Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 7(3): 774-84, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24746732

RESUMO

Spinal and bulbar muscular atrophy (SBMA) is caused by the polyglutamine androgen receptor (polyQ-AR), a protein expressed by both lower motor neurons and skeletal muscle. Although viewed as a motor neuronopathy, data from patients and mouse models suggest that muscle contributes to disease pathogenesis. Here, we tested this hypothesis using AR113Q knockin and human bacterial artificial chromosome/clone (BAC) transgenic mice that express the full-length polyQ-AR and display androgen-dependent weakness, muscle atrophy, and early death. We developed antisense oligonucleotides that suppressed AR gene expression in the periphery but not the CNS after subcutaneous administration. Suppression of polyQ-AR in the periphery rescued deficits in muscle weight, fiber size, and grip strength, reversed changes in muscle gene expression, and extended the lifespan of mutant males. We conclude that polyQ-AR expression in the periphery is an important contributor to pathology in SBMA mice and that peripheral administration of therapeutics should be explored for SBMA patients.


Assuntos
Transtornos Musculares Atróficos/metabolismo , Receptores Androgênicos/genética , Animais , Peso Corporal , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Força da Mão/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Transtornos Musculares Atróficos/mortalidade , Transtornos Musculares Atróficos/patologia , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Taxa de Sobrevida , Testosterona/sangue , Transcriptoma
2.
PLoS One ; 8(7): e66923, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922646

RESUMO

Obesity is a primary risk factor for multiple metabolic disorders. Many drugs for the treatment of obesity, which mainly act through CNS as appetite suppressants, have failed during development or been removed from the market due to unacceptable adverse effects. Thus, there are very few efficacious drugs available and remains a great unmet medical need for anti-obesity drugs that increase energy expenditure by acting on peripheral tissues without severe side effects. Here, we report a novel approach involving antisense inhibition of fibroblast growth factor receptor 4 (FGFR4) in peripheral tissues. Treatment of diet-induce obese (DIO) mice with FGFR4 antisense oligonucleotides (ASO) specifically reduced liver FGFR4 expression that not only resulted in decrease in body weight (BW) and adiposity in free-feeding conditions, but also lowered BW and adiposity under caloric restriction. In addition, combination treatment with FGFR4 ASO and rimonabant showed additive reduction in BW and adiposity. FGFR4 ASO treatment increased basal metabolic rate during free-feeding conditions and, more importantly, prevented adaptive decreases of metabolic rate induced by caloric restriction. The treatment increased fatty acid oxidation while decreased lipogenesis in both liver and fat. Mechanistic studies indicated that anti-obesity effect of FGFR4 ASO was mediated at least in part through an induction of plasma FGF15 level resulted from reduction of hepatic FGFR4 expression. The anti-obesity effect was accompanied by improvement in plasma glycemia, whole body insulin sensitivity, plasma lipid levels and liver steatosis. Therefore, FGFR4 could be a potential novel target and antisense reduction of hepatic FGFR4 expression could be an efficacious therapy as an adjunct to diet restriction or to an appetite suppressant for the treatment of obesity and related metabolic disorders.


Assuntos
Adiposidade/efeitos dos fármacos , Metabolismo Basal/efeitos dos fármacos , Obesidade/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Peso Corporal/efeitos dos fármacos , Restrição Calórica , Dieta , Quimioterapia Combinada , Ácidos Graxos/metabolismo , Fígado Gorduroso/sangue , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Comportamento Alimentar/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Insulina/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/sangue , Obesidade/tratamento farmacológico , Obesidade/genética , Oligonucleotídeos Antissenso/uso terapêutico , Oxirredução/efeitos dos fármacos , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Rimonabanto
3.
Cell Metab ; 10(6): 499-506, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19945407

RESUMO

In patients with poorly controlled type 2 diabetes mellitus (T2DM), hepatic insulin resistance and increased gluconeogenesis contribute to fasting and postprandial hyperglycemia. Since cAMP response element-binding protein (CREB) is a key regulator of gluconeogenic gene expression, we hypothesized that decreasing hepatic CREB expression would reduce fasting hyperglycemia in rodent models of T2DM. In order to test this hypothesis, we used a CREB-specific antisense oligonucleotide (ASO) to knock down CREB expression in liver. CREB ASO treatment dramatically reduced fasting plasma glucose concentrations in ZDF rats, ob/ob mice, and an STZ-treated, high-fat-fed rat model of T2DM. Surprisingly, CREB ASO treatment also decreased plasma cholesterol and triglyceride concentrations, as well as hepatic triglyceride content, due to decreases in hepatic lipogenesis. These results suggest that CREB is an attractive therapeutic target for correcting both hepatic insulin resistance and dyslipidemia associated with nonalcoholic fatty liver disease (NAFLD) and T2DM.


Assuntos
Colesterol/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Diabetes Mellitus Tipo 2/metabolismo , Fígado Gorduroso/genética , Glucose/metabolismo , Resistência à Insulina/genética , Fígado/metabolismo , Fígado/fisiopatologia , Triglicerídeos/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Mellitus Tipo 2/terapia , Modelos Animais de Doenças , Dislipidemias/terapia , Fígado Gorduroso/fisiopatologia , Fígado Gorduroso/terapia , Lipogênese/fisiologia , Masculino , Camundongos , Oligonucleotídeos Antissenso , Ratos
4.
Proc Natl Acad Sci U S A ; 106(27): 11288-93, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19549853

RESUMO

Hepatic gluconeogenesis is a major contributing factor to hyperglycemia in the fasting and postprandial states in type 2 diabetes mellitus (T2DM). Because Sirtuin 1 (SirT1) induces hepatic gluconeogenesis during fasting through the induction of phosphoenolpyruvate carboxylase kinase (PEPCK), fructose-1,6-bisphosphatase (FBPase), and glucose-6-phosphatase (G6Pase) gene transcription, we hypothesized that reducing SirT1, by using an antisense oligonucleotide (ASO), would decrease fasting hyperglycemia in a rat model of T2DM. SirT1 ASO lowered both fasting glucose concentration and hepatic glucose production in the T2DM rat model. Whole body insulin sensitivity was also increased in the SirT1 ASO treated rats as reflected by a 25% increase in the glucose infusion rate required to maintain euglycemia during the hyperinsulinemic-euglycemic clamp and could entirely be attributed to increased suppression of hepatic glucose production by insulin. The reduction in basal and clamped rates of glucose production could in turn be attributed to decreased expression of PEPCK, FBPase, and G6Pase due to increased acetylation of signal transducer and activator of transcription 3 (STAT3), forkhead box O1 (FOXO1), and peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha), known substrates of SirT1. In addition to the effects on glucose metabolism, SirT1 ASO decreased plasma total cholesterol, which was attributed to increased cholesterol uptake and export from the liver. These results indicate that inhibition of hepatic SirT1 may be an attractive approach for treatment of T2DM.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamento de Genes , Glucose/biossíntese , Insulina/metabolismo , Fígado/metabolismo , Sirtuínas/deficiência , Acetilação/efeitos dos fármacos , Animais , Colesterol/sangue , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Hiperinsulinismo/sangue , Hiperinsulinismo/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Oligonucleotídeos Antissenso/farmacologia , Ratos , Ratos Sprague-Dawley , Sirtuína 1 , Sirtuínas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos
5.
Cell Metab ; 9(3): 252-64, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19254570

RESUMO

Peroxisome proliferator-activated receptor gamma coactivator-1 beta (PGC-1beta) is known to be a transcriptional coactivator for SREBP-1, the master regulator of hepatic lipogenesis. Here, we evaluated the role of PGC-1beta in the pathogenesis of fructose-induced insulin resistance by using an antisense oligonucletoide (ASO) to knockdown PGC-1beta in liver and adipose tissue. PGC-1beta ASO improved the metabolic phenotype induced by fructose feeding by reducing expression of SREBP-1 and downstream lipogenic genes in liver. PGC-1beta ASO also reversed hepatic insulin resistance induced by fructose in both basal and insulin-stimulated states. Furthermore, PGC-1beta ASO increased insulin-stimulated whole-body glucose disposal due to a threefold increase in glucose uptake in white adipose tissue. These data support an important role for PGC-1beta in the pathogenesis of fructose-induced insulin resistance and suggest that PGC-1beta inhibition may be a therapeutic target for treatment of NAFLD, hypertriglyceridemia, and insulin resistance associated with increased de novo lipogenesis.


Assuntos
Frutose/metabolismo , Resistência à Insulina/fisiologia , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Tecido Adiposo/metabolismo , Animais , Dieta , Frutose/administração & dosagem , Expressão Gênica , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Masculino , Camundongos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Fatores de Transcrição/genética
6.
Am J Physiol Endocrinol Metab ; 295(2): E436-45, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18523126

RESUMO

To investigate the role of JNK1 in metabolism, male ob/ob and diet-induced obese mice were treated with a JNK1-specific antisense oligonucleotide (ASO) or control ASO at 25 mg/kg or saline twice/wk for 6 and 7 wk, respectively. JNK1 ASO reduced JNK1 mRNA and activity by 65-95% in liver and fat tissues in both models. Compared with controls, treatment with JNK1 ASO did not change food intake but lowered body weight, fat pad weight, and whole body fat content. The treatment increased metabolic rate. In addition, the treatment markedly reduced plasma cholesterol levels and improved liver steatosis and insulin sensitivity. These positive observations were accompanied by the following changes: 1) increased mRNA levels of AR-beta(3) and UCP1 by >60% in BAT, 2) reduced mRNA levels of ACC1, ACC2, FAS, SCD1, DGAT1, DGAT2, and RBP4 by 30-60% in WAT, and 3) reduced mRNA levels of ACC1, FAS, G-6-Pase, and PKCepsilon by 40-70% and increased levels of UCP2 and PPARalpha by more than twofold in liver. JNK1 ASO-treated mice demonstrated reduced levels of pIRS-1 Ser(302) and pIRS-1 Ser(307) and increased levels of pAkt Ser(473) in liver and fat in response to insulin. JNK1 ASO-transfected mouse hepatocytes showed decreased rates of de novo sterol and fatty acid synthesis and an increased rate of fatty acid oxidation. These results indicate that inhibition of JNK1 expression in major peripheral tissues can improve adiposity via increasing fuel combustion and decreasing lipogenesis and could therefore provide clinical benefit for the treatment of obesity and related metabolic abnormalities.


Assuntos
Proteína Quinase 8 Ativada por Mitógeno/biossíntese , Obesidade/enzimologia , Oligonucleotídeos Antissenso/genética , Adiposidade/efeitos dos fármacos , Animais , Glicemia/metabolismo , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Ingestão de Alimentos/efeitos dos fármacos , Fígado Gorduroso/metabolismo , Insulina/sangue , Insulina/metabolismo , Resistência à Insulina/fisiologia , Lipoproteínas/sangue , Lipoproteínas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Obesidade/sangue , Obesidade/terapia , Oligonucleotídeos Antissenso/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Am J Physiol Endocrinol Metab ; 294(3): E530-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18198353

RESUMO

To investigate the possible role of eukaryotic initiation factor 4E-binding protein-2 (4E-BP2) in metabolism and energy homeostasis, high-fat diet-induced obese mice were treated with a 4E-BP2-specific antisense oligonucleotide (ASO) or a control 4E-BP2 ASO at a dose of 25 mg/kg body wt or with saline twice a week for 6 wk. 4E-BP2 ASO treatment reduced 4E-BP2 levels by >75% in liver and white (WAT) and brown adipose (BAT) tissues. Treatment did not change food intake but lowered body weight by approximately 7% and body fat content by approximately 18%. Treatment decreased liver triglyceride (TG) content by >50%, normalized plasma glucose and insulin levels, and reduced glucose excursion during glucose tolerance test. 4E-BP2 ASO-treated mice showed >8.5% increase in metabolic rate, >40% increase in UCP1 levels in BAT, >45% increase in beta(3)-adrenoceptor mRNA, and 40-55% decrease in mitochondrial dicarboxylate carrier, fatty acid synthase, and diacylglycerol acyltransferase 2 mRNA levels in WAT. 4E-BP2 ASO-transfected mouse hepatocytes showed an increased fatty acid oxidation rate and a decreased TG synthesis rate. In addition, 4E-BP2 ASO-treated mice demonstrated approximately 60 and 29% decreases in hepatic glucose-6-phosphatase and phosphoenolpyruvate carboxykinase mRNA, respectively, implying decreased hepatic glucose output. Furthermore, increased phosphorylation of Akt(Ser473) in both liver and fat of 4E-BP2 ASO-treated mice and increased GLUT4 levels in plasma membrane in WAT of the ASO-treated mice were observed, indicating enhanced insulin signaling and increased glucose uptake as a consequence of reduced 4E-BP2 expression. These data demonstrate for the first time that peripheral 4E-BP2 plays an important role in metabolism and energy homeostasis.


Assuntos
Adiposidade/fisiologia , Fatores de Iniciação em Eucariotos/fisiologia , Insulina/farmacologia , Obesidade/fisiopatologia , Oligonucleotídeos Antissenso/farmacologia , Tecido Adiposo/química , Animais , Glicemia/análise , Gorduras na Dieta/administração & dosagem , Metabolismo Energético , Fatores de Iniciação em Eucariotos/antagonistas & inibidores , Fatores de Iniciação em Eucariotos/genética , Fígado Gorduroso/etiologia , Fígado Gorduroso/terapia , Expressão Gênica/efeitos dos fármacos , Homeostase , Fígado/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/genética , RNA Mensageiro/análise , Transfecção , Redução de Peso
8.
Hepatology ; 47(2): 625-35, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18000880

RESUMO

UNLABELLED: Retinyl ester (RE) stores decrease during hepatic stellate cell (HSC) activation and liver fibrosis. Although retinol esterification is mostly catalyzed by lecithin:retinol acyltransferase (LRAT), diacylglycerol acyltransferase (DGAT)1 also does this. In previous reports, LRAT(-/-) mice had reduced hepatic RE but neither excessive HSC activation nor liver fibrosis, and DGAT1(-/-) mice had increased liver levels of RE and retinol. We sought to clarify the role of DGAT1 in liver fibrosis. Expression of DGAT1/2 was compared by real time PCR in freshly isolated, primary mouse HSCs and hepatocytes. To induce nonalcoholic steatohepatitis (NASH) and liver fibrosis, adult male db/db mice were fed methionine choline-deficient (MCD) diets. Half were treated with DGAT1 antisense oligonucleotide (ASO); the rest were injected with saline. Results were compared with chow-fed controls. Inhibition of DGAT1 in liver had no effect on hepatic triglyceride content or liver necroinflammation but reduced HSC activation and liver fibrosis in mice with NASH. To evaluate the role of DGAT1 in HSC activation, HSC were isolated from healthy rats treated with DGAT1 ASO or saline. DGAT1 was expressed at relatively high levels in HSCs. HSC isolated from DGAT1 ASO-treated rats had reduced DGAT1 expression and increased messenger RNA (mRNA) levels of LRAT and cellular retinol binding protein-1. During culture, they retained more vitamin A, had repressed collagen a2 (I) transcriptional activity, and expressed less collagen a1 (I) and a2 (I) mRNA. CONCLUSION: DGAT1 may be a therapeutic target in NASH because inhibiting DGAT1 favorably altered. HSC retinoid homeostasis and inhibited hepatic fibrosis in mice with NASH.


Assuntos
Diacilglicerol O-Aciltransferase/genética , Fígado Gorduroso/prevenção & controle , Hepatite/prevenção & controle , Cirrose Hepática/prevenção & controle , Oligonucleotídeos Antissenso/uso terapêutico , Ração Animal , Animais , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Biol Chem ; 282(31): 22678-88, 2007 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-17526931

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a major contributing factor to hepatic insulin resistance in type 2 diabetes. Diacylglycerol acyltransferase (Dgat), of which there are two isoforms (Dgat1 and Dgat2), catalyzes the final step in triglyceride synthesis. We evaluated the metabolic impact of pharmacological reduction of DGAT1 and -2 expression in liver and fat using antisense oligonucleotides (ASOs) in rats with diet-induced NAFLD. Dgat1 and Dgat2 ASO treatment selectively reduced DGAT1 and DGAT2 mRNA levels in liver and fat, but only Dgat2 ASO treatment significantly reduced hepatic lipids (diacylglycerol and triglyceride but not long chain acyl CoAs) and improved hepatic insulin sensitivity. Because Dgat catalyzes triglyceride synthesis from diacylglycerol, and because we have hypothesized that diacylglycerol accumulation triggers fat-induced hepatic insulin resistance through protein kinase C epsilon activation, we next sought to understand the paradoxical reduction in diacylglycerol in Dgat2 ASO-treated rats. Within 3 days of starting Dgat2 ASO therapy in high fat-fed rats, plasma fatty acids increased, whereas hepatic lysophosphatidic acid and diacylglycerol levels were similar to those of control rats. These changes were associated with reduced expression of lipogenic genes (SREBP1c, ACC1, SCD1, and mtGPAT) and increased expression of oxidative/thermogenic genes (CPT1 and UCP2). Taken together, these data suggest that knocking down Dgat2 protects against fat-induced hepatic insulin resistance by paradoxically lowering hepatic diacylglycerol content and protein kinase C epsilon activation through decreased SREBP1c-mediated lipogenesis and increased hepatic fatty acid oxidation.


Assuntos
Diacilglicerol O-Aciltransferase/fisiologia , Fígado Gorduroso/patologia , Resistência à Insulina , Oligonucleotídeos Antissenso/química , Animais , Diacilglicerol O-Aciltransferase/metabolismo , Dieta , Ácidos Graxos/metabolismo , Fígado Gorduroso/terapia , Hepatócitos/metabolismo , Fígado/metabolismo , Masculino , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Triglicerídeos/metabolismo
10.
Hepatology ; 45(6): 1366-74, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17476695

RESUMO

UNLABELLED: In the early stages of nonalcoholic fatty liver disease (NAFLD), triglycerides accumulate in hepatocytes. Diacylglycerol acyltransferase 2 (DGAT2) catalyzes the final step in hepatocyte triglyceride biosynthesis. DGAT2 antisense oligonucleotide (ASO) treatment improved hepatic steatosis dramatically in a previous study of obese mice. According to the 2-hit hypothesis for progression of NAFLD, hepatic steatosis is a risk factor for nonalcoholic steatohepatitis (NASH) and fibrosis. To evaluate this hypothesis, we inhibited DGAT2 in a mouse model of NASH induced by a diet deficient in methionine and choline (MCD). Six-week-old genetically obese and diabetic male db/db mice were fed either the control or the MCD diet for 4 or 8 weeks. The MCD diet group was treated with either 25 mg/kg DGAT2 ASO or saline intraperitoneally twice weekly. Hepatic steatosis, injury, fibrosis, markers of lipid peroxidation/oxidant stress, and systemic insulin sensitivity were evaluated. Hepatic steatosis, necroinflammation, and fibrosis were increased in saline-treated MCD diet-fed mice compared to controls. Treating MCD diet-fed mice with DGAT2 ASO for 4 and 8 weeks decreased hepatic steatosis, but increased hepatic free fatty acids, cytochrome P4502E1, markers of lipid peroxidation/oxidant stress, lobular necroinflammation, and fibrosis. Progression of liver damage occurred despite reduced hepatic expression of tumor necrosis factor alpha, increased serum adiponectin, and striking improvement in systemic insulin sensitivity. CONCLUSION: Results from this mouse model would suggest accumulation of triglycerides may be a protective mechanism to prevent progressive liver damage in NAFLD.


Assuntos
Fígado Gorduroso/metabolismo , Cirrose Hepática/metabolismo , Obesidade/metabolismo , Triglicerídeos/biossíntese , Triglicerídeos/metabolismo , Adiponectina/sangue , Ração Animal , Animais , Glicemia/metabolismo , Colina/farmacologia , Citocromo P-450 CYP2E1/metabolismo , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Fígado Gorduroso/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Insulina/sangue , Peroxidação de Lipídeos/fisiologia , Cirrose Hepática/patologia , Masculino , Metionina/farmacologia , Camundongos , Camundongos Mutantes , Obesidade/complicações , Oligorribonucleotídeos Antissenso/farmacologia , Estresse Oxidativo/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
11.
J Biol Chem ; 282(19): 14291-9, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17353188

RESUMO

To investigate the role of low molecular weight protein-tyrosine phosphatase (LMW-PTP) in glucose metabolism and insulin action, a specific antisense oligonucleotide (ASO) was used to reduce its expression both in vitro and in vivo. Reduction of LMW-PTP expression with the ASO in cultured mouse hepatocytes and in liver and fat tissues of diet-induced obese (DIO) mice and ob/ob mice led to increased phosphorylation and activity of key insulin signaling intermediates, including insulin receptor-beta subunit, phosphatidylinositol 3-kinase, and Akt in response to insulin stimulation. The ASO-treated DIO and ob/ob animals showed improved insulin sensitivity, which was reflected by a lowering of both plasma insulin and glucose levels and improved glucose and insulin tolerance in DIO mice. The treatment did not decrease body weight or increase metabolic rate. These data demonstrate that LMW-PTP is a key negative regulator of insulin action and a potential novel target for the treatment of insulin resistance and type 2 diabetes.


Assuntos
Hiperglicemia/metabolismo , Resistência à Insulina , Insulina/metabolismo , Isoenzimas/metabolismo , Obesidade/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Glicemia/metabolismo , Western Blotting , Peso Corporal , Teste de Tolerância a Glucose , Hepatócitos/metabolismo , Imunoprecipitação , Proteínas Substratos do Receptor de Insulina , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais
12.
Cell Metab ; 3(2): 87-98, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16459310

RESUMO

Current understanding of microRNA (miRNA) biology is limited, and antisense oligonucleotide (ASO) inhibition of miRNAs is a powerful technique for their functionalization. To uncover the role of the liver-specific miR-122 in the adult liver, we inhibited it in mice with a 2'-O-methoxyethyl phosphorothioate ASO. miR-122 inhibition in normal mice resulted in reduced plasma cholesterol levels, increased hepatic fatty-acid oxidation, and a decrease in hepatic fatty-acid and cholesterol synthesis rates. Activation of the central metabolic sensor AMPK was also increased. miR-122 inhibition in a diet-induced obesity mouse model resulted in decreased plasma cholesterol levels and a significant improvement in liver steatosis, accompanied by reductions in several lipogenic genes. These results implicate miR-122 as a key regulator of cholesterol and fatty-acid metabolism in the adult liver and suggest that miR-122 may be an attractive therapeutic target for metabolic disease.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Obesidade/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Proteínas Quinases Ativadas por AMP , Animais , Análise Química do Sangue , Northern Blotting , Western Blotting , Linhagem Celular , Colesterol/sangue , Cromatografia Líquida de Alta Pressão , Primers do DNA , Ativação Enzimática/efeitos dos fármacos , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/citologia , Camundongos , Análise em Microsséries , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Oligonucleotídeos Antissenso/genética , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Clin Invest ; 116(3): 817-24, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16485039

RESUMO

Hepatic steatosis is a core feature of the metabolic syndrome and type 2 diabetes and leads to hepatic insulin resistance. Malonyl-CoA, generated by acetyl-CoA carboxylases 1 and 2 (Acc1 and Acc2), is a key regulator of both mitochondrial fatty acid oxidation and fat synthesis. We used a diet-induced rat model of nonalcoholic fatty liver disease (NAFLD) and hepatic insulin resistance to explore the impact of suppressing Acc1, Acc2, or both Acc1 and Acc2 on hepatic lipid levels and insulin sensitivity. While suppression of Acc1 or Acc2 expression with antisense oligonucleotides (ASOs) increased fat oxidation in rat hepatocytes, suppression of both enzymes with a single ASO was significantly more effective in promoting fat oxidation. Suppression of Acc1 also inhibited lipogenesis whereas Acc2 reduction had no effect on lipogenesis. In rats with NAFLD, suppression of both enzymes with a single ASO was required to significantly reduce hepatic malonyl-CoA levels in vivo, lower hepatic lipids (long-chain acyl-CoAs, diacylglycerol, and triglycerides), and improve hepatic insulin sensitivity. Plasma ketones were significantly elevated compared with controls in the fed state but not in the fasting state, indicating that lowering Acc1 and -2 expression increases hepatic fat oxidation specifically in the fed state. These studies suggest that pharmacological inhibition of Acc1 and -2 may be a novel approach in the treatment of NAFLD and hepatic insulin resistance.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Dieta , Inibidores Enzimáticos/uso terapêutico , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/enzimologia , Resistência à Insulina/fisiologia , Oligonucleotídeos Antissenso/uso terapêutico , Acetil-CoA Carboxilase/genética , Animais , Ácidos Graxos/metabolismo , Hepatócitos/metabolismo , Ratos , Transdução de Sinais/fisiologia , Triglicerídeos/biossíntese
14.
Hepatology ; 42(2): 362-71, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16001399

RESUMO

In this study, we investigated the role of acyl-coenzyme A:diacylglycerol acyltransferase 2 (DGAT2) in glucose and lipid metabolism in obese mice by reducing its expression in liver and fat with an optimized antisense oligonucleotide (ASO). High-fat diet-induced obese (DIO) C57BL/6J mice and ob/ob mice were treated with DGAT2 ASO, control ASO, or saline. DGAT2 ASO treatment reduced DGAT2 messenger RNA (mRNA) levels by more than 75% in both liver and fat but did not change DGAT1 mRNA levels in either of these tissues, which resulted in decreased DGAT activity in liver but not in fat. DGAT2 ASO treatment did not cause significant changes in body weight, adiposity, metabolic rate, insulin sensitivity, or skin microstructure. However, DGAT2 ASO treatment caused a marked reduction in hepatic triglyceride content and improved hepatic steatosis in both models, which was consistent with a dramatic decrease in triglyceride synthesis and an increase in fatty acid oxidation observed in primary mouse hepatocytes treated with DGAT2 ASO. In addition, the treatment lowered hepatic triglyceride secretion rate and plasma triglyceride levels, and improved plasma lipoprotein profile in DIO mice. The positive effects of the DGAT2 ASO were accompanied by a reduction in the mRNA levels of several hepatic lipogenic genes, including SCD1, FAS, ACC1, ACC2, ATP-citrate lyase, glycerol kinase, and HMG-CoA reductase. In conclusion, reduction of DGAT2 expression in obese animals can reduce hepatic lipogenesis and hepatic steatosis as well as attenuate hyperlipidemia, thereby leading to an improvement in metabolic syndrome.


Assuntos
Aciltransferases/antagonistas & inibidores , Fígado Gorduroso/tratamento farmacológico , Hiperlipidemias/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Aciltransferases/genética , Animais , Glicemia/análise , Diacilglicerol O-Aciltransferase , Ácidos Graxos/metabolismo , Fígado Gorduroso/metabolismo , Hepatócitos/metabolismo , Lipídeos/biossíntese , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Oxirredução , Pele/patologia , Triglicerídeos/biossíntese
15.
Endocrinology ; 145(6): 2594-603, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14976145

RESUMO

Hormonal control of metabolic rate can be important in regulating the imbalance between energy intake and expenditure that underlies the development of obesity. In mice fed a high-fat diet, human fibroblast growth factor 19 (FGF19) increased metabolic rate [1.53 +/- 0.06 liters O(2)/h.kg(0.75) (vehicle) vs. 1.93 +/- 0.05 liters O(2)/h.kg(0.75) (FGF19); P < 0.001] and decreased respiratory quotient [0.82 +/- 0.01 (vehicle) vs. 0.80 +/- 0.01 (FGF19); P < 0.05]. In contrast to the vehicle-treated mice that gained weight (0.14 +/- 0.05 g/mouse.d), FGF19-treated mice lost weight (-0.13 +/- 0.03 g/mouse.d; P < 0.001) without a significant change in food intake. Furthermore, in addition to a reduction in weight gain, treatment with FGF19 prevented or reversed the diabetes that develops in mice made obese by genetic ablation of brown adipose tissue or genetic absence of leptin. To explore the mechanisms underlying the FGF19-mediated increase in metabolic rate, we profiled the FGF19-induced gene expression changes in the liver and brown fat. In brown adipose tissue, chronic exposure to FGF19 led to a gene expression profile that is consistent with activation of this tissue. We also found that FGF19 acutely increased liver expression of the leptin receptor (1.8-fold; P < 0.05) and decreased the expression of acetyl coenzyme A carboxylase 2 (0.6-fold; P < 0.05). The gene expression changes were consistent with the experimentally determined increase in fat oxidation and decrease in liver triglycerides. Thus, FGF19 is able to increase metabolic rate concurrently with an increase in fatty acid oxidation.


Assuntos
Diabetes Mellitus/etiologia , Diabetes Mellitus/fisiopatologia , Dieta , Fatores de Crescimento de Fibroblastos/farmacologia , Leptina/deficiência , Proteínas Recombinantes/farmacologia , Acetil-CoA Carboxilase/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/fisiopatologia , Animais , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Metabolismo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Obesidade , Receptores de Superfície Celular/metabolismo , Receptores para Leptina
16.
FASEB J ; 16(2): 155-68, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11818363

RESUMO

A survey of genes differentially expressed in the brown adipose tissue (BAT) of mice exposed to a range of environmental temperatures was carried out to identify novel genes and pathways associated with the transition of this tissue toward an amplified thermogenic state. The current report focuses on an analysis of the expression patterns of 50 metabolic genes in BAT under control conditions (22 degrees C), cold exposure (4 degrees C, 1 to 48 h), warm acclimation (33 degrees C, 3 wk), or food restriction/meal feeding (animals fed the same amount as warm mice). In general, expression of genes encoding proteins involving glucose uptake and catabolism was significantly elevated in the BAT of cold-exposed mice. The levels of mRNAs encoding proteins critical to de novo lipogenesis were also increased. Gene expression for enzymes associated with procurement and combustion of long chain fatty acids (LCFAs) was increased in the cold. Thus, a model was proposed in which coordinated activation of glucose uptake, fatty acid synthesis, and fatty acid combustion occurs as part of the adaptive thermogenic processes in BAT. Confirmation emerged from in vivo assessments of cold-induced changes in BAT 2-deoxyglucose uptake (increased 2.7-fold), BAT lipogenesis (2.8-fold higher), and incorporation of LCFA carboxyl-carbon into BAT water-soluble metabolites (elevated approximately twofold). It is proposed that temperature-sensitive regulation of distinct intracellular malonyl-CoA pool sizes plays an important role in driving this unique metabolic profile via maintenance of the lipogenic pool but diminution of the carnitine palmitoyltransferase 1 inhibitory pool under cold conditions.


Assuntos
Tecido Adiposo Marrom/metabolismo , Temperatura Baixa , Ácidos Graxos/metabolismo , Aclimatação , Acetil-CoA Carboxilase/genética , Animais , Metabolismo dos Carboidratos , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Masculino , Camundongos , Oxirredução , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Adrenérgicos beta/genética , Receptores Citoplasmáticos e Nucleares/genética , Estearoil-CoA Dessaturase/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...