Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Commun ; 15(1): 3443, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658557

RESUMO

The hypothalamus contains a remarkable diversity of neurons that orchestrate behavioural and metabolic outputs in a highly plastic manner. Neuronal diversity is key to enabling hypothalamic functions and, according to the neuroscience dogma, it is predetermined during embryonic life. Here, by combining lineage tracing of hypothalamic pro-opiomelanocortin (Pomc) neurons with single-cell profiling approaches in adult male mice, we uncovered subpopulations of 'Ghost' neurons endowed with atypical molecular and functional identity. Compared to 'classical' Pomc neurons, Ghost neurons exhibit negligible Pomc expression and are 'invisible' to available neuroanatomical approaches and promoter-based reporter mice for studying Pomc biology. Ghost neuron numbers augment in diet-induced obese mice, independent of neurogenesis or cell death, but weight loss can reverse this shift. Our work challenges the notion of fixed, developmentally programmed neuronal identities in the mature hypothalamus and highlight the ability of specialised neurons to reversibly adapt their functional identity to adult-onset obesogenic stimuli.


Assuntos
Hipotálamo , Neurônios , Obesidade , Pró-Opiomelanocortina , Análise de Célula Única , Animais , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/genética , Neurônios/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Masculino , Camundongos , Hipotálamo/metabolismo , Hipotálamo/citologia , Modelos Animais de Doenças , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese , Camundongos Obesos
2.
Science ; 384(6694): 438-446, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38662831

RESUMO

Liver mitochondria play a central role in metabolic adaptations to changing nutritional states, yet their dynamic regulation upon anticipated changes in nutrient availability has remained unaddressed. Here, we found that sensory food perception rapidly induced mitochondrial fragmentation in the liver through protein kinase B/AKT (AKT)-dependent phosphorylation of serine 131 of the mitochondrial fission factor (MFFS131). This response was mediated by activation of hypothalamic pro-opiomelanocortin (POMC)-expressing neurons. A nonphosphorylatable MFFS131G knock-in mutation abrogated AKT-induced mitochondrial fragmentation in vitro. In vivo, MFFS131G knock-in mice displayed altered liver mitochondrial dynamics and impaired insulin-stimulated suppression of hepatic glucose production. Thus, rapid activation of a hypothalamus-liver axis can adapt mitochondrial function to anticipated changes of nutritional state in control of hepatic glucose metabolism.


Assuntos
Alimentos , Gluconeogênese , Glucose , Fígado , Proteínas de Membrana , Mitocôndrias Hepáticas , Dinâmica Mitocondrial , Proteínas Mitocondriais , Percepção , Animais , Masculino , Camundongos , Técnicas de Introdução de Genes , Glucose/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Neurônios/metabolismo , Fosforilação , Pró-Opiomelanocortina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Transgênicos
3.
Nat Metab ; 6(3): 473-493, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38378998

RESUMO

Agouti-related peptide (AgRP)-expressing and proopiomelanocortin (POMC)-expressing neurons reciprocally regulate food intake. Here, we combine non-interacting recombinases to simultaneously express functionally opposing chemogenetic receptors in AgRP and POMC neurons for comparing metabolic responses in male and female mice with simultaneous activation of AgRP and inhibition of POMC neurons with isolated activation of AgRP neurons or isolated inhibition of POMC neurons. We show that food intake is regulated by the additive effect of AgRP neuron activation and POMC neuron inhibition, while systemic insulin sensitivity and gluconeogenesis are differentially modulated by isolated-versus-simultaneous regulation of AgRP and POMC neurons. We identify a neurocircuit engaging Npy1R-expressing neurons in the paraventricular nucleus of the hypothalamus, where activated AgRP neurons and inhibited POMC neurons cooperate to promote food consumption and activate Th+ neurons in the nucleus tractus solitarii. Collectively, these results unveil how food intake is precisely regulated by the simultaneous bidirectional interplay between AgRP and POMC neurocircuits.


Assuntos
Neurônios , Pró-Opiomelanocortina , Camundongos , Masculino , Feminino , Animais , Pró-Opiomelanocortina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Neurônios/metabolismo , Hipotálamo/metabolismo
4.
Nat Commun ; 14(1): 7824, 2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38016943

RESUMO

Dysregulation of hypothalamic ceramides has been associated with disrupted neuronal pathways in control of energy and glucose homeostasis. However, the specific ceramide species promoting neuronal lipotoxicity in obesity have remained obscure. Here, we find increased expression of the C16:0 ceramide-producing ceramide synthase (CerS)6 in cultured hypothalamic neurons exposed to palmitate in vitro and in the hypothalamus of obese mice. Conditional deletion of CerS6 in hypothalamic neurons attenuates high-fat diet (HFD)-dependent weight gain and improves glucose metabolism. Specifically, CerS6 deficiency in neurons expressing pro-opiomelanocortin (POMC) or steroidogenic factor 1 (SF-1) alters feeding behavior and alleviates the adverse metabolic effects of HFD feeding on insulin sensitivity and glucose tolerance. POMC-expressing cell-selective deletion of CerS6 prevents the diet-induced alterations of mitochondrial morphology and improves cellular leptin sensitivity. Our experiments reveal functions of CerS6-derived ceramides in hypothalamic lipotoxicity, altered mitochondrial dynamics, and ER/mitochondrial stress in the deregulation of food intake and glucose metabolism in obesity.


Assuntos
Obesidade , Pró-Opiomelanocortina , Animais , Camundongos , Ceramidas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Camundongos Obesos , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo
5.
Science ; 381(6665): eabl7398, 2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37769095

RESUMO

Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic signals with numerous cues that predict future deviations in energy supply. Recently, our understanding of the neural pathways underlying these regulatory principles-as well as their convergence in the hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism-have been revealed. These advances have changed our view of brain-dependent control of metabolic physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and how this emerging knowledge may provide new targets for their treatment.


Assuntos
Eixo Encéfalo-Intestino , Diabetes Mellitus Tipo 2 , Ingestão de Alimentos , Metabolismo Energético , Hipotálamo , Vias Neurais , Obesidade , Humanos , Diabetes Mellitus Tipo 2/fisiopatologia , Homeostase , Hipotálamo/fisiologia , Obesidade/fisiopatologia , Vias Neurais/fisiopatologia
6.
Nat Metab ; 4(10): 1402-1419, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36266547

RESUMO

The hypothalamus plays a key role in coordinating fundamental body functions. Despite recent progress in single-cell technologies, a unified catalog and molecular characterization of the heterogeneous cell types and, specifically, neuronal subtypes in this brain region are still lacking. Here, we present an integrated reference atlas, 'HypoMap,' of the murine hypothalamus, consisting of 384,925 cells, with the ability to incorporate new additional experiments. We validate HypoMap by comparing data collected from Smart-Seq+Fluidigm C1 and bulk RNA sequencing of selected neuronal cell types with different degrees of cellular heterogeneity. Finally, via HypoMap, we identify classes of neurons expressing glucagon-like peptide-1 receptor (Glp1r) and prepronociceptin (Pnoc), and validate them using single-molecule in situ hybridization. Collectively, HypoMap provides a unified framework for the systematic functional annotation of murine hypothalamic cell types, and it can serve as an important platform to unravel the functional organization of hypothalamic neurocircuits and to identify druggable targets for treating metabolic disorders.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1 , Hipotálamo , Camundongos , Animais , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Análise de Sequência de RNA , Expressão Gênica
7.
Cell Metab ; 34(2): 269-284.e9, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108514

RESUMO

Obesity and type 2 diabetes are associated with cognitive dysfunction. Because the hypothalamus is implicated in energy balance control and memory disorders, we hypothesized that specific neurons in this brain region are at the interface of metabolism and cognition. Acute obesogenic diet administration in mice impaired recognition memory due to defective production of the neurosteroid precursor pregnenolone in the hypothalamus. Genetic interference with pregnenolone synthesis by Star deletion in hypothalamic POMC, but not AgRP neurons, deteriorated recognition memory independently of metabolic disturbances. Our data suggest that pregnenolone's effects on cognitive function were mediated via an autocrine mechanism on POMC neurons, influencing hippocampal long-term potentiation. The relevance of central pregnenolone on cognition was also confirmed in metabolically unhealthy patients with obesity. Our data reveal an unsuspected role for POMC neuron-derived neurosteroids in cognition. These results provide the basis for a framework to investigate new facets of POMC neuron biology with implications for cognitive disorders.


Assuntos
Diabetes Mellitus Tipo 2 , Doenças Metabólicas , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipotálamo/metabolismo , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pregnenolona/metabolismo , Pró-Opiomelanocortina/metabolismo
8.
Endocr Rev ; 43(2): 314-328, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-34490882

RESUMO

The central nervous system (CNS) receives information from afferent neurons, circulating hormones, and absorbed nutrients and integrates this information to orchestrate the actions of the neuroendocrine and autonomic nervous systems in maintaining systemic metabolic homeostasis. Particularly the arcuate nucleus of the hypothalamus (ARC) is of pivotal importance for primary sensing of adiposity signals, such as leptin and insulin, and circulating nutrients, such as glucose. Importantly, energy state-sensing neurons in the ARC not only regulate feeding but at the same time control multiple physiological functions, such as glucose homeostasis, blood pressure, and innate immune responses. These findings have defined them as master regulators, which adapt integrative physiology to the energy state of the organism. The disruption of this fine-tuned control leads to an imbalance between energy intake and expenditure as well as deregulation of peripheral metabolism. Improving our understanding of the cellular, molecular, and functional basis of this regulatory principle in the CNS could set the stage for developing novel therapeutic strategies for the treatment of obesity and metabolic syndrome. In this review, we summarize novel insights with a particular emphasis on ARC neurocircuitries regulating food intake and glucose homeostasis and sensing factors that inform the brain of the organismal energy status.


Assuntos
Núcleo Arqueado do Hipotálamo , Diabetes Mellitus , Núcleo Arqueado do Hipotálamo/metabolismo , Diabetes Mellitus/metabolismo , Metabolismo Energético/fisiologia , Glucose/metabolismo , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Obesidade/metabolismo
9.
Nat Metab ; 3(12): 1662-1679, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34931084

RESUMO

Insulin acts on neurons and glial cells to regulate systemic glucose metabolism and feeding. However, the mechanisms of insulin access in discrete brain regions are incompletely defined. Here we show that insulin receptors in tanycytes, but not in brain endothelial cells, are required to regulate insulin access to the hypothalamic arcuate nucleus. Mice lacking insulin receptors in tanycytes (IR∆Tan mice) exhibit systemic insulin resistance, while displaying normal food intake and energy expenditure. Tanycytic insulin receptors are also necessary for the orexigenic effects of ghrelin, but not for the anorexic effects of leptin. IR∆Tan mice exhibit increased agouti-related peptide (AgRP) neuronal activity, while displaying blunted AgRP neuronal adaptations to feeding-related stimuli. Lastly, a highly palatable food decreases tanycytic and arcuate nucleus insulin signalling to levels comparable to those seen in IR∆Tan mice. These changes are rooted in modifications of cellular stress responses and of mitochondrial protein quality control in tanycytes. Conclusively, we reveal a critical role of tanycyte insulin receptors in gating feeding-state-dependent regulation of AgRP neurons and systemic insulin sensitivity, and show that insulin resistance in tanycytes contributes to the pleiotropic manifestations of obesity-associated insulin resistance.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Células Ependimogliais/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Proteína Relacionada com Agouti/química , Animais , Biomarcadores , Barreira Hematoencefálica/metabolismo , Cálcio , Metabolismo Energético , Imunofluorescência , Grelina/metabolismo , Glucose/metabolismo , Resistência à Insulina , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Modelos Biológicos , Fragmentos de Peptídeos/metabolismo , Receptor de Insulina/metabolismo
10.
Nat Neurosci ; 24(7): 913-929, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34002087

RESUMO

Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus represent key regulators of metabolic homeostasis. Electrophysiological and single-cell sequencing experiments have revealed a remarkable degree of heterogeneity of these neurons. However, the exact molecular basis and functional consequences of this heterogeneity have not yet been addressed. Here, we have developed new mouse models in which intersectional Cre/Dre-dependent recombination allowed for successful labeling, translational profiling and functional characterization of distinct POMC neurons expressing the leptin receptor (Lepr) and glucagon like peptide 1 receptor (Glp1r). Our experiments reveal that POMCLepr+ and POMCGlp1r+ neurons represent largely nonoverlapping subpopulations with distinct basic electrophysiological properties. They exhibit a specific anatomical distribution within the arcuate nucleus and differentially express receptors for energy-state communicating hormones and neurotransmitters. Finally, we identify a differential ability of these subpopulations to suppress feeding. Collectively, we reveal a notably distinct functional microarchitecture of critical metabolism-regulatory neurons.


Assuntos
Comportamento Alimentar/fisiologia , Hipotálamo/fisiologia , Neurônios/fisiologia , Pró-Opiomelanocortina/metabolismo , Animais , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Hipotálamo/citologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia
11.
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
12.
Nat Commun ; 9(1): 3432, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143607

RESUMO

p53 is a well-known tumor suppressor that has emerged as an important player in energy balance. However, its metabolic role in the hypothalamus remains unknown. Herein, we show that mice lacking p53 in agouti-related peptide (AgRP), but not proopiomelanocortin (POMC) or steroidogenic factor-1 (SF1) neurons, are more prone to develop diet-induced obesity and show reduced brown adipose tissue (BAT) thermogenic activity. AgRP-specific ablation of p53 resulted in increased hypothalamic c-Jun N-terminal kinase (JNK) activity before the mice developed obesity, and central inhibition of JNK reversed the obese phenotype of these mice. The overexpression of p53 in the ARC or specifically in AgRP neurons of obese mice decreased body weight and stimulated BAT thermogenesis, resulting in body weight loss. Finally, p53 in AgRP neurons regulates the ghrelin-induced food intake and body weight. Overall, our findings provide evidence that p53 in AgRP neurons is required for normal adaptations against diet-induced obesity.


Assuntos
Dieta/efeitos adversos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/genética , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Ratos Sprague-Dawley , Fator Esteroidogênico 1/metabolismo , Proteína Supressora de Tumor p53/genética
13.
Cell Rep ; 23(6): 1728-1741, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29742429

RESUMO

Anorexigenic pro-opiomelanocortin (Pomc)/alpha-melanocyte stimulating hormone (αMSH) neurons of the hypothalamic melanocortin system function as key regulators of energy homeostasis, also controlling somatic growth across different species. However, the mechanisms of melanocortin-dependent growth control still remain ill-defined. Here, we reveal a thus-far-unrecognized structural and functional connection between Pomc neurons and the somatotropic hypothalamo-pituitary axis. Excessive feeding of larval zebrafish causes leptin resistance and reduced levels of the hypothalamic satiety mediator pomca. In turn, this leads to reduced activation of hypophysiotropic somatostatin (Sst)-neurons that express the melanocortin receptor Mc4r, elevated growth hormone (GH) expression in the pituitary, and enhanced somatic growth. Mc4r expression and αMSH responsiveness are conserved in Sst-expressing hypothalamic neurons of mice. Thus, acquired leptin resistance and attenuation of pomca transcription in response to excessive caloric intake may represent an ancient mechanism to promote somatic growth when food resources are plentiful.


Assuntos
Dieta , Hormônio do Crescimento/metabolismo , Crescimento e Desenvolvimento/efeitos dos fármacos , Leptina/farmacologia , Pró-Opiomelanocortina/metabolismo , Somatostatina/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Larva/efeitos dos fármacos , Larva/fisiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Peixe-Zebra , alfa-MSH/metabolismo
14.
Cell Metab ; 26(2): 375-393.e7, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768176

RESUMO

Beige adipocytes can interconvert between white and brown-like states and switch between energy storage versus expenditure. Here we report that beige adipocyte plasticity is important for feeding-associated changes in energy expenditure and is coordinated by the hypothalamus and the phosphatase TCPTP. A fasting-induced and glucocorticoid-mediated induction of TCPTP, inhibited insulin signaling in AgRP/NPY neurons, repressed the browning of white fat and decreased energy expenditure. Conversely feeding reduced hypothalamic TCPTP, to increase AgRP/NPY neuronal insulin signaling, white adipose tissue browning and energy expenditure. The feeding-induced repression of hypothalamic TCPTP was defective in obesity. Mice lacking TCPTP in AgRP/NPY neurons were resistant to diet-induced obesity and had increased beige fat activity and energy expenditure. The deletion of hypothalamic TCPTP in obesity restored feeding-induced browning and increased energy expenditure to promote weight loss. Our studies define a hypothalamic switch that coordinates energy expenditure with feeding for the maintenance of energy balance.


Assuntos
Ingestão de Alimentos/psicologia , Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 2/biossíntese , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Obesidade/genética , Obesidade/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética
15.
Dis Model Mech ; 10(6): 679-689, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28592656

RESUMO

The 'obesity epidemic' represents a major global socioeconomic burden that urgently calls for a better understanding of the underlying causes of increased weight gain and its associated metabolic comorbidities, such as type 2 diabetes mellitus and cardiovascular diseases. Improving our understanding of the cellular basis of obesity could set the stage for the development of new therapeutic strategies. The CNS plays a pivotal role in the regulation of energy and glucose homeostasis. Distinct neuronal cell populations, particularly within the arcuate nucleus of the hypothalamus, sense the nutrient status of the organism and integrate signals from peripheral hormones including pancreas-derived insulin and adipocyte-derived leptin to regulate calorie intake, glucose metabolism and energy expenditure. The arcuate neurons are tightly connected to other specialized neuronal subpopulations within the hypothalamus, but also to various extrahypothalamic brain regions, allowing a coordinated behavioral response. This At a Glance article gives an overview of the recent knowledge, mainly derived from rodent models, regarding the CNS-dependent regulation of energy and glucose homeostasis, and illustrates how dysregulation of the neuronal networks involved can lead to overnutrition and obesity. The potential impact of recent research findings in the field on therapeutic treatment strategies for human obesity is also discussed.


Assuntos
Apetite , Metabolismo Energético , Homeostase , Hipotálamo/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Animais , Humanos , Obesidade/genética
16.
J Clin Invest ; 127(1): 24-32, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28045396

RESUMO

Over the last years, hypothalamic inflammation has been linked to the development and progression of obesity and its sequelae. There is accumulating evidence that this inflammation not only impairs energy balance but also contributes to obesity-associated insulin resistance. Elevated activation of key inflammatory mediators such as JNK and IκB kinase (IKK) occurs rapidly upon consumption of a high-fat diet, even prior to significant weight gain. This activation of hypothalamic inflammatory pathways results in the uncoupling of caloric intake and energy expenditure, fostering overeating and further weight gain. In addition, these inflammatory processes contribute to obesity-associated insulin resistance and deterioration of glucose metabolism via altered neurocircuit functions. An understanding of the contributions of different neuronal and non-neuronal cell types to hypothalamic inflammatory processes, and delineation of the differences and similarities between acute and chronic activation of these inflammatory pathways, will be critical for the development of novel therapeutic strategies for the treatment of obesity and metabolic syndrome.


Assuntos
Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Animais , Gorduras na Dieta/efeitos adversos , Ingestão de Energia , Metabolismo Energético , Glucose/metabolismo , Humanos , Hipotálamo/patologia , Quinase I-kappa B/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , MAP Quinase Quinase 4/metabolismo , Síndrome Metabólica/patologia , Obesidade/patologia
17.
Cell Rep ; 17(10): 2512-2521, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27926856

RESUMO

Melanin-concentrating-hormone (MCH)-expressing neurons (MCH neurons) in the lateral hypothalamus (LH) are critical regulators of energy and glucose homeostasis. Here, we demonstrate that insulin increases the excitability of these neurons in control mice. In vivo, insulin promotes phosphatidylinositol 3-kinase (PI3K) signaling in MCH neurons, and cell-type-specific deletion of the insulin receptor (IR) abrogates this response. While lean mice lacking the IR in MCH neurons (IRΔMCH) exhibit no detectable metabolic phenotype under normal diet feeding, they present with improved locomotor activity and insulin sensitivity under high-fat-diet-fed, obese conditions. Similarly, obesity promotes PI3 kinase signaling in these neurons, and this response is abrogated in IRΔMCH mice. In turn, acute chemogenetic activation of MCH neurons impairs locomotor activity but not insulin sensitivity. Collectively, our experiments reveal an insulin-dependent activation of MCH neurons in obesity, which contributes via distinct mechanisms to the manifestation of impaired locomotor activity and insulin resistance.


Assuntos
Hormônios Hipotalâmicos/genética , Resistência à Insulina/genética , Insulina/metabolismo , Melaninas/genética , Obesidade/metabolismo , Hormônios Hipofisários/genética , Animais , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Glucose/metabolismo , Humanos , Hipotálamo/metabolismo , Insulina/administração & dosagem , Locomoção/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Obesidade/tratamento farmacológico , Obesidade/patologia , Fosfatidilinositol 3-Quinases/genética
18.
Curr Biol ; 26(11): R470-3, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27269725

RESUMO

Cutting-edge experiments show a new means to control the activity of specifically genetically targeted neurons in the hypothalamus using electromagnetic force. At the flip of a switch, the system bidirectionally regulates feeding behavior and glucose homeostasis, demonstrating wireless control over deep brain regions and their strong influence over energy balance.


Assuntos
Hipotálamo , Neuroendocrinologia , Metabolismo Energético , Comportamento Alimentar , Glucose , Homeostase
19.
Cell ; 162(6): 1404-17, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26359991

RESUMO

Activation of orexigenic AgRP-expressing neurons in the arcuate nucleus of the hypothalamus potently promotes feeding, thus defining new regulators of AgRP neuron activity could uncover potential novel targets for obesity treatment. Here, we demonstrate that AgRP neurons express the purinergic receptor 6 (P2Y6), which is activated by uridine-diphosphate (UDP). In vivo, UDP induces ERK phosphorylation and cFos expression in AgRP neurons and promotes action potential firing of these neurons in brain slice recordings. Consequently, central application of UDP promotes feeding, and this response is abrogated upon pharmacologic or genetic inhibition of P2Y6 as well as upon pharmacogenetic inhibition of AgRP neuron activity. In obese animals, hypothalamic UDP content is elevated as a consequence of increased circulating uridine concentrations. Collectively, these experiments reveal a potential regulatory pathway in obesity, where peripheral uridine increases hypothalamic UDP concentrations, which in turn can promote feeding via PY6-dependent activation of AgRP neurons.


Assuntos
Regulação do Apetite , Hipotálamo/metabolismo , Obesidade/metabolismo , Receptores Purinérgicos P2/metabolismo , Difosfato de Uridina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Modelos Animais de Doenças , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
Cell ; 156(3): 495-509, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24462248

RESUMO

Maternal metabolic homeostasis exerts long-term effects on the offspring's health outcomes. Here, we demonstrate that maternal high-fat diet (HFD) feeding during lactation predisposes the offspring for obesity and impaired glucose homeostasis in mice, which is associated with an impairment of the hypothalamic melanocortin circuitry. Whereas the number and neuropeptide expression of anorexigenic proopiomelanocortin (POMC) and orexigenic agouti-related peptide (AgRP) neurons, electrophysiological properties of POMC neurons, and posttranslational processing of POMC remain unaffected in response to maternal HFD feeding during lactation, the formation of POMC and AgRP projections to hypothalamic target sites is severely impaired. Abrogating insulin action in POMC neurons of the offspring prevents altered POMC projections to the preautonomic paraventricular nucleus of the hypothalamus (PVH), pancreatic parasympathetic innervation, and impaired glucose-stimulated insulin secretion in response to maternal overnutrition. These experiments reveal a critical timing, when altered maternal metabolism disrupts metabolic homeostasis in the offspring via impairing neuronal projections, and show that abnormal insulin signaling contributes to this effect.


Assuntos
Dieta Hiperlipídica , Hiperglicemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactação , Obesidade/metabolismo , Animais , Axônios/metabolismo , Feminino , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Gravidez , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA