Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Neuron ; 111(16): 2583-2600.e6, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37295418

RESUMO

Chronic stress fuels the consumption of palatable food and can enhance obesity development. While stress- and feeding-controlling pathways have been identified, how stress-induced feeding is orchestrated remains unknown. Here, we identify lateral habenula (LHb) Npy1r-expressing neurons as the critical node for promoting hedonic feeding under stress, since lack of Npy1r in these neurons alleviates the obesifying effects caused by combined stress and high fat feeding (HFDS) in mice. Mechanistically, this is due to a circuit originating from central amygdala NPY neurons, with the upregulation of NPY induced by HFDS initiating a dual inhibitory effect via Npy1r signaling onto LHb and lateral hypothalamus neurons, thereby reducing the homeostatic satiety effect through action on the downstream ventral tegmental area. Together, these results identify LHb-Npy1r neurons as a critical node to adapt the response to chronic stress by driving palatable food intake in an attempt to overcome the negative valence of stress.


Assuntos
Habenula , Camundongos , Animais , Vias Neurais/fisiologia , Habenula/fisiologia , Região Hipotalâmica Lateral , Área Tegmentar Ventral , Neurônios/fisiologia
2.
Cell Metab ; 35(6): 979-995.e7, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37201523

RESUMO

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.


Assuntos
Leptina , Pró-Opiomelanocortina , Camundongos , Animais , Leptina/metabolismo , Pró-Opiomelanocortina/metabolismo , Neuropeptídeo Y/metabolismo , Proteína Relacionada com Agouti/metabolismo , Neurônios/metabolismo , Núcleo Arqueado do Hipotálamo , Obesidade/metabolismo
3.
Neurochem Res ; 48(8): 2490-2501, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37017888

RESUMO

The nucleus accumbens shell is a critical node in reward circuitry, encoding environments associated with reward. Long-range inputs from the ventral hippocampus (ventral subiculum) to the nucleus accumbens shell have been identified, yet their precise molecular phenotype remains to be determined. Here we used retrograde tracing to identify the ventral subiculum as the brain region with the densest glutamatergic (VGluT1-Slc17a7) input to the shell. We then used circuit-directed translating ribosome affinity purification to examine the molecular characteristics of distinct glutamatergic (VGluT1, VGluT2-Slc17a6) ventral subiculum to nucleus accumbens shell projections. We immunoprecipitated translating ribosomes from this population of projection neurons and analysed molecular connectomic information using RNA sequencing. We found differential gene enrichment across both glutamatergic projection neuron subtypes. In VGluT1 projections, we found enrichment of Pfkl, a gene involved in glucose metabolism. In VGluT2 projections, we found a depletion of Sparcl1 and Dlg1, genes known to play a role in depression- and addiction-related behaviours. These findings highlight potential glutamatergic neuronal-projection-specific differences in ventral subiculum to nucleus accumbens shell projections. Together these data advance our understanding of the phenotype of a defined brain circuit.


Assuntos
Hipocampo , Núcleo Accumbens , Encéfalo , Hipocampo/metabolismo , Núcleo Accumbens/metabolismo , Recompensa , Animais , Camundongos
4.
Diabetologia ; 65(6): 984-996, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35316840

RESUMO

AIMS/HYPOTHESIS: Pancreatic beta cell dedifferentiation, transdifferentiation into other islet cells and apoptosis have been implicated in beta cell failure in type 2 diabetes, although the mechanisms are poorly defined. The endoplasmic reticulum stress response factor X-box binding protein 1 (XBP1) is a major regulator of the unfolded protein response. XBP1 expression is reduced in islets of people with type 2 diabetes, but its role in adult differentiated beta cells is unclear. Here, we assessed the effects of Xbp1 deletion in adult beta cells and tested whether XBP1-mediated unfolded protein response makes a necessary contribution to beta cell compensation in insulin resistance states. METHODS: Mice with inducible beta cell-specific Xbp1 deletion were studied under normal (chow diet) or metabolic stress (high-fat diet or obesity) conditions. Glucose tolerance, insulin secretion, islet gene expression, alpha cell mass, beta cell mass and apoptosis were assessed. Lineage tracing was used to determine beta cell fate. RESULTS: Deletion of Xbp1 in adult mouse beta cells led to beta cell dedifferentiation, beta-to-alpha cell transdifferentiation and increased alpha cell mass. Cell lineage-specific analyses revealed that Xbp1 deletion deactivated beta cell identity genes (insulin, Pdx1, Nkx6.1, Beta2, Foxo1) and derepressed beta cell dedifferentiation (Aldh1a3) and alpha cell (glucagon, Arx, Irx2) genes. Xbp1 deletion in beta cells of obese ob/ob or high-fat diet-fed mice triggered diabetes and worsened glucose intolerance by disrupting insulin secretory capacity. Furthermore, Xbp1 deletion increased beta cell apoptosis under metabolic stress conditions by attenuating the antioxidant response. CONCLUSIONS/INTERPRETATION: These findings indicate that XBP1 maintains beta cell identity, represses beta-to-alpha cell transdifferentiation and is required for beta cell compensation and prevention of diabetes in insulin resistance states.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Células Secretoras de Insulina , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Transdiferenciação Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina/genética , Células Secretoras de Insulina/metabolismo , Camundongos , Estresse Fisiológico , Proteína 1 de Ligação a X-Box/genética
5.
Mol Metab ; 59: 101455, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35167990

RESUMO

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.


Assuntos
Metabolismo Energético , Neuropeptídeo Y , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Ligantes , Camundongos , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-34592387

RESUMO

Independent from homeostatic needs, the consumption of foods originating from hyperpalatable diets is defined as hedonic eating. Hedonic eating can be observed in many forms of eating phenotypes, such as compulsive eating and stress-eating, heightening the risk of obesity development. For instance, stress can trigger the consumption of palatable foods as a type of coping strategy, which can become compulsive, particularly when developed as a habit. Although eating for pleasure is observed in multiple maladaptive eating behaviours, the current understanding of the neurobiology underlying hedonic eating remains deficient. Intriguingly, the combined orexigenic, anxiolytic and reward-seeking properties of Neuropeptide Y (NPY) ignited great interest and has positioned NPY as one of the core neuromodulators operating hedonic eating behaviours. While extensive literature exists exploring the homeostatic orexigenic and anxiolytic properties of NPY, the rewarding effects of NPY continue to be investigated. As deduced from a series of behavioural and molecular-based studies, NPY appears to motivate the consumption and enhancement of food-rewards. As a possible mechanism, NPY may modulate reward-associated monoaminergic pathways, such as the dopaminergic and serotoninergic neural networks, to modulate hedonic eating behaviours. Furthermore, potential direct and indirect NPYergic neurocircuitries connecting classical homeostatic and hedonic neuropathways may also exist involving the anti-reward centre the lateral habenula. Therefore, this review investigates the participation of NPY in orchestrating hedonic eating behaviours through the modulation of monoaminergic pathways.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Comportamento Alimentar/fisiologia , Vias Neurais/metabolismo , Neuropeptídeo Y/metabolismo , Recompensa , Neurônios Serotoninérgicos/metabolismo , Adaptação Psicológica , Homeostase , Humanos , Motivação , Obesidade/fisiopatologia
7.
FASEB J ; 35(11): e21980, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34694651

RESUMO

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.


Assuntos
Ansiedade/metabolismo , Comportamento de Ingestão de Líquido , Oligopeptídeos/fisiologia , Receptores de Neuropeptídeos/metabolismo , Animais , Peso Corporal , Metabolismo Energético , Feminino , Masculino , Camundongos , Camundongos Knockout
8.
Nat Commun ; 12(1): 2622, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976180

RESUMO

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.


Assuntos
Arginina/análogos & derivados , Obesidade/prevenção & controle , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Termogênese/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Adulto , Animais , Arginina/farmacologia , Arginina/uso terapêutico , Biópsia , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Obesidade/etiologia , Obesidade/metabolismo , Cultura Primária de Células , Receptores de Neuropeptídeo Y/metabolismo
9.
STAR Protoc ; 2(2): 100448, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33912847

RESUMO

Chronic stress has adverse consequences on many organ systems and physiological processes. However, existing protocols show large variability in response and are not suitable for female mice. Here, we provide a step-by-step protocol for establishing a reliable chronic stress model in mice that can be used in a variety of physiological settings. This protocol has been tested to be effective to produce a consistent response to stress in several mouse strains (C57BL/6J, 129X1/SvJ, B6.V-Lepob/J) and both sexes. For complete details on the use and execution of this protocol, please refer to Ip et al. (2019).


Assuntos
Doença Crônica , Modelos Animais de Doenças , Camundongos , Estresse Fisiológico , Animais , Encéfalo/patologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Transcriptoma/genética
10.
Peptides ; 140: 170534, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33757831

RESUMO

Cocaine- and amphetamine-related transcript (CART) is a neuropeptide first discovered in the striatum of the rat brain. Later, the genetic sequence and function of CART peptide (CARTp) was found to be conserved among multiple mammalian species. Over the 25 years, since its discovery, CART mRNA (Cartpt) expression has been reported widely throughout the central and peripheral nervous systems underscoring its role in diverse physiological functions. Here, we review the localization and function of CARTp as it relates to energy homeostasis. We summarize the expression changes of central and peripheral Cartpt in response to metabolic states and make use of available large data sets to gain additional insights into the anatomy of the Cartpt expressing vagal neurons and their expression patterns in the gut. Furthermore, we provide an overview of the role of CARTp as an anorexigenic signal and its effect on energy expenditure and body weight control with insights from both pharmacological and transgenic animal studies. Subsequently, we discuss the role of CARTp in the pathophysiology of obesity and review important new developments towards identifying a candidate receptor for CARTp signalling. Altogether, the field of CARTp research has made rapid and substantial progress recently, and we review the case for considering CARTp as a potential therapeutic target for stemming the obesity epidemic.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Peptídeos/metabolismo , Nervo Vago/metabolismo , Animais , Metabolismo Energético , Homeostase , Humanos
11.
FASEB J ; 35(4): e21435, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33749879

RESUMO

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.


Assuntos
Bactérias/classificação , Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal , Peptídeo YY/metabolismo , Animais , Composição Corporal , Camundongos , Camundongos Knockout , Peptídeo YY/genética
12.
Int J Obes (Lond) ; 44(10): 2149-2164, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32152498

RESUMO

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.


Assuntos
Tecido Adiposo/metabolismo , Osso e Ossos/metabolismo , Metabolismo Energético , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Adiposidade , Animais , Composição Corporal , Dieta Hiperlipídica , Ingestão de Energia , Feminino , Masculino , Camundongos , Receptores para Leptina
13.
Neuropeptides ; 80: 101994, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31740068

RESUMO

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.


Assuntos
Peso Corporal/fisiologia , Metabolismo Energético/fisiologia , Neurônios/metabolismo , Neuropeptídeo Y/deficiência , Neurotensina/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Homeostase/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Transgênicos , Neuropeptídeo Y/metabolismo , Fenótipo
15.
Cell Metab ; 30(1): 111-128.e6, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31031093

RESUMO

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.


Assuntos
Tonsila do Cerebelo/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Animais , Temperatura Corporal , Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos/fisiologia , Eletrofisiologia , Metabolismo Energético/fisiologia , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Insulina/metabolismo , Masculino , Camundongos , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real
16.
Nat Commun ; 9(1): 4722, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30413707

RESUMO

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.


Assuntos
Dieta , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais , Termogênese , Tecido Adiposo Marrom/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Animal , Osso e Ossos/metabolismo , Temperatura Baixa , Metabolismo Energético , Feminino , Homeostase , Ligantes , Masculino , Camundongos Knockout , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Neuropeptídeos/metabolismo , Osteogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética
17.
Elife ; 72018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30129922

RESUMO

Cocaine- and amphetamine-regulated transcript (CART) is widely expressed in the hypothalamus and an important regulator of energy homeostasis; however, the specific contributions of different CART neuronal populations to this process are not known. Here, we show that depolarization of mouse arcuate nucleus (Arc) CART neurons via DREADD technology decreases energy expenditure and physical activity, while it exerts the opposite effects in CART neurons in the lateral hypothalamus (LHA). Importantly, when stimulating these neuronal populations in the absence of CART, the effects were attenuated. In contrast, while activation of CART neurons in the LHA stimulated feeding in the presence of CART, endogenous CART inhibited food intake in response to Arc CART neuron activation. Taken together, these results demonstrate anorexigenic but anabolic effects of CART upon Arc neuron activation, and orexigenic but catabolic effects upon LHA-neuron activation, highlighting the complex and nuclei-specific functions of CART in controlling feeding and energy homeostasis.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético , Região Hipotalâmica Lateral/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Comportamento Animal , Temperatura Corporal/efeitos dos fármacos , Clozapina/análogos & derivados , Clozapina/farmacologia , Dependovirus/metabolismo , Ingestão de Alimentos , Metabolismo Energético/efeitos dos fármacos , Injeções , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurotransmissores/metabolismo , Condicionamento Físico Animal , Reprodutibilidade dos Testes , Aumento de Peso/efeitos dos fármacos
18.
Nat Commun ; 8(1): 490, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28887564

RESUMO

Failure to secrete sufficient quantities of insulin is a pathological feature of type-1 and type-2 diabetes, and also reduces the success of islet cell transplantation. Here we demonstrate that Y1 receptor signaling inhibits insulin release in ß-cells, and show that this can be pharmacologically exploited to boost insulin secretion. Transplanting islets with Y1 receptor deficiency accelerates the normalization of hyperglycemia in chemically induced diabetic recipient mice, which can also be achieved by short-term pharmacological blockade of Y1 receptors in transplanted mouse and human islets. Furthermore, treatment of non-obese diabetic mice with a Y1 receptor antagonist delays the onset of diabetes. Mechanistically, Y1 receptor signaling inhibits the production of cAMP in islets, which via CREB mediated pathways results in the down-regulation of several key enzymes in glycolysis and ATP production. Thus, manipulating Y1 receptor signaling in ß-cells offers a unique therapeutic opportunity for correcting insulin deficiency as it occurs in the pathological state of type-1 diabetes as well as during islet transplantation.Islet transplantation is considered one of the potential treatments for T1DM but limited islet survival and their impaired function pose limitations to this approach. Here Loh et al. show that the Y1 receptor is expressed in ß- cells and inhibition of its signalling, both genetic and pharmacological, improves mouse and human islet function.


Assuntos
Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais
19.
Curr Top Dev Biol ; 117: 497-521, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26969997

RESUMO

The embryonic head is the first major body part to be constructed during embryogenesis. The allocation and the assembly of the progenitor tissues, which start at gastrulation, are accompanied by the spatiotemporal activity of transcription factors and signaling pathways that drives lineage specification, germ layer formation, and cell/tissue movement. The morphogenesis, regionalization, and patterning of the brain and craniofacial structures rely on the function of LIM-domain, homeodomain, and basic helix-loop-helix transcription factors. These factors constitute the central nodes of a gene regulatory network (GRN) which encompasses and intersects with signaling pathways involved with head formation. It is predicted that the functional output of this "head GRN" impacts on cellular function and cell-cell interactions that are essential for lineage differentiation and tissue modeling, which are key processes underpinning the formation of the head.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Cabeça/embriologia , Animais , Embrião de Mamíferos/metabolismo , Camundongos
20.
Int J Cardiol ; 191: 138-48, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25965621

RESUMO

BACKGROUND: In patients at high risk of atherosclerotic cardiovascular diseases (ASCVDs), residual cardiovascular risk persists despite the achievement of target LDL cholesterol levels with statin therapy. It is still unclear whether adding lipid-modifying agent to statin treatment can further improve clinical outcomes. METHODS: Randomized controlled trials (RCTs) in terms of adding lipid-modifying agent to statin versus statin monotherapy in patients at high risk of ASCVD were identified by electronic and manual searches. Results were expressed as relative risk (RR) with 95% confidence intervals (CIs). RESULTS: Eleven RCTs with 109,244 patients were included in this meta-analysis. Overall, the incidences of major adverse cardiovascular events (MACEs) were 9.70% in the statin combination groups and 9.92% in the statin monotherapy groups. No significant difference was observed in the risk of MACEs either in overall (RR 0.99, 95% CI 0.93-1.05, P=0.76) or subgroup analysis (CETP inhibitor: RR 1.07, 95% CI 0.93-1.23, P=0.37; niacin: RR 1.03, 95% CI 0.85-1.25, P=0.79; n-3 fatty acid: RR 0.98, 95% CI 0.88-1.09, P=0.70; fenofibrate: RR 0.93, 95% CI 0.80-1.09, P=0.38), with the exception of the statin/ezetimibe combination subgroup (RR 0.92, 95% CI 0.87-0.97, P=0.004). Adding lipid-modifying agent to statin significantly increased liver injury risk. Adding ezetimibe to statin did not alter side effect profile. CONCLUSION: Adding niacin, CETP inhibitors, n-3 fatty acid or fibrates to statin therapy has all failed to achieve a clinical benefit. Adding ezetimibe to statin therapy further lowers LDL-cholesterol safely and translates into a clinical benefit in patients at high risk of ASCVD.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/sangue , Ensaios Clínicos Controlados Aleatórios como Assunto , Doenças Cardiovasculares/sangue , Humanos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...