Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Cell Endocrinol ; 455: 33-40, 2017 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-27884781

RESUMO

Aging constitutes the central risk factor for major diseases including many forms of cancer, neurodegeneration, and cardiovascular diseases. The aging process is characterized by both global and tissue-specific changes in gene expression across taxonomically diverse species. While aging has historically been thought to entail cell-autonomous, even stochastic changes, recent evidence suggests that modulation of this process can be hierarchal, wherein manipulations of nutrient-sensing neurons (e.g., in the hypothalamus) produce peripheral effects that may modulate the aging process itself. The most robust intervention extending lifespan, plausibly impinging on the aging process, involves different modalities of dietary restriction (DR). Lifespan extension by DR is associated with broad protection against diseases (natural and engineered). Here we review potential epigenetic processes that may link lifespan to age-related diseases, particularly in the context of DR and (other) ketogenic diets, focusing on brain and hypothalamic mechanisms.


Assuntos
Restrição Calórica , Dieta Cetogênica , Epigênese Genética , Inibidores de Histona Desacetilases/farmacologia , Hidroxibenzoatos/farmacologia , Longevidade/efeitos dos fármacos , Animais , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Metilação de DNA , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Longevidade/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo
2.
PLoS One ; 11(11): e0166381, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27832201

RESUMO

We have reported a correlation between hypothalamic expression of Creb-binding protein (Cbp) and lifespan, and that inhibition of Cbp prevents protective effects of dietary restriction during aging, suggesting that hypothalamic Cbp plays a role in responses to nutritional status and energy balance. Recent GWAS and network analyses have also implicated Cbp as the most connected gene in protein-protein interactions in human Type 2 diabetes. The present studies address mechanisms mediating the role of Cbp in diabetes by inhibiting hypothalamic Cbp using a Cre-lox strategy. Inhibition of hypothalamic Cbp results in profound obesity and impaired glucose homeostasis, increased food intake, and decreased body temperature. In addition, these changes are accompanied by molecular evidence in the hypothalamus for impaired leptin and insulin signaling, a shift from glucose to lipid metabolism, and decreased Pomc mRNA, with no effect on locomotion. Further assessment of the significance of the metabolic switch demonstrated that enhanced expression of hypothalamic Cpt1a, which promotes lipid metabolism, similarly resulted in increased body weight and reduced Pomc mRNA.


Assuntos
Proteína de Ligação a CREB/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Animais , Temperatura Corporal , Fator Neurotrófico Derivado do Encéfalo/genética , Proteína de Ligação a CREB/genética , Carnitina O-Palmitoiltransferase/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Regulação para Baixo , Ingestão de Alimentos , Metabolismo Energético , Feminino , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Hipotálamo/patologia , Insulina/metabolismo , Leptina/metabolismo , Metabolismo dos Lipídeos , Masculino , Camundongos , Obesidade/genética , Obesidade/patologia , Pró-Opiomelanocortina/genética , Transdução de Sinais , Aumento de Peso
4.
Neurobiol Dis ; 85: 25-34, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26485309

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disease characterized by metabolic, cognitive, and motor deficits. HD is caused by an expanded CAG repeat in the first exon of the HTT gene, resulting in an expanded polyglutamine section. Dietary restriction (DR) increases lifespan and ameliorates age-related pathologies, including in a model of HD, but the mechanisms mediating these protective effects are unknown. We report metabolic and behavioral effects of DR in the full-length YAC128 HD mouse model, and associated transcriptional changes in hypothalamus and striatum. DR corrected many effects of the transgene including increased body weight, decreased blood glucose, and impaired motor function. These changes were associated with reduced striatal human (but not mouse) HTT expression, as well as alteration in gene expression regulating histone acetylation modifications, particularly Hdac2. Other mRNAs related to Huntington's pathology in striatal tissue showed significant modulation by the transgene, dietary restriction or both. These results establish a protective role of DR in a transgenic model that contains the complete human HTT gene and for the first time suggest a role for DR in lowering HTT level, which correlates with severity of symptoms.


Assuntos
Jejum/metabolismo , Histonas/metabolismo , Doença de Huntington/dietoterapia , Doença de Huntington/metabolismo , Acetilação , Animais , Glicemia/fisiologia , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Histona Desacetilase 2/metabolismo , Proteína Huntingtina , Doença de Huntington/genética , Hipotálamo/metabolismo , Camundongos Transgênicos , Atividade Motora/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Transcrição Gênica/fisiologia
5.
Trends Endocrinol Metab ; 24(10): 488-94, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23791973

RESUMO

We propose that energy balance, glucose homeostasis, and aging are all regulated largely by the same nutrient-sensing neurons in the ventromedial hypothalamus (VMH). Although the central role of these neurons in regulating energy balance is clear, their role in regulating glucose homeostasis has only recently become more clear. This latter function may be most relevant to aging and lifespan by controlling the rate of glucose metabolism. Specifically, glucose-sensing neurons in VMH promote peripheral glucose metabolism, and dietary restriction, by reducing glucose metabolism in these neurons, reduces glucose metabolism of the rest of the body, thereby increasing lifespan. Here we discuss recent studies demonstrating the key role of hypothalamic neurons in driving aging and age-related diseases.


Assuntos
Envelhecimento/metabolismo , Diabetes Mellitus/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Animais , Humanos
6.
Endocrinol Metab Clin North Am ; 42(1): 67-80, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23391240

RESUMO

All organisms must adapt to changing nutrient availability, with nutrient surplus promoting glucose metabolism and nutrient deficit promoting alternative fuels (in mammals, mainly free fatty acids). A major function of glucose-sensing neurons in the hypothalamus is to regulate blood glucose. When these neurons sense glucose levels are too low, they activate robust counterregulatory responses to enhance glucose production, primarily from liver, and reduce peripheral metabolism. Some hypothalamic neurons can metabolize free fatty acids via ß-oxidation, and ß-oxidation generally opposes effects of glucose on hypothalamic neurons. Thus hypothalamic ß-oxidation promotes obese phenotypes, including enhanced hepatic glucose output.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Animais , Glucose/metabolismo , Humanos , Hipotálamo/citologia , Fígado/metabolismo
7.
Front Neuroendocrinol ; 34(2): 95-106, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23262258

RESUMO

Dietary restriction (DR) can extend lifespan and reduce disease burden across a wide range of animals and yeast but the mechanisms mediating these remarkably protective effects remain to be elucidated despite extensive efforts. Although it has generally been assumed that protective effects of DR are cell-autonomous, there is considerable evidence that many whole-body responses to nutritional state, including DR, are regulated by nutrient-sensing neurons. In this review, we explore the hypothesis that nutrient sensing neurons in the ventromedial hypothalamus hierarchically regulate the protective responses of dietary restriction. We describe multiple peripheral responses that are hierarchically regulated by the hypothalamus and we present evidence for non-cell autonomous signaling of dietary restriction gathered from a diverse range of models including invertebrates, mammalian cell culture, and rodent studies.


Assuntos
Envelhecimento/fisiologia , Restrição Calórica , Hipotálamo/fisiologia , Longevidade/fisiologia , Animais , Neurônios/fisiologia
8.
Am J Physiol Endocrinol Metab ; 302(8): E987-91, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22318949

RESUMO

To discover hypothalamic genes that might play a role in regulating energy balance, we carried out a microarray screen for genes induced by a 48-h fast in male C57Bl/6J mouse hypothalamus. One such gene was Fkbp51 (FK506 binding protein 5; Locus NP_034350). The product of this gene is of interest because it blocks glucocorticoid action, suggesting that fasting-induced elevation of this gene in the hypothalamus may reduce glucocorticoid negative feedback, leading to elevated glucocorticoid levels, thus promoting obese phenotypes. Subsequent analysis demonstrated that a 48-h fast induces Fkbp51 in ventromedial, paraventricular, and arcuate hypothalamic nuclei of mice and rats. To assess if hypothalamic Fkbp51 promotes obesity, the gene was transferred to the hypothalamus via an adeno-associated virus vector. Within 2 wk following Fkbp51 overexpression, mice on a high-fat diet exhibited elevated body weight, without hyperphagia, relative to mice receiving the control mCherry vector. Body weight remained elevated for more than 8 wk and was associated with elevated corticosterone and impaired glucose tolerance. These studies suggest that elevated hypothalamic Fkbp51 promotes obese phenotypes.


Assuntos
Jejum/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Regulação para Cima , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Corticosterona/sangue , Ingestão de Energia , Perfilação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/sangue , Análise de Sequência com Séries de Oligonucleotídeos , Núcleo Hipotalâmico Paraventricular/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas de Ligação a Tacrolimo/antagonistas & inibidores , Proteínas de Ligação a Tacrolimo/genética , Núcleo Hipotalâmico Ventromedial/metabolismo , Aumento de Peso
9.
Nat Med ; 17(9): 1121-7, 2011 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-21873987

RESUMO

Previous studies have proposed roles for hypothalamic reactive oxygen species (ROS) in the modulation of circuit activity of the melanocortin system. Here we show that suppression of ROS diminishes pro-opiomelanocortin (POMC) cell activation and promotes the activity of neuropeptide Y (NPY)- and agouti-related peptide (AgRP)-co-producing (NPY/AgRP) neurons and feeding, whereas ROS-activates POMC neurons and reduces feeding. The levels of ROS in POMC neurons were positively correlated with those of leptin in lean and ob/ob mice, a relationship that was diminished in diet-induced obese (DIO) mice. High-fat feeding resulted in proliferation of peroxisomes and elevated peroxisome proliferator-activated receptor γ (PPAR-γ) mRNA levels within the hypothalamus. The proliferation of peroxisomes in POMC neurons induced by the PPAR-γ agonist rosiglitazone decreased ROS levels and increased food intake in lean mice on high-fat diet. Conversely, the suppression of peroxisome proliferation by the PPAR antagonist GW9662 increased ROS concentrations and c-fos expression in POMC neurons. Also, it reversed high-fat feeding-triggered elevated NPY/AgRP and low POMC neuronal firing, and resulted in decreased feeding of DIO mice. Finally, central administration of ROS alone increased c-fos and phosphorylated signal transducer and activator of transcription 3 (pStat3) expression in POMC neurons and reduced feeding of DIO mice. These observations unmask a previously unknown hypothalamic cellular process associated with peroxisomes and ROS in the central regulation of energy metabolism in states of leptin resistance.


Assuntos
Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , PPAR gama/metabolismo , Peroxissomos/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Relacionada com Agouti/metabolismo , Anilidas/farmacologia , Animais , Linhagem Celular , Ingestão de Alimentos/fisiologia , Eletrofisiologia , Proteínas de Fluorescência Verde , Hipotálamo/citologia , Camundongos , Camundongos Obesos , Neuropeptídeo Y/metabolismo , PPAR gama/antagonistas & inibidores , Reação em Cadeia da Polimerase , Pró-Opiomelanocortina/metabolismo
10.
Endocrinology ; 151(11): 5206-17, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20881243

RESUMO

Nutrient-sensitive hypothalamic neurons regulate energy balance and glucose homeostasis, but the molecular mechanisms mediating hypothalamic responses to nutritional state remain incompletely characterized. To address these mechanisms, the present studies used quantitative PCR to characterize the expression of a panel of genes the hypothalamic expression by nutritional status of which had been suggested by DNA microarray studies. Although these genes regulate a variety of function, the most prominent set regulate intermediary metabolism, and the overall pattern clearly indicated that a 48-h fast produced a metabolic reprogramming away from glucose metabolism and toward the utilization of alternative fuels, particularly lipid metabolism. This general reprogramming of intermediary metabolism by fasting was observed both in cortex and hypothalamus but most prominently in hypothalamus. The effect of fasting on the expression of these genes may be mediated by reduction in plasma glucose or glucose metabolism, rather than leptin, because they were generally recapitulated by hypoglycemia even in the presence of elevated insulin and in vitro by low glucose but were not recapitulated in ob/ob mice. These studies suggest that fasting reduces glucose metabolism and thus minimizes the production of hypothalamic malonyl-coenzyme A. However, because the reprogramming of glucose metabolism by fasting was also observed in cortex, this apparent substrate competition may mediate more general responses to nutritional deprivation, including those responsible for the protective effects of dietary restriction. The present studies also provide a large panel of novel glucose-regulated genes that can be used as markers of glucose action to address mechanisms mediating hypothalamic responses to nutritional state.


Assuntos
Jejum/metabolismo , Glicólise/fisiologia , Hipotálamo/metabolismo , Metabolismo dos Lipídeos/fisiologia , Neurônios/metabolismo , Estado Nutricional/fisiologia , Análise de Variância , Animais , Córtex Cerebral/metabolismo , Expressão Gênica , Insulina/metabolismo , Leptina/metabolismo , Masculino , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Brain Res ; 1280: 77-83, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19445909

RESUMO

In rats and humans estradiol attenuates neuroendocrine responses to hypoglycemia. Since neuroendocrine responses to hypoglycemia are mediated by hypothalamic neurons, we assessed if estradiol attenuates hypoglycemia-induced gene expression in the hypothalamus in female ovariectomized mice. As expected, estradiol-implanted ovariectomized mice exhibited increased plasma estradiol, increased uterine weight, decreased body weight, decreased visceral adiposity, and enhanced glucose tolerance with decreased plasma insulin. Estradiol-implanted mice exhibited attenuated hypoglycemia-induced gene expression of both glucose transporter 1 (Glut1) and inhibitor of kappa beta signaling (IkappaB) in the hypothalamus but not in the liver. Estradiol also attenuated hypoglycemia-induced plasma glucagon, pituitary proopiomelanocortin (POMC), and adrenal c-fos, consistent with impaired counterregulatory responses to hypoglycemia. In addition, estradiol inhibited hypothalamic expression of carnitine palmitoyltransferase (CPT1a and CPT1c) and pyruvate dehydrogenase kinase 4 (PDK4), effects that would be expected to enhance the accumulation of long-chain fatty acids and glycolysis. Taken together, these findings suggest hypothalamic mechanisms mediating attenuation of hypoglycemia-induced neuroendocrine responses.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Hipoglicemia/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Carnitina O-Palmitoiltransferase/metabolismo , Estradiol/sangue , Estrogênios/sangue , Feminino , Expressão Gênica/efeitos dos fármacos , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 1/metabolismo , Hipoglicemia/tratamento farmacológico , Hipoglicemia/genética , Proteínas I-kappa B/metabolismo , Insulina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Ovariectomia , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Distribuição Aleatória , Útero/efeitos dos fármacos , Útero/patologia
12.
Endocrinology ; 149(2): 703-10, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17974626

RESUMO

The regulation of neuroendocrine electrical activity and gene expression by glucose is mediated through several distinct metabolic pathways. Many studies have implicated AMP and ATP as key metabolites mediating neuroendocrine responses to glucose, especially through their effects on AMP-activated protein kinase (AMPK), but other studies have suggested that glycolysis, and in particular the cytoplasmic conversion of nicotinamide adenine dinucleotide (NAD+) to reduced NAD (NADH), may play a more important role than oxidative phosphorylation for some effects of glucose. To address these molecular mechanisms further, we have examined the regulation of agouti-related peptide (AgRP) in a clonal hypothalamic cell line, N-38. AgRP expression was induced monotonically as glucose concentrations decreased from 10 to 0.5 mm glucose and with increasing concentrations of glycolytic inhibitors. However, neither pyruvate nor 3-beta-hydroxybutyrate mimicked the effect of glucose to reduce AgRP mRNA, but on the contrary, produced the opposite effect of glucose and actually increased AgRP mRNA. Nevertheless, 3beta-hydroxybutyrate mimicked the effect of glucose to increase ATP and to decrease AMPK phosphorylation. Similarly, inhibition of AMPK by RNA interference increased, and activation of AMPK decreased, AgRP mRNA. Additional studies demonstrated that neither the hexosamine nor the pentose/carbohydrate response element-binding protein pathways mediate the effects of glucose on AgRP expression. These studies do not support that either ATP or AMPK mediate effects of glucose on AgRP in this hypothalamic cell line but support a role for glycolysis and, in particular, NADH. These studies support that cytoplasmic or nuclear NADH, uniquely produced by glucose metabolism, mediates effects of glucose on AgRP expression.


Assuntos
Proteína Relacionada com Agouti/genética , Glicólise/fisiologia , Hipotálamo/citologia , Neurônios/metabolismo , Fosforilação Oxidativa , Proteínas Quinases Ativadas por AMP , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Desoxiglucose/farmacologia , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucosamina/farmacologia , Glucose/farmacologia , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Hexosaminas/metabolismo , Iodoacetatos/farmacologia , Corpos Cetônicos/farmacologia , Camundongos , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , NAD/metabolismo , Neurônios/citologia , Via de Pentose Fosfato/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ácido Pirúvico/farmacologia , Interferência de RNA
13.
Endocrinology ; 148(4): 1928-32, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17218412

RESUMO

Because appetite, hypothalamic gene expression, reproductive function, and adrenal function are highly sensitive to acute changes in plasma glucose levels, it has been hypothesized hypothalamic neurons sensitive to glucose play a role in regulating these functions. To assess this hypothesis, we examined these neuronendocrine functions in mice in which the glucokinase gene, which plays an essential role in neuroendocrine glucose sensing, has been ablated. Haploinsufficiency in heterozygous glucokinase knockout mice produced effects similar to those produced by hypoglycemia: impaired reproductive function, elevated plasma corticosterone, increased food intake, and hypothalamic gene expression similar to that observed in fasted or leptin-deficient obese mice (increased hypothalamic neuropeptide Y mRNA and reduced hypothalamic proopiomelanocortin mRNA). Plasma glucose was elevated 2-fold in glucokinase knockout mice, consistent with a maturity-onset diabetes of the young phenotype, but plasma insulin and leptin levels were normal. These data support the hypothesis that glucokinase plays a key role in the neuroendocrine regulation of metabolic economy.


Assuntos
Ingestão de Alimentos/genética , Glucocorticoides/metabolismo , Glucoquinase/fisiologia , Hipotálamo/metabolismo , Reprodução/genética , Animais , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Glucoquinase/genética , Hiperglicemia/genética , Insulina/sangue , Insulina/genética , Leptina/sangue , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas
14.
Cell Metab ; 5(1): 1-2, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17189199

RESUMO

Mechanisms by which the hypothalamus senses nutritional status are important for many metabolic diseases, including obesity and diabetes. Now, report that hypothalamic neurons sense nutritional deficit through a cascade of events involving leptin, corticosterone, and glial production of thyroid hormone, leading to neuronal induction of uncoupling protein.


Assuntos
Fome , Hipotálamo/metabolismo , Neurônios/metabolismo , Termogênese , Trifosfato de Adenosina/metabolismo , Animais , Metabolismo Energético , Jejum , Hipotálamo/citologia , Canais Iônicos/metabolismo , Leptina/metabolismo , Camundongos , Proteínas Mitocondriais/metabolismo , Tri-Iodotironina/metabolismo , Proteína Desacopladora 2
15.
Physiol Behav ; 85(1): 3-23, 2005 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-15924903

RESUMO

Since nutrition-sensitive feedback signals normally act to maintain relatively stable levels of both available and stored nutritional resources, failure in one or more of these feedback signals could plausibly lead to obese phenotypes. The glucostatic hypothesis in its original form posited that glucose serves as a physiological satiety factor (in the sense that post-prandial increases in plasma glucose cause meal termination), but in this form the hypothesis has been difficult to prove, and, especially since the discovery of leptin, the glucostatic hypothesis has largely been abandoned. Nevertheless, reduction of plasma glucose levels or glucose signaling produces a profile of neuroendocrine responses similar to those produced by leptin deficiency. Since leptin is not a physiological satiety factor (because it does not increase before meal termination), yet leptin deficiency causes obesity, we suggest that the glucostatic hypothesis be re-formulated without reference to satiety (i.e., short-term effects on food intake). Instead we argue that like leptin signaling, glucose signaling regulates long-term energy balance, in part by regulating metabolic rate but also by chronically regulating food intake. We further speculate that high-fat diets produce obesity in part because carbohydrates are, per calorie, more effective than lipids to reduce food intake and increase metabolic rate. In support of this glucoadipostatic hypothesis, the 5 present review examines evidence that obesity and the metabolic syndrome may be due to reduction in neuroendocrine sensitivity to glucose leading to increased metabolic efficiency.


Assuntos
Metabolismo Energético , Glucose/metabolismo , Obesidade/etiologia , Transdução de Sinais , Adenilato Quinase/metabolismo , Animais , Glicemia/metabolismo , Dieta , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Insulina/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/fisiologia , Resposta de Saciedade/fisiologia , Termogênese/fisiologia
16.
Neurochem Res ; 29(6): 1093-103, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15176466

RESUMO

DNA microarray analysis has been used to investigate relative changes in the level of gene expression in the CNS, including changes that are associated with disease, injury, psychiatric disorders, drug exposure or withdrawal, and memory formation. We have used oligonucleotide microarrays to identify hypothalamic genes that respond to nutritional manipulation. In addition to commonly used microarray analysis based on criteria such as fold-regulation, we have also found that simply carrying out multiple t tests then sorting by P value constitutes a highly reliable method to detect true regulation, as assessed by real-time polymerase chain reaction (PCR), even for relatively low abundance genes or relatively low magnitude of regulation. Such analyses directly suggested novel mechanisms that mediate effects of nutritional state on neuroendocrine function and are being used to identify regulated gene products that may elucidate the metabolic pathology of obese ob/ob, lean Vgf-/Vgf-, and other models with profound metabolic impairments.


Assuntos
Regulação da Expressão Gênica/genética , Hipotálamo/fisiologia , Metabolismo/genética , Fenômenos Fisiológicos da Nutrição , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenômenos Fisiológicos da Nutrição Animal , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
17.
Ann Med ; 35(6): 425-33, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14572167

RESUMO

The central melanocortin system plays an important role in the regulation of energy homeostasis both in rodents and humans, and melanocortin receptors appear to be the core of this system. Alpha-melanocyte-stimulating hormone (alpha-MSH) inhibits feeding through melanocrtin 3 and 4 receptors (MC3-R and MC4-R) as an endogenous agonist. Although mutations in the agouti gene cause an over-expression of agouti peptide which antagonizes effects of alpha-MSH at MC4-R in the brain and causes obese phenotypes, there was no evidence for the presence of an endogenous antagonist for MC3-R and MC4-R until agouti related protein (AGRP) was identified. AGRP is expressed primarily in the hypothalamic arcuate nucleus and central administration of AGRP stimulates feeding and weight gain, and decreases metabolic rate. Although a complete deletion of the AGRP gene does not produce any significant metabolic phenotypes, reduction in AGRP expression by RNA interference is associated with increased metabolic rate along with reduced weight gain. The currently available data suggest that elevated AGRP mRNA along with reduced proopiomelanocortin (POMC) mRNA is associated with many types of obesity and agents antagonizing the effect of AGRP may be a potential therapeutic target in treating obesity and obesity-associated disorders in which endogenous hypothalamic AGRP is elevated.


Assuntos
Metabolismo Energético/fisiologia , Hipotálamo/fisiologia , Neuropeptídeos/genética , Proteínas/genética , Proteína Relacionada com Agouti , Jejum/fisiologia , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Obesidade/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular
18.
Brain Res ; 985(1): 1-12, 2003 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12957363

RESUMO

Hypothalamic POMC neurons mediate catabolic responses such as decreased food intake and increased energy expenditure by, in part, monitoring levels of metabolic factors such as glucose, insulin and leptin. Recently, fatty acid synthase inhibitors were reported to reduce body weight, inhibit food intake, and increase metabolic rate, possibly by acting on hypothalamic neurons through a mechanism involving malonyl-CoA accumulation. Given the observation that leptin mediates similar catabolic effects by, in part, activating hypothalamic POMC neurons, it is possible that other catabolic signals such as feeding and fatty acid synthase inhibition may also activate POMC neurons. To test this hypothesis, hypothalamic sections from mice that were fed or injected with the fatty acid synthase inhibitor cerulenin were examined for Fos (a marker for neuronal activation) and POMC product immunoreactivity and compared with similarly processed sections from leptin-injected mice. Feeding increased Fos immunoreactivity in the lateral peri-arcuate area of the hypothalamus of both wild-type and leptin-deficient ob/ob mice (P<0.05), indicating that nutritional activation of the hypothalamus can be leptin-independent. Furthermore, feeding significantly induced Fos immunoreactivity in neurons expressing POMC (P<0.003), indicating that feeding, like leptin, activates POMC neurons. Injection with cerulenin, like feeding and leptin, also increased Fos immunoreactivity in the lateral peri-arcuate area (P<0.03) and, more specifically, in neurons expressing POMC. In contrast, injection with cerulenin had no grossly observable effects on cortical Fos immunoreactivity and appeared to suppress fasting-induced Fos immunoreactivity by about 35% (although the decrease did not reach statistical significance) in the medial arcuate nucleus, an area associated with anabolic responses such as increased food intake. Injection with cerulenin also decreased Fos immunoreactivity in the granular layer of the dentate gyrus of the hippocampus by about 30% (P<0.05), further suggesting that cerulenin does not non-specifically activate wide varieties of neurons. These results suggest that activation of hypothalamic POMC neurons may help to mediate some of the catabolic effects associated with feeding, cerulenin and leptin.


Assuntos
Antifúngicos/farmacologia , Cerulenina/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Animais , Ingestão de Alimentos/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Imuno-Histoquímica , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos/genética , Neurônios/fisiologia , Pró-Opiomelanocortina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
19.
Diabetes ; 52(8): 1951-7, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12882910

RESUMO

Zucker (fa/fa) rats with defective leptin receptors are obese, hyperphagic, and hyperinsulinemic. For testing whether chronic activation of the central melanocortin pathway can bypass the defective leptin signaling and normalize altered energy homeostasis in these rats, recombinant adeno-associated virus encoding pro-opiomelanocortin (rAAV-POMC) or control vector was delivered bilaterally into the basal hypothalamus with coordinates targeting the arcuate nucleus. Thirty-eight days after POMC gene delivery, hypothalamic POMC expression increased fourfold and melanocortin signaling (indicated by phosphorylation of CREB) increased by 62% with respect to controls. There was a sustained reduction in food intake, a moderate but significant attenuation of weight gain, and a 24% decrease in visceral adiposity in rAAV-POMC rats. POMC gene delivery enhanced uncoupling protein 1 in brown adipose tissue (BAT) by more than fourfold. Fasting serum leptin, insulin, and cholesterol levels were also significantly reduced by rAAV-POMC treatment. This study demonstrates that targeted POMC gene delivery in the hypothalamus suppresses food intake and weight gain and reduces visceral adiposity and hyperinsulinemia in leptin-resistant obese Zucker rats. The mechanisms may involve the sustained hypophagia and the augmentation of thermogenesis in BAT.


Assuntos
Tecido Adiposo Marrom/fisiologia , Diabetes Mellitus/fisiopatologia , Comportamento Alimentar/fisiologia , Resistência à Insulina/fisiologia , Obesidade , Pró-Opiomelanocortina/genética , Proteína Relacionada com Agouti , Animais , Glicemia , Proteínas de Transporte/genética , Linhagem Celular , Colesterol/sangue , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Diabetes Mellitus/terapia , Ingestão de Alimentos/fisiologia , Jejum , Ácidos Graxos não Esterificados/sangue , Terapia Genética , Humanos , Hipotálamo/metabolismo , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intercelular , Canais Iônicos , Rim/citologia , Leptina/sangue , Proteínas de Membrana/genética , Proteínas Mitocondriais , Fosforilação , Proteínas/genética , Ratos , Ratos Zucker , Receptores de Superfície Celular/genética , Receptores para Leptina , Transdução de Sinais/fisiologia , Proteína Desacopladora 1 , Vísceras
20.
BMC Physiol ; 3: 5, 2003 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-12848900

RESUMO

BACKGROUND: Fasting and diabetes are characterized by elevated glucocorticoids and reduced insulin, leptin, elevated hypothalamic AGRP and NPY mRNA, and reduced hypothalamic POMC mRNA. Although leptin replacement can reverse changes in hypothalamic gene expression associated with fasting and diabetes, leptin also normalizes corticosterone; therefore the extent to which the elevated corticosterone contributes to the regulation of hypothalamic gene expression in fasting and diabetes remains unclear. To address if elevated corticosterone is necessary for hypothalamic responses to fasting and diabetes, we assessed the effects of adrenalectomy on hypothalamic gene expression in 48-hour-fasted or diabetic mice. To assess if elevated corticosterone is sufficient for the hypothalamic responses to fasting and diabetes, we assessed the effect of corticosterone pellets implanted for 48 hours on hypothalamic gene expression. RESULTS: Fasting and streptozotocin-induced diabetes elevated plasma glucocorticoid levels and reduced serum insulin and leptin levels. Adrenalectomy prevented the rise in plasma glucocorticoids associated with fasting and diabetes, but not the associated reductions in insulin or leptin. Adrenalectomy blocked the effects of fasting and diabetes on hypothalamic AGRP, NPY, and POMC expression. Conversely, corticosterone implants induced both AGRP and POMC mRNA (with a non-significant trend toward induction of NPY mRNA), accompanied by elevated insulin and leptin (with no change in food intake or body weight). CONCLUSION: These data suggest that elevated plasma corticosterone mediate some effects of fasting and diabetes on hypothalamic gene expression. Specifically, elevated plasma corticosterone is necessary for the induction of NPY mRNA with fasting and diabetes; since corticosterone implants only produced a non-significant trend in NPY mRNA, it remains uncertain if a rise in corticosterone may be sufficient to induce NPY mRNA. A rise in corticosterone is necessary to reduce hypothalamic POMC mRNA with fasting and diabetes, but not sufficient for the reduction of hypothalamic POMC mRNA. Finally, elevated plasma corticosterone is both necessary and sufficient for the induction of hypothalamic AGRP mRNA with fasting and diabetes.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Jejum/fisiologia , Regulação da Expressão Gênica/fisiologia , Glucocorticoides/fisiologia , Hipotálamo/fisiologia , Hipotálamo/fisiopatologia , Medula Suprarrenal/fisiologia , Medula Suprarrenal/fisiopatologia , Medula Suprarrenal/cirurgia , Adrenalectomia/métodos , Proteína Relacionada com Agouti , Animais , Corticosterona/administração & dosagem , Corticosterona/farmacologia , Diabetes Mellitus Experimental/sangue , Implantes de Medicamento/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/sangue , Hipotálamo/efeitos dos fármacos , Insulina/deficiência , Insulina/genética , Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/genética , Pró-Opiomelanocortina/genética , Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA