Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 68(12): 2210-2222, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31530579

RESUMO

Melanin-concentrating hormone (MCH) is an important regulator of food intake, glucose metabolism, and adiposity. However, the mechanisms mediating these actions remain largely unknown. We used pharmacological and genetic approaches to show that the sirtuin 1 (SIRT1)/FoxO1 signaling pathway in the hypothalamic arcuate nucleus (ARC) mediates MCH-induced feeding, adiposity, and glucose intolerance. MCH reduces proopiomelanocortin (POMC) neuronal activity, and the SIRT1/FoxO1 pathway regulates the inhibitory effect of MCH on POMC expression. Remarkably, the metabolic actions of MCH are compromised in mice lacking SIRT1 specifically in POMC neurons. Of note, the actions of MCH are independent of agouti-related peptide (AgRP) neurons because inhibition of γ-aminobutyric acid receptor in the ARC did not prevent the orexigenic action of MCH, and the hypophagic effect of MCH silencing was maintained after chemogenetic stimulation of AgRP neurons. Central SIRT1 is required for MCH-induced weight gain through its actions on the sympathetic nervous system. The central MCH knockdown causes hypophagia and weight loss in diet-induced obese wild-type mice; however, these effects were abolished in mice overexpressing SIRT1 fed a high-fat diet. These data reveal the neuronal basis for the effects of MCH on food intake, body weight, and glucose metabolism and highlight the relevance of SIRT1/FoxO1 pathway in obesity.


Assuntos
Adiposidade/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Intolerância à Glucose/metabolismo , Hiperfagia/metabolismo , Hormônios Hipotalâmicos/farmacologia , Melaninas/farmacologia , Neurônios/efeitos dos fármacos , Hormônios Hipofisários/farmacologia , Pró-Opiomelanocortina/metabolismo , Sirtuína 1/metabolismo , Adiposidade/fisiologia , Animais , Proteína Forkhead Box O1/genética , Intolerância à Glucose/genética , Hiperfagia/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos Sprague-Dawley , Sirtuína 1/genética
2.
Nat Med ; 23(12): 1444-1453, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29106398

RESUMO

Asprosin is a recently discovered fasting-induced hormone that promotes hepatic glucose production. Here we demonstrate that asprosin in the circulation crosses the blood-brain barrier and directly activates orexigenic AgRP+ neurons via a cAMP-dependent pathway. This signaling results in inhibition of downstream anorexigenic proopiomelanocortin (POMC)-positive neurons in a GABA-dependent manner, which then leads to appetite stimulation and a drive to accumulate adiposity and body weight. In humans, a genetic deficiency in asprosin causes a syndrome characterized by low appetite and extreme leanness; this is phenocopied by mice carrying similar mutations and can be fully rescued by asprosin. Furthermore, we found that obese humans and mice had pathologically elevated concentrations of circulating asprosin, and neutralization of asprosin in the blood with a monoclonal antibody reduced appetite and body weight in obese mice, in addition to improving their glycemic profile. Thus, in addition to performing a glucogenic function, asprosin is a centrally acting orexigenic hormone that is a potential therapeutic target in the treatment of both obesity and diabetes.


Assuntos
Regulação do Apetite/genética , Hipotálamo/metabolismo , Proteínas dos Microfilamentos/fisiologia , Fragmentos de Peptídeos/fisiologia , Hormônios Peptídicos/fisiologia , Adolescente , Adulto , Animais , Depressores do Apetite/metabolismo , Feminino , Fibrilina-1 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Neurônios/metabolismo , Fragmentos de Peptídeos/genética , Hormônios Peptídicos/genética , Ratos , Transdução de Sinais , Adulto Jovem
3.
Neuron ; 95(4): 944-954.e4, 2017 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-28817806

RESUMO

The somatosensory system provides animals with the ability to detect, distinguish, and respond to diverse thermal, mechanical, and irritating stimuli. While there has been progress in defining classes of neurons underlying temperature sensation and gentle touch, less is known about the neurons specific for mechanical pain. Here, we use in vivo functional imaging to identify a class of cutaneous sensory neurons that are selectively activated by high-threshold mechanical stimulation (HTMRs). We show that their optogenetic excitation evokes rapid protective and avoidance behaviors. Unlike other nociceptors, these HTMRs are fast-conducting Aδ-fibers with highly specialized circumferential endings wrapping the base of individual hair follicles. Notably, we find that Aδ-HTMRs innervate unique but overlapping fields and can be activated by stimuli as precise as the pulling of a single hair. Together, the distinctive features of this class of Aδ-HTMRs appear optimized for accurate and rapid localization of mechanical pain. VIDEO ABSTRACT.


Assuntos
Vias Aferentes/fisiologia , Cabelo , Mecanorreceptores/fisiologia , Nociceptores/fisiologia , Células Receptoras Sensoriais/fisiologia , Tato/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Antineoplásicos Hormonais/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Channelrhodopsins , Diterpenos/farmacologia , Feminino , Cabelo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurotoxinas/farmacologia , Pele/inervação , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Tamoxifeno/farmacologia , Gânglio Trigeminal/diagnóstico por imagem , Gânglio Trigeminal/fisiologia
4.
Nature ; 545(7655): 477-481, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28514446

RESUMO

In humans and other mammalian species, lesions in the preoptic area of the hypothalamus cause profound sleep impairment, indicating a crucial role of the preoptic area in sleep generation. However, the underlying circuit mechanism remains poorly understood. Electrophysiological recordings and c-Fos immunohistochemistry have shown the existence of sleep-active neurons in the preoptic area, especially in the ventrolateral preoptic area and median preoptic nucleus. Pharmacogenetic activation of c-Fos-labelled sleep-active neurons has been shown to induce sleep. However, the sleep-active neurons are spatially intermingled with wake-active neurons, making it difficult to target the sleep neurons specifically for circuit analysis. Here we identify a population of preoptic area sleep neurons on the basis of their projection target and discover their molecular markers. Using a lentivirus expressing channelrhodopsin-2 or a light-activated chloride channel for retrograde labelling, bidirectional optogenetic manipulation, and optrode recording, we show that the preoptic area GABAergic neurons projecting to the tuberomammillary nucleus are both sleep active and sleep promoting. Furthermore, translating ribosome affinity purification and single-cell RNA sequencing identify candidate markers for these neurons, and optogenetic and pharmacogenetic manipulations demonstrate that several peptide markers (cholecystokinin, corticotropin-releasing hormone, and tachykinin 1) label sleep-promoting neurons. Together, these findings provide easy genetic access to sleep-promoting preoptic area neurons and a valuable entry point for dissecting the sleep control circuit.


Assuntos
Técnicas de Rastreamento Neuroanatômico , Neurônios/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Sono/fisiologia , Transcriptoma , Animais , Biomarcadores/análise , Channelrhodopsins , Canais de Cloreto/metabolismo , Canais de Cloreto/efeitos da radiação , Colecistocinina/análise , Colecistocinina/genética , Hormônio Liberador da Corticotropina/análise , Hormônio Liberador da Corticotropina/genética , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/efeitos da radiação , Região Hipotalâmica Lateral/fisiologia , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Optogenética , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/efeitos da radiação , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ribossomos/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Sono/efeitos dos fármacos , Sono/efeitos da radiação , Taquicininas/análise , Taquicininas/genética , Vigília/fisiologia , Vigília/efeitos da radiação
5.
Nat Commun ; 7: 10268, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26743492

RESUMO

Agouti-related peptide (AgRP) neurons of the hypothalamus play a key role in regulating food intake and body weight, by releasing three different orexigenic molecules: AgRP; GABA; and neuropeptide Y. AgRP neurons express various G protein-coupled receptors (GPCRs) with different coupling properties, including Gs-linked GPCRs. At present, the potential role of Gs-coupled GPCRs in regulating the activity of AgRP neurons remains unknown. Here we show that the activation of Gs-coupled receptors expressed by AgRP neurons leads to a robust and sustained increase in food intake. We also provide detailed mechanistic data linking the stimulation of this class of receptors to the observed feeding phenotype. Moreover, we show that this pathway is clearly distinct from other GPCR signalling cascades that are operative in AgRP neurons. Our data suggest that drugs able to inhibit this signalling pathway may become useful for the treatment of obesity.


Assuntos
Proteína Relacionada com Agouti/genética , Ingestão de Alimentos/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Células HEK293 , Humanos , Hipotálamo/citologia , Imuno-Histoquímica , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neuropeptídeo Y/metabolismo , Técnicas de Patch-Clamp , Fosfoproteínas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo
6.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R41-54, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491097

RESUMO

The parabrachial nucleus is important for thermoregulation because it relays skin temperature information from the spinal cord to the hypothalamus. Prior work in rats localized thermosensory relay neurons to its lateral subdivision (LPB), but the genetic and neurochemical identity of these neurons remains unknown. To determine the identity of LPB thermosensory neurons, we exposed mice to a warm (36°C) or cool (4°C) ambient temperature. Each condition activated neurons in distinct LPB subregions that receive input from the spinal cord. Most c-Fos+ neurons in these LPB subregions expressed the transcription factor marker FoxP2. Consistent with prior evidence that LPB thermosensory relay neurons are glutamatergic, all FoxP2+ neurons in these subregions colocalized with green fluorescent protein (GFP) in reporter mice for Vglut2, but not for Vgat. Prodynorphin (Pdyn)-expressing neurons were identified using a GFP reporter mouse and formed a caudal subset of LPB FoxP2+ neurons, primarily in the dorsal lateral subnucleus (PBdL). Warm exposure activated many FoxP2+ neurons within PBdL. Half of the c-Fos+ neurons in PBdL were Pdyn+, and most of these project into the preoptic area. Cool exposure activated a separate FoxP2+ cluster of neurons in the far-rostral LPB, which we named the rostral-to-external lateral subnucleus (PBreL). These findings improve our understanding of LPB organization and reveal that Pdyn-IRES-Cre mice provide genetic access to warm-activated, FoxP2+ glutamatergic neurons in PBdL, many of which project to the hypothalamus.


Assuntos
Febre/metabolismo , Hipotermia/metabolismo , Neurônios/metabolismo , Núcleos Parabraquiais/metabolismo , Temperatura Cutânea , Sensação Térmica , Animais , Modelos Animais de Doenças , Encefalinas/genética , Encefalinas/metabolismo , Febre/genética , Febre/fisiopatologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Genótipo , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipotermia/genética , Hipotermia/fisiopatologia , Integrases/genética , Integrases/metabolismo , Sítios Internos de Entrada Ribossomal , Masculino , Camundongos Transgênicos , Técnicas de Rastreamento Neuroanatômico , Núcleos Parabraquiais/fisiopatologia , Fenótipo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Repressoras/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
7.
J Clin Invest ; 125(7): 2576-8, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098209

RESUMO

Activation of estrogen receptor α (ERα) in the brain prevents obesity as the result of increased energy expenditure and decreased food intake. While ERα is present on several neural populations, it is not clear how different regions of the brain mediate the weight-regulating effects of ERα activation. In this issue of the JCI, Xu and colleagues provide extensive evidence that ERα is abundant on neurons expressing single-minded-1 (SIM1) in the medial amygdala (MeA) and that loss of ERα in these cells enhances weight gain in both male and female mice, as the result of reduced physical activity. Moreover, focal deletion of ERα from the MeA recapitulated these alterations in energy homeostasis. Conversely, overexpression of ERα in the MeA partially prevented mice from diet-induced obesity, while chemogenetic activation of SIM1-expressing neurons in the MeA transiently promoted physical activity. The results of this study provide important insight into the regions of the brain that mediate ERα-dependent energy homeostasis.


Assuntos
Peso Corporal/fisiologia , Complexo Nuclear Corticomedial/metabolismo , Receptor alfa de Estrogênio/metabolismo , Animais , Feminino , Masculino
8.
Proc Natl Acad Sci U S A ; 111(36): 13193-8, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25157144

RESUMO

Activation of melanocortin-4 receptors (MC4Rs) restrains feeding and prevents obesity; however, the identity, location, and axonal projections of the neurons bearing MC4Rs that control feeding remain unknown. Reexpression of MC4Rs on single-minded 1 (SIM1)(+) neurons in mice otherwise lacking MC4Rs is sufficient to abolish hyperphagia. Thus, MC4Rs on SIM1(+) neurons, possibly in the paraventricular hypothalamus (PVH) and/or amygdala, regulate food intake. It is unknown, however, whether they are also necessary, a distinction required for excluding redundant sites of action. Hence, the location and nature of obesity-preventing MC4R-expressing neurons are unknown. Here, by deleting and reexpressing MC4Rs from cre-expressing neurons, establishing both necessity and sufficiency, we demonstrate that the MC4R-expressing neurons regulating feeding are SIM1(+), located in the PVH, glutamatergic and not GABAergic, and do not express oxytocin, corticotropin-releasing hormone, vasopressin, or prodynorphin. Importantly, these excitatory MC4R-expressing PVH neurons are synaptically connected to neurons in the parabrachial nucleus, which relays visceral information to the forebrain. This suggests a basis for the feeding-regulating effects of MC4Rs.


Assuntos
Comportamento Alimentar , Glutamatos/metabolismo , Neurônios/metabolismo , Núcleos Parabraquiais/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Sinapses/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Peso Corporal , Dependovirus/metabolismo , Metabolismo Energético , Neurônios GABAérgicos/metabolismo , Deleção de Genes , Injeções , Integrases/metabolismo , Camundongos , Neuropeptídeos/metabolismo , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes , Técnicas Estereotáxicas , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
9.
Nature ; 507(7491): 238-42, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24487620

RESUMO

Hunger is a hard-wired motivational state essential for survival. Agouti-related peptide (AgRP)-expressing neurons in the arcuate nucleus (ARC) at the base of the hypothalamus are crucial to the control of hunger. They are activated by caloric deficiency and, when naturally or artificially stimulated, they potently induce intense hunger and subsequent food intake. Consistent with their obligatory role in regulating appetite, genetic ablation or chemogenetic inhibition of AgRP neurons decreases feeding. Excitatory input to AgRP neurons is important in caloric-deficiency-induced activation, and is notable for its remarkable degree of caloric-state-dependent synaptic plasticity. Despite the important role of excitatory input, its source(s) has been unknown. Here, through the use of Cre-recombinase-enabled, cell-specific neuron mapping techniques in mice, we have discovered strong excitatory drive that, unexpectedly, emanates from the hypothalamic paraventricular nucleus, specifically from subsets of neurons expressing thyrotropin-releasing hormone (TRH) and pituitary adenylate cyclase-activating polypeptide (PACAP, also known as ADCYAP1). Chemogenetic stimulation of these afferent neurons in sated mice markedly activates AgRP neurons and induces intense feeding. Conversely, acute inhibition in mice with caloric-deficiency-induced hunger decreases feeding. Discovery of these afferent neurons capable of triggering hunger advances understanding of how this intense motivational state is regulated.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Fome/fisiologia , Vias Neurais/fisiologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteína Relacionada com Agouti/deficiência , Animais , Apetite/efeitos dos fármacos , Apetite/fisiologia , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Mapeamento Encefálico , Rastreamento de Células , Clozapina/análogos & derivados , Clozapina/farmacologia , Dependovirus/genética , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Feminino , Privação de Alimentos , Fome/efeitos dos fármacos , Integrases/metabolismo , Masculino , Camundongos , Vias Neurais/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Vírus da Raiva/genética , Resposta de Saciedade/fisiologia , Hormônio Liberador de Tireotropina/metabolismo
10.
J Neurosci ; 28(12): 3103-13, 2008 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-18354013

RESUMO

In Drosophila, formation of aversive olfactory long-term memory (LTM) requires multiple training sessions pairing odor and electric shock punishment with rest intervals. In contrast, here we show that a single 2 min training session pairing odor with a more ethologically relevant sugar reinforcement forms long-term appetitive memory that lasts for days. Appetitive LTM has some mechanistic similarity to aversive LTM in that it can be disrupted by cycloheximide, the dCreb2-b transcriptional repressor, and the crammer and tequila LTM-specific mutations. However, appetitive LTM is completely disrupted by the radish mutation that apparently represents a distinct mechanistic phase of consolidated aversive memory. Furthermore, appetitive LTM requires activity in the dorsal paired medial neuron and mushroom body alpha'beta' neuron circuit during the first hour after training and mushroom body alphabeta neuron output during retrieval, suggesting that appetitive middle-term memory and LTM are mechanistically linked. Last, experiments feeding and/or starving flies after training reveals a critical motivational drive that enables appetitive LTM retrieval.


Assuntos
Comportamento Apetitivo/fisiologia , Condicionamento Clássico/fisiologia , Memória/fisiologia , Odorantes , Biossíntese de Proteínas/fisiologia , Raphanus/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal , Condicionamento Clássico/efeitos dos fármacos , AMP Cíclico/farmacologia , Cicloeximida/farmacologia , Drosophila , Proteínas de Drosophila/genética , Privação de Alimentos/fisiologia , Memória/efeitos dos fármacos , Corpos Pedunculados/citologia , Mutação/fisiologia , Neurônios/fisiologia , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Temperatura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA