Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Psychophysiology ; 61(2): e14443, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37737514

RESUMO

The gut hormone ghrelin drives food motivation and increases food intake, but it is also involved in the anticipation of and response to rewards other than food. This pre-registered study investigated how naturally varying ghrelin concentrations affect the processing of touch as a social reward in humans. Sixty-seven volunteers received slow caressing touch (so-called CT-targeted touch) as a social reward and control touch on their shins during 3T functional imaging on two test days. On one occasion, participants were fasted, and on another, they received a meal. On each occasion, plasma ghrelin was measured at three time points. All touch was rated as more pleasant after the meal, but there was no association between ghrelin concentrations and pleasantness. CT-targeted touch was rated as the most pleasant and activated somatosensory and reward networks (whole brain). A region-of-interest in the right medial orbitofrontal cortex (mOFC) showed lower activation during all touches, the higher the ghrelin concentrations were. During CT-targeted touch, a larger satiety response (ghrelin decrease after the meal) was associated with higher mOFC activation, and this mOFC activation was associated with higher experienced pleasantness. Overall, higher ghrelin concentrations appear to be related to a lower reward value for touch. Ghrelin may reduce the value of social stimuli, such as touch, to promote food search and intake in a state of low energy. This suggests that the role of ghrelin goes beyond assigning value to food reward.


Assuntos
Percepção do Tato , Tato , Humanos , Tato/fisiologia , Grelina , Percepção do Tato/fisiologia , Encéfalo/diagnóstico por imagem , Recompensa
2.
Obes Rev ; 19(4): 435-451, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29363272

RESUMO

Western diets, with high consumption of simple sugars and saturated fats, contribute to the rise in the prevalence of obesity. It now seems clear that high-fat diets cause obesity, at least in part, by modifying the composition and function of the microorganisms that colonize in the gastrointestinal tract, the microbiota. The exact pathways by which intestinal microbiota contribute to obesity remain largely unknown. High-fat diet-induced alterations in intestinal microbiota have been suggested to increase energy extraction, intestinal permeability and systemic inflammation while decreasing the capability to generate obesity-suppressing short-chain fatty acids. Moreover, by increasing systemic inflammation, microglial activation and affecting vagal nerve activity, 'obese microbiota' indirectly influence hypothalamic gene expression and promote overeating. Because the potential of intestinal microbiota to induce obesity has been recognized, multiple ways to modify its composition and function are being investigated to provide novel preventive and therapeutic strategies against diet-induced obesity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Disbiose/fisiopatologia , Microbioma Gastrointestinal/imunologia , Hipotálamo/fisiopatologia , Inflamação/microbiologia , Obesidade/microbiologia , Disbiose/etiologia , Disbiose/imunologia , Ingestão de Energia , Ácidos Graxos Voláteis/metabolismo , Expressão Gênica , Humanos , Inflamação/etiologia , Inflamação/fisiopatologia , Microglia/metabolismo , Obesidade/complicações , Obesidade/fisiopatologia , Prebióticos/administração & dosagem , Probióticos/administração & dosagem , Nervo Vago/metabolismo
3.
J Neuroendocrinol ; 29(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28219000

RESUMO

Ghrelin, an orexigenic hormone released from the empty stomach, provides a gut-brain signal that promotes many appetitive behaviours, including anticipatory and goal-directed behaviours for palatable treats high in sugar and/or fat. In the present study, we aimed to determine whether ghrelin is able to influence and/or may even have a role in binge-like eating behaviour in rodents. Accordingly, we used a palatable scheduled feeding (PSF) paradigm in which ad lib. chow-fed rodents are trained to 'binge' on a high-fat diet (HFD) offered each day for a limited period of 2 hours. After 2 weeks of habituation to this paradigm, on the test day and immediately prior to the 2-hour PSF, rats were administered ghrelin or vehicle solution by the i.c.v. route. Remarkably and unexpectedly, during the palatable scheduled feed, when rats normally only binge on the HFD, those injected with i.c.v. ghrelin started to eat more chow and chow intake remained above baseline for the rest of the 24-hour day. We identify the ventral tegmental area (VTA) (a key brain area involved in food reward) as a substrate involved because these effects could be reproduced, in part, by intra-VTA delivery of ghrelin. Fasting, which increases endogenous ghrelin, immediately prior to a palatable schedule feed also increased chow intake during/after the schedule feed but, in contrast to ghrelin injection, did not reduce HFD intake. Chronic continuous central ghrelin infusion over several weeks enhanced binge-like behaviour in palatable schedule fed rats. Over a 4-week period, GHS-R1A-KO mice were able to adapt and maintain large meals of HFD in a manner similar to wild-type mice, suggesting that ghrelin signalling may not have a critical role in the acquisition or maintenance in this kind of feeding behaviour. In conclusion, ghrelin appears to act as a modulating factor for binge-like eating behaviour by shifting food preference towards a more nutritious choice (from HFD to chow), with these effects being somewhat divergent from fasting.


Assuntos
Bulimia/fisiopatologia , Dieta Hiperlipídica , Preferências Alimentares , Grelina/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Comportamento de Escolha , Ingestão de Alimentos , Jejum , Grelina/administração & dosagem , Masculino , Camundongos , Camundongos Knockout , Ratos Sprague-Dawley , Receptores de Grelina/genética , Receptores de Grelina/fisiologia
4.
Mol Metab ; 4(6): 437-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26042199

RESUMO

BACKGROUND: The gastrointestinal peptide hormone ghrelin was discovered in 1999 as the endogenous ligand of the growth hormone secretagogue receptor. Increasing evidence supports more complicated and nuanced roles for the hormone, which go beyond the regulation of systemic energy metabolism. SCOPE OF REVIEW: In this review, we discuss the diverse biological functions of ghrelin, the regulation of its secretion, and address questions that still remain 15 years after its discovery. MAJOR CONCLUSIONS: In recent years, ghrelin has been found to have a plethora of central and peripheral actions in distinct areas including learning and memory, gut motility and gastric acid secretion, sleep/wake rhythm, reward seeking behavior, taste sensation and glucose metabolism.

5.
J Neuroendocrinol ; 27(6): 424-34, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25377898

RESUMO

'Hunger is the best spice' is an old and wise saying that acknowledges the fact that almost any food tastes better when we are hungry. The neurobiological underpinnings of this lore include activation of the brain's reward system and the stimulation of this system by the hunger-promoting hormone ghrelin. Ghrelin is produced largely from the stomach and levels are higher preprandially. The ghrelin receptor is expressed in many brain areas important for feeding control, including not only the hypothalamic nuclei involved in energy balance regulation, but also reward-linked areas such as the ventral tegmental area. By targeting the mesoaccumbal dopamine neurones of the ventral tegmental area, ghrelin recruits pathways important for food reward-related behaviours that show overlap with but are also distinct from those important for food intake. We review a variety of studies that support the notion that ghrelin signalling at the level of the mesolimbic system is one of the key molecular substrates that provides a physiological signal connecting gut and reward pathways.


Assuntos
Trato Gastrointestinal/fisiologia , Grelina/metabolismo , Sistema Límbico/fisiologia , Recompensa , Transdução de Sinais , Humanos
6.
Somnologie (Berl) ; 17(2): 111-114, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23807868

RESUMO

BACKGROUND: We hypothesized that acutely sleep-deprived participants would rate ascending concentrations of sucrose as more intense and pleasant, than they would do after one night of normal sleep. Such a finding would offer a potential mechanism through which acute sleep loss could promote overeating in humans. METHOD: A total of 16 healthy normal-weight men participated in 2 conditions: sleep (permitted between 22:30 and 06:30 h) and total sleep deprivation (TSD) respectively. On the morning after regular sleep and TSD, circulating concentrations of ghrelin and glucose were measured. In addition, participants hunger level was assessed by means of visual analogue scales, both before and after a caloric preload. Finally, following the preload, participants rated both intensity and pleasantness of six orally presented yogurt probes with varying sucrose concentrations (2-29 %). RESULTS: Feelings of hunger were significantly more intense under both fasted and sated conditions when subjects were sleep-deprived. In contrast, the change in hunger induced by the preload was similar between the sleep and TSD conditions. Plasma concentrations of ghrelin were significantly higher under conditions of TSD, whereas plasma glucose did not differ between the conditions. No effects were found either on sweet taste intensity or on pleasantness after TSD. CONCLUSION: One night of TSD increases morning plasma concentrations of the hunger-promoting hormone ghrelin in healthy young men. In contrast, sweet taste perception was not affected by nocturnal wakefulness. This suggests that an altered sweet taste perception is an unlikely mechanism by which TSD enhances food intake.

7.
Obesity (Silver Spring) ; 21(8): 1548-53, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23585292

RESUMO

OBJECTIVE: Cognitive factors and anticipation are known to influence food intake. The current study examined the effect of anticipation and actual consumption of food on hormone (ghrelin, cortisol, and insulin) and glucose levels, appetite and ad libitum intake, to assess whether changes in hormone levels might explain the predicted differences in subsequent food intake. DESIGN AND METHODS: During four breakfast sessions, participants consumed a yogurt preload that was either low caloric (LC: 180 kcal/300 g) or high caloric (HC: 530 kcal/300 g) and was provided with either consistent or inconsistent calorie information (i.e., stating the caloric content of the preload was low or high). Appetite ratings and hormone and glucose levels were measured at baseline (t = 0), after providing the calorie information about the preload (t = 20), after consumption of the preload (t = 40), and just before ad libitum intake (t = 60). RESULTS: Ad libitum intake was lower after HC preloads (as compared to LC preloads; P < 0.01). Intake after LC preloads was higher when provided with (consistent) LC information (467±254 kcal) as compared to (inconsistent) HC information (346±210 kcal), but intake after the HC preloads did not depend on the information provided (LC information: 290±178 kcal, HC information: 333±179 kcal; caloric load*information P = 0.03). Hormone levels did not respond in an anticipatory manner, and the post-prandial responses depended on actual calories consumed. CONCLUSIONS: These results suggest that both cognitive and physiological information determine food intake. When actual caloric intake was sufficient to produce physiological satiety, cognitive factors played no role; however, when physiological satiety was limited, cognitively induced satiety reduced intake to comparable levels.


Assuntos
Ingestão de Alimentos/fisiologia , Ingestão de Energia/fisiologia , Apetite/fisiologia , Glicemia/metabolismo , Estudos Cross-Over , Feminino , Grelina/sangue , Humanos , Fome/fisiologia , Hidrocortisona/sangue , Insulina/sangue , Saciação/fisiologia , Paladar , Iogurte , Adulto Jovem
8.
Neuroscience ; 180: 201-11, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21303683

RESUMO

The orexigenic and pro-obesity hormone ghrelin targets key hypothalamic and mesolimbic circuits involved in energy balance, appetite and reward. Given that such circuits are closely integrated with those regulating mood and cognition, we sought to determine whether chronic (>2 weeks) CNS exposure to ghrelin alters anxiety- and depression-like behaviour in rats as well as some physiological correlates. Rats bearing chronically implanted i.c.v. catheters were treated with ghrelin (10 µg/d) or vehicle for 4 weeks. Tests used to assess anxiety- and depression-like behaviour were undertaken during weeks 3-4 of the infusion. These revealed an increase in anxiety- and depression-like behaviour in the ghrelin-treated rats relative to controls. At the end of the 4-week infusion, brains were removed and the amygdala dissected for subsequent qPCR analysis that revealed changes in expression of a number of genes representing key systems implicated in these behavioural changes. Finally, given the key role of the dorsal raphe serotonin system in emotional reactivity, we examined the electrophysiological response of dorsal raphe neurons after a ghrelin challenge, and found mainly inhibitory responses in this region. We demonstrate that the central ghrelin signalling system is involved in emotional reactivity in rats, eliciting pro-anxiety and pro-depression effects and have begun to explore novel target systems for ghrelin that may be of importance for these effects.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/fisiologia , Emoções/fisiologia , Expressão Gênica/fisiologia , Grelina/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Eletrofisiologia , Emoções/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Grelina/administração & dosagem , Injeções Intraventriculares , Masculino , Aprendizagem em Labirinto/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Neuroscience ; 180: 129-37, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21335062

RESUMO

Ghrelin, a circulating orexigenic stomach-derived hormone, has recently been implicated in extra-homeostatic feeding, increasing food reward and food-motivated behavior. The precise target site(s) for ghrelin's effects on food reward have yet to be elucidated. The neurocircuitry underpinning food-motivated behavior involves, in particular, the dopamine cells of the ventral tegmental area (VTA) that project to the nucleus accumbens (NAcc). Ghrelin stimulation in both of these mesolimbic reward areas increases chow intake. Here we sought to determine if ghrelin acts directly within these mesolimbic reward areas to increase food reward/motivation in studies that combine feeding behavior, pharmacology, and neuroanatomy. We found that motivated behavior for a sucrose reward, assessed in an operant conditioning paradigm in rats, was increased when ghrelin was microinjected directly into the VTA but not into the NAcc. By contrast, ghrelin administration to both areas increased the free feeding of chow. Importantly, in a state of overnight food restriction, where endogenous levels of ghrelin are increased, ghrelin receptor (GHS-R1A) blockade in the VTA was sufficient to decrease the motivation to work for a sugar reward. Blockade of the GHS-R1A in VTA or NAcc was not sufficient to reduce fasting-induced chow hyperphagia. Taken together our data identify the VTA but not the NAcc as a direct, necessary, and sufficient target site for ghrelin's action on food motivation.


Assuntos
Comportamento Alimentar/fisiologia , Grelina/metabolismo , Motivação/fisiologia , Área Tegmentar Ventral/metabolismo , Animais , Condicionamento Operante , Alimentos , Grelina/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Área Tegmentar Ventral/efeitos dos fármacos
10.
Neuroscience ; 171(4): 1180-6, 2010 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20933579

RESUMO

Here we sought to determine whether ghrelin's central effects on food intake can be interrupted by nicotine acetylcholine receptor (nAChR) blockade. Ghrelin regulates mesolimbic dopamine neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens, partly via cholinergic VTA afferents originating in the laterodorsal tegmental area (LDTg). Given that these cholinergic projections to the VTA have been implicated in natural as well as drug-induced reinforcement, we sought to investigate the role of cholinergic signaling in ghrelin-induced food intake as well as fasting-induced food intake, for which endogenous ghrelin has been implicated. We found that i.p. treatment with the non-selective centrally active nAChR antagonist, mecamylamine decreased fasting-induced food intake in both mice and rats. Moreover, central administration of mecamylamine decreased fasting-induced food intake in rats. I.c.v. ghrelin-induced food intake was suppressed by mecamylamine i.p. but not by hexamethonium i.p., a peripheral nAChR antagonist. Furthermore, mecamylamine i.p. blocked food intake following ghrelin injection into the VTA. Expression of the ghrelin receptor, the growth hormone secretagogue receptor 1A, was found to co-localize with choline acetyltransferase, a marker of cholinergic neurons, in the LDTg. Finally, mecamylamine treatment i.p. decreased the ability of palatable food to condition a place preference. These data suggest that ghrelin-induced food intake is partly mediated via nAChRs and that nicotinic blockade decreases the rewarding properties of food.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Grelina/farmacologia , Receptores Nicotínicos/fisiologia , Transdução de Sinais/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos , Análise de Variância , Animais , Colina O-Acetiltransferase/metabolismo , Condicionamento Operante/efeitos dos fármacos , Vias de Administração de Medicamentos , Interações Medicamentosas , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Preferências Alimentares/efeitos dos fármacos , Preferências Alimentares/fisiologia , Hexametônio/farmacologia , Masculino , Mecamilamina/farmacologia , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Antagonistas Nicotínicos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Grelina/deficiência , Receptores Nicotínicos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/citologia
11.
J Neuroendocrinol ; 21(9): 777-85, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19703102

RESUMO

In the present study, we explore the central nervous system mechanism underlying the chronic central effects of ghrelin with respect to increasing body weight and body fat. Specifically, using a recently developed ghrelin receptor antagonist, GHS-R1A (JMV2959), we investigate the role of GHS-R1A in mediating the effects of ghrelin on energy balance and on hypothalamic gene expression. As expected, in adult male rats, chronic central treatment with ghrelin for 14 days, when compared to vehicle-treated control rats, resulted in an increased body weight, lean mass and fat mass (assessed by dual X-ray absorptiometry), dissected white fat pad weight, cumulative food intake, food efficiency, respiratory exchange ratio and a decrease of energy expenditure. Co-administration of the ghrelin receptor antagonist JMV2959 suppressed/blocked the majority of these effects, with the notable exception of ghrelin-induced food intake and food efficiency. The hypothesis emerging from these data, namely that GHS-R1A mediates the chronic effects of ghrelin on fat accumulation, at least partly independent of food intake, is discussed in light of the accompanying data regarding the hypothalamic genes coding for peptides and receptors involved in energy balance regulation, which were found to have altered expression in these studies.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Grelina/farmacologia , Antagonistas de Hormônios/farmacologia , Obesidade/induzido quimicamente , Receptores de Grelina/antagonistas & inibidores , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Grelina/administração & dosagem , Grelina/efeitos adversos , Grelina/antagonistas & inibidores , Hormônios/sangue , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraventriculares , Masculino , Obesidade/sangue , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
12.
J Neuroendocrinol ; 21(7): 620-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19490366

RESUMO

Interleukin (IL)-6 is a pro-inflammatory cytokine that also affects metabolic function because IL-6 depleted (IL-6(-/-)) mice develop late-onset obesity. IL-6 appears to act in the central nervous system, presumably in the hypothalamus, to increase energy expenditure that appears to involve stimulation of the sympathetic nervous system. In the present study, we explored possible central mechanisms for the effects exerted by IL-6 on body fat. Therefore, we measured the effects of IL-6 depletion in IL-6(-/-) mice on expression of key hypothalamic peptide genes involved in energy balance by the real time polymerase chain reaction. Additionally, co-localisation between such peptides and IL-6 receptor alpha was investigated by immunohistochemistry. IL-6 deficiency decreased the expression of several peptides found in the paraventricular nucleus (PVN), which is a nucleus that has been attributed an adipostatic function. For example, corticotrophin-releasing hormone (CRH), which is reported to stimulate the sympathetic nervous system, was decreased by 40% in older IL-6(-/-) mice. Oxytocin, which is reported to prevent obesity, was also decreased in older IL-6(-/-) animals, as was arginine vasopressin (AVP). The IL-6 receptor alpha was abundantly expressed in the PVN, but also in the supraoptic nucleus, and was shown to be co-expressed to a high extent with CRH, AVP, oxytocin and thyrotrophin-releasing hormone. These data indicate that depletion of endogenous IL-6, a body fat suppressing cytokine, is associated with the decreased expression of CRH and oxytocin (i.e. energy balance regulating peptides) as well as AVP in the PVN. Because IL-6 receptor alpha is co-expressed with CRH, oxytocin and AVP, IL-6 could stimulate the expression of these peptides directly.


Assuntos
Interleucina-6/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Supraóptico/metabolismo , Adiposidade/genética , Adiposidade/fisiologia , Animais , Arginina Vasopressina/metabolismo , Contagem de Células , Hormônio Liberador da Corticotropina/metabolismo , Imuno-Histoquímica , Interleucina-6/deficiência , Interleucina-6/genética , Subunidade alfa de Receptor de Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ocitocina/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Hormônio Liberador de Tireotropina/metabolismo
13.
Eur Surg Res ; 40(3): 279-88, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18253047

RESUMO

BACKGROUND/AIM: Gastric bypass (GB) is usually designed to restrict food intake and to induce malabsorption. Gastric hormones have been thought to play a role in the regulation of food intake and body weight. The aim of the present study was to analyze feeding behavior after total gastrectomy (Gx) or GB in rats. METHODS: Animals were subjected to Gx, GB, or sham operations. Eating and drinking behaviors after surgeries were assessed by a comprehensive laboratory animal monitoring system. Gastric hormones were measured by radioimmunoassay and energy density in feces by adiabatic bomb calorimeter. RESULTS: Compared with sham operation, both Gx and GB reduced the body weight as measured during 3-8 weeks postoperatively, which was associated with increased energy expenditure per 100 g body weight. Daily accumulated food intake and meal size (during nighttime) were reduced following Gx, but not GB. The water intake (during daytime) was increased after Gx and GB. The energy density in feces was unchanged. Serum concentrations of ghrelin, obestatin, leptin, gastrin, and pancreastatin were greatly reduced after Gx. CONCLUSIONS: Control of food intake and meal size was independent of the food reservoir function of the stomach. Surgical depletion of gastric hormones is associated with reduced meal size, but increased water intake.


Assuntos
Comportamento Alimentar , Gastrectomia , Derivação Gástrica , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
14.
J Neuroendocrinol ; 17(6): 387-93, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15929744

RESUMO

The hypothalamus appears to be more responsive to ghrelin and growth hormone secretagogues (GHS) in fasting, as reflected by a two- to three-fold increase in the number of cells detected that express Fos protein in the arcuate nucleus, in 48-h fasted rats compared to fed controls. Moreover, this increased hypothalamic responsiveness to GHS in fasting is regulated by the central action of exogenous leptin and insulin, although it is unknown whether these hormones mediate the changes in hypothalamic responsiveness to GHS associated with the fasting/fed state. In the present study, we show that refeeding with normal rat chow for only 2 h at the end of a 48-h fast reversed the potentiation of the Fos response to GHRP-6 observed in fasted rats. Circulating leptin and insulin levels remained significantly lower in refed rats compared to ad lib-fed rats, suggesting that the change in the hypothalamic sensitivity brought about by refeeding was independent of these hormones. By contrast, 2 h of chow refeeding at the end of a fast restored plasma glucose levels to those of the fed state. Refeeding with sugar alone for 2 h at the end of a 48-h fast also reduced the potentiated Fos response in fasting, indicating that elevated blood glucose can influence the central responsiveness to ghrelin/GHS. By contrast, infusion of the ileal satiety factor, PYY(3-36) (known to increase postprandially) did not alter the central responsiveness to GHRP-6, although it suppressed feeding and body weight as expected. This study highlights the importance of nutritional status in regulating the action of exogenous GHS (and presumably endogenous ghrelin) on the hypothalamic circuits controlling food intake.


Assuntos
Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Oligopeptídeos/farmacologia , Ração Animal , Animais , Glicemia , Carboidratos/farmacologia , Grelina , Insulina/sangue , Leptina/sangue , Masculino , Mimetismo Molecular , Oligopeptídeos/química , Fragmentos de Peptídeos , Hormônios Peptídicos/química , Peptídeo YY/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar
15.
FASEB J ; 19(10): 1302-4, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15928196

RESUMO

Peg3 encodes a C2H2 type zinc finger protein that is implicated in a novel physiological pathway regulating core body temperature, feeding behavior, and obesity in mice. Peg3+/- mutant mice develop an excess of abdominal, subcutaneous, and intra-scapular fat, despite a lifetime of lower food intake than wild-type animals. However, they start life with reduced fat reserves and are slower to enter puberty. These mice maintain a lower core body temperature, fail to respond to a cold challenge, and have lower metabolic activity as measured by oxygen consumption. Plasma leptin levels are significantly higher than in wild types, and Peg3+/- mice appear to have developed leptin resistance. Administration of exogenous leptin resulted in a significant reduction in food intake in wild-type mice that was not observed in Peg3+/- mutants. This mutation, which is strongly expressed in hypothalamic tissue during development, has the capacity to regulate multiple events relating to energy homeostasis.


Assuntos
Tecido Adiposo/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Animais , Composição Corporal , Temperatura Corporal , Peso Corporal , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético , Feminino , Hipotálamo/fisiologia , Fatores de Transcrição Kruppel-Like , Leptina/sangue , Leptina/farmacologia , Masculino , Camundongos , Atividade Motora , Mutação , Neuropeptídeo Y/genética , Obesidade/etiologia , Consumo de Oxigênio , Pró-Opiomelanocortina/genética , RNA Mensageiro/análise , Maturidade Sexual
16.
Gut ; 54(7): 907-13, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15849166

RESUMO

BACKGROUND AND AIMS: The gastric hormone ghrelin has been reported to stimulate food intake, increase weight gain, and cause obesity but its precise physiological role remains unclear. We investigated the long term effects of gastrectomy evoked ghrelin deficiency and of daily ghrelin injections on daily food intake, body weight, fat mass, lean body mass, and bone mass in mice. METHODS: Ghrelin was given by subcutaneous injections (12 nmol/mouse once daily) for eight weeks to young female mice subjected to gastrectomy or sham operation one week previously. RESULTS: Gastrectomy reduced plasma concentrations of total ghrelin (octanoylated and des-octanoylated) and active (octanoylated) ghrelin by approximately 80%. Immediately after injection of ghrelin, the plasma concentration was supraphysiological and was still elevated 16 hours later. Daily food intake was not affected by either gastrectomy or ghrelin treatment. The effect of ghrelin on meal initiation was not studied. At the end point of the study, mean body weight was 15% lower in gastrectomised mice than in sham operated mice (p<0.001); daily ghrelin injections for eight weeks partially prevented this weight loss. In sham operated mice, ghrelin had no effect on body weight. The weight of fat was reduced in gastrectomised mice (-30%; p<0.01). This effect was reversed by ghrelin, enhancing the weight of fat in sham operated mice also (+20%; p<0.05). Gastrectomy reduced lean body mass (-10%; p<0.01) and bone mass (-20%; p<0.001) compared with sham operated mice. Ghrelin replacement prevented the gastrectomy induced decrease in lean body mass but did not affect bone. In sham operated mice, ghrelin affected neither of these two parameters. CONCLUSIONS: Ghrelin replacement partially reversed the gastrectomy induced reduction in body weight, lean body mass, and body fat but not in bone mass. In sham operated mice, ghrelin only increased fat mass. Our results suggest that ghrelin is mainly concerned with the control of fat metabolism and that ghrelin replacement therapy may alleviate the weight loss associated with gastrectomy.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Gastrectomia , Hormônios Peptídicos/farmacologia , Aumento de Peso/efeitos dos fármacos , Tecido Adiposo/patologia , Animais , Composição Corporal/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Fêmur/patologia , Grelina , Camundongos , Hormônios Peptídicos/sangue , Hormônios Peptídicos/deficiência , Período Pós-Operatório
17.
Eur J Endocrinol ; 150(6): 905-11, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15191362

RESUMO

OBJECTIVE: Chronic administration of GH secretagogues (GHSs) induces a state of positive energy balance in rodents by a GH-independent mechanism. Here we sought to determine to what extent the GHS effects to increase food intake and increase fat accumulation are glucocorticoid-dependent. DESIGN: The effects of twice-daily s.c. injections of GH-releasing peptide-6 (GHRP-6) (250 microg/kg) for 2 weeks on body weight, food intake and fat pad weight were determined in both adrenalectomised (ADX) rats (with or without basal corticosterone replacement) and adrenal-intact rats. RESULTS: All GHS-injected rats had a significantly increased body weight at the end of 2 weeks of treatment compared with saline controls. However, increased fat accumulation was only seen in adrenal-intact rats, with a 15% increase in s.c. inguinal (P<0.05 vs saline controls) and 20% increase in visceral mesenteric (P<0.05) fat pad weights following GHS treatment. The increased body weight observed in ADX rats following GHS treatment was not due to increased fat mass or increased weight of other organs measured. Food intake was increased for up to 7 h following a single injection of GHRP-6 in both the adrenal-intact (P<0.01) and corticosterone-replacement groups (P<0.05). This stimulating effect on food intake was not observed at any time point in the ADX rats without corticosterone replacement. CONCLUSION: These data suggest that GHS-induced body weight gain is glucocorticoid-independent. However, basal levels of glucocorticoids are permissive for the GHS-induced increase in food intake whilst activation of the hypothalamo-pituitary-adrenal axis appears to contribute to the GHS-induced accumulation of fat mass.


Assuntos
Tecido Adiposo , Apetite/efeitos dos fármacos , Composição Corporal/efeitos dos fármacos , Glucocorticoides/fisiologia , Oligopeptídeos/administração & dosagem , Hormônios Peptídicos/agonistas , Adrenalectomia , Animais , Peso Corporal/efeitos dos fármacos , Corticosterona/administração & dosagem , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Grelina , Cinética , Ratos , Ratos Wistar
18.
Proc Natl Acad Sci U S A ; 101(13): 4695-700, 2004 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-15070780

RESUMO

Inactivating mutations of the pro-opiomelanocortin (POMC) gene in both mice and humans leads to hyperphagia and obesity. To further examine the mechanisms whereby POMC-deficiency leads to disordered energy homeostasis, we have generated mice lacking all POMC-derived peptides. Consistent with a previously reported model, Pomc(-/-) mice were obese and hyperphagic. They also showed reduced resting oxygen consumption associated with lowered serum levels of thyroxine. Hypothalami from Pomc(-/-) mice showed markedly increased expression of melanin-concentrating hormone mRNA in the lateral hypothalamus, but expression of neuropeptide Y mRNA in the arcuate nucleus was not altered. Provision of a 45% fat diet increased energy intake and body weight in both Pomc(-/-) and Pomc(+/-) mice. The effects of leptin on food intake and body weight were blunted in obese Pomc(-/-) mice whereas nonobese Pomc(-/-) mice were sensitive to leptin. Surprisingly, we found that Pomc(-/-) mice maintained their acute anorectic response to peptide-YY(3-36) (PYY(3-36)). However, 7 days of PYY(3-36) administration had no effect on cumulative food intake or body weight in wild-type or Pomc(-/-) mice. Thus, POMC peptides seem to be necessary for the normal response of energy balance to high-fat feeding, but not for the acute anorectic effect of PYY(3-36) or full effects of leptin on feeding. The finding that the loss of only one copy of the Pomc gene is sufficient to render mice susceptible to the effects of high fat feeding emphasizes the potential importance of this locus as a site for gene-environment interactions predisposing to obesity.


Assuntos
Depressores do Apetite/farmacologia , Gorduras na Dieta/farmacologia , Leptina/farmacologia , Peptídeo YY/farmacologia , Pró-Opiomelanocortina/deficiência , Pró-Opiomelanocortina/genética , Animais , Sequência de Bases , Peso Corporal/efeitos dos fármacos , Primers do DNA , Ingestão de Energia , Hormônios Hipotalâmicos/genética , Hipotálamo/fisiologia , Cinética , Melaninas/genética , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Neuropeptídeo Y/genética , Obesidade/genética , Fragmentos de Peptídeos , Fenótipo , Hormônios Hipofisários/genética , Reação em Cadeia da Polimerase , Transcrição Gênica
19.
J Neuroendocrinol ; 16(3): 191-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15049849

RESUMO

The set points for vasopressin release in response to increasing plasma osmolality and hypovolaemia alter with reproductive status. Here, we studied stimulated vasopressin release following ovariectomy and oestrogen replacement, neuronal activity being measured in terms of immediate early gene expression. Observations were carried out on three groups of female Sprague-Dawley rats. The first group were ovariectomized. The second group were given a subcutaneous oestrogen implant (20 microg/ml oestradiol-17 beta) at the time of ovariectomy. The final group were left intact and observations performed at oestrus. Two weeks after ovariectomy, vascular cannulae were implanted under anaesthesia and at least 48 h allowed for recovery before hormone release was stimulated by infusion of 1.5 M NaCl for 90 min, or hypovolaemia induced by the removal of 10 mg/kg body weight taken in 1-ml aliquots. Blood pressure was monitored, and blood samples were taken for determination of packed cell volume and plasma vasopressin and osmolality. After a minimum of 48 h, the challenge was repeated, the rats anaesthetized, and perfused with 4% paraformaldehyde. Brain sections were processed for immunocytochemical detection of Fos protein. Vasopressin release in response to both stimuli was reduced in ovariectomized compared to intact rats and the response could be substantially restored by oestradiol replacement. The number of Fos positive cells in the supraoptic nucleus of oestrogen-replaced rats was significantly higher than in the ovariectomized group and not statistically different from the intact group.


Assuntos
Estradiol/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Supraóptico/metabolismo , Vasopressinas/sangue , Equilíbrio Hidroeletrolítico/fisiologia , Adaptação Fisiológica , Análise de Variância , Animais , Volume Sanguíneo/fisiologia , Ciclo Estral/fisiologia , Feminino , Genes Precoces/fisiologia , Hipovolemia/sangue , Hipovolemia/fisiopatologia , Imuno-Histoquímica , Concentração Osmolar , Ovariectomia , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
20.
Biochem Biophys Res Commun ; 311(4): 915-9, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14623268

RESUMO

It has recently been suggested that gut-derived PYY(3-36) may be involved in the central mediation of post-prandial satiety signals. We have examined the acute effects of peripherally administered PYY(3-36) on food intake and hypothalamic gene expression of neuropeptides in mice. A single intraperitoneal injection of PYY(3-36) to mice that had been fasted for 24h resulted in a highly significant reduction in food intake at 6 and 24h post-injection but not at 48h. However, in freely fed mice, food intake was unaltered by PYY(3-36) administration. In the arcuate nucleus POMC mRNA expression was significantly elevated at 6h and remained elevated at 24h following PYY(3-36) injection. By contrast NPY mRNA expression in the arcuate nucleus was suppressed at 6h but not at 24h post-injection. In the lateral hypothalamus there were no differences in MCH mRNA expression at either time point. In conclusion, peripherally administered PYY(3-36) has a suppressive effect on food intake that is more prominent in recently fasted mice and lasts up to 24 h. This is associated with a short-lived suppression of NPY mRNA, a longer lasting increase in POMC mRNA but no change in MCH mRNA expression.


Assuntos
Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Peptídeo YY/administração & dosagem , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/citologia , Injeções Intraperitoneais , Melaninas/metabolismo , Camundongos , Neuropeptídeo Y/metabolismo , Hormônios Hipofisários/metabolismo , Pró-Opiomelanocortina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...