Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nutr Neurosci ; 27(2): 106-119, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36634108

RESUMO

The hypothalamus controls food intake by integrating nutrient signals, of which one of the most important is glucose. Consequently, impairments in hypothalamic glucose-sensing mechanisms are associated with hyperphagia and obesity. Environmental enrichment (EE) is an animal housing protocol that provides complex sensory, motor, and social stimulations and has been proven to reduce adiposity in laboratory mice. However, the mechanism by which EE promotes adiposity-suppressing effect remains incompletely understood. Neurotrophic factors play an important role in the development and maintenance of the nervous system, but they are also involved in the hypothalamic regulation of feeding. Brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) are expressed in the hypothalamus and their expression is stimulated by glucose. EE is associated with increased expression of Bdnf mRNA in the hypothalamus. Therefore, we hypothesized that EE potentiates the anorectic action of glucose by altering the expression of neurotrophic factor genes in the hypothalamus. Male C57BL/6 mice were maintained under standard or EE conditions to investigate the feeding response to glucose and the associated expression of feeding-related neurotrophic factor genes in the hypothalamus. Intraperitoneal glucose injection reduced food intake in both control and EE mice with a significantly greater reduction in the EE group compared to the control group. EE caused a significantly enhanced response of Gdnf mRNA expression to glucose without altering basal Gdnf mRNA expression and Bdnf mRNA response to glucose. These findings suggest that EE enhances glucose-induced feeding suppression, at least partly, by enhancing hypothalamic glucose-sensing ability that involves GDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Glucose , Animais , Masculino , Camundongos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Glucose/metabolismo , Hipotálamo/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Int J Mol Sci ; 23(13)2022 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-35806073

RESUMO

Feeding-regulating neurotrophic factors are expressed in both neurons and glial cells. However, nutritional regulation of anorexigenic glial cell line-derived neurotrophic factor (GDNF) and orexigenic mesencephalic astrocyte-derived neurotrophic factor (MANF) expression in specific cell types remains poorly understood. Hypothalamic glucose sensing plays a critical role in the regulation of food intake. It has been theorized that local glucose concentration modulates microglial activity partially via glucose transporter 5 (GLUT5). We hypothesized that an increased local glucose concentration stimulates GDNF expression while inhibiting MANF expression in the hypothalamus and microglia via GLUT5. The present study investigated the effect of glucose on Gdnf and Manf mRNA expression in the mouse hypothalamus and murine microglial cell line SIM-A9. Intracerebroventricular glucose treatment significantly increased Gdnf mRNA levels in the hypothalamus without altering Manf mRNA levels. Exposure to high glucose caused a significant increase in Gdnf mRNA expression and a time-dependent change in Manf mRNA expression in SIM-A9 cells. GLUT5 inhibitor treatment did not block glucose-induced Gdnf mRNA expression in these cells. These findings suggest that microglia are responsive to changes in the local glucose concentration and increased local glucose availability stimulates the expression of microglial GNDF through a GLUT5-independent mechanism, contributing to glucose-induced feeding suppression.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial , Microglia , Animais , Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Transportador de Glucose Tipo 5/metabolismo , Camundongos , Microglia/metabolismo , Fatores de Crescimento Neural/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Int J Mol Sci ; 22(23)2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34884473

RESUMO

Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body's metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) is a fructose transporter primarily expressed in microglia within the central nervous system. However, little is known about the nutritional regulation of Slc2a5 expression in microglia and its role in the regulation of metabolism. The present study aimed to address the hypothesis that nutrients affect microglial activity by altering the expression of glucose transporter genes. Murine microglial cell line SIM-A9 cells and primary microglia from mouse brain were exposed to different concentrations of glucose and levels of microglial activation markers and glucose transporter genes were measured. High concentration of glucose increased levels of the immediate-early gene product c-Fos, a marker of cell activation, Slc2a5 mRNA, and pro-inflammatory cytokine genes in microglial cells in a time-dependent manner, while fructose failed to cause these changes. Glucose-induced changes in pro-inflammatory gene expression were partially attenuated in SIM-A9 cells treated with the GLUT5 inhibitor. These findings suggest that an increase in local glucose availability leads to the activation of microglia by controlling their carbohydrate sensing mechanism through both GLUT5-dependent and -independent mechanisms.


Assuntos
Transportador de Glucose Tipo 5/genética , Glucose/farmacologia , Microglia/citologia , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética
4.
Biochem Biophys Res Commun ; 498(4): 842-848, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29530527

RESUMO

Xenin is a gastrointestinal hormone that belongs to the neurotensin family. Central administration of xenin to obese mice reduces food intake and body weight gain and causes alterations in the expression of lipid metabolism-related genes and proteins in white adipose tissue (WAT). However, it has not been tested whether or not xenin directly acts on adipose tissue and alters lipid metabolism. The present study was performed to address this possibility by examining the effect of xenin treatment on the levels of glycerol and free fatty acids (FFA) and expression levels of lipolysis marker proteins ex vivo in cultured mouse WAT. Xenin treatment significantly increased concentrations of glycerol and FFA in culture media and increased phosphorylation of hormone sensitive lipase (HSL) in ex vivo cultured WAT. These findings support the hypothesis that xenin directly acts on adipose tissues and stimulates lipolysis. Thus, enhancement of xenin action and its downstream signaling may offer a novel and effective therapy for obese patients by reducing the amount of stored fat in adipose tissue.


Assuntos
Tecido Adiposo Branco/metabolismo , Neurotensina/farmacologia , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/efeitos dos fármacos , Animais , Células Cultivadas , Meios de Cultura/análise , Ácidos Graxos não Esterificados/metabolismo , Glicerol/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Esterol Esterase/metabolismo
5.
Neuroreport ; 33(13): 583-589, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-36049163

RESUMO

Brain-derived neurotrophic factor (BDNF) is expressed in both hypothalamic neurons and microglia, and plays a critical role in the regulation of metabolism. Although hypothalamic expression of BDNF is regulated by metabolic signals such as nutrients and hormones, it remains unknown whether these signals differentially regulate BDNF expression in different cell types. The present study aimed to determine whether glucose and fructose regulate BDNF expression in microglia via the specific glucose transporter. To determine the effect of glucose and fructose on Bdnf mRNA and protein expression, murine microglial cell line SIM-A9 cells were exposed to the maintenance concentration of glucose (17.5 mmol/l), high glucose (25 mmol/l), or fructose (7.5 mmol/l) for 40 min to 24 h. To determine whether the blockade of glucose transporter 5 (GLUT5) negates the effect of glucose on Bdnf mRNA expression, cells were exposed to 25 mmol/l glucose in the presence or absence of the GLUT5 inhibitor for 4 h. Levels of Bdnf mRNA and protein were measured by real-time PCR and ELISA, respectively. High glucose caused a significant increase in both pan-Bdnf and long-form Bdnf (L-Bdnf) mRNA as well as protein levels when compared with the maintenance of concentration of glucose in a time-dependent manner. Fructose treatment also increased L-Bdnf mRNA expression. Pharmacological blockade of GLUT5 did not affect glucose-induced Bdnf mRNA expression. These findings suggest that glucose and fructose directly stimulate Bdnf mRNA expression in microglia and these responses may mediate the metabolic actions of glucose and fructose.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Frutose , Glucose , Microglia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Frutose/metabolismo , Frutose/farmacologia , Expressão Gênica , Glucose/metabolismo , Glucose/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/farmacologia , Camundongos , Microglia/metabolismo , RNA Mensageiro/metabolismo
6.
Behav Brain Res ; 433: 113986, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35798208

RESUMO

Social and environmental factors influence behavior via modulation of brain physiological functions. Environmental enrichment (EE) is an animal housing technique that provides complex sensory, motor, and social stimulation, leading to modifications in the innate aggressiveness in group-housed laboratory mice. Brain-derived neurotrophic factor (BDNF) is encoded by multiple splice variants and plays a critical role in controlling aggressive behavior in a transcript variant-specific manner. BDNF mediates the beneficial effects of EE on a variety of pathophysiological conditions. These findings led to the hypothesis that EE reduces aggressive behavior by altering the expression of Bdnf mRNA in a transcript variant-specific manner. To test this hypothesis, 3-4-week-old male C57BL/6 mice were randomly group-housed (5 mice per cage) under standard or EE conditions for 6-8 weeks. Aggressive behavior was monitored and levels of Bdnf mRNA variants in aggression-related brain regions were measured. Mice housed in EE cages displayed a significantly lower frequency of aggressive interactions compared to control mice. EE increased levels of Bdnf mRNA variant I (Bdnf I) in the amygdala while it reduced levels of Bdnf I in the hypothalamus, hippocampus, prefrontal cortex, parietal cortex, and brainstem. Meanwhile, EE did not significantly alter levels of Bdnf mRNA variants IIc, IV, and VI in all brain regions examined. These findings support the hypothesis that EE diminishes inter-male aggression by altering Bdnf mRNA expression in a transcript variant-specific and brain region-specific manner. Specifically, brain region-specific alterations in Bdnf I expression may partly mediate EE-induced suppression of inter-male aggression.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Meio Ambiente , Agressão/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo
7.
Biochem Biophys Res Commun ; 400(4): 713-7, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20816934

RESUMO

Common variants in the fat mass and obesity associated (FTO) gene are associated with obesity and type 2 diabetes. Fto-deficient mice develop hepatic insulin resistance, leading to the hypothesis that hepatic Fto plays a role in the regulation of glucose metabolism and that hepatic Fto expression is regulated by metabolic states. We found that hepatic Fto mRNA levels were increased by fasting in mice. Intraperitoneal glucose injection reduced hepatic Fto mRNA levels without significant changes in body weight in fasted mice. The inverse correlation between Fto mRNA and glucose remained significant after adjusting for body weight. There were positive correlations between hepatic Fto mRNA expression and gluconeogenic gene expression. These data support the hypothesis that hepatic Fto expression changes in response to metabolic states and glucose reduces hepatic Fto mRNA expression independently of body weight. Hepatic Fto may participate in the feedback regulation of glucose metabolism via gluconeogenesis.


Assuntos
Glicemia/genética , Gluconeogênese/genética , Fígado/metabolismo , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Peso Corporal/genética , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
J Neurochem ; 109(2): 562-72, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19222701

RESUMO

Transgenic mice that express human equilibrative nucleoside transporter subtype 1 (hENT1) under the control of a neuron-specific enolase promoter have been generated. Southern blot and PCR revealed the presence of the transgene in five founder mice. Mice from each founder line were examined by reverse transcriptase (RT)-PCR and found to express hENT1 in RNA isolated from whole brain, cerebral cortex, striatum, hippocampus, and cerebellum but not liver, kidney, heart, lung or skeletal muscle. Cortical synaptosomes prepared from transgenic mice had significantly increased [(3)H]adenosine uptake and [(3)H]nitrobenzylthioinosine binding, relative to samples from wild-type mice. In behavioral tests, transgenic mice had altered responses to caffeine and ethanol, two drugs that inhibit and enhance, respectively, adenosine receptor activity. Caffeine-induced locomotor stimulation was attenuated whereas the hypnotic effect of ethanol was enhanced in transgenic mice. Caffeine was more potent in inhibiting ethanol-induced motor incoordination in wild-type than in transgenic mice. No differences in expression of mouse genes for adenosine receptors, nucleoside transporters, or purine metabolizing enzymes were detected by RT-PCR analyses. These data indicate that expression of hENT1 in neurons does not trigger adaptive changes in expression of adenosine-related genes. Instead, hENT1 expression affects dynamic changes in endogenous adenosine levels, as revealed by altered behavioral responses to drugs that affect adenosine receptor signalling.


Assuntos
Transportador Equilibrativo 1 de Nucleosídeo/biossíntese , Transportador Equilibrativo 1 de Nucleosídeo/genética , Regulação da Expressão Gênica/fisiologia , Neurônios/metabolismo , Adenosina/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Cafeína/administração & dosagem , Cafeína/antagonistas & inibidores , Transportador Equilibrativo 1 de Nucleosídeo/fisiologia , Etanol/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Células PC12 , Ratos , Receptores Purinérgicos P1/biossíntese , Receptores Purinérgicos P1/genética , Regulação para Cima/genética
9.
Can J Physiol Pharmacol ; 87(10): 839-49, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20052010

RESUMO

Space travelers experience anorexia and body weight loss in a microgravity environment, and microgravity-like situations cause changes in hypothalamic activity. Hypothalamic melanocortins play a critical role in the regulation of metabolism. Therefore, we hypothesized that microgravity affects metabolism through alterations in specific hypothalamic signaling pathways, including melanocortin signaling. To address this hypothesis, the microgravity-like situation was produced by an antiorthostatic tail suspension in wild-type and agouti mice, and the effect of tail suspension on energy expenditure and hypothalamic gene expression was examined. Energy expenditure was measured using indirect calorimetry before and during the tail suspension protocol. Hypothalamic tissues were collected for gene expression analysis at the end of the 3 h tail suspension period. Tail suspension significantly increased oxygen consumption, carbon dioxide production, and heat production in wild-type mice. Tail suspension-induced increases in energy expenditure were not attenuated in agouti mice. Although tail suspension did not alter hypothalamic proopiomelanocortin (POMC) and agouti-related protein (AGRP) mRNA levels, it significantly increased hypothalamic interleukin 6 (Il-6) mRNA levels. These data are consistent with the hypothesis that microgravity increases energy expenditure and suggest that these effects are mediated through hypothalamic signaling pathways that are independent of melanocortins, but possibly used by Il-6.


Assuntos
Metabolismo Energético/fisiologia , Elevação dos Membros Posteriores/fisiologia , Melanocortinas/biossíntese , Tecido Adiposo Marrom/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Análise Química do Sangue , Western Blotting , Expressão Gênica/fisiologia , Hormônios/sangue , Hipotálamo/fisiologia , Interleucina-6/biossíntese , Cinética , Masculino , Melanocortinas/genética , Metabolismo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Pró-Opiomelanocortina/metabolismo , RNA/biossíntese , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Ausência de Peso
10.
Biochem Biophys Res Commun ; 376(2): 384-8, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18789887

RESUMO

The central melanocortin system regulates hepatic lipid metabolism. Hepatic lipogenic gene expression is regulated by transcription factors including sterol regulatory element-binding protein 1c (SREBP-1c), carbohydrate responsive element-binding protein (ChREBP), and peroxisome proliferator-activated receptor gamma2 (PPARgamma2). However, it is unclear if central melanocortin signaling regulates hepatic lipogenic gene expression through the activation of these transcription factors. To delineate the molecular mechanisms by which the melanocortin system regulates hepatic lipid metabolism, we examined the effect of intracerebroventricular injection of SHU9119, a melanocortin receptor antagonist, on hepatic expression levels of genes involved in lipid metabolism in mice. SHU9119 treatment increased hepatic triglyceride content and mRNA levels of lipogenic genes, SREBP-1c, and PPARgamma2, whereas it did not cause any changes in hepatic ChREBP mRNA levels. These findings suggest that reduced central melanocortin signaling increases hepatic lipid deposition by stimulating hepatic lipogenic gene expression at least partly through the activation of SREBP-1c and PPARgamma2.


Assuntos
Regulação da Expressão Gênica , Lipogênese/genética , Fígado/metabolismo , Melanocortinas/metabolismo , PPAR gama/genética , Tecido Adiposo Branco/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores de Melanocortina/antagonistas & inibidores , Receptores de Melanocortina/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Triglicerídeos
11.
Behav Brain Res ; 194(1): 66-71, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18639588

RESUMO

Neurotensin plays a role in regulating feeding behavior. Central injection of neurotensin reduces food intake and the anorectic effect of neurotensin is mediated through neurotensin receptor 1 (Ntsr1). Ntsr1-deficient mice are characterized by mild hyperphagia and overweight without hyperleptinemia. The mechanism by which Ntsr1-deficient mice develop these metabolic abnormalities is not well understood. Leptin, secreted by adipocytes, regulates food intake by acting on hypothalamic neurons including neurotensin-producing neurons. Since the anorectic effect of leptin is blocked by neurotensin receptor antagonist, we hypothesized that the anorectic effect of leptin is mediated through Ntsr1 in the central nervous system and that decreased sensitivity to the anorectic effect of leptin contributes to metabolic perturbations in Ntsr1-deficient mice. To address this hypothesis, we examined the effect of intracerebroventricular (i.c.v.) administration of leptin on food intake in Ntsr1-deficient mice. A single i.c.v. injection of leptin caused robust reductions in food intake in wild-type mice. These effects were markedly attenuated in Ntsr1-deficient mice. These data are consistent with our hypothesis that the anorectic effect of leptin is at least partly mediated through central Ntsr1 and that the leptin-Ntsr1 signaling pathway is involved in the regulation of food intake. Our data also suggest that the lack of Ntsr1 reduces sensitivity to the anorectic action of leptin, causing hyperphagia and abnormal weight gain.


Assuntos
Depressores do Apetite/uso terapêutico , Hiperfagia/tratamento farmacológico , Hiperfagia/genética , Leptina/uso terapêutico , Receptores de Neurotensina/deficiência , Análise de Variância , Animais , Comportamento Animal , Glicemia/efeitos dos fármacos , Glicemia/genética , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/genética , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Calorimetria , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Neurotensina/genética , Neurotensina/metabolismo , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo
12.
Nutrients ; 10(11)2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388740

RESUMO

Common genetic variants of the fat mass and obesity associated (FTO) gene are strongly associated with obesity and type 2 diabetes. FTO is ubiquitously expressed. Earlier studies have focused on the role of hypothalamic FTO in the regulation of metabolism. However, recent studies suggest that expression of hepatic FTO is regulated by metabolic signals, such as nutrients and hormones, and altered FTO levels in the liver affect glucose and lipid metabolism. This review outlines recent findings on hepatic FTO in the regulation of metabolism, with particular focus on hepatic glucose and lipid metabolism. It is proposed that abnormal activity of hepatic signaling pathways involving FTO links metabolic impairments such as obesity, type 2 diabetes and nonalcoholic fatty liver disease (NAFLD). Therefore, a better understanding of these pathways may lead to therapeutic approaches to treat these metabolic diseases by targeting hepatic FTO. The overall goal of this review is to place FTO within the context of hepatic regulation of metabolism.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Glucose/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Variação Genética , Humanos
13.
Free Radic Biol Med ; 115: 458-470, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277394

RESUMO

Debilitating muscle-disuse atrophy in aging or obesity has huge socioeconomic impact. Since nitric oxide (NO) mediates muscle satellite cell activation and induces hypertrophy with exercise in old mice, we tested whether treatment with the NO donor, isosorbide dinitrate (ISDN), during hind limb suspension would reduce atrophy. Mice were suspended 18 days, with or without daily ISDN (66mg/kg). Muscles were examined for atrophy (weight, fiber diameter); regulatory changes in atrogin-1 (a negative regulator of muscle mass), myostatin (inhibits myogenesis), and satellite cell proliferation; and metabolic responses in myosin heavy chains (MyHCs), liver lipid, and hypothalamic gene expression. Suspension decreased muscle weight and weight relative to body weight between 25-55%, and gastrocnemius fiber diameter vs. CONTROLS: In young-adult mice, ISDN attenuated atrophy by half or more. In quadriceps, ISDN completely prevented the suspension-induced rise in atrogin-1 and drop in myostatin precursor, and attenuated the changes in MyHCs 1 and 2b observed in unloaded muscles without treatment. Fatty liver in suspended young-adult mice was also reduced by ISDN; suspended young mice had higher hypothalamic expression of the orexigenic agouti-related protein, Agrp than controls. Notably, a suspension-induced drop in muscle satellite cell proliferation by 25-58% was completely prevented (young mice) or attenuated (halved, in young-adult mice) by ISDN. NO-donor treatment has potential to attenuate atrophy and metabolic changes, and prevent regulatory changes during disuse and offset/prevent wasting in age-related sarcopenia or space travel. Increases in precursor proliferation resulting from NO treatment would also amplify benefits of physical therapy and exercise.


Assuntos
Envelhecimento/fisiologia , Membro Posterior/patologia , Dinitrato de Isossorbida/uso terapêutico , Transtornos Musculares Atróficos/terapia , Óxido Nítrico/metabolismo , Sarcopenia/terapia , Células Satélites de Músculo Esquelético/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Membro Posterior/efeitos dos fármacos , Elevação dos Membros Posteriores , Humanos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miostatina/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo
14.
Endocrinology ; 148(4): 1928-32, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17218412

RESUMO

Because appetite, hypothalamic gene expression, reproductive function, and adrenal function are highly sensitive to acute changes in plasma glucose levels, it has been hypothesized hypothalamic neurons sensitive to glucose play a role in regulating these functions. To assess this hypothesis, we examined these neuronendocrine functions in mice in which the glucokinase gene, which plays an essential role in neuroendocrine glucose sensing, has been ablated. Haploinsufficiency in heterozygous glucokinase knockout mice produced effects similar to those produced by hypoglycemia: impaired reproductive function, elevated plasma corticosterone, increased food intake, and hypothalamic gene expression similar to that observed in fasted or leptin-deficient obese mice (increased hypothalamic neuropeptide Y mRNA and reduced hypothalamic proopiomelanocortin mRNA). Plasma glucose was elevated 2-fold in glucokinase knockout mice, consistent with a maturity-onset diabetes of the young phenotype, but plasma insulin and leptin levels were normal. These data support the hypothesis that glucokinase plays a key role in the neuroendocrine regulation of metabolic economy.


Assuntos
Ingestão de Alimentos/genética , Glucocorticoides/metabolismo , Glucoquinase/fisiologia , Hipotálamo/metabolismo , Reprodução/genética , Animais , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Glucoquinase/genética , Hiperglicemia/genética , Insulina/sangue , Insulina/genética , Leptina/sangue , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas
15.
Neuropeptides ; 63: 67-73, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28190525

RESUMO

Xenin is a gastrointestinal hormone that reduces food intake when administered centrally and it has been hypothesized that central action of xenin participates in the regulation of whole-body metabolism. The present study was performed to address this hypothesis by investigating the central effect of xenin on the expression of genes and proteins that are involved in the regulation of lipid metabolism in white adipose tissue (WAT). Male obese ob/ob mice received intracerebroventricular (i.c.v.) injections of xenin (5µg) twice 12h apart. Food intake and body weight change during a 24-h period after the first injection were measured. Epididymal WAT was collected at the end of the 24-h treatment period and levels of lipid metabolism-related genes and proteins were measured. Xenin treatment caused significant reductions in food intake and body weight compared to control vehicle treatment. Levels of fatty acid synthase (FASN) protein were significantly reduced by xenin treatment, while levels of adipose triglyceride lipase (Atgl) and beta-3 adrenergic receptor (Adrb3) mRNA and phosphorylated hormone sensitive lipase (Ser660-pHSL and Ser563-pHSL) were significantly increased by xenin treatment. These findings suggest that central action of xenin causes alterations in lipid metabolism in adipose tissue toward reduced lipogenesis and increased lipolysis, possibly contributing to xenin-induced body weight reduction. Thus, enhancing central action of xenin and its downstream targets may be possible targets for the treatment of obesity by reducing the amount of stored fat in adipose tissue.


Assuntos
Tecido Adiposo Branco/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Neurotensina/farmacologia , Obesidade/genética , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Ácidos Graxos não Esterificados/sangue , Glicerol/sangue , Lipase/genética , Lipase/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo
16.
Life Sci ; 170: 50-55, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27914922

RESUMO

AIMS: To investigate the role of glucose and insulin in the regulation of hepatic fat mass and obesity associated (Fto) gene expression and the role of hepatic Fto in the regulation of gluconeogenic gene expression. MAIN METHODS: To determine the effect of hyperglycemia on hepatic Fto expression, levels of Fto mRNA in liver were compared between normoglycemic/normoinsulinemic, hypereglycemic/hyperinsulinemic, and hyperglycemic/hypoinsulinemic mice. To determine the direct effect of insulin on Fto expression, levels of Fto, glucose-6-phosphatase (G6pase), and phosphoenolpyruvate carboxykinase (Pepck) mRNA levels were compared between control and insulin-treated mouse liver tissues cultured ex vivo and immortalized mouse hepatocytes AML12. To determine the role of Fto in the regulation of gluconeogenic gene expression, we examined the effect of enhanced Fto expression on G6pase and Pepck mRNA levels in AML12 cells. KEY FINDINGS: Fto mRNA levels were significantly reduced in hyperglycemic/hyperinsulinemic mice compared to normoglycemic/normoinsulinemic mice, while they were indistinguishable between hyperglycemic/hypoinsulinemic mice and normoglycemic/normoinsulinemic mice. Insulin treatment reduced Fto, G6pase, and Pepck mRNA levels compared to control vehicle treatment in both ex vivo cultured mouse liver tissues and AML12 cells. Enhanced Fto expression significantly increased G6pase and Pepck mRNA level in AML12 cells. SIGNIFICANCE: Our findings support the hypothesis that hepatic Fto participates in the maintenance of glucose homeostasis possibly by mediating the inhibitory effect of glucose and insulin on gluconeogenic gene expression in liver. It is further suggested that impairments in nutritional and hormonal regulation of hepatic Fto expression may lead to impairments in glycemic control in diabetes.


Assuntos
Tecido Adiposo/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Hiperglicemia/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Animais , Glicemia/metabolismo , Linhagem Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Gluconeogênese , Glucose/metabolismo , Glucose-6-Fosfatase/metabolismo , Hepatócitos/citologia , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , RNA Mensageiro/metabolismo
17.
Behav Brain Res ; 312: 118-26, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27316340

RESUMO

Xenin is a gut hormone that reduces food intake by partly acting through the hypothalamus via neurotensin receptor 1 (Ntsr1). However, specific signaling pathways that mediate xenin-induced feeding suppression are not fully understood. Activation of Ntsr1 leads to the activation of the extracellular signal-regulated kinase (ERK). Hypothalamic ERK participates in the regulation of food intake by mediating the effect of hormonal signals. Therefore, we hypothesized that the anorectic effect of xenin is mediated by hypothalamic ERK signaling. To address this hypothesis, we compared levels of phosphorylation of ERK1/2 (pERK1/2) in the hypothalamus of both control and xenin-treated mice. The effect of xenin on ERK1/2 phosphorylation was also examined in mouse hypothalamic neuronal cell lines with or without Ntsr1. We also examined the effect of the blockade of central ERK signaling on xenin-induced feeding suppression in mice. The intraperitoneal (i.p.) injection of xenin caused a significant increase in the number of pERK1/2-immunoreactive cells in the hypothalamic arcuate nucleus. The majority of pERK1/2-positive cells expressed neuronal nuclei (NeuN), a marker for neurons. Xenin treatment increased pERK1/2 levels in one cell line expressing Ntsr1 but not another line without Ntsr1 expression. Both i.p. and intracerebroventricular (i.c.v.) injections of xenin reduced food intake in mice. The i.c.v. pre-treatment with U0126, a selective inhibitor of ERK1/2 upstream kinases, did not affect xenin-induced reduction in food intake. These findings suggest that although xenin activates ERK signaling in subpopulations of hypothalamic neurons, xenin does not require the activation of hypothalamic ERK signaling pathway to elicit feeding suppression.


Assuntos
Ingestão de Alimentos , Hipotálamo/metabolismo , Sistema de Sinalização das MAP Quinases , Neurotensina/metabolismo , Animais , Linhagem Celular , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotensina/administração & dosagem , Fosforilação
18.
J Neurosci ; 22(16): 6929-38, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12177191

RESUMO

Targeted deletion of the gene encoding the neuronal and neuroendocrine secreted polypeptide VGF (nonacronymic) produces a lean, hypermetabolic mouse. Consistent with this phenotype, VGF mRNA levels are regulated in the hypothalamic arcuate nucleus in response to fasting. To gain insight into the site(s) and mechanism(s) of action of VGF, we further characterized VGF expression in the hypothalamus. Double-label studies indicated that VGF and pro-opiomelanocortin were coexpressed in lateral arcuate neurons in the fed state, and that VGF expression was induced after fasting in medial arcuate neurons that synthesize neuropeptide Y (NPY). Like NPY, VGF mRNA induction in this region of the hypothalamus in fasted mice was inhibited by exogenous leptin. In leptin-deficient ob/ob and receptor-mutant db/db mice, VGF mRNA levels in the medial arcuate were elevated. To identify neural pathways that are functionally compromised by Vgf ablation, VGF mutant mice were crossed with obese A(y)/a (agouti) and ob/ob mice. VGF deficiency completely blocked the development of obesity in A(y)/a mice, whereas deletion of Vgf in ob/ob mice attenuated weight gain but had no impact on adiposity. Hypothalamic levels of NPY and agouti-related polypeptide mRNAs in both double-mutant lines were dramatically elevated 10- to 15-fold above those of wild-type mice. VGF-deficient mice were also found to resist diet- and gold thioglucose-induced obesity. These data and the susceptibility of VGF mutant mice to monosodium glutamate-induced obesity are consistent with a role for VGF in outflow pathways, downstream of hypothalamic and/or brainstem melanocortin 4 receptors, that project via the autonomic nervous system to peripheral metabolic tissues and regulate energy homeostasis.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Dieta , Peptídeos e Proteínas de Sinalização Intercelular , Neurônios/metabolismo , Obesidade/metabolismo , Proteínas/metabolismo , Proteína Agouti Sinalizadora , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/patologia , Aurotioglucose , Modelos Animais de Doenças , Metabolismo Energético/genética , Jejum/metabolismo , Marcação de Genes , Imuno-Histoquímica , Hibridização In Situ , Leptina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Fatores de Crescimento Neural , Vias Neurais/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neuropeptídeo Y/biossíntese , Neuropeptídeos , Obesidade/induzido quimicamente , Obesidade/patologia , Pró-Opiomelanocortina/biossíntese , Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Resposta de Saciedade , Glutamato de Sódio
19.
Endocrinology ; 146(12): 5151-63, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16141392

RESUMO

Targeted deletion of the gene encoding the neuronal and endocrine secreted peptide precursor called VGF (nonacronymic) produces a lean, hypermetabolic, hyperactive mouse. Because VGF mutant mice are resistant to specific forms of diet-, lesion-, and genetically induced obesity, we investigated the role that this polypeptide plays in glucose homeostasis. We report that VGF mutant mice have increased insulin sensitivity by hyperinsulinemic euglycemic clamp analysis, and by insulin and glucose tolerance testing. Blunted counterregulatory responses in VGF-deficient mice were likely influenced by their significantly lower liver glycogen levels. VGF deficiency lowered circulating glucose and insulin levels in several murine models of obesity that are also susceptible to adult onset diabetes mellitus, including A(y)/a agouti, ob/ob, and MC4R(-)/MC4R(-) mice. Interestingly, ablation of Vgf in ob/ob mice decreased circulating glucose and insulin levels but did not affect adiposity, whereas MC4R(-)/MC4R(-) mice that are additionally deficient in VGF have improved insulin responsiveness at 7-8 wk of age, when lean MC4R(-)/MC4R(-) mice already have impaired insulin tolerance but are not yet obese. VGF mutant mice also resisted developing obesity and hyperglycemia in response to a high-fat/high-carbohydrate diet, and after gold thioglucose treatment, which is toxic to hypothalamic glucose-sensitive neurons. Lastly, circulating adiponectin, an adipose-synthesized protein the levels of which are correlated with improved insulin sensitivity, increased in VGF mutant compared with wild-type mice. Modulation of VGF levels and/or VGF signaling may consequently represent an alternative means to regulate circulating glucose levels and insulin sensitivity.


Assuntos
Hiperglicemia/etiologia , Hiperinsulinismo/etiologia , Obesidade/complicações , Obesidade/metabolismo , Proteínas/metabolismo , Tecido Adiposo/metabolismo , Animais , Dieta/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Ingestão de Energia , Glucagon/metabolismo , Glucose/administração & dosagem , Glucose/metabolismo , Glucose/farmacocinética , Glucose/farmacologia , Homeostase , Hipoglicemia/induzido quimicamente , Hipoglicemia/fisiopatologia , Injeções Intraperitoneais , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural , Neuropeptídeos , Obesidade/genética , Proteínas/genética , Receptor Tipo 4 de Melanocortina/deficiência
20.
Diabetes ; 52(11): 2675-83, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14578285

RESUMO

Hypothalamic proopiomelanocortin (POMC) gene expression is reduced in many forms of obesity and diabetes, particularly in those attributable to deficiencies in leptin or its receptor. To assess the functional significance of POMC in mediating metabolic phenotypes associated with leptin deficiency, leptin-deficient mice bearing a transgene expressing the POMC gene under control of the neuron-specific enolase promoter were produced. The POMC transgene attenuated fasting-induced hyperphagia in wild-type mice. Furthermore, the POMC transgene partially reversed obesity, hyperphagia, and hypothermia and effectively normalized hyperglycemia, glucosuria, glucose intolerance, and insulin resistance in leptin-deficient mice. Effects of the POMC transgene on glucose homeostasis were independent of the partial correction of hyperphagia and obesity. Furthermore, the POMC transgene normalized the profile of hepatic and adipose gene expression associated with gluconeogenesis, glucose output, and insulin sensitivity. These results indicate that central POMC is a key modulator of glucose homeostasis and that agonists of POMC products may provide effective therapy in treating impairments in glucose homeostasis when hypothalamic POMC expression is reduced, as occurs with leptin deficiency, hypothalamic damage, and aging.


Assuntos
Jejum/fisiologia , Hiperfagia/prevenção & controle , Leptina/deficiência , Neurônios/fisiologia , Obesidade/genética , Pró-Opiomelanocortina/genética , Tecido Adiposo/anatomia & histologia , Animais , Sequência de Bases , Peso Corporal , Clonagem Molecular , Primers do DNA , Teste de Tolerância a Glucose , Insulina/sangue , Leptina/genética , Leptina/fisiologia , Camundongos , Camundongos Transgênicos , Fosfopiruvato Hidratase/genética , Pró-Opiomelanocortina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA