Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 443(7109): 289-95, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16988703

RESUMO

The capacity to adjust food intake in response to changing energy requirements is essential for survival. Recent progress has provided an insight into the molecular, cellular and behavioural mechanisms that link changes of body fat stores to adaptive adjustments of feeding behaviour. The physiological importance of this homeostatic control system is highlighted by the severe obesity that results from dysfunction of any of several of its key components. This new information provides a biological context within which to consider the global obesity epidemic and identifies numerous potential avenues for therapeutic intervention and future research.


Assuntos
Peso Corporal/fisiologia , Sistema Nervoso Central/fisiologia , Comportamento Alimentar/fisiologia , Tecido Adiposo/metabolismo , Plasticidade Neuronal/fisiologia , Resposta de Saciedade/fisiologia
2.
Brain Res ; 964(1): 107-15, 2003 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-12573518

RESUMO

Recent studies have demonstrated that the metabolic hormones insulin and leptin can modulate behavioral performance in reward-related paradigms. However, specific anatomical substrate(s) within the CNS for these effects remain to be identified. We hypothesize that midbrain dopamine neurons, which have been implicated to be critical in the mediation of motivational and reward aspects of stimuli, contribute to these behavioral effects of insulin and leptin. As one approach to evaluate this hypothesis, we used double-labeling fluorescence immunohistochemistry to determine whether the midbrain dopamine neurons express insulin receptors or leptin receptors. Extensive co-expression of tyrosine hydroxylase (a marker for dopamine neurons) with both the insulin receptor and the leptin receptor was observed in the ventral tegmentum and substantia nigra. These findings suggest that midbrain dopamine neurons are direct targets of insulin and leptin, and that they participate in mediating the effects of these hormones on reward-seeking behavior.


Assuntos
Comportamento Alimentar/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Receptor de Insulina/metabolismo , Receptores de Superfície Celular/metabolismo , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Comportamento Animal/fisiologia , Dopamina/metabolismo , Imunofluorescência , Masculino , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores para Leptina , Recompensa , Substância Negra/citologia , Área Tegmentar Ventral/citologia
3.
Am J Physiol Endocrinol Metab ; 281(4): E655-64, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11551840

RESUMO

To develop an index of the activation of abdominal sympathetic nerves, we used Fos immunostaining of the celiac ganglion (CG) taken from rats receiving nicotine, preganglionic nerve stimulation, or glucopenic agents. Subcutaneous nicotine injection moderately increased Fos expression in the principal ganglionic cells of the CG (17 +/- 4 Fos+ per mm(2), approximately 12% of all principal CG cells), whereas subcutaneous saline had no effect (0 +/- 0 Fos+ per mm(2); n = 7; P < 0.01). Greater Fos expression was obtained by applying nicotine topically to the CG (71 +/- 8 Fos+ per mm(2); 52% of all principal CG cells, n = 5; P < 0.01 vs. topical saline, n = 4) and by preganglionic nerve stimulation (126 +/- 9 Fos+ per mm(2); 94% of all principal CG cells, n = 11; P < 0.01 vs. nerve isolation, n = 7). Moderate Fos expression was also observed in the CG after intraperitoneal 2-deoxy-D-glucose (2DG) injection (21 +/- 2 Fos+ per mm(2); 16% of all principal CG cells, n = 5; P < 0.01 vs. saline ip) or insulin injection (16 +/- 2 Fos+ per mm(2); 12% of all principal CG cells, n = 6; P < 0.01 vs. saline ip). Furthermore, Fos expression induced by 2DG was dose and time dependent. These data demonstrate significant Fos expression in the CG in response to chemical, electrical, and reflexive stimulation. Thus Fos expression in the CG may be a useful index to describe various levels of activation of its postganglionic sympathetic neurons.


Assuntos
Desoxiglucose/farmacologia , Gânglios Simpáticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes fos , Nicotina/farmacologia , Sistema Nervoso Simpático/fisiologia , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/fisiologia , Desoxiglucose/administração & dosagem , Relação Dose-Resposta a Droga , Estimulação Elétrica , Gânglios Simpáticos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Injeções Intraperitoneais , Injeções Subcutâneas , Masculino , Nicotina/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Wistar , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/fisiologia , Sistema Nervoso Simpático/efeitos dos fármacos
4.
J Neurosci ; 21(10): 3639-45, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11331393

RESUMO

The CNS melanocortin (MC) system is implicated as a mediator of the central effects of leptin, and reduced activity of the CNS MC system promotes obesity in both rodents and humans. Because activation of CNS MC receptors has direct effects on autonomic outflow and metabolism, we hypothesized that food intake-independent mechanisms contribute to development of obesity induced by pharmacological blockade of MC receptors in the brain and that changes in hypothalamic neuropeptidergic systems known to regulate weight gain [i.e., corticotropin-releasing hormone (CRH), cocaine-amphetamine-related transcript (CART), proopiomelanocortin (POMC), and neuropeptide Y (NPY)] would trigger this effect. Relative to vehicle-treated controls, third intracerebroventricular (i3vt) administration of the MC receptor antagonist SHU9119 to rats for 11 d doubled food and water intake (toward the end of treatment) and increased body weight ( approximately 14%) and fat content ( approximately 90%), hepatic glycogen content ( approximately 40%), and plasma levels of cholesterol ( approximately 48%), insulin ( approximately 259%), glucagon ( approximately 80%), and leptin ( approximately 490%), whereas spontaneous locomotor activity and body temperature were reduced. Pair-feeding of i3vt SHU9119-treated animals to i3vt vehicle-treated controls normalized plasma levels of insulin, glucagon, and hepatic glycogen content, but only partially reversed the elevations of plasma cholesterol ( approximately 31%) and leptin ( approximately 104%) and body fat content ( approximately 27%). Reductions in body temperature and locomotor activity induced by i3vt SHU9119 were not reversed by pair feeding, but rather were more pronounced. None of the effects found can be explained by peripheral action of the compound. The obesity effects occurred despite a lack in neuropeptide expression responses in the neuroanatomical range selected across the arcuate (i.e., CART, POMC, and NPY) and paraventricular (i.e., CRH) hypothalamus. The results indicate that reduced activity of the CNS MC pathway promotes fat deposition via both food intake-dependent and -independent mechanisms.


Assuntos
Comportamento Animal/fisiologia , Hipotálamo/metabolismo , Obesidade/metabolismo , Receptores da Corticotropina/metabolismo , Transdução de Sinais/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Composição Corporal/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Colesterol/sangue , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Glucagon/sangue , Hipotálamo/efeitos dos fármacos , Injeções Intraventriculares , Insulina/sangue , Leptina/sangue , Masculino , Hormônios Estimuladores de Melanócitos/administração & dosagem , Atividade Motora/efeitos dos fármacos , Neurotransmissores/genética , Neurotransmissores/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores da Corticotropina/antagonistas & inibidores , Receptores de Melanocortina , Transdução de Sinais/efeitos dos fármacos
5.
J Pediatr Endocrinol Metab ; 14 Suppl 6: 1417-29, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11837495

RESUMO

The brain plays a key role in the regulation of energy homeostasis, balancing food intake and energy expenditure to maintain adipose tissue mass. A widely accepted model proposes that energy homeostasis is modulated by hormones that circulate in the blood in proportion to adipose tissue mass. A major candidate 'adiposity signal' to the brain is the adipocyte hormone, leptin; this inhibits neuropeptide circuits that promote anabolic metabolism, and stimulates those that promote catabolic metabolism. It is hypothesized that leptin-responsive circuits in the hypothalamus project to caudal brainstem neuronal groups that integrate satiety signals converging on the brain from the stomach and intestine following ingestion of food. Leptin signaling to the brainstem via hypothalamic pathways potentially increases the brain's motor and autonomic responses to satiety signals, leading to smaller individual meals, reduced cumulative food intake, and a lower body weight. This mechanism explains how leptin deficiency or defects in the brain's processing of leptin signaling can result in a sustained increase in food intake and obesity.


Assuntos
Peso Corporal/fisiologia , Encéfalo/fisiologia , Ingestão de Alimentos/fisiologia , Leptina/fisiologia , Tecido Adiposo/fisiologia , Animais , Anorexia/etiologia , Humanos , Transdução de Sinais/fisiologia
6.
J Histochem Cytochem ; 48(12): 1593-99, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11101627

RESUMO

To increase the sensitivity of fluorescence in situ hybridization (FISH) for detection of low-abundance mRNAs, we performed FISH on cryostat sections of rat hypothalamus with biotin-labeled riboprobes to leptin receptor (ObRb) and amplified the signal by combining tyramide signal amplification (TSA) and Enzyme-Labeled Fluorescent alkaline phosphatase substrate (ELF) methods. First, TSA amplification was done with biotinylated tyramide. Second, streptavidin-alkaline phosphatase was followed by the ELF substrate, producing a bright green fluorescent reaction product. FISH signal for ObRb was undetectable when TSA or ELF methods were used alone, but intense ELF FISH signal was visible in hypothalamic neurons when the ELF protocol was preceded by TSA. The TSA-ELF was combined with FISH for pro-opiomelanocortin (POMC) and neuropeptide Y (NPY) mRNAs by hybridizing brain sections in a cocktail containing digoxigenin-labeled riboprobes to NPY or POMC mRNA and biotin-labeled riboprobes to ObRb mRNA. Dioxigenin-labeled NPY or POMC mRNA hybrids were subsequently detected first with IgG-Cy3. Then biotin-labeled leptin receptor hybrids were detected with the TSA-ELF method. Combining the ELF and TSA amplification techniques enabled FISH detection of scarce leptin receptor mRNAs and permitted the identification of leptin receptor mRNA in cells that also express NPY and POMC gene products.


Assuntos
Biotina/análogos & derivados , Proteínas de Transporte/análise , Leptina/metabolismo , RNA Mensageiro/análise , Receptores de Superfície Celular , Tiramina/análogos & derivados , Fosfatase Alcalina , Animais , Proteínas de Transporte/metabolismo , Hipotálamo/metabolismo , Hibridização in Situ Fluorescente/métodos , Masculino , Microscopia de Fluorescência , Ratos , Ratos Wistar , Receptores para Leptina , Estreptavidina
7.
J Neurosci ; 20(22): 8637-42, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11069973

RESUMO

Melanin-concentrating hormone (MCH) is an orexigenic neuropeptide produced by neurons of the lateral hypothalamic area (LHA). Because genetic MCH deficiency induces hypophagia and loss of body fat, we hypothesized that MCH neurons may represent a specific LHA pathway that, when inhibited, contributes to the pathogenesis of certain anorexia syndromes. To test this hypothesis, we measured behavioral, hormonal, and hypothalamic neuropeptide responses in two models of hyperestrogenemia in male rats, a highly reproducible anorexia paradigm. Whereas estrogen-induced weight loss engaged multiple systems that normally favor recovery of lost weight, the expected increase of MCH mRNA expression induced by energy restriction was selectively and completely abolished. These findings identify MCH neurons as specific targets of estrogen action and suggest that inhibition of these neurons may contribute to the hypophagic effect of estrogen.


Assuntos
Anorexia/metabolismo , Estrogênios/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Tumor de Células de Leydig/metabolismo , Melaninas/metabolismo , Hormônios Hipofisários/metabolismo , Redução de Peso/fisiologia , Proteína Relacionada com Agouti , Animais , Anorexia/induzido quimicamente , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Modelos Animais de Doenças , Implantes de Medicamento , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Hormônios/sangue , Hipotálamo/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Transplante de Neoplasias , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Redução de Peso/efeitos dos fármacos
8.
Regul Pept ; 92(1-3): 9-15, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-11024559

RESUMO

Treatment of rodents with exogenous leptin increases SOCS-3 mRNA levels in the arcuate nucleus (ARC) and dorsomedial nucleus (DMN) of the hypothalamus. To determine if SOCS-3 gene activity in the hypothalamus could be influenced by changes in physiological levels of circulating leptin, we performed in situ hybridization (ISH) and immunostaining for SOCS-3 expression in fed vs. fasted (48 h) rats. The ARC and DMN were the only regions of the diencephalon that showed SOCS-3 ISH and the autoradiographic ISH signal for SOCS-3 mRNA was visibly less in the ARC and DMN of fasted rats. The ISH signal for SOCS-3 mRNA was decreased 70% in the ARC and 90% in the DMN (to background levels) when animals were fasted (P<0.01), consistent with decreased immunostaining for SOCS-3 protein observed in the fasted rats. Double fluorescence ISH (FISH) analyses showed colocalization of SOCS-3 mRNA with mRNAs for NPY and POMC in the ARC. These findings are consistent with increased leptin signaling to the NPY and POMC neurons in the ARC by physiological levels of circulating leptin during normal feeding. Therefore, changes in SOCS-3 mRNA levels in the ARC and DMN can be viewed as an indicator of relative physiological leptin signaling to the hypothalamus and also identify cells responding directly to leptin signaling through its cognate receptor.


Assuntos
Jejum/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas/genética , Proteínas Repressoras , Fatores de Transcrição , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Expressão Gênica , Hipotálamo/patologia , Hibridização in Situ Fluorescente/métodos , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Núcleo Mediodorsal do Tálamo/patologia , Neurônios/metabolismo , Neuropeptídeos/genética , Pró-Opiomelanocortina/genética , Biossíntese de Proteínas , Ratos , Ratos Wistar , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina
9.
Diabetes ; 49(2): 244-52, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10868941

RESUMO

Hypothalamic melanocortins are among several neuropeptides strongly implicated in the control of food intake. Agonists for melanocortin 4 (MC-4) receptors such as alpha-melanocyte-stimulating hormone (alpha-MSH), a product of proopiomelanocortin (POMC), reduce food intake, whereas hypothalamic agouti-related protein (AgRP) is a MC-4 receptor antagonist that increases food intake. To investigate whether reduced melanocortin signaling contributes to hyperphagia induced by uncontrolled diabetes, male Sprague-Dawley rats were studied 7 days after administration of streptozotocin (STZ) or vehicle. In addition, we wished to determine the effect of diabetes on muscle uncoupling protein 3 (UCP-3), a potential regulator of muscle energy metabolism. STZ diabetic rats were markedly hyperglycemic (31.3 +/- 1.0 mmol/l; P < 0.005) compared with nondiabetic controls (9.3 +/- 0.2 mmol/l). Insulin treatment partially corrected the hyperglycemia (18.8 +/- 2.5 mol/l; P < 0.005). Plasma leptin was markedly reduced in STZ diabetic rats (0.4 +/- 0.1 ng/ml; P < 0.005) compared with controls (3.0 +/- 0.4 ng/ml), an effect that was also partially reversed by insulin treatment (1.8 +/- 0.3 ng/ml). Untreated diabetic rats were hyperphagic, consuming 40% more food (48 +/- 1 g/day; P < 0.005) than controls (34 +/- 1 g/day). Hyperphagia was prevented by insulin treatment (32 +/- 2 g/day). In untreated diabetic rats, hypothalamic POMC mRNA expression (measured by in situ hybridization) was reduced by 80% (P < 0.005), whereas AgRP mRNA levels were increased by 60% (P < 0.01), suggesting a marked decrease of hypothalamic melanocortin signaling. The change in POMC, but not in AgRP, mRNA levels was partially reversed by insulin treatment. By comparison, the effects of diabetes to increase hypothalamic neuropeptide Y (NPY) expression and to decrease corticotropin-releasing hormone (CRH) expression were normalized by insulin treatment, whereas the expression of mRNA encoding the long form of the leptin receptor in the arcuate nucleus was unaltered by diabetes or insulin treatment. UCP-3 mRNA expression in gastrocnemius muscle from diabetic rats was increased fourfold (P < 0.005), and the increase was prevented by insulin treatment. The effect of uncontrolled diabetes to decrease POMC, while increasing AgRP gene expression, suggests that reduced hypothalamic melanocortin signaling, along with increased NPY and decreased CRH signaling, could contribute to diabetic hyperphagia. These responses, in concert with increased muscle UCP-3 expression, may also contribute to the catabolic effects of uncontrolled diabetes on fuel metabolism in peripheral tissues.


Assuntos
Proteínas de Transporte/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Hipotálamo/metabolismo , Insulina/uso terapêutico , Pró-Opiomelanocortina/metabolismo , Receptores de Superfície Celular , Animais , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Ingestão de Alimentos/efeitos dos fármacos , Hormônios/sangue , Canais Iônicos , Masculino , Proteínas Mitocondriais , Proteínas Musculares/metabolismo , Neuropeptídeo Y/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores para Leptina , Proteína Desacopladora 3
10.
Nature ; 404(6778): 661-71, 2000 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-10766253

RESUMO

New information regarding neuronal circuits that control food intake and their hormonal regulation has extended our understanding of energy homeostasis, the process whereby energy intake is matched to energy expenditure over time. The profound obesity that results in rodents (and in the rare human case as well) from mutation of key signalling molecules involved in this regulatory system highlights its importance to human health. Although each new signalling pathway discovered in the hypothalamus is a potential target for drug development in the treatment of obesity, the growing number of such signalling molecules indicates that food intake is controlled by a highly complex process. To better understand how energy homeostasis can be achieved, we describe a model that delineates the roles of individual hormonal and neuropeptide signalling pathways in the control of food intake and the means by which obesity can arise from inherited or acquired defects in their function.


Assuntos
Sistema Nervoso Central/fisiologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Monoaminas Biogênicas/fisiologia , Homeostase , Humanos , Hipotálamo/fisiologia , Insulina/fisiologia , Leptina/fisiologia , Modelos Biológicos , Neuropeptídeos/fisiologia , Obesidade/tratamento farmacológico , Obesidade/etiologia , Saciação/fisiologia , Transdução de Sinais
12.
Obes Rev ; 1(1): 37-46, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-12119644

RESUMO

In the field of obesity research, two separate lines of study have emerged which explore the mechanism by which food intake is regulated: short-term control of food intake, and the central regulation of energy balance. The former studies the satiety response during consumption of meals, whereby satiety signalling originating in the gut is transduced into a neural signal that modulates satiety pathways in the brainstem. This review describes a neuroanatomically based model in which leptin and insulin signalling in the hypothalamus governs long-term regulation of energy balance via mechanisms that are integrated with satiety hormone signalling in the brainstem. The functional outcome of this integration is a cumulative meal-to-meal regulation of food intake, that over relatively long intervals serves to maintain stable adipose stores. Our model provides a context within which continued investigation of neuroendocrine mechanisms that control food intake and body weight can be explored, and has potential application to our current understanding of clinical obesity and its treatment.


Assuntos
Peso Corporal , Ingestão de Alimentos , Sistemas Neurossecretores/fisiologia , Encéfalo/fisiologia , Tronco Encefálico/fisiologia , Metabolismo Energético , Homeostase , Humanos , Hipotálamo/fisiologia , Insulina/fisiologia , Leptina/fisiologia , Saciação/fisiologia , Transdução de Sinais
13.
Annu Rev Psychol ; 51: 255-77, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10751972

RESUMO

This chapter reviews the recent literature on hormonal and neural signals critical to the regulation of individual meals and body fat. Rather than eating in response to acute energy deficits, animals eat when environmental conditions (social and learned factors, food availability, opportunity, etc.) are optimal. Hence, eating patterns are idiosyncratic. Energy homeostasis, the long-term matching of food intake to energy expenditure, is accomplished via controls over the size of meals. Individuals who have not eaten sufficient food to maintain their normal weight have lower levels of adiposity signals (leptin and insulin) in the blood and brain, and one consequence is that meal-generated signals (such as CCK) are less efficacious at reducing meal size. The converse is true if individuals are above their normal weight, when they tend to eat smaller meals. The final section reviews how these signals are received and integrated by the CNS, as well as the neural circuits and transmitters involved.


Assuntos
Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/psicologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Animais , Humanos , Hipotálamo/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Rede Nervosa/fisiologia , Neuropeptídeos/metabolismo , Obesidade/metabolismo
14.
Brain Res ; 848(1-2): 114-23, 1999 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-10612703

RESUMO

Insulin and leptin are hypothesized to be 'adiposity signals' for the long-term regulation of body weight by the brain. Accordingly, a change in the plasma levels of leptin or insulin indicates a state of altered energy homeostasis and adiposity, and the brain responds by adjusting food intake to restore adipose tissue mass to a regulated level. The candidate site for the brain's detection of leptin adiposity signaling is the hypothalamic arcuate nucleus, where leptin inhibits expression neuropeptide Y and increases expression of the pro-opiomelanocortin (POMC) precursor of alphaMSH. Insulin also inhibits arcuate nucleus expression of neuropeptide Y but its effects on other hypothalamic signaling systems are not known. Leptin-responsive neurons in the arcuate nucleus are hypothesized to project to the paraventricular nucleus and lateral hypothalamic area where they are proposed to influence the expression of peptides that regulate food intake. Future development of this model will incorporate brain pathways for integration of leptin and insulin adiposity signaling to the hypothalamus with meal-related signals that act in the caudal brainstem. Recent research showing that leptin and insulin enhance the satiety action of peripheral CCK, thereby causing meals to be terminated earlier and reducing cumulative food intake, suggests that hypothalamic pathways that are sensitive to leptin and insulin adiposity signals have anatomical connections with caudal brainstem neurons that respond to meal-related signals and regulate meal size. The recent findings that insulin alters the expression and function of neural transporters for dopamine and norepinephrine indicate that adiposity signals may influence food intake by acting on non-peptide neurotransmitter systems.


Assuntos
Tecido Adiposo/fisiologia , Peso Corporal/fisiologia , Encéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Insulina/fisiologia , Leptina/fisiologia , Transdução de Sinais/fisiologia , Animais , Encéfalo/anatomia & histologia , Humanos
15.
Horm Metab Res ; 31(5): 345-50, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10422733

RESUMO

A major paradigm in the field of obesity research is the existence of an adipose tissue-brain endocrine axis for the regulation of body weight. Leptin, the peptide mediator of this axis, is secreted by adipose cells. It lowers food intake and body weight by acting in the hypothalamus, a region expressing an abundance of leptin receptors and a variety of neuropeptides that influence food intake and energy balance. Among the most promising candidates for leptin-sensitive cells in the hypothalamus are arcuate nucleus neurons that co-express the anabolic neuropeptides, neuropeptide Y (NPY) and agouti-related peptide (AGRP), and those that express proopiomelanocortin (POMC), the precursor of the catabolic peptide, alphaMSH. These cell types contain mRNA encoding leptin receptors and show changes in neuropeptide gene expression in response to changes in food intake and circulating leptin levels. Decreased leptin signaling in the arcuate nucleus is hypothesized to increase the expression of NPY and AGRP. Levels of leptin receptor mRNA and leptin binding are increased in the arcuate nucleus during fasting, principally in NPY/AGRP neurons. These findings suggest that changes in leptin receptor expression in the arcuate nucleus are inversely associated with changes in leptin signaling, and that the arcuate nucleus is an important target of leptin action in the brain.


Assuntos
Hipotálamo/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Proteínas/farmacologia , Proteína Relacionada com Agouti , Animais , Núcleo Arqueado do Hipotálamo/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Leptina , Neuropeptídeo Y/fisiologia , Pró-Opiomelanocortina/fisiologia , Proteínas/fisiologia
16.
Horm Metab Res ; 31(5): 351-4, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10422734

RESUMO

Our laboratory has investigated the role of the neuropeptide galanin in the sympathetic neural control of both the canine endocrine pancreas and liver. Galanin mRNA and peptide were found in the neuronal cell bodies of the celiac ganglion, which projects fibers to both organs. Galanin fibers formed dense networks around the islets. Galanin was released from these nerves and the amount released appeared sufficient to markedly inhibit basal insulin secretion. We therefore propose that galanin is a sympathetic neurotransmitter in canine endocrine pancreas. Galanin was also found in hepatic nerves usually co-localized with tyrosine hydroxylase, a sympathetic marker. Further, intraportal administration of the sympathetic neurotoxin, 6-hydroxydopamine, abolished galanin staining in the hepatic parenchyma. We evaluated the role of galanin in mediating the actions of sympathetic nerves to increase hepatic glucose production and decrease hepatic arterial conductance. Local infusion of synthetic galanin had little effect by itself, but it did potentiate the action of norepinephrine to stimulate hepatic glucose production, demonstrating a neuromodulatory action. In contrast, galanin had no effect on hepatic arterial blood flow. We therefore propose that in the liver galanin functions as a neuromodulator of norepinephrine's metabolic action.


Assuntos
Galanina/fisiologia , Ilhotas Pancreáticas/inervação , Fígado/inervação , Neurotransmissores/fisiologia , Animais , Cães
17.
Brain Res ; 828(1-2): 154-8, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10320735

RESUMO

The arcuate nucleus (ARC) mediates the anorexic effects of leptin and expresses the long form (Ob-Rb) of the leptin receptor. To determine whether ARC leptin binding increases when plasma leptin levels are low during fasting, [125I]-leptin specific binding to rat brain slices was measured by quantitative autoradiography. [125I]-leptin specific binding was dense in the ARC and increased 2-fold after a 48-h fast (P<0.001). These findings suggest that leptin receptor binding in the ARC is upregulated during fasting and that fasting changes the sensitivity of the ARC to leptin.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Proteínas de Transporte/metabolismo , Jejum/fisiologia , Proteínas/farmacologia , Receptores de Superfície Celular , Animais , Ingestão de Alimentos/fisiologia , Radioisótopos do Iodo , Leptina , Masculino , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteínas/metabolismo , Ensaio Radioligante , Ratos , Ratos Wistar , Receptores para Leptina
18.
Am J Physiol ; 276(5): R1425-33, 1999 05.
Artigo em Inglês | MEDLINE | ID: mdl-10233036

RESUMO

To investigate whether brain leptin involves neuropeptidergic pathways influencing ingestion, metabolism, and gastrointestinal functioning, leptin (3.5 micrograms) was infused daily into the third cerebral ventricular of rats for 3 days. To distinguish between direct leptin effects and those secondary to leptin-induced anorexia, we studied vehicle-infused rats with food available ad libitum and those that were pair-fed to leptin-treated animals. Although body weight was comparably reduced (-8%) and plasma glycerol was comparably increased (142 and 17%, respectively) in leptin-treated and pair-fed animals relative to controls, increases in plasma fatty acids and ketones were only detected (132 and 234%, respectively) in pair-fed rats. Resting energy expenditure (-15%) and gastrointestinal fill (-50%) were reduced by pair-feeding relative to the ad libitum group, but they were not reduced by leptin treatment. Relative to controls, leptin increased hypothalamic mRNA for corticotropin-releasing hormone (CRH; 61%) and for proopiomelanocortin (POMC; 31%) but did not reduce mRNA for neuropeptide Y. These results suggest that CNS leptin prevents metabolic/gastrointestinal responses to caloric restriction by activating hypothalamic CRH- and POMC-containing pathways and raise the possibility that these peripheral responses to CNS leptin administration contribute to leptin's anorexigenic action.


Assuntos
Sistema Digestório/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Proteínas/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Animais , Nível de Alerta/efeitos dos fármacos , Glicemia , Peso Corporal/fisiologia , Corticosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Sistema Digestório/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Gorduras/metabolismo , Expressão Gênica/fisiologia , Glicogênio/sangue , Asseio Animal/efeitos dos fármacos , Hipotálamo/metabolismo , Hibridização In Situ , Insulina/sangue , Leptina , Masculino , Oxirredução , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Long-Evans , Descanso/fisiologia , Sistema Nervoso Simpático/metabolismo , Triglicerídeos/sangue
19.
Diabetes ; 48(4): 828-33, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10102700

RESUMO

The decline of leptin (Ob protein) concentrations during fasting is implicated as a signal for increasing the expression of the orexigenic peptide neuropeptide Y (NPY) in the hypothalamus. To test the hypothesis that the effects of food intake on arcuate nucleus NPY activation are mediated by leptin, we performed simultaneous triple in situ hybridization colocalization studies to determine whether the subset of NPY neurons that are activated by fasting preferentially expresses the long form of the leptin receptor (Ob-Rb). Thus, mRNAs encoding NPY and pro-opiomelanocortin (POMC) were colocalized in the arcuate nucleus of fed and fasted rats by fluorescence in situ hybridization in combination with isotopic in situ hybridization for Ob-Rb mRNA. In fed animals, 47% of arcuate nucleus neurons containing NPY mRNA also contained Ob-Rb mRNA, compared with 79% of POMC neurons (P < 0.01). After a 2-day fast, the number of arcuate nucleus neurons with NPY mRNA increased 50% (P < 0.05); the number of these that coexpressed Ob-Rb increased twofold (P = 0.013). Furthermore, Ob-Rb mRNA hybridization in individual NPY neurons increased by 64% (P < 0.02). In contrast, the number of POMC neurons that coexpressed Ob-Rb was unchanged. A significant interpretation of these findings is that the NPY neurons that do not express detectable levels of Ob-Rb mRNA are not activated by fasting, whereas the NPY neurons that are activated by fasting are the ones that express Ob-Rb. These data demonstrate a significant physiological difference between NPY neurons that express Ob-Rb and those that do not. The results support the conclusion that the effect of food intake on NPY neurons is mediated by the direct action of leptin via Ob-Rb receptors expressed by these NPY cells. The results also indicate that expression of Ob-Rb is a defining phenotypic characteristic of the subset of arcuate nucleus NPY neurons that are activated by fasting and play a central role in the adaptive response to negative energy balance.


Assuntos
Proteínas de Transporte/genética , Jejum/fisiologia , Hipotálamo/metabolismo , Neurônios/fisiologia , Neuropeptídeo Y/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular , Animais , Proteínas de Transporte/química , Hipotálamo/citologia , Hibridização In Situ , Hibridização in Situ Fluorescente , Isomerismo , Masculino , Neurônios/metabolismo , Pró-Opiomelanocortina/genética , Ratos , Ratos Wistar , Receptores para Leptina
20.
Am J Clin Nutr ; 69(4): 584-96, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10197558

RESUMO

In 1995, we described a new model for adiposity regulation. Since then, data regarding the biology of body weight regulation has accumulated at a remarkable rate and has both modified and strengthened our understanding of this homeostatic system. In this review we integrate new information into a revised model for further understanding this important regulatory process. Our model of energy homeostasis proposes that long-term adiposity-related signals such as insulin and leptin influence the neuronal activity of central effector pathways that serve as controllers of energy balance.


Assuntos
Tecido Adiposo/metabolismo , Metabolismo Energético , Homeostase/fisiologia , Modelos Biológicos , Animais , Estimulantes do Apetite , Peso Corporal , Humanos , Hipotálamo/fisiologia , Neuropeptídeo Y/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA