Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neural Eng ; 15(1): 016001, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28745303

RESUMO

OBJECTIVE: Vagal nerve stimulation (VNS) has shown potential benefits for obesity treatment; however, current devices lack physiological feedback, which limit their efficacy. Changes in extracellular pH (pHe) have shown to be correlated with neural activity, but have traditionally been measured with glass microelectrodes, which limit their in vivo applicability. APPROACH: Iridium oxide has previously been shown to be sensitive to fluctuations in pH and is biocompatible. Iridium oxide microelectrodes were inserted into the subdiaphragmatic vagus nerve of anaesthetised rats. Introduction of the gut hormone cholecystokinin (CCK) or distension of the stomach was used to elicit vagal nerve activity. MAIN RESULTS: Iridium oxide microelectrodes have sufficient pH sensitivity to readily detect changes in pHe associated with both CCK and gastric distension. Furthermore, a custom-made Matlab script was able to use these changes in pHe to automatically trigger an implanted VNS device. SIGNIFICANCE: This is the first study to show pHe changes in peripheral nerves in vivo. In addition, the demonstration that iridium oxide microelectrodes are sufficiently pH sensitive as to measure changes in pHe associated with physiological stimuli means they have the potential to be integrated into closed-loop neurostimulating devices.


Assuntos
Líquido Extracelular/fisiologia , Irídio/fisiologia , Estimulação do Nervo Vago/métodos , Nervo Vago/fisiologia , Animais , Líquido Extracelular/química , Irídio/química , Masculino , Microeletrodos , Ratos , Ratos Wistar , Estimulação do Nervo Vago/instrumentação
2.
Cell Rep ; 19(11): 2202-2209, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28614708

RESUMO

The obesity epidemic is a significant global health issue. Improved understanding of the mechanisms that regulate appetite and body weight will provide the rationale for the design of anti-obesity therapies. Thyroid hormones play a key role in metabolic homeostasis through their interaction with thyroid hormone receptors (TRs), which function as ligand-inducible transcription factors. The TR-beta isoform (TRß) is expressed in the ventromedial hypothalamus (VMH), a brain area important for control of energy homeostasis. Here, we report that selective knockdown of TRß in the VMH of adult mice results in severe obesity due to hyperphagia and reduced energy expenditure. The observed increase in body weight is of a similar magnitude to murine models of the most extreme forms of monogenic obesity. These data identify TRß in the VMH as a major physiological regulator of food intake and energy homeostasis.


Assuntos
Peso Corporal/genética , Ingestão de Alimentos/genética , Receptores beta dos Hormônios Tireóideos/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Peso Corporal/fisiologia , Masculino , Camundongos
3.
Endocrinology ; 158(8): 2680-2693, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28586439

RESUMO

Endogenous satiety hormones provide an attractive target for obesity drugs. Glucagon causes weight loss by reducing food intake and increasing energy expenditure. To further understand the cellular mechanisms by which glucagon and related ligands activate the glucagon receptor (GCGR), we investigated the interaction of the GCGR with receptor activity modifying protein (RAMP)2, a member of the family of receptor activity modifying proteins. We used a combination of competition binding experiments, cell surface enzyme-linked immunosorbent assay, functional assays assessing the Gαs and Gαq pathways and ß-arrestin recruitment, and small interfering RNA knockdown to examine the effect of RAMP2 on the GCGR. Ligands tested were glucagon; glucagonlike peptide-1 (GLP-1); oxyntomodulin; and analog G(X), a GLP-1/glucagon coagonist developed in-house. Confocal microscopy was used to assess whether RAMP2 affects the subcellular distribution of GCGR. Here we demonstrate that coexpression of RAMP2 and the GCGR results in reduced cell surface expression of the GCGR. This was confirmed by confocal microscopy, which demonstrated that RAMP2 colocalizes with the GCGR and causes significant GCGR cellular redistribution. Furthermore, the presence of RAMP2 influences signaling through the Gαs and Gαq pathways, as well as recruitment of ß-arrestin. This work suggests that RAMP2 may modify the agonist activity and trafficking of the GCGR, with potential relevance to production of new peptide analogs with selective agonist activities.


Assuntos
Glucagon/metabolismo , Proteína 2 Modificadora da Atividade de Receptores/metabolismo , Receptores de Glucagon/metabolismo , Animais , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , DNA , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Ligação Proteica , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Proteína 2 Modificadora da Atividade de Receptores/química , Receptores de Glucagon/genética , Transdução de Sinais , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
4.
Am J Physiol Endocrinol Metab ; 311(1): E42-55, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27189932

RESUMO

Glucokinase is a key component of the neuronal glucose-sensing mechanism and is expressed in brain regions that control a range of homeostatic processes. In this review, we detail recently identified roles for neuronal glucokinase in glucose homeostasis and counterregulatory responses to hypoglycemia and in regulating appetite. We describe clinical implications from these advances in our knowledge, especially for developing novel treatments for diabetes and obesity. Further research required to extend our knowledge and help our efforts to tackle the diabetes and obesity epidemics is suggested.


Assuntos
Apetite/fisiologia , Encéfalo/metabolismo , Diabetes Mellitus/metabolismo , Glucoquinase/metabolismo , Glucose/metabolismo , Hipoglicemia/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Animais , Homeostase , Humanos
6.
Peptides ; 32(11): 2290-4, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21835215

RESUMO

Ghrelin is a peptide hormone secreted into circulation from the stomach. It has been postulated to act as a signal of hunger. Ghrelin administration acutely increases energy intake in lean and obese humans and chronically induces weight gain and adiposity in rodents. Circulating ghrelin levels are elevated by fasting and suppressed following a meal. Inhibiting ghrelin signaling therefore appears an attractive target for anti-obesity therapies. A number of different approaches to inhibiting the ghrelin system to treat obesity have been explored. Despite this, over a decade after its discovery, no ghrelin based anti-obesity therapies are close to reaching the market. This article discusses the role of ghrelin in appetite control in humans, examines different approaches to inhibiting the ghrelin system and assesses their potential as anti-obesity therapies.


Assuntos
Aciltransferases/metabolismo , Regulação do Apetite/fisiologia , Apetite/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Grelina , Obesidade/tratamento farmacológico , Receptores de Grelina/metabolismo , Aciltransferases/antagonistas & inibidores , Tecido Adiposo , Adiposidade/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/uso terapêutico , Apetite/fisiologia , Regulação do Apetite/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Jejum , Grelina/antagonistas & inibidores , Grelina/sangue , Grelina/genética , Grelina/metabolismo , Humanos , Fome/efeitos dos fármacos , Camundongos , Obesidade/sangue , Obesidade/fisiopatologia , Hormônios Peptídicos/administração & dosagem , Hormônios Peptídicos/uso terapêutico , Peptídeo YY/metabolismo , Peptídeos/administração & dosagem , Peptídeos/uso terapêutico , Ratos , Receptores de Grelina/antagonistas & inibidores , Receptores de Grelina/genética , Resposta de Saciedade/efeitos dos fármacos , Substância P/administração & dosagem , Substância P/análogos & derivados , Substância P/uso terapêutico , Aumento de Peso
7.
Gastroenterology ; 138(7): 2468-76, 2476.e1, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20178795

RESUMO

BACKGROUND & AIMS: Ghrelin is the only peripheral hormone known to increase food intake. It is released from the stomach and is thought to function as a signal of energy deficit and a meal initiator. We generated transgenic mice in which levels of bioactive ghrelin are increased in the stomach and circulation. These mice, as expected, are hyperphagic and glucose intolerant. We investigated whether exposure to a high-fat diet (HFD) would exacerbate this phenotype. METHODS: We investigated the effect of HFD on energy and glucose homeostasis in ghrelin transgenic mice. We determined dietary preference; expression of hypothalamic neuropeptides that control food intake; and, using fast-performance liquid chromatography, the circulating forms of ghrelin. We measured food intake during continuous administration of ghrelin in wild-type mice fed either regular chow or an HFD. RESULTS: Ghrelin transgenic mice were resistant to diet-induced obesity because of their reduced food intake. This was not caused by alterations to food preference, hypothalamic signaling of neuropeptides that control food intake, or the form of circulating acylated ghrelin. Long-term administration of ghrelin to wild-type mice failed to increase ingestion of an HFD but, as expected, increased intake of regular chow. CONCLUSIONS: This is the first report that diets high in fat inhibit the hyperphagic effect of ghrelin; these findings indicate that features of the diet are important determinants of ghrelin's function. This information is important for the development of anti-obesity drugs that target ghrelin signaling.


Assuntos
Gorduras na Dieta/administração & dosagem , Grelina/farmacologia , Hiperfagia/prevenção & controle , Animais , Ingestão de Energia , Metabolismo Energético , Hiperfagia/induzido quimicamente , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Atividade Motora
8.
Diabetes ; 59(2): 397-406, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19933997

RESUMO

OBJECTIVE: Prokineticin 2 (PK2) is a hypothalamic neuropeptide expressed in central nervous system areas known to be involved in food intake. We therefore hypothesized that PK2 plays a role in energy homeostasis. RESEARCH DESIGN AND METHODS: We investigated the effect of nutritional status on hypothalamic PK2 expression and effects of PK2 on the regulation of food intake by intracerebroventricular (ICV) injection of PK2 and anti-PK2 antibody. Subsequently, we investigated the potential mechanism of action by determining sites of neuronal activation after ICV injection of PK2, the hypothalamic site of action of PK2, and interaction between PK2 and other hypothalamic neuropeptides regulating energy homeostasis. To investigate PK2's potential as a therapeutic target, we investigated the effect of chronic administration in lean and obese mice. RESULTS: Hypothalamic PK2 expression was reduced by fasting. ICV administration of PK2 to rats potently inhibited food intake, whereas anti-PK2 antibody increased food intake, suggesting that PK2 is an anorectic neuropeptide. ICV administration of PK2 increased c-fos expression in proopiomelanocortin neurons of the arcuate nucleus (ARC) of the hypothalamus. In keeping with this, PK2 administration into the ARC reduced food intake and PK2 increased the release of alpha-melanocyte-stimulating hormone (alpha-MSH) from ex vivo hypothalamic explants. In addition, ICV coadministration of the alpha-MSH antagonist agouti-related peptide blocked the anorexigenic effects of PK2. Chronic peripheral administration of PK2 reduced food and body weight in lean and obese mice. CONCLUSIONS: This is the first report showing that PK2 has a role in appetite regulation and its anorectic effect is mediated partly via the melanocortin system.


Assuntos
Ingestão de Energia/efeitos dos fármacos , Hormônios Gastrointestinais/farmacologia , Hormônios Gastrointestinais/fisiologia , Neuropeptídeos/farmacologia , Neuropeptídeos/fisiologia , Obesidade/fisiopatologia , Animais , Relação Dose-Resposta a Droga , Hormônios Gastrointestinais/genética , Regulação da Expressão Gênica , Hipotálamo/fisiologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Neuropeptídeos/genética , RNA Mensageiro/genética , Ratos , Ratos Wistar
9.
Diabetes ; 58(4): 840-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19151202

RESUMO

OBJECTIVE: Ghrelin is the only known peripheral hormone to increase ingestive behavior. However, its role in the physiological regulation of energy homeostasis is unclear because deletion of ghrelin or its receptor does not alter food intake or body weight in mice fed a normal chow diet. We hypothesized that overexpression of ghrelin in its physiological tissues would increase food intake and body weight. RESEARCH DESIGN AND METHODS: We used bacterial artificial chromosome transgenesis to generate a mouse model with increased ghrelin expression and production in the stomach and brain. We investigated the effect of ghrelin overexpression on food intake and body weight. We also measured energy expenditure and determined glucose tolerance, glucose stimulated insulin release, and peripheral insulin sensitivity. RESULTS: Ghrelin transgenic (Tg) mice exhibited increased circulating bioactive ghrelin, which was associated with hyperphagia, increased energy expenditure, glucose intolerance, decreased glucose stimulated insulin secretion, and reduced leptin sensitivity. CONCLUSIONS: This is the first report of a Tg approach suggesting that ghrelin regulates appetite under normal feeding conditions and provides evidence that ghrelin plays a fundamental role in regulating beta-cell function.


Assuntos
Grelina/genética , Hiperfagia/genética , Leptina/genética , Animais , Apetite/genética , Apetite/fisiologia , Glicemia/metabolismo , Encéfalo/metabolismo , Cromossomos Artificiais Bacterianos , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica , Hiperfagia/metabolismo , Camundongos , Camundongos Transgênicos
10.
Obesity (Silver Spring) ; 16(10): 2239-44, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18719668

RESUMO

OBJECTIVE: Cocaine- and amphetamine-regulated transcript (CART) codes for a hypothalamic neuropeptide, CART (55-102), which inhibits food intake. Intracerebroventricular injection of CART (55-102) reduces appetite, but also results in motor abnormalities. More recently, studies have demonstrated that administration of CART directly into the paraventricular nucleus (PVN) increases food intake. To investigate the role of CART in the regulation of energy balance in the PVN, we used recombinant adeno-associated virus (rAAV) to overexpress CART in the PVN. METHODS AND PROCEDURES: Male Wistar rats were injected with either rAAV-encoding CART (rAAV-CART) or rAAV-encoding enhanced green fluorescent protein (rAAV-EGFP) as a control. Food intake and body weight were measured regularly. Animals were fed on normal-chow diet for the first 93 days of the study. After this point, they were fed on high-fat diet. Animals were killed 138 days postinjection and blood and tissues were collected for analysis. RESULTS: Overexpression of CART in the PVN resulted in increased cumulative food intake and body weight gain compared with rAAV-EGFP controls when fed normal chow. These changes became significant at day 65 and 79, respectively and were accentuated on a high-fat diet. A 4% increase in food intake was observed in rAAV-CART animals on a normal-chow diet and a 6% increase when fed a high-fat diet. At the end of the study, rAAV-CART-treated animals had higher circulating leptin concentrations in accord with their higher body weight. DISCUSSION: These data provide further evidence that hypothalamic CART plays an orexigenic role.


Assuntos
Regulação do Apetite , Comportamento Animal , Ingestão de Alimentos , Proteínas do Tecido Nervoso/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Aumento de Peso , Tecido Adiposo/metabolismo , Animais , Dependovirus/genética , Gorduras na Dieta/administração & dosagem , Vetores Genéticos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Leptina/sangue , Masculino , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/sangue , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Tireotropina/sangue , Fatores de Tempo , Transdução Genética , Proteína Desacopladora 1 , Regulação para Cima
11.
Endocrinology ; 147(7): 3510-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16574794

RESUMO

Neuropeptide S (NPS) is a recently discovered peptide shown to be involved in the modulation of arousal and fear responses. It has also been shown that lateral ventricle administration of NPS causes a significant decrease in food intake. Neuropeptides involved in the modulation of arousal have been shown to be involved in the regulation of the hypothalamo-pituitary adrenal (HPA) axis and food intake. In this study, we have examined the effect of intracerebroventricular (ICV) administration of NPS on behavior, regulation of the HPA axis, and food intake. ICV NPS significantly increased plasma ACTH and corticosterone 10 and 40 min after injection, respectively. A single ICV injection of NPS caused a significant increase in rearing activity as well as ambulatory movement for up to 45 min after injection. We then studied the effect of paraventricular nucleus (PVN) administration of NPS on the regulation of the HPA axis, behavior, and food intake. There was a significant increase in plasma ACTH and corticosterone after a single NPS PVN injection. Incubation of hypothalamic explants with increasing concentrations of NPS caused a significant increase in CRH and arginine vasopressin release. In addition, PVN administration of NPS dose-dependently inhibited food intake in the first hour after injection, although no effect on food intake was seen after this time. PVN administration of NPS caused a significant increase in rearing activity. These data demonstrate a novel role for NPS in the stimulation of the HPA axis.


Assuntos
Comportamento Alimentar , Hipotálamo/metabolismo , Neuropeptídeos/fisiologia , Sistema Hipófise-Suprarrenal/metabolismo , Animais , Comportamento Animal , Corticosterona/sangue , Corticosterona/metabolismo , Ingestão de Alimentos , Humanos , Masculino , Núcleo Hipotalâmico Paraventricular/metabolismo , Hipófise/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
12.
FASEB J ; 19(12): 1680-2, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16099943

RESUMO

Agouti-related protein (AgRP) and neuropeptide Y (NPY) are colocalized in arcuate nucleus (arcuate) neurons implicated in the regulation of energy balance. Both AgRP and NPY stimulate food intake when administered into the third ventricle and are up-regulated in states of negative energy balance. However, mice with targeted deletion of either NPY or AgRP or both do not have major alterations in energy homeostasis. Using bacterial artificial chromosome (BAC) transgenesis we have targeted expression of a neurotoxic CAG expanded form of ataxin-3 to AgRP-expressing neurons in the arcuate. This resulted in a 47% loss of AgRP neurons by 16 weeks of age, a significantly reduced body weight, (wild-type mice (WT) 34.7+/-0.7 g vs. transgenic mice (Tg) 28.6+/-0.6 g, P<0.001), and reduced food intake (WT 5.0+/-0.2 vs. Tg 3.6+/-0.1 g per day, P<0.001). Transgenic mice had significantly reduced total body fat, plasma insulin, and increased brown adipose tissue UCP1 expression. Transgenic mice failed to respond to peripherally administered ghrelin but retained sensitivity to PYY 3-36. These data suggest that postembryonic partial loss of AgRP/NPY neurons leads to a lean, hypophagic phenotype.


Assuntos
Neurônios/metabolismo , Neuropeptídeo Y/genética , Proteínas/genética , Tecido Adiposo/metabolismo , Proteína Relacionada com Agouti , Fenômenos Fisiológicos da Nutrição Animal , Animais , Glicemia/metabolismo , Northern Blotting , Composição Corporal , Peso Corporal , Proteínas de Transporte/metabolismo , Cromossomos Artificiais Bacterianos/metabolismo , DNA Complementar/metabolismo , Éxons , Comportamento Alimentar , Deleção de Genes , Grelina , Hipotálamo/metabolismo , Hipotálamo/patologia , Hibridização In Situ , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intercelular , Canais Iônicos , Leptina/sangue , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais , Modelos Genéticos , Neuropeptídeo Y/metabolismo , Hormônios Peptídicos/metabolismo , Fenótipo , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Radioimunoensaio , Magreza/genética , Fatores de Tempo , Transgenes , Proteína Desacopladora 1 , Regulação para Cima
13.
Endocrinology ; 146(8): 3526-34, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15890775

RESUMO

Nociceptin or orphanin FQ (N/OFQ) and its receptor NOP1 are expressed in hypothalamic nuclei involved in energy homeostasis. N/OFQ administered by intracerebroventricular or arcuate nucleus (ARC) injection increases food intake in satiated rats. The mechanisms by which N/OFQ increases food intake are unknown. We hypothesized that N/OFQ may regulate hypothalamic neurons containing peptides involved in the control of food intake such as cocaine- and amphetamine-regulated transcript (CART), alphaMSH, neuropeptide Y (NPY), and agouti-related protein (AgRP). We investigated the ability of N/OFQ to alter the release of CART, alphaMSH, NPY, and AgRP using ex vivo medial basal hypothalamic explants. Incubation of hypothalamic explants with N/OFQ (1, 10, 100 nM) resulted in significant changes in CART and AgRP release. One hundred nanomoles N/OFQ caused a 33% decrease in release of CART (55-102) immunoreactivity (IR) and increased release of AgRP-IR to 163% but produced no change in either alphaMSH-IR or NPY-IR. Double immunocytochemistry/in situ hybridization demonstrated that CART-IR and NOP1 mRNA are colocalized throughout the hypothalamus, in particular in the paraventricular nucleus, lateral hypothalamus, zona incerta, and ARC, providing an anatomical basis for N/OFQ action on CART release. Dual in situ hybridization demonstrated that AgRP neurons in the ARC also express the NOP1 receptor. Our data suggest that nociceptin via the NOP1 receptor may increase food intake by decreasing the release of the anorectic peptide CART and increasing the release of the orexigenic peptide AgRP.


Assuntos
Regulação da Expressão Gênica , Hipotálamo/fisiologia , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Peptídeos Opioides/genética , Peptídeos Opioides/farmacologia , Hormônios Peptídicos/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada com Agouti , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Energia , Injeções Intraventriculares , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Neuropeptídeo Y/metabolismo , Técnicas de Cultura de Órgãos , Hormônios Peptídicos/metabolismo , Ratos , Ratos Wistar , Receptores Opioides , Ribonucleases , Receptor de Nociceptina , Nociceptina
14.
Regul Pept ; 122(3): 169-72, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15491787

RESUMO

The melanocortin system is an important regulator of body weight and the hypothalamo-pituitary-thyroid (HPT) axis. The pro-opiomelanocortin (POMC)-null mouse, deficient in all POMC-derived peptides, including alpha-melanocyte stimulating hormone (alpha-MSH), has an obese phenotype. We studied the HPT axis of POMC-null mice, which has not been previously investigated. Because alpha-MSH has a stimulatory effect on the HPT axis, we hypothesised that these mice would have a down-regulated thyroid axis, consistent with a recent study of POMC-null humans. The activity of the HPT axis was studied by collecting blood, pituitaries and hypothalami from ad libitum fed, adult POMC-null, heterozygous and wild-type mice. POMC-null mice had significantly elevated plasma total T(4) (TT(4)) and free T(3) (fT(3)) with reduced plasma thyroid stimulating hormone (TSH), pituitary TSH content and hypothalamic thyrotrophin stimulating hormone (TRH) content compared to wild-type mice. No significant differences between heterozygous and wild-type mice were observed. POMC-null mice have an abnormal HPT axis, which may contribute to their hyperphagia and obesity. These abnormalities are in contrast to those observed in POMC-null humans. These findings support a role for the melanocortin system in the regulation of the HPT axis.


Assuntos
Hipotálamo/metabolismo , Obesidade/sangue , Hipófise/metabolismo , Pró-Opiomelanocortina/deficiência , Glândula Tireoide/metabolismo , Hormônio Liberador de Tireotropina/sangue , Tireotropina/sangue , Animais , Hipotálamo/anormalidades , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/patologia , Hipófise/anormalidades , Glândula Tireoide/anormalidades
15.
Endocrinology ; 145(11): 5252-8, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15297436

RESUMO

Increased food intake is characteristic of hyperthyroidism, although this is presumed to compensate for a state of negative energy balance. However, here we show that the thyroid hormone T(3) directly stimulates feeding at the level of the hypothalamus. Peripheral administration of T(3) doubled food intake in ad libitum-fed rats over 2 h and induced expression of the immediate early gene, early growth response-1, in the hypothalamic ventromedial nucleus (VMN), whereas maintaining plasma-free T(3) levels within the normal range. T(3)-induced feeding occurred without altering energy expenditure or locomotion. Injection of T(3) directly into the VMN produced a 4-fold increase in food intake in the first hour. The majority of T(3) in the brain is reported to be produced by tissue-specific conversion of T(4) to T(3) by the enzyme type 2 iodothyronine deiodinase (D2). Hypothalamic D2 mRNA expression showed a diurnal variation, with a peak in the nocturnal feeding phase. Hypothalamic D2 mRNA levels also increased after a 12- and 24-h fast, suggesting that local production of T(3) may play a role in this T(3) feeding circuit. Thus, we propose a novel hypothalamic feeding circuit in which T(3), from the peripheral circulation or produced by local conversion, stimulates food intake via the VMN.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Hipertireoidismo/fisiopatologia , Tri-Iodotironina/farmacologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Jejum/fisiologia , Regulação Enzimológica da Expressão Gênica , Iodeto Peroxidase/genética , Leptina/sangue , Masculino , RNA Mensageiro/análise , Ratos , Ratos Wistar , Tiroxina/sangue , Iodotironina Desiodinase Tipo II
16.
Clin Biochem ; 37(3): 217-23, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14972644

RESUMO

OBJECTIVE: In this experiment, we studied the chronic effects of NPY, as there were no data on long-term effects of NPY in vivo. METHODS: Complementary DNA encoding NPY was isolated, sequenced and cloned into the expression vector, pCEP4. The 6-23 clone 6 cell line was transfected with this clone. Two groups of 10 adult male WAG rats (180-250 g body weight) were injected with either untransfected 6-23 clone 6 or 6-23 clone 6 transfected with NPY cDNA [6-23 (NPY)]. After 8 weeks, the animals were killed, their plasma assayed for insulin. Pancreatic glucagon (PG), by RIA, and plasma glucose were measured. RESULTS: The transfected cells were shown to be producing fully processed, bioactive NPY. The expression of NPY was also confirmed by Northern blot analysis. The animals injected with 6-23 (NPY) cells gained significantly more weight than the controls, (on day 54, 31.89 +/- 3.56 vs. 24.1 +/- 4.12 g, n = 10, P < 0.05). Plasma insulin and PG increased significantly in NPY animals compared to controls. The total RNA extracted from tumours was analysed by Northern blotting and showed NPY mRNA expression in NPY animals, but not in controls. CONCLUSION: The long-term effects of NPY was confirmed by injection of the cells producing this peptide.


Assuntos
Transplante de Células/métodos , Neuropeptídeo Y/administração & dosagem , Transfecção/métodos , Animais , Sequência de Bases/genética , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Linhagem Celular Tumoral , Clonagem Molecular/métodos , Humanos , Injeções Subcutâneas , Masculino , Dados de Sequência Molecular , Neuropeptídeo Y/genética , RNA Mensageiro/genética , Ratos , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/fisiologia
17.
Endocrinology ; 144(4): 1420-5, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12639925

RESUMO

Calcitonin gene-related protein (CGRP) inhibits food intake and stimulates the hypothalamo-pituitary-adrenal (HPA) axis after intracerebroventricular injection in rats. However, the hypothalamic site and mechanism of action are unknown. We investigated the effects of intraparaventricular nucleus administration (iPVN) of CGRP on food intake and the HPA axis in rats and the effect of CGRP on the release of hypothalamic neuropeptides in vitro. In addition, we investigated the effects of food deprivation on hypothalamic CGRP expression. CGRP dose-dependently reduced food intake in the first hour after iPVN injection in fasted male rats (saline, 5.1 +/- 0.8 g; 0.3 nmol CGRP, 1.1 +/- 0.5 g; P < 0.001 vs. saline). iPVN injection of CGRP(8-37) (a CGRP(1) receptor antagonist) alone had no effect on food intake. However, the reduction in food intake by iPVN CGRP was attenuated by prior administration of CGRP(8-37) [CGRP(8-37) (10 nmol)/CGRP (0.3 nmol), 3.0 +/- 0.8 g; P < 0.05 vs. 0.3 nmol CGRP]. CGRP (100 nM) stimulated the release of alpha-melanocyte stimulating hormone, cocaine- and amphetamine-related transcript, corticotropin-releasing hormone, and arginine vasopressin from hypothalamic explants to 127 +/- 19%, 148 +/- 10%, 158 +/- 17%, and 198 +/- 21% of basal levels, respectively (P < 0.05 vs. basal), but did not alter the release of either neuropeptide Y or agouti-related protein. Hypothalamic CGRP mRNA levels in 24-h fasted rats were increased to 130 +/- 8% of control levels [CGRP mRNA (arbitrary units), 4.75 +/- 0.4; controls, 3.65 +/- 0.34; P < 0.05]. Our data suggest that CGRP administered to the PVN inhibits food intake and stimulates the HPA axis.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Fragmentos de Peptídeos/farmacologia , Sistema Hipófise-Suprarrenal/fisiologia , Hormônio Adrenocorticotrópico/sangue , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Cateterismo , Corticosterona/sangue , Jejum/fisiologia , Expressão Gênica/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Técnicas In Vitro , Masculino , Microinjeções , Neuropeptídeos/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , RNA Mensageiro/análise , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...