Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Neuroendocrinol ; 29(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28477438

RESUMEN

In common forms of obesity, leptin fails to convey its regulatory effect. This so called "leptin resistance" is not well understood, and solving this puzzle is a key to understanding how obesity develops. In the present study, we investigated the temporal and regional onset of leptin resistance in response to a diet enriched with long-chain saturated fatty acids (high-fat diet; HFD) in mice. Mice were exposed to either a low-fat diet (LFD) or a HFD for 4 hours, 24 hours, 10 days and 28 days. Mice in each group received an i.p. injection of either phosphate-buffered saline or leptin and the number of phosphorylated signal transducer and activator of transcription-3 (pSTAT3) immunoreactive (-IR) cells in the arcuate nucleus (ARC), ventromedial nucleus of the hypothalamus (VMH) and dorsomedial nucleus of the hypothalamus (DMH) was analysed 30 or 120 minutes after treatment. In the ARC, as soon as 24 hours of HFD, the molecular leptin response was reduced by 40% (P≤.01). Compared to at 24 hours, after 10 days, the number of leptin-induced pSTAT3-IR cells was elevated after 120 minutes, suggesting a sustained response and a partial return of leptin sensitivity. After 28 days, leptin failed to induce the number of pSTAT3-IR over control levels, suggesting a markedly reduced sensitivity to leptin. In the VMH after 24 hours, we observed a 50% reduction in leptin-induced pSTAT-3-IR cells, followed by a further decline after 10 days. However, after 28 days, there was a significant increase in pSTAT-3-IR cells (P≤.05), indicating partial recovery of leptin sensitivity. By contrast to these two regions, in the DMH, no loss of leptin sensitivity was observed at any time-point. These findings demonstrate that a loss of sensitivity to leptin occurs rapidly after exposure to HFD in the ARC and VMH but not the DMH. However, there appears to be a biphasic pattern of leptin responsiveness, with a partial return of leptin sensitivity occurring after 10 days in the arcuate nucleus, and after 28 days in the VMH. By 28 days, the response to leptin in the arcuate nucleus was completely lost. These findings suggest that the molecular responses to leptin are altered after high-fat feeding in a time- and region-specific manner.


Asunto(s)
Dieta Alta en Grasa , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Modelos Animales de Enfermedad , Núcleo Hipotalámico Dorsomedial/metabolismo , Leptina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Fosforilación , Factor de Transcripción STAT3/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo
2.
J Neuroendocrinol ; 28(3): 12368, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26802435

RESUMEN

Wnt signalling and its downstream effectors are well known for their roles in embryogenesis and tumourigenesis, including the regulation of cell proliferation, survival and differentiation. In the nervous system, Wnt signalling has been described mainly during embryonic development, although accumulating evidence suggests that it also plays a major role in adult brain morphogenesis and function. Studies have predominantly concentrated on memory formation in the hippocampus, although recent data indicate that Wnt signalling is also critical for neuroendocrine control of the developed hypothalamus, a brain centre that is key in energy balance regulation and whose dysfunction is implicated in metabolic disorders such as type 2 diabetes and obesity. Based on scattered findings that report the presence of Wnt molecules in the tanycytes and ependymal cells lining the third ventricle and arcuate nucleus neurones of the hypothalamus, their potential importance in key regions of food intake and body weight regulation has been investigated in recent studies. The present review brings together current knowledge on Wnt signalling in the hypothalamus of adult animals and discusses the evidence suggesting a key role for members of the Wnt signalling family in glucose and energy balance regulation in the hypothalamus in diet-induced and genetically obese (leptin deficient) mice. Aspects of Wnt signalling in seasonal (photoperiod sensitive) rodents are also highlighted, given the recent evidence indicating that the Wnt pathway in the hypothalamus is not only regulated by diet and leptin, but also by photoperiod in seasonal animals, which is connected to natural adaptive changes in food intake and body weight. Thus, Wnt signalling appears to be critical as a modulator for normal functioning of the physiological state in the healthy adult brain, and is also crucial for normal glucose and energy homeostasis where its dysregulation can lead to a range of metabolic disorders.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Vía de Señalización Wnt/fisiología , Adulto , Animales , Regulación del Apetito/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Homeostasis/genética , Humanos , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo , Fotoperiodo
3.
J Neuroendocrinol ; 26(2): 58-67, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24382295

RESUMEN

The occurrence of type II diabetes is highly correlated with obesity, although the mechanisms linking the two conditions are incompletely understood. Leptin is a potent insulin sensitiser and, in leptin-deficient, insulin insensitive, Lep(ob/ob) mice, leptin improves glucose tolerance, indicating that leptin resistance may link obesity to insulin insensitivity. Leptin resistance occurs in response to a high-fat diet (HFD) and both hyperleptinaemia and inflammation have been proposed as causative mechanisms. Scrutinising the role of hyperleptinaemia in this process, central hyperleptinaemia in Lep(ob/ob) mice was induced by chronic i.c.v. infusion of leptin (4.2 µg/day) over 10 days. This treatment led to a dramatic decline in body weight and food intake, as well as an improvement in glucose tolerance. Transfer to HFD for 4 days markedly arrested the beneficial effects of leptin on these parameters. Because Lep(ob/ob) mice are exquisitely sensitive to leptin, the possibility that leptin could reverse HFD-induced glucose intolerance in these animals was investigated. HFD led to increased body weight and glucose intolerance compared to a low-fat diet (LFD). Older and heavier Lep(ob/ob) mice were used as body weight-matched controls. Mice in each group received either i.p. leptin (1.25 mg/kg) or vehicle, and glucose tolerance, food intake and the number of phosphorylated signal transducer and activator of transcription (pSTAT)3 immunoreactive cells in the arcuate nucleus (ARC) and ventromedial hypothalamus (VMH) were analysed. Leptin improved glucose tolerance (P = 0. 019) and reduced food intake in Lep(ob/ob) mice on LFD (P ≤ 0.001) but was ineffective in mice on HFD. Furthermore, when leptin was administered centrally, the glucose tolerance of Lep(ob/ob) mice on HFD was significantly impaired (P = 0.007). Although leptin induced the number of pSTAT3 immunoreactive cells in the ARC and VMH of Lep(ob/ob) mice on LFD, HFD was associated with elevated pSTAT3 immunoreactivity in vehicle-treated Lep(ob/ob) mice that was unaffected by leptin treatment, suggesting central leptin resistance. Negating central inflammation by co-administering a c-Jun n-terminal kinase (JNK) inhibitor reinstated the glucose-lowering effects of leptin. These findings demonstrate that Lep(ob/ob) mice develop leptin resistance on a HFD independent of hyperleptinaemia and also indicate that the JNK inflammatory pathway plays a key role in the induction of diet-induced glucose intolerance.


Asunto(s)
Dieta Alta en Grasa , Leptina/fisiología , Animales , Inflamación/fisiopatología , Leptina/administración & dosificación , Leptina/sangre , Leptina/genética , Ratones , Ratones Transgénicos , Fosforilación , Factor de Transcripción STAT3/metabolismo
4.
J Neuroendocrinol ; 25(5): 446-54, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23301857

RESUMEN

The hypothalamus has been identified as a main insulin target tissue for regulating normal body weight and glucose metabolism. Recent observations suggest that c-Jun-N-terminal kinase (JNK)-signalling plays a crucial role in the development of obesity and insulin resistance because neuronal JNK-1 ablation in the mouse prevented high-fat diet-induced obesity (DIO) and increased energy expenditure, as well as insulin sensitivity. In the present study, we investigated whether central JNK inhibition is associated with sensitisation of hypothalamic insulin signalling in mice fed a high-fat diet for 3 weeks and in leptin-deficient mice. We determined whether i.c.v. injection of a pharmacological JNK-inhibitor (SP600125) improved impaired glucose homeostasis. By immunohistochemistry, we first observed that JNK activity was increased in the arcuate nucleus (ARC) and the ventromedial hypothalamus (VMH) in both mouse models, relative to normoglycaemic controls. This suggests that up-regulation of JNK in these regions is associated with glucose intolerance and obesity, independent of leptin levels. Acute i.c.v. injection of SP600125 ameliorated glucose tolerance within 30 min in both leptin-deficient and DIO mice. Given the acute nature of i.c.v. injections, these effects cannot be attributed to changes in food intake or energy balance. In a hypothalamic cell line, and in the ARC and VMH of leptin-deficient mice, JNK inhibition by SP600125 consistently improved impaired insulin signalling. This was determined by a reduction of phospho-insulin receptor substrate-1 [IRS-1(Ser612)] protein in a hypothalamic cell line and a decline in the number of pIRS-1(Ser612) immunoreactive cells in the ARC and VMH. Serine 612 phosphorylation of IRS-1 is assumed to negatively regulate insulin signalling. In leptin-deficient mice, in both nuclei, central inhibition of JNK increased the number of cells immunoreactive for phospho-Akt (Ser473) and phospho-GSK-3ß (Ser9), which are important markers of insulin signalling. Collectively, our data suggest that the acute inhibition of central JNK improves impaired glucose homeostasis and is associated with sensitisation of hypothalamic insulin signalling.


Asunto(s)
Conducta Alimentaria , Neuronas/fisiología , Oxitocina/fisiología , Hormona Liberadora de Prolactina/fisiología , Animales , Ratones , Péptidos/análisis , Ratas
5.
J Neuroendocrinol ; 22(6): 534-42, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20236230

RESUMEN

Both insulin and leptin action in the brain are considered to involve activation of phosphoinositide 3-kinase (PI3K), although the roles of different PI3K isoforms in insulin signalling in the hypothalamus are unknown. In the present study, we characterised the roles of these isoforms in hypothalamic insulin and leptin signalling and investigated the cross-talk of both hormones. To evaluate PI3K levels in the hypothalamus, PI3K was immunoprecipitated using an antibody directed against the p85 subunit, and then total PI3K activity was measured in the presence of novel isoform-selective pharmacological inhibitors of each isoform of PI3K. Subsequently, these inhibitors were administered into the lateral ventricle of male Sprague-Dawley rats, followed by vehicle, insulin, leptin or both hormones 45 min later. PI3K activity was determined by immunohistochemical detection of phosphorylated AKT (S473). In a separate study, the effects of the inhibitors on the anorexigenic action of insulin and leptin were determined. Hypothalamic insulin signalling was specifically mediated by the combined actions of the class Ia isoforms p110alpha and p110beta. Total hypothalamic PI3K activity was inhibited 65% by a p110alpha inhibitor, and 35% by a p110beta inhibitor, with a combination of inhibitors being equally effective as the broad-spectrum PI3K inhibitor wortmannin. Individual i.c.v. administration of p110alpha and p110beta inhibitors partly prevented insulin-induced phosphorylated AKT (S473) in the arcuate nucleus, whereas simultaneous application completely blocked insulin action. Unlike insulin, leptin did not induce phosphorylated AKT in the hypothalamus, as detected by immunohistochemistry, and the anorectic effects of leptin were not affected by pre-treatment with a combination of p110alpha and p110beta inhibitors. The enhanced anorectic effect of a combined i.c.v. application of both insulin and leptin could be prevented by pre-treatment with the combination of p110alpha and p110beta inhibitors. The data suggest that p110alpha and p110beta isoforms of PI3K are necessary to mediate insulin action in the hypothalamus. The role of PI3K in leptin action is less clear, but it may be involved by means of an insulin-dependent sensitisation of leptin action.


Asunto(s)
Hipotálamo/metabolismo , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Isoformas de Proteínas/fisiología , Transducción de Señal/fisiología , Animales , Anorexia/etiología , Fosfatidilinositol 3-Quinasa Clase I , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Cinética , Masculino , Fosfatidilinositol 3-Quinasas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
6.
J Neuroendocrinol ; 21(11): 961-71, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19732287

RESUMEN

In common forms of obesity, animals and humans become leptin resistant associated with impaired regulation of energy homeostasis. Over the last decade, significant advances in delineating the underlying mechanisms have been achieved. As well as the obvious scientific progress obtained by novel transgenic animals, natural and physiological models of leptin resistance such as the Siberian hamster (Phodoups sungorus), the field vole (Microtus agrestis) or the rat during pregnancy have also provided invaluable insight into the dynamic long-term control of energy homeostasis. Seasonal (in the hamster) and pregnancy-induced leptin resistance are characterised by a modulation of the leptin signalling cascade downstream of its receptor in the hypothalamus. In this state, leptin-induced phosphorylation of the important transcription factor, signal transducer and activator of transcription 3 (STAT3), is impaired in the arcuate nucleus and the ventromedial hypothalamus (only during pregnancy). This is accompanied by elevated levels of leptin signalling inhibitors such as the suppressor of cytokine signalling (SOCS3) and the protein tyrosine phosphatase 1B (PTP1B). The janus kinase 2 (JAK2)-STAT3 signalling pathway might be modulated by a dual function of the tyrosine residue Tyr(985) in the intracellular domain of the leptin receptor. In seasonal animals, SOCS3, most importantly seems to act as a 'molecular switch' enabling a photoperiod-induced alteration in leptin signalling and subsequent adjustments in energy homeostasis to allow attainment of a new body weight set-point. These physiological models show that animals can exhibit leptin resistance as an adaptive response to meet new physiological or environmental challenges, promoting the survival of the species during times of increased metabolic demand. The molecular mechanisms mediating physiological and/or pathological leptin resistance, like during diet induced obesity, might be very similar involving hypothalamic SOCS3. Investigation of these models might further provide new insight into the dynamic complexity of energy homeostasis.


Asunto(s)
Leptina/fisiología , Modelos Biológicos , Animales , Femenino , Fosforilación , Embarazo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
7.
J Neuroendocrinol ; 21(5): 449-56, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19302191

RESUMEN

Hypothalamic leptin resistance during pregnancy is an important adaptation that facilitates the state of positive energy balance required for fat deposition in preparation for lactation. Within the arcuate nucleus, pro-opiomelanocortin (POMC) neurones and neuropeptide Y (NPY)/agouti-related gene protein (AgRP) neurones are first-order leptin responsive neurones involved in the regulation of energy balance. The present study aimed to investigate whether the regulation of these neuropeptides is disrupted during pregnancy in association with the development of leptin resistance. As measured by quantitative in situ hybridisation, POMC and AgRP mRNA levels were not significantly different during pregnancy, whereas NPY mRNA levels increased such that, by day 21 of pregnancy, levels were significantly higher than in nonpregnant, animals. These data suggest that these neurones were not responding normally to the elevated leptin found during pregnancy. To further characterise the melanocortin system during pregnancy, double-label immunohistochemistry was used to quantify leptin-induced phosphorylation of signal transducer and activator of transcription 3 (pSTAT3) in POMC neurones, using α-melanocyte-stimulating hormone (MSH) as a marker. The percentage of α-MSH neurones containing leptin-induced pSTAT3 did not significantly differ from nonpregnant animals, indicating that there was no change in the number of POMC neurones that respond to leptin during pregnancy. Treatment with α-MSH significantly reduced food intake in nonpregnant rats, but not in pregnant rats, indicating resistance to the satiety actions of α-MSH during pregnancy. The data suggest that multiple mechanisms contribute to leptin resistance during pregnancy. As well as a loss of responses in first-order leptin-responsive neurones in the arcuate nucleus, there is also a downstream disruption in the melanocortin system.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/fisiología , Leptina/metabolismo , Melanocortinas/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Cricetinae , Femenino , Hipotálamo/citología , Hibridación in Situ , Lactancia/fisiología , Neuronas/citología , Neuronas/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Embarazo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , alfa-MSH/metabolismo , alfa-MSH/farmacología
8.
J Neuroendocrinol ; 19(2): 83-94, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17214870

RESUMEN

We have previously shown that cold-acclimated (8 degrees C) male field voles (Microtus agrestis) transferred from short day (SD, 8 h light) to long day (LD, 16 h light) photoperiod exhibit an increase in body mass lasting 4 weeks, after which they stabilise at a new plateau approximately 7.5 g (24.8%) higher than animals maintained in SD. By infusing voles with exogenous leptin, we have also demonstrated that SD voles respond to the hormone by reducing body mass and food intake, whereas LD animals increasing body mass are resistant to leptin treatment. In the present study, we investigated whether seasonal changes in body mass could be linked to modulation of the leptin signal by suppressor of cytokine signalling-3 (SOCS3). We used in situ hybridisation to examine hypothalamic arcuate nucleus (ARC) expression of SOCS3, neuropeptide Y (NPY), agouti-related peptide (AgRP), pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) genes in 90 voles exposed to either SD or LD for up to 11 weeks. LD voles increasing body mass had significantly higher levels of SOCS3 mRNA than SD or LD voles with a stable body mass. There were no associated changes in expression of NPY, AgRP, POMC and CART genes. These results suggest that voles that regulate body mass at either the lower (SD) or upper (LD) plateau remain sensitive to leptin action, whereas SOCS3-mediated leptin resistance is a short-term mechanism that enables animals to move between the stable body mass plateaus. Our data provide evidence that expression of SOCS3 in the ARC is involved in the modulation of the strength of the leptin signal to facilitate seasonal cycles in body mass and adiposity.


Asunto(s)
Aclimatación/fisiología , Núcleo Arqueado del Hipotálamo/metabolismo , Arvicolinae/metabolismo , Leptina/fisiología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteína Relacionada con Agouti , Animales , Peso Corporal/fisiología , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Fotoperiodo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Mensajero/análisis , Estaciones del Año , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/genética
9.
J Neuroendocrinol ; 18(6): 413-25, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16684131

RESUMEN

A remarkable feature of the seasonal adaptation displayed by the Siberian hamster (Phodopus sungorus) is the ability to decrease food intake and body weight (by up to 40%) in response to shortening photoperiod. The regulating neuroendocrine systems involved in this adaptation and their neuroanatomical and molecular bases are poorly understood. We investigated the effect of photoperiod on the expression of prohormone convertases 1 (PC1/3) and 2 (PC2) and the endoproteolytic processing of the neuropeptide precursor pro-opiomelanocortin (POMC) within key energy balance regulating centres of the hypothalamus. We compared mRNA levels and protein distribution of PC1/3, PC2, POMC, adrenocorticotrophic hormone (ACTH), alpha-melanocyte-stimulating hormone (MSH), beta-endorphin and orexin-A in selected hypothalamic areas of long day (LD, 16:8 h light:dark), short day (SD, 8:16 h light:dark) and natural-day (ND, photoperiod depending on time of the year) acclimated Siberian hamsters. The gene expression of PC2 was significantly higher within the arcuate nucleus (ARC, P < 0.01) in SD and in ND (versus LD), and is reflected in the day length profile between October and April in the latter. PC1/3 gene expression in the ARC and lateral hypothalamus was higher in ND but not in SD compared to the respective LD controls. The immunoreactivity of PC1/3 cleaved neuropeptide ACTH in the ARC and PC1/3-colocalised orexin-A in the lateral hypothalamus were not affected by photoperiod changes. However, increased levels of PC2 mRNA and protein were associated with higher abundance of the mature neuropeptides alpha-MSH and beta-endorphin (P < 0.01) in SD. This study provides a possible explanation for previous paradoxical findings showing lower food intake in SD associated with decreased POMC mRNA levels. Our results suggest that a major part of neuroendocrine body weight control in seasonal adaptation may be effected by post-translational processing mediated by the prohormone convertases PC1/3 and PC2, in addition to regulation of gene expression of neuropeptide precursors.


Asunto(s)
Adaptación Fisiológica/genética , Fotoperiodo , Proopiomelanocortina/genética , Proproteína Convertasa 1/genética , Proproteína Convertasa 2/genética , Hormona Adrenocorticotrópica/metabolismo , Animales , Peso Corporal/fisiología , Cricetinae , Femenino , Regulación Enzimológica de la Expresión Génica/fisiología , Área Hipotalámica Lateral/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neuropéptidos/genética , Neuropéptidos/metabolismo , Orexinas , Phodopus , Proopiomelanocortina/metabolismo , Proproteína Convertasa 1/metabolismo , Proproteína Convertasa 2/metabolismo , Precursores de Proteínas/genética , Procesamiento Proteico-Postraduccional/fisiología , ARN Mensajero/análisis , Estaciones del Año , alfa-MSH/metabolismo , betaendorfina/metabolismo
10.
J Neuroendocrinol ; 18(2): 139-45, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16420283

RESUMEN

The Siberian hamster, Phodopus sungorus, exhibits a remarkable cycle of body weight, reproduction and leptin sensitivity in response to a seasonal change in photoperiod. In the present study, we investigated the hypothesis that the suppressor of cytokine signalling 3 (SOCS3) plays a critical role in the regulation of the seasonal body weight cycle. We analysed arcuate nucleus SOCS3 gene expression in short day length (SD; 8 : 16 h light/dark) acclimated Siberian hamsters that were transferred back to long day length (LD; 16 : 8 h light/dark) and in hamsters that spontaneously became photorefractory to SD induced by prolonged exposure. SD acclimated hamsters that were transferred back to LD for 1, 2, 3, 4 or 6 weeks, increased arcuate nucleus SOCS3 gene expression to the LD level within 2 weeks, and maintained this higher level thereafter. The early increase of SOCS3 gene expression preceded the LD-induced rise in body weight by approximately 3 weeks. Hamsters kept in SD for an extended period (25 weeks), began to become refractory to SD and to increase body weight. By this time, there was no difference in level of SOCS3 gene expression between LD and SD photoperiods, although body weight was still suppressed in SD hamsters. Finally, we addressed whether SOCS3 gene expression is related to SD-induced gonadal regression or to body weight decrease by comparing Siberian hamsters with Syrian hamsters. The latter exhibited substantial SD-induced gonadal regression but only limited seasonal changes in body weight. Acclimation to either LD or SD for 14 weeks had no effect on SOCS3 gene expression. This implies that arcuate nucleus SOCS3 gene expression is unlikely to be related to seasonal cycles in reproductive activity. Taken together, the findings further strengthen our hypothesis that SOCS3 may be one molecular trigger of seasonal cycles in body weight.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Relojes Biológicos/fisiología , Peso Corporal/fisiología , Fotoperiodo , Estaciones del Año , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Aclimatación/fisiología , Análisis de Varianza , Animales , Relojes Biológicos/genética , Cricetinae , Regulación de la Expresión Génica , Masculino , Mesocricetus , ARN Mensajero/análisis , Estadísticas no Paramétricas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética
11.
J Neuroendocrinol ; 16(11): 922-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15584933

RESUMEN

Ghrelin is an endogenous ligand for the growth hormone secretagogue receptor (GHSR). However, the functional interaction of ligand and receptor is not very well understood. We demonstrate that GHSR mRNA is up-regulated after food deprivation (48 h) in the hypothalamic arcuate nucleus and ventromedial nucleus of the seasonal Siberian hamster, Phodopus sungorus. This increase is accompanied by a two-fold elevation of circulating ghrelin concentration. Chronic changes in feeding state imposed by food restriction over a period of 12 weeks during long day-length induced increased GHSR gene expression, whereas food restriction for 6 weeks had no effect. Phodopus sungorus reveals remarkable seasonal changes in body weight, fat mass and circulating leptin levels. Ghrelin is generally regarded as having opposing effects on appetite and body weight with respect to those exhibited by leptin. However, our study revealed that seasonal adaptations were not accompanied by changes in either GHSR gene expression or circulating ghrelin concentration. Therefore, we suggest that ghrelin only plays a minor role in modulating long-term seasonal body weight cycles. Our findings imply that ghrelin predominantly acts as a short-term regulator of feeding.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Alimentaria/fisiología , Privación de Alimentos/fisiología , Hormonas Peptídicas/sangre , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Adaptación Fisiológica , Animales , Composición Corporal/fisiología , Peso Corporal/fisiología , Cricetinae , Ghrelina , Phodopus , Fotoperiodo , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G/genética , Receptores de Ghrelina , Estaciones del Año , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA