Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 30(4): 735-753.e4, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577934

RESUMO

Dietary sugars, fructose and glucose, promote hepatic de novo lipogenesis and modify the effects of a high-fat diet (HFD) on the development of insulin resistance. Here, we show that fructose and glucose supplementation of an HFD exert divergent effects on hepatic mitochondrial function and fatty acid oxidation. This is mediated via three different nodes of regulation, including differential effects on malonyl-CoA levels, effects on mitochondrial size/protein abundance, and acetylation of mitochondrial proteins. HFD- and HFD plus fructose-fed mice have decreased CTP1a activity, the rate-limiting enzyme of fatty acid oxidation, whereas knockdown of fructose metabolism increases CPT1a and its acylcarnitine products. Furthermore, fructose-supplemented HFD leads to increased acetylation of ACADL and CPT1a, which is associated with decreased fat metabolism. In summary, dietary fructose, but not glucose, supplementation of HFD impairs mitochondrial size, function, and protein acetylation, resulting in decreased fatty acid oxidation and development of metabolic dysregulation.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Açúcares da Dieta/efeitos adversos , Ácidos Graxos/metabolismo , Frutose/efeitos adversos , Fígado/metabolismo , Proteínas Mitocondriais , Obesidade/metabolismo , Animais , Linhagem Celular , Glucose/efeitos adversos , Lipogênese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Processamento de Proteína Pós-Traducional , Transcrição Gênica
3.
J Clin Invest ; 127(11): 4059-4074, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972537

RESUMO

Overconsumption of high-fat diet (HFD) and sugar-sweetened beverages are risk factors for developing obesity, insulin resistance, and fatty liver disease. Here we have dissected mechanisms underlying this association using mice fed either chow or HFD with or without fructose- or glucose-supplemented water. In chow-fed mice, there was no major physiological difference between fructose and glucose supplementation. On the other hand, mice on HFD supplemented with fructose developed more pronounced obesity, glucose intolerance, and hepatomegaly as compared to glucose-supplemented HFD mice, despite similar caloric intake. Fructose and glucose supplementation also had distinct effects on expression of the lipogenic transcription factors ChREBP and SREBP1c. While both sugars increased ChREBP-ß, fructose supplementation uniquely increased SREBP1c and downstream fatty acid synthesis genes, resulting in reduced liver insulin signaling. In contrast, glucose enhanced total ChREBP expression and triglyceride synthesis but was associated with improved hepatic insulin signaling. Metabolomic and RNA sequence analysis confirmed dichotomous effects of fructose and glucose supplementation on liver metabolism in spite of inducing similar hepatic lipid accumulation. Ketohexokinase, the first enzyme of fructose metabolism, was increased in fructose-fed mice and in obese humans with steatohepatitis. Knockdown of ketohexokinase in liver improved hepatic steatosis and glucose tolerance in fructose-supplemented mice. Thus, fructose is a component of dietary sugar that is distinctively associated with poor metabolic outcomes, whereas increased glucose intake may be protective.


Assuntos
Frutose/farmacologia , Glucose/farmacologia , Insulina/fisiologia , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Adolescente , Animais , Dieta Hiperlipídica/efeitos adversos , Indução Enzimática , Ácidos Graxos/biossíntese , Frutoquinases/genética , Frutoquinases/metabolismo , Intolerância à Glucose , Humanos , Resistência à Insulina , Fígado/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/enzimologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transcriptoma , Regulação para Cima
4.
Proc Natl Acad Sci U S A ; 114(34): E7111-E7120, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28784777

RESUMO

Brown and beige adipocytes convert chemical energy into heat through uncoupled respiration to defend against cold stress. Beyond thermogenesis, brown and beige fats engage other metabolic tissues via secreted factors to influence systemic energy metabolism. How the protein and long noncoding RNA (lncRNA) regulatory networks act in concert to regulate key aspects of thermogenic adipocyte biology remains largely unknown. Here we developed a genome-wide functional screen to interrogate the transcription factors and cofactors in thermogenic gene activation and identified zinc finger and BTB domain-containing 7b (Zbtb7b) as a potent driver of brown fat development and thermogenesis and cold-induced beige fat formation. Zbtb7b is required for activation of the thermogenic gene program in brown and beige adipocytes. Genetic ablation of Zbtb7b impaired cold-induced transcriptional remodeling in brown fat, rendering mice sensitive to cold temperature, and diminished browning of inguinal white fat. Proteomic analysis revealed a mechanistic link between Zbtb7b and the lncRNA regulatory pathway through which Zbtb7b recruits the brown fat lncRNA 1 (Blnc1)/heterogeneous nuclear ribonucleoprotein U (hnRNPU) ribonucleoprotein complex to activate thermogenic gene expression in adipocytes. These findings illustrate the emerging concept of a protein-lncRNA regulatory network in the control of adipose tissue biology and energy metabolism.


Assuntos
Tecido Adiposo Bege/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas de Ligação a DNA/metabolismo , Termogênese , Fatores de Transcrição/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo Bege/crescimento & desenvolvimento , Tecido Adiposo Marrom/crescimento & desenvolvimento , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Longo não Codificante , Fatores de Transcrição/genética
5.
Mol Metab ; 6(8): 863-872, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28752050

RESUMO

OBJECTIVE: Brown and white adipose tissue exerts pleiotropic effects on systemic energy metabolism in part by releasing endocrine factors. Neuregulin 4 (Nrg4) was recently identified as a brown fat-enriched secreted factor that ameliorates diet-induced metabolic disorders, including insulin resistance and hepatic steatosis. However, the physiological mechanisms through which Nrg4 regulates energy balance and glucose and lipid metabolism remain incompletely understood. The aims of the current study were: i) to investigate the regulation of adipose Nrg4 expression during obesity and the physiological signals involved, ii) to elucidate the mechanisms underlying Nrg4 regulation of energy balance and glucose and lipid metabolism, and iii) to explore whether Nrg4 regulates adipose tissue secretome gene expression and adipokine secretion. METHODS: We examined the correlation of adipose Nrg4 expression with obesity in a cohort of diet-induced obese mice and investigated the upstream signals that regulate Nrg4 expression. We performed metabolic cage and hyperinsulinemic-euglycemic clamp studies in Nrg4 transgenic mice to dissect the metabolic pathways regulated by Nrg4. We investigated how Nrg4 regulates hepatic lipid metabolism in the fasting state and explored the effects of Nrg4 on adipose tissue gene expression, particularly those encoding secreted factors. RESULTS: Adipose Nrg4 expression is inversely correlated with adiposity and regulated by pro-inflammatory and anti-inflammatory signaling. Transgenic expression of Nrg4 increases energy expenditure and augments whole body glucose metabolism. Nrg4 protects mice from diet-induced hepatic steatosis in part through activation of hepatic fatty acid oxidation and ketogenesis. Finally, Nrg4 promotes a healthy adipokine profile during obesity. CONCLUSIONS: Nrg4 exerts pleiotropic beneficial effects on energy balance and glucose and lipid metabolism to ameliorate obesity-associated metabolic disorders. Biologic therapeutics based on Nrg4 may improve both type 2 diabetes and non-alcoholic fatty liver disease (NAFLD) in patients.


Assuntos
Adipocinas/sangue , Ácidos Graxos/metabolismo , Neurregulinas/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , Adipócitos/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Glucose/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurregulinas/genética , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/etiologia
6.
Trends Endocrinol Metab ; 26(5): 231-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25843910

RESUMO

Brown fat is highly active in fuel oxidation and dissipates chemical energy through uncoupling protein (UCP)1-mediated heat production. Activation of brown fat leads to increased energy expenditure, reduced adiposity, and lower plasma glucose and lipid levels, thus contributing to better homeostasis. Uncoupled respiration and thermogenesis have been considered to be responsible for the metabolic benefits of brown adipose tissue. Recent studies have demonstrated that brown adipocytes also secrete factors that act locally and systemically to influence fuel and energy metabolism. This review discusses the evidence supporting a thermogenesis-independent role of brown fat, particularly through its release of secreted factors, and their implications in physiology and therapeutic development.


Assuntos
Tecido Adiposo Marrom/metabolismo , Termogênese , Adipócitos Marrons/metabolismo , Adiponectina/fisiologia , Tecido Adiposo Marrom/inervação , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Metabolismo Energético , Fatores de Crescimento de Fibroblastos/fisiologia , Homeostase , Humanos , Canais Iônicos/fisiologia , Proteínas Mitocondriais/fisiologia , Fator de Crescimento Neural/fisiologia , Neurregulinas/fisiologia , Obesidade , Proteína Desacopladora 1 , Fator A de Crescimento do Endotélio Vascular/fisiologia
7.
Nat Med ; 20(12): 1436-1443, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25401691

RESUMO

Brown fat activates uncoupled respiration in response to cold temperature and contributes to systemic metabolic homeostasis. To date, the metabolic action of brown fat has been primarily attributed to its role in fuel oxidation and uncoupling protein 1 (UCP1)-mediated thermogenesis. Whether brown fat engages other tissues through secreted factors remains largely unexplored. Here we show that neuregulin 4 (Nrg4), a member of the epidermal growth factor (EGF) family of extracellular ligands, is highly expressed in adipose tissues, enriched in brown fat and markedly increased during brown adipocyte differentiation. Adipose tissue Nrg4 expression was reduced in rodent and human obesity. Gain- and loss-of-function studies in mice demonstrated that Nrg4 protects against diet-induced insulin resistance and hepatic steatosis through attenuating hepatic lipogenic signaling. Mechanistically, Nrg4 activates ErbB3 and ErbB4 signaling in hepatocytes and negatively regulates de novo lipogenesis mediated by LXR and SREBP1c in a cell-autonomous manner. These results establish Nrg4 as a brown fat-enriched endocrine factor with therapeutic potential for the treatment of obesity-associated disorders, including type 2 diabetes and nonalcoholic fatty liver disease (NAFLD).


Assuntos
Tecido Adiposo Marrom/metabolismo , Lipogênese , Fígado/metabolismo , Neurregulinas/genética , Obesidade/genética , RNA Mensageiro/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4/metabolismo , Células 3T3-L1 , Adipócitos Marrons/metabolismo , Adipogenia , Animais , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Células HEK293 , Humanos , Resistência à Insulina , Receptores X do Fígado , Camundongos , Neurregulinas/metabolismo , Obesidade/metabolismo , Receptores Nucleares Órfãos/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
8.
Mol Cell ; 55(3): 372-82, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25002143

RESUMO

Brown and beige/brite fats generate heat via uncoupled respiration to defend against cold. The total mass and activity of thermogenic adipose tissues are also tightly linked to systemic energy and nutrient homeostasis. Despite originating from distinct progenitors, brown and beige adipocytes acquire remarkably similar molecular and metabolic characteristics during differentiation through the action of a network of transcription factors and cofactors. How this regulatory network interfaces with long noncoding RNAs (lncRNAs), an emerging class of developmental regulators, remains largely unexplored. Here, we globally profiled lncRNA gene expression during thermogenic adipocyte formation and identified Brown fat lncRNA 1 (Blnc1) as a nuclear lncRNA that promotes brown and beige adipocyte differentiation and function. Blnc1 forms a ribonucleoprotein complex with transcription factor EBF2 to stimulate the thermogenic gene program. Further, Blnc1 itself is a target of EBF2, thereby forming a feedforward regulatory loop to drive adipogenesis toward thermogenic phenotype.


Assuntos
Adipogenia , Tecido Adiposo Marrom/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/genética , Ribonucleoproteínas/metabolismo
9.
Diabetes ; 63(4): 1340-52, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24379350

RESUMO

Chronic low-grade inflammation is emerging as a pathogenic link between obesity and metabolic disease. Persistent immune activation in white adipose tissue (WAT) impairs insulin sensitivity and systemic metabolism, in part, through the actions of proinflammatory cytokines. Whether obesity engages an adaptive mechanism to counteract chronic inflammation in adipose tissues has not been elucidated. Here we identified otopetrin 1 (Otop1) as a component of a counterinflammatory pathway that is induced in WAT during obesity. Otop1 expression is markedly increased in obese mouse WAT and is stimulated by tumor necrosis factor-α in cultured adipocytes. Otop1 mutant mice respond to high-fat diet with pronounced insulin resistance and hepatic steatosis, accompanied by augmented adipose tissue inflammation. Otop1 attenuates interferon-γ (IFN-γ) signaling in adipocytes through selective downregulation of the transcription factor STAT1. Using a tagged vector, we found that Otop1 physically interacts with endogenous STAT1. Thus, Otop1 defines a unique target of cytokine signaling that attenuates obesity-induced adipose tissue inflammation and plays an adaptive role in maintaining metabolic homeostasis in obesity.


Assuntos
Tecido Adiposo/patologia , Inflamação/prevenção & controle , Proteínas de Membrana/farmacologia , Obesidade/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica , Homeostase/efeitos dos fármacos , Inflamação/imunologia , Resistência à Insulina/fisiologia , Interferon gama/efeitos dos fármacos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Fator de Transcrição STAT1/metabolismo
10.
PLoS One ; 8(8): e70109, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23990898

RESUMO

The biological clock plays an important role in integrating nutrient and energy metabolism with other cellular processes. Previous studies have demonstrated that core clock genes are rhythmically expressed in peripheral tissues, including the liver, skeletal muscle, pancreatic islets, and white and brown adipose tissues. These peripheral clocks are entrained by physiological cues, thereby aligning the circadian pacemaker to tissue functions. The mechanisms that regulate brown adipose tissue clock in response to physiological signals remain poorly understood. Here we found that the expression of core clock genes is highly responsive to cold exposure in brown fat, but not in white fat. This cold-inducible regulation of the clock network is mediated by adrenergic receptor activation and the transcriptional coactivator PGC-1α. Brown adipocytes in mice lacking a functional clock contain large lipid droplets accompanied by dysregulation of genes involved in lipid metabolism and adaptive thermogenesis. Paradoxically, the "clockless" mice were competent in maintaining core body temperature during cold exposure. These studies elucidated the presence of adrenergic receptor/clock crosstalk that appears to be required for normal thermogenic gene expression in brown fat.


Assuntos
Tecido Adiposo Marrom/metabolismo , Relógios Biológicos , Regulação da Expressão Gênica , Receptores Adrenérgicos/metabolismo , Transdução de Sinais , Termogênese , Fatores de Transcrição ARNTL/metabolismo , Adipócitos/citologia , Animais , Temperatura Corporal , Diferenciação Celular , Ritmo Circadiano , Temperatura Baixa , Dioxóis/farmacologia , Perfilação da Expressão Gênica , Lipídeos/química , Masculino , Camundongos , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Reação em Cadeia da Polimerase , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...