Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Horm Behav ; 66(1): 196-207, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24560890

RESUMO

This article is part of a Special Issue "Energy Balance". The classical estrogen receptors, estrogen receptor-α and estrogen receptor-ß are well established in the regulation of body weight and energy homeostasis in both male and female mice, whereas, the role for G protein-coupled estrogen receptor 1 (GPER) as a modulator of energy homeostasis remains controversial. This study sought to determine whether gene deletion of GPER (GPER KO) alters body weight, body adiposity, food intake, and energy homeostasis in both males and females. Male mice lacking GPER developed moderate obesity and larger adipocyte size beginning at 8 weeks of age, with significant reductions in energy expenditure, but not food intake or adipocyte number. Female GPER KO mice developed increased body weight relative to WT females a full 6 weeks later than the male GPER KO mice. Female GPER KO mice also had reductions in energy expenditure, but no significant increases in body fat content. Consistent with their decrease in energy expenditure, GPER KO males and females showed significant reductions in two brown fat thermogenic proteins. GPER KO females, prior to their divergence in body weight, were less sensitive than WT females to the feeding-inhibitory effects of leptin and CCK. Additionally, body weight was not as modulated by ovariectomy or estradiol replacement in GPER KO mice. Estradiol treatment activated phosphorylated extracellular signal-regulated kinase (pERK) in WT but not GPER KO females. For the first time, GPER expression was found in the adipocyte but not the stromal fraction of adipose tissue. Together, these results provide new information elucidating a sexual dimorphism in GPER function in the development of postpubertal energy balance.


Assuntos
Adiposidade/fisiologia , Metabolismo Energético/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Caracteres Sexuais , Animais , Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Estradiol/farmacologia , Feminino , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Knockout , Ovariectomia
2.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1351-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19710389

RESUMO

Maternal obesity accentuates offspring obesity in dams bred to develop diet-induced obesity (DIO) on a 31% fat, high-sucrose, high-energy (HE) diet but has no effect on offspring of diet-resistant (DR) dams. Also, only DIO dams become obese when they and DR dams are fed HE diet throughout gestation and lactation. We assessed glucose and oleic acid (OA) sensitivity of dissociated ventromedial hypothalamic nucleus (VMN) neurons from 3- to 4-wk old offspring of DIO and DR dams fed chow or HE diet using fura-2 calcium imaging to monitor intracellular calcium fluctuations as an index of neuronal activity. Offspring of DIO dams fed chow had approximately 2-fold more glucose-inhibited (GI) neurons than did DR offspring. This difference was eliminated in offspring of DIO dams fed HE diet. At 2.5 mM glucose, offspring of chow-fed DIO dams had more GI neurons that were either excited or inhibited by OA than did DR offspring. Maternal HE diet intake generally increased the percentage of neurons that were excited and decreased the percentage that were inhibited by OA in both DIO and DR offspring. However, this effect was more pronounced in DIO offspring. These data, as well as concentration-dependent differences in OA sensitivity, suggest that genotype, maternal obesity, and dietary content can all affect the sensitivity of offspring VMN neurons to glucose and long-chain fatty acids. Such altered sensitivities may underlie the propensity of DIO offspring to become obese when fed high-fat, high-sucrose diets.


Assuntos
Glicemia/metabolismo , Gorduras na Dieta/farmacologia , Sacarose Alimentar/farmacologia , Ácidos Graxos/metabolismo , Neurônios/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Peso Corporal/fisiologia , Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Genótipo , Insulina/sangue , Leptina/sangue , Masculino , Obesidade/metabolismo , Ácido Oleico/farmacologia , Gravidez , Prenhez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Ratos , Ratos Endogâmicos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
3.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R655-64, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19535676

RESUMO

We assessed the mechanisms by which specialized hypothalamic ventromedial nucleus (VMN) neurons utilize both glucose and long-chain fatty acids as signaling molecules to alter their activity as a potential means of regulating energy homeostasis. Fura-2 calcium (Ca(2+)) and membrane potential dye imaging, together with pharmacological agents, were used to assess the mechanisms by which oleic acid (OA) alters the activity of dissociated VMN neurons from 3- to 4-wk-old rats. OA excited up to 43% and inhibited up to 29% of all VMN neurons independently of glucose concentrations. In those neurons excited by both 2.5 mM glucose and OA, OA had a concentration-dependent effective excitatory concentration (EC(50)) of 13.1 nM. Neurons inhibited by both 2.5 mM glucose and OA had an effective inhibitory concentration (IC(50)) of 93 nM. At 0.5 mM glucose, OA had markedly different effects on these same neurons. Inhibition of carnitine palmitoyltransferase, reactive oxygen species formation, long-chain acetyl-CoA synthetase and ATP-sensitive K(+) channel activity or activation of uncoupling protein 2 (UCP2) accounted for only approximately 20% of OA's excitatory effects and approximately 40% of its inhibitory effects. Inhibition of CD36, a fatty acid transporter that can alter cell function independently of intracellular fatty acid metabolism, reduced the effects of OA by up to 45%. Thus OA affects VMN neuronal activity through multiple pathways. In glucosensing neurons, its effects are glucose dependent. This glucose-OA interaction provides a potential mechanism whereby such "metabolic sensing" neurons can respond to differences in the metabolic states associated with fasting and feeding.


Assuntos
Sinalização do Cálcio , Metabolismo Energético , Glucose/metabolismo , Neurônios/metabolismo , Ácido Oleico/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Carnitina O-Palmitoiltransferase/metabolismo , Coenzima A Ligases/antagonistas & inibidores , Coenzima A Ligases/metabolismo , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Homeostase , Canais Iônicos/metabolismo , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Potenciais da Membrana , Microscopia de Fluorescência , Proteínas Mitocondriais/metabolismo , Inibição Neural , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Proteína Desacopladora 2 , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/enzimologia
4.
Endocrinology ; 149(10): 5146-54, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18556346

RESUMO

Neurons in the ventromedial and arcuate hypothalamic nuclei (VMN and ARC, respectively) mediate many of leptin's effects on energy homeostasis. Some are also glucosensing, whereby they use glucose as a signaling molecule to regulate their firing rate. We used fura-2 calcium (Ca2+) imaging to determine the interactions between these two important mediators of peripheral metabolism on individual VMN neurons and the mechanisms by which leptin regulates neuronal activity in vitro. Leptin excited 24%, inhibited 20%, and had a biphasic response in 10% of VMN neurons. Excitation occurred with a EC50 of 5.2 fmol/liter and inhibition with a IC50 of 4.2 fmol/liter. These effects were independent of the ambient glucose levels, and both glucosensing and non-glucosensing neurons were affected by leptin. In contrast, the ARC showed a very different distribution of leptin-responsive neurons, with 40% leptin excited, 10% leptin inhibited, and 2% having a biphasic response (chi2=60.2; P<0.0001). Using pharmacological manipulations we found that leptin inhibits VMN neurons via activation of phosphoinositol-3 kinase and activation of the ATP-sensitive K+ channel. In addition, leptin inhibition was antagonized by 5'-AMP-activated protein kinase activation in 39% of neurons but was unaffected by 5'-AMP-activated protein kinase inhibition. No mechanism was delineated for leptin-induced excitation. Thus, within the physiological range of brain glucose levels, leptin has a differential effect on VMN vs. ARC neurons, and acts on both glucosensing and non-glucosensing VMN neurons in a glucose-independent fashion with inhibition primarily dependent upon activation of the ATP-sensitive K+ channel.


Assuntos
Leptina/metabolismo , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Ventromedial/citologia , Proteínas Quinases Ativadas por AMP , Animais , Núcleo Arqueado do Hipotálamo/citologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Glucose/metabolismo , Canais KATP/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Endocrinology ; 148(1): 310-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17023527

RESUMO

Rats with a genetic predisposition to develop diet-induced obesity (DIO) have a preexisting reduction in central leptin and insulin sensitivity. High-fat diets also reduce sensitivity to leptin, insulin, and melanocortin agonists. We postulated that such reduced sensitivities would be associated with decreased binding to the hypothalamic leptin, insulin, and melanocortin receptors in selectively bred DIO rats and in rats fed a high-energy (HE; 31% fat) diet for 7 wk. On HE diet, DIO rats gained 15% more weight and had 121% heavier fat pads and 70% higher leptin levels than low fat chow-fed DIO rats. Diet-resistant (DR) rats gained no more weight on HE diet but had 48% heavier fat pads and 70% higher leptin levels than chow-fed DR rats. Compared with DR rats, DIO (125)I-leptin binding was 41, 36, and 40% lower in the hypothalamic dorsomedial, arcuate, and dorsomedial portion of the ventromedial nuclei, respectively, and arcuate (125)I-insulin binding was 31% lower independent of diet. In contrast, hypothalamic melanocortin binding did not differ between DIO and DR rats. However, HE diet intake lowered lateral hypothalamic melanocortin-3 and melanocortin-4 receptor and hippocampal insulin binding of both DIO and DR rats and hypothalamic paraventricular nucleus melanocortin-4 receptor binding only in DR rats. Neither genotype nor diet affected substantia nigra or ventral tegmental area binding. These results corroborate our previous findings demonstrating a preexisting decrease in DIO hypothalamic leptin and insulin signaling and demonstrate that HE diet intake reduces hypothalamic melanocortin and hippocampal insulin binding.


Assuntos
Hipotálamo/metabolismo , Insulina/farmacocinética , Leptina/farmacocinética , Melanocortinas/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Glicemia , Gorduras na Dieta/farmacologia , Núcleo Hipotalâmico Dorsomedial/metabolismo , Insulina/sangue , Radioisótopos do Iodo , Leptina/sangue , Masculino , Obesidade/genética , Ratos , Ratos Mutantes , Receptor Tipo 4 de Melanocortina/metabolismo , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Aumento de Peso/fisiologia
6.
Peptides ; 26(10): 1788-99, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16046247

RESUMO

The process of energy homeostasis is a highly regulated process involving interacting signals between a variety of anorexigenic and orexigenic peptides, proteins and signaling molecules. The melanocortin system is an important component of this complex regulatory network. Involvement of the melanocortin pathway in the control of food intake and body weight regulation has been studied extensively in the past two decades. Previous studies that involve central administration of melanocortin molecules and examination of molecules that effect food intake in melanocortin knockout (KO) mice (MC3R, MC4R, POMC, AGRP and NPY) have been examined. In this review, we have summarized feeding studies that have resulted in the recognition of the melanocortin system as a major contributor to the complex neuroendocrine system regulating energy homeostasis.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Melanocortinas/metabolismo , Receptores de Melanocortina/metabolismo , Animais , Humanos , Ligantes , Melanocortinas/agonistas , Melanocortinas/antagonistas & inibidores , Melanocortinas/fisiologia , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inibidores , Receptores de Melanocortina/fisiologia
7.
Peptides ; 26(10): 1720-7, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16005545

RESUMO

Ghrelin stimulates food intake in part by activating hypothalamic neuropeptide Y (NPY) neurons/agouti related peptide (AGRP) neurons. We investigated the role of AGRP/melanocortin signaling in ghrelin-induced food intake by studying melanocortin 3 and 4 receptor knockout (MC3R KO and MC4R KO) mice. We also determined whether reduced ghrelin levels and/or an altered sensitivity to the GH-stimulating effects of ghrelin accompany the obesity syndromes of MC3R KO and MC4R KO mice. Compared to wild-type (WT) mice, the effects of ghrelin on food intake were reduced in MC3R KO and MC4R KO mice and circulating ghrelin levels were reduced in female MC4R KO mice. Female MC3R KO and MC4R KO mice exhibited a diminished responsiveness to the GH-releasing effects of ghrelin. Thus, deletion of the MC3R or MC4R results in a decreased sensitivity to ghrelin and verifies the involvement in the melanocortin system in ghrelin-induced food intake.


Assuntos
Ingestão de Alimentos/fisiologia , Grelina/fisiologia , Hormônio do Crescimento/metabolismo , Receptor Tipo 3 de Melanocortina/deficiência , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/deficiência , Receptor Tipo 4 de Melanocortina/genética , Animais , Ingestão de Alimentos/genética , Feminino , Grelina/sangue , Humanos , Masculino , Camundongos , Camundongos Knockout , Ratos
8.
Curr Pharm Des ; 10(28): 3443-79, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15579046

RESUMO

The melanocortin pathway consists of endogenous agonists, antagonists, G-protein coupled receptors (GPCRs), and auxiliary proteins. This pathway has been identified to participate physiologically in numerous biological pathways including energy homeostasis, pigmentation, sexual function, inflammation, cardiovascular function, adrenal function, sebaceous gland lipid production, just to list a few. During this past decade, a clear link between the melanocortin-4 receptor (MC4R) and obesity, in both mice and humans via the regulation of food intake and energy homeostasis, has made this pathway the target of many academic and industrial research endeavors in attempts to develop potent and selective MC4R small molecules as anti-obesity therapeutic agents. Herein, we attempt to summarize the known proteins that constitute the melanocortin system and discuss advances in peptide and non-peptide drug discovery.


Assuntos
Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inibidores , Animais , Humanos , Ligantes , Peptídeos/química , Peptídeos/farmacologia , Receptores de Melanocortina/genética , Relação Estrutura-Atividade
10.
Cell Metab ; 14(4): 453-65, 2011 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21982706

RESUMO

Estrogens regulate body weight and reproduction primarily through actions on estrogen receptor-α (ERα). However, ERα-expressing cells mediating these effects are not identified. We demonstrate that brain-specific deletion of ERα in female mice causes abdominal obesity stemming from both hyperphagia and hypometabolism. Hypometabolism and abdominal obesity, but not hyperphagia, are recapitulated in female mice lacking ERα in hypothalamic steroidogenic factor-1 (SF1) neurons. In contrast, deletion of ERα in hypothalamic pro-opiomelanocortin (POMC) neurons leads to hyperphagia, without directly influencing energy expenditure or fat distribution. Further, simultaneous deletion of ERα from both SF1 and POMC neurons causes hypometabolism, hyperphagia, and increased visceral adiposity. Additionally, female mice lacking ERα in SF1 neurons develop anovulation and infertility, while POMC-specific deletion of ERα inhibits negative feedback regulation of estrogens and impairs fertility in females. These results indicate that estrogens act on distinct hypothalamic ERα neurons to regulate different aspects of energy homeostasis and reproduction.


Assuntos
Metabolismo Energético/fisiologia , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Animais , Estradiol/sangue , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Feminino , Hiperfagia/etiologia , Infertilidade Feminina/etiologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/etiologia , Pró-Opiomelanocortina/metabolismo , Fator Esteroidogênico 1/metabolismo
11.
Eur J Pharmacol ; 660(1): 80-7, 2011 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-21199647

RESUMO

The melanocortin system is well recognized to be involved in the regulation of food intake, body weight, and energy homeostasis. To probe the role of the MC(3) in the regulation of food intake, JRH322-18 a mixed MC(3) partial agonist/antagonist and MC(4) agonist tetrapeptide was examined in wild type (WT) and melanocortin 4 receptor (MC(4)) knockout mice and shown to reduce food intake in both models. In the wild type mice, 2.0 nmol of JRH322-18 statistically reduced food intake 4h post icv treatment into satiated nocturnally feeding wild type mice. The same dose in the MC(4)KO mice significantly reduced cumulative food intake 24h post treatment. Conditioned taste aversion as well as activity studies supports that the decreased food intake was not due to visceral illness. Since these studies resulted in loss-of-function results, the SHU9119 and agouti-related protein (AGRP) melanocortin receptor antagonists were administered to wild type as well as the MC(3) and MC(4) knockout mice in anticipation of gain-of-function results. The SHU9119 ligand produced an increase in food intake in the wild type mice as anticipated, however no effect was observed in the MC(3) and MC(4) knockout mice as compared to the saline control. The AGRP ligand however, produced a significant increase in food intake in the wild type as well as the MC(3) and MC(4) knockout mice and it had a prolonged affect for several days. These data support the hypothesis that the MC(3) plays a subtle role in the regulation of food intake, however the mechanism by which this is occurring remains to be determined.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Receptor Tipo 3 de Melanocortina/metabolismo , Sequência de Aminoácidos , Animais , Ingestão de Alimentos/genética , Técnicas de Inativação de Genes , Humanos , Ligantes , Camundongos , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Receptor Tipo 3 de Melanocortina/agonistas , Receptor Tipo 3 de Melanocortina/antagonistas & inibidores , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Saciação , alfa-MSH/análogos & derivados , alfa-MSH/química , alfa-MSH/farmacologia
12.
Endocrinology ; 151(9): 4270-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20668022

RESUMO

Because rearing rats in large litters (LLs) protects them from becoming obese, we postulated that LL rearing would protect rats selectively bred to develop diet-induced obesity (DIO) from becoming obese by overcoming their inborn central leptin resistance. Male and female DIO rats were raised in normal litters (NLs; 10 pups/dam) or LLs (16 pups/dam) and assessed for anatomical, biochemical, and functional aspects of leptin sensitivity at various ages when fed low-fat chow or a 31% fat high-energy (HE) diet. LL rearing reduced plasma leptin levels by postnatal day 2 (P2) and body weight gain by P8. At P16, LL DIO neonates had increased arcuate nucleus (ARC) binding of leptin to its extracellular receptors and at P28 an associated increase of their agouti-related peptide and alpha-MSH axonal projections to the paraventricular nucleus. Reduced body weight persisted and was associated with increased ARC leptin receptor binding and sensitivity to the anorectic effects of leptin, reduced adiposity, and enhanced insulin sensitivity in LL DIO rats fed chow until 10 wk of age. The enhanced ARC leptin receptor binding and reduced adiposity of LL DIO rats persisted after an additional 5 wk on the HE diet. Female LL DIO rats had similar reductions in weight gain on both chow and HE diet vs. normal litter DIO rats. We postulate that LL rearing enhances DIO leptin sensitivity by lowering plasma leptin levels and thereby increasing leptin receptor availability and that this both enhances the ARC-paraventricular nucleus pathway development and protects them from becoming obese.


Assuntos
Dieta , Leptina/sangue , Tamanho da Ninhada de Vivíparos/fisiologia , Obesidade/fisiopatologia , Adiposidade/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Cruzamento , Ingestão de Alimentos/fisiologia , Feminino , Idade Gestacional , Imuno-Histoquímica , Lactação/fisiologia , Leptina/metabolismo , Masculino , Obesidade/sangue , Obesidade/etiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ligação Proteica , Ratos , Receptores para Leptina/metabolismo , Fatores de Tempo , Desmame , Aumento de Peso/fisiologia , alfa-MSH/metabolismo
13.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R521-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19118094

RESUMO

Maternal obesity accentuates offspring obesity in dams bred to develop diet-induced obesity (DIO) on a 31% fat, high energy (HE) diet but has no effect on offspring of diet-resistant (DR) dams. Only DIO dams became obese on HE diet when they and DR dams were fed 5% fat chow or HE diets throughout gestation and lactation. Leptin sensitivity of dissociated arcuate (ARC) and ventromedial (VMN) hypothalamic nucleus neurons from the 3- to 4-wk-old offspring was assessed using fura-2 calcium imaging to monitor leptin-induced changes in intracellular calcium ([Ca(2+)](i)) as an index of neuronal activity. At 0.1, 1, 10 fmol/l leptin, approximately 4 times more VMN and ARC neurons were excited than inhibited by leptin. In the VMN, leptin excited up to 41% fewer neurons, and these excited neurons were less sensitive to increasing doses of leptin in DIO compared with DR offspring. Also, maternal HE diet intake decreased the percentage of leptin-excited VMN neurons in both DIO and DR offspring and decreased the percentage of leptin-inhibited VMN neurons by 36% only in DIO offspring. In the ARC, there were no genotype or maternal diet effects on the percentage of ARC neurons excited by leptin. However, those DR neurons that were leptin excited were more sensitive to leptin than were those from DIO offspring. These data suggest that reduced responsiveness of DIO VMN neurons to leptin's excitatory effects may be an important contributing factor to the reduced anorectic and thermogenic leptin responsiveness of DIO rats in vivo.


Assuntos
Dieta , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Obesidade/etiologia , Obesidade/genética , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Glicemia/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Energia/efeitos dos fármacos , Genótipo , Insulina/sangue , Leptina/sangue , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Núcleo Hipotalâmico Ventromedial/citologia , Aumento de Peso/efeitos dos fármacos
14.
J Clin Invest ; 119(9): 2577-89, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19726875

RESUMO

Insulin signaling can be modulated by several isoforms of PKC in peripheral tissues. Here, we assessed whether one specific isoform, PKC-theta, was expressed in critical CNS regions that regulate energy balance and whether it mediated the deleterious effects of diets high in fat, specifically palmitic acid, on hypothalamic insulin activity in rats and mice. Using a combination of in situ hybridization and immunohistochemistry, we found that PKC-theta was expressed in discrete neuronal populations of the arcuate nucleus, specifically the neuropeptide Y/agouti-related protein neurons and the dorsal medial nucleus in the hypothalamus. CNS exposure to palmitic acid via direct infusion or by oral gavage increased the localization of PKC-theta to cell membranes in the hypothalamus, which was associated with impaired hypothalamic insulin and leptin signaling. This finding was specific for palmitic acid, as the monounsaturated fatty acid, oleic acid, neither increased membrane localization of PKC-theta nor induced insulin resistance. Finally, arcuate-specific knockdown of PKC-theta attenuated diet-induced obesity and improved insulin signaling. These results suggest that many of the deleterious effects of high-fat diets, specifically those enriched with palmitic acid, are CNS mediated via PKC-theta activation, resulting in reduced insulin activity.


Assuntos
Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Resistência à Insulina/fisiologia , Isoenzimas/metabolismo , Ácido Palmítico/toxicidade , Proteína Quinase C/metabolismo , Animais , Sequência de Bases , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/toxicidade , Gluconeogênese/efeitos dos fármacos , Isoenzimas/deficiência , Isoenzimas/genética , Leptina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido Palmítico/administração & dosagem , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C-theta , RNA Interferente Pequeno/genética , Ratos , Ratos Long-Evans , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA