Proteolisis , Distribución Aleatoria , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/sangre , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Eliminación Renal , Pruebas de Toxicidad Subaguda , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Biol Chem ; 286(23): 20845-60, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21402696

RESUMEN

Based on the conformationally constrained D-Trp-Phe-D-Trp (wFw) core of the prototype inverse agonist [D-Arg(1),D-Phe(5),D-Trp(7,9),Leu(11)]substance P, a series of novel, small, peptide-mimetic agonists for the ghrelin receptor were generated. By using various simple, ring-constrained spacers connecting the D-Trp-Phe-D-Trp motif with the important C-terminal carboxyamide group, 40 nm agonism potency was obtained and also in one case (wFw-Isn-NH(2), where Isn is isonipecotic acid) ~80% efficacy. However, in contrast to all previously reported ghrelin receptor agonists, the piperidine-constrained wFw-Isn-NH(2) was found to be a functionally biased agonist. Thus, wFw-Isn-NH(2) mediated potent and efficacious signaling through the Gα(q) and ERK1/2 signaling pathways, but in contrast to all previous ghrelin receptor agonists it did not signal through the serum response element, conceivably the Gα(12/13) pathway. The recognition pattern of wFw-Isn-NH(2) with the ghrelin receptor also differed significantly from that of all previously characterized unbiased agonists. Most importantly, wFw-Isn-NH(2) was not dependent on GluIII:09 (Glu3.33), which otherwise is an obligatory TM III anchor point residue for ghrelin agonists. Molecular modeling and docking experiments indicated that wFw-Isn-NH(2) binds in the classical agonist binding site between the extracellular segments of TMs III, VI, and VII, interacting closely with the aromatic cluster between TMs VI and VII, but that it does so in an opposite orientation as compared with, for example, the wFw peptide agonists. It is concluded that the novel peptide-mimetic ligand wFw-Isn-NH(2) is a biased ghrelin receptor agonist and that the selective signaling pattern presumably is due to its unique receptor recognition pattern lacking interaction with key residues especially in TM III.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Peptidomiméticos/farmacología , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/metabolismo , Sustancia P , Secuencias de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Moleculares , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Receptores de Ghrelina/genética
5.
Amino Acids ; 43(3): 1265-75, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22200933

RESUMEN

L-Arginine (L-Arg) is a conditionally essential amino acid and a natural constituent of dietary proteins. Studies in obese rats and type 2 diabetic humans have indicated that dietary supplementation with L-Arg can diminish gain in white adipose tissue (WAT) and improve insulin sensitivity. However, the effects of L-Arg on glucose homeostasis, body composition and energy metabolism remain unclear. In addition, no studies have, to our knowledge, examined whether L-Arg has beneficial effects as a dietary supplement in the mouse model. In the present study, we investigated the effects of L-Arg supplementation to male C57BL/6 mice on an array of physiological parameters. L-Arg supplemented mice were maintained on a low-protein diet and body composition, appetite regulation, glucose tolerance, insulin sensitivity and energy expenditure were evaluated. A significant reduction in epididymal WAT was observed in L-Arg supplemented mice compared with mice fed an isocaloric control diet. Surprisingly, the L-Arg supplemented animals were hyperphagic corresponding to a highly significant decrease in feed efficiency, as body weight developed in a similar pattern in both experimental groups. Glucose homeostasis experiments revealed a major effect of L-Arg supplementation on glucose tolerance and insulin sensitivity, interestingly, independent of a parallel regulation in whole-body adiposity. Increased L-Arg ingestion also raised energy expenditure; however, no concurrent effect on locomotor activity, substrate metabolism or expression of uncoupling proteins (UCP1 and UCP2) in adipose tissues was displayed. In conclusion, dietary L-Arg supplementation substantially affects an array of metabolic-associated parameters including a reduction in WAT, hyperphagia, improved insulin sensitivity and increased energy expenditure in mice fed a low-protein diet.


Asunto(s)
Arginina/administración & dosificación , Hipoglucemiantes/administración & dosificación , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/patología , Adiposidad/efectos de los fármacos , Animales , Arginina/efectos adversos , Glucemia , Dieta con Restricción de Proteínas/efectos adversos , Suplementos Dietéticos , Ingestión de Energía/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Genes Mitocondriales , Glucosa/metabolismo , Homeostasis , Hiperfagia/inducido químicamente , Hipoglucemiantes/efectos adversos , Insulina/sangre , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos
6.
Neurosci Lett ; 449(1): 20-3, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18926875

RESUMEN

The selective CB1 receptor antagonist rimonabant is a novel weight control agent. Although CB1 receptors and binding sites are present in both the rodent central and peripheral nervous systems, including the afferent vagus nerve, the role of gut afferents in mediating anorexia following CB1R blockade is still debated. In the present study we examined rimonabant-induced anorexia in male C57BL/6J mice with subdiaphragmatic vagotomy (VGX) as well as in male Sprague-Dawley rats subjected to either subdiaphragmatic vagal deafferentation (SDA) alone or in combination with a complete celiac-superior mesenteric ganglionectomy (CGX). Irrespective of the operational procedure, rimonabant (10mg/kg) effectively reduced standard chow as well as palatable diet (ensure) intake. In conclusion, the data clearly demonstrate that neither vagal gut afferents, nor gut afferents traveling via the sympathetic nervous system, are required for rimonabant to inhibit food intake leading to the hypothesis that centrally located CB1 receptors are the prime mediators of rimonabant-induced anorexia.


Asunto(s)
Anorexia/inducido químicamente , Anorexia/fisiopatología , Ganglios Simpáticos/fisiología , Piperidinas , Pirazoles , Nervio Vago/fisiología , Análisis de Varianza , Animales , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Ganglionectomía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Rimonabant , Estilbamidinas/metabolismo , Vagotomía/métodos , Nervio Vago/cirugía
7.
J Neuroendocrinol ; 31(7): e12699, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30776164

RESUMEN

Huntington's disease (HD) is a heritable neurodegenerative disorder, characterised by metabolic disturbances, along with cognitive and psychiatric impairments. Targeting metabolic HD dysfunction via the maintenance of body weight and fat mass and restoration of peripheral energy metabolism can improve the progression of neurological symptoms. In this respect, we focused on the therapeutic potential of the orexigenic peptide hormone ghrelin, which plays an important role in promoting a positive energy balance. In the present study, we found a significant disruption of circadian metabolic regulation in a R6/2 mouse HD model in the late stage of disease. Daily circadian rhythms of activity, energy expenditure, respiratory exchange ratio and feeding were strongly attenuated in R6/2 mice. During the rest phase, R6/2 mice had a higher total activity, elevated energy expenditure and excessive water consumption compared to control mice. We also found that, in the late stage of disease, R6/2 mice had ghrelin axis deficiency as a result of low circulating ghrelin levels, in addition to down-regulation of the ghrelin receptor and several key signalling molecules in the hypothalamus, as well as a reduced responsiveness to exogenous peripheral ghrelin. We demonstrated that, in pre-symptomatic mice, responsiveness to ghrelin is preserved. Chronic ghrelin treatment efficiently increased lean body mass and decreased the energy expenditure and fat utilisation of R6/2 mice in the early stage of disease. In addition, ghrelin treatment was also effective in the normalisation of drinking behaviour and the rest activity of these mice. Ghrelin treatment could provide a novel therapeutic possibility for delaying disease progression; however, deficiency in ghrelin receptor expression could limit its therapeutic potential in the late stage of disease.


Asunto(s)
Ghrelina/metabolismo , Enfermedad de Huntington/metabolismo , Animales , Composición Corporal , Ritmo Circadiano , Modelos Animales de Enfermedad , Ingestión de Alimentos , Metabolismo Energético , Femenino , Ratones Transgénicos , Actividad Motora , Fenotipo
8.
Cell Rep ; 22(1): 175-188, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29298420

RESUMEN

Apolipoprotein M (apoM) is the carrier of sphingosine-1-phosphate (S1P) in plasma high-density lipoproteins. S1P is a bioactive lipid interacting with five receptors (S1P1-5). We show that lack of apoM in mice increases the amount of brown adipose tissue (BAT), accelerates the clearance of postprandial triglycerides, and protects against diet-induced obesity (i.e., a phenotype similar to that induced by cold exposure or ß3-adrenergic stimulation). Moreover, the data suggest that the phenotype of apoM-deficient mice is S1P dependent and reflects diminished S1P1 stimulation. The results reveal a link between the apoM/S1P axis and energy metabolism.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Apolipoproteínas M/metabolismo , Metabolismo Energético/fisiología , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Triglicéridos/metabolismo , Tejido Adiposo Pardo/citología , Animales , Apolipoproteínas M/genética , Lisofosfolípidos/genética , Ratones , Ratones Noqueados , Esfingosina/genética , Esfingosina/metabolismo
9.
Mol Cell Endocrinol ; 449: 64-73, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27908836

RESUMEN

GPR40 is generally known to signal through Gq. However, in transfected cells, certain synthetic agonists can make the receptor signal also through Gs and cAMP (Hauge et al., 2015). Here we find that, in colonic crypt cultures, the GLP-1 secretion induced by such Gq + Gs GPR40 agonists is indeed inhibited by blockers of both Gq and Gs and is eliminated by combining these. This in contrast to Gq-only GPR40 agonists which only are affected by the Gq inhibitor. Importantly, Gq-only GPR40 agonists in combination with low doses of selective synthetic agonists for Gs coupled receptors, e.g. GPR119 and TGR5 provide more than additive GLP-1 secretion both ex vivo and in vivo in mice. It is concluded that under physiological circumstances triglyceride metabolites, i.e. long chain fatty acids and 2-monoacyl glycerol plus bile acids, act synergistically through their respective receptors, GPR40, GPR119 and TGR5 to stimulate GLP-1 secretion robustly by combining Gq and Gs signaling pathways.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Transducción de Señal , Administración Oral , Animales , Colon/metabolismo , Ratones Endogámicos C57BL
10.
Mol Cell Endocrinol ; 448: 108-121, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28390953

RESUMEN

Pannexins (Panx's) are membrane proteins involved in a variety of biological processes, including cell death signaling and immune functions. The role and functions of Panx's in pancreatic ß-cells remain to be clarified. Here, we show Panx1 and Panx2 expression in isolated islets, primary ß-cells, and ß-cell lines. The expression of Panx2, but not Panx1, was downregulated by interleukin-1ß (IL-1ß) plus interferon-γ (IFNγ), two pro-inflammatory cytokines suggested to contribute to ß-cell demise in type 1 diabetes (T1D). siRNA-mediated knockdown (KD) of Panx2 aggravated cytokine-induced apoptosis in rat INS-1E cells and primary rat ß-cells, suggesting anti-apoptotic properties of Panx2. An anti-apoptotic function of Panx2 was confirmed in isolated islets from Panx2-/- mice and in human EndoC-ßH1 cells. Panx2 KD was associated with increased cytokine-induced activation of STAT3 and higher expression of inducible nitric oxide synthase (iNOS). Glucose-stimulated insulin release was impaired in Panx2-/- islets, and Panx2-/- mice subjected to multiple low-dose Streptozotocin (MLDS) treatment, a model of T1D, developed more severe diabetes compared to wild type mice. These data suggest that Panx2 is an important regulator of the insulin secretory capacity and apoptosis in pancreatic ß-cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Conexinas/deficiencia , Citocinas/farmacología , Intolerancia a la Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Conexinas/metabolismo , Técnicas de Silenciamiento del Gen , Intolerancia a la Glucosa/patología , Humanos , Hiperglucemia/patología , Inflamación/patología , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Factor de Transcripción STAT3/metabolismo , Estreptozocina
11.
Endocrinology ; 157(1): 176-94, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26469136

RESUMEN

The 2 gut hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) are well known to be coexpressed, costored, and released together to coact in the control of key metabolic target organs. However, recently, it became clear that several other gut hormones can be coexpressed in the intestinal-specific lineage of enteroendocrine cells. Here, we focus on the anatomical and functional consequences of the coexpression of neurotensin with GLP-1 and PYY in the distal small intestine. Fluorescence-activated cell sorting analysis, laser capture, and triple staining demonstrated that GLP-1 cells in the crypts become increasingly multihormonal, ie, coexpressing PYY and neurotensin as they move up the villus. Proglucagon promoter and pertussis toxin receptor-driven cell ablation and reappearance studies indicated that although all the cells die, the GLP-1 cells reappear more quickly than PYY- and neurotensin-positive cells. High-resolution confocal fluorescence microscopy demonstrated that neurotensin is stored in secretory granules distinct from GLP-1 and PYY storing granules. Nevertheless, the 3 peptides were cosecreted from both perfused small intestines and colonic crypt cultures in response to a series of metabolite, neuropeptide, and hormonal stimuli. Importantly, neurotensin acts synergistically, ie, more than additively together with GLP-1 and PYY to decrease palatable food intake and inhibit gastric emptying, but affects glucose homeostasis in a more complex manner. Thus, neurotensin is a major gut hormone deeply integrated with GLP-1 and PYY, which should be taken into account when exploiting the enteroendocrine regulation of metabolism pharmacologically.


Asunto(s)
Células Enteroendocrinas/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Neurotensina/metabolismo , Péptido YY/metabolismo , Animales , Biomarcadores/metabolismo , Bombesina/farmacología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/ultraestructura , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Péptido 1 Similar al Glucagón/genética , Humanos , Íleon/efectos de los fármacos , Íleon/ultraestructura , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/ultraestructura , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurotensina/genética , Fragmentos de Péptidos/farmacología , Péptido YY/genética , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura , Técnicas de Cultivo de Tejidos , Proteína Fluorescente Roja
12.
Endocrinology ; 157(12): 4561-4569, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779915

RESUMEN

Triglycerides (TGs) are among the most efficacious stimulators of incretin secretion; however, the relative importance of FFA1 (G Protein-coupled Receptor [GPR] 40), FFA4 (GPR120), and GPR119, which all recognize TG metabolites, ie, long-chain fatty acid and 2-monoacylglycerol, respectively, is still unclear. Here, we find all 3 receptors to be highly expressed and highly enriched in fluorescence-activated cell sorting-purified GLP-1 and GIP cells isolated from transgenic reporter mice. In vivo, the TG-induced increase in plasma GIP was significantly reduced in FFA1-deficient mice (to 34%, mean of 4 experiments each with 8-10 animals), in GPR119-deficient mice (to 24%) and in FFA1/FFA4 double deficient mice (to 15%) but not in FFA4-deficient mice. The TG-induced increase in plasma GLP-1 was only significantly reduced in the GPR119-deficient and the FFA1/FFA4 double deficient mice, but not in the FFA1, and FFA4-deficient mice. In mouse colonic crypt cultures the synthetic FFA1 agonists, TAK-875 stimulated GLP-1 secretion to a similar extent as the prototype GLP-1 secretagogue neuromedin C; this, however, only corresponded to approximately half the maximal efficiency of the GPR119 agonist AR231453, whereas the GPR120 agonist Metabolex-209 had no effect. Importantly, when the FFA1 agonist was administered on top of appropriately low doses of the GPR119 agonist, a clear synergistic, ie, more than additive, effect was observed. It is concluded that the 2-monoacylglycerol receptor GPR119 is at least as important as the long-chain fatty acid receptor FFA1 in mediating the TG-induced secretion of incretins and that the 2 receptors act in synergy, whereas FFA4 plays a minor if any role.


Asunto(s)
Colon/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Triglicéridos/metabolismo , Animales , Benzofuranos/farmacología , Bombesina/farmacología , Colon/efectos de los fármacos , Grasas de la Dieta , Péptido 1 Similar al Glucagón/metabolismo , Ratones , Ratones Noqueados , Fragmentos de Péptidos/farmacología , Receptores Acoplados a Proteínas G/genética , Sulfonas/farmacología
13.
Endocrinology ; 157(9): 3482-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27580810

RESUMEN

Neurotensin (NT) is a peptide expressed in the brain and in the gastrointestinal tract. Brain NT inhibits food intake, but the effects of peripheral NT are less investigated. In this study, peripheral NT decreased food intake in both mice and rats, which was abolished by a NT antagonist. Using c-Fos immunohistochemistry, we found that peripheral NT activated brainstem and hypothalamic regions. The anorexigenic effect of NT was preserved in vagotomized mice but lasted shorter than in sham-operated mice. This in combination with a strong increase in c-Fos activation in area postrema after ip administration indicates that NT acts both through the blood circulation and the vagus. To improve the pharmacokinetics of NT, we developed a pegylated NT peptide, which presumably prolonged the half-life, and thus, the effect on feeding was extended compared with native NT. On a molecular level, the pegylated NT peptide increased proopiomelanocortin mRNA in the arcuate nucleus. We also investigated the importance of NT for the decreased food intake after gastric bypass surgery in a rat model of Roux-en-Y gastric bypass (RYGB). NT was increased in plasma and in the gastrointestinal tract in RYGB rats, and pharmacological antagonism of NT increased food intake transiently in RYGB rats. Taken together, our data suggest that NT is a metabolically active hormone, which contributes to the regulation of food intake.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Derivación Gástrica , Neurotensina/administración & dosificación , Animales , Ingestión de Alimentos/efectos de los fármacos , Femenino , Masculino , Ratones Endogámicos C57BL , Neurotensina/antagonistas & inhibidores , Neurotensina/sangre , Ratas Sprague-Dawley , Sacarosa , Vagotomía
14.
Cell Metab ; 23(2): 335-43, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26724858

RESUMEN

The liver is an important integrator of nutrient metabolism, yet no liver-derived factors regulating nutrient preference or carbohydrate appetite have been identified. Here we show that the liver regulates carbohydrate intake through production of the hepatokine fibroblast growth factor 21 (FGF21), which markedly suppresses consumption of simple sugars, but not complex carbohydrates, proteins, or lipids. Genetic loss of FGF21 in mice increases sucrose consumption, whereas acute administration or overexpression of FGF21 suppresses the intake of both sugar and non-caloric sweeteners. FGF21 does not affect chorda tympani nerve responses to sweet tastants, instead reducing sweet-seeking behavior and meal size via neurons in the hypothalamus. This liver-to-brain hormonal axis likely represents a negative feedback loop as hepatic FGF21 production is elevated by sucrose ingestion. We conclude that the liver functions to regulate macronutrient-specific intake by producing an endocrine satiety signal that acts centrally to suppress the intake of "sweets."


Asunto(s)
Sistema Endocrino/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Preferencias Alimentarias/efectos de los fármacos , Hígado/metabolismo , Sacarosa/farmacología , Gusto/efectos de los fármacos , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Sistema Endocrino/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Ratones Noqueados , Proteínas Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
15.
J Nutr Biochem ; 26(1): 9-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25315863

RESUMEN

Whey protein consumption reportedly alleviates parameters of the metabolic syndrome. Here, we investigated the effects of whey protein isolate (whey) in young mice fed a high-fat diet. We hypothesized that whey as the sole protein source reduced early weight gain associated with retarded growth and decreased concentration of insulin-like growth factor-1. Moreover, we hypothesized that these changes were explained by increased nitrogen loss via elevated urea production and/or increased energy expenditure. Male 5-week-old C57BL/6 mice were fed high-fat diets with the protein source being either whey, casein or a combination of both for 5 weeks. After 1, 3 or 5 weeks, respectively, the mice were subjected to a meal challenge with measurements of blood and urinary urea before and 1 and 3 h after eating a weighed meal of their respective diets. In a subset of mice, energy expenditure was measured by indirect calorimetry during the first week of dietary intervention. Observed exclusively during the first week of intervention, whey significantly reduced body length (P<.01) and weight gain (P<.001) correlating positively with plasma concentrations of insulin-like growth factor-1. The combination diet displayed intermediate results indicating an interactive effect. Urea production, urea cycle activity, food intake and energy expenditure were unaffected by protein source. In conclusion, whey decreased growth-related parameters exclusively during the first week of dietary intervention. The early effect of whey could not be explained by food intake, energy expenditure, urea production or urea cycle activity but was correlated with plasma levels of insulin-like growth factor-1.


Asunto(s)
Caseínas/farmacología , Dieta Alta en Grasa/efectos adversos , Proteínas de la Leche/farmacología , Pérdida de Peso/efectos de los fármacos , Animales , Composición Corporal , Ingestión de Energía , Metabolismo Energético/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Urea/sangre , Urea/orina , Aumento de Peso/efectos de los fármacos , Proteína de Suero de Leche
16.
J Endocrinol ; 225(2): 77-88, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25767056

RESUMEN

Erythropoietin (EPO) ameliorates glucose metabolism through mechanisms not fully understood. In this study, we investigated the effect of EPO on glucose metabolism and insulin signaling in skeletal muscle. A 2-week EPO treatment of rats fed with a high-fat diet (HFD) improved fasting glucose levels and glucose tolerance, without altering total body weight or retroperitoneal fat mass. Concomitantly, EPO partially rescued insulin-stimulated AKT activation, reduced markers of oxidative stress, and restored heat-shock protein 72 expression in soleus muscles from HFD-fed rats. Incubation of skeletal muscle cell cultures with EPO failed to induce AKT phosphorylation and had no effect on glucose uptake or glycogen synthesis. We found that the EPO receptor gene was expressed in myotubes, but was undetectable in soleus. Together, our results indicate that EPO treatment improves glucose tolerance but does not directly activate the phosphorylation of AKT in muscle cells. We propose that the reduced systemic inflammation or oxidative stress that we observed after treatment with EPO could contribute to the improvement of whole-body glucose metabolism.


Asunto(s)
Eritropoyetina/metabolismo , Intolerancia a la Glucosa/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Intolerancia a la Glucosa/enzimología , Intolerancia a la Glucosa/genética , Proteínas del Choque Térmico HSP72/genética , Proteínas del Choque Térmico HSP72/metabolismo , Humanos , Insulina/metabolismo , Masculino , Ratones , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo
17.
Mol Metab ; 4(1): 3-14, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25685685

RESUMEN

OBJECTIVES: GPR40 (FFAR1), a clinically proven anti-diabetes target, is a Gq-coupled receptor for long chain fatty acids (LCFA) stimulating insulin secretion directly and mediating a major part of the dietary triglyceride-induced secretion of the incretins GLP-1 and GIP. In phase-II studies the GPR40 agonist TAK-875 decreased blood glucose but surprisingly without stimulating incretins. METHODS AND RESULTS: Here we find that GPR40 can signal through not only Gq and IP3 but also Gs and cAMP when stimulated with certain agonists such as AM-1638 and AM-5262 in contrast to the endogenous LCFA ligands and agonists such as TAK-875 and AM-837, which only signal through Gq. In competition binding against [3H]AM-1638 and [3H]L358 the Gq + Gs and the Gq-only agonists either competed for or showed positive cooperativity by increasing the binding of the two different radio-ligands, in opposite ways. Nevertheless, both the Gq-only and the Gq + Gs agonists all docked surprisingly well into the binding site for TAK-875 in the X-ray structure of GPR40. In murine intestinal primary cell-cultures the endogenous LCFAs and the Gq-only agonists stimulated GLP-1 secretion with rather poor efficacy as compared with the high efficacy Gq + Gs GPR40 agonists and a prototype GPR119 agonist. Similarly, in fasting both male and female mice the Gq + Gs agonists showed significantly higher efficacy than the Gq-only agonists in respect of increasing plasma GLP-1 and plasma GIP in a GPR40-dependent manner. CONCLUSIONS: It is concluded that stimulation of GPR40 by endogenous LCFAs or by Gq-only synthetic agonists result in a rather limited incretin response, whereas Gq + Gs GPR40 agonists stimulate incretin secretion robustly.

18.
PLoS One ; 9(6): e100365, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24959837

RESUMEN

BACKGROUND AND AIMS: Oleoylethanolamide and several other N-acylethanolamines (NAEs), e.g. linoleoylethanolamide and palmitoylethanolamide, have anorectic properties in rats, and prolonged intake of a high-fat diet decreases the levels of the anorectic NAEs in jejunum. Jejunal anorectic NAEs are thought to add to the control of food intake via activation of PPARalpha and the vagus nerve. The fat-induced decrease may explain part of the hyperphagic effect of high-fat diets. In the present study, we investigated 1) whether the reduced levels of anorectic NAEs were reversible in rats, 2) whether mice respond to dietary fat (olive oil) by reducing levels of anorectic NAEs, and 3) whether dietary non-esterified oleic acid also can decrease levels of anorectic NAEs in mice. We are searching for the fat sensor in the intestine, which mediates the decreased levels of anorectic NAEs. METHODS: Male rats and mice were fed diets high (45 energy% fat) in either triacylglycerol or free fatty acids for 7-14 days, and jejunal NAE and N-acylphosphatidylethanolamine (NAPE) levels were determined by liquid-chromatography mass spectrometry. RESULTS: In rats, reduced levels of anorectic NAEs could be reversed after 3 days from changing the diet from high-fat to chow. Corresponding NAPE levels tended to show the same changes. In mice, jejunal levels of anorectic NAEs were also reduced when fed a high-fat diet. In addition, we found that non-esterified oleic acid were also able to reduce levels of anorectic NAEs in mice. CONCLUSIONS: These results suggest that the down-regulation of the jejunal level of anorectic NAEs by dietary fat is not restricted to rats, and that the fatty acid component oleic acid, in dietary olive oil may be sufficient to mediate this regulation. Thus, a fatty acid sensor may mediate this effect of dietary fat.


Asunto(s)
Grasas de la Dieta/metabolismo , Etanolaminas/metabolismo , Yeyuno/metabolismo , Ácido Oléico/metabolismo , Animales , Dieta Alta en Grasa , Grasas de la Dieta/administración & dosificación , Masculino , Ratones , Ácido Oléico/administración & dosificación , Aceite de Oliva , Aceites de Plantas/administración & dosificación , Aceites de Plantas/metabolismo , Ratas , Triglicéridos/metabolismo
19.
J Endocrinol ; 217(2): 151-60, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23428581

RESUMEN

The recently identified G protein-coupled receptor GPRC6A is activated by dietary amino acids and expressed in multiple tissues. Although the receptor is hypothesised to exert biological impact on metabolic and endocrine-related parameters, the role of the receptor in obesity and metabolic complications is still elusive. In the present study, we investigated the impact of GPRC6A deficiency in a murine model of diet-induced obesity (DIO). Male Gprc6a knockout (KO) mice and WT littermates were subjected to a high-fat diet (HFD) for 25 weeks and exposed to comprehensive metabolic phenotyping. A significant increase in body weight, corresponding to a selective increase in body fat, was observed in Gprc6a KO mice exposed to an HFD relative to WT controls. The obese phenotype was linked to subtle perturbations in energy homoeostasis as GPRC6A deficiency resulted in chronic hyperphagia and decreased locomotor activity. Moreover, diet-induced obese Gprc6a KO mice had increased circulating insulin and leptin levels relative to WT animals, thereby demonstrating that endocrine abnormalities associate with the reported disturbances in energy balance. The phenotype was further accompanied by disruptions in glucose metabolism showing that Gprc6a KO mice on an HFD display increased susceptibility to develop metabolic-related disorders. Altogether, these data suggest that the amino acid sensing receptor GPRC6A plays an important role in resistance to DIO and metabolic complications. Future studies will illuminate the underlying molecular mechanisms mediating the herein reported findings and potentially facilitate the development of novel therapeutic compounds targeting the GPRC6A receptor.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Predisposición Genética a la Enfermedad/genética , Obesidad/etiología , Obesidad/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Animales , Composición Corporal/fisiología , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Glucosa/metabolismo , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Fenotipo , Receptores Acoplados a Proteínas G/metabolismo
20.
Diabetes ; 62(9): 3064-74, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23670974

RESUMEN

Circulating interleukin (IL)-18 is elevated in obesity, but paradoxically causes hypophagia. We hypothesized that IL-18 may attenuate high-fat diet (HFD)-induced insulin resistance by activating AMP-activated protein kinase (AMPK). We studied mice with a global deletion of the α-isoform of the IL-18 receptor (IL-18R(-/-)) fed a standard chow or HFD. We next performed gain-of-function experiments in skeletal muscle, in vitro, ex vivo, and in vivo. We show that IL-18 is implicated in metabolic homeostasis, inflammation, and insulin resistance via mechanisms involving the activation of AMPK in skeletal muscle. IL-18R(-/-) mice display increased weight gain, ectopic lipid deposition, inflammation, and reduced AMPK signaling in skeletal muscle. Treating myotubes or skeletal muscle strips with IL-18 activated AMPK and increased fat oxidation. Moreover, in vivo electroporation of IL-18 into skeletal muscle activated AMPK and concomitantly inhibited HFD-induced weight gain. In summary, IL-18 enhances AMPK signaling and lipid oxidation in skeletal muscle implicating IL-18 in metabolic homeostasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Resistencia a la Insulina/fisiología , Interleucina-18/metabolismo , Músculo Esquelético/enzimología , Aumento de Peso/fisiología , Proteínas Quinasas Activadas por AMP/genética , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Calorimetría Indirecta , Femenino , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-18/deficiencia , Receptores de Interleucina-18/genética , Aumento de Peso/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA