Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Am J Physiol Endocrinol Metab ; 323(3): E319-E332, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35767699

RESUMEN

Osteoglycin (OGN) and lipocalin-2 (LCN2) are hormones that can be secreted by bone and have been linked to glucose homeostasis in rodents. However, the endocrine role of these hormones in humans is contradictory and unclear. We examined the effects of exercise and meal ingestion on circulating serum OGN and LCN2 levels in eight healthy males {age: 28 [25, 30] years [median ± interquartile range (IQR)] and body mass index [BMI]: 24.3 [23.6, 25.5] kg/m2}. In a randomized crossover design, participants ingested a high-glucose (1.1 g glucose/kg body wt) mixed-nutrient meal (45% carbohydrate, 20% protein, and 35% fat) on a rest-control day and 3 and 24 h after aerobic cycling exercise (1 h at 70%-75% V̇o2peak). Acute aerobic exercise increased serum LCN2 levels immediately after exercise (∼61%), which remained elevated 3-h postexercise (∼55%). In contrast, serum OGN remained similar to baseline levels throughout the 3-h postexercise recovery period. The ingestion of a high-glucose mixed-nutrient meal led to a decrease in serum OGN at 90-min (approximately -17%) and 120-min postprandial (approximately -44%), and a decrease in LCN2 at 120-min postprandial (approximately -26%). Compared with the control meal, prior exercise elevated serum OGN and LCN2 levels at 120-min postprandial when the meal was ingested 3-h (OGN: ∼74% and LCN2: ∼68%) and 24-h postexercise (OGN: ∼56% and LCN2: ∼16%). Acute exercise increases serum LCN2 and attenuates the postprandial decrease in OGN and LCN2 following high-glucose mixed-nutrient meal ingestion. The potential endocrine role of circulating OGN and LCN2 in humans warrants further investigation.NEW & NOTEWORTHY We provide novel evidence that OGN and LCN2 decrease 120 min after ingesting a high-glucose mixed-nutrient meal in healthy adults. Acute aerobic exercise increases circulating LCN2 for up to 3-h postexercise, whereas circulating OGN remains similar to baseline. Despite differing postexercise responses, postprandial LCN2 and OGN are elevated when the high-glucose meal is ingested 3-h and 24-h postexercise. Findings support that OGN and LCN2 are dynamically linked to energy homeostasis in humans.


Asunto(s)
Ejercicio Físico , Periodo Posprandial , Adulto , Glucemia/metabolismo , Ingestión de Alimentos , Ejercicio Físico/fisiología , Glucosa , Hormonas , Humanos , Insulina/metabolismo , Lipocalina 2 , Masculino , Nutrientes , Periodo Posprandial/fisiología
2.
Int J Obes (Lond) ; 46(10): 1749-1758, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35794191

RESUMEN

BACKGROUND/OBJECTIVE: Female mice are often excluded from diet-induced obesity studies as they are more resistant to the obesifying effects of a high-fat diet (HFD). However, the underlying mechanisms behind this sex disparity may actually have important implications for the development and management of obesity in humans. Therefore, we systematically investigated the immediate sex-specific effects of transitioning to a HFD in C57BL/6J mice as well as monitored whether these effects are altered after sustained HFD feeding and whether sex affects the response to a return to chow, representative of dieting. METHODS: Dual X-ray absorptiometry (DXA) analysis of body composition, indirect calorimetry measurements, and qPCR analysis of hypothalamic and brainstem regions were performed on male and female C57BL/6J mice. RESULTS: HFD had immediate and dramatic effects in males, increasing fat mass by 58% in the first 3 days. The resistance to the obesifying effect of HFD in females was linked both to an ability to maintain activity levels as well as to an immediate and significantly enhanced reduction in respiratory quotient (RQ), suggesting a greater ability to utilise fat in the diet as a source of fuel. Mechanistically, this sex disparity may be at least partially due to inherent sex differences in the catabolic (POMC/CART) versus anabolic (NPY/AgRP) neurological signalling pathways. Interestingly, the reintroduction of chow following HFD had immediate and consistent responses between the sexes with body composition and most metabolic parameters normalised within 3 days. However, both sexes displayed elevated hypothalamic Npy levels reminiscent of starvation. The difference in RQ seen between the sexes on HFD was immediately abolished suggesting similar abilities to burn fat reserves for fuel. CONCLUSIONS: C57BL/6J mice have markedly different sex-specific behavioural and metabolic responses to the introduction as well as the sustained intake of a HFD, but consistent responses to a dieting situation.


Asunto(s)
Dieta Alta en Grasa , Grasas de la Dieta , Obesidad , Proteína Relacionada con Agouti/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Factores Sexuales
3.
FASEB J ; 35(11): e21980, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34694651

RESUMEN

Although best known for their involvement in modulating nociception, Neuropeptide FF (NPFF) group peptides have been suggested to fulfil a variety of biological functions such as feeding, anxiety behaviors and thermogenesis. However, evidence supporting these functions of NPFF is mostly pharmacological, leaving the physiological relevance unaddressed. Here we examined the physiological impact of lack of NPFF signalling in both genders using a Npff-/- mouse model. NPFF expression in the mouse is restricted to the spinal cord and brainstem while its cognate receptor NPFFR2 has wider distribution throughout the brain. Both male and female Npff-/- mice showed reduced repetitive behaviors evidenced in the marble burying test and self-grooming test. A decrease in anxiety-related behaviors in the Npff-/- mice was also observe in the open field test and to a lesser degree in an elevated plus maze test. Moreover, both male and female Npff-/- mice exhibited increased water intake resulting from increases in drinking size, rather than number of drinking events. During a fasting-refeeding challenge, Npff-/- mice of both genders displayed alterations in reparatory exchange ratio that reflect a greater fuel type flexibility. Npff-/- mice were otherwise wild-type-like regarding body weight, body composition, feeding behaviors, locomotion or energy expenditure. Together, these findings reveal the important physiological roles of NPFF signalling in the regulation of anxiety-related and repetitive behaviors, fluid homeostasis and oxidative fuel selection, highlighting the therapeutical potential of the NPFF system in a number of behavioral and metabolic disorders.


Asunto(s)
Ansiedad/metabolismo , Conducta de Ingestión de Líquido , Oligopéptidos/fisiología , Receptores de Neuropéptido/metabolismo , Animales , Peso Corporal , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Noqueados
4.
Int J Obes (Lond) ; 44(10): 2149-2164, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32152498

RESUMEN

BACKGROUND/OBJECTIVES: Maintaining energy balance is important to ensure a healthy organism. However, energy partitioning, coordinating the distribution of sufficient energy to different organs and tissues is equally important, but the control of this process is largely unknown. In obesity, an increase in fat mass necessitates the production of additional bone mass to cope with the increase in bodyweight and processes need to be in place to communicate this new weight bearing demand. Here, we investigate the interaction between leptin and NPY, two factors critically involved in the regulation of both energy metabolism and bone mass, in this process. METHODS: We assessed the co-localization of leptin receptors on NPY neurons using RNAScope followed by a systematic examination of body composition and energy metabolism profiling in male and female mice lacking leptin receptors specifically in NPY neurons (Leprlox/lox;NPYCre/+). The effect of short-term switching between chow and high-fat diet was also examined in these mice. RESULTS: We uncovered that leptin receptor expression is greater on a subpopulation of NPY neurons in the arcuate that do not express AgRP. We further show that Leprlox/lox;NPYCre/+ mice exhibit significantly increased adiposity while bone mass is diminished. These body composition changes occur in the absence of alterations in food intake or energy expenditure, demonstrating a prominent role for leptin signaling in NPY neurons in the control of energy partitioning. Importantly however, when fed a high-fat diet, these mice display a switch in energy partitioning whereby they exhibit a significantly enhanced ability to increase their bone mass to match the increased bodyweight caused by higher caloric intake concurrent with attenuated adiposity. CONCLUSIONS: Taken together, these results demonstrate that leptin signaling in NPY neurons is critical for coordinating energy partitioning between fat and bone mass especially during situations of changes in energy balance.


Asunto(s)
Tejido Adiposo/metabolismo , Huesos/metabolismo , Metabolismo Energético , Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Adiposidad , Animales , Composición Corporal , Dieta Alta en Grasa , Ingestión de Energía , Femenino , Masculino , Ratones , Receptores de Leptina
5.
Nat Med ; 13(11): 1333-40, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17982462

RESUMEN

Anorexia and weight loss are part of the wasting syndrome of late-stage cancer, are a major cause of morbidity and mortality in cancer, and are thought to be cytokine mediated. Macrophage inhibitory cytokine-1 (MIC-1) is produced by many cancers. Examination of sera from individuals with advanced prostate cancer showed a direct relationship between MIC-1 abundance and cancer-associated weight loss. In mice with xenografted prostate tumors, elevated MIC-1 levels were also associated with marked weight, fat and lean tissue loss that was mediated by decreased food intake and was reversed by administration of antibody to MIC-1. Additionally, normal mice given systemic MIC-1 and transgenic mice overexpressing MIC-1 showed hypophagia and reduced body weight. MIC-1 mediates its effects by central mechanisms that implicate the hypothalamic transforming growth factor-beta receptor II, extracellular signal-regulated kinases 1 and 2, signal transducer and activator of transcription-3, neuropeptide Y and pro-opiomelanocortin. Thus, MIC-1 is a newly defined central regulator of appetite and a potential target for the treatment of both cancer anorexia and weight loss, as well as of obesity.


Asunto(s)
Anorexia/metabolismo , Citocinas/fisiología , Familia de Multigenes/inmunología , Neoplasias de la Próstata/metabolismo , Pérdida de Peso , Animales , Anorexia/genética , Anorexia/inmunología , Anorexia/fisiopatología , Anticuerpos/administración & dosificación , Anticuerpos/fisiología , Línea Celular Tumoral , Citocinas/sangre , Citocinas/genética , Citocinas/inmunología , Factor 15 de Diferenciación de Crecimiento , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/fisiopatología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/fisiología , Pérdida de Peso/genética , Pérdida de Peso/inmunología
6.
Mol Metab ; 76: 101790, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37562743

RESUMEN

OBJECTIVE: One of leptin's main targets in the hypothalamus are neuropeptide Y (NPY) neurons, with selective deletion of leptin receptors (Lepr) specifically in Npy neurons resulting in major alterations of energy partitioning between fat and bone mass. However, the specific action of these Npy+/Lepr+ neurons compared to Npy-negative Lepr (Npy-/Lepr+) neurons in regard to energy homeostasis regulation is unknown. METHODS: Specific AAV viral vectors were generated using DREADD and INTRSECT technology and used in male LeprCre/+ and LeprCre/+;NpyFlp/+ mice to assess the effect of activating either all Lepr neurons or specifically Npy+/Lepr+ or Npy-/Lepr+ neurons only on feeding, energy homeostasis control, and body composition. RESULTS: Selective stimulation of Npy+/Lepr+ neurons led to an immediate decrease in respiratory quotient followed by a delayed increase in food intake in standard chow fed, but interestingly not in high fat diet (HFD) fed mice. In addition, stimulation of Npy+/Lepr+ neurons led to a robust increase in brown adipose tissue thermogenesis and improved glucose tolerance. These effects were not observed in standard chow fed mice when Npy-/Lepr+ expressing neurons were specifically activated, suggesting the effects of leptin on these parameters are driven by NPY. However, under HFD condition when leptin levels are elevated, the stimulation of the Npy-/Lepr+ neurons increased food intake, physical activity and energy expenditure. Interestingly, chronic stimulation of Npy-positive Lepr neurons was able to increase bone mass independently of bodyweight, whilst chronic stimulation of the Npy-/Lepr+ neurons resulted in increased bodyweight and fat mass with proportionate increases in bone mass. CONCLUSIONS: Together, these data indicate that leptin signalling through Npy-positive Lepr-expressing neurons controls energy partitioning via stimulation of thermogenesis, energy expenditure, and the use of fat as a fuel source. However, under prolonged HFD, leptin resistance may occur and actions of leptin signalling through Npy-negative Lepr hypothalamic neurons may exacerbate excess food intake.


Asunto(s)
Leptina , Neuropéptido Y , Ratones , Masculino , Animales , Leptina/metabolismo , Neuropéptido Y/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Neuronas/metabolismo , Metabolismo Energético
7.
Cell Metab ; 35(6): 979-995.e7, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37201523

RESUMEN

Neuropeptide Y (NPY) in the arcuate nucleus (ARC) is known as one of the most critical regulators of feeding. However, how NPY promotes feeding under obese conditions is unclear. Here, we show that positive energy balance, induced by high-fat diet (HFD) or in genetically obese leptin-receptor-deficient mice, leads to elevated Npy2r expression especially on proopiomelanocortin (POMC) neurons, which also alters leptin responsiveness. Circuit mapping identified a subset of ARC agouti-related peptide (Agrp)-negative NPY neurons that control these Npy2r expressing POMC neurons. Chemogenetic activation of this newly discovered circuitry strongly drives feeding, while optogenetic inhibition reduces feeding. Consistent with that, lack of Npy2r on POMC neurons leads to reduced food intake and fat mass. This suggests that under energy surplus conditions, when ARC NPY levels generally drop, high-affinity NPY2R on POMC neurons is still able to drive food intake and enhance obesity development via NPY released predominantly from Agrp-negative NPY neurons.


Asunto(s)
Leptina , Proopiomelanocortina , Ratones , Animales , Leptina/metabolismo , Proopiomelanocortina/metabolismo , Neuropéptido Y/metabolismo , Proteína Relacionada con Agouti/metabolismo , Neuronas/metabolismo , Núcleo Arqueado del Hipotálamo , Obesidad/metabolismo
8.
Mol Metab ; 59: 101455, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35167990

RESUMEN

OBJECTIVE: Aguti-related protein (AGRP) neurons in the arcuate nucleus of the hypothalamus (ARC), which co-express neuropeptide Y (NPY), are key regulators of feeding and energy homeostasis. However, the precise role NPY has within these neurons and the specific pathways that it control are still unclear. In this article, we aimed to determine what aspects of feeding behaviour and energy homeostasis are controlled by NPY originating from AGRP neurons and which Y-receptor pathways are utilised to fulfil this function. METHODS: Novel conditional Agrpcre/+;Npylox/lox knockout mice were generated and comprehensively phenotyped, both under standard chow as well as high-fat-diet conditions. Designer receptor exclusively activated by designer drugs (DREADD) technology was used to assess the altered responses on feeding and energy homeostasis control in the absence of NPY in these neurons. Rescue experiments utilising Npy1r- and Npy2r-selective NPY ligands were performed to assess which component of the energy homeostasis control is dependent by which specific Y-receptor pathway. RESULTS: We show that the specific deletion of Npy only in AGRP neurons leads to a paradoxical mild obese phenotype associated with reduced locomotion and energy expenditure and increased feeding and Respiratory Quotient (RQ) that remain elevated under a positive energy balance. The activation of Npy-deficient AGRP neurons via DREADD's is still able to drive feeding, yet with a delayed onset. Additionally, Clozapine-N-oxide (CNO) treatment reduces locomotion without impacting on energy expenditure. Rescue experiments re-introducing Npy1r- and Npy2r-selective NPY ligands revealed that the increased feeding and RQ are mostly driven by Npy1r, whereas energy expenditure and locomotion are controlled by Npy2r signalling. CONCLUSION: Together, these results demonstrate that NPY originating from AGRP neurons is not only critical to initiate but also for continuously driving feeding, and we for the first time identify which Y-receptor controls which pathway.


Asunto(s)
Metabolismo Energético , Neuropéptido Y , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Ligandos , Ratones , Neuronas/metabolismo , Neuropéptido Y/metabolismo
9.
J Neuroendocrinol ; 33(5): e12978, 2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33913541

RESUMEN

The central nervous system is an active and major regulator of bone structure and remodelling. Specifically, signalling within the hypothalamus has been shown to be critical to ensuring that skeletal functions align with whole body metabolic supply and demand. Here, we identify agouti-related peptide (AgRP), an orexigenic peptide exclusively co-expressed with neuropeptide Y (NPY) in the arcuate nucleus (ARC) of the hypothalamus, as another critical player in the central control of bone homeostasis. Using novel mouse models, we show that AgRP deletion leads to an increase in cortical and trabecular bone mass as a result of an increase in bone thickness despite a lean phenotype, particularly in male mice. Interestingly, male AgRP deficient mice display a significant decrease in pro-opiomelanocortin (POMC) expression in the ARC, but no change in NPY or CART expression, suggesting that the increase in bone mass in AgRP-deficient mice is unlikely to be a result of altered NPY signalling. This is consistent with the observation that bone mass is unchanged in response to the specific deletion of NPY from AgRP expressing neurones. By contrast, POMC expression in the ARC is significantly increased in female AgRP deficient mice, although AgRP deletion results in altered respiratory exchange ratio regulation in response to re-feeding after a fast in both sexes. Taken together, the present study identifies AgRP as being directly involved in the regulation of bone mass and highlights the complexity intrinsic to the neuropeptide regulation of the skeleton.

10.
J Neuroendocrinol ; 33(4): e12925, 2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33427385

RESUMEN

Insulin is a major contributor to many important physiological processes. Although its function in the periphery has been studied in detail, the contributions that it makes to functions in the brain are far less understood. The neuropeptide Y (NPY) neurones comprise a major target of insulin in the brain and are inhibited by its action. In particular, NPY neurones in the arcuate nucleus of the hypothalamus are critical control centres for insulin's central action on control energy homeostasis, as well as glucose homeostasis regulation. However, the colocalisation of insulin receptors with NPY neurones is also found in many other brain areas, although very little is known about their interactions and control functions. In this review, we explore the recent advances that have been made to further the understanding of the hypothalamic insulin receptor-NPY network, as well as provide insights from other lesser explored areas, such as the amygdala and hippocampus. We will also look at the peripheral interaction of the NPY system with insulin release, thereby closing the loop between these two energy and glucose homeostasis controlling systems and highlighting the critical interaction points that may be dysregulated in conditions of obesity and diabetes.

11.
Neuropeptides ; 80: 101994, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31740068

RESUMEN

Neuropeptide Y (NPY) producing neurons in the arcuate nucleus (Arc) of the hypothalamus are essential to the regulation of food intake and energy homeostasis. Whilst they have classically been thought to co-express agouti-related peptide (AgRP), it is now clear that there is a sub-population of NPY neurons in the Arc that do not. Here, we show that a subset of AgRP-negative, NPY-positive neurons in the Arc also express neurotensin (NTS) and we use an NTS-Cre line to investigate the function of this sub-population of NPY neurons. The lack of NPY in NTS-positive neurons led to a marked reduction in fat mass and bodyweight as well as a significant reduction in food intake in male NPYlox/lox; NTScre/+ mice compared to controls. Despite the reduction in food intake, overall energy expenditure was similar between genotypes due to concomitant reduction in activity in NPYlox/lox; NTScre/+ mice. Furthermore, cortical bone mass was significantly reduced in NPYlox/lox;NTScre/+ mice with no evident alterations in the cancellous bone compartment, likely due to reduced leptin levels as a result of their reduced adiposity. Taken together, these data suggest that the sub-population of Arc NPY neurons expressing NTS are critical for regulating food intake, activity and fat mass but are not directly involved in the control of bone mass.


Asunto(s)
Peso Corporal/fisiología , Metabolismo Energético/fisiología , Neuronas/metabolismo , Neuropéptido Y/deficiencia , Neurotensina/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Homeostasis/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones Transgénicos , Neuropéptido Y/metabolismo , Fenotipo
12.
J Neuroendocrinol ; 31(2): e12687, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30633834

RESUMEN

The RANKL pathway is known to be an important aspect of the pathogenesis of oestrogen deficiency-induced bone loss. RANK deletion specifically in neuropeptide Y (NPY) neurones has been shown to enhance the ability of the skeleton to match increases in body weight caused by high-fat diet feeding, likely via the modulation of NPY levels. In the present study, we used ovariectomy in female mice to show that RANK deletion in NPY neurones attenuates bone loss caused by long-term oestrogen deficiency, particularly in the vertebral compartment. Ovariectomy led to a reduction in NPY expression levels in the arcuate nucleus of NPYcre/+ ;RANKlox/lox mice compared to NPYcre/+ ;RANKlox/+ controls. Because NPY deficient mice also displayed a similar protection against ovariectomy-induced bone loss, modulation of hypothalamic NPY signalling is the likely mechanism behind the protection from bone loss in the NPYcre/+ ;RANKlox/lox mice.


Asunto(s)
Resorción Ósea/metabolismo , Estrógenos/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Femenino , Ratones Noqueados , Ovariectomía , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal
13.
Cell Metab ; 30(1): 111-128.e6, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31031093

RESUMEN

Neuropeptide Y (NPY) exerts a powerful orexigenic effect in the hypothalamus. However, extra-hypothalamic nuclei also produce NPY, but its influence on energy homeostasis is unclear. Here we uncover a previously unknown feeding stimulatory pathway that is activated under conditions of stress in combination with calorie-dense food; NPY neurons in the central amygdala are responsible for an exacerbated response to a combined stress and high-fat-diet intervention. Central amygdala NPY neuron-specific Npy overexpression mimics the obese phenotype seen in a combined stress and high-fat-diet model, which is prevented by the selective ablation of Npy. Using food intake and energy expenditure as readouts, we demonstrate that selective activation of central amygdala NPY neurons results in increased food intake and decreased energy expenditure. Mechanistically, it is the diminished insulin signaling capacity on central amygdala NPY neurons under combined stress and high-fat-diet conditions that leads to the exaggerated development of obesity.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Obesidad/metabolismo , Animales , Temperatura Corporal , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/fisiología , Electrofisiología , Metabolismo Energético/fisiología , Inmunohistoquímica , Hibridación Fluorescente in Situ , Insulina/metabolismo , Masculino , Ratones , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Neuropeptides ; 42(1): 19-30, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18164057

RESUMEN

The gut-derived hormone, peptide YY (PYY) reduces food intake and enhances satiety in both humans and animals. Obese individuals also have a deficiency in circulating peptide YY, although whether this is a cause or a consequence of obesity is unclear. Our aims were to determine whether peptide YY (PYY) over-expression may have therapeutic effects for the treatment of obesity by altering energy balance and glucose homeostasis. We generated PYY transgenic mice and measured body weight, food intake, temperature, adiposity, glucose tolerance, circulating hormone and lipid concentrations and hypothalamic neuropeptide levels (neuropeptide Y; proopiomelanocortin, and thyrotropin-releasing hormone) under chow and high-fat feeding and after crossing these mice onto the genetically obese leptin-deficient ob/ob mouse background. PYY transgenic mice were protected against diet-induced obesity in association with increased body temperature (indicative of increased thermogenesis) and sustained expression of thyrotropin-releasing hormone in the paraventricular nucleus of the hypothalamus. Moreover, PYY transgenic mice crossed onto the genetically obese ob/ob background had significantly decreased weight gain and adiposity, reduced serum triglyceride levels and improved glucose tolerance compared to ob/ob controls. There was no effect of PYY transgenic over expression on basal or fasting-induced food intake measured at 11-12 weeks of age. Together, these findings suggest that long-term administration of PYY, PYY-like compounds or agents that stimulate PYY synthesis in vivo can reduce excess adiposity and improve glucose tolerance, possibly via effects on the hypothalamo-pituitary-thyroid axis and thermogenesis.


Asunto(s)
Obesidad/etiología , Obesidad/genética , Péptido YY/genética , Péptido YY/fisiología , Adiposidad/genética , Animales , Peso Corporal/genética , Dieta , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis/genética , Homeostasis/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Hibridación in Situ , Leptina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleo Hipotalámico Paraventricular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Termogénesis/genética , Termogénesis/fisiología , Glándula Tiroides/fisiología , Tirotropina/biosíntesis , Tirotropina/genética
15.
Nutrition ; 24(9): 892-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18662863

RESUMEN

OBJECTIVE: Neuropeptide Y regulates numerous processes including food intake, body composition, and reproduction by at least five different Y receptors. We previously demonstrated a synergistic interaction between Y2 and Y4 receptors in reducing adiposity in chow- or fat-fed Y2Y4-receptor double-knockout mice. In the present study, we investigated whether this synergy could reduce the massive obesity of leptin-deficient ob/ob mice. METHODS: Mice with germline deletions of Y2 and Y4 receptors were crossed onto the ob/ob strain. Body weight was measured weekly until 15-18 wk of age before decapitation for collection of trunk blood and tissues. RESULTS: Male and female Y24ob triple mutants showed highly significant reductions in body weight and white adipose tissue mass compared with ob/ob mice. This reduction in body weight was not evident in Y2ob or Y4ob double mutants, and the effect on adiposity was significantly greater than that seen in Y2ob or Y4ob mice. These changes were associated with significant attenuation of the increased brown adipose tissue mass and small intestinal hypertrophy seen in ob/ob mice and with normalization of the low circulating free thyroxine concentrations seen in female ob/ob mice and the high circulating corticosterone concentrations seen in male ob/ob mice. CONCLUSION: These data reveal a synergistic interaction between Y2 and Y4 receptors in attenuating the massive obesity of ob/ob mice, possibly mediated by stimulation of thyroid function and inhibition of intestinal nutrient absorption. Dual pharmacologic antagonism of Y2 and Y4 receptors could help people to attain and maintain a healthy weight.


Asunto(s)
Obesidad/metabolismo , Obesidad/prevención & control , Receptores de Neuropéptido Y/metabolismo , Tejido Adiposo/metabolismo , Animales , Peso Corporal , Densidad Ósea , Modelos Animales de Enfermedad , Femenino , Fémur , Masculino , Ratones , Ratones Noqueados
16.
Mol Endocrinol ; 21(7): 1722-35, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17456788

RESUMEN

Endogenous opioids, particularly dynorphins, have been implicated in regulation of energy balance, but it is not known how they mediate this in vivo. We investigated energy homeostasis in dynorphin knockout mice (Dyn(-/-) mice) and probed the interactions between dynorphins and the neuropeptide Y (NPY) system. Dyn(-/-) mice were no different from wild types with regards to body weight and basal and fasting-induced food intake, but fecal output was increased, suggesting decreased nutrient absorption, and they had significantly less white fat and lost more weight during a 24-h fast. The neuroendocrine and thermal responses to fasting were at least as pronounced in Dyn(-/-) as in wild types, and there was no stimulatory effect of dynorphin knockout on 24-h energy expenditure (kilocalories of heat produced) or physical activity. However, Dyn(-/-) mice showed increased circulating concentrations of 3,4-dihydroxyphenlacetic acid and 3,4-dihydroxyphenylglycol, suggesting increased activity of the sympathetic nervous system. The respiratory exchange ratio of male but not female Dyn(-/-) mice was reduced, demonstrating increased fat oxidation. Interestingly, expression of the orexigenic acting NPY in the hypothalamic arcuate nucleus was reduced in Dyn(-/-) mice. However, fasting-induced increases in pre-prodynorphin expression in the arcuate nucleus, the paraventricular nucleus, and the ventromedial hypothalamus but not the lateral hypothalamus were abolished by deletion of Y(1) but not Y(2) receptors. Therefore, ablation of dynorphins results in increases in fatty acid oxidation in male mice, reductions in adiposity, and increased weight loss during fasting, possibly via increases in sympathetic activity, decreases in intestinal nutrient absorption, and interactions with the NPYergic system.


Asunto(s)
Dinorfinas/deficiencia , Dinorfinas/genética , Pérdida de Peso/fisiología , Tejido Adiposo/patología , Animales , Peso Corporal/fisiología , Dinorfinas/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético , Ayuno/metabolismo , Femenino , Glucosa/metabolismo , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptido Y/genética , Sistemas Neurosecretores/fisiología , Esfuerzo Físico , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
Curr Osteoporos Rep ; 6(2): 62-6, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18778565

RESUMEN

Leptin and its actions in bone came to prominence in 2000, with the publication of two landmark articles identifying a novel interaction between energy and bone homeostasis, as well as a novel hypothalamic circuit to the skeleton. However, they also revealed the dichotomous nature of leptin's effect on the skeleton. Subsequent research has increased understanding of the factors critical to interpretation of the leptin-bone signaling. These include opposing effects in cortical and cancellous bone, central and peripheral effects, involvement of other neural and endocrine factors, and leptin receptor polymorphisms in human populations. It is clear that leptin can markedly influence the regulation of bone mass, and that study of this pathway continues to increase our knowledge of the biology of skeletal tissue and its interactions with other tissues. However, this relationship is complex and requires careful interpretation.


Asunto(s)
Densidad Ósea/fisiología , Huesos/fisiología , Leptina/fisiología , Animales , Densidad Ósea/genética , Humanos , Modelos Animales , Neuropéptido Y/fisiología , Polimorfismo Genético , Receptores de Leptina/genética
18.
Bone ; 106: 167-178, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26055106

RESUMEN

Brown adipose tissue (BAT), largely controlled by the sympathetic nervous system (SNS), has the ability to dissipate energy in the form of heat through the actions of uncoupling protein-1 (UCP-1), thereby critically influencing energy expenditure. Besides BAT, the SNS also strongly influences bone, and recent studies have demonstrated a positive correlation between BAT activity and bone mass, albeit the interactions between BAT and bone remain unclear. Here we show that UCP-1 is critical for protecting bone mass in mice under conditions of permanent mild cold stress for this species (22°C). UCP-1-/- mice housed at 22°C showed significantly lower cancellous bone mass, with lower trabecular number and thickness, a lower bone formation rate and mineralising surface, but unaltered osteoclast number, compared to wild type mice housed at the same temperature. UCP-1-/- mice also displayed shorter femurs than wild types, with smaller cortical periosteal and endocortical perimeters. Importantly, these altered bone phenotypes were not observed when UCP-1-/- and wild type mice were housed in thermo-neutral conditions (29°C), indicating a UCP-1 dependent support of bone mass and bone formation at the lower temperature. Furthermore, at 22°C UCP-1-/- mice showed elevated hypothalamic expression of neuropeptide Y (NPY) relative to wild type, which is consistent with the lower bone formation and mass of UCP-1-/- mice at 22°C caused by the catabolic effects of hypothalamic NPY-induced SNS modulation. The results from this study suggest that during mild cold stress, when BAT-dependent thermogenesis is required, UCP-1 activity exerts a protective effect on bone mass possibly through alterations in central NPY pathways known to regulate SNS activity.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteína Desacopladora 1/metabolismo , Animales , Western Blotting , Composición Corporal/fisiología , Peso Corporal/fisiología , Calorimetría Indirecta , Frío , Metabolismo Energético/fisiología , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Neuropéptido Y/metabolismo , Proteína Desacopladora 1/genética , Microtomografía por Rayos X
19.
Nat Commun ; 9(1): 4722, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413707

RESUMEN

Excess caloric intake results in increased fat accumulation and an increase in energy expenditure via diet-induced adaptive thermogenesis; however, the underlying mechanisms controlling these processes are unclear. Here we identify the neuropeptide FF receptor-2 (NPFFR2) as a critical regulator of diet-induced thermogenesis and bone homoeostasis. Npffr2-/- mice exhibit a stronger bone phenotype and when fed a HFD display exacerbated obesity associated with a failure in activating brown adipose tissue (BAT) thermogenic response to energy excess, whereas the activation of cold-induced BAT thermogenesis is unaffected. NPFFR2 signalling is required to maintain basal arcuate nucleus NPY mRNA expression. Lack of NPFFR2 signalling leads to a decrease in BAT thermogenesis under HFD conditions with significantly lower UCP-1 and PGC-1α levels in the BAT. Together, these data demonstrate that NPFFR2 signalling promotes diet-induced thermogenesis via a novel hypothalamic NPY-dependent circuitry thereby coupling energy homoeostasis with energy partitioning to adipose and bone tissue.


Asunto(s)
Dieta , Receptores de Neuropéptido/metabolismo , Transducción de Señal , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Animal , Huesos/metabolismo , Frío , Metabolismo Energético , Femenino , Homeostasis , Ligandos , Masculino , Ratones Noqueados , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Neuropéptido/deficiencia , Receptores de Neuropéptido/genética
20.
Endocrinology ; 148(5): 2056-65, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17272395

RESUMEN

Neuropeptide Y, a neuropeptide abundantly expressed in the brain, has been implicated in the regulation of the hypothalamo-pituitary-somatotropic axis and the hypothalamo-pituitary-gonadotropic axis. Elevated hypothalamic neuropeptide Y expression, such as that occurs during fasting, is known to inhibit both of these axes. However, it is not known which Y receptor(s) mediate these effects. Here we demonstrate, using Y receptor knockout mice, that Y2 and Y4 receptors are separately involved in the regulation of these axes. Fasting-induced inhibition of hypothalamic GHRH mRNA expression and reduction of circulating IGF-I levels were observed in wild-type and Y4(-/-) mice but not Y2(-/-) or Y2(-/-)Y4(-/-) mice. In contrast, fasting-induced reduction of GnRH expression in the medial preoptic area and testis testosterone content were abolished in the absence of Y4 receptors. Colocalization of Y2 receptors and GHRH in the arcuate nucleus (Arc) suggests that GHRH mRNA expression in this region might be directly regulated by Y2 receptors. Indeed, hypothalamic-specific deletion of Y2 receptors in conditional knockout mice prevented the fasting-induced reduction in Arc GHRH mRNA expression. On the other hand, fasting-induced decrease in GnRH mRNA expression in the medial preoptic area is more likely indirectly influenced by Y4 receptors because no Y4 receptors could be detected on GnRH neurons in this region. Together these data show that fasting inhibits the somatotropic axis via direct action on Y2 receptors in the Arc and indirectly inhibits the gonadotropic axis via Y4 receptors.


Asunto(s)
Ayuno/fisiología , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario/fisiología , Receptores de Neuropéptido Y/metabolismo , Reproducción/fisiología , Animales , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/fisiología , Gónadas/crecimiento & desarrollo , Gónadas/fisiología , Hormona del Crecimiento/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipófisis/crecimiento & desarrollo , Hipófisis/fisiología , Área Preóptica/crecimiento & desarrollo , Área Preóptica/fisiología , ARN Mensajero/metabolismo , Receptores de Neuropéptido/genética , Receptores de Neuropéptido Y/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/fisiología , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA