Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Regul Pept ; 167(2-3): 201-8, 2011 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21315111

RESUMO

The aim of this study was to investigate the central actions of the stable pansomatostatin peptide agonist, ODT8-SST on body weight. ODT8-SST or vehicle was acutely (1µg/rat) injected or chronically infused (5µg/rat/d, 14d) intracerebroventricularly and daily food intake, body weight and composition were monitored. In lean rats, neither acute nor chronic ODT8-SST influenced daily food intake while body weight was reduced by 2.2% after acute injection and there was a 14g reduction of body weight gain after 14d compared to vehicle (p<0.01). In diet-induced obese (DIO) rats, chronic ODT8-SST increased cumulative 2-week food intake compared to vehicle (+14%, p<0.05) and also blunted body weight change (-11g, p<0.05). ODT8-SST for 14d reduced lean mass (-22g and -25g respectively, p<0.001) and total water (-19g and -22g respectively, p<0.001) in lean and DIO rats and increased fat mass in DIO (+16g, p<0.001) but not lean rats (+1g, p>0.05) compared to vehicle. In DIO rats, ODT8-SST reduced ambulatory (-27%/24h, p<0.05) and fine movements (-38%, p<0.01) which was associated with an increased positive energy balance compared to vehicle (+50g, p<0.01). Chronic central somatostatin receptor activation in lean rats reduces body weight gain and lean mass independently of food intake which is likely related to growth hormone inhibition. In DIO rats, ODT8-SST reduces lean mass but promotes food intake and fat mass, indicating differential responsiveness to somatostatin under obese conditions.


Assuntos
Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Somatostatina/análogos & derivados , Somatostatina/agonistas , Aumento de Peso/efeitos dos fármacos , Animais , Dieta , Injeções , Masculino , Obesidade/etiologia , Obesidade/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/metabolismo , Somatostatina/administração & dosagem , Somatostatina/farmacologia , Magreza/metabolismo
2.
Endocrinology ; 151(9): 4224-35, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20610566

RESUMO

Somatostatin and octreotide injected into the brain have been reported to modulate food intake. However, little is known regarding the underlying mechanisms. The stable oligosomatostatin analog, des-AA(1,2,4,5,12,13)-[DTrp(8)]-somatostatin (ODT8-SST), like somatostatin, binds to all five somatostatin receptors (sst(1-5)). We characterized the effects of ODT8-SST injected intracerebroventricularly (i.c.v.) on food consumption and related mechanisms of action in freely fed rats. ODT8-SST (0.3 and 1 microg per rat, i.c.v.) injected during the light or dark phase induced an early onset (within 1 h) and long-lasting (4 h) increase in food intake in nonfasted rats. By contrast, i.p. injection (0.3-3 mg/kg) or i.c.v. injection of selective sst(1) or sst(4) agonists (1 microg per rat) had no effect. The 2 h food intake response during the light phase was blocked by i.c.v. injection of a sst(2) antagonist, the neuropeptide Y (NPY) Y(1) receptor antagonist, BIBP-3226, and ip injection of the mu-opioid receptor antagonist, naloxone, and not associated with changes in plasma ghrelin levels. ODT8-SST (1 microg per rat, i.c.v.) stimulated gastric emptying of a solid meal which was also blocked by naloxone. The increased food intake was accompanied by a sustained increase in respiratory quotient, energy expenditure, and drinking as well as mu-opioid receptor-independent grooming behavior and hyperthermia, while ambulatory movements were not altered after ODT8-SST (1 microg per rat, i.c.v.). These data show that ODT8-SST acts primarily through brain sst(2) receptors to induce a long-lasting orexigenic effect that involves the activation of Y(1) and opiate-receptors, accompanied by enhanced gastric transit and energy expenditure suggesting a modulation of NPYergic and opioidergic orexigenic systems by brain sst(2) receptors.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Somatostatina/fisiologia , Somatostatina/análogos & derivados , Animais , Ansiolíticos/administração & dosagem , Ansiolíticos/farmacologia , Arginina/administração & dosagem , Arginina/análogos & derivados , Arginina/farmacologia , Temperatura Corporal/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Metabolismo Energético/efeitos dos fármacos , Esvaziamento Gástrico/efeitos dos fármacos , Asseio Animal/efeitos dos fármacos , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naloxona/administração & dosagem , Naloxona/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/fisiologia , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/fisiologia , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores , Somatostatina/administração & dosagem , Somatostatina/farmacologia
3.
J Pharmacol Exp Ther ; 329(2): 429-38, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19182070

RESUMO

The mammalian neuropeptide, melanin-concentrating hormone, interacts with two G protein-coupled receptors, melanin-concentrating hormone receptor (MCHR) 1 and MCHR2; however, only MCHR1 is expressed in rats and mice. In the present study, we evaluated MCHR1 antagonism in preclinical models believed to be predictive of antiobesity and antidepressant activity. Central activity of the selective MCHR1 antagonist, GW803430 [6-(4-chloro-phenyl)-3-[3-methoxy-4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-3H-thieno[3,2-d]pyrimidin-4-one], was evaluated using ex vivo binding with autoradiography. Effective doses of GW803430 (1 and 3 mg/kg p.o.) were correlated with antiobesity activity in a 14-day study of diet-induced obese rats. GW803430 was evaluated subsequently for antidepressant-like effects in mice and rats. Acute and subchronic administration reduced immobility time in the mouse forced-swim test at doses of 3 (acute) and 3 and 10 (chronic) mg/kg p.o., an effect that was absent in MCHR1(-/-) mice. Combined subeffective doses of GW803430 (0.3 and 1 mg/kg p.o.) and imipramine (5 mg/kg) produced a robust antidepressant-like response. The compound was also active in the tail suspension test at a dose of 10 mg/kg p.o. GW803430 (30 mg/kg p.o.) significantly reduced submissive behaviors at weeks 2 and 3, a model of submissive behavior that may predict antidepressant onset. GW803430 decreased marble burying in mice at doses of 3, 10, and 30 mg/kg p.o., an assay that detects anxiolytic-like effects. Thus, GW803430 produces robust antiobesity and antidepressant-like effects in rats and mice at doses that compete for central MCHR1 in vivo. As such, MCHR1 should be considered as a promising target for future drug discovery efforts.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Obesidade/tratamento farmacológico , Pirimidinonas/uso terapêutico , Receptores de Somatostatina/antagonistas & inibidores , Tiofenos/uso terapêutico , Animais , Fármacos Antiobesidade/administração & dosagem , Fármacos Antiobesidade/farmacologia , Antidepressivos/administração & dosagem , Antidepressivos/farmacologia , Autorradiografia , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Depressão/metabolismo , Depressão/fisiopatologia , Depressão/psicologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade/psicologia , Ligação Proteica , Pirimidinonas/administração & dosagem , Pirimidinonas/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Receptores de Somatostatina/genética , Predomínio Social , Natação , Tiofenos/administração & dosagem , Tiofenos/farmacologia
4.
Obesity (Silver Spring) ; 15(11): 2643-52, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18070755

RESUMO

OBJECTIVES: Obestatin has been initially characterized as a new peptide derived from the ghrelin precursor, which suppresses food intake and inhibits the orexigenic and prokinetic actions of ghrelin when injected peripherally or centrally in lean mice. However, reproducing these data remains controversial. Reasons for the disparity may be the use of different doses, routes, and animal models. We aimed to investigate the effects of peripheral and intracisternal (IC) injection of obestatin on feeding, gastric motility, and blood glucose in rats as well as in diet-induced obese (DIO) mice. RESEARCH METHODS AND PROCEDURES: Food intake and gastric emptying of a semi-liquid caloric meal were measured after intraperitoneal (IP) injection of obestatin in rats and DIO mice. Gastric phasic motility and blood glucose were monitored in urethane-anesthetized rats after IC or intravenous (IV) injection of obestatin. RESULTS: Obestatin injected intraperitoneally at doses ranging from 0.1 to 3 mg/kg influenced neither acute food intake nor gastric emptying in rats. Obestatin injected intravenously at 0.3 or 3 mg/kg and IC at 7.5 or 30 microg/rat modified neither fasted gastric phasic motility nor blood glucose levels, while ghrelin (30 microg/kg, IV) increased and vagotomy suppressed gastric motility, and an oligosomatostatin analog (3 microg/rat, IC) decreased blood glucose. Obestatin, injected intraperitoneally (0.3 mg/kg) in DIO mice, did not alter feeding response to a fast, while urocortin 1 (10 microg/kg, IP) induced a 73.3% inhibition at 2 hours. DISCUSSION: Our data demonstrate that peripheral administration of obestatin did not modify food intake in rats or obese mice or gastric motor function in rats.


Assuntos
Ingestão de Alimentos/fisiologia , Grelina/fisiologia , Obesidade/fisiopatologia , Magreza/fisiopatologia , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Esvaziamento Gástrico/fisiologia , Grelina/administração & dosagem , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Somatostatina/fisiologia , Urocortinas/fisiologia
5.
Peptides ; 26(10): 1988-96, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15985311

RESUMO

Human beta-MSH(1-22) was first isolated from human pituitary as a 22-amino acid (aa) peptide derived from a precursor protein, pro-opiomelanocortin (POMC). However, Bertagna et al. demonstrated that a shorter human beta-MSH(5-22), (DEGPYRMEHFRWGSPPKD), is a true endogenous peptide produced in human hypothalamus. In this report, we demonstrated that in vitro enzymatic cleavage of native human beta-MSH(5-22) with two ubiquitous dipeptidyl peptidases (DPP), DPP-I and DPP-IV, generated two potent MC3/4R peptide analogues, beta-MSH(7-22) (GPYRMEHFRWGSPPKD) and beta-MSH(9-22) (YRMEHFRWGSPPKD). In fact, the MC4R binding affinity and functional potency of beta-MSH(7-22) (Ki=4.6 nM, EC50=0.6 nM) and beta-MSH(9-22) (Ki=5.7 nM, EC50=0.6 nM) are almost an order of magnitude greater than those of their parent peptide, beta-MSH(5-22) (MC4R, Ki=23 nM, EC50= 3nM). Furthermore, the DPP-I/DPP-IV cleaved peptide, beta-MSH(9-22), when administered intracerebroventricularly (ICV) at a dose of 3 nmol/rat, potently induced an acute negative energy balance in a diet-induced obese rat model, while its parent molecule, beta-MSH(5-22), administered at the same dose did not have any effect. These data suggest that DPP-I and DPP-IV may play a role in converting the endogenous beta-MSH(5-22) to more potent peptides that regulate energy homeostasis in the hypothalamus.


Assuntos
Catepsina C/fisiologia , Dipeptidil Peptidase 4/fisiologia , Peptídeos/agonistas , Receptor Tipo 3 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/agonistas , beta-MSH/metabolismo , Animais , Catepsina C/química , Linhagem Celular , Dipeptidil Peptidase 4/química , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Peptídeos/metabolismo , Ratos , Ratos Long-Evans , Receptor Tipo 3 de Melanocortina/química , Receptor Tipo 3 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/química , Receptor Tipo 4 de Melanocortina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA