Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Brain Res ; 1829: 148770, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38266888

RESUMO

The brain and peripheral organs communicate through hormones and neural connections. Proper communication is required to maintain normal whole-body energy homeostasis. In addition to endocrine system, from the perspective of neural connections for metabolic homeostasis, the role of the sympathetic nervous system has been extensively studied, but understanding of the parasympathetic nervous system is limited. The liver plays a central role in glucose and lipid metabolism. This study aimed to clarify the innervation of parasympathetic nervous system in the liver and its functional roles in metabolic homeostasis. The liver-specific parasympathetic nervous system innervation (PNS) was shown by tissue clearing, immunofluorescence and transgenic mice at the three-dimensional histological level. The parasympathetic efferent signals were manipulated using a chemogenetic technique and the activation of ChAT+ parasympathetic neurons in dorsal motor vagus (DMV) results in the increased blood glucose through the elevated hepatic gluconeogenic and lipogenic gene expression in the liver. Thus, our study showed the evidence of ChAT+ parasympathetic neurons in the liver and its role for hepatic parasympathetic nervous signaling in glucose homeostasis through the regulation of hepatic gene expression.


Assuntos
Glicemia , Nervo Vago , Camundongos , Animais , Glicemia/metabolismo , Nervo Vago/fisiologia , Neurônios/metabolismo , Fígado/metabolismo , Glucose/metabolismo , Camundongos Transgênicos , Expressão Gênica
2.
BMB Rep ; 57(3): 149-154, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37817436

RESUMO

The stomach has emerged as a crucial endocrine organ in the regulation of feeding since the discovery of ghrelin. Gut-derived hormones, such as ghrelin and cholecystokinin, can act through the vagus nerve. We previously reported the satiety effect of hypothalamic clusterin, but the impact of peripheral clusterin remains unknown. In this study, we administered clusterin intraperitoneally to mice and observed its ability to suppress fasting-driven food intake. Interestingly, we found its synergism with cholecystokinin and antagonism with ghrelin. These effects were accompanied by increased c-fos immunoreactivity in nucleus tractus solitarius, area postrema, and hypothalamic paraventricular nucleus. Notably, truncal vagotomy abolished this response. The stomach expressed clusterin at high levels among the organs, and gastric clusterin was detected in specific enteroendocrine cells and the submucosal plexus. Gastric clusterin expression decreased after fasting but recovered after 2 hours of refeeding. Furthermore, we confirmed that stomachspecific overexpression of clusterin reduced food intake after overnight fasting. These results suggest that gastric clusterin may function as a gut-derived peptide involved in the regulation of feeding through the gut-brain axis. [BMB Reports 2024; 57(3): 149-154].


Assuntos
Ingestão de Alimentos , Grelina , Camundongos , Animais , Grelina/farmacologia , Ingestão de Alimentos/fisiologia , Clusterina/farmacologia , Colecistocinina/farmacologia , Estômago , Comportamento Alimentar
4.
Nat Commun ; 14(1): 4321, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37468558

RESUMO

Small humanin-like peptide 2 (SHLP2) is a mitochondrial-derived peptide implicated in several biological processes such as aging and oxidative stress. However, its functional role in the regulation of energy homeostasis remains unclear, and its corresponding receptor is not identified. Hereby, we demonstrate that both systemic and intracerebroventricular (ICV) administrations of SHLP2 protected the male mice from high-fat diet (HFD)-induced obesity and improved insulin sensitivity. In addition, the activation of pro-opiomelanocortin (POMC) neurons by SHLP2 in the arcuate nucleus of the hypothalamus (ARC) is involved in the suppression of food intake and the promotion of thermogenesis. Through high-throughput structural complementation screening, we discovered that SHLP2 binds to and activates chemokine receptor 7 (CXCR7). Taken together, our study not only reveals the therapeutic potential of SHLP2 in metabolic disorders but also provides important mechanistic insights into how it exerts its effects on energy homeostasis.


Assuntos
Hipotálamo , Neurônios , Masculino , Animais , Camundongos , Hipotálamo/metabolismo , Neurônios/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Peptídeos/farmacologia , Peptídeos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Homeostase , Mitocôndrias/metabolismo , Pró-Opiomelanocortina/metabolismo , Metabolismo Energético/fisiologia
5.
Life Sci ; 321: 121533, 2023 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-36863487

RESUMO

AIMS: The role of the parasympathetic nervous system (PNS) in the pathogenesis of nonalcoholic steatohepatitis (NASH) is largely unknown. In this study, the effect of PNS modulation on NASH was investigated using chemogenetics. MAIN METHODS: A streptozotocin (STZ) and high-fat diet (HFD)-induced NASH mouse model was used. To activate or inhibit the PNS, chemogenetic human M3-muscarinic receptor coupled with either Gq or Gi protein-containing viruses was injected into the dorsal motor nucleus of the vagus at week 4 and clozapine N-oxide was administered intraperitoneally for a week from week 11. Three groups (PNS-stimulation, PNS-inhibition, and control) were compared in terms of heart rate variability (HRV), histological lipid droplet area, nonalcoholic fatty liver disease activity score (NAS), the area of F4/80-positive macrophages, and biochemical responses. KEY FINDINGS: The STZ/HFD-treated mouse model showed typical histological characteristics of NASH. HRV analysis confirmed that PNS-stimulation and PNS-inhibition groups had significantly higher and lower PNS activity, respectively (both P < 0.05). The PNS-stimulation group showed a significantly smaller hepatic lipid droplet area (14.3 % vs. 20.6 %, P = 0.02) and lower NAS (5.2 vs. 6.3, P = 0.047) than the control group. The area of F4/80-positive macrophages was significantly smaller in the PNS-stimulation group than in the control group (4.1 % vs. 5.6 %, P = 0.04). The PNS-stimulation group showed a lower serum aspartate aminotransferase level than the control group (119.0 vs. 356.0 U/L, P = 0.04). SIGNIFICANCE: In STZ/HFD-treated mice, chemogenetic stimulation of the PNS significantly reduced hepatic fat accumulation and inflammation. The hepatic PNS may play a pivotal role in the pathogenesis of NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Humanos , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/patologia , Fígado/patologia , Inflamação/patologia , Modelos Animais de Doenças , Dieta Hiperlipídica/efeitos adversos , Sistema Nervoso Parassimpático , Lipídeos , Camundongos Endogâmicos C57BL
6.
Biomed Res Int ; 2021: 3501770, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34840970

RESUMO

The hypothalamus plays a central role in the integrated regulation of feeding and energy homeostasis. The hypothalamic arcuate nucleus (ARC) contains a population of neurons that express orexigenic and anorexigenic factors and is thought to control feeding behavior via several neuronal circuits. In this study, a comparative proteomic analysis of low-fat control diet- (LFD-) and high-fat diet- (HFD-) induced hypothalamic ARC was performed to identify differentially expressed proteins (DEPs) related to changes in body weight. In the ARC in the hypothalamus, 6621 proteins (FDR < 0.01) were detected, and 178 proteins were categorized as DEPs (89 upregulated and 89 downregulated in the HFD group). Among the Gene Ontology molecular function terms associated with the DEPs, protein binding was the most significant. Fibroblast growth factor receptor substrate 2 (Frs2) and SHC adaptor protein 3 (Shc3) were related to protein binding and involved in the neurotrophin signaling pathway according to Kyoto Encyclopedia of Genes and Genomes analysis. Furthermore, high-precision quantitative proteomic analysis revealed that the protein profile of the ARC in mice with HFD-induced obesity differed from that in LFD mice, thereby offering insight into the molecular basis of feeding regulation and suggesting Frs2 and Shc3 as novel treatment targets for central anorexigenic signal induction.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Obesidade/metabolismo , Proteoma/metabolismo , Animais , Peso Corporal , Dieta com Restrição de Gorduras , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Regulação para Baixo , Comportamento Alimentar , Ontologia Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/etiologia , Obesidade/genética , Ligação Proteica , Proteoma/genética , Proteômica , Transdução de Sinais , Regulação para Cima
7.
Diabetes ; 70(1): 62-75, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33115827

RESUMO

Circulating branched-chain amino acids (BCAAs) are elevated in obesity and diabetes, and recent studies support a causal role for BCAAs in insulin resistance and defective glycemic control. The physiological mechanisms underlying BCAA regulation are poorly understood. Here we show that insulin signaling in the mediobasal hypothalamus (MBH) of rats is mandatory for lowering plasma BCAAs, most probably by inducing hepatic BCAA catabolism. Insulin receptor deletion only in agouti-related protein (AgRP)-expressing neurons (AgRP neurons) in the MBH impaired hepatic BCAA breakdown and suppression of plasma BCAAs during hyperinsulinemic clamps in mice. In support of this, chemogenetic stimulation of AgRP neurons in the absence of food significantly raised plasma BCAAs and impaired hepatic BCAA degradation. A prolonged fasting or ghrelin treatment recapitulated designer receptors exclusively activated by designer drugs-induced activation of AgRP neurons and increased plasma BCAAs. Acute stimulation of vagal motor neurons in the dorsal motor nucleus was sufficient to decrease plasma BCAAs. Notably, elevated plasma BCAAs were associated with impaired glucose homeostasis. These findings suggest a critical role of insulin signaling in AgRP neurons for BCAA regulation and raise the possibility that this control may be mediated primarily via vagal outflow. Furthermore, our results provide an opportunity to closely examine the potential mechanistic link between central nervous system-driven BCAA control and glucose homeostasis.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Neurônios/metabolismo , Aminoácidos de Cadeia Ramificada/sangue , Animais , Glicemia/metabolismo , Grelina/farmacologia , Técnica Clamp de Glucose , Hipotálamo/efeitos dos fármacos , Resistência à Insulina/fisiologia , Masculino , Camundongos , Neurônios Motores/metabolismo , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Nervo Vago/metabolismo
8.
Heliyon ; 6(10): e05190, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33088957

RESUMO

Glucagon-like peptide-1 (GLP-1), whose agonists are widely prescribed, is a peptide proven effective in reducing obesity. Similarly, oxytocin (OXT) is a peptide known to increase satiety and help reduce body weight. In the present study, we aimed to examine the metabolic effects of co-administration of GLP-1 and OXT in diet-induced obesity (DIO) mice to elucidate their functions and interactions in the central nervous system. To this end, 40 DIO mice were subjected to stereotaxic surgery for the installation of an osmotic minipump and intracerebroventricular administration of GLP-1, OXT, or both. Initially, it was anticipated that co-administration of these anorexigenic peptides would be as effective as, if not more than, either GLP-1 or OXT alone in providing metabolic benefits to the obese mice. Interestingly, co-administration of OXT and GLP-1 offset the reductions in body weight and food intake promoted by either peptide alone. Co-administration also negated the decrease in fat and increase in lean mass produced by either peptide alone. Moreover, co-administration showed an equivalent calorimetric benefit as either peptide alone. Therefore, these results suggest antagonistic, rather than synergistic or additive, effects of centrally administered GLP-1 and OXT that attenuate the metabolic benefits of either peptide.

9.
Neurosci Lett ; 712: 134356, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31470043

RESUMO

Parasympathetic nervous system (PNS) innervates with several peripheral organs such as liver, pancreas and regulates energy metabolism. However, the direct role of PNS on food intake has been poorly understood. In the present study, we investigated the role of parasympathetic nervous system in regulation of feeding by chemogenetic methods. Adeno associated virus carrying DREADD (designer receptors exclusively activated by designer drugs) infused into the target brain region by stereotaxic surgery. The stimulatory hM3Dq or inhibitory hM4Di DREADD was over-expressed in selective population of dorsal motor nucleus of the vagus (DMV) neurons by Cre-recombinase-dependent manners. Activation of parasympathetic neuron by intraperitoneal injection of the M3-muscarinic receptor ligand clozapine-N-oxide (CNO) (1 mg/kg) suppressed food intake and resulted in body weight loss in ChAT-Cre mice. Parasympathetic neurons activation resulted in improved glucose tolerance while inhibition of the neurons resulted in impaired glucose tolerance. Stimulation of parasympathetic nervous system by injection of CNO (1 mg/kg) increased oxygen consumption and energy expenditure. Within the hypothalamus, in the arcuate nucleus (ARC) changed AGRP/POMC neurons. These results suggest that direct activation of parasympathetic nervous system decreases food intake and body weight with improved glucose tolerance.


Assuntos
Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/fisiologia , Neurônios/fisiologia , Sistema Nervoso Parassimpático/fisiologia , Nervo Vago/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Clozapina/análogos & derivados , Clozapina/farmacologia , Dependovirus , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Sistema Nervoso Parassimpático/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
10.
Biochem Biophys Res Commun ; 490(2): 247-252, 2017 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-28610922

RESUMO

Glucagon-like peptide-1 amide (GLP-1) and gastric inhibitory polypeptide (GIP) are incretin hormones regulating energy metabolism. GLP-1 and GIP combination is suggested as a promising therapeutic strategy for treatment of obesity and diabetes. However, the neuronal mechanisms are not yet investigated. In the present study, we investigated the role of central GLP-1 and GIP in regulation of body weight homeostasis. The effect of GLP-1 with GIP on food intake, body weight, locomotor activity were determined following intracerebroventricular (ICV) administration of GLP-1 and/or GIP in mice. ICV administration of low dose GLP-1 (0.3 nmol) and GIP (1 and 3 nmol) did not change food intake. However, ICV administration of higher doses GLP-1 (1 and 3 nmol) and GIP (6 nmol) significantly decreased food intake and body weight. To investigate the synergic effect of ICV GLP-1 and GIP, subeffective dose GLP-1 (0.3 nmol) and subeffective dose GIP (1 nmol) were chosen for further co-administration study. ICV co-administration of GLP-1 and GIP significantly decreased food intake, body weight and drinking. ICV co-administration of GLP-1 and GIP significantly increased neuronal activation and pro-opiomelanocortin (POMC) expression in hypothalamic arcuate nucleus. The neuronal activation and POMC expression were observed in two distinct neuronal populations. These results provide neuronal mechanisms supporting the development of GLP-1 and GIP combination therapeutics for treatment of obesity and diabetes.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Polipeptídeo Inibidor Gástrico/administração & dosagem , Polipeptídeo Inibidor Gástrico/farmacologia , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Animais , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Biochem Biophys Res Commun ; 485(2): 409-413, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213131

RESUMO

Neuroendocrine regulatory peptide (NERP)-2 is derived from a distinct region of VGF, a neurosecretory protein originally identified as a product of a nerve growth factor-responsive gene in rat PC12 cells. Colocalization of NERP-2 with orexin-A in the lateral hypothalamus increases orexin-A-induced feeding and energy expenditure in both rats and mice. Orexigenic and anorectic peptides in the hypothalamus modulate gastric function. In this study, we investigated the effect of NERP-2 on gastric function in rats. Intracerebroventricular administration of NERP-2 to rats increased gastric acid secretion and gastric emptying, whereas peripheral administration did not affect gastric function. NERP-2-induced gastric acid secretion and gastric emptying were blocked by an orexin 1 receptor antagonist, SB334867. NERP-2 also induced Fos expression in the lateral hypothalamus and the dorsomotor nucleus of the vagus X, which are key sites in the central nervous system for regulation of gastric function. Atropine, a blocker of vagal efferent signal transduction, completely blocked NERP-2-induced gastric acid secretion. These results demonstrate that central administration of NERP-2 activates the orexin pathway, resulting in elevated gastric acid secretion and gastric emptying.


Assuntos
Ácido Gástrico/metabolismo , Esvaziamento Gástrico/efeitos dos fármacos , Proteínas do Tecido Nervoso/farmacologia , Receptores de Orexina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Atropina/farmacologia , Benzoxazóis/farmacologia , Injeções Intraventriculares , Masculino , Naftiridinas , Proteínas do Tecido Nervoso/administração & dosagem , Parassimpatolíticos/farmacologia , Ratos Sprague-Dawley , Ratos Transgênicos , Ratos Wistar , Ureia/análogos & derivados , Ureia/farmacologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/fisiologia
12.
Biochem Biophys Res Commun ; 464(4): 1157-1162, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26208455

RESUMO

A high-fat diet (HFD) induces inflammation in systemic organs including the hypothalamus, resulting in obesity and diabetes. The vagus nerve connects the visceral organs and central nervous system, and the gastric-derived orexigenic peptide ghrelin transmits its starvation signals to the hypothalamus via the vagal afferent nerve. Here we investigated the inflammatory response in vagal afferent neurons and the hypothalamus in mice following one day of HFD feeding. This treatment increased the number of macrophages/microglia in the nodose ganglion and hypothalamus. Furthermore, one-day HFD induced expression of Toll-like receptor 4 in the goblet cells of the colon and upregulated mRNA expressions of the proinflammatory biomarkers Emr1, Iba1, Il6, and Tnfα in the nodose ganglion and hypothalamus. Both subcutaneous administration of ghrelin and celiac vagotomy reduced HFD-induced inflammation in these tissues. HFD intake triggered inflammatory responses in the gut, nodose ganglion, and subsequently in the hypothalamus within 24 h. These findings suggest that the vagal afferent nerve may transfer gut-derived inflammatory signals to the hypothalamus via the nodose ganglion, and that ghrelin may protect against HFD-induced inflammation.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Encefalite/imunologia , Grelina/imunologia , Hipotálamo/imunologia , Gânglio Nodoso/imunologia , Doenças do Nervo Vago/imunologia , Animais , Encefalite/etiologia , Encefalite/patologia , Hipotálamo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gânglio Nodoso/patologia , Doenças do Nervo Vago/etiologia , Doenças do Nervo Vago/patologia
13.
J Endocrinol ; 226(1): 81-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26016745

RESUMO

Ghrelin, a stomach-derived orexigenic peptide, transmits starvation signals to the hypothalamus via the vagus afferent nerve. Peripheral administration of ghrelin does not induce food intake in high fat diet (HFD)-induced obese mice. We investigated whether this ghrelin resistance was caused by dysfunction of the vagus afferent pathway. Administration (s.c.) of ghrelin did not induce food intake, suppression of oxygen consumption, electrical activity of the vagal afferent nerve, phosphorylation of ERK2 and AMP-activated protein kinase alpha in the nodose ganglion, or Fos expression in hypothalamic arcuate nucleus of mice fed a HFD for 12 weeks. Administration of anti-ghrelin IgG did not induce suppression of food intake in HFD-fed mice. Expression levels of ghrelin receptor mRNA in the nodose ganglion and hypothalamus of HFD-fed mice were reduced. Inflammatory responses, including upregulation of macrophage/microglia markers and inflammatory cytokines, occurred in the nodose ganglion and hypothalamus of HFD-fed mice. A HFD blunted ghrelin signaling in the nodose ganglion via a mechanism involving in situ activation of inflammation. These results indicate that ghrelin resistance in the obese state may be caused by dysregulation of ghrelin signaling via the vagal afferent.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Grelina/fisiologia , Obesidade/etiologia , Obesidade/fisiopatologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Grelina/administração & dosagem , Grelina/sangue , Hipotálamo/fisiopatologia , Inflamação/etiologia , Inflamação/genética , Inflamação/fisiopatologia , Leptina/administração & dosagem , Leptina/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gânglio Nodoso/fisiopatologia , Obesidade/genética , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Grelina/genética , Transdução de Sinais
14.
J Biol Chem ; 285(13): 9706-9715, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20097762

RESUMO

Anorexia and weight loss are prevalent in infectious diseases. To investigate the molecular mechanisms underlying these phenomena, we established animal models of infection-associated anorexia by administrating bacterial and viral products, lipopolysaccharide (LPS) and human immunodeficiency virus-1 transactivator protein (Tat). In these models, we found that the nuclear factor-kappaB (NF-kappaB), a pivotal transcription factor for inflammation-related proteins, was activated in the hypothalamus. In parallel, administration of LPS and Tat increased hypothalamic pro-inflammatory cytokine production, which was abrogated by inhibition of hypothalamic NF-kappaB. In vitro, NF-kappaB activation directly stimulated the transcriptional activity of pro-opiomelanocortin (POMC), a precursor of anorexigenic melanocortin, and mediated the stimulatory effects of LPS, Tat, and pro-inflammatory cytokines on POMC transcription, implying the involvement of NF-kappaB in controlling feeding behavior. Consistently, hypothalamic injection of LPS and Tat caused a significant reduction in food intake and body weight, which was prevented by blockade of NF-kappaB and melanocortin. Furthermore, disruption of I kappaB kinase-beta, an upstream kinase of NF-kappaB, in POMC neurons attenuated LPS- and Tat-induced anorexia. These findings suggest that infection-associated anorexia and weight loss are mediated via NF-kappaB activation in hypothalamic POMC neurons. In addition, hypothalamic NF-kappaB was activated by leptin, an important anorexigenic hormone, and mediates leptin-stimulated POMC transcription, indicating that hypothalamic NF-kappaB also serves as a downstream signaling pathway of leptin.


Assuntos
Anorexia/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Lipopolissacarídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Modelos Biológicos
15.
Nat Neurosci ; 9(7): 901-6, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16783365

RESUMO

Insulin signaling in the hypothalamus plays a role in maintaining body weight. Studies suggest that the forkhead transcription factor Foxo1 is an important mediator of insulin signaling in peripheral tissues. Here we demonstrate that in normal mice, hypothalamic Foxo1 expression is reduced by the anorexigenic hormones insulin and leptin. These hormones' effects on feeding are inhibited when hypothalamic Foxo1 is activated, establishing a new signaling pathway through which insulin and leptin regulate food intake in hypothalamic neurons. Moreover, activation of Foxo1 in the hypothalamus increases food intake and body weight, whereas inhibition of Foxo1 decreases both. Foxo1 stimulates the transcription of the orexigenic neuropeptide Y and Agouti-related protein through the phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway, but suppresses the transcription of anorexigenic proopiomelanocortin by antagonizing the activity of signal transducer-activated transcript-3 (STAT3). Our data suggest that hypothalamic Foxo1 is an important regulator of food intake and energy balance.


Assuntos
Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Análise de Variância , Animais , Western Blotting/métodos , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina/métodos , Ingestão de Alimentos/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Metabolismo Energético/efeitos dos fármacos , Proteína Forkhead Box O1 , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Hipotálamo/efeitos dos fármacos , Imuno-Histoquímica/métodos , Insulina/farmacologia , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma , RNA Interferente Pequeno/farmacologia
16.
Nat Med ; 10(7): 727-33, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15195087

RESUMO

AMP-activated protein kinase (AMPK) functions as a fuel sensor in the cell and is activated when cellular energy is depleted. Here we report that alpha-lipoic acid (alpha-LA), a cofactor of mitochondrial enzymes, decreases hypothalamic AMPK activity and causes profound weight loss in rodents by reducing food intake and enhancing energy expenditure. Activation of hypothalamic AMPK reverses the effects of alpha-LA on food intake and energy expenditure. Intracerebroventricular (i.c.v.) administration of glucose decreases hypothalamic AMPK activity, whereas inhibition of intracellular glucose utilization through the administration of 2-deoxyglucose increases hypothalamic AMPK activity and food intake. The 2-deoxyglucose-induced hyperphagia is reversed by inhibiting hypothalamic AMPK. Our findings indicate that hypothalamic AMPK is important in the central regulation of food intake and energy expenditure and that alpha-LA exerts anti-obesity effects by suppressing hypothalamic AMPK activity.


Assuntos
Fármacos Antiobesidade/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Hipotálamo/efeitos dos fármacos , Ácido Tióctico/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Hipotálamo/enzimologia , Hipotálamo/fisiologia , Leptina/fisiologia , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA