Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
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
2.
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
3.
FASEB J ; 35(4): e21435, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749879

RESUMEN

Peptide YY (PYY), produced by endocrine L cells in the gut, is known for its critical role in regulating gastrointestinal functions as well as satiety. However, how these processes are integrated with maintaining a healthy gut microbiome composition is unknown. Here, we show that lack of PYY in mice leads to distinct changes in gut microbiome composition that are diet-dependent. While under chow diet only slight differences in gut microbiome composition could be observed, high-fat diet (HFD) aggravated these differences. Specifically an increased abundance of the Bacteroidetes phylum with a corresponding decrease of the Firmicutes/Bacteroidetes ratio could be detected in Pyy-knockout (KO) mice in response to HFD. Detailed analysis of the Bacteroidetes phylum further revealed that the Alistipes genus belonging to the Rikenellaceae family, the Parabacteroides belonging to the Tannerellaceae family, as well as Muribaculum were increased in Pyy-KO mice. In order to investigate whether these changes are associated with changed markers of gut barrier and immunity, we analyzed the colonic expression of various pro-inflammatory cytokines, as well as tight junction proteins and mucin 2, and identified increased mRNA expression of the tight junction proteins Cldn2 and Ocel1 in Pyy-KO mice, while pro-inflammatory cytokine expression was not significantly altered. Together these results highlight a critical gene-environment interaction between diet and the gut microbiome and its impact on homeostasis of the intestinal epithelium under conditions of reduced PYY signaling which is commonly seen under obese conditions.


Asunto(s)
Bacterias/clasificación , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal , Péptido YY/metabolismo , Animales , Composición Corporal , Ratones , Ratones Noqueados , Péptido YY/genética
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.
Calcif Tissue Int ; 107(5): 510-523, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32804252

RESUMEN

The maintenance of whole body energy homeostasis is critical to survival and mechanisms exist whereby an organism can adapt to its environment and the stresses placed upon it. Environmental temperature and thermogenesis are key components known to affect energy balance. However, little is known about how these processes are balanced against the overall energy balance. We show that even mild cold exposure has a significant effect on energy expenditure and UCP-1 levels which increase by 43% and 400%, respectively, when wild-type (WT) mice at thermoneutral (29 °C) were compared to mice at room temperature (22 °C) conditions. Interestingly, bone mass was lower in cold-stressed WT mice with significant reductions in femoral bone mineral content (- 19%) and bone volume (- 13%). Importantly, these cold-induced skeletal changes were absent in mice lacking NPY, one of the main controllers of energy homeostasis, highlighting the critical role of NPY in this process. However, energy expenditure was significantly greater in cold-exposed NPY null mice, indicating that suppression of non-thermogenic tissues, like bone, contributes to the adaptive responses to cold exposure. Altogether, this work identifies NPY as being crucial in coordinating energy and bone homeostasis where it suppresses energy expenditure, UCP-1 levels and lowers bone mass under conditions of cold exposure.


Asunto(s)
Densidad Ósea , Frío , Metabolismo Energético , Neuropéptido Y , Animales , Homeostasis , Ratones , Neuropéptido Y/genética , Proteína Desacopladora 1
6.
J Biol Chem ; 293(15): 5731-5745, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29440390

RESUMEN

Obesity is associated with metabolic dysfunction, including insulin resistance and hyperinsulinemia, and with disorders such as cardiovascular disease, osteoporosis, and neurodegeneration. Typically, these pathologies are examined in discrete model systems and with limited temporal resolution, and whether these disorders co-occur is therefore unclear. To address this question, here we examined multiple physiological systems in male C57BL/6J mice following prolonged exposure to a high-fat/high-sucrose diet (HFHSD). HFHSD-fed mice rapidly exhibited metabolic alterations, including obesity, hyperleptinemia, physical inactivity, glucose intolerance, peripheral insulin resistance, fasting hyperglycemia, ectopic lipid deposition, and bone deterioration. Prolonged exposure to HFHSD resulted in morbid obesity, ectopic triglyceride deposition in liver and muscle, extensive bone loss, sarcopenia, hyperinsulinemia, and impaired short-term memory. Although many of these defects are typically associated with aging, HFHSD did not alter telomere length in white blood cells, indicating that this diet did not generally promote all aspects of aging. Strikingly, glucose homeostasis was highly dynamic. Glucose intolerance was evident in HFHSD-fed mice after 1 week and was maintained for 24 weeks. Beyond 24 weeks, however, glucose tolerance improved in HFHSD-fed mice, and by 60 weeks, it was indistinguishable from that of chow-fed mice. This improvement coincided with adaptive ß-cell hyperplasia and hyperinsulinemia, without changes in insulin sensitivity in muscle or adipose tissue. Assessment of insulin secretion in isolated islets revealed that leptin, which inhibited insulin secretion in the chow-fed mice, potentiated glucose-stimulated insulin secretion in the HFHSD-fed mice after 60 weeks. Overall, the excessive calorie intake was accompanied by deteriorating function of numerous physiological systems.


Asunto(s)
Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Enfermedades Metabólicas , Sacarosa/efectos adversos , Homeostasis del Telómero/efectos de los fármacos , Animales , Carbohidratos de la Dieta/farmacología , Grasas de la Dieta/farmacología , Masculino , Enfermedades Metabólicas/inducido químicamente , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Ratones , Sacarosa/farmacología , Factores de Tiempo
7.
Int J Obes (Lond) ; 42(11): 1925-1938, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29523877

RESUMEN

OBJECTIVE: To determine whether age and neuropeptide Y (NPY) were involved in the skeletal response to extended periods of diet-induced obesity. METHODS: Male wild-type (WT) and NPY null (NPYKO) mice were fed a mild (23% fat) high-fat diet for 10 weeks from 6 or 16 weeks of age. Metabolism and bone density were assessed during feeding. Skeletal changes were assessed by microCT and histomorphometry. RESULTS: High-fat feeding in 6-week-old WT mice led to significantly increased body weight, adiposity and serum leptin levels, accompanied with markedly suppressed cortical bone accrual. NPYKO mice were less susceptible to fat accrual but, importantly, displayed a complete lack of suppression of bone accrual or cortical bone loss. In contrast, when skeletally mature (16 week old) mice underwent 10 weeks of fat feeding, the metabolic response to HFD was similar to younger mice, however bone mass was not affected in either WT or NPYKO. Thus, growing mice are particularly susceptible to the detrimental effects of HFD on bone mass, through suppression of bone accrual involving NPY signalling. CONCLUSION: This study provides new insights into the relationship between the opposing processes of a positive weight/bone relationship and the negative 'metabolic' effect of obesity on bone mass. This negative effect is particularly active in growing skeletons, which have heightened sensitivity to changes in obesity. In addition, NPY is identified as a fundamental driver of this negative 'metabolic' pathway to bone.


Asunto(s)
Remodelación Ósea/fisiología , Hueso Cortical/patología , Neuropéptido Y/deficiencia , Obesidad/patología , Aumento de Peso/fisiología , Animales , Densidad Ósea , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos , Neuropéptido Y/fisiología , Obesidad/metabolismo
8.
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
9.
Adv Sci (Weinh) ; 11(23): e2307963, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38602451

RESUMEN

In recent decades, the role of tumor biomechanics on cancer cell behavior at the primary site has been increasingly appreciated. However, the effect of primary tumor biomechanics on the latter stages of the metastatic cascade, such as metastatic seeding of secondary sites and outgrowth remains underappreciated. This work sought to address this in the context of triple negative breast cancer (TNBC), a cancer type known to aggressively disseminate at all stages of disease progression. Using mechanically tuneable model systems, mimicking the range of stiffness's typically found within breast tumors, it is found that, contrary to expectations, cancer cells exposed to softer microenvironments are more able to colonize secondary tissues. It is shown that heightened cell survival is driven by enhanced metabolism of fatty acids within TNBC cells exposed to softer microenvironments. It is demonstrated that uncoupling cellular mechanosensing through integrin ß1 blocking antibody effectively causes stiff primed TNBC cells to behave like their soft counterparts, both in vitro and in vivo. This work is the first to show that softer tumor microenvironments may be contributing to changes in disease outcome by imprinting on TNBC cells a greater metabolic flexibility and conferring discrete cell survival advantages.


Asunto(s)
Ácidos Grasos , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Humanos , Femenino , Ácidos Grasos/metabolismo , Ratones , Línea Celular Tumoral , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Metástasis de la Neoplasia
10.
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
11.
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
12.
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
13.
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.

14.
Nat Commun ; 12(1): 2622, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976180

RESUMEN

Obesity is caused by an imbalance between food intake and energy expenditure (EE). Here we identify a conserved pathway that links signalling through peripheral Y1 receptors (Y1R) to the control of EE. Selective antagonism of peripheral Y1R, via the non-brain penetrable antagonist BIBO3304, leads to a significant reduction in body weight gain due to enhanced EE thereby reducing fat mass. Specifically thermogenesis in brown adipose tissue (BAT) due to elevated UCP1 is enhanced accompanied by extensive browning of white adipose tissue both in mice and humans. Importantly, selective ablation of Y1R from adipocytes protects against diet-induced obesity. Furthermore, peripheral specific Y1R antagonism also improves glucose homeostasis mainly driven by dynamic changes in Akt activity in BAT. Together, these data suggest that selective peripheral only Y1R antagonism via BIBO3304, or a functional analogue, could be developed as a safer and more effective treatment option to mitigate diet-induced obesity.


Asunto(s)
Arginina/análogos & derivados , Obesidad/prevención & control , Receptores de Neuropéptido Y/antagonistas & inhibidores , Termogénesis/efectos de los fármacos , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Adulto , Animales , Arginina/farmacología , Arginina/uso terapéutico , Biopsia , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Obesidad/etiología , Obesidad/metabolismo , Cultivo Primario de Células , Receptores de Neuropéptido Y/metabolismo
15.
Am J Physiol Regul Integr Comp Physiol ; 299(6): R1618-28, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20881101

RESUMEN

Neuropeptide Y receptors are critical regulators of energy homeostasis and are well known for their powerful influence on feeding, but their roles in other important aspects of energy homeostasis, such as energy expenditure and their functional interactions in these processes, are largely unknown. Here we show that mice lacking both Y2 and Y4 receptors exhibited a reduction in adiposity, more prominent in intra-abdominal vs. subcutaneous fat, and an increase in lean mass as determined by dual-energy X-ray absorptiometry. These changes were more pronounced than those seen in mice with Y2 or Y4 receptor single deletion, demonstrating the important roles and synergy of Y2 and Y4 signaling in the regulation of body composition. These changes in body composition occurred without significant changes in food intake, but energy expenditure and physical activity were significantly increased in Y4(-/-) and particularly in Y2(-/-)Y4(-/-) but not in Y2(-/-) mice, suggesting a critical role of Y4 signaling and synergistic interactions with Y2 signaling in the regulation of energy expenditure and physical activity. Y2(-/-) and Y4(-/-) mice also exhibited a decrease in respiratory exchange ratio with no further synergistic decrease in Y2(-/-)Y4(-/-) mice, suggesting that Y2 and Y4 signaling each play important and independent roles in the regulation of substrate utilization. The synergy between Y2 and Y4 signaling in regulating fat mass may be related to differences in mitochondrial oxidative capacity, since Y2(-/-)Y4(-/-) but not Y2(-/-) or Y4(-/-) mice showed significant increases in muscle protein levels of peroxisome proliferator-activated receptor (PPAR)γ coactivator (PGC)-1α, and mitochondrial respiratory chain complexes I and III. Taken together, this work demonstrates the critical roles of Y2 and Y4 receptors in the regulation of body composition and energy metabolism, highlighting dual antagonism of Y2 and Y4 receptors as a potentially effective anti-obesity treatment.


Asunto(s)
Metabolismo Energético/fisiología , Actividad Motora/fisiología , Receptores de Neuropéptido Y/metabolismo , Absorciometría de Fotón , Análisis de Varianza , Animales , Western Blotting , Composición Corporal/fisiología , Calorimetría Indirecta , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Noqueados , Receptores de Neuropéptido Y/genética , Transducción de Señal/fisiología
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA