Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 6: 21721, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26883167

RESUMO

Although obesity is undoubtedly major risk for non-alcoholic steatohepatitis (NASH), the presence of lean NASH patients with normal body mass index has been recognized. Here, we report that the insufficiency of phosphatidylethanolamine N-methyltransferase (PEMT) is a risk for the lean NASH. The Pemt-/- mice fed high fat-high sucrose (HFHS) diet were protected from diet-induced obesity and diabetes, while they demonstrated prominent steatohepatitis and developed multiple liver tumors. Pemt exerted inhibitory effects on p53-driven transcription by forming the complex with clathrin heavy chain and p53, and Pemt-/- mice fed HFHS diet demonstrated prominent apoptosis of hepatocytes. Furthermore, hypermethylation and suppressed mRNA expression of F-box protein 31 and hepatocyte nuclear factor 4α resulted in the prominent activation of cyclin D1. PEMT mRNA expression in liver tissues of NASH patients was significantly lower than those with simple steatosis and we postulated the distinct clinical entity of lean NASH with insufficiency of PEMT activities.


Assuntos
Hepatócitos/citologia , Hepatopatia Gordurosa não Alcoólica/patologia , Fosfatidiletanolamina N-Metiltransferase/genética , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Animais , Apoptose , Células Cultivadas , Cadeias Pesadas de Clatrina/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Humanos , Camundongos , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/prevenção & controle , Proteína Supressora de Tumor p53/metabolismo
2.
PLoS One ; 9(3): e92647, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667182

RESUMO

Phosphatidylethanolamine N-methyltransferase (Pemt) catalyzes the methylation of phosphatidylethanolamine (PE) to phosphatidylcholine (PC) mainly in the liver. Under an obese state, the upregulation of Pemt induces endoplasmic reticulum (ER) stress by increasing the PC/PE ratio in the liver. We targeted the Pemt gene in mice to explore the therapeutic impact of Pemt on the progression of diabetic nephropathy and diabetes, which was induced by the injection of streptozotocin (STZ). Although the blood glucose levels were similar in STZ-induced diabetic Pemt+/+ and Pemt-/-mice, the glomerular hypertrophy and albuminuria in Pemt-/- mice were significantly reduced. Pemt deficiency reduced the intraglomerular F4/80-positive macrophages, hydroethidine fluorescence, tubulointerstitial fibrosis and tubular atrophy. The expression of glucose-regulated protein-78 (GRP78) was enriched in the renal tubular cells in STZ-induced diabetic mice, and this was ameliorated by Pemt deficiency. In mProx24 renal proximal tubular cells, the treatment with ER-stress inducers, tunicamycin and thapsigargin, increased the expression of GRP78, which was reduced by transfection of a shRNA lentivirus for Pemt (shRNA-Pemt). The number of apoptotic cells in the renal tubules was significantly reduced in Pemt-/- diabetic mice, and shRNA-Pemt upregulated the phosphorylation of Akt and decreased the cleavage of caspase 3 and 7 in mProx24 cells. Taken together, these findings indicate that the inhibition of Pemt activity ameliorates the ER stress associated with diabetic nephropathy in a model of type 1 diabetes and corrects the functions of the three major pathways downstream of ER stress, i.e. oxidative stress, inflammation and apoptosis.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatias Diabéticas/metabolismo , Estresse do Retículo Endoplasmático , Túbulos Renais Proximais/metabolismo , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Animais , Apoptose/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Knockout , Estresse Oxidativo/genética , Fosfatidiletanolamina N-Metiltransferase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
J Biol Chem ; 286(43): 37458-69, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21862590

RESUMO

Krüppel-like factor 15 (KLF15), a member of the Krüppel-like factor family of transcription factors, has been found to play diverse roles in adipocytes in vitro. However, little is known of the function of KLF15 in adipocytes in vivo. We have now found that the expression of KLF15 in adipose tissue is down-regulated in obese mice, and we therefore generated adipose tissue-specific KLF15 transgenic (aP2-KLF15 Tg) mice to investigate the possible contribution of KLF15 to various pathological conditions associated with obesity in vivo. The aP2-KLF15 Tg mice manifest insulin resistance and are resistant to the development of obesity induced by maintenance on a high fat diet. However, they also exhibit improved glucose tolerance as a result of enhanced insulin secretion. Furthermore, this enhancement of insulin secretion was shown to result from down-regulation of the expression of stearoyl-CoA desaturase 1 (SCD1) in white adipose tissue and a consequent reduced level of oxidative stress. This is supported by the findings that restoration of SCD1 expression in white adipose tissue of aP2-KLF15 Tg mice exhibited increased oxidative stress in white adipose tissue and reduced insulin secretion with hyperglycemia. Our data thus provide an example of cross-talk between white adipose tissue and pancreatic ß cells mediated through modulation of oxidative stress.


Assuntos
Adipócitos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Insulina/metabolismo , Estearoil-CoA Dessaturase/biossíntese , Fatores de Transcrição/metabolismo , Adipócitos/patologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Comunicação Celular/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Glucose/genética , Insulina/genética , Resistência à Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Estresse Oxidativo/genética , Ratos , Estearoil-CoA Dessaturase/genética , Fatores de Transcrição/genética
4.
Diabetes ; 59(7): 1608-15, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20393151

RESUMO

OBJECTIVE: An increase in the rate of gluconeogenesis is largely responsible for the hyperglycemia in individuals with type 2 diabetes, with the antidiabetes action of metformin being thought to be achieved at least in part through suppression of gluconeogenesis. RESEARCH DESIGN AND METHODS: We investigated whether the transcription factor KLF15 has a role in the regulation of gluconeogenesis and whether KLF15 participates in the antidiabetes effect of metformin. RESULTS: Here we show that KLF15 regulates the expression of genes for gluconeogenic or amino acid-degrading enzymes in coordination with the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1alpha. Liver-specific ablation of KLF15 in diabetic mice resulted in downregulation of the expression of genes for gluconeogenic or amino acid catabolic enzymes and in amelioration of hyperglycemia. Exposure of cultured hepatocytes to metformin reduced the abundance of KLF15 through acceleration of its degradation and downregulation of its mRNA. Metformin suppressed the expression of genes for gluconeogenic or amino acid-degrading enzymes in cultured hepatocytes, and these effects of metformin were attenuated by restoration of KLF15 expression. Administration of metformin to mice inhibited both the expression of KLF15 and glucose production in the liver, the latter effect also being attenuated by restoration of hepatic KLF15 expression. CONCLUSIONS: KLF15 plays an important role in regulation of the expression of genes for gluconeogenic and amino acid-degrading enzymes and that the inhibitory effect of metformin on gluconeogenesis is mediated at least in part by downregulation of KLF15 and consequent attenuation of the expression of such genes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Gluconeogênese/genética , Hepatócitos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Metformina/metabolismo , Animais , Glicemia/metabolismo , Western Blotting , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Expressão Gênica/efeitos dos fármacos , Gluconeogênese/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hiperglicemia/genética , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Masculino , Metformina/farmacologia , Camundongos , Camundongos Transgênicos , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Biochem Biophys Res Commun ; 381(4): 537-43, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19233136

RESUMO

The transcriptional regulator peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) controls mitochondrial biogenesis and energy homeostasis. Although physical exercise induces PGC-1alpha expression in muscle, the underlying mechanism of this effect has remained incompletely understood. We recently identified a novel muscle-enriched isoform of PGC-1alpha transcript (designated PGC-1alpha-b) that is derived from a previously unidentified first exon. We have now cloned and characterized the human PGC-1alpha-b promoter. The muscle-specific transcription factors MyoD and MRF4 transactivated this promoter through interaction with a proximal E-box motif. Furthermore, either forced expression of Ca(2+)- and calmodulin-dependent protein kinase IV (CaMKIV), calcineurin A, or the p38 mitogen-activated protein kinase (p38 MAPK) kinase MKK6 or the intracellular accumulation of cAMP activated the PGC-1alpha-b promoter in cultured myoblasts through recruitment of cAMP response element (CRE)-binding protein (CREB) to a putative CRE located downstream of the E-box. Our results thus reveal a potential molecular basis for isoform-specific regulation of PGC-1alpha expression in contracting muscle.


Assuntos
Proteínas de Choque Térmico/genética , Contração Muscular/genética , Músculo Esquelético/fisiologia , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional , Sequência de Bases , Cálcio/farmacologia , AMP Cíclico/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Humanos , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição de p300-CBP/metabolismo
6.
Biochem Biophys Res Commun ; 379(1): 98-103, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-19094967

RESUMO

KLF15 (Krüppel-like factor 15) plays a key role in adipocyte differentiation and glucose transport in adipocytes through activation of its target genes. We have now identified six target genes regulated directly by KLF15 in 3T3-L1 mouse adipocytes with the use of a combination of microarray-based chromatin immunoprecipitation and gene expression analyses. We confirmed the direct regulation by KLF15 of one of these genes, that for adrenomedullin, with the use of a luciferase reporter assay in 3T3-L1 preadipocytes and adipocytes. Such analysis revealed that the most proximal CACCC element in the promoter of the human adrenomedullin gene (located in the region spanning nucleotides -70 and -29) is required for trans-inhibition by KLF15. Furthermore, chromatin immunoprecipitation showed that KLF15 binds to this region of the human adrenomedullin gene promoter in cultured human adipocytes. These results thus implicate KLF15 in the regulation of adrenomedullin expression in adipose tissue.


Assuntos
Adipócitos/metabolismo , Adrenomedulina/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição/metabolismo , Células 3T3 , Adrenomedulina/antagonistas & inibidores , Animais , Imunoprecipitação da Cromatina , Regulação para Baixo , Humanos , Fatores de Transcrição Kruppel-Like , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transcrição Gênica
7.
J Mol Endocrinol ; 42(2): 161-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19060180

RESUMO

Gene related to anergy in lymphocytes (GRAIL) is an E3 ubiquitin ligase that regulates energy in T-lymphocytes. Whereas, the relevance of GRAIL to T lymphocyte function is well established, the role of this protein in other cell types remains unknown. Given that GRAIL is abundant in the liver, we investigated the potential function of GRAIL in nutrient metabolism by generating mice in which the expression of GRAIL is reduced specifically in the liver. Adenovirus-mediated transfer of a short hairpin RNA specific for GRAIL mRNA markedly reduced the amounts of GRAIL mRNA and protein in the liver. Blood glucose levels of the mice with hepatic GRAIL deficiency did not differ from those of control animals in the fasted or fed states. However, these mice manifested glucose intolerance in association with a normal increase in plasma insulin levels during glucose challenge. The mice also manifested an increase in the serum concentration of free fatty acids, whereas the serum levels of cholesterol and triglyceride were unchanged. The hepatic abundance of mRNAs for glucose-6-phosphatase, catalytic (a key enzyme in hepatic glucose production) and for sterol regulatory element-binding transcription factor 1 (an important transcriptional regulator of lipogenesis) was increased in the mice with hepatic GRAIL deficiency, possibly contributing to the metabolic abnormalities of these animals. Our results thus demonstrate that GRAIL in the liver is essential for maintenance of normal glucose and lipid metabolism in living animals.


Assuntos
Anergia Clonal/imunologia , Glucose/metabolismo , Metabolismo dos Lipídeos , Fígado/enzimologia , Linfócitos/enzimologia , Linfócitos/imunologia , Ubiquitina-Proteína Ligases/genética , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/metabolismo
8.
Biochem Biophys Res Commun ; 379(2): 249-54, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19109928

RESUMO

Skp2, the substrate-binding subunit of an SCF ubiquitin ligase complex, is a key regulator of cell cycle progression that targets substrates for degradation by the 26S proteasome. We have now shown that ablation of Skp2 in primary mouse embryonic fibroblasts (MEFs) results both in impairment of adipocyte differentiation and in the accumulation of the cyclin-dependent kinase inhibitor p27(Kip1), a principal target of the SCF(Skp2) complex. Genetic ablation of p27(Kip1) in MEFs promoted both lipid accumulation and adipocyte-specific gene expression. However, depletion of p27(Kip1) by adenovirus-mediated RNA interference failed to correct the impairment of adipocyte differentiation in Skp2(-/-) MEFs. In contrast, troglitazone, a high-affinity ligand for peroxisome proliferator-activated receptor gamma (PPARgamma), largely restored lipid accumulation and PPARgamma gene expression in Skp2(-/-) MEFs. Our data suggest that Skp2 plays an essential role in adipogenesis in MEFs in a manner that is at least in part independent of regulation of p27(Kip1) expression.


Assuntos
Adipócitos/fisiologia , Adipogenia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fibroblastos/citologia , Proteínas Quinases Associadas a Fase S/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Cromanos/farmacologia , Embrião de Mamíferos/citologia , Camundongos , Camundongos Knockout , Proteínas Quinases Associadas a Fase S/genética , Tiazolidinedionas/farmacologia , Troglitazona
9.
J Biol Chem ; 283(25): 17702-11, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18430722

RESUMO

PDK1 (3-phosphoinositide-dependent protein kinase 1) is a key mediator of signaling by phosphoinositide 3-kinase. To gain insight into the physiological importance of PDK1 in cell proliferation and cell cycle control, we established immortalized mouse embryonic fibroblasts (MEFs) from mice homozygous for a "floxed" allele of Pdk1 and from wild-type mice. Introduction of Cre recombinase by retrovirus-mediated gene transfer resulted in the depletion of PDK1 in Pdk1(lox/lox) MEFs but not in Pdk1(+/+) MEFs. The insulin-like growth factor-1-induced phosphorylation of various downstream effectors of PDK1, including Akt, glycogen synthase kinase 3, ribosomal protein S6, and p70 S6 kinase, was markedly inhibited in the PDK1-depleted (Pdk1-KO) MEFs. The rate of serum-induced cell proliferation was reduced; progression of the cell cycle from the G(0)-G(1) phase to the S phase was delayed, and cell cycle progression at G(2)-M phase was impaired in Pdk1-KO MEFs. These cells also manifested an increased level of p27(Kip1) expression and a reduced level of cyclin D1 expression during cell cycle progression. The defect in cell cycle progression from the G(0)-G(1) to the S phase in Pdk1-KO MEFs was rescued by forced expression of cyclin D1, whereas rescue of the defect in G(2)-M progression in these cells required both overexpression of cyclin D1 and depletion of p27(Kip1) by RNA interference. These data indicate that PDK1 plays an important role in cell proliferation and cell cycle progression by controlling the expression of both cyclin D1 and p27(Kip1).


Assuntos
Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Adenoviridae/metabolismo , Animais , Apoptose , Ciclo Celular , Proliferação de Células , DNA/química , Fibroblastos/metabolismo , Homozigoto , Camundongos , Camundongos Knockout , Modelos Biológicos , Oligonucleotídeos/química , Proteínas Serina-Treonina Quinases/genética , Fatores de Tempo
10.
Kobe J Med Sci ; 54(4): E200-8, 2008 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19258740

RESUMO

Regulation of hepatic gene expression is largely responsible for the control of nutrient metabolism. We previously showed that the transcription factor STAT3 regulates glucose homeostasis by suppressing the expression of gluconeogenic genes in the liver. However, the role of STAT3 in the control of lipid metabolism has remained unknown. We have now investigated the effects of hepatic overexpression of STAT3, achieved by adenovirus-mediated gene transfer, on glucose and lipid metabolism in insulin-resistant diabetic mice. Forced expression of STAT3 reduced blood glucose and plasma insulin concentrations as well as the hepatic abundance of mRNA for phosphoenolpyruvate carboxykinase. However, it also increased the plasma levels of triglyceride and total cholesterol without affecting those of low density lipoprotein- or high density lipoprotein-cholesterol. The hepatic abundance of mRNAs for fatty acid synthase and acetyl-CoA carboxylase, both of which catalyze the synthesis of fatty acids, was increased by overexpression of STAT3, whereas that of mRNAs for sterol regulatory element-binding proteins 1a, 1c, or 2 was unaffected. Moreover, the amount of mRNA for acyl-CoA oxidase, which contributes to beta-oxidation, was decreased by forced expression of STAT3. These results indicate that forced activation of STAT3 signaling in the liver of insulin-resistant diabetic mice increased the circulating levels of atherogenic lipids through changes in the hepatic expression of genes involved in lipid metabolism. Furthermore, these alterations in hepatic gene expression likely occurred through a mechanism independent of sterol regulatory element-binding proteins.


Assuntos
Metabolismo dos Lipídeos , Fígado/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Glicemia/metabolismo , Regulação da Expressão Gênica , Gluconeogênese/genética , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/genética
12.
Diabetes ; 56(4): 1000-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17267763

RESUMO

Phosphoinositide-dependent kinase-1 (PDK1) is implicated in the metabolic effects of insulin as a key mediator of phosphoinositide 3-kinase-dependent signaling. Here we show that mice with liver-specific PDK1 deficiency manifest various defects in the metabolic actions of insulin in the liver as well as a type 2 diabetes-like phenotype characterized by marked hyperinsulinemia and postprandial hyperglycemia. The hepatic abundance of glucokinase, an important determinant of glucose flux and glucose-evoked signaling in hepatocytes, was substantially reduced in these mice. Restoration of hepatic glucokinase expression, with the use of an adenoviral vector, induced insulin-like effects in the liver and almost completely normalized the fasting hyperinsulinemia and postprandial hyperglycemia in these animals. These results indicate that, if the hepatic abundance of glucokinase is maintained, ingested glucose is normally disposed of even in the absence of acute activation of proximal insulin signaling, such as the activation of Akt, in the liver.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucoquinase/genética , Fígado/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Primers do DNA , Glucose/metabolismo , Teste de Tolerância a Glucose , Insulina/farmacologia , Camundongos , Camundongos Transgênicos , Período Pós-Prandial , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Biol Chem ; 282(3): 2038-46, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17082193

RESUMO

The increase in the mass of adipose tissue during the development of obesity can arise through an increase in cell size, an increase in cell number, or both. Here we show that long term maintenance of C57BL/6 mice on a high fat diet (for approximately 25 weeks) induces an initial increase in adipocyte size followed by an increase in adipocyte number in white adipose tissue. The latter effect was found to be accompanied by up-regulation of expression of the gene for the F-box protein Skp2 as well as by downregulation of the cyclin-dependent kinase inhibitor p27(Kip1), a principal target of the SCF(Skp2) ubiquitin ligase, in white adipose tissue. Ablation of Skp2 protected mice from the development of obesity induced either by a high fat diet or by the lethal yellow agouti (A(y)) mutation, and this protective action was due to inhibition of the increase in adipocyte number without an effect on adipocyte hypertrophy. The reduction in the number of adipocyte caused by Skp2 ablation also inhibited the development of obesity-related insulin resistance in the A(y) mutant mice, although the reduced number of beta cells and reduced level of insulin secretion in Skp2-deficient mice resulted in glucose intolerance. Our observations thus indicate that Skp2 controls adipocyte proliferation during the development of obesity.


Assuntos
Adipócitos/citologia , Obesidade/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação para Baixo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , RNA Mensageiro/metabolismo , Regulação para Cima
14.
Cell Metab ; 3(4): 267-75, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16581004

RESUMO

STAT3 regulates glucose homeostasis by suppressing the expression of gluconeogenic genes in the liver. The mechanism by which hepatic STAT3 is regulated by nutritional or hormonal status has remained unknown, however. Here, we show that an increase in the plasma insulin concentration, achieved either by glucose administration or by intravenous insulin infusion, stimulates tyrosine phosphorylation of STAT3 in the liver. This effect of insulin was mediated by the hormone's effects in the brain, and the increase in hepatic IL-6 induced by the brain-insulin action is essential for the activation of STAT3. The inhibition of hepatic glucose production and of expression of gluconeogenic genes induced by intracerebral ventricular insulin infusion was impaired in mice with liver-specific STAT3 deficiency or in mice with IL-6 deficiency. These results thus indicate that IL-6-STAT3 signaling in the liver contributes to insulin action in the brain, leading to the suppression of hepatic glucose production.


Assuntos
Encéfalo/metabolismo , Glucose/metabolismo , Insulina/fisiologia , Fígado/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Ativação Enzimática , Gluconeogênese , Glucose/farmacologia , Técnica Clamp de Glucose , Glucose-6-Fosfatase/fisiologia , Homeostase , Insulina/administração & dosagem , Insulina/sangue , Insulina/farmacologia , Resistência à Insulina , Interleucina-6/análise , Interleucina-6/fisiologia , Células de Kupffer/química , Células de Kupffer/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoenolpiruvato Carboxilase/fisiologia , Fosforilação , Receptor de Insulina/fisiologia , Transdução de Sinais
15.
Proc Natl Acad Sci U S A ; 102(30): 10610-5, 2005 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16030142

RESUMO

There is a rapid global rise in obesity, and the link between obesity and diabetes remains somewhat obscure. We identified an adipocytokine, designated as visceral adipose tissue-derived serpin (vaspin), which is a member of serine protease inhibitor family. Vaspin cDNA was isolated by from visceral white adipose tissues (WATs) of Otsuka Long-Evans Tokushima fatty (OLETF) rat, an animal model of abdominal obesity with type 2 diabetes. Rat, mouse, and human vaspins are made up of 392, 394, and 395 amino acids, respectively; exhibit approximately 40% homology with alpha1-antitrypsin; and are related to serine protease inhibitor family. Vaspin was barely detectable in rats at 6 wk and was highly expressed in adipocytes of visceral WATs at 30 wk, the age when obesity, body weight, and insulin levels peak in OLETF rats. The tissue expression of vaspin and its serum levels decrease with worsening of diabetes and body weight loss at 50 wk. The expression and serum levels were normalized with the treatment of insulin or insulin-sensitizing agent, pioglitazone, in OLETF rats. Administration of vaspin to obese CRL:CD-1 (ICR) (ICR) mice fed with high-fat high-sucrose chow improved glucose tolerance and insulin sensitivity reflected by normalized serum glucose levels. It also led to the reversal of altered expression of genes relevant to insulin resistance, e.g., leptin, resistin, TNFalpha, glucose transporter-4, and adiponectin. In DNA chip analyses, vaspin treatment resulted in the reversal of expression in approximately 50% of the high-fat high-sucrose-induced genes in WATs. These findings indicate that vaspin exerts an insulin-sensitizing effect targeted toward WATs in states of obesity.


Assuntos
Adipócitos/metabolismo , Diabetes Mellitus Tipo 1/genética , Regulação da Expressão Gênica , Obesidade/genética , Serpinas/genética , Fatores Etários , Sequência de Aminoácidos , Animais , Sequência de Bases , Glicemia , Northern Blotting , Peso Corporal , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Insulina/metabolismo , Resistência à Insulina/genética , Camundongos , Dados de Sequência Molecular , Obesidade/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Pioglitazona , Ratos , Ratos Endogâmicos OLETF , Análise de Sequência de DNA , Homologia de Sequência , Serpinas/metabolismo , Tiazolidinedionas
16.
Biochem Biophys Res Commun ; 327(3): 920-6, 2005 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-15649433

RESUMO

Regulation of hepatic gene expression is important for energy homeostasis. We now show that hepatic expression of the gene for the transcription factor Kruppel-like factor 15 (KLF15) is increased by food deprivation and reduced by feeding in mice. Expression of the KLF15 gene in mouse liver was also down-regulated by a euglycemic-hyperinsulinemic clamp and was increased by inhibition of phosphatidylinositol 3-kinase. In cultured rat hepatocytes, KLF15 gene expression was induced by dexamethasone and a non-hydrolyzing analog of cAMP, and this effect was inhibited by insulin in a manner dependent on phosphatidylinositol 3-kinase signaling. Forced expression of KLF15 in cultured hepatocytes increased both the expression and the promoter activity of the gene for phosphoenolpyruvate carboxykinase (PEPCK). These results suggest that insulin and its counteracting hormones regulate the hepatic expression of KLF15, and that this transcription factor contributes to the regulation of PEPCK gene expression in the liver.


Assuntos
Regulação Enzimológica da Expressão Gênica , Fígado/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas de Ligação a DNA , Dexametasona/farmacologia , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Técnica Clamp de Glucose/métodos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Fatores de Transcrição Kruppel-Like , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Fatores de Transcrição/química
17.
Science ; 307(5708): 426-30, 2005 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-15604363

RESUMO

Fat tissue produces a variety of secreted proteins (adipocytokines) with important roles in metabolism. We isolated a newly identified adipocytokine, visfatin, that is highly enriched in the visceral fat of both humans and mice and whose expression level in plasma increases during the development of obesity. Visfatin corresponds to a protein identified previously as pre-B cell colony-enhancing factor (PBEF), a 52-kilodalton cytokine expressed in lymphocytes. Visfatin exerted insulin-mimetic effects in cultured cells and lowered plasma glucose levels in mice. Mice heterozygous for a targeted mutation in the visfatin gene had modestly higher levels of plasma glucose relative to wild-type littermates. Surprisingly, visfatin binds to and activates the insulin receptor. Further study of visfatin's physiological role may lead to new insights into glucose homeostasis and/or new therapies for metabolic disorders such as diabetes.


Assuntos
Tecido Adiposo/metabolismo , Citocinas/metabolismo , Insulina/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Sítios de Ligação , Glicemia/análise , Linhagem Celular , Células Cultivadas , Citocinas/sangue , Citocinas/genética , Citocinas/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Relação Dose-Resposta a Droga , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Humanos , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Mimetismo Molecular , Células Musculares/metabolismo , Nicotinamida Fosforribosiltransferase , Fosforilação , Receptor de Insulina/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Tela Subcutânea , Vísceras
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...