Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Nature ; 599(7885): 436-441, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34732894

RESUMO

The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.


Assuntos
Desenvolvimento Infantil/fisiologia , Estado Nutricional/fisiologia , Puberdade/fisiologia , Receptor Tipo 3 de Melanocortina/metabolismo , Maturidade Sexual/fisiologia , Adolescente , Idoso de 80 Anos ou mais , Animais , Criança , Ciclo Estral/genética , Ciclo Estral/fisiologia , Feminino , Homozigoto , Humanos , Hipotálamo/citologia , Hipotálamo/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Melanocortinas/metabolismo , Menarca/genética , Menarca/fisiologia , Camundongos , Fenótipo , Puberdade/genética , Receptor Tipo 3 de Melanocortina/deficiência , Receptor Tipo 3 de Melanocortina/genética , Maturidade Sexual/genética , Fatores de Tempo , Aumento de Peso
2.
Mol Psychiatry ; 19(10): 1085-94, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24514567

RESUMO

Anorexia nervosa (AN) is a complex and heritable eating disorder characterized by dangerously low body weight. Neither candidate gene studies nor an initial genome-wide association study (GWAS) have yielded significant and replicated results. We performed a GWAS in 2907 cases with AN from 14 countries (15 sites) and 14 860 ancestrally matched controls as part of the Genetic Consortium for AN (GCAN) and the Wellcome Trust Case Control Consortium 3 (WTCCC3). Individual association analyses were conducted in each stratum and meta-analyzed across all 15 discovery data sets. Seventy-six (72 independent) single nucleotide polymorphisms were taken forward for in silico (two data sets) or de novo (13 data sets) replication genotyping in 2677 independent AN cases and 8629 European ancestry controls along with 458 AN cases and 421 controls from Japan. The final global meta-analysis across discovery and replication data sets comprised 5551 AN cases and 21 080 controls. AN subtype analyses (1606 AN restricting; 1445 AN binge-purge) were performed. No findings reached genome-wide significance. Two intronic variants were suggestively associated: rs9839776 (P=3.01 × 10(-7)) in SOX2OT and rs17030795 (P=5.84 × 10(-6)) in PPP3CA. Two additional signals were specific to Europeans: rs1523921 (P=5.76 × 10(-)(6)) between CUL3 and FAM124B and rs1886797 (P=8.05 × 10(-)(6)) near SPATA13. Comparing discovery with replication results, 76% of the effects were in the same direction, an observation highly unlikely to be due to chance (P=4 × 10(-6)), strongly suggesting that true findings exist but our sample, the largest yet reported, was underpowered for their detection. The accrual of large genotyped AN case-control samples should be an immediate priority for the field.


Assuntos
Anorexia Nervosa/genética , Povo Asiático/genética , Calcineurina/genética , Proteínas de Transporte/genética , Estudos de Casos e Controles , Proteínas Culina/genética , Feminino , Estudo de Associação Genômica Ampla , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Japão , Masculino , Metanálise como Assunto , Proteínas Nucleares/genética , Polimorfismo de Nucleotídeo Único , População Branca/genética
3.
Int J Obes (Lond) ; 37(8): 1147-53, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23229731

RESUMO

CONTEXT: Patients with pseudohypoparathyroidism type 1a (PHP-1a) develop early-onset obesity. The abnormality in energy expenditure and/or energy intake responsible for this weight gain is unknown. OBJECTIVE: The aim of this study was to evaluate energy expenditure in children with PHP-1a compared with obese controls. PATIENTS: We studied 6 obese females with PHP-1a and 17 obese female controls. Patients were recruited from a single academic center. MEASUREMENTS: Resting energy expenditure (REE) and thermogenic effect of a high fat meal were measured using whole room indirect calorimetry. Body composition was assessed using whole body dual energy x-ray absorptiometry. Fasting glucose, insulin, and hemoglobin A1C were measured. RESULTS: Children with PHP-1a had decreased REE compared with obese controls (P<0.01). After adjustment for fat-free mass, the PHP-1a group's REE was 346.4 kcals day(-1) less than obese controls (95% CI (-585.5--106.9), P<0.01). The thermogenic effect of food (TEF), expressed as percent increase in postprandial energy expenditure over REE, was lower in PHP-1a patients than obese controls, but did not reach statistical significance (absolute reduction of 5.9%, 95% CI (-12.2-0.3%), P=0.06). CONCLUSIONS: Our data indicate that children with PHP-1a have decreased REE compared with the obese controls, and that may contribute to the development of obesity in these children. These patients may also have abnormal diet-induced thermogenesis in response to a high-fat meal. Understanding the causes of obesity in PHP-1a may allow for targeted nutritional or pharmacologic treatments in the future.


Assuntos
Glicemia/metabolismo , Hemoglobinas Glicadas/metabolismo , Insulina/sangue , Obesidade Pediátrica/metabolismo , Pseudo-Hipoparatireoidismo/metabolismo , Aumento de Peso , Absorciometria de Fóton , Adolescente , Idade de Início , Metabolismo Basal , Composição Corporal , Calorimetria Indireta , Criança , Suscetibilidade a Doenças , Metabolismo Energético/genética , Feminino , Humanos , Obesidade Pediátrica/epidemiologia , Obesidade Pediátrica/genética , Fenótipo , Polimorfismo de Nucleotídeo Único , Período Pós-Prandial , Pseudo-Hipoparatireoidismo/epidemiologia , Pseudo-Hipoparatireoidismo/genética , Descanso , Termogênese , Estados Unidos/epidemiologia
4.
Kidney Int ; 69(5): 794-7, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16518340

RESUMO

Leptin is an adipocyte-derived hormone that acts as a major regulator of food intake and energy homeostasis. It circulates both as a free and as a protein-bound entity. Leptin is released into the blood in proportion to the amount of body fat and exerts sustained inhibitory effects on food intake while increasing energy expenditure. The leptin receptor belongs to the class I cytokine receptor superfamily and possesses strong homology to the signal-transducing subunits of the IL-6 receptor. The hypothalamic melanocortin system, and specifically the melanocortin-4 receptor (MC-4R), is critical in mediating leptin's effect on appetite and metabolism. Serum leptin concentrations are elevated in patients with chronic kidney disease (CKD) and correlate with C-reactive protein levels suggesting that inflammation is an important factor that contributes to hyperleptinemia in CKD. Hyperleptinemia may be important in the pathogenesis of inflammation-associated cachexia in CKD. We showed that experimental uremic cachexia was attenuated in db/db mice, a model of leptin receptor deficiency. Nephrectomy in these animals did not result in any change in weight gain, body composition, resting metabolic rate, and efficiency of food consumption. Furthermore, experimental uremic cachexia could be ameliorated by blocking leptin signaling through the hypothalamic MC-4R. MC-4R knockout mice or mice administered the MC-4R and MC-3R antagonist, agouti-related peptide, resisted uremia-induced loss of lean body mass and maintained normal basal metabolic rates. Thus, melanocortin receptor antagonism may provide a novel therapeutic strategy for inflammation-associated cachexia in CKD.


Assuntos
Caquexia/fisiopatologia , Inflamação/fisiopatologia , Falência Renal Crônica/fisiopatologia , Leptina/fisiologia , Animais , Apetite/fisiologia , Caquexia/etiologia , Humanos , Inflamação/complicações , Mediadores da Inflamação/fisiologia , Falência Renal Crônica/complicações , Hormônios Estimuladores de Melanócitos/fisiologia , Camundongos , Receptores de Superfície Celular/fisiologia , Receptores para Leptina , Receptores de Melanocortina/fisiologia , Transdução de Sinais
5.
Neuropeptides ; 36(2-3): 77-84, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12359499

RESUMO

Identifying the role of the melanocortin system in regulating energy homeostasis has relied on both genetic and pharmacological studies. The key findings included 1) that the coat color phenotype in the lethal yellow (A(Y)/a) mouse is due to antagonism of the melanocortin-1 receptor (MC1R) by the agouti gene product; 2) the MC3R and MC4R are expressed in CNS centers involved in energy homeostasis, and 3) the combined results of pharmacological studies showing that agouti is an antagonist of the MC4R and transgenic studies showing that inhibition or loss of the MC4R recapitulate the lethal yellow phenotype. Pro-opiomelanocortin (POMC), MC3R, and MC4R knockouts are obese and are now being used to further analyze melanocortin receptor function. The obesity phenotype observed in the MC3R and MC4R knockouts (KO) differ markedly. MC4RKO mice are hyperphagic, do not regulate pathways that increase energy expenditure (diet-induced thermogenesis) and physical activity in response to hyperphagia, and can develop type 2 diabetes. In contrast, MC3R deficient mice are not hyperphagic, have a normal metabolic response to increased energy consumption, and do not develop diabetes. The mechanism underlying the increased adiposity in the MC3R knockout remains unclear, but might be related to changes in nutrient partitioning or physical activity.


Assuntos
Receptores da Corticotropina/genética , Animais , Hipotálamo/fisiologia , Leptina/fisiologia , Camundongos , Camundongos Knockout , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 3 de Melanocortina , Receptor Tipo 4 de Melanocortina , Receptores da Corticotropina/fisiologia , Receptores de Melanocortina
7.
Trends Genet ; 17(10): S50-4, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11585677

RESUMO

Our understanding of body weight regulation has been greatly advanced by the characterization of previously existing mutations in mice that cause obesity. Subsequent analysis of a number of mouse knockout models has greatly expanded the number of genes known to influence adiposity by affecting metabolic rate, physical activity, and/or appetite.


Assuntos
Obesidade/genética , Animais , Modelos Animais de Doenças , Hormônios Esteroides Gonadais/genética , Humanos , Camundongos , Camundongos Knockout , Neuropeptídeo Y/genética , Neurotransmissores/genética , Obesidade/etiologia , Pró-Opiomelanocortina/genética , Receptores Adrenérgicos beta 3/genética , Receptores da Corticotropina/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Melanocortina , Receptores de Neuropeptídeo Y/genética , Fatores de Transcrição/genética
8.
Neuroendocrinology ; 74(3): 193-201, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11528221

RESUMO

Mutations in the melanocortin-4 receptor (MC4-R) cause obesity in both mice and humans, and the receptor is presumed to have an important role in the regulation of energy homeostasis. The MC4-R is expressed in discrete sets of neurons in the central nervous system, and thus it has been technically difficult to study the regulation of expression and the signaling mechanisms of this receptor. We report here a neuronal cell line that exhibits endogenous functional expression for the MC4-R. Initially, RT-PCR analysis showed the presence of MC4-R RNA in the hypothalamic GT1-1 and GT1-7 cells. In addition, GT1-7 cells expressed melanocortin-3 receptor while the GT1-1 subclone specifically expressed predominantly the MC4-R RNA. High-affinity binding sites were demonstrated in the GT1-1 and GT1-7 cells for NDP-alpha melanocyte-stimulating hormone (MSH; K(i) = 1.1 x 10(-10) and 1.8 x 10(-10) M) and agouti-related protein (AGRP; K(i) = 1.548 x 10(-9) and 1.663(-9) M). alpha-MSH-stimulated cAMP production in GT1-1 cells with an EC(50) of 2.2 x 10(-8) M, and cAMP production was inhibited in the presence of AGRP, an endogenous antagonist of the MC4-R. Stimulation of gonadotropin-releasing hormone (GnRH) secretion was achieved with 1 nM to 1 microM concentrations of NDP-alpha-MSH while no GnRH secretion was observed when the GT1-1 cells were treated with AGRP. The data presented here show that GT1-1 cells specifically express a functional MC4-R that couples to GnRH release.


Assuntos
Hipotálamo/metabolismo , Receptores de Peptídeos/metabolismo , alfa-MSH/análogos & derivados , Proteína Relacionada com Agouti , Animais , Linhagem Celular , Cricetinae , AMP Cíclico/metabolismo , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Mesocricetus , Camundongos , Células PC12 , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Receptor Tipo 4 de Melanocortina , alfa-MSH/metabolismo , alfa-MSH/farmacologia
9.
Biochemistry ; 40(20): 6164-79, 2001 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-11352754

RESUMO

In vitro mutagenesis of the mouse melanocortin-4 receptor (mMC4R) has been performed, based upon homology molecular modeling and previous melanocortin receptor mutagenesis studies that identified putative ligand-receptor interactions. Twenty-three mMC4 receptor mutants were generated and pharmacologically characterized using several melanocortin-based ligands [alpha-MSH, NDP-MSH, MTII, DNal (1')(7)-MTII, Nal(2')(7)-MTII, SHU9119, and SHU9005]. Selected mutant receptors possessing significant differences in the melanocortin-based peptide agonist and/or antagonist pharmacology were further evaluated using the endogenous antagonist agouti-related protein fragment hAGRP(83-132) and hAGRP(109-118) molecules. These studies of the mouse MC4R provide further experimental data suggesting that the conserved melanocortin receptor residues Glu92 (TM2), Asp114 (TM3), and Asp118 (TM3) (mouse MC4R numbering) are important for melanocortin-based peptide molecular recognition. Additionally, the Glu92 and Asp118 mMC4R residues are important for molecular recognition and binding of AGRP(83-132). We have identified the Phe176 (TM4), Tyr179 (TM4), Phe254 (TM6), and Phe259 (TM6) receptor residues as putatively interacting with the melanocortin-based ligand Phe(7) by differences between alpha-MSH and NDP-MSH agonist potencies. The Glu92, Asp118, and Phe253 mMC4R receptor residues appear to be critical for hAGRP(83-132) molecular recognition and binding while Phe176 appears to be important for functional antagonism of AGRP(83-132) and AGRP(109-118) but not molecular recognition. The Phe253 mMC4R residue appears to be important for AGRP(83-132) molecular recognition and general mMC4 receptor stimulation. The Phe254 and Phe259 mMC4R amino acids may participate in the differentiation of agonist versus antagonist activity of the melanocortin-based peptide antagonists SHU9119 and SHU9005, but not AGRP(83-132) or AGRP(109-118). The Met192 side chain when mutated to a Phe results in a constitutively active mMC4R that does not effect agonist ligand binding or potency. Melanocortin-based peptides modified at the 7 position of MTII with DPhe, DNal(1'), Nal(2'), and DNal(2') have been pharmacologically characterized at these mutant mouse MC4Rs. These data suggest a revised hypothesis for the mechanism of SHU9119 antagonism at the MC4R which may be attributed to the presence of a "bulky" naphthyl moiety at the 7 position (original hypothesis), and additionally that both the stereochemistry and naphthyl ring position (2' versus 1') are important for positioning of the ligand Arg(8) residue with the corresponding mMC4R amino acids.


Assuntos
Mutagênese Sítio-Dirigida , Peptídeos/farmacologia , Proteínas/metabolismo , Receptores da Corticotropina/química , Receptores da Corticotropina/metabolismo , Receptores de Peptídeos/química , Receptores de Peptídeos/metabolismo , alfa-MSH/agonistas , Proteína Relacionada com Agouti , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Lisina/genética , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/síntese química , Peptídeos/metabolismo , Fenilalanina/genética , Ligação Proteica/genética , Proteínas/química , Proteínas/farmacologia , Receptor Tipo 4 de Melanocortina , Receptores da Corticotropina/antagonistas & inibidores , Receptores da Corticotropina/genética , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/genética , Serina/genética , Relação Estrutura-Atividade , Transfecção , alfa-MSH/análogos & derivados , alfa-MSH/antagonistas & inibidores , alfa-MSH/química , alfa-MSH/metabolismo , alfa-MSH/farmacologia
10.
Nature ; 411(6836): 480-4, 2001 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-11373681

RESUMO

The administration of leptin to leptin-deficient humans, and the analogous Lepob/Lepob mice, effectively reduces hyperphagia and obesity. But common obesity is associated with elevated leptin, which suggests that obese humans are resistant to this adipocyte hormone. In addition to regulating long-term energy balance, leptin also rapidly affects neuronal activity. Proopiomelanocortin (POMC) and neuropeptide-Y types of neurons in the arcuate nucleus of the hypothalamus are both principal sites of leptin receptor expression and the source of potent neuropeptide modulators, melanocortins and neuropeptide Y, which exert opposing effects on feeding and metabolism. These neurons are therefore ideal for characterizing leptin action and the mechanism of leptin resistance; however, their diffuse distribution makes them difficult to study. Here we report electrophysiological recordings on POMC neurons, which we identified by targeted expression of green fluorescent protein in transgenic mice. Leptin increases the frequency of action potentials in the anorexigenic POMC neurons by two mechanisms: depolarization through a nonspecific cation channel; and reduced inhibition by local orexigenic neuropeptide-Y/GABA (gamma-aminobutyric acid) neurons. Furthermore, we show that melanocortin peptides have an autoinhibitory effect on this circuit. On the basis of our results, we propose an integrated model of leptin action and neuronal architecture in the arcuate nucleus of the hypothalamus.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Leptina/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Pró-Opiomelanocortina/fisiologia , Potenciais de Ação , Animais , Animais Geneticamente Modificados , Anorexia , Eletrofisiologia , Potenciais Evocados , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Neuropeptídeo Y/metabolismo , Ácido gama-Aminobutírico/metabolismo
11.
Nat Neurosci ; 4(6): 605-11, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11369941

RESUMO

In response to moderately increased dietary fat content, melanocortin-4 receptor-null mutant (MC4R-/-) mice exhibit hyperphagia and accelerated weight gain compared to wild-type mice. An increased feed efficiency (weight gain/kcal consumed) argues that mechanisms in addition to hyperphagia are instrumental in causing weight gain. We report two specific defects in coordinating energy expenditure with food intake in MC4R-/- mice. Wild-type mice respond to an increase in the fat content of the diet by rapidly increasing diet-induced thermogenesis and by increasing physical activity, neither of which are observed in MC4R-/- mice. Leptin-deficient and MC3R-/- mice regulate metabolic rate similarly to wild-type mice in this protocol. Melanocortinergic pathways involving MC4-R-regulated neurons, which rapidly respond to signals not requiring changes in leptin, thus seem to be important in regulating metabolic and behavioral responses to dietary fat.


Assuntos
Gorduras na Dieta/farmacologia , Hiperfagia/genética , Receptores da Corticotropina/fisiologia , Tecido Adiposo Marrom/fisiologia , Animais , Cruzamentos Genéticos , Metabolismo Energético , Comportamento Alimentar , Feminino , Homeostase , Leptina/deficiência , Leptina/genética , Leptina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esforço Físico , Receptor Tipo 3 de Melanocortina , Receptor Tipo 4 de Melanocortina , Receptores da Corticotropina/deficiência , Receptores da Corticotropina/genética , Valores de Referência , Termogênese , Aumento de Peso
12.
Cancer Res ; 61(4): 1432-8, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11245447

RESUMO

Individuals affected with either acute or chronic diseases often show disorders of nutrient balance. In some cases, a devastating state of malnutrition known as cachexia arises, brought about by a synergistic combination of a dramatic decrease in appetite and an increase in metabolism of fat and lean body mass. Stimulation of the hypothalamic melanocortin 4 receptor (MC4-R) produces relative anorexia and increased metabolic rate, even in a relatively starved state. Here we demonstrate that cachexia induced by lipopolysaccharide administration and by tumor growth is ameliorated by central MC4-R blockade. MC4-R knock-out mice or mice administered the MC3-R/MC4-R antagonist, agouti-related peptide, resist tumor-induced loss of lean body mass, and maintain normal circadian activity patterns during tumor growth. The final tumor mass is not affected in these animals, providing further support for the potential role of MC4-R antagonism in the treatment of cachexia in disease states.


Assuntos
Caquexia/prevenção & controle , Receptores de Peptídeos/antagonistas & inibidores , Proteína Relacionada com Agouti , Animais , Caquexia/induzido quimicamente , Caquexia/etiologia , Carcinoma Pulmonar de Lewis/complicações , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Proteínas/farmacologia , Receptor Tipo 3 de Melanocortina , Receptor Tipo 4 de Melanocortina , Receptores da Corticotropina/antagonistas & inibidores , Receptores de Peptídeos/genética , Receptores de Peptídeos/fisiologia , Sarcoma Experimental/complicações , Transdução de Sinais/fisiologia
13.
Recent Prog Horm Res ; 56: 359-75, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11237221

RESUMO

Recent advances in our understanding of the regulation of body weight, appetite, and metabolic rate have highlighted the role of the adipose-derived hormone leptin and its receptor as fundamental modulators of these processes. Investigations of the neural targets for leptin action--as well as characterization of the agouti obesity syndrome--have, in turn, led to the discovery of fundamental neural pathways involved in the central regulation of energy homeostasis. In particular, the central melanocortin system has been shown to regulate appetite and metabolic rate in rodents; mutations in this system have been demonstrated to result in obesity in humans. Overall, the melanocortin system appears to function as a bidirectional rheostat in the regulation of energy intake and expenditure in rodents and potentially in humans. The first section of this chapter will focus on the development of our understanding of melanocortin physiology in the context of obesity. In particular, recent data regarding the interplay between melanocortin and neuropeptide Y (NPY) signaling at a cellular level will be discussed. The following section will discuss the hypothesis that melanocortin signaling plays a role in pathological weight loss and hypermetabolism observed in murine cachexia models. The potential role of this system in integrating a variety of anorexic and cachexic signals, as well as the potential for its pharmacological manipulation in the treatment of human cachexia, will be discussed.


Assuntos
alfa-MSH/metabolismo , alfa-MSH/fisiologia , Animais , Caquexia/etiologia , Citocinas/metabolismo , Humanos , Camundongos , Modelos Biológicos , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Ligação Proteica , Ratos , Transdução de Sinais
14.
Int J Obes Relat Metab Disord ; 25 Suppl 5: S63-7, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11840218

RESUMO

Arcuate nucleus neurons are known to be responsive to a wide array of hormones and nutrients, including leptin, insulin, gonadal steroids and glucose. In addition to potential transport mechanisms, peripheral substances may access these neurons via arcuate cell bodies in and projections to the median eminence, a region considered to be a circumventricular organ. The arcuate is a potent site of leptin action, probably mediating a component of leptin's effects via arcuate neuropeptide Y/agouti-related peptide (NPY/AgRP) and pro-opiomelanocortin (POMC) neurons, and implicating this structure in the long-term control of energy stores. However, ghrelin, the endogenous ligand of the growth hormone secretagogue receptor, may also stimulate feeding and weight gain, in part through action on receptors in arcuate NPY neurons. Since ghrelin is secreted by the stomach upon content depletion, with a half-life of no more than an hour, the arcuate nucleus may also be important in sensing and responding to acute changes in nutrients. We have developed a system for recording from arcuate POMC neurons using a mouse containing a transgene in which the POMC promoter is driving expression of the green fluorescent protein (GFP). In these mice, 99% of the beta-endorphin positive neurons express GFP, making whole cell patch clamp recordings from the sparsely distributed POMC neurons facile. All of the POMC neurons appear to be activated by leptin, via two different mechanisms, while approximately 30-50% of the neurons appear to be inhibited by a gamma-melanocyte stimulating hormone (MSH) specific agonist. The latter result suggests that the melanocortin-3 receptor (MC3-R) may act as an autoinhibitory receptor on some POMC neurons. This hypothalamic slice preparation also confirms the responsiveness of arcuate POMC neurons to a wide variety of nutrients and hormones. Thus the arcuate melanocortin system is best described as a conduit of many diverse signals involved in energy homeostasis, with leptin acting tonically to regulate the responsiveness of the circuit to a wide variety of hormones and nutrients.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Metabolismo Energético , Homeostase , Transdução de Sinais , Animais , Humanos , Leptina/fisiologia , Pró-Opiomelanocortina/fisiologia
15.
Annu Rev Genet ; 34: 687-745, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11092843

RESUMO

Obesity is a health problem of epidemic proportions in the industrialized world. The cloning and characterization of the genes for the five naturally occurring monogenic obesity syndromes in the mouse have led to major breakthroughs in understanding the physiology of energy balance and the contribution of genetics to obesity in the human population. However, the regulation of energy balance is an extremely complex process, and it is quickly becoming clear that hundreds of genes are involved. In this article, we review the naturally occurring monogenic and polygenic obese mouse strains, as well as the large number of transgenic and knockout mouse models currently available for the study of obesity and energy balance.


Assuntos
Metabolismo Energético/genética , Modelos Genéticos , Obesidade/genética , Animais , Predisposição Genética para Doença , Camundongos , Mutação
16.
Neuroendocrinology ; 72(2): 126-32, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10971147

RESUMO

A study of the effects of insulin-induced hypoglycemia in the obese yellow agouti A(y) mouse was initiated to test the hypothesis that the central melanocortin pathways are required for a normal sympathetic response to hypoglycemia. An experimental protocol was performed in which young nonobese male mice were isolated and fasted beginning on day 1, then tested for glucose responses to insulin-induced hypoglycemia on day 2. Normal mice demonstrated the expected glucose rebound to hypoglycemia, exceeding baseline glucose levels by 2-3 times as a consequence of increased gluconeogenesis and glycogenolysis before returning to baseline levels. A(y) animals lacked the rebound, exhibiting instead a gradual restoration of baseline glucose levels. The results suggested a defective sympathetic response to hypoglycemia in the A(y) mouse. However, a more detailed analysis demonstrated that the lack of a hyperglycemic rebound was due to an acute inhibition of feeding specifically in the A(y) mouse, which resulted not from the hypoglycemia stressor, but rather from the stress of isolation. Handling and intraperitoneal administration of saline also specifically inhibited food intake in the A(y) but not the wild-type mouse, while restraint stress had an equivalent inhibitory effect on food intake on wild-type and A(y) mice. Since the A(y) mouse has defective hypothalamic melanocortin-4 receptor (MC4-R) signaling, these data imply that the central melanocortin pathway is necessary for regulating the effects of stress on feeding behavior. Furthermore, these data demonstrate the need for exercising extreme caution in designing experiments to analyze feeding behavior and metabolism in genetic or pharmacological models involving perturbation of the melanocortin system.


Assuntos
Comportamento Alimentar/fisiologia , Estresse Psicológico/psicologia , Animais , Glicemia/metabolismo , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Hipoglicemiantes/farmacologia , Insulina/sangue , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/genética , Restrição Física , Isolamento Social , Estresse Psicológico/genética
18.
Endocrinology ; 141(9): 3072-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10965876

RESUMO

The central melanocortin system has been demonstrated to play a pivotal role in energy homeostasis. Genetic disruption of this system causes obesity in both humans and mice. Previous experiments have shown that centrally-administered melanocortin agonists inhibit food intake and stimulate oxygen consumption. Here we report that centrally-administered melanocortin agonists also inhibit basal insulin release, and alter glucose tolerance. Furthermore, increased plasma insulin levels occur in the young lean MC4-R knockout (MC4-RKO) mouse, and impaired insulin tolerance takes place before the onset of detectable hyperphagia or obesity. These data suggest that the central melanocortin system regulates not only energy intake and expenditure, but also processes related to energy partitioning, as indicated by effects on insulin release and peripheral insulin responsiveness. Previous studies emphasize the role of excess adipose mass in the development of tissue insulin resistance, leading to type II diabetes. The data presented here show that defects in the central control of glucose homeostasis may be an additional factor in some types of obesity-associated type II diabetes.


Assuntos
Sistema Nervoso Central/fisiologia , Insulina/sangue , Hormônios Estimuladores de Melanócitos/fisiologia , Animais , Glicemia/metabolismo , Glicemia/fisiologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Ácidos Graxos não Esterificados/sangue , Feminino , Teste de Tolerância a Glucose , Homeostase/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Oligopeptídeos/farmacologia , Receptor Tipo 4 de Melanocortina , Receptores de Peptídeos/agonistas , Receptores de Peptídeos/genética , Transdução de Sinais/fisiologia , alfa-MSH/análogos & derivados
19.
Endocrinology ; 141(9): 3518-21, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10965927

RESUMO

The central melanocortin system is critical for the long term regulation of energy homeostasis. Null mutations of the melanocortin-4 receptor (MC4-R) are associated with hyperphagia, obesity, and accelerated longitudinal growth in mice and humans. However, little is known about the function of another central melanocortin receptor, the MC3-R. To assess the role of the MC3-R in energy homeostasis, the majority of the mc3r coding sequence was deleted from the mouse genome. In contrast to the MC4-R knockout, which exhibits increased food intake, increased somatic growth, and defects in metabolism, mc3r-/- mice exhibit an exclusively metabolic syndrome. Homozygous null mc3r mice, while not significantly overweight, exhibit an approximately 50% to 60% increase in adipose mass. Mc3r-/- mice also exhibit an unusual increase in respiratory quotient when transferred onto high fat chow, suggesting a reduced ratio of fat/carbohydrate oxidation. Furthermore, male mc3r-/- mice also exhibit an approximately 50% reduction in locomotory behavior on the running wheel, suggesting reduced energy expenditure.


Assuntos
Obesidade/genética , Receptores da Corticotropina/deficiência , Receptores da Corticotropina/genética , Absorciometria de Fóton , Tecido Adiposo/metabolismo , Animais , Calorimetria Indireta , Clonagem Molecular , Dieta , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Marcação de Genes , Vetores Genéticos , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 3 de Melanocortina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...