Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nutr Res ; 34(6): 491-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25026916

RESUMO

Periodontal disease is related to aging, smoking habits, diabetes mellitus, and systemic inflammation. However, there remains limited evidence about causality from intervention studies. An effective diet for prevention of periodontal disease has not been well established. The current study was an intervention study examining the effects of a high-fiber, low-fat diet on periodontal disease markers in high-risk subjects. Forty-seven volunteers were interviewed for recruitment into the study. Twenty-one volunteers with a body mass index of at least 25.0 kg/m(2) or with impaired glucose tolerance were enrolled in the study. After a 2- to 3-week run-in period, subjects were provided with a test meal consisting of high fiber and low fat (30 kcal/kg of ideal body weight) 3 times a day for 8 weeks and followed by a regular diet for 24 weeks. Four hundred twenty-five teeth from 17 subjects were analyzed. Periodontal disease markers assessed as probing depth (2.28 vs 2.21 vs 2.13 mm; P < .0001), clinical attachment loss (6.11 vs 6.06 vs 5.98 mm; P < .0001), and bleeding on probing (16.2 vs 13.2 vs 14.6 %; P = .005) showed significant reductions after the test-meal period, and these improvements persisted until the follow-up period. Body weight (P < .0001), HbA1c (P < .0001), and high-sensitivity C-reactive protein (P = .038) levels showed improvement after the test-meal period; they returned to baseline levels after the follow-up period. In conclusion, treatment with a high-fiber, low-fat diet for 8 weeks effectively improved periodontal disease markers as well as metabolic profiles, at least in part, by effects other than the reduction of total energy intake.


Assuntos
Biomarcadores/sangue , Dieta com Restrição de Gorduras , Fibras na Dieta/administração & dosagem , Doenças Periodontais/sangue , Doenças Periodontais/dietoterapia , Adulto , Glicemia/metabolismo , Índice de Massa Corporal , Peso Corporal , Proteína C-Reativa/metabolismo , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Proteínas Alimentares/administração & dosagem , Ingestão de Energia , Comportamento Alimentar , Feminino , Intolerância à Glucose , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Triglicerídeos/sangue , Circunferência da Cintura
2.
Metabolism ; 63(7): 930-40, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24850465

RESUMO

OBJECTIVE: The beneficial effects of fish and n-3 polyunsaturated fatty acids (PUFAs) consumption on atherosclerosis have been reported in numerous epidemiological studies. However, to the best of our knowledge, the effects of a fish-based diet intervention on endothelial function have not been investigated. Therefore, we studied these effects in postmenopausal women with type 2 diabetes mellitus (T2DM). MATERIALS/METHODS: Twenty-three postmenopausal women with T2DM were assigned to two four-week periods of either a fish-based diet (n-3 PUFAs ≧ 3.0 g/day) or a control diet in a randomized crossover design. Endothelial function was measured with reactive hyperemia using strain-gauge plethysmography and compared with the serum levels of fatty acids and their metabolites. Endothelial function was determined with peak forearm blood flow (Peak), duration of reactive hyperemia (Duration) and flow debt repayment (FDR). RESULTS: A fish-based dietary intervention improved Peak by 63.7%, Duration by 27.9% and FDR by 70.7%, compared to the control diet. Serum n-3 PUFA levels increased after the fish-based diet period and decreased after the control diet, compared with the baseline (1.49 vs. 0.97 vs. 1.19 mmol/l, p < 0.0001). There was no correlation between serum n-3 PUFA levels and endothelial function. An increased ratio of epoxyeicosatrienoic acid/dihydroxyeicosatrienoic acid was observed after a fish-based diet intervention, possibly due to the inhibition of the activity of soluble epoxide hydrolase. CONCLUSIONS: A fish-based dietary intervention improves endothelial function in postmenopausal women with T2DM. Dissociation between the serum n-3 PUFA concentration and endothelial function suggests that the other factors may contribute to this phenomenon.


Assuntos
Envelhecimento , Aterosclerose/prevenção & controle , Diabetes Mellitus Tipo 2/dietoterapia , Angiopatias Diabéticas/prevenção & controle , Endotélio Vascular/fisiopatologia , Peixes , Alimentos Marinhos , Idoso , Animais , Aterosclerose/complicações , Estudos de Coortes , Estudos Cross-Over , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Registros de Dieta , Gorduras na Dieta/análise , Gorduras na Dieta/sangue , Gorduras na Dieta/metabolismo , Gorduras na Dieta/uso terapêutico , Eicosanoides/sangue , Eicosanoides/metabolismo , Ácidos Graxos Ômega-3/análise , Ácidos Graxos Ômega-3/sangue , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-3/uso terapêutico , Feminino , Humanos , Japão , Pessoa de Meia-Idade , Pós-Menopausa , Alimentos Marinhos/análise
3.
Obesity (Silver Spring) ; 22(4): 1070-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24123834

RESUMO

OBJECTIVE: The aim of this study was to search for novel markers of visceral adiposity. METHODS: Visceral (omental) and subcutaneous adipose tissues were obtained from 43 Japanese men. Microarray analysis using total RNA from visceral and subcutaneous adipose tissues obtained from five men with abdominal obesity and five nonobese men was first conducted. Then the expression pattern of candidate genes identified in the human study in mouse models of adiposity was examined. RESULTS: Among 30,500 genes evaluated, the mRNA expression of CCDC3 (encoding coiled-coil domain-containing protein 3) was upregulated in omental adipose tissues from abdominally obese subjects (3.07-fold) but not in subcutaneous adipose tissues (0.89-fold). Similar expression patterns were found in two distinct mouse models of obesity. In the analysis of all 43 men, CCDC3 mRNA levels in omental, but not in subcutaneous adipose tissue, were positively correlated with waist circumference and body mass index. CCDC3 was predicted to be a secretory protein, which was confirmed by western blotting, as overexpressed CCDC3 was secreted into the culture media. CONCLUSIONS: The expression of CCDC3 is specifically increased in visceral adipose tissues in abdominally obese subjects. These results suggest that CCDC3 is a potential biomarker for estimating visceral adiposity.


Assuntos
Gordura Abdominal/metabolismo , Proteínas de Transporte/metabolismo , Obesidade Abdominal/metabolismo , Omento/metabolismo , RNA Mensageiro/metabolismo , Regulação para Cima , Adipócitos/metabolismo , Adipócitos/patologia , Idoso , Animais , Biomarcadores/metabolismo , Índice de Massa Corporal , Proteínas de Transporte/genética , Estudos de Casos e Controles , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Humanos , Japão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Obesidade Abdominal/etiologia , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/genética , Gordura Subcutânea/metabolismo , Circunferência da Cintura/fisiologia
4.
PLoS One ; 8(7): e69415, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936010

RESUMO

Recent studies have proposed that n-3 polyunsaturated fatty acids (n-3 PUFAs) have direct antioxidant and anti-inflammatory effects in vascular tissue, explaining their cardioprotective effects. However, the molecular mechanisms are not yet fully understood. We tested whether n-3 PUFAs showed antioxidant activity through the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), a master transcriptional factor for antioxidant genes. C57BL/6 or Nrf2(-/-) mice were fed a fish-oil diet for 3 weeks. Fish-oil diet significantly increased the expression of heme oxygenase-1 (HO-1), and endothelium-dependent vasodilation in the aorta of C57BL/6 mice, but not in the Nrf2(-/-) mice. Furthermore, we observed that 4-hydroxy hexenal (4-HHE), an end-product of n-3 PUFA peroxidation, was significantly increased in the aorta of C57BL/6 mice, accompanied by intra-aortic predominant increase in docosahexaenoic acid (DHA) rather than that in eicosapentaenoic acid (EPA). Human umbilical vein endothelial cells were incubated with DHA or EPA. We found that DHA, but not EPA, markedly increased intracellular 4-HHE, and nuclear expression and DNA binding of Nrf2. Both DHA and 4-HHE also increased the expressions of Nrf2 target genes including HO-1, and the siRNA of Nrf2 abolished these effects. Furthermore, DHA prevented oxidant-induced cellular damage or reactive oxygen species production, and these effects were disappeared by an HO-1 inhibitor or the siRNA of Nrf2. Thus, we found protective effects of DHA through Nrf2 activation in vascular tissue, accompanied by intra-vascular increases in 4-HHE, which may explain the mechanism of the cardioprotective effects of DHA.


Assuntos
Aldeídos/farmacologia , Citoproteção/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/química , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aldeídos/metabolismo , Animais , Antioxidantes/farmacologia , Aorta/efeitos dos fármacos , Aorta/fisiologia , Peso Corporal/efeitos dos fármacos , Dano ao DNA , Dieta , Ácido Eicosapentaenoico/química , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1 , Vasodilatação/efeitos dos fármacos
5.
Am J Physiol Endocrinol Metab ; 305(2): E293-304, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23715726

RESUMO

Nonalcoholic fatty liver disease is the most frequent liver disease. Ezetimibe, an inhibitor of intestinal cholesterol absorption, has been reported to ameliorate hepatic steatosis in human and animal models. To explore how ezetimibe reduces hepatic steatosis, we investigated the effects of ezetimibe on the expression of lipogenic enzymes and intestinal lipid metabolism in mice fed a high-fat or a high-fructose diet. CBA/JN mice were fed a high-fat diet or a high-fructose diet for 8 wk with or without ezetimibe. High-fat diet induced hepatic steatosis accompanied by hyperinsulinemia. Treatment with ezetimibe reduced hepatic steatosis, insulin levels, and glucose production from pyruvate in mice fed the high-fat diet, suggesting a reduction of insulin resistance in the liver. In the intestinal analysis, ezetimibe reduced the expression of fatty acid transfer protein-4 and apoB-48 in mice fed the high-fat diet. However, treatment with ezetimibe did not prevent hepatic steatosis, hyperinsulinemia, and intestinal apoB-48 expression in mice fed the high-fructose diet. Ezetimibe decreased liver X receptor-α binding to the sterol regulatory element-binding protein-1c promoter but not expression of carbohydrate response element-binding protein and fatty acid synthase in mice fed the high-fructose diet, suggesting that ezetimibe did not reduce hepatic lipogenesis induced by the high-fructose diet. Elevation of hepatic and intestinal lipogenesis in mice fed a high-fructose diet may partly explain the differences in the effect of ezetimibe.


Assuntos
Anticolesterolemiantes/uso terapêutico , Azetidinas/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Dieta , Fígado Gorduroso/prevenção & controle , Frutose/efeitos adversos , Animais , Apolipoproteínas B/metabolismo , Compostos Azo , Western Blotting , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Ezetimiba , Fígado Gorduroso/etiologia , Teste de Tolerância a Glucose , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Ácido Pirúvico/metabolismo , RNA/biossíntese , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real
6.
Biochem Biophys Res Commun ; 430(1): 225-30, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23131562

RESUMO

Oxidative stress is produced in adipose tissue of obese subjects and has been associated with obesity-related disorders. Recent studies have shown that omega-3 polyunsaturated fatty acid (ω3-PUFA) has beneficial effects in preventing atherosclerotic diseases and insulin resistance in adipose tissue. However, the role of ω3-PUFA on adipocytes has not been elucidated. In this study, 3T3-L1 adipocytes were treated with ω3-PUFA and its metabolites, eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), or 4-hydroxy hexenal (4-HHE). ω3-PUFA and its metabolites dose-dependently increased mRNA and protein levels of the anti-oxidative enzyme, heme oxygenase-1 (HO-1); whereas no changes in the well-known anti-oxidant molecules, superoxide dismutase, catalase, and glutathione peroxidase, were observed. Knockdown of nuclear factor erythroid 2-related factor 2 (Nrf-2) significantly reduced EPA, DHA or 4-HHE-induced HO-1 mRNA and protein expression. Also, pretreatment with ω3-PUFA prevented H(2)O(2)-induced cytotoxicity in a HO-1 dependent manner. In conclusion, treatment with EPA and DHA induced HO-1 through the activation of Nrf-2 and prevented oxidative stress in 3T3-L1 adipocytes. This anti-oxidant defense may be of high therapeutic value for clinical conditions associated with systemic oxidative stress.


Assuntos
Adipócitos/efeitos dos fármacos , Antioxidantes/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Heme Oxigenase-1/metabolismo , Proteínas de Membrana/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Heme Oxigenase-1/biossíntese , Proteínas de Membrana/biossíntese , Camundongos , Fator 2 Relacionado a NF-E2/biossíntese , Água/farmacologia
7.
Am J Physiol Endocrinol Metab ; 303(12): E1419-27, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23047984

RESUMO

MicroRNAs (miRNAs) are important posttranscriptional regulators of various biological pathways. In this study, we focused on the role of miRNAs during mitochondrial biogenesis in skeletal muscle. The expression of miR-494 was markedly decreased in murine myoblast C2C12 cells during myogenic differentiation, accompanied by an increase in mtDNA. Furthermore, the expression of predicted target genes for miR-494, including mitochondrial transcription factor A (mtTFA) and Forkhead box j3 (Foxj3), was posttranscriptionally increased during myogenic differentiation. Knockdown of miR-494 resulted in increased mitochondrial content and upregulation of mtTFA and Foxj3 at the protein level. A 3'-untranslated region reporter assay revealed that miR-494 knockdown directly upregulated the luciferase activity of mtTFA and Foxj3. All of these observations were reversed by overexpression of miR-494. Furthermore, the miR-494 content significantly decreased after endurance exercise in C57BL/6J mice, accompanied by an increase in expression of mtTFA and Foxj3 proteins. These results suggest that miR-494 regulates mitochondrial biogenesis by downregulating mtTFA and Foxj3 during myocyte differentiation and skeletal muscle adaptation to physical exercise.


Assuntos
Proteínas de Ligação a DNA/metabolismo , MicroRNAs/metabolismo , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais/metabolismo , Renovação Mitocondrial , Músculo Esquelético/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Fatores de Transcrição Forkhead , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas Mitocondriais/genética , Atividade Motora , Mioblastos/citologia , Mioblastos/metabolismo , Oligorribonucleotídeos Antissenso/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética , Regulação para Cima
9.
Biochem Biophys Res Commun ; 417(1): 352-7, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22155234

RESUMO

Autophagy is an essential process for both the maintenance and the survival of cells, with homeostatic low levels of autophagy being critical for intracellular organelles and proteins. In insulin resistant adipocytes, various dysfunctional/damaged molecules, organelles, proteins, and end-products accumulate. However, the role of autophagy (in particular, whether autophagy is activated or not) is poorly understood. In this study we found that in adipose tissue of insulin resistant mice and hypertrophic 3T3-L1 adipocytes autophagy was suppressed. Also in hypertrophic adipocytes, autophagy-related gene expression, such as LAMP1, LAMP2, and Atg5 was reduced, whereas gene expression in the inflammatory-related genes, such as MCP-1, IL-6, and IL-1ß was increased. To find out whether suppressed autophagy was linked to inflammation we used the autophagy inhibitor, 3-methyladenine, to inhibit autophagy. Our results suggest that such inhibition leads to an increase in inflammatory gene expression and causes endoplasmic reticulum (ER) stress (which can be attenuated by treatment with the ER stress inhibitor, Tauroursodeoxycholic Acid). Conversely, the levels of inflammatory gene expression were reduced by the activation of autophagy or by the inhibition of ER stress. The results indicate that the suppression of autophagy increases inflammatory responses via ER stress, and also defines a novel role of autophagy as an important regulator of adipocyte inflammation in systemic insulin resistance.


Assuntos
Adipócitos/patologia , Autofagia , Estresse do Retículo Endoplasmático , Inflamação/patologia , Células 3T3-L1 , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Inflamação/genética , Resistência à Insulina , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética
10.
Int J Mol Med ; 28(1): 95-100, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21424113

RESUMO

Abnormal secretion of adipocytokines promotes atherosclerosis, diabetes and insulin resistance, and is mainly induced by adipocyte hypertrophy. Recently, the circulating adipocytokine concentrations were reported to change in the postprandial period, as the levels of TNFα, IL-6 IL-8 and MCP-1 increased after a meal, whereas that of adiponectin decreased. These data suggest that prandial modulation of cytokines may be involved in the pathogenesis of atherosclerosis in type 2 diabetes. However, the regulatory mechanism of such change is still unclear. In the present study, we identified this mechanism with a special focus on the functions of protein kinase C (PKC) and of the transcription factor AP-2ß, both of which are associated with the pathophysiology of adipocytokine regulation. PKCµ was highly phosphorylated in the re-feeding condition compared to the fasting condition in mouse adipose tissue, while other PKC isoforms remained unchanged. Furthermore, overexpression of PKCµ in 3T3-L1 adipocytes, but not other PKC isoforms, positively regulated the mRNA expression and promoter activity of MCP-1 and IL-6, and negatively regulated those of adiponectin. AP-2ß had similar effects on the expression and promoter activity of these adipocytokines. Interestingly, overexpression of PKCµ enhanced the stimulatory and inhibitory effects of AP-2ß on the expression of these adipocytokines. Finally, PKCµ could not activate a mutant MCP-1 promoter lacking the AP-2ß binding domain. Our results suggest that postprandial activation of PKCµ plays a role in disordered postprandial adipocytokine expression through AP-2ß.


Assuntos
Adipocinas/metabolismo , Período Pós-Prandial , Proteína Quinase C/metabolismo , Fator de Transcrição AP-2/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adipocinas/genética , Adiponectina/genética , Adiponectina/metabolismo , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase C/genética , Fator de Transcrição AP-2/genética , Ativação Transcricional
11.
Diabetes Res Clin Pract ; 92(2): 174-80, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21288590

RESUMO

AIM: The aim of our study was to investigate whether serum levels of soluble tumor necrosis factor α receptor (sTNFR) 1 and 2 are markers for renal dysfunction in type 2 diabetic patients without overt proteinuria. METHODS: Japanese type 2 diabetic patients without overt proteinuria (n = 168) enrolled in the prospective observational follow-up study in 2001 were retrospectively analyzed. At baseline, the serum levels of sTNFR1 and sTNFR2 were measured by sandwich ELISA. The associations between these markers and change in estimated glomerular filtration rate (eGFR) after 5 years were evaluated. RESULTS: The levels of sTNFR1 and sTNFR2 closely correlated. At baseline, sTNFR1 and sTNFR2 associated inversely with eGFR. After 5 years, patients with high level of both sTNFR1 and sTNFR2 showed a greater decline in eGFR (-13.8 ± 15.5% versus -8.5 ± 11.8%, P = 0.027) and a 4-fold higher risk for a GFR decline of ≥ 25% than those with high level of only one receptor or low level of both receptors. These associations were enhanced in diabetic women. CONCLUSIONS: The higher levels of sTNFR1 and sTNFR2 were associated with a greater decline in eGFR in type 2 diabetic patients without proteinuria, especially in diabetic women.


Assuntos
Proteinúria/patologia , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Fator de Necrose Tumoral alfa/sangue , Idoso , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Mellitus Tipo 2/urina , Ensaio de Imunoadsorção Enzimática , Feminino , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Fatores Sexuais
12.
Biochem Biophys Res Commun ; 392(4): 526-32, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20097161

RESUMO

Down-regulation of insulin receptor substrate-1 (IRS-1) expression could modify the ability of IRS-1 to fulfill its functions. It has been proposed that the phosphorylation of IRS-1 on serine residues could promote its degradation. However, few studies have investigated the transcriptional regulation of IRS-1 in the pathogenesis of insulin resistance. Genotyping for genome-wide single nucleotide polymorphisms revealed that the transcription factor activating enhancer-binding protein-2beta (AP-2beta) is a novel candidate gene for conferring susceptibility to obesity and type 2 diabetes. AP-2beta is expressed in adipose tissue and its expression is increased during the maturation of adipocytes. Overexpression of AP-2beta leads to adipocyte hypertrophy, directly inhibits adiponectin expression, and enhanced the expression of inflammatory adipokines such as IL-6 and MCP-1. In this study, we found that overexpression of AP-2beta in 3T3-L1 adipocytes impaired the promoter activity of IRS-1, and subsequently decreased mRNA and protein expression. Electrophoretic mobility shift assays showed that AP-2beta bound specifically to the IRS-1 promoter region. Furthermore, site-directed mutagenesis of the AP-2 binding site located at -362 to -351, relative to the transcription start site, markedly decreased AP-2-induced suppression of IRS-1 promoter activity, whereas other putative AP-2 binding sites did not. Our results clearly showed that AP-2beta directly decreased IRS-1 expression by binding to its promoter. Based on these findings, we speculate that the AP-2beta transcriptional factor is a unique regulator of IRS-1 and a candidate gene for insulin resistance.


Assuntos
Regulação da Expressão Gênica , Proteínas Substratos do Receptor de Insulina/genética , Fator de Transcrição AP-2/metabolismo , Células 3T3-L1 , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Técnicas de Silenciamento de Genes , Resistência à Insulina/genética , Camundongos , Camundongos Endogâmicos , Mutação , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/química , Elementos de Resposta , Fator de Transcrição AP-2/genética
13.
Am J Physiol Endocrinol Metab ; 298(3): E419-28, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19996381

RESUMO

Chronic inflammation is an important etiology underlying obesity-related disorders such as insulin resistance and type 2 diabetes, and recent findings indicate that the macrophage can be the initiating cell type responsible for this chronic inflammatory state. The mammalian silent information regulator 2 homolog SIRT1 modulates several physiological processes important for life span, and a potential role of SIRT1 in the regulation of insulin sensitivity has been shown. However, with respect to inflammation, the role of SIRT1 in regulating the proinflammatory pathway within macrophages is poorly understood. Here, we show that knockdown of SIRT1 in the mouse macrophage RAW264.7 cell line and in intraperitoneal macrophages broadly activates the JNK and IKK inflammatory pathways and increases LPS-stimulated TNFalpha secretion. Moreover, gene expression profiles reveal that SIRT1 knockdown leads to an increase in inflammatory gene expression. We also demonstrate that SIRT1 activators inhibit LPS-stimulated inflammatory pathways, as well as secretion of TNFalpha, in a SIRT1-dependent manner in RAW264.7 cells and in primary intraperitoneal macrophages. Treatment of Zucker fatty rats with a SIRT1 activator leads to greatly improved glucose tolerance, reduced hyperinsulinemia, and enhanced systemic insulin sensitivity during glucose clamp studies. These in vivo insulin-sensitizing effects were accompanied by a reduction in tissue inflammation markers and a decrease in the adipose tissue macrophage proinflammatory state, fully consistent with the in vitro effects of SIRT1 in macrophages. In conclusion, these results define a novel role for SIRT1 as an important regulator of macrophage inflammatory responses in the context of insulin resistance and raise the possibility that targeting of SIRT1 might be a useful strategy for treating the inflammatory component of metabolic diseases.


Assuntos
Inflamação/metabolismo , Resistência à Insulina/genética , Insulina/metabolismo , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Sirtuína 1/metabolismo , Animais , Células Cultivadas , Expressão Gênica , Masculino , Camundongos , Ratos , Ratos Zucker , Transdução de Sinais
14.
Obesity (Silver Spring) ; 18(7): 1277-82, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20019683

RESUMO

To determine the potential role of the transcriptional factor-activating enhancer-binding protein-2beta (TFAP2B) in the regulation of expression of adipokines, adiponectin, leptin, and interleukin-6 (IL-6) in vivo, we quantified the mRNA expression levels of these adipokines and TFAP2B in visceral (omental) and abdominal subcutaneous adipose tissues of 66 individuals with variable degree of adiposity and studied their correlations with BMI and their plasma concentrations. We found that BMI correlated negatively with plasma adiponectin levels and positively with those of leptin. Adiponection mRNA expression in subcutaneous fat correlated negatively with BMI, whereas leptin mRNA levels in the omentum correlated with plasma leptin levels and BMI. In contrast, IL-6 mRNA levels in subcutaneous and omental fat did not correlate with BMI. IL-6 mRNA levels in the omental fat correlated with plasma IL-6 levels. Whereas TFAP2B mRNA expression did not correlate with BMI, it correlated negatively with adiponectin expression in the subcutaneous adipose tissue. Furthermore, TFAP2B mRNA expression correlated negatively with leptin and positively with IL-6 expression in both subcutaneous and omental adipose tissues. These relationships are consistent with our in vitro observations and indicate that TFAP2B seems to regulate the expression of various adipokines in vivo.


Assuntos
Leptina/genética , Síndrome Metabólica/genética , Omento/fisiologia , Gordura Subcutânea/fisiologia , Fator de Transcrição AP-2/genética , Gordura Abdominal/fisiologia , Adiponectina/sangue , Adiponectina/genética , Idoso , Idoso de 80 Anos ou mais , Índice de Massa Corporal , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Humanos , Interleucina-6/sangue , Interleucina-6/genética , Leptina/sangue , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Fator de Transcrição AP-2/metabolismo
15.
J Biochem ; 146(4): 541-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19605464

RESUMO

Protein-tyrosine phosphatase 1B (PTP1B) is a major regulator of insulin sensitivity. We have described a novel action of PTP1B in the induction of sterol regulatory element-binding protein-1 (SREBP-1) gene expression through activation of protein phosphatase 2A (PP2A). PTP1B is anchored to the endoplasmic reticulum membrane via its C-terminal tail. We have previously reported that membrane localization of PTP1B is essential for PP2A activation, which is crucial for enhancing SREBP-1 gene expression in in vitro experiments. In this study, we further investigated the physiological importance of membrane localization of PTP1B in vivo. We found that transient liver-specific overexpression of wild-type PTP1B (PTP1B-WT) using adenovirus-mediated gene transfer was associated with hypertriglyceridaemia and enhanced hepatic SREBP-1 gene expression in mice. However, overexpression of the C-terminal truncated PTP1B (PTP1BDeltaCT) failed to increase hepatic SREBP-1 expression or serum triglyceride levels, despite causing insulin resistance. Our results indicate that activation of PTP1B in the liver could induce hypertriglyceridaemia and that anchoring of PTP1B to the membrane is crucial for its action.


Assuntos
Membrana Celular/enzimologia , Hipertrigliceridemia/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Animais , Membrana Celular/química , Membrana Celular/metabolismo , Retículo Endoplasmático/química , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Proteína Fosfatase 2/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/química , Ratos , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
16.
Endocr J ; 56(6): 767-72, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19506323

RESUMO

Type 1A diabetes is an autoimmune disease characterized by the destruction of insulin-producing beta-cells in the pancreas. The HLA-DR and -DQ genes are well established as being associated with increased risk for type 1 diabetes. Moreover, polymorphisms in CTLA4 have been reported to be associated with susceptibility to type 1 diabetes and autoimmune thyroid disease (AITD). In both Caucasian and Japanese populations, the lifetime risk in siblings of type 1 diabetic probands is much higher than that in general populations. However, in Japan, where the prevalence of type 1 diabetes is less than one-tenth that of most Caucasian populations, it is rare for type 1 diabetes to develop in three or more siblings within a family. Here, we report a Japanese family in which type 1 diabetes occurred in three siblings amongst four sisters. Three probands of type 1 diabetes had the same combination of HLA haplotypes, DRB1(*)0405-DQB1(*)0401/ DRB1(*)0802-DQB1(*)0302, which occurs significantly more often in type 1 diabetes patients than in control subjects in the Japanese population. With respect to the rs3087243 (+6230G>A) polymorphism of CTLA4, the first sister had type 1 diabetes and AITD and had the GG genotype, whereas the second and third sisters, who had type 1 diabetes without AITD, had the AG genotype. This is the first report of a family in which type 1A diabetes developed in three siblings. We performed genetic analysis of HLA-DR, -DQ, and CTLA4 in all family members. Even in a country where the prevalence of type 1 diabetes is low, diabetic proband siblings should be monitored for the onset of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Irmãos , Adulto , Autoanticorpos/sangue , Doenças Autoimunes/complicações , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/imunologia , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Japão
17.
Mol Cell Biol ; 29(5): 1363-74, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19103747

RESUMO

SIRT1 is a prominent member of a family of NAD(+)-dependent enzymes and affects a variety of cellular functions ranging from gene silencing, regulation of the cell cycle and apoptosis, to energy homeostasis. In mature adipocytes, SIRT1 triggers lipolysis and loss of fat content. However, the potential effects of SIRT1 on insulin signaling pathways are poorly understood. To assess this, we used RNA interference to knock down SIRT1 in 3T3-L1 adipocytes. SIRT1 depletion inhibited insulin-stimulated glucose uptake and GLUT4 translocation. This was accompanied by increased phosphorylation of JNK and serine phosphorylation of insulin receptor substrate 1 (IRS-1), along with inhibition of insulin signaling steps, such as tyrosine phosphorylation of IRS-1, and phosphorylation of Akt and ERK. In contrast, treatment of cells with specific small molecule SIRT1 activators led to an increase in glucose uptake and insulin signaling as well as a decrease in serine phosphorylation of IRS-1. Moreover, gene expression profiles showed that SIRT1 expression was inversely related to inflammatory gene expression. Finally, we show that treatment of 3T3-L1 adipocytes with a SIRT1 activator attenuated tumor necrosis factor alpha-induced insulin resistance. Taken together, these data indicate that SIRT1 is a positive regulator of insulin signaling at least partially through the anti-inflammatory actions in 3T3-L1 adipocytes.


Assuntos
Inflamação , Resistência à Insulina , Insulina/fisiologia , Sirtuínas/fisiologia , Células 3T3-L1 , Adipócitos , Animais , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Insulina/metabolismo , Camundongos , Interferência de RNA , Transdução de Sinais , Sirtuína 1
18.
Proc Natl Acad Sci U S A ; 105(18): 6614-9, 2008 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-18445652

RESUMO

Glucagon-like peptide-1 (GLP-1) is a polypeptide hormone secreted from enteroendocrine L cells and potentiates glucose-dependent insulin secretion in pancreatic beta cells. Recently the GLP-1 receptor (GLP-1 R) has been a focus for new anti-diabetic therapy with the introduction of GLP-1 analogues and DPP-IV inhibitors, and this has stimulated additional interest in the mechanisms of GLP-1 signaling. Here we identify a mechanism for GLP-1 action, showing that the scaffold protein beta-arrestin-1 mediates the effects of GLP-1 to stimulate cAMP production and insulin secretion in beta cells. Using a coimmunoprecipitation technique, we also found a physical association between the GLP-1 R and beta-arrestin-1 in cultured INS-1 pancreatic beta cells. beta-Arrestin-1 knockdown broadly attenuated GLP-1 signaling, causing decreased ERK and CREB activation and IRS-2 expression as well as reduced cAMP levels and impaired insulin secretion. However, beta-arrestin-1 knockdown did not affect GLP-1 R surface expression and ligand-induced GLP-1 R internalization/desensitization. Taken together, these studies indicate that beta-arrestin-1 plays a role in GLP-1 signaling leading to insulin secretion, defining a previously undescribed mechanism for GLP-1 action.


Assuntos
Arrestinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Endocitose/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Proteínas Substratos do Receptor de Insulina , Secreção de Insulina , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos , Receptores de Glucagon/metabolismo , beta-Arrestina 1 , beta-Arrestinas
19.
J Biol Chem ; 282(39): 28549-28556, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17664271

RESUMO

Tumor necrosis factor-alpha (TNFalpha) is a proinflammatory cytokine secreted from macrophages and adipocytes. It is well known that chronic TNFalpha exposure can lead to insulin resistance both in vitro and in vivo and that elevated blood levels of TNFalpha are observed in obese and/or diabetic individuals. TNFalpha has many acute biologic effects, mediated by a complex intracellular signaling pathway. In these studies we have identified new G-protein signaling components to this pathway in 3T3-L1 adipocytes. We found that beta-arrestin-1 is associated with TRAF2 (TNF receptor-associated factor 2), an adaptor protein of TNF receptors, and that TNFalpha acutely stimulates tyrosine phosphorylation of G alpha(q/11) with an increase in G alpha(q/11) activity. Small interfering RNA-mediated knockdown of beta-arrestin-1 inhibits TNFalpha-induced tyrosine phosphorylation of G alpha(q/11) by interruption of Src kinase activation. TNFalpha stimulates lipolysis in 3T3-L1 adipocytes, and beta-arrestin-1 knockdown blocks the effects of TNFalpha to stimulate ERK activation and glycerol release. TNFalpha also led to activation of JNK with increased expression of the proinflammatory gene, monocyte chemoattractant protein-1 and matrix metalloproteinase 3, and beta-arrestin-1 knockdown inhibited both of these effects. Taken together these results reveal novel elements of TNFalpha action; 1) the trimeric G-protein component G alpha(q/11) and the adapter protein beta-arrestin-1 can function as signaling molecules in the TNFalpha action cascade; 2) beta-arrestin-1 can couple TNFalpha stimulation to ERK activation and lipolysis; 3) beta-arrestin-1 and G alpha(q/11) can mediate TNFalpha-induced phosphatidylinositol 3-kinase activation and inflammatory gene expression.


Assuntos
Adipócitos/metabolismo , Arrestinas/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Complexos Multiproteicos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Células 3T3-L1 , Animais , Complicações do Diabetes/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glicerol/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Lipólise/efeitos dos fármacos , Camundongos , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , beta-Arrestina 1 , beta-Arrestinas
20.
Mol Cell Biol ; 27(14): 5172-83, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17515613

RESUMO

Phosphatidylinositol 3-kinase activation of Akt signaling is critical to insulin-stimulated glucose transport and GLUT4 translocation. However, the downstream signaling events following Akt activation which mediate glucose transport stimulation remain relatively unknown. Here we identify an Akt consensus phosphorylation motif in the actin-based motor protein myosin 5a and show that insulin stimulation leads to phosphorylation of myosin 5a at serine 1650. This Akt-mediated phosphorylation event enhances the ability of myosin 5a to interact with the actin cytoskeleton. Small interfering RNA-induced inhibition of myosin 5a and expression of dominant-negative myosin 5a attenuate insulin-stimulated glucose transport and GLUT4 translocation. Furthermore, knockdown of Akt2 or expression of dominant-negative Akt (DN-Akt) abolished insulin-stimulated phosphorylation of myosin 5a, inhibited myosin 5a binding to actin, and blocked insulin-stimulated glucose transport. Taken together, these data indicate that myosin 5a is a newly identified direct substrate of Akt2 and, upon insulin stimulation, phosphorylated myosin 5a facilitates anterograde movement of GLUT4 vesicles along actin to the cell surface.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Insulina/farmacologia , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vesículas Transportadoras/efeitos dos fármacos , Vesículas Transportadoras/metabolismo , Células 3T3-L1 , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Genes Dominantes , Glucose/metabolismo , Humanos , Isoenzimas/metabolismo , Camundongos , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/química , Miosina Tipo V/química , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteína Quinase C/metabolismo , Interferência de RNA , Especificidade por Substrato/efeitos dos fármacos , Proteínas rab4 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA