Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1450, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36922513

RESUMO

Disruption of brain-expressed G protein-coupled receptor-10 (GPR10) causes obesity in animals. Here, we identify multiple rare variants in GPR10 in people with severe obesity and in normal weight controls. These variants impair ligand binding and G protein-dependent signalling in cells. Transgenic mice harbouring a loss of function GPR10 variant found in an individual with obesity, gain excessive weight due to decreased energy expenditure rather than increased food intake. This evidence supports a role for GPR10 in human energy homeostasis. Therapeutic targeting of GPR10 may represent an effective weight-loss strategy.


Assuntos
Obesidade , Receptores Acoplados a Proteínas G , Animais , Humanos , Camundongos , Metabolismo Energético , Camundongos Transgênicos , Obesidade/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Aumento de Peso/genética
2.
Mol Metab ; 55: 101407, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34844019

RESUMO

OBJECTIVE: Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are effective medications to reduce appetite and body weight. These actions are centrally mediated; however, the neuronal substrates involved are poorly understood. METHODS: We employed a combination of neuroanatomical, genetic, and behavioral approaches in the mouse to investigate the involvement of caudal brainstem cholecystokinin-expressing neurons in the effect of the GLP-1RA exendin-4. We further confirmed key neuroanatomical findings in the non-human primate brain. RESULTS: We found that cholecystokinin-expressing neurons in the caudal brainstem are required for the anorectic and body weight-lowering effects of GLP-1RAs and for the induction of GLP-1RA-induced conditioned taste avoidance. We further show that, while cholecystokinin-expressing neurons are not a direct target for glucose-dependent insulinotropic peptide (GIP), GIP receptor activation results in a reduced recruitment of these GLP-1RA-responsive neurons and a selective reduction of conditioned taste avoidance. CONCLUSIONS: In addition to disclosing a neuronal population required for the full appetite- and body weight-lowering effect of GLP-1RAs, our data also provide a novel framework for understanding and ameliorating GLP-1RA-induced nausea - a major factor for withdrawal from treatment.


Assuntos
Colecistocinina/farmacologia , Polipeptídeo Inibidor Gástrico/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Animais , Apetite/efeitos dos fármacos , Depressores do Apetite/farmacologia , Glicemia/efeitos dos fármacos , Exenatida/farmacologia , Feminino , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/fisiologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Liraglutida/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo
3.
PLoS One ; 17(10): e0275604, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36251705

RESUMO

Here we show that central administration of pyroglutamylated arginine-phenylamine-amide peptide (QRFP/26RFa) increases both food intake and locomotor activity, without any significant effect on energy expenditure, thermogenesis or reward. Germline knock out of either of the mouse QRFP receptor orthologs, Gpr103a and Gpr103b, did not produce a metabolic phenotype. However, both receptors are required for the effect of centrally administered QRFP to increase feeding and locomotor activity. As central injection of QRFP activated orexin/hypocretin neurons in the lateral hypothalamus, we compared the action of QRFP and orexin on behaviour. Both peptides increased arousal and locomotor activity. However, while orexin increased consummatory behaviour, QRFP also affected other appetitive behaviours. Furthermore, the feeding but not the locomotor response to QRFP, was blocked by co-administration of an orexin receptor 1 antagonist. These results suggest that QRFP agonism induces both appetitive and consummatory behaviour, but only the latter is dependent on orexin/hypocretin receptor signalling.


Assuntos
Receptores de Orexina , Peptídeos , Receptores Acoplados a Proteínas G , Animais , Camundongos , Amidas , Compostos de Anilina , Arginina , Peptídeos e Proteínas de Sinalização Intercelular , Locomoção , Neuropeptídeos , Receptores de Orexina/metabolismo , Orexinas , Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Comportamento Alimentar
4.
Curr Biol ; 32(21): 4699-4706.e4, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36182699

RESUMO

Loss of appetite and negative energy balance are common features of endotoxemia in all animals and are thought to have protective roles by reducing nutrient availability to host and pathogen metabolism. Accordingly, fasting and caloric restriction have well-established anti-inflammatory properties. However, in response to reduced nutrient availability at the cellular and organ levels, negative energy balance also recruits distinct energy-sensing brain circuits, but it is not known whether these neuronal systems have a role in its anti-inflammatory effects. Here, we report that hypothalamic AgRP neurons-a critical neuronal population for the central representation of negative energy balance-have parallel immunoregulatory functions. We found that when endotoxemia occurs in fasted mice, the activity of AgRP neurons remains sustained, but this activity does not influence feeding behavior and endotoxemic anorexia. Furthermore, we found that endotoxemia acutely desensitizes AgRP neurons, which also become refractory to inhibitory signals. Mimicking this sustained AgRP neuron activity in fed mice by chemogenetic activation-a manipulation known to recapitulate core behavioral features of fasting-results in reduced acute tumor necrosis factor alpha (TNF-α) release during endotoxemia. Mechanistically, we found that endogenous glucocorticoids play an important role: glucocorticoid receptor deletion from AgRP neurons prevents their endotoxemia-induced desensitization, and importantly, it counteracts the fasting-induced suppression of TNF-α release, resulting in prolonged sickness. Together, these findings provide evidence directly linking AgRP neuron activity to the acute response during endotoxemia, suggesting that these neurons are a functional component of the immunoregulatory effects associated with negative energy balance and catabolic metabolism.


Assuntos
Endotoxemia , Fator de Necrose Tumoral alfa , Camundongos , Animais , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Fator de Necrose Tumoral alfa/genética , Endotoxemia/metabolismo , Endotoxemia/patologia , Hipotálamo/metabolismo , Neurônios/fisiologia , Metabolismo Energético
5.
J Neurosci ; 30(21): 7369-76, 2010 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-20505104

RESUMO

Hemopressin is a short, nine amino acid peptide (H-Pro-Val-Asn-Phe-Lys-Leu-Leu-Ser-His-OH) isolated from rat brain that behaves as an inverse agonist at the cannabinoid receptor CB(1), and is shown here to inhibit agonist-induced receptor internalization in a heterologous cell model. Since this peptide occurs naturally in the rodent brain, we determined its effect on appetite, an established central target of cannabinoid signaling. Hemopressin dose-dependently decreases night-time food intake in normal male rats and mice, as well as in obese ob/ob male mice, when administered centrally or systemically, without causing any obvious adverse side effects. The normal, behavioral satiety sequence is maintained in male mice fasted overnight, though refeeding is attenuated. The anorectic effect is absent in CB(1) receptor null mutant male mice, and hemopressin can block CB(1) agonist-induced hyperphagia in male rats, providing strong evidence for antagonism of the CB(1) receptor in vivo. We speculate that hemopressin may act as an endogenous functional antagonist at CB(1) receptors and modulate the activity of appetite pathways in the brain.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Hemoglobinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Análise de Variância , Animais , Comportamento Animal , Benzoxazinas/farmacologia , Células COS , Chlorocebus aethiops , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Cicloexanóis , Relação Dose-Resposta a Droga , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Dronabinol/farmacologia , Vias de Administração de Medicamentos , Ingestão de Alimentos/genética , Privação de Alimentos/fisiologia , Proteínas de Fluorescência Verde/genética , Hiperfagia/induzido quimicamente , Hiperfagia/tratamento farmacológico , Leptina/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfolinas/farmacologia , Naftalenos/farmacologia , Piperidinas/farmacologia , Transporte Proteico/efeitos dos fármacos , Psicotrópicos/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/deficiência , Rimonabanto , Fatores de Tempo , Transfecção/métodos
6.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34265067

RESUMO

The ventromedial nucleus of the hypothalamus (VMH) is a complex brain structure that is integral to many neuroendocrine functions, including glucose regulation, thermogenesis, and appetitive, social, and sexual behaviors. As such, it is of little surprise that the nucleus is under intensive investigation to decipher the mechanisms which underlie these diverse roles. Developments in genetic and investigative tools, for example the targeting of steroidogenic factor-1-expressing neurons, have allowed us to take a closer look at the VMH, its connections, and how it affects competing behaviors. In the current review, we aim to integrate recent findings into the literature and contemplate the conclusions that can be drawn.


Assuntos
Hipotálamo/fisiologia , Neurônios/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Agressão , Animais , Glicemia/metabolismo , Peso Corporal , Ingestão de Alimentos/genética , Metabolismo Energético , Comportamento Alimentar , Feminino , Fluorescência , Glucose/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Neurônios/metabolismo , Comportamento Sexual Animal , Comportamento Social , Fator Esteroidogênico 1/metabolismo , Termogênese
7.
J Neuroendocrinol ; 33(11): e13035, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34495565

RESUMO

There has been a long history of research on the effects of oxytocin on feeding behaviour. The classic-held view is that the neurohormone is anorexigenic at least in rodents, although the data for humans are not so clear cut. Likewise, a physiological role for oxytocin is disputed. Thus, although pharmacological, anatomical and physiological data suggest oxytocin may have a function in satiety signalling, this view is not supported by the latest research using the genetic recording and manipulation of oxytocin neurones. Here, we avoid a discussion of the pharmacological effects of oxytocin and examine evidence, from both sides of the argument, concerning whether the endogenous oxytocin system has a role in the regulation of normal feeding.


Assuntos
Comportamento Alimentar , Neurônios , Ocitocina , Animais , Comportamento Alimentar/fisiologia , Humanos , Neurônios/fisiologia , Ocitocina/fisiologia , Roedores , Saciação
8.
Br J Pharmacol ; 178(10): 2096-2110, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32627171

RESUMO

Given the high-energy requirements to sustain immune responses and healing processes, it is intriguing that lack of appetite (i.e., anorexia) is a cardinal feature of sickness behaviour. While our understanding of the brain mechanisms that control appetite is rapidly growing, how inflammation affects these mechanisms is not fully understood. Here, we discuss advances in our understanding of discrete appetite controlling mechanisms and how inflammation influences their function. We further discuss the pathophysiological significance of anorexia and negative energy balance during the immune regulatory response. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.


Assuntos
Apetite , Inflamação , Encéfalo , Humanos , Imunidade
9.
J Neurosci ; 29(47): 14828-35, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19940178

RESUMO

The adipose-derived hormone, leptin, was discovered over 10 years ago, but only now are we unmasking its downstream pathways which lead to reduced energy intake (feeding) and increased energy expenditure (thermogenesis). Recent transgenic models have challenged the long-standing supposition that the hypothalamic arcuate nucleus (Arc) is omnipotent in the central response to leptin, and research focus is beginning to shift to examine roles of extra-arcuate sites. Dhillon et al. (2006) demonstrated that targeted knock out of the signaling form of the leptin receptor (lepr-B) in steroidogenic factor 1 (SF-1) cells of the hypothalamic ventromedial nucleus (VMN) produces obesity of a similar magnitude to the pro-opiomelanocortin (POMC)-driven lepr-B deleted mouse, via a functionally distinct mechanism. These findings reveal that SF-1 cells of the VMN could be equally as important as POMC cells in mediating leptin's anti-obesity effects. However, the identification of molecular and cellular correlates of this relationship remains tantalizingly unknown. Here, we have shown that mRNA expression of the VMN-expressed neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is regulated according to energy status and that it exerts catabolic effects when administered centrally to mice. Furthermore, we have shown that SF-1 and PACAP mRNAs are colocalized in the VMN, and that leptin signaling via lepr-B is required for normal PACAP expression in these cells. Finally, blocking endogenous central PACAP signaling with the antagonist PACAP(6-38) markedly attenuates leptin-induced hypophagia and hyperthermia in vivo. Thus, it appears that PACAP is an important mediator of central leptin effects on energy balance.


Assuntos
Leptina/metabolismo , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Fator Esteroidogênico 1/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Regulação do Apetite/fisiologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Febre/tratamento farmacológico , Febre/metabolismo , Febre/fisiopatologia , Leptina/farmacologia , Masculino , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Receptores para Leptina/efeitos dos fármacos , Receptores para Leptina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator Esteroidogênico 1/genética , Núcleo Hipotalâmico Ventromedial/citologia
10.
J Neurochem ; 113(5): 1123-32, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20236391

RESUMO

The glucose analogue, 2-deoxy-D-glucose (2-DG) is an inhibitor of glycolysis and, when administered systemically or centrally, induces glucoprivation leading to counter-regulatory responses, including increased feeding behaviour. Investigations into how the brain responds to glucoprivation could have important therapeutic potential, as disruptions or defects in the defence of the brain's 'glucostatic' circuitry may be partly responsible for pathological conditions resulting from diabetes and obesity. To define the 'glucostat' brain circuitry further we have combined blood-oxygen-level-dependent pharmacological-challenge magnetic resonance imaging (phMRI) with whole-brain c-Fos functional activity mapping to characterise brain regions responsive to an orexigenic dose of 2-DG [200 mg/kg; subcutaneous (s.c.)]. For phMRI, rats were imaged using a T(2)*-weighted gradient echo in a 7T magnet for 60 min under alpha-chloralose anaesthesia, whereas animals for immunohistochemistry were unanaesthetised and freely behaving. These complementary methods demonstrated functional brain activity in a number of previously characterised glucose-sensing brain regions such as those in the hypothalamus and brainstem following administration of 2-DG compared with vehicle. As the study mapped whole-brain functional responses, it also identified the orbitofrontal cortex and striatum (nucleus accumbens and ventral pallidum) as novel 2-DG-responsive brain regions. These regions make up a corticostriatal connection with the hypothalamus, by which aspects of motivation, salience and reward can impinge on the hypothalamic control of feeding behaviour. This study, therefore, provides further evidence for a common integrated circuit involved in the induction of feeding behaviour, and illustrates the valuable potential of phMRI in investigating central pharmacological actions.


Assuntos
Antimetabólitos/farmacologia , Encéfalo/fisiologia , Desoxiglucose/farmacologia , Glucose/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Gasometria , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Glucose/antagonistas & inibidores , Hipotálamo/anatomia & histologia , Hipotálamo/fisiologia , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Vias Neurais/anatomia & histologia , Vias Neurais/efeitos dos fármacos , Oxigênio/sangue , Ratos , Ratos Sprague-Dawley
11.
Trends Endocrinol Metab ; 31(9): 691-703, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32563715

RESUMO

Impaired awareness of hypoglycaemia (IAH) affects around a quarter of patients with diabetes who receive insulin treatment. This condition is characterised by a progressive reduction in symptomatic and behavioural responses to hypoglycaemia, increasing risk of deeper drops in blood glucose, unconsciousness, and collapse. Thus, patients with IAH experience severe hypoglycaemic episodes more frequently, resulting in significant morbidity and mortality. IAH is thought to develop as a consequence of whole-body adaptations to repeated insulin-induced hypoglycaemia (RH), with widespread deficits in the hypoglycaemia counter-regulatory response (CRR). Despite this important insight, the precise pathophysiology by which RH leads to an attenuated CRR is unknown. Studies into the underlying mechanisms of IAH have employed a variety of protocols in humans and experimental species. The use of animal models has many investigational benefits, including the unprecedented increase in the availability of transgenic strains. However, modelling impaired hypoglycaemia-associated counter-regulation remains challenging and appropriate interpretation of findings across species and protocols even more so. Here, we review the experimental modelling of IAH and impaired hypoglycaemia-associated counter-regulation, with a focus on understanding species-specific variation in glucose homeostasis. This review will aid investigators in interpreting outputs from different studies in IAH and aid progress in the field.


Assuntos
Hipoglicemia/tratamento farmacológico , Animais , Glicemia/efeitos dos fármacos , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico
12.
Elife ; 92020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32723474

RESUMO

The cytokine, GDF15, is produced in pathological states which cause cellular stress, including cancer. When over expressed, it causes dramatic weight reduction, suggesting a role in disease-related anorexia. Here, we demonstrate that the GDF15 receptor, GFRAL, is located in a subset of cholecystokinin neurons which span the area postrema and the nucleus of the tractus solitarius of the mouse. GDF15 activates GFRALAP/NTS neurons and supports conditioned taste and place aversions, while the anorexia it causes can be blocked by a monoclonal antibody directed at GFRAL or by disrupting CCK neuronal signalling. The cancer-therapeutic drug, cisplatin, induces the release of GDF15 and activates GFRALAP/NTS neurons, as well as causing significant reductions in food intake and body weight in mice. These metabolic effects of cisplatin are abolished by pre-treatment with the GFRAL monoclonal antibody. Our results suggest that GFRAL neutralising antibodies or antagonists may provide a co-treatment opportunity for patients undergoing chemotherapy.


Assuntos
Anorexia/genética , Tronco Encefálico/fisiologia , Fator 15 de Diferenciação de Crescimento/genética , Neurônios/fisiologia , Pica/genética , Transdução de Sinais , Animais , Colecistocinina/metabolismo , Fator 15 de Diferenciação de Crescimento/administração & dosagem , Fator 15 de Diferenciação de Crescimento/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem
13.
Am J Physiol Endocrinol Metab ; 297(2): E545-51, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19531638

RESUMO

Neuromedin U (NMU) is known to have potent actions on appetite and energy expenditure. Deletion of the NMU gene in mice leads to an obese phenotype, characterized by hyperphagia and decreased energy expenditure. Conversely, transgenic mice that overexpress proNMU exhibit reduced body weight and fat storage. Here, we show that central administration of NMU or the related peptide neuromedin S (NMS) dose-dependently decreases food intake, increases metabolic rate, and leads to significant weight loss in mice. The effects of NMU and NMS on both feeding and metabolism are almost completely lost in mice lacking the putative CNS receptor for NMU and NMS, NMUr2. However, NMUr2 knockout mice do not exhibit overt differences in body weight or energy expenditure compared with wild-type mice, suggesting that the dramatic phenotype of the NMU gene knockout mouse is not due simply to the loss of NMU/NMUr2 signaling. Putative proteolytic cleavage sites indicate that an additional peptide is produced from the NMU precursor protein, which is extremely well conserved between human, mouse, and rat. Here, we demonstrate that this peptide, proNMU(104-136), has a pronounced effect on energy balance in mice. Specifically, central administration of proNMU(104-136) causes a significant but transient ( approximately 4 h) increase in feeding, yet both food intake and body weight are decreased over the following 24 h. proNMU(104-136) administration also significantly increased metabolic rate. These results suggest that proNMU(104-136) is a novel modulator of energy balance and may contribute to the phenotype exhibited by NMU knockout mice.


Assuntos
Regulação do Apetite/efeitos dos fármacos , Neuropeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Sequência de Aminoácidos , Animais , Anorexia/induzido quimicamente , Metabolismo Energético/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neuropeptídeos/química , Neuropeptídeos/genética , Fragmentos de Peptídeos/química , Precursores de Proteínas/química , Receptores de Neurotransmissores/genética , Homologia de Sequência de Aminoácidos
14.
Endocrinology ; 160(11): 2737-2747, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31074796

RESUMO

To study the effects of an analog of the gut-produced hormone peptide YY (PYY3-36), which has increased selectivity for the Y2 receptor; specifically, to record its effects on food intake and on hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neuron activity. NNC0165-1273, a modified form of the peptide hormone PYY3-36 with potent selectivity at Y2 receptor (>5000-fold over Y1, 1250-fold over Y4, and 650-fold over Y5 receptor), was tested in vivo and in vitro in mouse models. NNC0165-1273 has fivefold lower relative affinity for Y2 compared with PYY3-36, but >250-, 192-, and 400-fold higher selectivity, respectively, for the Y1, Y4, and Y5 receptors. NNC0165-1273 produced a reduction in nighttime feeding at a dose at which PYY3-36 loses efficacy. The normal behavioral satiety sequence observed suggests that NNC0165-1273 is not nauseating and, instead, reduces food intake by producing early satiety. Additionally, NNC0165-1273 blocked ghrelin-induced cFos expression in NPY/AgRP neurons. In vitro electrophysiological recordings showed that, opposite to ghrelin, NNC0165-1273 hyperpolarized NPY/AgRP neurons and reduced action potential frequency. Administration of NNC0165-1273 via subcutaneous osmotic minipump caused a dose-dependent decrease in body weight and fat mass in an obese mouse model. Finally, NNC0165-1273 attenuated the feeding response when NPY/AgRP neurons were activated using ghrelin or more selectively with designer receptors. NNC0165-1273 is nonnauseating and stimulates a satiety response through, at least in part, a direct action on hypothalamic NPY/AgRP neurons. Modification of PYY3-36 to produce compounds with increased affinity to Y2 receptors may be useful as antiobesity therapies in humans.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Fragmentos de Peptídeos/química , Peptídeo YY/química , Receptores de Neuropeptídeo Y/agonistas , Resposta de Saciedade/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/citologia , Grelina , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia
15.
Endocrinology ; 149(3): 1004-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18063679

RESUMO

Kisspeptins are extraordinarily potent in stimulating gonadotropic hormone secretion via an action on the hypothalamic GnRH neural system. Because the physiological frequency of the GnRH pulse generator is a critical component of the control system that governs reproductive processes, the aim of this study was to examine the effect of kisspeptin-10 on pulsatile LH secretion and on the electrophysiological manifestation of GnRH pulse generator activity to determine frequency modulatory effects. Adult Sprague Dawley rats were ovariectomized and chronically implanted with electrodes in the arcuate nucleus to record the characteristic increases in hypothalamic multiunit electrical activity volleys coincident with the initiation of each LH pulse measured in peripheral blood and/or indwelling cardiac catheters for the collection of blood samples (25 microl) every 5 min for 6-7 h for the measurement of LH. Intravenous infusion of kisspeptin-10 (7.5, 35, and 100 nmol) induced a dose-dependent increase in LH secretion. The stimulatory effect of kisspeptin-10 (100 nmol) on LH secretion was blocked by the GnRH antagonist cetrorelix, precluding a singular action on gonadotropes. Unexpectedly, however, the marked increase in LH release in response to kisspeptin-10 (100 nmol) administration was not accompanied by any change in multiunit electrical activity volley frequency. It seem unlikely, therefore, that kisspeptin-10 has an appreciable frequency modulatory effect on GnRH pulse generator activity in the female rat.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormônio Luteinizante/sangue , Oligopeptídeos/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Relação Dose-Resposta a Droga , Eletrofisiologia , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/farmacologia , Antagonistas de Hormônios/farmacologia , Hipotálamo/efeitos dos fármacos , Infusões Intravenosas , Kisspeptinas , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley
16.
Eur J Neurosci ; 27(2): 457-65, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18190518

RESUMO

In this study, pharmacological-challenge magnetic resonance imaging was used to further characterize the central action of serotonin on feeding. In both feeding and pharmacological-challenge magnetic resonance imaging experiments, we combined 5-HT(1B/2C) agonist m-chlorophenylpiperazine (mCPP) challenge with pre-treatment with the selective 5-HT(1B) and 5-HT(2C) receptor antagonists, SB 224289 (2.5 mg/kg) and SB 242084 (2 mg/kg), respectively. Subcutaneous injection of mCPP (3 mg/kg) completely blocked fast-induced refeeding in freely behaving, non-anaesthetized male rats, an effect that was not modified by the 5-HT(1B) receptor antagonist but was partially reversed by the 5-HT(2C) receptor antagonist. mCPP alone induced both positive and negative blood oxygen level-dependent (BOLD) responses in the brains of anaesthetized rats, including in the limbic system and basal ganglia. Overall, the 5-HT(2C) antagonist SB 242084 reversed the effects elicited by mCPP, whereas the 5-HT(1B) antagonist SB 224289 had virtually no impact. SB 242084 eliminated BOLD signal in nuclei associated with the limbic system and diminished activation in basal ganglia. In addition, BOLD signal was returned to baseline levels in the cortical regions and cerebellum. These results suggest that mCPP may reduce food intake by acting specifically on brain circuits that are modulated by 5-HT(2C) receptors in the rat.


Assuntos
Encéfalo/metabolismo , Comportamento Alimentar/fisiologia , Imageamento por Ressonância Magnética/métodos , Consumo de Oxigênio/fisiologia , Antagonistas do Receptor 5-HT2 de Serotonina , Antagonistas da Serotonina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptor 5-HT2C de Serotonina/fisiologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-30425681

RESUMO

Background: Glucose-sensing neurons are located in several parts of the brain, but are concentrated in the ventromedial nucleus of the hypothalamus (VMH). The importance of these VMH neurons in glucose homeostasis is well-established, however, little is known about their individual identity. In the present study, we identified a distinct glucose-sensing population in the VMH and explored its place in the glucose-regulatory network. Methods: Using patch-clamp electrophysiology on Pacap-cre::EYFP cells, we explored the glucose-sensing ability of the pituitary adenylate cyclase-activating peptide (PACAP) neurons both inside and outside the VMH. We also mapped the efferent projections of these neurons using anterograde and retrograde tracing techniques. Finally, to test the functionality of PACAPVMH in vivo, we used DREADD technology and measured systemic responses. Results: We demonstrate that PACAP neurons inside (PACAPVMH), but not outside the VMH are intrinsically glucose inhibited (GI). Anatomical tracing techniques show that PACAPVMH neurons project to several areas that can influence autonomic output. In vivo, chemogenetic stimulation of these neurons inhibits insulin secretion leading to reduced glucose tolerance, implicating their role in systemic glucose regulation. Conclusion: These findings are important as they identify, for the first time, a specific VMH neuronal population involved in glucose homeostasis. Identifying the different glucose-sensing populations in the VMH will help piece together the different arms of glucose regulation providing vital information regarding central responses to glucose metabolic disorders including hypoglycaemia.

18.
J Endocrinol ; 192(1): 3-15, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17210738

RESUMO

In the three decades since FMRFamide was isolated from the clam Macrocallista nimbosa, the list of RFamide peptides has been steadily growing. These peptides occur widely across the animal kingdom, including five groups of RFamide peptides identified in mammals. Although there is tremendous diversity in structure and biological activity in the RFamides, the involvement of these peptides in the regulation of energy balance and feeding behaviour appears consistently through evolution. Even so, questions remain as to whether feeding-related actions represent a primary function of the RFamides, especially within mammals. However, as we will discuss here, the study of RFamide function is rapidly expanding and with it so is our understanding of how these peptides can influence food intake directly as well as related aspects of feeding behaviour and energy expenditure.


Assuntos
Sistema Nervoso Central/fisiologia , Ingestão de Alimentos , Comportamento Alimentar , Mamíferos/fisiologia , Neuropeptídeos/fisiologia , Analgésicos Opioides/metabolismo , Animais , Metabolismo Energético , Humanos , Invertebrados/fisiologia
19.
Physiol Behav ; 90(1): 180-5, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17078977

RESUMO

Ghrelin, the endogenous ligand for the growth hormone secretagogue (GHS) receptor, stimulates feeding and increases body weight. Systemic ghrelin administration induces the immediate-early gene protein product, c-Fos, in the arcuate nucleus of the hypothalamus (ARC) of satiated rats and this increase is potentiated in fasted rats. The aim of this study was to determine whether potentiation was seen in fasted animals after intracerebroventricular (i.c.v) administration of ghrelin and to identify the hypothalamic nuclei activated by this peptide. In addition we investigated if allowing fasted animals to re-feed for 1 h prior to i.c.v. ghrelin injection affected the c-Fos response. Using c-Fos immunocytochemistry, we demonstrated that i.c.v. ghrelin activated several hypothalamic nuclei, including the ARC, paraventricular nucleus (PVH) and the lateral hypothalamus (LH). The c-Fos response was greater in fasted animals compared with satiated animals. Fasted rats allowed access to food for 1 h prior to central ghrelin administration showed an attenuated response in the ARC, similar to the response seen in fed animals. However, the response in the LH (including in the orexin neurons) was further potentiated. The latter may reflect a connection between the hypothalamus and regions of the brain responding to the reward value of the meal.


Assuntos
Jejum/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Estado Nutricional/fisiologia , Hormônios Peptídicos/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Grelina , Hipotálamo/citologia , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Hormônios Peptídicos/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Saciação/fisiologia
20.
Eur Neuropsychopharmacol ; 27(8): 809-815, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28647450

RESUMO

Feelings of hunger carry a negative-valence (emotion) signal that appears to be conveyed through agouti-related peptide (AgRP) neurons in the hypothalamic arcuate nucleus. The circulating hunger hormone, ghrelin, activates these neurons although it remains unclear whether it also carries a negative-valence signal. Given that ghrelin also activates pathways in the midbrain that are important for reward, it remains possible that ghrelin could act as a positive reinforcer and hence, carry a positive-valence signal. Here we used condition preference/avoidance tests to explore the reinforcing/aversive properties of ghrelin, delivered by intracerebroventricular (ICV) injection (2µg/injection once a day for 4 days). We found that ICV ghrelin produces conditioned avoidance, both in a conditioned place preference/avoidance test (CPP/CPA, in which the animals avoid a chamber previously paired to ghrelin injection) and in a conditioned flavor preference/avoidance test (CFP/CFA, in which the animals consume/avoid a taste previously paired to ghrelin injection). These effects of ghrelin to induce a CPA were observed when conditioning to ghrelin occurred in the absence or presence of food. We did not find evidence, however, that brain ghrelin delivery to rats induces malaise (in the pica test). Our data indicate that ICV ghrelin carries a negative-valence signal consistent with its role as a circulating hunger hormone and with its effects to activate AgRP neurones.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Grelina/farmacologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA