Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 146(6): 992-1003, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925320

RESUMO

Synaptic plasticity in response to changes in physiologic state is coordinated by hormonal signals across multiple neuronal cell types. Here, we combine cell-type-specific electrophysiological, pharmacological, and optogenetic techniques to dissect neural circuits and molecular pathways controlling synaptic plasticity onto AGRP neurons, a population that regulates feeding. We find that food deprivation elevates excitatory synaptic input, which is mediated by a presynaptic positive feedback loop involving AMP-activated protein kinase. Potentiation of glutamate release was triggered by the orexigenic hormone ghrelin and exhibited hysteresis, persisting for hours after ghrelin removal. Persistent activity was reversed by the anorexigenic hormone leptin, and optogenetic photostimulation demonstrated involvement of opioid release from POMC neurons. Based on these experiments, we propose a memory storage device for physiological state constructed from bistable synapses that are flipped between two sustained activity states by transient exposure to hormones signaling energy levels.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Pareamento Cromossômico , Retroalimentação Fisiológica , Fome , Memória , Neurônios/metabolismo , Proteína Relacionada com Agouti/metabolismo , Analgésicos Opioides/metabolismo , Animais , Cálcio/metabolismo , Grelina/metabolismo , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal , Pró-Opiomelanocortina/metabolismo , Rianodina/metabolismo , Transdução de Sinais
2.
Physiol Rev ; 98(1): 391-418, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351511

RESUMO

Chemogenetic technologies enable selective pharmacological control of specific cell populations. An increasing number of approaches have been developed that modulate different signaling pathways. Selective pharmacological control over G protein-coupled receptor signaling, ion channel conductances, protein association, protein stability, and small molecule targeting allows modulation of cellular processes in distinct cell types. Here, we review these chemogenetic technologies and instances of their applications in complex tissues in vivo and ex vivo.


Assuntos
Engenharia Genética/métodos , Canais Iônicos de Abertura Ativada por Ligante/genética , Técnicas de Sonda Molecular , Neurônios , Receptores Acoplados a Proteínas G/genética , Animais , Humanos , Canais Iônicos de Abertura Ativada por Ligante/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos
3.
Neuroendocrinology ; 114(5): 439-452, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38271999

RESUMO

INTRODUCTION: Postweaning social isolation (PWSI) in rodents is an advanced psychosocial stress model in early life. Some psychosocial stress, such as restrain and isolation, disrupts reproductive physiology in young and adult periods. Mechanisms of early-life stress effects on central regulation of reproduction need to be elucidated. We have investigated the effects of PWSI on function of arcuate kisspeptin (ARCKISS1) neurons by using electrophysiological techniques combining with monitoring of puberty onset and estrous cycle in male and female Kiss1-Cre mice. METHODS: Female mice were monitored for puberty onset with vaginal opening examination during social isolation. After isolation, the estrous cycle of female mice was monitored with vaginal cytology. Anxiety-like behavior of mice was determined by an elevated plus maze test. Effects of PWSI on electrophysiology of ARCKISS1 neurons were investigated by the patch clamp method after intracranial injection of AAV-GFP virus into arcuate nucleus of Kiss1-Cre mice after the isolation period. RESULTS: We found that both male and female isolated mice showed anxiety-like behavior. PWSI caused delay in vaginal opening and extension in estrous cycle length. Spontaneous-firing rates of ARCKISS1 neurons were significantly lower in the isolated male and female mice. The peak amplitude of inhibitory postsynaptic currents to ARCKISS1 neurons was higher in the isolated mice, while frequency of excitatory postsynaptic currents was higher in group-housed mice. CONCLUSION: These findings demonstrate that PWSI alters pre- and postpubertal reproductive physiology through metabolic and electrophysiological pathways.


Assuntos
Núcleo Arqueado do Hipotálamo , Ciclo Estral , Kisspeptinas , Neurônios , Maturidade Sexual , Isolamento Social , Animais , Kisspeptinas/metabolismo , Feminino , Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios/fisiologia , Neurônios/metabolismo , Masculino , Maturidade Sexual/fisiologia , Camundongos , Ciclo Estral/fisiologia , Camundongos Transgênicos , Ansiedade/fisiopatologia , Estresse Psicológico/fisiopatologia
4.
Hum Mol Genet ; 27(18): 3129-3136, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29878108

RESUMO

Prader-Willi Syndrome (PWS) is a neurodevelopmental disorder causing social and learning deficits, impaired satiety and severe childhood obesity. Genetic underpinning of PWS involves deletion of a chromosomal region with several genes, including MAGEL2, which is abundantly expressed in the hypothalamus. Of appetite regulating hypothalamic cell types, both AGRP and POMC-expressing neurons contain Magel2 transcripts but the functional impact of its deletion on these cells has not been fully characterized. Here, we investigated these key neurons in Magel2-null mice in terms of the activity levels at different energy states as well as their behavioral function. Using cell type specific ex vivo electrophysiological recordings and in vivo chemogenetic activation approaches we evaluated impact of Magel2 deletion on AGRP and POMC-neuron induced changes in appetite. Our results suggest that POMC neuron activity profile as well as its communication with downstream targets is significantly compromised, while AGRP neuron function with respect to short term feeding is relatively unaffected in Magel2 deficiency.


Assuntos
Proteína Relacionada com Agouti/genética , Antígenos de Neoplasias/genética , Apetite/genética , Síndrome de Prader-Willi/genética , Pró-Opiomelanocortina/genética , Proteínas/genética , Animais , Apetite/fisiologia , Deleção Cromossômica , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Camundongos Knockout , Neurônios/patologia , Obesidade/complicações , Obesidade/genética , Obesidade/fisiopatologia , Síndrome de Prader-Willi/complicações , Síndrome de Prader-Willi/fisiopatologia
5.
Neuroendocrinology ; 110(3-4): 258-270, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31154452

RESUMO

BACKGROUND: Melanin-concentrating hormone (MCH)-expressing neurons have been implicated in regulation of energy homeostasis and reward, yet the role of their electrical activity in short-term appetite and reward modulation has not been fully understood. OBJECTIVES: We investigated short-term behavioral and physiological effects of MCH neuron activity manipulations. METHODS: We used optogenetic and chemogenetic approaches in Pmch-cre transgenic mice to acutely stimulate/inhibit MCH neuronal activity while probing feeding, locomotor activity, anxiety-like behaviors, glucose homeostasis, and reward. RESULTS: MCH neuron activity is neither required nor sufficient for short-term appetite unless stimulation is temporally paired with consumption. MCH neuronal activation does not affect short-term locomotor activity, but inhibition improves glucose tolerance and is mildly anxiolytic. Finally, using two different operant tasks, we showed that activation of MCH neurons alone is sufficient to induce reward. CONCLUSIONS: Our results confirm diverse behavioral/physiological functions of MCH neurons and suggest a direct role in reward function.


Assuntos
Apetite/fisiologia , Comportamento Animal/fisiologia , Glicemia/metabolismo , Comportamento Alimentar/fisiologia , Hormônios Hipotalâmicos/metabolismo , Locomoção/fisiologia , Melaninas/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Recompensa , Animais , Feminino , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Optogenética
6.
Neurobiol Dis ; 121: 58-64, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30240706

RESUMO

Prader-Willi and the related Schaaf-Yang Syndromes (PWS/SYS) are rare neurodevelopmental disorders characterized by overlapping phenotypes of high incidence of autism spectrum disorders (ASD) and neonatal feeding difficulties. Based on clinical and basic studies, oxytocin pathway defects are suggested to contribute disease pathogenesis but the mechanism has been poorly understood. Specifically, whether the impairment in oxytocin system is limited to neuropeptide levels and how the functional properties of broader oxytocin neuron circuits affected in PWS/SYS have not been addressed. Using cell type specific electrophysiology, we investigated basic synaptic and cell autonomous properties of oxytocin neurons in the absence of MAGEL2; a hypothalamus enriched ubiquitin ligase regulator that is inactivated in both syndromes. We observed significant suppression of overall ex vivo oxytocin neuron activity, which was largely contributed by altered synaptic input profile; with reduced excitatory and increased inhibitory currents. Our results suggest that dysregulation of oxytocin system goes beyond altered neuropeptide expression and synaptic excitation inhibition imbalance impairs overall oxytocin pathway function.


Assuntos
Antígenos de Neoplasias/fisiologia , Hipotálamo/fisiologia , Potenciais da Membrana , Neurônios/fisiologia , Ocitocina/fisiologia , Proteínas/fisiologia , Potenciais de Ação , Animais , Antígenos de Neoplasias/genética , Potenciais Pós-Sinápticos Excitadores , Feminino , Potenciais Pós-Sinápticos Inibidores , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas/genética , Receptores de AMPA/metabolismo
7.
Nature ; 488(7410): 172-7, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22801496

RESUMO

Hunger is a complex behavioural state that elicits intense food seeking and consumption. These behaviours are rapidly recapitulated by activation of starvation-sensitive AGRP neurons, which present an entry point for reverse-engineering neural circuits for hunger. Here we mapped synaptic interactions of AGRP neurons with multiple cell populations in mice and probed the contribution of these distinct circuits to feeding behaviour using optogenetic and pharmacogenetic techniques. An inhibitory circuit with paraventricular hypothalamus (PVH) neurons substantially accounted for acute AGRP neuron-evoked eating, whereas two other prominent circuits were insufficient. Within the PVH, we found that AGRP neurons target and inhibit oxytocin neurons, a small population that is selectively lost in Prader-Willi syndrome, a condition involving insatiable hunger. By developing strategies for evaluating molecularly defined circuits, we show that AGRP neuron suppression of oxytocin neurons is critical for evoked feeding. These experiments reveal a new neural circuit that regulates hunger state and pathways associated with overeating disorders.


Assuntos
Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Fome/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Axônios/metabolismo , Feminino , Privação de Alimentos , Masculino , Camundongos , Modelos Neurológicos , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Pró-Opiomelanocortina/metabolismo , Inanição , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo
8.
Surg Endosc ; 30(10): 4624-5, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26902612

RESUMO

Complete mesocolic excision (CME) with central vascular ligation for right-sided colon cancer has been proven to provide superior oncologic outcomes and survival advantage when compared to standard lymphadenectomy [1]. A number of studies comparing conventional laparoscopic versus open CME have shown feasibility and safety of the laparoscopic approach with acceptable oncological profile and postoperative outcomes [2, 3]. The introduction of robotic systems with its technical advantages, including improved vision, better ergonomics and precise dissection, has further revolutionized minimally invasive approach in colorectal surgery. However, there seems to be a relatively slow adoption of robotic approach in the CME technique for right-sided colon cancer. This video demonstrates our detailed operative technique and feasibility for performing right-sided CME robotically. The surgical procedure is performed with a medial-to-lateral approach through four 8-mm robotic and one assistant ports. First, the ileocolic vessels are isolated, clipped and transected near their origins. Cephalad dissection continues along the ventral aspect of the superior mesenteric vein. Staying in the embryological planes between the mesocolon and retroperitoneal structures, mesenteric dissection is extended up to the root of the right colic vessels, if present, and the middle colic vessels, which are clipped and divided individually near their origins. After the terminal ileum is transected using an endolinear staple, the colon is mobilized fully from gastrocolic tissue and then from its lateral attachments. The transverse colon is transected under the guidance of near-infrared fluorescence imaging. Creation of an intracorporeal side-to-side ileotransversostomy anastomosis and extraction of the specimen complete the operation. We consider robotic CME to be feasible, safe and oncologically adequate for the treatment of right-sided colon cancer. Its technical advantages may lead to further dissemination of the robotic approach and better standardization of this surgical technique.


Assuntos
Adenocarcinoma/cirurgia , Colectomia/métodos , Neoplasias do Colo/cirurgia , Mesocolo/cirurgia , Procedimentos Cirúrgicos Robóticos/métodos , Idoso de 80 Anos ou mais , Colo Ascendente/cirurgia , Colo Transverso/cirurgia , Dissecação/métodos , Humanos , Laparoscopia/métodos , Ligadura , Excisão de Linfonodo/métodos , Masculino , Veias Mesentéricas
9.
Neuroendocrinology ; 100(2-3): 95-102, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25402352

RESUMO

New tools for mapping and manipulating molecularly defined neural circuits have improved the understanding of how the central nervous system regulates appetite. Studies that focused on Agouti-related protein neurons, a starvation-sensitive hypothalamic population, have identified multiple circuit elements that can elicit or suppress feeding behavior. Distinct axon projections of this neuron population point to different circuits that regulate long-term appetite, short-term feeding, or visceral malaise-mediated anorexia. Here, we review recent studies examining these neural circuits that control food intake.


Assuntos
Regulação do Apetite/fisiologia , Encéfalo/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Anorexia/fisiopatologia , Comportamento Apetitivo/fisiologia , Encéfalo/fisiopatologia , Vias Neurais/fisiologia , Neurônios/fisiologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-38326456

RESUMO

Stress is thought to be an important contributing factor for eating disorders; however, neural substrates underlying the complex relationship between stress and appetite are not fully understood. Using in vivo recordings from awake behaving mice, we show that various acute stressors activate catecholaminergic nucleus tractus solitarius (NTSTH) projections in the paraventricular hypothalamus (PVH). Remarkably, the resulting adrenergic tone inhibits MC4R-expressing neurons (PVHMC4R), which are known for their role in feeding suppression. We found that PVHMC4R silencing encodes negative valence in sated mice and is required for avoidance induced by visceral malaise. Collectively, these findings establish PVHMC4R neurons as an effector of stress-activated brainstem adrenergic input in addition to the well-established hypothalamic-pituitary-adrenal axis. Convergent modulation of stress and feeding by PVHMC4R neurons implicates NTSTH → PVHMC4R input in stress-associated appetite disorders.

11.
Nat Neurosci ; 27(1): 102-115, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37957320

RESUMO

Food intake follows a predictable daily pattern and synchronizes metabolic rhythms. Neurons expressing agouti-related protein (AgRP) read out physiological energetic state and elicit feeding, but the regulation of these neurons across daily timescales is poorly understood. Using a combination of neuron dynamics measurements and timed optogenetic activation in mice, we show that daily AgRP-neuron activity was not fully consistent with existing models of homeostatic regulation. Instead of operating as a 'deprivation counter', AgRP-neuron activity primarily followed the circadian rest-activity cycle through a process that required an intact suprachiasmatic nucleus and synchronization by light. Imposing novel feeding patterns through time-restricted food access or periodic AgRP-neuron stimulation was sufficient to resynchronize the daily AgRP-neuron activity rhythm and drive anticipatory-like behavior through a process that required DMHPDYN neurons. These results indicate that AgRP neurons integrate time-of-day information of past feeding experience with current metabolic needs to predict circadian feeding time.


Assuntos
Neurônios , Núcleo Supraquiasmático , Animais , Camundongos , Proteína Relacionada com Agouti , Comportamento Alimentar/fisiologia , Neurônios/fisiologia
12.
Cell Rep ; 43(1): 113630, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38165803

RESUMO

Opioids are generally known to promote hedonic food consumption. Although much of the existing evidence is primarily based on studies of the mesolimbic pathway, endogenous opioids and their receptors are widely expressed in hypothalamic appetite circuits as well; however, their role in homeostatic feeding remains unclear. Using a fluorescent opioid sensor, deltaLight, here we report that mediobasal hypothalamic opioid levels increase by feeding, which directly and indirectly inhibits agouti-related protein (AgRP)-expressing neurons through the µ-opioid receptor (MOR). AgRP-specific MOR expression increases by energy surfeit and contributes to opioid-induced suppression of appetite. Conversely, its antagonists diminish suppression of AgRP neuron activity by food and satiety hormones. Mice with AgRP neuron-specific ablation of MOR expression have increased fat preference without increased motivation. These results suggest that post-ingestion release of endogenous opioids contributes to AgRP neuron inhibition to shape food choice through MOR signaling.


Assuntos
Analgésicos Opioides , Neurônios , Animais , Camundongos , Proteína Relacionada com Agouti/metabolismo , Analgésicos Opioides/farmacologia , Ingestão de Alimentos , Hipotálamo/metabolismo , Neurônios/metabolismo , Transdução de Sinais
13.
Mol Metab ; 82: 101904, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38395148

RESUMO

OBJECTIVE: The prevalence of obesity has increased over the past three decades. Proopiomelanocortin (POMC) neurons in the hypothalamic arcuate nucleus (ARC) play a vital role in induction of satiety. Chronic consumption of high-fat diet is known to reduce hypothalamic neuronal sensitivity to hormones like leptin, thus contributing to the development and persistence of obesity. The functional and morphological effects of a high-calorie diet on POMC neurons and how these effects contribute to the development and maintenance of the obese phenotype are not fully understood. For this purpose, POMC-Cre transgenic mice model was exposed to high-fat diet (HFD) and at the end of a 3- and 6-month period, electrophysiological and morphological changes, and the role of POMC neurons in homeostatic nutrition and their response to leptin were thoroughly investigated. METHODS: Effects of HFD on POMC-satiety neurons in transgenic mice models exposed to chronic high-fat diet were investigated using electrophysiological (patch-clamp), chemogenetic and Cre recombinase advanced technological methods. Leptin, glucose and lipid profiles were determined and analyzed. RESULTS: In mice exposed to a high-fat diet for 6 months, no significant changes in POMC dendritic spine number or projection density from POMC neurons to the paraventricular hypothalamus (PVN), lateral hypothalamus (LH), and bed nucleus stria terminalis (BNST) were observed. It was revealed that leptin hormone did not change the electrophysiological activities of POMC neurons in mice fed with HFD for 6 months. In addition, chemogenetic stimulation of POMC neurons increased HFD consumption. In the 3-month HFD-fed group, POMC activation induced an orexigenic response in mice, whereas switching to a standard diet was found to abolish orexigenic behavior in POMC mice. CONCLUSIONS: Chronic high fat consumption disrupts the regulation of POMC neuron activation by leptin. Altered POMC neuron activation abolished the neuron's characteristic behavioral anorexigenic response. Change in nutritional content contributes to the reorganization of developing maladaptations.


Assuntos
Dieta Hiperlipídica , Leptina , Camundongos , Animais , Dieta Hiperlipídica/efeitos adversos , Leptina/metabolismo , Pró-Opiomelanocortina/metabolismo , Hipotálamo/metabolismo , Obesidade , Neurônios/metabolismo , Camundongos Transgênicos
14.
Hepatogastroenterology ; 60(124): 896-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23321061

RESUMO

BACKGROUND/AIMS: The efficacy of endoscopic ultrasound with fine-needle aspiration (EUS-FNA) in the diagnosis and staging of pancreatic malignancy is quite well established. The aim of this study is to describe a single-centre's experience. METHODOLOGY: Data were collected retrospectively on all patients with solid pancreatic masses undergoing EUS-FNA from January 2006 to March 2011. In tumor cases, TNM staging using EUS was compared with the results of histopathological staging. RESULTS: EUS-FNA of pancreatic lesions was performed in 125 patients. Of these patients, data of 75 were available (69% men, mean age 59.97±11.12 (SD) years); 58 (76%) of the lesions were ductal adenocarcinoma, 11 (14.5%) were chronic pancreatitis, 4 (%5) were intraductal papillary mucinous carcinoma (IPMN) and 2 (%3) were insulinoma. Diagnostic yield of the EUS-FNA procedure was 74.7% (56/75). Specimens from six patients were inadequate. In multivariate analysis, lesion diameter below 2cm was an independent predictor for the inadequacy of material (p=0.04). CONCLUSIONS: In patients with pancreatic mass with suspected malignancy, EUS-FNA provided a diagnosis with accuracy rate of 75%. Inadequate material with EUS-FNA was significantly more frequent in lesions below 2cm.


Assuntos
Biópsia por Agulha Fina , Endossonografia , Pancreatopatias/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pancreatopatias/diagnóstico por imagem , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/patologia , Estudos Retrospectivos , Sensibilidade e Especificidade
15.
Cell Metab ; 35(5): 723-725, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37137283

RESUMO

Dieting often fails in the long run becuase of an ever-growing urge to eat. In this issue of Cell Metabolism, Grzelka et al. unveil a brain circuit that is potentiated during caloric restriction and incites rebound increases in food consumption and body weight.


Assuntos
Ingestão de Alimentos , Fome , Humanos , Peso Corporal , Restrição Calórica , Sistema Nervoso Central , Ingestão de Energia
16.
Mol Metab ; 69: 101676, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36682413

RESUMO

OBJECTIVE: Serotonin (5HT) is a well-known anorexigenic molecule, and 5HT neurons of dorsal raphe nucleus (DRN) have been implicated in suppression of feeding; however, the downstream circuitry is poorly understood. Here we explored major projections of DRN5HT neurons for their capacity to modulate feeding. METHODS: We used optogenetics to selectively activate DRN5HT axonal projections in hypothalamic and extrahypothalamic areas and monitored food intake. We next used fiber photometry to image the activity dynamics of DRN5HT axons and 5HT levels in projection areas in response feeding and metabolic hormones. Finally, we used electrophysiology to determine how DRN5HT axons affect downstream neuron activity. RESULTS: We found that selective activation of DRN5HT axons in (DRN5HT → LH) and (DRN5HT → BNST) suppresses feeding whereas activating medial hypothalamic projections has no effect. Using in vivo imaging, we found that food access and satiety hormones activate DRN5HT projections to LH where they also rapidly increase extracellular 5HT levels. Optogenetic mapping revealed that DRN5HT → LHvGAT and DRN5HT → LHvGlut2 connections are primarily inhibitory and excitatory respectively. Further, in addition to its direct action on LH neurons, we found that 5HT suppresses GABA release from presynaptic terminals arriving from AgRP neurons. CONCLUSIONS: These findings define functionally redundant forebrain circuits through which DRN5HT neurons suppress feeding and reveal that these projections can be modulated by metabolic hormones.


Assuntos
Núcleo Dorsal da Rafe , Neurônios Serotoninérgicos , Núcleo Dorsal da Rafe/metabolismo , Neurônios Serotoninérgicos/metabolismo , Serotonina/metabolismo , Hipotálamo/metabolismo , Hormônios
17.
Endocrinology ; 164(11)2023 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-37823477

RESUMO

The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.


Assuntos
Hormônio Liberador da Corticotropina , Grelina , Camundongos , Masculino , Animais , Hormônio Liberador da Corticotropina/metabolismo , Grelina/farmacologia , Grelina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Neurônios/metabolismo
18.
Nat Commun ; 14(1): 6602, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37857606

RESUMO

Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.


Assuntos
Fome , Melanocortinas , Melanocortinas/metabolismo , Adrenérgicos/metabolismo , Apetite , Núcleo Hipotalâmico Paraventricular/metabolismo , Norepinefrina/metabolismo , Hipoglicemiantes/metabolismo
19.
Cell Rep ; 42(8): 112935, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37540598

RESUMO

Resting metabolic rate (RMR) adaptation occurs during obesity and is hypothesized to contribute to failed weight management. Angiotensin II (Ang-II) type 1 (AT1A) receptors in Agouti-related peptide (AgRP) neurons contribute to the integrative control of RMR, and deletion of AT1A from AgRP neurons causes RMR adaptation. Extracellular patch-clamp recordings identify distinct cellular responses of individual AgRP neurons from lean mice to Ang-II: no response, inhibition via AT1A and Gαi, or stimulation via Ang-II type 2 (AT2) receptors and Gαq. Following diet-induced obesity, a subset of Ang-II/AT1A-inhibited AgRP neurons undergo a spontaneous G-protein "signal switch," whereby AT1A stop inhibiting the cell via Gαi and instead begin stimulating the cell via Gαq. DREADD-mediated activation of Gαi, but not Gαq, in AT1A-expressing AgRP cells stimulates RMR in lean and obese mice. Thus, loss of AT1A-Gαi coupling within the AT1A-expressing AgRP neuron subtype represents a molecular mechanism contributing to RMR adaptation.


Assuntos
Neurônios , Obesidade , Receptor Tipo 1 de Angiotensina , Animais , Camundongos , Proteína Relacionada com Agouti/metabolismo , Angiotensina II/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo
20.
Cell Metab ; 34(2): 317-328.e6, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108517

RESUMO

Excessive alcohol consumption is a major health and social issue in our society. Pharmacologic administration of the endocrine hormone fibroblast growth factor 21 (FGF21) suppresses alcohol consumption through actions in the brain in rodents, and genome-wide association studies have identified single nucleotide polymorphisms in genes involved with FGF21 signaling as being associated with increased alcohol consumption in humans. However, the neural circuit(s) through which FGF21 signals to suppress alcohol consumption are unknown, as are its effects on alcohol consumption in higher organisms. Here, we demonstrate that administration of an FGF21 analog to alcohol-preferring non-human primates reduces alcohol intake by 50%. Further, we reveal that FGF21 suppresses alcohol consumption through a projection-specific subpopulation of KLB-expressing neurons in the basolateral amygdala. Our results illustrate how FGF21 suppresses alcohol consumption through a specific population of neurons in the brain and demonstrate its therapeutic potential in non-human primate models of excessive alcohol consumption.


Assuntos
Fatores de Crescimento de Fibroblastos , Estudo de Associação Genômica Ampla , Consumo de Bebidas Alcoólicas , Animais , Sistema Endócrino/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA