Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 30: 72-130, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31767182

RESUMEN

BACKGROUND: The glucagon-like peptide-1 (GLP-1) is a multifaceted hormone with broad pharmacological potential. Among the numerous metabolic effects of GLP-1 are the glucose-dependent stimulation of insulin secretion, decrease of gastric emptying, inhibition of food intake, increase of natriuresis and diuresis, and modulation of rodent ß-cell proliferation. GLP-1 also has cardio- and neuroprotective effects, decreases inflammation and apoptosis, and has implications for learning and memory, reward behavior, and palatability. Biochemically modified for enhanced potency and sustained action, GLP-1 receptor agonists are successfully in clinical use for the treatment of type-2 diabetes, and several GLP-1-based pharmacotherapies are in clinical evaluation for the treatment of obesity. SCOPE OF REVIEW: In this review, we provide a detailed overview on the multifaceted nature of GLP-1 and its pharmacology and discuss its therapeutic implications on various diseases. MAJOR CONCLUSIONS: Since its discovery, GLP-1 has emerged as a pleiotropic hormone with a myriad of metabolic functions that go well beyond its classical identification as an incretin hormone. The numerous beneficial effects of GLP-1 render this hormone an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, and neurodegenerative disorders.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Receptores de Glucagón/metabolismo
2.
Diabetes Obes Metab ; 14(6): 531-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22226053

RESUMEN

AIM: Previous studies with the novel once daily glucagon-like peptide-1 (GLP-1) analogue liraglutide and the GLP-1 receptor agonist exenatide have revealed profound insulinotrophic and antidiabetic effects, but also potent effects on gastric emptying (GE) and long-term and lasting reductions in body weight. In this study, we examined the acute and chronic effects of two different GLP-1 analogues with different pharmacokinetic profiles on GE, food intake and body weight. METHODS: On the basis of a series of dose-finding studies, the doses of exenatide and liraglutide with similar acute anorectic effects were identified. GE was assessed using a standard acetaminophen release assay. After the acute test, rats were dosed bi-daily for 14 days in which period food intake and body weight was monitored. On day 14, the GE rate was reassessed. RESULTS: While both compounds exerted robust acute reductions in GE, the effect was markedly diminished following 14 days of dosing with liraglutide. In contrast, exenatide-treated rats still displayed a profound reduction in GE at the 14-day time-point. Both compounds exerted similar effects on body weight. CONCLUSION: The data suggest that the 'gastric inhibitory' GLP-1 receptors in rats are subject to desensitization/tachyphylaxis but that this effect is dependent on full 24-h exposure as obtained by liraglutide. The body weight-lowering effects of GLP-1 receptor stimulation are not subject to desensitization. These data indicate that regulation of appetite signals in the brain, and not GE, is the main mechanism for liraglutide-induced weight loss.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Vaciamiento Gástrico/efectos de los fármacos , Péptido 1 Similar al Glucagón/análogos & derivados , Hipocampo/efectos de los fármacos , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Ingestión de Alimentos/efectos de los fármacos , Exenatida , Péptido 1 Similar al Glucagón/farmacocinética , Péptido 1 Similar al Glucagón/farmacología , Inyecciones Intravenosas , Liraglutida , Masculino , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Ponzoñas/farmacología
3.
J Neuroendocrinol ; 14(11): 880-6, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12421341

RESUMEN

In the hypothalamic arcuate nucleus, neurones that coexpress cocaine-amphetamine-regulated transcript (CART) and alpha-melanocyte-stimulating hormone [alpha-MSH; pro-opiomelanocortin (POMC) derived] peptides exert catabolic actions and are stimulated by leptin. However, leptin treatment also affects other circulating factors that influence hypothalamic gene expression. Notably, the hypercorticosteronaemia of ob/ob mice is lowered by leptin treatment. To examine the interaction between glucocorticoids and leptin on POMC/CART mRNA expression, an experiment combining leptin and adrenalectomy (ADX) in leptin deficient ob/ob mice was carried out. Obese ob/ob and lean littermate Ob/? mice were ADX or sham-operated. ADX mice received a pellet containing 25% corticosterone subcutaneously. Seven days postoperatively, mice were injected intraperitoneally for 5 days with either recombinant human leptin or vehicle. On the sixth day, the mice were decapitated and the brains removed and trunk blood was collected for corticosterone analysis. Plasma concentrations of corticosterone were elevated in all ob/ob groups compared to Ob/?. For both ob/ob and Ob/? groups, corticosterone concentrations exhibited a decline across groups: vehicle-sham>leptin-sham>ADX-vehicle>ADX-leptin. Leptin inhibited food intake and bodyweight in ob/ob-sham and ob/ob-ADX to a similar extent, whereas no effect of leptin was observed in Ob/? mice. Similarly, leptin caused an identical increase in arcuate POMC and CART mRNA expression in ob/ob-sham and ob/ob-ADX compared to vehicle. The present data support the view that leptin influences arcuate POMC and CART mRNA expression directly, and that the effect is not modulated by corticosterone across a wide range of circulating corticosterone concentrations.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Corticosterona/sangre , Leptina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Proopiomelanocortina/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL
5.
Diabetes ; 50(11): 2530-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11679431

RESUMEN

Postprandial release of the incretin glucagon-like peptide-1 (GLP-1) has been suggested to act as an endogenous satiety factor in humans. In rats, however, the evidence for this is equivocal probably because of very high endogenous activity of the GLP-1 degrading enzyme dipeptidyl peptidase-IV. In the present study, we show that intravenously administered GLP-1 (100 and 500 microg/kg) decreases food intake for 60 min in hungry rats. This effect is pharmacologically specific as it is inhibited by previous administration of 100 microg/kg exendin(9-39), and biologically inactive GLP-1(1-37) had no effect on food intake when administered alone (500 microg/kg). Acute intravenous administration of GLP-1 also caused dose-dependent inhibition of water intake, and this effect was equally well abolished by previous administration of exendin(9-39). A profound increase in diuresis was observed after intravenous administration of both 100 and 500 microg/kg GLP-1. Using a novel long-acting injectable GLP-1 derivative, NN2211, the acute and subchronic anorectic potentials of GLP-1 and derivatives were studied in both normal rats and rats made obese by neonatal monosodium glutamate treatment (MSG). We showed previously that MSG-treated animals are insensitive to the anorectic effects of centrally administered GLP-1(7-37). Both normal and MSG-lesioned rats were randomly assigned to groups to receive NN2211 or vehicle. A single bolus injection of NN2211 caused profound dose-dependent inhibition of overnight food and water intake and increased diuresis in both normal and MSG-treated rats. Subchronic multiple dosing of NN2211 (200 microg/kg) twice daily for 10 days to normal and MSG-treated rats caused profound inhibition of food intake. The marked decrease in food intake was accompanied by reduced body weight in both groups, which at its lowest stabilized at approximately 85% of initial body weight. Initial excursions in water intake and diuresis were transient as they were normalized within a few days of treatment. Lowered plasma levels of triglycerides and leptin were observed during NN2211 treatment in both normal and MSG-treated obese rats. In a subsequent study, a 7-day NN2211 treatment period of normal rats ended with measurement of energy expenditure (EE) and body composition determined by indirect calorimetry and dual energy X-ray absorptiometry, respectively. Compared with vehicle-treated rats, NN2211 and pair-fed rats decreased their total EE corresponding to the observed weight loss, such that EE per weight unit of lean body mass was unaffected. Despite its initial impact on body fluid balance, NN2211 had no debilitating effects on body water homeostasis as confirmed by analysis of body composition, plasma electrolytes, and hematocrit. This is in contrast to pair-fed animals, which displayed hemoconcentration and tendency toward increased percentage of fat mass. The present series of experiments show that GLP-1 is fully capable of inhibiting food intake in rats via a peripherally accessible site. The loss in body weight is accompanied by decreased levels of circulating leptin indicative of loss of body fat. The profound weight loss caused by NN2211 treatment was without detrimental effects on body water homeostasis. Thus, long-acting GLP-1 derivatives may prove efficient as weight-reducing therapeutic agents for overweight patients with type 2 diabetes.


Asunto(s)
Peso Corporal/efectos de los fármacos , Glucagón/farmacología , Obesidad/patología , Animales , Glucemia/análisis , Defecación/efectos de los fármacos , Diuresis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Líquidos/efectos de los fármacos , Esquema de Medicación , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón , Lípidos/sangre , Liraglutida , Masculino , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/farmacología , Ratas , Ratas Wistar , Valores de Referencia , Factores de Tiempo
6.
Ugeskr Laeger ; 163(3): 287-91, 2001 Jan 15.
Artículo en Danés | MEDLINE | ID: mdl-11219107

RESUMEN

We report here that glucagon-like peptide 2(GLP-2) and its receptor constitute a distinct projection system connecting the nucleus of the solitary tract with the dorsomedial hypothalamic nucleus (DMH). The DMH contains a dense plexus of GLP-2 immunoreactive fibres and is the only hypothalamic nucleus expressing GLP-2 receptor mRNA. Consistent with this, central application of GLP-2 activates the expression of neurones solely in the DMH. Furthermore, central administration of GLP-2 causes a dose-related, a pharmacologically and behaviourally specific inhibition of food intake in rats. Surprisingly, the alleged GLP-1 receptor antagonist, Exending (9-39), proved a functional antagonist of centrally applied GLP-2. These data implicate GLP-2 as an important neurotransmitter in the regulation of food intake and likely bodyweight. Our data therefore point to the DMH as a crossroad for endocrine and visceral information affecting feeding behaviour.


Asunto(s)
Regulación del Apetito/fisiología , Hormonas Gastrointestinales/fisiología , Neurotransmisores/fisiología , Péptidos/fisiología , Receptores de Glucagón/fisiología , Animales , Regulación del Apetito/efectos de los fármacos , Tronco Encefálico/inmunología , Tronco Encefálico/metabolismo , Núcleo Hipotalámico Dorsomedial/inmunología , Núcleo Hipotalámico Dorsomedial/metabolismo , Hormonas Gastrointestinales/administración & dosificación , Hormonas Gastrointestinales/inmunología , Expresión Génica , Péptido 1 Similar al Glucagón , Péptido 2 Similar al Glucagón , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Neurotransmisores/administración & dosificación , Neurotransmisores/inmunología , Péptidos/administración & dosificación , Péptidos/inmunología , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores de Glucagón/genética , Receptores de Glucagón/inmunología
7.
Int J Obes Relat Metab Disord ; 25 Suppl 5: S42-7, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11840214

RESUMEN

The pre-proglucagon derived peptides, glucagon-like peptide-1 (GLP-1) and glucagon-like peptide-2 (GLP-2) are both involved in a wide variety of peripheral functions, such as glucose homeostasis, gastric emptying, intestinal growth, insulin secretion as well as the regulation of food intake. Pre-proglucagon is also found in the brainstem in a small population of nerve cells in the nucleus of the solitary tract (NTS) that process the pre-propeptide as in the gut to yield GLP-1 and GLP-2. GLP-1 containing nerve fibres and the GLP-1 receptor are found predominantly in hypothalamic midline nuclei. GLP-1 given centrally to naive rats results in a marked induction of c-Fos protein in the supraoptic nucleus, paraventricular nucleus of the hypothalamus (PVN) and central nucleus of the amygdala, but only a moderate increase in the arcuate nucleus. The pattern of c-Fos activation is compatible with the appetite suppressing effects of GLP-1. This anorectic effect of GLP-1 appears to be mediated by the PVN, as direct injections of GLP-1 into this nucleus cause anorexia without concomitant taste aversion, suggesting a specific action upon neuronal circuits involved in the regulation of feeding. Recent experiments have also shown that GLP-1 is implicated in mediating signals from the gastrointestinal tract pertaining to discomfort and malaise. The distribution of the co-localised peptide, GLP-2, displays a perfect overlap with GLP-1 in the CNS with the highest concentration in the diffuse ventral part of the dorsomedial nucleus (DMHv). In contrast to the widely distributed GLP-1 receptor mRNA, GLP-2 receptor mRNA is exclusively expressed in the compact part of the DMH (DMHc). Interestingly, the DMHc is also the only nucleus responding to central administration of GLP-2 with a significant increase in the number of c-Fos positive cells. When injected into the lateral ventricle, GLP-2 has a marked inhibitory effect on feeding. The effect of GLP-2 on feeding is both behaviourally and pharmacologically specific. Future experiments will elucidate whether or not GLP-1 and GLP-2 are involved in the long-term or short-term regulation of feeding behaviour and hence have an impact on bodyweight.


Asunto(s)
Ingestión de Alimentos/fisiología , Glucagón/fisiología , Homeostasis , Fragmentos de Péptidos/fisiología , Precursores de Proteínas/fisiología , Angiotensina II/farmacología , Animales , Conducta Animal/fisiología , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Líquidos/fisiología , Ayuno/fisiología , Péptido 1 Similar al Glucagón , Masculino , Neuropéptido Y/farmacología , Ratas
8.
Nat Med ; 6(7): 802-7, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10888930

RESUMEN

The dorsomedial hypothalamic nucleus harbors leptin sensitive neurons and is intrinsically connected to hypothalamic nuclei involved in feeding behavior. However, it also receives ascending input from the visceroceptive neurons of the brainstem. We have identified a unique glucagon-like-peptide-2 containing neuronal pathway connecting the nucleus of the solitary tract with the dorsomedial hypothalamic nucleus. A glucagon-like-peptide-2 fiber plexus targets neurons expressing its receptor within the dorsomedial hypothalamic nucleus. Pharmacological and behavioral studies confirmed that glucagon-like-peptide-2 signaling is a specific transmitter inhibiting rodent feeding behavior and with potential long-term effects on body weight homeostasis. The glucagon-like-peptide-1 receptor antagonist, Exendin (9-39) is also a functional antagonist of centrally applied glucagon-like-peptide-2.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Glucagón/metabolismo , Neurotransmisores/farmacología , Péptidos/farmacología , Precursores de Proteínas/metabolismo , Animales , Condicionamiento Operante , Núcleo Hipotalámico Dorsomedial/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Péptido 1 Similar al Glucagón , Péptido 2 Similar al Glucagón , Hipotálamo/anatomía & histología , Masculino , Neuropéptido Y/farmacología , Fragmentos de Péptidos/farmacología , Péptidos/genética , Péptidos/aislamiento & purificación , Proglucagón , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Ratas , Ratas Wistar , Gusto
9.
Endocrinology ; 141(2): 794-801, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10650962

RESUMEN

We have recently shown that intracerebroventricular (i.c.v.) administration of the hypothalamic neuropeptide cocaine-amphetamine-regulated transcript (CART) inhibits food intake and induces the expression of c-fos in several nuclei involved in the regulation of food intake. A high number of CART-induced c-Fos-positive nuclei in the paraventricular nucleus of the hypothalamus prompted us to examine the effect of i.c.v. recombinant CART-(42-89) on activation of CRH-, oxytocin-, and vasopressin-synthesizing neuroendocrine cells in the paraventricular nucleus (PVN). In addition, plasma levels of glucose were examined after central administration of CART-(42-89). Seventy-six male Wistar rats were fitted with i.c.v. cannulas and singly housed under 12-h light, 12-h dark conditions. One week postsurgery the animals were injected i.c.v. in the morning with 0.5 microg recombinant CART-(42-89) or saline. Trunk blood was collected by decapitation at 0 (baseline), 10, 20, 40, 60, 120, or 240 min. CART caused a strong increase in circulating corticosterone that was significantly different from saline at 20, 40, 60, and 120 min postinjection (P<0.05). Furthermore, CART caused a transient rise in plasma oxytocin levels (P<0.05 at 10 and 20 min postinjection), whereas plasma vasopressin levels were unaffected by i.c.v. CART. Animals injected i.c.v. with CART showed a rise in blood glucose levels 10 min postinjection (P<0.05). To examine whether the stimulatory effect of i.c.v. CART on corticosterone and oxytocin secretion is caused by activation of paraventricular nucleus/supraoptic nucleus (PVN/SON) neuroendocrine neurons, we used c-Fos as a marker of neuronal activity. Animals injected with CART showed a strong increase in c-Fos-immunoreactive nuclei in the PVN. Double immunohistochemistry revealed that a high (89+/-0.4%) number of CRH-immunoreactive neurons in the PVN contained c-Fos after CART i.c.v.. c-Fos expression was also observed in oxytocinergic cells (in both magnocellular and parvicellular PVN neurons as well as in the supraoptic nuclei) 120 min after CART administration, whereas none of the vasopressinergic neurons contained c-Fos. Triple immunofluorescence microscopy revealed that CART-immunoreactive fibers closely apposed c-Fos-positive CRH neurons, suggestive of a direct action of CART on PVN CRH neurons. In summary, i.c.v. CART activates central CRH neurons as well as both magnocellular (presumably neurohypophysial) and parvicellular (presumably descending) oxytocinergic neurons of the PVN. The effect of CART on CRH neurons most likely leads to corticosterone secretion from the adrenal gland, which may contribute to the inhibitory effects of CART on feeding behavior.


Asunto(s)
Ventrículos Cerebrales/fisiología , Proteínas del Tejido Nervioso/farmacología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Animales , Arginina Vasopresina/sangre , Arginina Vasopresina/metabolismo , Glucemia/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ventrículos Cerebrales/efectos de los fármacos , Corticosterona/sangre , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/sangre , Hormona Liberadora de Corticotropina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Proteínas del Tejido Nervioso/administración & dosificación , Neuronas/efectos de los fármacos , Oxitocina/sangre , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Ratas Wistar
10.
J Clin Endocrinol Metab ; 84(10): 3781-91, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10523030

RESUMEN

The orexigenic role of central neuropeptide Y (NPY) in nonhuman primates has been questioned. Therefore, we have studied the effect of central NPY on feeding in ad libitum-fed male rhesus macaques. NPY dose-dependently increased food intake, with the maximal effect obtained by 50 microg (960 min food intake +/- SEM, 104 +/- 5 to 188 +/- 11 g; vehicle vs. NPY; n = 6). Blood glucose levels were unaffected by intracerebroventricular administration of NPY, but animals receiving either 20 or 50 microg displayed increased plasma levels of insulin and cortisol at few time points. To assess the pharmacological specificity of this response, a novel Y1 antagonist, [(Ile,Glu,Pro,Daba,Tyr,Arg,Leu,Arg,Tyr-NH2)2 cyclic (2,4'),(2',4)-diamide] (Y1ANT), was synthesized. Receptor binding experiments demonstrated that Y1ANT preferentially binds to Y1 and Y4 receptors (pKi 10.12 +/- 0.06 and 9.11 +/- 0.05 nmol/L, respectively). Functional analysis revealed that Y1ANT is a Y1 antagonist and a partial Y4 agonist. Central administration of Y1ANT blocked NPY-induced feeding. In food-deprived monkeys, Y1ANT attenuated the feeding response. However, Y1ANT had no effect on food intake in satiated monkeys. Thus, endogenous NPY is likely to be involved in the regulation of food intake in the nonhuman primate, and this effect is at least partially mediated via Y1-like receptors.


Asunto(s)
Encéfalo/metabolismo , Ingestión de Alimentos/fisiología , Receptores de Neuropéptido Y/fisiología , Animales , Clonidina/farmacología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Hidrocortisona/sangre , Inyecciones Intraventriculares , Insulina/sangre , Macaca mulatta , Masculino , Neuropéptido Y/agonistas , Neuropéptido Y/antagonistas & inhibidores , Neuropéptido Y/farmacología , Péptidos Cíclicos/farmacología , Receptores de Neuropéptido Y/antagonistas & inhibidores
11.
Neuroreport ; 10(6): 1183-7, 1999 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-10363921

RESUMEN

The adipose tissue hormone leptin, which is secreted to the general circulation and transported into the brain in a facilitated manner, possibly acts via hypothalamic neurones to reduce food intake and increase energy expenditure. To evaluate the involvement of importance of the arcuate nucleus in leptin induced anorexia, groups of rats treated neonatally with monosodium-glutamate (MSG; arcuate lesioned) and littermate controls were injected centrally with 5 microg recombinant leptin or saline daily for three consecutive days. Leptin significantly inhibited food intake and caused weight-loss in non-MSG rats (-14.5+/-3.0 g vs. 10.2+/-4.3 g; mean +/-s.e.m.; leptin vs. vehicle) whereas MSG-treated rats were unresponsive to leptin treatment (5.0+/-2.2 g vs. 0.8+/-3.8 g; leptin vs. vehicle). The present data indicate that an intact arcuate nucleus is necessary for leptins actions on food intake and body weight.


Asunto(s)
Anorexia/inducido químicamente , Núcleo Arqueado del Hipotálamo/fisiología , Ventrículos Cerebrales/fisiología , Proteínas/farmacología , Tejido Adiposo/fisiología , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Corticosterona/sangre , Inyecciones Intraventriculares , Leptina , Masculino , Proteínas/administración & dosificación , Proteínas/farmacocinética , Ratas , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacología , Glutamato de Sodio/toxicidad
12.
Brain Res ; 818(2): 499-509, 1999 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-10082837

RESUMEN

We have recently shown that the hypothalamic neuropeptide CART (cocaine-amphetamine-regulated-transcript) is a leptin dependent endogenous satiety factor in the rat. In the present study we confirm and extend our previous observations by showing that intracerebroventricular (i.c.v.) administered CART(42-89) dose-dependently inhibits 3-h food intake in food restricted rats with a lowest effective dose of 0.5 microgram. CART also potently inhibits NPY-induced food intake in satiated rats as well as nighttime food intake in free feeding animals. To identify brain areas potentially involved in mediating the anorectic effects of CART, the temporal expression pattern of the immediate early gene c-fos was examined in the central nervous system by immunohistochemistry in rats receiving recombinant CART. Compared to vehicle, CART induced c-Fos expression in several hypothalamic and brainstem structures implicated in the central control of food intake. In the hypothalamus, high numbers of c-Fos immunoreactive (-ir) cells were observed in the medial parvocellular part of the paraventricular nucleus and in the posterior part of the dorsomedial nucleus. Lower numbers of c-Fos positive nuclei were found in the supraoptic and arcuate nuclei. A relatively high number of c-Fos-ir cells was found in the central nucleus of the amygdala. In the brainstem, c-Fos-positive nuclei were found in the parabrachial nucleus, and in the nucleus of the solitary tract. Notably both the area postrema and the dorsal motor nucleus of the vagus were virtually devoid of c-Fos-ir cells. The present experiments suggest that CART peptide exerts its inhibitory effects on appetite by activating hypothalamic and brainstem neurones implicated in the central control of feeding behaviour and metabolism.


Asunto(s)
Depresores del Apetito/farmacología , Conducta Alimentaria/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Neuropéptidos/farmacología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , ARN Mensajero/biosíntesis , Anfetamina/farmacología , Animales , Cocaína/farmacología , Inmunohistoquímica , Leptina , Masculino , Obesidad/sangre , Proteínas/fisiología , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología , Respuesta de Saciedad/efectos de los fármacos
13.
J Clin Endocrinol Metab ; 84(2): 711-7, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10022442

RESUMEN

The present study was performed to determine the effects of central administration of leptin on food intake and sympathetic nervous system activity in a nonrodent species, the rhesus monkey. Peripheral administration of leptin at doses (1 and 3 microg/kg, s.c.) that produced increments of circulating leptin concentrations within a physiological range did not inhibit food intake over the subsequent 3 days. In contrast, leptin (1 microg/kg, intracerebroventricularly) had no acute effect on food intake, but caused a significant and sustained suppression (40-50%) of food intake during the entire following day (P < 0.01). In addition, circulating norepinephrine levels increased by 55 +/- 16% (P < 0.02) 1 h after intracerebroventricular leptin administration, but did not increase after artificial cerebrospinal fluid administration. These results indicate that leptin can provide a signal to the central nervous system that decreases food intake in primates and in addition acutely activates the sympathetic nervous system. However, the results showing an acute increase in circulating leptin concentrations after peripheral administration of human leptin suggest that in primates, increases in circulating leptin within the physiological range do not acutely regulate food intake. Leptin may be more important in regulating food intake when there are sustained changes in circulating concentrations of leptin (e.g. with obesity, prolonged energy restriction, or diabetes).


Asunto(s)
Encéfalo/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Proteínas/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Epinefrina/sangre , Inyecciones Intraventriculares , Cinética , Leptina , Macaca mulatta , Masculino , Norepinefrina/sangre , Proteínas/administración & dosificación , Sistema Nervioso Simpático/fisiología
14.
J Endocrinol ; 159(2): 307-12, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9795372

RESUMEN

A number of neuropeptide Y (NPY) receptor subtypes, including the recently cloned Y5 receptor, have been implicated in the stimulation of food intake. In the present study, Y5 receptor antisense oligodeoxynucleotides (ODNs) were used to assess the potential involvement of the Y5 receptor in the regulation of spontaneous as well as NPY-induced food intake. Repeated central administration of Y5 antisense ODN significantly decreased spontaneous food intake and subsequently resulted in a significant weight loss. Furthermore, Y5 antisense ODN pre-treatment significantly inhibited the robust feeding response elicited by central administration of NPY (5.3+/-0. 8 vs 1.08+/-0.28 g, vehicle+/-s.e.m. vs Y5 ODN+/-s.e.m.). The present results provide evidence that central Y5 receptors are involved in both spontaneous as well as NPY-induced food intake, which may prove to be a new therapeutic route in the treatment of obesity and other disorders of appetite.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Neuropéptido Y/farmacología , Oligonucleótidos Antisentido/administración & dosificación , Receptores de Neuropéptido Y/fisiología , Receptores de Neuropéptido/fisiología , Análisis de Varianza , Animales , Peso Corporal/efectos de los fármacos , Insulina/sangre , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores de Neuropéptido Y/genética
15.
Diabetes ; 47(4): 530-7, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9568683

RESUMEN

In the rat, the glucagon-like peptide 1 (GLP-1)(7-36) amide inhibits neurones in the central nervous system responsible for food and water intake. GLP-1-induced inhibition of food intake may involve the hypothalamic arcuate nucleus, whereas rostral sensory circumventricular organs may be responsible for the inhibitory action of GLP-1 on drinking. To further investigate the role of these blood-brain-barrier-free areas in GLP-1-induced inhibition of ingestive behavior, neonatal Wistar rats were subjected to monosodium glutamate (MSG) treatment, which causes extensive damage to the arcuate nucleus as well as to parts of the sensory circumventricular organs. The inhibitory effect of GLP-1 on feeding induced by food deprivation was completely abolished in MSG-lesioned rats. This effect was not due to either a loss of sensitivity to anorectic agents or a loss of taste aversion because MSG-treated animals displayed normal anorectic responses to central administration of corticotropin-releasing factor and normal aversive responses to peripheral administration of both lithium chloride and D-amphetamine. In non-lesioned rats, neuropeptide Y (NPY)-induced feeding was significantly reduced by concomitant GLP-1 administration. In contrast, GLP-1 had no effect on NPY-induced feeding in MSG-lesioned rats, suggesting that the GLP-1 receptors that mediate inhibition of feeding are localized upstream to the NPY-sensitive neurones inducing feeding behavior. The inhibitory effect of GLP-1 on water intake was tested using an ANG II-elicited drinking paradigm. Central administration of GLP-1 inhibited ANG II drinking in both MSG-treated rats and their nontreated littermates. In contrast, peripheral administration of GLP-1 did not inhibit ANG II-induced drinking behavior in MSG-treated rats. Thus it is evident that centrally acting GLP-1 modulates feeding and drinking behavior via neurones sensitive to MSG lesioning in the arcuate nucleus and circumventricular organs, respectively.


Asunto(s)
Encéfalo/fisiología , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Aditivos Alimentarios/toxicidad , Fragmentos de Péptidos/metabolismo , Glutamato de Sodio/toxicidad , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/fisiología , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Glucemia/metabolismo , Barrera Hematoencefálica/fisiología , Encéfalo/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/fisiología , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Privación de Alimentos , Glucagón , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón , Masculino , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Ratas , Ratas Wistar , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/fisiología , Gusto/efectos de los fármacos , Gusto/fisiología
16.
Endocrinology ; 138(10): 4445-55, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9322962

RESUMEN

Within the central nervous system, glucagon-like peptide-1-(7-36) amide (GLP-1) acts as a transmitter, inhibiting feeding and drinking behavior. Hypothalamic neuroendocrine neurons are centrally involved in the regulatory mechanisms controlling these behaviors, and high densities of GLP-1 binding sites are present in the rat hypothalamus. In the present study we have, over a period of 4 h, followed the effect of centrally injected GLP-1 on plasma levels of the neurohypophysial hormones vasopressin and oxytocin. Plasma levels of corticosterone and glucose were also followed across time after central administration of GLP-1. In conscious, freely moving, and unstressed rats, central injection of GLP-1 significantly elevated plasma levels of vasopressin 15 and 30 min after administration (basal, 0.8 +/- 0.2 pg/ml; 15 min, 7.5 +/- 2.0 pg/ml; 30 min, 5.6 +/- 1.1 pg/ml; mean +/- SEM) and elevated corticosterone 15 min after administration (52 +/- 13 vs. 447 +/- 108 ng/ml, basal vs. 15 min; mean +/- SEM). In contrast, plasma oxytocin levels were unaffected by intracerebroventricular (icv) injections of GLP-1 over a period of 4 h after the injection. The animals given a central injection of GLP-1 developed transient hypoglycemia 20 min after the injection, which was fully restored to normal levels at 30 min. Furthermore, we used c-fos immunocytochemistry as an index of stimulated neuronal activity. The distribution and quantity of GLP-1-induced c-fos immunoreactivity were evaluated in a number of hypothalamic neuroendocrine areas, including the magnocellular neurons of the paraventricular (PVN) and supraoptic (SON) nuclei and the parvicellular neurons of the medial parvicellular subregion of the PVN. The number of c-fos-expressing nuclei in those areas was assessed 30, 60, and 90 min after icv administration of GLP-1. Intracerebroventricular injection of GLP-1 induced c-fos expression in the medial parvicellular subregion of the PVN as well as in magnocellular neurons of the PVN and SON. A slight induction of c-fos expression was seen in the arcuate nucleus and the nucleus of the solitary tract, including the area postrema. In contrast, the subfornical organ, which is a rostrally situated circumventricular organ, was free of c-fos-positive cells after central administration of GLP-1. When the GLP-1 antagonist exendin-(9-39) was given before the GLP-1, c-fos expression in these neuroendocrine areas was almost completely abolished, suggesting that the effect of GLP-1 on c-fos expression is mediated via specific receptors. A dual labeling immunocytochemical technique was used to identify the phenotypes of some of the neurons containing c-fos-immunoreactive nuclei. Approximately 80% of the CRH-positive neurons in the hypophysiotropic medial parvicellular part of the PVN coexpressed c-fos 90 min after icv GLP-1 administration. In contrast, very few (approximately 10%) of the vasopressinergic magnocellular neurons of the PVN/SON contained c-fos-positive nuclei, whereas approximately 38% of the magnocellular oxytocinergic neurons expressed c-fos-positive nuclei in response to GLP-1 administration. This study demonstrates that central administration of the anorectic neuropeptide GLP-1 activates the central CRH-containing neurons of the hypothalamo-pituitary-adrenocortical axis as well as oxytocinergic neurons of the hypothalamo-neurohypophysial tract. Therefore, we conclude that GLP-1 activates the hypothalamo-pituitary-adrenocortical axis primarily through stimulation of CRH neurons, and this activation may also be responsible for the inhibition of feeding behavior.


Asunto(s)
Hipotálamo/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Animales , Núcleo Arqueado del Hipotálamo/química , Núcleo Arqueado del Hipotálamo/citología , Glucemia/análisis , Glucemia/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/análisis , Hormona Liberadora de Corticotropina/genética , Regulación de la Expresión Génica , Glucagón , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón , Sistema Hipotálamo-Hipofisario/fisiología , Hipotálamo/química , Inmunohistoquímica , Inyecciones Intraventriculares , Masculino , Neuronas/química , Oxitocina/análisis , Oxitocina/sangre , Oxitocina/genética , Núcleo Hipotalámico Paraventricular/química , Núcleo Hipotalámico Paraventricular/citología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/fisiología , Fenotipo , Sistema Hipófiso-Suprarrenal/fisiología , Proteínas Proto-Oncogénicas c-fos/análisis , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Ratas Wistar , Núcleo Supraóptico/química , Núcleo Supraóptico/citología , Factores de Tiempo , Vasopresinas/análisis , Vasopresinas/sangre , Vasopresinas/genética
17.
Neuroscience ; 77(1): 257-70, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9044391

RESUMEN

Central administration of the preproglucagon-derived peptide glucagon-like peptide-1 significantly inhibits ingestion of food and water, and glucagon-like peptide-1 binding sites are present in a multitude of central areas involved in the regulation of ingestional behaviour. To evaluate further the neuroanatomical organization of central glucagon-like peptide-1 containing neuronal circuits with potential implications on ingestional behaviour, we carried out a series of experiments in the rat demonstrating the topographical sites of synthesis and processing of the preproglucagon precursor followed by a chromatographic analysis of the processed fragments. In situ hybridization histochemistry revealed that preproglucagon encoding messenger RNA was expressed in a single population of neurons in the caudal portion of the nucleus of the solitary tract. Gel chromatographic analysis of hypothalamic and brainstem tissue extracts revealed that the preproglucagon precursor is processed in a fashion similar to that seen in the small intestine, preferentially giving rise to glicentin, glucagon-like peptide-1 and glucagon-like peptide-2. This single brain site of glucagon-like peptide-1 synthesis was subsequently confirmed by immunohistochemical demonstration of glucagon-like peptide-1-immunoreactive perikarya in the central and caudal parts of the nucleus of the solitary tract. Numerous sites containing glucagon-like peptide-1 immunoreactive fibres were, however, discovered in the forebrain including hypothalamic, thalamic and cortical areas. The densest innervation by glucagon-like peptide-1 immunoreactive nerve fibres was seen in the hypothalamic dorsomedial and paraventricular nuclei, but numerous glucagon-like peptide-1 immunoreactive fibres were also seen throughout the periventricular strata of the third ventricle. Dual-labelling immunohistochemistry for tyrosine hydroxylase and glucagon-like peptide-1 gave no evidence for co-localization of catecholamines and glucagon-like peptide-1 in neurons of the lower brainstem. To identify neurons of the nucleus of the solitary tract that project to the hypothalamic paraventricular nucleus, the retrograde tracer FluoroGold was injected into this hypothalamic target and dual immunocytochemical identification of glucagon-like peptide-1 and tyrosine hydroxylase-positive neurons was performed on brainstem sections containing retrogradely labelled perikarya. From this experiment it was seen that many of the retrogradely labelled neurons in the central portion of the nucleus of the solitary tract are catecholaminergic, while none is glucagon-like peptide-1 immunoreactive. In contrast, most of the retrogradely labelled neurons of the caudal portion of the nucleus of the solitary tract contain glucagon-like peptide-1. These observations further substantiate that glucagon-like peptide-1 neurons of the solitary tract constitute a distinct non-catecholaminergic cell group which projects to many targets, one of which is the hypothalamic paraventricular nucleus.


Asunto(s)
Glucagón/análisis , Glucagón/genética , Núcleo Hipotalámico Paraventricular/química , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/genética , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Núcleo Solitario/química , Núcleo Supraóptico/química , Animales , Cromatografía en Gel , Péptido 1 Similar al Glucagón , Inmunohistoquímica , Hibridación in Situ , Masculino , Fibras Nerviosas/química , Neurosecreción/fisiología , Núcleo Hipotalámico Paraventricular/citología , ARN Mensajero/análisis , Radioinmunoensayo , Ratas , Ratas Wistar , Núcleo Solitario/citología , Núcleo Supraóptico/citología
18.
Am J Physiol ; 271(4 Pt 2): R848-56, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8897973

RESUMEN

Glucagon-like peptide (GLP)-1-(7-36) amide and its pancreatic receptors are important for control of blood glucose levels. However, rat GLP-1 receptors are also localized in the brain, in hypothalamus, and in areas without a blood-brain barrier. When rats were kept on a food restriction schedule, intracerebroventricular injection of GLP-1 just before food was offered inhibited food intake. However, peripheral GLP-1 administration by intraperitoneal injection had little effect. GLP-1 effects on water intake and output were also investigated. Intracerebroventricular GLP-1 profoundly inhibited angiotensin II-induced drinking behavior in rats, and water intake was suppressed by exogenous GLP-1 in rats habituated to a water restriction schedule. These effects were reproduced by intraperitoneal administration of GLP-1. Furthermore, intracerebroventricular GLP-1 stimulated urinary excretion of water and sodium. The centrally elicited effects were blocked by the GLP-1 antagonist exendin-(9-39) amide, whereas the N-terminally extended and inactive GLP-1-(1-36) amide had no effect on feeding and drinking. GLP-1 had no effect in behavioral assays measuring exploratory locomotor activity and conditioned taste aversion. In conclusion, GLP-1 may play a physiological role in regulation of both ingestion and the water and salt homeostasis.


Asunto(s)
Encéfalo/fisiología , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Animales , Conducta Animal/efectos de los fármacos , Glucagón , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón , Inyecciones Intraventriculares , Masculino , Natriuresis/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA