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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 152(3): 612-9, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374353

RESUMO

Melanocortin 4 receptors (MC4Rs) in the central nervous system are key regulators of energy and glucose homeostasis. Notably, obese patients with MC4R mutations are hyperinsulinemic and resistant to obesity-induced hypertension. Although these effects are probably dependent upon the activity of the autonomic nervous system, the cellular effects of MC4Rs on parasympathetic and sympathetic neurons remain undefined. Here, we show that MC4R agonists inhibit parasympathetic preganglionic neurons in the brainstem. In contrast, MC4R agonists activate sympathetic preganglionic neurons in the spinal cord. Deletion of MC4Rs in cholinergic neurons resulted in elevated levels of insulin. Furthermore, re-expression of MC4Rs specifically in cholinergic neurons (including sympathetic preganglionic neurons) restores obesity-associated hypertension in MC4R null mice. These findings provide a cellular correlate of the autonomic side effects associated with MC4R agonists and demonstrate a role for MC4Rs expressed in cholinergic neurons in the regulation of insulin levels and in the development of obesity-induced hypertension.


Assuntos
Tronco Encefálico/metabolismo , Insulina/metabolismo , Neurônios/metabolismo , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Pressão Sanguínea , Tronco Encefálico/citologia , Neurônios Colinérgicos/metabolismo , AMP Cíclico/metabolismo , Fenômenos Eletrofisiológicos , Humanos , Canais KATP/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Obesidade/fisiopatologia , Sistema Nervoso Parassimpático/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Medula Espinal/metabolismo , Sistema Nervoso Simpático/metabolismo
2.
PLoS Biol ; 21(8): e3002252, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37594983

RESUMO

It is well known that the neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons increase appetite and decrease thermogenesis. Previous studies demonstrated that optogenetic and/or chemogenetic manipulations of NPY/AgRP neuronal activity alter food intake and/or energy expenditure (EE). However, little is known about intrinsic molecules regulating NPY/AgRP neuronal excitability to affect long-term metabolic function. Here, we found that the G protein-gated inwardly rectifying K+ (GIRK) channels are key to stabilize NPY/AgRP neurons and that NPY/AgRP neuron-selective deletion of the GIRK2 subunit results in a persistently increased excitability of the NPY/AgRP neurons. Interestingly, increased body weight and adiposity observed in the NPY/AgRP neuron-selective GIRK2 knockout mice were due to decreased sympathetic activity and EE, while food intake remained unchanged. The conditional knockout mice also showed compromised adaptation to coldness. In summary, our study identified GIRK2 as a key determinant of NPY/AgRP neuronal excitability and driver of EE in physiological and stress conditions.


Assuntos
Adiposidade , Proteína Relacionada com Agouti , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Obesidade , Animais , Camundongos , Proteína Relacionada com Agouti/genética , Peso Corporal , Camundongos Knockout , Neurônios , Peptídeos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética
3.
EMBO J ; 40(15): e107121, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34013588

RESUMO

Glia contribute to synapse elimination through phagocytosis in the central nervous system. Despite the important roles of this process in development and neurological disorders, the identity and regulation of the "eat-me" signal that initiates glia-mediated phagocytosis of synapses has remained incompletely understood. Here, we generated conditional knockout mice with neuronal-specific deletion of the flippase chaperone Cdc50a, to induce stable exposure of phosphatidylserine, a well-known "eat-me" signal for apoptotic cells, on the neuronal outer membrane. Surprisingly, acute Cdc50a deletion in mature neurons causes preferential phosphatidylserine exposure in neuronal somas and specific loss of inhibitory post-synapses without effects on other synapses, resulting in abnormal excitability and seizures. Ablation of microglia or the deletion of microglial phagocytic receptor Mertk prevents the loss of inhibitory post-synapses and the seizure phenotype, indicating that microglial phagocytosis is responsible for inhibitory post-synapse elimination. Moreover, we found that phosphatidylserine is used for microglia-mediated pruning of inhibitory post-synapses in normal brains, suggesting that phosphatidylserine serves as a general "eat-me" signal for inhibitory post-synapse elimination.


Assuntos
Microglia/metabolismo , Fosfatidilserinas/metabolismo , Convulsões/fisiopatologia , Sinapses/fisiologia , c-Mer Tirosina Quinase/metabolismo , Animais , Encéfalo/fisiopatologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fagocitose/fisiologia , Convulsões/genética , c-Mer Tirosina Quinase/genética
4.
Ann Neurol ; 90(2): 285-299, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34180075

RESUMO

OBJECTIVE: Low-level somatic mosaicism in the brain has been shown to be a major genetic cause of intractable focal epilepsy. However, how a relatively few mutation-carrying neurons are able to induce epileptogenesis at the local network level remains poorly understood. METHODS: To probe the origin of epileptogenesis, we measured the excitability of neurons with MTOR mutation and nearby nonmutated neurons recorded by whole-cell patch-clamp and array-based electrodes comparing the topographic distribution of mutation. Computational simulation is used to understand neural network-level changes based on electrophysiological properties. To examine the underlying mechanism, we measured inhibitory and excitatory synaptic inputs in mutated neurons and nearby neurons by electrophysiological and histological methods using the mouse model and postoperative human brain tissue for cortical dysplasia. To explain non-cell-autonomous hyperexcitability, an inhibitor of adenosine kinase was injected into mice to enhance adenosine signaling and to mitigate hyperactivity of nearby nonmutated neurons. RESULTS: We generated mice with a low-level somatic mutation in MTOR presenting spontaneous seizures. The seizure-triggering hyperexcitability originated from nonmutated neurons near mutation-carrying neurons, which proved to be less excitable than nonmutated neurons. Interestingly, the net balance between excitatory and inhibitory synaptic inputs onto mutated neurons remained unchanged. Additionally, we found that inhibition of adenosine kinase, which affects adenosine metabolism and neuronal excitability, reduced the hyperexcitability of nonmutated neurons. INTERPRETATION: This study shows that neurons carrying somatic mutations in MTOR lead to focal epileptogenesis via non-cell-autonomous hyperexcitability of nearby nonmutated neurons. ANN NEUROL 2021;90:285-299.


Assuntos
Epilepsias Parciais/genética , Epilepsias Parciais/fisiopatologia , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/fisiopatologia , Serina-Treonina Quinases TOR/genética , Adolescente , Animais , Criança , Pré-Escolar , Eletroencefalografia/métodos , Epilepsias Parciais/diagnóstico por imagem , Feminino , Humanos , Masculino , Malformações do Desenvolvimento Cortical/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Gravidez
5.
Int J Mol Sci ; 22(7)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810547

RESUMO

The hypothalamic regulation of appetite governs whole-body energy balance. Satiety is regulated by endocrine factors including leptin, and impaired leptin signaling is associated with obesity. Despite the anorectic effect of leptin through the regulation of the hypothalamic feeding circuit, a distinct downstream mediator of leptin signaling in neuron remains unclear. Angiopoietin-like growth factor (AGF) is a peripheral activator of energy expenditure and antagonizes obesity. However, the regulation of AGF expression in brain and localization to mediate anorectic signaling is unknown. Here, we demonstrated that AGF is expressed in proopiomelanocortin (POMC)-expressing neurons located in the arcuate nucleus (ARC) of the hypothalamus. Unlike other brain regions, hypothalamic AGF expression is stimulated by leptin-induced signal transducers and activators of transcription 3 (STAT3) phosphorylation. In addition, leptin treatment to hypothalamic N1 cells significantly enhanced the promoter activity of AGF. This induction was abolished by the pretreatment of ruxolitinib, a leptin signaling inhibitor. These results indicate that hypothalamic AGF expression is induced by leptin and colocalized to POMC neurons.


Assuntos
Proteínas Semelhantes a Angiopoietina/genética , Proteínas Semelhantes a Angiopoietina/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Transdução de Sinais , Proteína 6 Semelhante a Angiopoietina , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Encéfalo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fosforilação , Pró-Opiomelanocortina/metabolismo , Fator de Transcrição STAT3/metabolismo
6.
Pflugers Arch ; 472(11): 1547-1561, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32960363

RESUMO

Glucose is a major source of energy in animals. Maintaining blood glucose levels within a physiological range is important for facilitating glucose uptake by cells, as required for optimal functioning. Glucose homeostasis relies on multiple glucose-sensing cells in the body that constantly monitor blood glucose levels and respond accordingly to adjust its glycemia. These include not only pancreatic ß-cells and α-cells that secrete insulin and glucagon, but also central and peripheral neurons regulating pancreatic endocrine function. Different types of cells respond distinctively to changes in blood glucose levels, and the mechanisms involved in glucose sensing are diverse. Notably, recent studies have challenged the currently held views regarding glucose-sensing mechanisms. Furthermore, peripheral and central glucose-sensing cells appear to work in concert to control blood glucose level and maintain glucose and energy homeostasis in organisms. In this review, we summarize the established concepts and recent advances in the understanding of cellular and systemic mechanisms that regulate glucose sensing and its homeostasis.


Assuntos
Glucose/metabolismo , Transdução de Sinais , Animais , Homeostase , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Potenciais da Membrana
7.
J Physiol ; 596(17): 4141-4156, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29870060

RESUMO

KEY POINTS: High-frequency stimulation (HFS) of the Schaffer collateral pathway activates metabotropic glutamate receptor 5 (mGluR5) signalling in the proximal apical dendrites of CA1 pyramidal neurons. The synaptic activation of mGluR5-mediated calcium signalling causes a significant increase in persistent sodium current (INa,P ) in the dendrites. Increased INa,P by HFS underlies potentiation of synaptic inputs at both the proximal and distal dendrite, leading to an enhanced probability of action potential firing associated with decreased action potential thresholds. Therefore, HFS-induced activation of intracellular mGluR5 serves an important role as an instructive signal for potentiation of upcoming inputs by increasing dendritic excitability. ABSTRACT: Dendritic Na+ channels in pyramidal neurons are known to amplify synaptic signals, thereby facilitating action potential (AP) generation. However, the mechanisms that modulate dendritic Na+ channels have remained largely uncharacterized. Here, we report a new form of short-term plasticity in which proximal excitatory synaptic inputs to hippocampal CA1 pyramidal neurons transiently elevate dendritic excitability. High-frequency stimulations (HFS) to the Schaffer collateral (SC) pathway activate mGluR5-dependent Ca2+ signalling in the apical dendrites, which, with calmodulin, upregulates specifically Nav1.6 channel-mediated persistent Na+ currents (INa,P ) in the dendrites. This HFS-induced increase in dendritic INa,P results in transient increases in the amplitude of excitatory postsynaptic potentials induced by both proximal SC and distal perforant path stimulation, leading to the enhanced probability of AP firing associated with decreased AP thresholds. Taken together, our study identifies dendritic INa,P as a novel target for mediating activity-dependent modulation of dendritic integration and neuronal output.


Assuntos
Potenciais de Ação , Região CA1 Hipocampal/fisiologia , Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores , Células Piramidais/fisiologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Sódio/metabolismo , Animais , Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.6/química , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Células Piramidais/citologia , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
8.
Pharmacol Res ; 129: 10-19, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29329999

RESUMO

It is well known that melanocortin-4 receptors (MC4Rs) and central melanocortin pathways regulate food intake, energy expenditure, and glucose homeostasis. Importantly, MC4R deficiency is the most common monogenic cause of human obesity. Interestingly, MC4Rs expressed by distinct central nuclei are responsible for the different physiological function of MC4R stimulation. In addition, MC4Rs activate multiple intracellular and/or synaptic signaling molecules for the regulation of neuronal circuits. Therefore, MC4Rs and the downstream signal molecules are plausible targets for development of novel therapeutics against obesity and obesity-related metabolic disorders. In this review, we discuss recent findings on the neuronal circuits and signaling molecules that are responsible for MC4R control energy balance and autonomic function. Further, we review status of MC4R agonists as novel therapeutics for obesity syndrome. We believe that comprehensive understanding of signaling molecules involved in MC4R control of neuronal circuits will help to design MC4R agonists as safe and effective anti-obesity drugs.


Assuntos
Neurônios/fisiologia , Obesidade/tratamento farmacológico , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Humanos , Ligantes , Obesidade/fisiopatologia , Receptor Tipo 4 de Melanocortina/agonistas , Transmissão Sináptica , Síndrome
9.
J Neurosci ; 35(16): 6401-12, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904792

RESUMO

Glutamate, a major neurotransmitter in the brain, activates ionotropic and metabotropic glutamate receptors (iGluRs and mGluRs, respectively). The two types of glutamate receptors interact with each other, as exemplified by the modulation of iGluRs by mGluRs. However, the other way of interaction (i.e., modulation of mGluRs by iGluRs) has not received much attention. In this study, we found that group I mGluR-specific agonist (RS)-3,5-dihydroxyphenylglycine (DHPG) alone is not sufficient to activate phospholipase C (PLC) in rat hippocampus, while glutamate robustly activates PLC. These results suggested that additional mechanisms provided by iGluRs are involved in group I mGluR-mediated PLC activation. A series of experiments demonstrated that glutamate-induced PLC activation is mediated by mGluR5 and is facilitated by local Ca(2+) signals that are induced by AMPA-mediated depolarization and L-type Ca(2+) channel activation. Finally, we found that PLC and L-type Ca(2+) channels are involved in hippocampal mGluR-dependent long-term depression (mGluR-LTD) induced by paired-pulse low-frequency stimulation, but not in DHPG-induced chemical LTD. Together, we propose that AMPA receptors initiate Ca(2+) influx via the L-type Ca(2+) channels that facilitate mGluR5-PLC signaling cascades, which underlie mGluR-LTD in rat hippocampus.


Assuntos
Ácido Glutâmico/fisiologia , Hipocampo/enzimologia , Hipocampo/metabolismo , Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores de AMPA/agonistas , Receptores de Glutamato Metabotrópico/agonistas , Fosfolipases Tipo C/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Ratos , Receptor de Glutamato Metabotrópico 5/agonistas , Receptores de AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Resorcinóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
10.
Proc Natl Acad Sci U S A ; 110(31): 12673-8, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858470

RESUMO

Leptin is a pivotal regulator of energy and glucose homeostasis, and defects in leptin signaling result in obesity and diabetes. The ATP-sensitive potassium (K(ATP)) channels couple glucose metabolism to insulin secretion in pancreatic ß-cells. In this study, we provide evidence that leptin modulates pancreatic ß-cell functions by promoting K(ATP) channel translocation to the plasma membrane via AMP-activated protein kinase (AMPK) signaling. K(ATP) channels were localized mostly to intracellular compartments of pancreatic ß-cells in the fed state and translocated to the plasma membrane in the fasted state. This process was defective in leptin-deficient ob/ob mice, but restored by leptin treatment. We discovered that the molecular mechanism of leptin-induced AMPK activation involves canonical transient receptor potential 4 and calcium/calmodulin-dependent protein kinase kinase ß. AMPK activation was dependent on both leptin and glucose concentrations, so at optimal concentrations of leptin, AMPK was activated sufficiently to induce K(ATP) channel trafficking and hyperpolarization of pancreatic ß-cells in a physiological range of fasting glucose levels. There was a close correlation between phospho-AMPK levels and ß-cell membrane potentials, suggesting that AMPK-dependent K(ATP) channel trafficking is a key mechanism for regulating ß-cell membrane potentials. Our results present a signaling pathway whereby leptin regulates glucose homeostasis by modulating ß-cell excitability.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células Secretoras de Insulina/metabolismo , Leptina/metabolismo , Potenciais da Membrana/fisiologia , Transdução de Sinais/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Glucose/metabolismo , Homeostase/fisiologia , Células Secretoras de Insulina/citologia , Leptina/genética , Camundongos , Camundongos Obesos , Transporte Proteico/fisiologia , ATPase Trocadora de Sódio-Potássio/genética , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo
11.
Soa Chongsonyon Chongsin Uihak ; 35(2): 150, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38601102

RESUMO

[This corrects the article on p. 57 in vol. 35, PMID: 38204745.].

12.
Soa Chongsonyon Chongsin Uihak ; 35(1): 57-65, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38204745

RESUMO

Autism spectrum disorder (ASD) can be associated with eating problems. However, currently, there is a lack of established guidelines for assessing and addressing eating behaviors in individuals with ASD. This gap in research exists due to the challenges associated with using traditional assessment methods, which may lead to discrepancies in responses and unintentional potential biases from caregivers. In this review, we provided a comprehensive overview of various eating behaviors commonly observed in individuals with ASD. These behaviors include 1) food neophobia, 2) selective eating, 3) binge eating, 4) food avoidance, 5) chewing and swallowing problems, 6) pica, 7) rumination, 8) rituals, and 9) problematic behaviors. Furthermore, we provide a perspective of utilizing digital tools: 1) augmentative and alternative communication; 2) ecological momentary assessment; and 3) video analysis, behavioral analysis, and facial expression analysis. This review explores existing assessment methods and suggests novel assessment aiding together.

13.
Sci Adv ; 10(27): eadj4433, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38959322

RESUMO

Memory processes rely on a molecular signaling system that balances the interplay between positive and negative modulators. Recent research has focused on identifying memory-regulating genes and their mechanisms. Phospholipase C beta 1 (PLCß1), highly expressed in the hippocampus, reportedly serves as a convergence point for signal transduction through G protein-coupled receptors. However, the detailed role of PLCß1 in memory function has not been elucidated. Here, we demonstrate that PLCß1 in the dentate gyrus functions as a memory suppressor. We reveal that mice lacking PLCß1 in the dentate gyrus exhibit a heightened fear response and impaired memory extinction, and this excessive fear response is repressed by upregulation of PLCß1 through its overexpression or activation using a newly developed optogenetic system. Last, our results demonstrate that PLCß1 overexpression partially inhibits exaggerated fear response caused by traumatic experience. Together, PLCß1 is crucial in regulating contextual fear memory formation and potentially enhancing the resilience to trauma-related conditions.


Assuntos
Giro Denteado , Medo , Memória , Neurônios , Fosfolipase C beta , Animais , Fosfolipase C beta/metabolismo , Fosfolipase C beta/genética , Medo/fisiologia , Giro Denteado/metabolismo , Giro Denteado/fisiologia , Memória/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Camundongos Knockout , Masculino , Optogenética , Camundongos Endogâmicos C57BL
14.
STAR Protoc ; 4(2): 102345, 2023 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-37270782

RESUMO

The spinal cord is a part of the central nervous system located within the spinal canal of the vertebrae. Here, we present a protocol to prepare mouse spinal cord sections for patch-clamp and histology experiments. We describe steps for isolating spinal cord from the spinal canal and obtaining acute slices for patch-clamp experiments. For histology experiments, we detail fixing spinal cord for cryosectioning and imaging. This protocol provides procedures to assess neuronal activity and protein expression of sympathetic preganglionic neurons. For complete details on the use and execution of this protocol, please refer to Ju et al.1.

15.
J Neuroendocrinol ; 35(9): e13328, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37525500

RESUMO

Serotonin is a neurotransmitter that is synthesized and released from the brainstem raphe nuclei to affect many brain functions. It is well known that the activity of raphe serotonergic neurons is changed in response to the changes in feeding status to regulate appetite via the serotonin receptors. Likewise, changes in volume status are known to alter the activity of raphe serotonergic neurons and drugs targeting serotonin receptors were shown to affect sodium appetite. Therefore, the central serotonin system appears to regulate ingestion of both food and salt, although neural mechanisms that induce appetite in response to hunger and sodium appetite in response to volume depletion are largely distinct from each other. In this review, we discuss our current knowledge regarding the regulation of ingestion - appetite and sodium appetite - by the central serotonin system.


Assuntos
Apetite , Sódio , Apetite/fisiologia , Serotonina/fisiologia , Núcleos da Rafe , Tronco Encefálico , Regulação do Apetite/fisiologia
16.
Endocrinol Metab (Seoul) ; 38(5): 545-556, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37749826

RESUMO

BACKGRUOUND: Insulin is a peptide hormone that regulates post-prandial physiology, and it is well known that insulin controls homeostasis at least in part via the central nervous system. In particular, insulin alters the activity of neurons within the autonomic nervous system. However, currently available data are mostly from unidentified brainstem neurons of the dorsal motor nucleus of the vagus nerve (DMV). METHODS: In this study, we used several genetically engineered mouse models to label distinct populations of neurons within the brainstem and the spinal cord for whole-cell patch clamp recordings and to assess several in vivo metabolic functions. RESULTS: We first confirmed that insulin directly inhibited cholinergic (parasympathetic preganglionic) neurons in the DMV. We also found inhibitory effects of insulin on both the excitatory and inhibitory postsynaptic currents recorded in DMV cholinergic neurons. In addition, GABAergic neurons of the DMV and nucleus tractus solitarius were inhibited by insulin. However, insulin had no effects on the cholinergic sympathetic preganglionic neurons of the spinal cord. Finally, we obtained results suggesting that the insulininduced inhibition of parasympathetic preganglionic neurons may not play a critical role in the regulation of glucose homeostasis and gastrointestinal motility. CONCLUSION: Our results demonstrate that insulin inhibits parasympathetic neuronal circuitry in the brainstem, while not affecting sympathetic neuronal activity in the spinal cord.


Assuntos
Insulina , Nervo Vago , Camundongos , Animais , Insulina/farmacologia , Insulina/metabolismo , Nervo Vago/metabolismo , Neurônios/metabolismo , Tronco Encefálico/metabolismo , Colinérgicos/metabolismo , Colinérgicos/farmacologia
17.
Nat Metab ; 5(9): 1506-1525, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37653043

RESUMO

The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.


Assuntos
Astrócitos , Obesidade , Masculino , Animais , Camundongos , Aumento de Peso , Neurônios , Modelos Animais de Doenças , Monoaminoxidase , Ácido gama-Aminobutírico
18.
J Neurosci ; 31(37): 13147-56, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917798

RESUMO

Evidence suggests that the role played by the adipocyte-derived hormone leptin in female reproductive physiology is mediated in part by neurons located within the ventral premammillary nucleus (PMV). Leptin activates PMV neurons; however, the intracellular signaling pathway and channel(s) involved remain undefined. Notably, leptin's excitatory and inhibitory effects within hypothalamic and brainstem nuclei share the intracellular signaling cascade phosphoinositide 3 kinase (PI3K). Therefore, we assessed whether PI3K signaling is required for the acute effect of leptin to alter cellular activity of PMV neurons that express leptin receptors (LepR PMV neurons). Leptin caused a rapid depolarization in the majority of LepR PMV neurons in patch-clamp recordings of hypothalamic slices, while a subset of LepR PMV neurons were hyperpolarized in response to leptin. Data were obtained from both male and female mice and results demonstrate that the acute effect of leptin on LepR PMV neurons was identical for both sexes. Pharmacological inhibition of PI3K prevented the acute leptin-induced change in neuronal activity of LepR PMV neurons, indicating a PI3K-dependent mechanism of leptin action. Similarly, mice with genetically disrupted PI3K signaling in LepR PMV neurons failed to alter cellular activity in response to leptin. Moreover, the leptin-induced depolarization was dependent on a putative TRPC channel. In contrast, the leptin-induced-hyperpolarization required the activation of a putative Katp channel. Collectively, these results suggest that PI3K signaling in LepR PMV neurons is essential for leptin-induced alteration in cellular activity, and these data may suggest a cellular correlate in which leptin contributes to the initiation of reproductive development.


Assuntos
Hipotálamo/fisiologia , Leptina/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Androstadienos/farmacologia , Animais , Compostos de Boro/farmacologia , Cromonas/farmacologia , Feminino , Genes Reporter/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Imidazóis/farmacologia , Canais KATP/fisiologia , Leptina/administração & dosagem , Leptina/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Subunidades Proteicas/biossíntese , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/fisiologia , Wortmanina
19.
Exp Mol Med ; 54(4): 370-376, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35474336

RESUMO

Neurons in the central nervous system (CNS) communicate with peripheral organs largely via the autonomic nervous system (ANS). Through such communications, the sympathetic and parasympathetic efferent divisions of the ANS may affect thermogenesis and blood glucose levels. In contrast, peripheral organs send feedback to the CNS via hormones and autonomic afferent nerves. These humoral and neural feedbacks, as well as neural commands from higher brain centers directly or indirectly shape the metabolic function of autonomic neurons. Notably, recent developments in mouse genetics have enabled more detailed studies of ANS neurons and circuits, which have helped elucidate autonomic control of metabolism. Here, we will summarize the functional organization of the ANS and discuss recent updates on the roles of neural and humoral factors in the regulation of energy balance and glucose homeostasis by the ANS.


Assuntos
Sistema Nervoso Autônomo , Vias Autônomas , Animais , Sistema Nervoso Autônomo/metabolismo , Vias Autônomas/fisiologia , Sistema Nervoso Central , Glucose/metabolismo , Homeostase , Camundongos
20.
Anim Cells Syst (Seoul) ; 26(3): 92-98, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35784392

RESUMO

The lateral parabrachial nucleus (LPBN) has been shown to be involved in the suppression of appetite at the pharmacological, optogenetic and chemogenetic levels. However, the signalling that mediates activation of these neurons in physiological conditions has been hindered by difficulties in segregating different cell populations in this region. Using reporter mice, we identify at the electrophysiological level the effects of an anorexic hormone, leptin, on leptin receptor (ObR)-expressing neurons in the LPBN (LPBNObR neurons). Application of leptin caused inhibition in a subpopulation of LPBNObR neurons. This effect was mediated by an increased potassium conductance and was also accompanied by a decrease in excitatory synaptic input onto these neurons. However, mimicking the inhibitory effects of leptin on LPBNObR neurons through chemogenetics led to no changes in feeding or glucose levels, which suggests that leptin action on LPBNObR neurons may not be sufficient to regulate these metabolic aspects.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA