Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurosci Lett ; 714: 134550, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31634502

RESUMO

BACKGROUND: Cannabinoid receptor 1 (CB1R) is a GPCR expressed widely in the brain as well as in peripheral metabolic organs. Although pharmacological blockade of CB1R has been effective for the treatment of obesity and tobacco addiction, precise distribution of CB1R within the brain and potential changes by obesity or nicotine exposure have not been thoroughly addressed. METHODS: To examine CB1R distribution within the central energy center, we performed immunostaining and qPCR analysis of micro-dissected hypothalamic nuclei from male C57BL/6 mice. To address the effect of nicotine on food intake and body weight, and on potential changes of CB1R levels in the hypothalamus, mice kept on a high fat diet (HFD) for four weeks were challenged with nicotine intraperitoneally. RESULTS: Validity of the micro-dissected samples was confirmed by the expression of established nucleus-enriched genes. The expression levels of CB1R in the arcuate and lateral nuclei of the hypothalamus were higher than paraventricular and ventral-dorsal medial nuclei. Nicotine administration led to a significant suppression of food intake and body weight either under standard or high fat diet. Neither HFD nor nicotine alone altered CB1R levels in any nucleus tested. By contrast, treatment of HFD-fed mice with nicotine led to a significant increase in CB1R levels in the arcuate, paraventricular and lateral nuclei. CONCLUSIONS: CB1R was widely distributed in multiple hypothalamic nuclei. The expression of CB1R was augmented only when mice were treated with HFD and nicotine in combination. These data suggest that the exposure to nicotine may provoke an enhanced endocannabinoid response in diet-induced obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Dieta Hiperlipídica , Núcleo Hipotalâmico Dorsomedial/metabolismo , Região Hipotalâmica Lateral/metabolismo , Nicotina/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor CB1 de Canabinoide/biossíntese , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Masculino , Camundongos , Microdissecção/métodos , Neuropeptídeo Y/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo
2.
Peptides ; 87: 12-19, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27825986

RESUMO

There is an increasing interest in elucidating the molecular mechanisms by which voluntary exercise is regulated. In this study, we examined how the central nervous system regulates exercise. We used SPORTS rats, which were established in our laboratory as a highly voluntary murine exercise model. SPORTS rats showed lower levels of serum ghrelin compared with those of the parental line of Wistar rats. Intracerebroventricular and intraperitoneal injection of ghrelin decreased wheel-running activity in SPORTS rats. In addition, daily injection of the ghrelin inhibitor JMV3002 into the lateral ventricles of Wistar rats increased wheel-running activity. Co-administration of obestatin inhibited ghrelin-induced increases in food intake but did not inhibit ghrelin-induced suppression of voluntary exercise in rats. Growth hormone secretagogue receptor (GHSR) in the hypothalamus and hippocampus of SPORTS rats was not difference that in control rats. We created an arcuate nucleus destruction model by administering monosodium glutamate (MSG) to neonatal SPORTS rats. Injection of ghrelin into MSG-treated rats decreased voluntary exercise but did not increase food intake, suggesting that wheel-running activity is not controlled by the arcuate nucleus neurons that regulate feeding. These results provide new insights into the mechanism by which ghrelin regulates voluntary activity independent of arcuate nucleus neurons.


Assuntos
Grelina/metabolismo , Atividade Motora/efeitos dos fármacos , Condicionamento Físico Animal , Corrida/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Grelina/administração & dosagem , Infusões Intraventriculares , Atividade Motora/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Glutamato de Sódio/administração & dosagem
3.
Nutr Metab (Lond) ; 13: 7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26839577

RESUMO

BACKGROUND: Obesity increases the risk for malignancies in various tissues including the stomach. Atrophic gastritis with precancerous lesions is an obesity-associated disease; however, the mechanisms that underlie the development of obesity-associated atrophic gastritis are unknown. Leptin is a hormone derived from stomach as well as adipose tissue and gastric leptin is involved in the development of gastric cancer. The aim of the current study is to investigate the involvement of leptin receptor signaling in the development of atrophic gastritis during diet-induced obesity. METHODS: Male C57BL/6, ob/ob and db/db mice were fed a high-fat diet (HFD) or a control diet (CD) from 1 week to 5 months. Pathological changes of the gastric mucosa and the expression of molecules associated with atrophic gastritis were evaluated in these mice. RESULTS: HFD feeding induced gastric mucosal hyperplasia with increased gastric leptin expression. Mucosal hyperplasia was accompanied by a higher frequency of Ki67-positive proliferating cells and atrophy of the gastric glands in the presence of inflammation, which increased following HFD feeding. Activation of ObR signaling-associated molecules such as ObR, STAT3, Akt, and ERK was detected in the gastric mucosa of mice fed the HFD for 1 week. The morphological alterations associated with gastric mucosal atrophy and the expression of Muc2 and Cdx2 resemble those associated with human intestinal metaplasia. In contrast to wild-type mice, leptin-deficient ob/ob mice and leptin receptor-mutated db/db mice did not show increased Cdx2 expression in response to HFD feeding. CONCLUSION: Together, these results suggest that activation of the leptin signaling pathway in the stomach is required to develop obesity-associated atrophic gastritis.

4.
Endocrinology ; 157(1): 245-57, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26505115

RESUMO

Dmbx1 is a brain-specific homeodomain transcription factor expressed primarily during embryogenesis, and its systemic disruption (Dmbx1(-/-)) in the ICR mouse strain resulted in leanness associated with impaired long-lasting orexigenic effect of agouti-related peptide (AgRP). Because spatial and temporal expression patterns of Dmbx1 change dramatically during embryogenesis, it remains unknown when and where Dmbx1 plays a critical role in energy homeostasis. In the present study, the physiological roles of Dmbx1 were examined by its conditional disruption (Dmbx1(loxP/loxP)) in the C57BL/6 mouse strain. Although Dmbx1 disruption in fetal brain resulted in neonatal lethality, its disruption by synapsin promoter-driven Cre recombinase, which eliminated Dmbx1 expression postnatally, exempted the mice (Syn-Cre;Dmbx1(loxP/loxP) mice) from lethality. Syn-Cre;Dmbx1(loxP/loxP) mice show mild leanness and impaired long-lasting orexigenic action of AgRP, demonstrating the physiological relevance of Dmbx1 in the adult. Visualization of Dmbx1-expressing neurons in adult brain using the mice harboring tamoxifen-inducible Cre recombinase in the Dmbx1 locus (Dmbx1(CreERT2/+) mice) revealed Dmbx1 expression in small numbers of neurons in restricted regions, including the lateral parabrachial nucleus (LPB). Notably, c-Fos expression in LPB was increased at 48 hours after AgRP administration in Dmbx1(loxP/loxP) mice but not in Syn-Cre;Dmbx1(loxP/loxP) mice. These c-Fos-positive neurons in LPB did not coincide with neurons expressing Dmbx1 or melanocortin 4 receptor but did coincide with those expressing calcitonin gene-related peptide. Accordingly, Dmbx1 in the adult LPB is required for the long-lasting orexigenic effect of AgRP via the neural circuitry involving calcitonin gene-related peptide neurons.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Regulação do Apetite , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Fatores de Transcrição Otx/metabolismo , Núcleos Parabraquiais/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Depressores do Apetite/farmacologia , Regulação do Apetite/efeitos dos fármacos , Biomarcadores/metabolismo , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Cruzamentos Genéticos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Loci Gênicos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fatores de Transcrição Otx/genética , Núcleos Parabraquiais/citologia , Núcleos Parabraquiais/efeitos dos fármacos , Núcleos Parabraquiais/crescimento & desenvolvimento , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Tamoxifeno/farmacologia
5.
Sci Rep ; 5: 17565, 2015 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-26615883

RESUMO

High-fat diet (HFD) triggers insulin resistance and diabetes mellitus, but their link remains unclear. Characterization of overt hyperglycemia in insulin receptor mutant (Insr(P1195L/+)) mice exposed to HFD (Insr(P1195L/+)/HFD mice) revealed increased glucose-6-phosphatase (G6pc) expression in liver and increased gluconeogenesis from glycerol. Lipolysis in white adipose tissues (WAT) and lipolysis-induced blood glucose rise were increased in Insr(P1195L/+)/HFD mice, while wild-type WAT transplantation ameliorated the hyperglycemia and the increased G6pc expression. We found that the expressions of genes involved in bile acid (BA) metabolism were altered in Insr(P1195L/+)/HFD liver. Among these, the expression of Cyp7a1, a BA synthesis enzyme, was insulin-dependent and was markedly decreased in Insr(P1195L/+)/HFD liver. Reduced Cyp7a1 expression in Insr(P1195L/+)/HFD liver was rescued by WAT transplantation, and the expression of Cyp7a1 was suppressed by glycerol administration in wild-type liver. These findings suggest that unsuppressed lipolysis in adipocytes elicited by HFD feeding is linked with enhanced gluconeogenesis from glycerol and with alterations in BA physiology in Insr(P1195L/+)/HFD liver.


Assuntos
Adipócitos/metabolismo , Ácidos e Sais Biliares/metabolismo , Dieta Hiperlipídica , Gluconeogênese , Lipólise , Receptor de Insulina/genética , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/transplante , Animais , Glicemia , Peso Corporal , Modelos Animais de Doenças , Metabolismo Energético , Gorduras/metabolismo , Genótipo , Glicerol/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Insulina/metabolismo , Resistência à Insulina , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Pirúvico/metabolismo
6.
Endocrinology ; 156(10): 3680-94, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26132918

RESUMO

Adipose tissue macrophages (ATMs) play an important role in the inflammatory response in obese animals. How ATMs are regulated in lean animals has remained elusive, however. We now show that the sympathetic nervous system (SNS) is necessary to maintain the abundance of the mRNA for the proinflammatory cytokine TNF-α at a low level in ATMs of lean mice. Intracerebroventricular injection of agouti-related neuropeptide increased the amount of TNF-α mRNA in epididymal (epi) white adipose tissue (WAT), but not in interscapular brown adipose tissue (BAT), through inhibition of sympathetic nerve activity in epiWAT. The surgical denervation and ß-adrenergic antagonist propranolol up-regulated TNF-α mRNA in both epiWAT and BAT in vivo. Signaling by the ß2-adrenergic receptor (AR) and protein kinase A down-regulated TNF-α mRNA in epiWAT explants and suppressed lipopolysaccharide-induced up-regulation of TNF-α mRNA in the stromal vascular fraction of this tissue. ß-AR-deficient (ß-less) mice manifested an increased plasma TNF-α concentration and increased TNF-α mRNA abundance in epiWAT and BAT. TNF-α mRNA abundance was greater in ATMs (CD11b(+) cells of the stromal vascular fraction) from epiWAT or BAT of wild-type mice than in corresponding CD11b(-) cells, and ß2-AR mRNA abundance was greater in ATMs than in CD11b(-) cells of epiWAT. Our results show that the SNS and ß2-AR-protein kinase A pathway maintain an anti-inflammatory state in ATMs of lean mice in vivo, and that the brain melanocortin pathway plays a role in maintaining this state in WAT of lean mice via the SNS.


Assuntos
Tecido Adiposo Branco/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Sistema Nervoso Simpático/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/inervação , Antagonistas Adrenérgicos beta/farmacologia , Proteína Relacionada com Agouti/administração & dosagem , Animais , Linhagem Celular , Epididimo/efeitos dos fármacos , Epididimo/metabolismo , Expressão Gênica/efeitos dos fármacos , Immunoblotting , Injeções Intraventriculares , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/administração & dosagem , Propranolol/farmacologia , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simpatectomia , Fator de Necrose Tumoral alfa/genética
7.
Cell Mol Life Sci ; 70(6): 1123-33, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23124190

RESUMO

Insulin is the main glucoregulator that promotes the uptake of glucose by tissues and the subsequent utilization of glucose as an energy source. In this paper, we describe a novel glucoregulator, the alpha-synuclein (SNCA) protein, that has previously been linked to Parkinson's disease. Treatment with recombinant SNCA promotes glucose uptake in vitro in preadipocytes and in vivo in the adipose tissues and skeletal muscles of mice through the LPAR2/Gab1/PI3K/Akt pathway; these effects occur independently of the insulin receptor. This function of SNCA represents a new mechanistic insight that creates novel avenues of research with respect to the process of glucose regulation.


Assuntos
Adipócitos/metabolismo , Glucose/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/fisiologia , alfa-Sinucleína/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adipócitos/fisiologia , Análise de Variância , Animais , Calorimetria Indireta , Linhagem Celular , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
PLoS One ; 6(4): e18324, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21694754

RESUMO

Insulin and leptin intracellular signaling pathways converge and act synergistically on the hypothalamic phosphatidylinositol-3-OH kinase/3-phosphoinositide-dependent protein kinase 1 (PDK1). However, little is known about whether PDK1 in agouti-related peptide (AGRP) neurons contributes to energy homeostasis. We generated AGRP neuron-specific PDK1 knockout (AGRPPdk1(-/-)) mice and mice with selective expression of transactivation-defective Foxo1 (Δ256Foxo1(AGRP)Pdk1(-/-)). The AGRPPdk1(-/-) mice showed reductions in food intake, body length, and body weight. The Δ256Foxo1(AGRP)Pdk1(-/-) mice showed increased body weight, food intake, and reduced locomotor activity. After four weeks of calorie-restricted feeding, oxygen consumption and locomotor activity were elevated in AGRPPdk1(-/-) mice and reduced in Δ256Foxo1(AGRP)Pdk1(-/-) mice. In vitro, ghrelin-induced changes in [Ca(2+)](i) and inhibition of ghrelin by leptin were significantly attenuated in AGRPPdk1(-/-) neurons compared to control neurons. However, ghrelin-induced [Ca(2+)](i) changes and leptin inhibition were restored in Δ256Foxo1(AGRP)Pdk1(-/-) mice. These results suggested that PDK1 and Foxo1 signaling pathways play important roles in the control of energy homeostasis through AGRP-independent mechanisms.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Fatores de Transcrição Forkhead/metabolismo , Melanocortinas/metabolismo , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Cálcio/metabolismo , Restrição Calórica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Feminino , Proteína Forkhead Box O1 , Técnicas de Inativação de Genes , Grelina/farmacologia , Homeostase/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Fenótipo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
9.
Endocrinology ; 152(5): 2100-12, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21343250

RESUMO

The nuclear receptor steroidogenic factor 1/adrenal 4 binding protein (SF-1/Ad4BP) is an essential regulator of endocrine development and function, and the expression of the corresponding gene (sf-1/ad4bp) is precisely regulated in a time- and tissue-dependent manner. We previously demonstrated that the basal promoter of sf-1/ad4bp is controlled by DNA methylation and that its methylation status reflects the expression pattern of SF-1/Ad4BP. Recently, three intronic enhancers were identified in the sf-1/ad4bp gene that target SF-1/Ad4BP expression to the fetal adrenal (FAdE; fetal adrenal-specific enhancer), to pituitary gonadotropes (PGE; pituitary gonadotrope-specific enhancer), and to the ventromedial hypothalamic nucleus (VMHE; ventromedial hypothalamic nucleus-specific enhancer). Here, we demonstrate that the activity of these enhancers is correlated with their DNA methylation status. We show that they are hypomethylated in tissues where they are active and generally hypermethylated in tissues where they are not active. Furthermore, we demonstrate in transient transfection experiments that forced DNA methylation represses reporter gene activity driven by these enhancers. These data directly demonstrate a functional significance for the enhancers' methylation status. Intriguingly, further analyses of the basal promoter in gonadotropes revealed that it is methylated in these cells, in contrast to other SF-1/Ad4BP-expressing tissues. Consistent with this, sf-1/ad4bp is transcribed from an alternative promoter in gonadotropes. Taken together, our experiments show that the tissue-specific expression of SF-1/Ad4BP is epigenetically regulated and identify tissue-specific differentially methylated regions within the sf-1/ad4bp locus that are essential for its transcriptional control.


Assuntos
Metilação de DNA , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Fator Esteroidogênico 1/genética , Glândulas Suprarrenais/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Ilhas de CpG/genética , Feminino , Imunofluorescência , Gonadotrofos/metabolismo , Íntrons/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética
10.
Diabetes ; 60(1): 119-26, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20921208

RESUMO

OBJECTIVE: TRPM2 is a Ca²(+)-permeable nonselective cation channel activated by adenosine dinucleotides. We previously demonstrated that TRPM2 is activated by coapplication of heat and intracellular cyclic adenosine 5'-diphosphoribose, which has been suggested to be involved in intracellular Ca²(+) increase in immunocytes and pancreatic ß-cells. To clarify the involvement of TRPM2 in insulin secretion, we analyzed TRPM2 knockout (TRPM2-KO) mice. RESEARCH DESIGN AND METHODS: Oral and intraperitoneal glucose tolerance tests (OGTT and IPGTT) were performed in TRPM2-KO and wild-type mice. We also measured cytosolic free Ca²(+) in single pancreatic cells using fura-2 microfluorometry and insulin secretion from pancreatic islets. RESULTS: Basal blood glucose levels were higher in TRPM2-KO mice than in wild-type mice without any difference in plasma insulin levels. The OGTT and IPGTT demonstrated that blood glucose levels in TRPM2-KO mice were higher than those in wild-type mice, which was associated with an impairment in insulin secretion. In isolated ß-cells, smaller intracellular Ca²(+) increase was observed in response to high concentrations of glucose and incretin hormone in TRPM2-KO cells than in wild-type cells. Moreover, insulin secretion from the islets of TRPM2-KO mice in response to glucose and incretin hormone treatment was impaired, whereas the response to tolbutamide, an ATP-sensitive potassium channel inhibitor, was not different between the two groups. CONCLUSIONS: These results indicate that TRPM2 is involved in insulin secretion stimulated by glucose and that further potentiated by incretins. Thus, TRPM2 may be a new target for diabetes therapy.


Assuntos
Glicemia/metabolismo , Insulina/metabolismo , Canais de Cátion TRPM/deficiência , Animais , Glicemia/efeitos dos fármacos , Cálcio/metabolismo , Teste de Tolerância a Glucose , Temperatura Alta , Insulina/sangue , Insulina/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Canais de Cátion TRPM/fisiologia
11.
J Mol Neurosci ; 43(2): 217-24, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21086064

RESUMO

Orexins are synthesized by lateral hypothalamic neurons and are suggested to be implicated in feeding behavior. Recent studies have shown that intracerebroventricular administration of orexin-A increases intake of sweet-tasting solution. Effects of suppressing the orexin system on consumption of sweet-tasting solution and sensory processing with sweet taste inputs, however, have yet to be examined. We examined the effects of orexin deficiency on sucrose solution intake, locomotor activity, and preference for sweet solution using male orexin knockout (OxKO) and littermate wild-type (WT) mice. In the dark and over 24-h periods, OxKO mice showed significantly less sucrose intake and lower locomotor activity than WT mice without alteration in food intake whereas preferences for 100 mM sucrose were not different between the genotypes. Moreover, sucrose intake of OxKO mice was significantly less than sucrose intake of a subgroup of WT mice with similar locomotor activity compared to that of OxKO mice. These results suggest that factors other than the lower energy expenditure due to lower locomotor activity are likely responsible for the decreased sucrose intake of OxKO mice. Orexin deficiency may lower the satiety threshold resulting in reduced sucrose intake, without altering food intake.


Assuntos
Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Preferências Alimentares/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Camundongos Knockout , Atividade Motora/fisiologia , Neuropeptídeos/deficiência , Sacarose/administração & dosagem , Animais , Metabolismo Energético , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeos/genética , Orexinas , Paladar
12.
Cell Metab ; 12(5): 483-95, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21035759

RESUMO

The liver may regulate glucose homeostasis by modulating the sensitivity/resistance of peripheral tissues to insulin, by way of the production of secretory proteins, termed hepatokines. Here, we demonstrate that selenoprotein P (SeP), a liver-derived secretory protein, causes insulin resistance. Using serial analysis of gene expression (SAGE) and DNA chip methods, we found that hepatic SeP mRNA levels correlated with insulin resistance in humans. Administration of purified SeP impaired insulin signaling and dysregulated glucose metabolism in both hepatocytes and myocytes. Conversely, both genetic deletion and RNA interference-mediated knockdown of SeP improved systemic insulin sensitivity and glucose tolerance in mice. The metabolic actions of SeP were mediated, at least partly, by inactivation of adenosine monophosphate-activated protein kinase (AMPK). In summary, these results demonstrate a role of SeP in the regulation of glucose metabolism and insulin sensitivity and suggest that SeP may be a therapeutic target for type 2 diabetes.


Assuntos
Resistência à Insulina , Fígado/metabolismo , Selenoproteína P/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Glucose/metabolismo , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido Palmítico/metabolismo , Fosforilação , Interferência de RNA , RNA Mensageiro/genética , Ratos , Selenoproteína P/genética
13.
Endocrinology ; 151(6): 2556-66, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375183

RESUMO

Silent information regulator (SIR)2 is an nicotinamide adenine dinucleotide dependent deacetylase implicated in the regulation of life span in species as diverse as yeast, worms, and flies. Mammalian Sirt1 is the most closely related homolog of the SIR2 gene. Pharmacological activators of Sirt1 have been reported to increase the life span and improve the health of mice fed a high-fat diet and to reverse diabetes in rodents. Sirt1 links the energy availability status with cellular metabolism in peripheral organs including liver, pancreas, muscle, and white adipose tissue. Insulin and leptin signaling regulate food intake by controlling the expression of orexigenic and anorexigenic neuropeptides in the arcuate nucleus of the hypothalamus via Forkhead box O (Foxo)-1 and signal transducer and activator of transcription-3. Sirt1 has been reported to improve insulin sensitivity in vitro, but the role of hypothalamic Sirt1 in regulating feeding has not been addressed. We found that hypothalamic Sirt1 protein levels increase on feeding, and this induction is abrogated in diet-induced obese mice and db/db mice. We also demonstrate for the first time that Sirt1 protein turnover is regulated by the proteasome and ubiquitination in a hypothalamic cell line and in vivo by feeding, and this regulation is not seen in a pituitary cell line AtT20. Forced expression of wild-type Sirt1 in the mediobasal hypothalamus by adenovirus microinjection suppressed Foxo1-induced hyperphagia, a model for central insulin resistance. Moreover, Sirt1 suppressed Foxo1-dependent expression of the orexigenic neuropeptide Agouti-related peptide in vitro. We propose that on feeding, Sirt1 protein is stabilized in the hypothalamus, leading to decreased Foxo1-dependent expression of orexigenic neuropeptide Agouti-related peptide and cessation of feeding.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Sirtuína 1/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Western Blotting , Linhagem Celular , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Hiperfagia/metabolismo , Hiperfagia/fisiopatologia , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/genética , Aumento de Peso/genética , Aumento de Peso/fisiologia
14.
Nutrition ; 24(9): 786-90, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18725075

RESUMO

Adenosine monophosphate-activated protein kinase (AMPK) functions as a cellular fuel gauge that regulates metabolic pathways in nutrient metabolism. Recent studies have strongly implicated that AMPK in the hypothalamus regulates energy metabolism by integrating inputs from multiple hormones, peptides, neurotransmitters, and nutrients. Leptin is an adipocyte hormone that regulates food intake and energy expenditure in peripheral tissues. Leptin inhibits AMPK activity in the arcuate and paraventricular hypothalamus, and its inhibition is necessary for the anorexic effect of leptin. Alteration of hypothalamic AMPK activity is sufficient to change food intake and body weight. Furthermore, fasting/refeeding, glucose, and melanocortin receptor alter AMPK activity in the hypothalamus. Adiponectin has also been shown to increase food intake by activating AMPK in the arcuate hypothalamus. Recent data have shown that acetyl-coenzyme A carboxylase/malonyl-coenzyme A/carnitine palmitoyltransferase-1/fatty acid oxidation and mammalian target of rapamycin signalings are putative downstream pathways for food intake regulation in response to hypothalamic AMPK. Thus, these results suggest that food intake and nutrient metabolism are coordinately regulated by the common signaling pathway of AMPK in the hypothalamus.


Assuntos
Adenilato Quinase/metabolismo , Comportamento Alimentar/fisiologia , Hipotálamo/enzimologia , Hipotálamo/fisiologia , Animais , Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Hormônios/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos , Transdução de Sinais/fisiologia
15.
J Biol Chem ; 282(42): 30794-803, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17724031

RESUMO

In obese individuals, white adipose tissue (WAT) is infiltrated by large numbers of macrophages, resulting in enhanced inflammatory responses that contribute to insulin resistance. Here we show that expression of the CXC motif chemokine ligand-14 (CXCL14), which targets tissue macrophages, is elevated in WAT of obese mice fed a high fat diet (HFD) compared with lean mice fed a regular diet. We found that HFD-fed CXCL14-deficient mice have impaired WAT macrophage mobilization and improved insulin responsiveness. Insulin-stimulated phosphorylation of Akt kinase in skeletal muscle was severely attenuated in HFD-fed CXCL14+/- mice but not in HFD-fed CXCL14-/- mice. The insulin-sensitive phenotype of CXCL14-/- mice after HFD feeding was prominent in female mice but not in male mice. HFD-fed CXCL14-/- mice were protected from hyperglycemia, hyperinsulinemia, and hypoadiponectinemia and did not exhibit increased levels of circulating retinol-binding protein-4 and increased expression of interleukin-6 in WAT. Transgenic overexpression of CXCL14 in skeletal muscle restored obesity-induced insulin resistance in CXCL14-/- mice. CXCL14 attenuated insulin-stimulated glucose uptake in cultured myocytes and to a lesser extent in cultured adipocytes. These results demonstrate that CXCL14 is a critical chemoattractant of WAT macrophages and a novel regulator of glucose metabolism that functions mainly in skeletal muscle.


Assuntos
Tecido Adiposo Branco/metabolismo , Quimiocinas CXC/metabolismo , Glucose/metabolismo , Resistência à Insulina , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo Branco/patologia , Animais , Movimento Celular/genética , Quimiocinas CXC/genética , Gorduras na Dieta/administração & dosagem , Feminino , Regulação da Expressão Gênica/genética , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patologia , Inflamação/metabolismo , Inflamação/patologia , Insulina/metabolismo , Resistência à Insulina/genética , Interleucina-6/biossíntese , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Obesidade/genética , Obesidade/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Plasmáticas de Ligação ao Retinol/metabolismo , Fatores Sexuais
16.
Cell Metab ; 6(1): 55-68, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17618856

RESUMO

Adiponectin has been shown to stimulate fatty acid oxidation and enhance insulin sensitivity through the activation of AMP-activated protein kinase (AMPK) in the peripheral tissues. The effects of adiponectin in the central nervous system, however, are still poorly understood. Here, we show that adiponectin enhances AMPK activity in the arcuate hypothalamus (ARH) via its receptor AdipoR1 to stimulate food intake; this stimulation of food intake by adiponectin was attenuated by dominant-negative AMPK expression in the ARH. Moreover, adiponectin also decreased energy expenditure. Adiponectin-deficient mice showed decreased AMPK phosphorylation in the ARH, decreased food intake, and increased energy expenditure, exhibiting resistance to high-fat-diet-induced obesity. Serum and cerebrospinal fluid levels of adiponectin and expression of AdipoR1 in the ARH were increased during fasting and decreased after refeeding. We conclude that adiponectin stimulates food intake and decreases energy expenditure during fasting through its effects in the central nervous system.


Assuntos
Adiponectina/fisiologia , Ingestão de Alimentos , Hipotálamo/enzimologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Adenoviridae/genética , Adiponectina/líquido cefalorraquidiano , Adiponectina/genética , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético , Feminino , Hipotálamo/patologia , Técnicas Imunoenzimáticas , Hibridização In Situ , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Sondas RNA , Receptores de Adiponectina , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores para Leptina
17.
Mol Cell Biol ; 27(12): 4317-27, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17420279

RESUMO

Leptin stimulates fatty acid oxidation in skeletal muscle through the activation of AMP-activated protein kinase (AMPK) and the induction of gene expression, such as that for peroxisome proliferator-activated receptor alpha (PPARalpha). We now show that leptin stimulates fatty acid oxidation and PPARalpha gene expression in the C2C12 muscle cell line through the activation of AMPK containing the alpha2 subunit (alpha2AMPK) and through changes in the subcellular localization of this enzyme. Activated alpha2AMPK containing the beta1 subunit was shown to be retained in the cytoplasm, where it phosphorylated acetyl coenzyme A carboxylase and thereby stimulated fatty acid oxidation. In contrast, alpha2AMPK containing the beta2 subunit transiently increased fatty acid oxidation but underwent rapid translocation to the nucleus, where it induced PPARalpha gene transcription. A nuclear localization signal and Thr(172) phosphorylation of alpha2 were found to be essential for nuclear translocation of alpha2AMPK, whereas the myristoylation of beta1 anchors alpha2AMPK in the cytoplasm. The prevention of alpha2AMPK activation and the change in its subcellular localization inhibited the metabolic effects of leptin. Our data thus suggest that the activation of and changes in the subcellular localization of alpha2AMPK are required for leptin-induced stimulation of fatty acid oxidation and PPARalpha gene expression in muscle cells.


Assuntos
Ácidos Graxos/metabolismo , Leptina/fisiologia , Complexos Multienzimáticos/metabolismo , Mioblastos/efeitos dos fármacos , PPAR alfa/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células 3T3-L1 , Proteínas Quinases Ativadas por AMP , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Leptina/administração & dosagem , Leptina/farmacologia , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Complexos Multienzimáticos/química , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Neoplasias de Tecido Muscular/patologia , Sinais de Localização Nuclear , Oxirredução , Proteínas Serina-Treonina Quinases/química , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
18.
Mol Cell Biol ; 25(21): 9713-23, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227617

RESUMO

Mice with muscle-specific knockout of the Glut4 glucose transporter (muscle-G4KO) are insulin resistant and mildly diabetic. Here we show that despite markedly reduced glucose transport in muscle, muscle glycogen content in the fasted state is increased. We sought to determine the mechanism(s). Basal glycogen synthase activity is increased by 34% and glycogen phosphorylase activity is decreased by 17% (P < 0.05) in muscle of muscle-G4KO mice. Contraction-induced glycogen breakdown is normal. The increased glycogen synthase activity occurs in spite of decreased signaling through the insulin receptor substrate 1 (IRS-1)-phosphoinositide (PI) 3-kinase-Akt pathway and increased glycogen synthase kinase 3beta (GSK3beta) activity in the basal state. Hexokinase II is increased, leading to an approximately twofold increase in glucose-6-phosphate levels. In addition, the levels of two scaffolding proteins that are glycogen-targeting subunits of protein phosphatase 1 (PP1), the muscle-specific regulatory subunit (RGL) and the protein targeting to glycogen (PTG), are strikingly increased by 3.2- to 4.2-fold in muscle of muscle-G4KO mice compared to wild-type mice. The catalytic activity of PP1, which dephosphorylates and activates glycogen synthase, is also increased. This dominates over the GSK3 effects, since glycogen synthase phosphorylation on the GSK3-regulated site is decreased. Thus, the markedly reduced glucose transport in muscle results in increased glycogen synthase activity due to increased hexokinase II, glucose-6-phosphate, and RGL and PTG levels and enhanced PP1 activity. This, combined with decreased glycogen phosphorylase activity, results in increased glycogen content in muscle in the fasted state when glucose transport is reduced.


Assuntos
Transportador de Glucose Tipo 4/fisiologia , Glicogênio/metabolismo , Músculo Esquelético/metabolismo , Animais , Jejum/metabolismo , Feminino , Transportador de Glucose Tipo 4/genética , Glucose-6-Fosfato/metabolismo , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Hexoquinase/metabolismo , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicogênio Hepático/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteínas Proto-Oncogênicas c-akt/metabolismo
19.
Cell Metab ; 1(2): 107-19, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16054052

RESUMO

Fatty acid binding proteins (FABPs) are cytosolic fatty acid chaperones whose biological role and mechanisms of action are not well understood. Here, we developed mice with targeted mutations in two related adipocyte FABPs, aP2 and mal1, to resolve their role in systemic lipid, glucose, and energy metabolism. Mice lacking aP2 and mal1 exhibited a striking phenotype with strong protection from diet-induced obesity, insulin resistance, type 2 diabetes, and fatty liver disease. These mice have altered cellular and systemic lipid transport and composition, leading to enhanced insulin receptor signaling, enhanced muscle AMP-activated kinase (AMP-K) activity, and dramatically reduced liver stearoyl-CoA desaturase-1 (SCD-1) activity underlying their phenotype. Taken together with the previously reported strong protection against atherosclerosis, these results demonstrate that adipocyte/macrophage FABPs have a robust impact on multiple components of metabolic syndrome, integrating metabolic and inflammatory responses in mice and constituting a powerful target for the treatment of these diseases.


Assuntos
Adipócitos/citologia , Proteínas de Transporte/metabolismo , Diabetes Mellitus/metabolismo , Macrófagos/citologia , Obesidade/metabolismo , Proteínas Quinases Ativadas por AMP , Adipócitos/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Arteriosclerose/metabolismo , Peso Corporal , Citocinas/metabolismo , Proteínas de Ligação a Ácido Graxo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Immunoblotting , Inflamação , Insulina/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexos Multienzimáticos/metabolismo , Mutação , Oxigênio/metabolismo , Fenótipo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Receptor de Insulina/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Fatores de Tempo , Distribuição Tecidual , Triglicerídeos/metabolismo
20.
Diabetes ; 54(8): 2365-74, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16046303

RESUMO

Leptin augments glucose and lipid metabolism independent of its effect on satiety. Administration of leptin in rodents increases skeletal muscle beta-oxidation by activating AMP-activated protein kinase (AMPK). We previously reported that, as hyperleptinemic as obese human subjects, transgenic skinny mice overexpressing leptin in liver (LepTg) exhibit enhanced insulin sensitivity and lipid clearance. To assess skeletal muscle AMPK activity in leptin-sensitive and -insensitive states, we examined phosphorylation of AMPK and its target, acetyl CoA carboxylase (ACC), in muscles from LepTg under dietary modification. Here we show that phosphorylation of AMPK and ACC are chronically augmented in LepTg soleus muscle, with a concomitant increase in the AMP-to-ATP ratio and a significant decrease in tissue triglyceride content. Despite preexisting hyperleptinemia, high-fat diet (HFD)-fed LepTg develop obesity, insulin-resistance, and hyperlipidemia. In parallel, elevated soleus AMPK and ACC phosphorylation in regular diet-fed LepTg is attenuated, and tissue triglyceride content is increased in those given HFD. Of note, substitution of HFD with regular diet causes a robust recovery of soleus AMPK and ACC phosphorylation in LepTg, with a higher rate of body weight reduction and a regain of insulin sensitivity. In conclusion, soleus AMPK and ACC phosphorylation in LepTg changes in parallel with its insulin sensitivity under dietary modification, suggesting a close association between skeletal muscle AMPK activity and sensitivity to leptin.


Assuntos
Dieta , Leptina/genética , Complexos Multienzimáticos/metabolismo , Músculo Esquelético/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Acetil-CoA Carboxilase/metabolismo , Monofosfato de Adenosina/análise , Trifosfato de Adenosina/análise , Animais , Proteínas de Transporte/genética , Gorduras na Dieta/administração & dosagem , Intolerância à Glucose/genética , Hiperlipidemias/genética , Resistência à Insulina/genética , Canais Iônicos , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mitocondriais , Músculo Esquelético/química , Obesidade/genética , Fosforilação , RNA Mensageiro/análise , Estearoil-CoA Dessaturase/genética , Triglicerídeos/análise , Proteína Desacopladora 1 , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA