Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(13)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-39000586

RESUMO

Visceral adipose tissue (VAT) dysfunction has been recently recognized as a potential contributor to the development of Alzheimer's disease (AD). This study aimed to explore the relationship between VAT metabolism and cerebral glucose metabolism in patients with cognitive impairment. This cross-sectional prospective study included 54 patients who underwent 18F-fluorodeoxyglucose (18F-FDG) brain and torso positron emission tomography/computed tomography (PET/CT), and neuropsychological evaluations. VAT metabolism was measured by 18F-FDG torso PET/CT, and cerebral glucose metabolism was measured using 18F-FDG brain PET/CT. A voxel-based analysis revealed that the high-VAT-metabolism group exhibited a significantly lower cerebral glucose metabolism in AD-signature regions such as the parietal and temporal cortices. In the volume-of-interest analysis, multiple linear regression analyses with adjustment for age, sex, and white matter hyperintensity volume revealed that VAT metabolism was negatively associated with cerebral glucose metabolism in AD-signature regions. In addition, higher VAT metabolism was correlated with poorer outcomes on cognitive assessments, including the Korean Boston Naming Test, Rey Complex Figure Test immediate recall, and the Controlled Oral Word Association Test. In conclusion, our study revealed significant relationships among VAT metabolism, cerebral glucose metabolism, and cognitive function. This suggests that VAT dysfunction actively contributes to the neurodegenerative processes characteristic of AD, making VAT dysfunction targeting a novel AD therapy approach.


Assuntos
Encéfalo , Disfunção Cognitiva , Fluordesoxiglucose F18 , Glucose , Gordura Intra-Abdominal , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Humanos , Masculino , Feminino , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/diagnóstico por imagem , Glucose/metabolismo , Idoso , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/diagnóstico por imagem , Fluordesoxiglucose F18/metabolismo , Estudos Transversais , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem , Pessoa de Meia-Idade , Estudos Prospectivos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Testes Neuropsicológicos
2.
J Biol Chem ; 298(12): 102619, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36272644

RESUMO

Thermoregulation is a process by which core body temperature is maintained in mammals. Males typically have a lower body temperature than females. However, the effects of androgens, which show higher levels in males, on adrenergic receptor-mediated thermogenesis remain unclear. Here, we demonstrate that androgen-androgen receptor (AR) signaling suppresses the ß-adrenergic agonist-induced rise of core body temperature using castrated and AR knockout (ARKO) male mice. Furthermore, in vitro mechanistic studies show that activated AR inhibits cAMP response element (CRE)-mediated transcription by suppressing cAMP response element-binding protein (CREB) phosphorylation. The elevation of body temperature induced by the ß-adrenergic agonist CL316243 was higher in ARKO and castrated mice than in the control mice. Similarly, CL316243 induced a greater increase in Uncoupling protein 1 (Ucp1) expression and CREB phosphorylation in the brown adipose tissue of ARKO mice than in that of controls. We determined that activation of AR by dihydrotestosterone suppressed ß3-agonist- or forskolin-induced CRE-mediated transcription, which was prevented by AR antagonist. AR activation also suppressed CREB phosphorylation induced by forskolin. Moreover, we found AR nuclear localization, but not transcriptional activity, was necessary for the suppression of CRE-mediated transcription. Finally, modified mammalian two-hybrid and immunoprecipitation analyses suggest nuclear AR and CREB form a protein complex both in the presence and absence of dihydrotestosterone and forskolin. These results suggest androgen-AR signaling suppresses ß-adrenoceptor-induced UCP1-mediated brown adipose tissue thermogenesis by suppressing CREB phosphorylation, presumably owing to a protein complex with AR and CREB. This mechanism explains sexual differences in body temperature, at least partially.


Assuntos
Tecido Adiposo Marrom , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Receptores Androgênicos , Termogênese , Animais , Feminino , Masculino , Camundongos , Tecido Adiposo Marrom/metabolismo , Androgênios/metabolismo , Colforsina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Di-Hidrotestosterona/farmacologia , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteína Desacopladora 1/genética , Temperatura Corporal
3.
J Biol Chem ; 295(7): 2034-2042, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31919095

RESUMO

Prolonged cold exposure stimulates the formation of brownlike adipocytes expressing UCP1 (uncoupling-protein-1) in subcutaneous white adipose tissue which, together with classical brown adipose tissue, contributes to maintaining body temperature in mammals through nonshivering thermogenesis. The mechanisms that regulate the formation of these cells, alternatively called beige or brite adipocytes, are incompletely understood. Here we report that mice lacking CD137, a cell surface protein used in several studies as a marker for beige adipocytes, showed elevated levels of thermogenic markers, including UCP1, increased numbers of beige adipocyte precursors, and expanded UCP1-expressing cell clusters in inguinal white adipose tissue after chronic cold exposure. CD137 knockout mice also showed enhanced cold resistance. These results indicate that CD137 functions as a negative regulator of "browning" in white adipose tissue and call into question the use of this protein as a functional marker for beige adipocytes.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Proteína Desacopladora 1/genética , Adipócitos Bege/metabolismo , Animais , Temperatura Corporal/genética , Temperatura Baixa , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Knockout , Termogênese/genética
4.
J Biol Chem ; 294(17): 6899-6911, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30842262

RESUMO

Sclerostin exerts profound local control over bone acquisition and also mediates endocrine communication between fat and bone. In bone, sclerostin's anti-osteoanabolic activity is enhanced by low-density lipoprotein receptor-related protein 4 (Lrp4), which facilitates its interaction with the Lrp5 and Lrp6 Wnt co-receptors. To determine whether Lrp4 similarly affects sclerostin's endocrine function, we examined body composition as well as glucose and fatty acid metabolism in mice rendered deficient of Lrp4 in the adipocyte (AdΔLrp4) or the osteoblast (ObΔLrp4). AdΔLrp4 mice exhibit a reduction in adipocyte hypertrophy and improved glucose and lipid homeostasis, marked by increased glucose and insulin tolerance and reduced serum fatty acids, and mirror the effect of sclerostin deficiency on whole-body metabolism. Indeed, epistasis studies place adipocyte-expressed Lrp4 and sclerostin in the same genetic cascade that regulates adipocyte function. Intriguingly, ObΔLrp4 mice, which exhibit dramatic increases in serum sclerostin, accumulate body fat and develop impairments in glucose tolerance and insulin sensitivity despite development of a high bone mass phenotype. These data indicate that expression of Lrp4 by both the adipocyte and osteoblast is required for normal sclerostin endocrine function and that the impact of sclerostin deficiency on adipocyte physiology is distinct from the effect on osteoblast function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos/metabolismo , Composição Corporal , Proteínas Relacionadas a Receptor de LDL/metabolismo , Osteoblastos/metabolismo , Animais , Células Cultivadas , Epistasia Genética , Ácidos Graxos/metabolismo , Homeostase , Proteínas Relacionadas a Receptor de LDL/genética , Camundongos
5.
J Biol Chem ; 294(41): 14896-14910, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31413114

RESUMO

The metabolic consequences and sequelae of obesity promote life-threatening morbidities. PKCδI is an important elicitor of inflammation and apoptosis in adipocytes. Here we report increased PKCδI activation via release of its catalytic domain concurrent with increased expression of proinflammatory cytokines in adipocytes from obese individuals. Using a screening strategy of dual recognition of PKCδI isozymes and a caspase-3 binding site on the PKCδI hinge domain with Schrödinger software and molecular dynamics simulations, we identified NP627, an organic small-molecule inhibitor of PKCδI. Characterization of NP627 by surface plasmon resonance (SPR) revealed that PKCδI and NP627 interact with each other with high affinity and specificity, SPR kinetics revealed that NP627 disrupts caspase-3 binding to PKCδI, and in vitro kinase assays demonstrated that NP627 specifically inhibits PKCδI activity. The SPR results also indicated that NP627 affects macromolecular interactions between protein surfaces. Of note, release of the PKCδI catalytic fragment was sufficient to induce apoptosis and inflammation in adipocytes. NP627 treatment of adipocytes from obese individuals significantly inhibited PKCδI catalytic fragment release, decreased inflammation and apoptosis, and significantly improved mitochondrial metabolism. These results indicate that PKCδI is a robust candidate for targeted interventions to manage obesity-associated chronic inflammatory diseases. We propose that NP627 may also be used in other biological systems to better understand the impact of caspase-3-mediated activation of kinase activity.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Obesidade/patologia , Proteína Quinase C-delta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Adipócitos/patologia , Tecido Adiposo/patologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Respiração Celular/efeitos dos fármacos , Humanos , Obesidade/metabolismo , Proteína Quinase C-delta/metabolismo , Hormônio Liberador de Tireotropina/análogos & derivados , Hormônio Liberador de Tireotropina/farmacologia
6.
J Biol Chem ; 294(50): 19306-19321, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31727739

RESUMO

Cholesterol plays essential structural and signaling roles in mammalian cells, but too much cholesterol can cause cytotoxicity. Acyl-CoA:cholesterol acyltransferases 1 and 2 (ACAT1/2) convert cholesterol into its storage form, cholesteryl esters, regulating a key step in cellular cholesterol homeostasis. Adipose tissue can store >50% of whole-body cholesterol. Interestingly, however, almost no ACAT activity is present in adipose tissue, and most adipose cholesterol is stored in its free form. We therefore hypothesized that increased cholesterol esterification may have detrimental effects on adipose tissue function. Here, using several approaches, including protein overexpression, quantitative RT-PCR, immunofluorescence, and various biochemical assays, we found that ACAT1 expression is significantly increased in the adipose tissue of the ob/ob mice. We further demonstrated that ACAT1/2 overexpression partially inhibited the differentiation of 3T3-L1 preadipocytes. In mature adipocytes, increased ACAT activity reduced the size of lipid droplets (LDs) and inhibited lipolysis and insulin signaling. Paradoxically, the amount of free cholesterol increased on the surface of LDs in ACAT1/2-overexpressing adipocytes, accompanied by increased LD localization of caveolin-1. Moreover, cholesterol depletion in adipocytes by treating the cells with cholesterol-deficient media or ß-cyclodextrins induced changes in cholesterol distribution that were similar to those caused by ACAT1/2 overexpression. Our results suggest that ACAT1/2 overexpression increases the level of free cholesterol on the LD surface, thereby impeding adipocyte function. These findings provide detailed insights into the role of free cholesterol in LD and adipocyte function and suggest that ACAT inhibitors have potential utility for managing disorders associated with extreme obesity.


Assuntos
Acetil-CoA C-Acetiltransferase/metabolismo , Adipócitos/metabolismo , Colesterol/metabolismo , Gotículas Lipídicas/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Tamanho da Partícula , Propriedades de Superfície
7.
J Dairy Sci ; 103(4): 3708-3718, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32008773

RESUMO

Adipose tissue plays an important role in a cow's ability to adapt to the metabolic demands of lactation, because of its central involvement in energy metabolism and immunity. High adiposity and adipose tissue resistance to insulin are associated with excessive lipid mobilization. We hypothesized that the response to a glucose challenge differs between cows of different body condition 21 d before and after calving and that the responses are explainable by gene expression in subcutaneous adipose tissue (SAT). In addition, we aimed to investigate insulin resistance with gene expression in SAT and lipid mobilization around parturition. Multiparous Holstein cows were grouped according to body conditions score (BCS) 4 wk before calving, as follows: BCS ≤ 3.0 = thin (T, n = 14); BCS 3.25 to 3.5 = optimal (O, n = 14); BCS ≥ 3.75 = over-conditioned (OC, n = 14). We collected SAT on d -21 and d 21 relative to calving. A reverse-transcriptase quantitative (RT-q)PCR was used to measure gene expression related to lipid metabolism. One hour after the collection of adipose tissue, an intravenous glucose tolerance test was carried out, with administration of 0.15 g of glucose per kg of body weight (with a 40% glucose solution). Once weekly from the first week before calving to the third week after calving, a blood sample was taken. The transition to lactation was associated with intensified release of energy stored in adipose tissue, a decrease in the lipogenic genes lipoprotein lipase (LPL) and diacylglycerol O-acyltransferase 2 (DGAT2), and an increase in the lipolytic gene hormone-sensitive lipase (LIPE). On d -21, compared with T cows, OC cows had lower mRNA abundance of LPL and DGAT2, and the latency of fatty acid response after glucose infusion was also longer (8.5 vs. 23.3 min) in OC cows. Cows with higher insulin area under the curve on d -21 had concurrently lower LPL and DGAT2 gene expression and greater concentration of fatty acids on d -7, d 7, and d 14. In conclusion, high adiposity prepartum lowers the whole-body lipid metabolism response to insulin and causes reduced expression of lipogenic genes in SAT 3 weeks before calving. In addition, more pronounced insulin release after glucose infusion on d -21 is related to higher lipid mobilization around calving, indicating an insulin-resistant state, and is associated with lower expression of lipogenic genes in SAT.


Assuntos
Tecido Adiposo/metabolismo , Expressão Gênica , Resistência à Insulina/fisiologia , Metabolismo dos Lipídeos/fisiologia , Período Pós-Parto/metabolismo , Animais , Bovinos , Dieta/veterinária , Metabolismo Energético/fisiologia , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Insulina/sangue , Lactação/fisiologia , Lipogênese/genética , Período Pós-Parto/genética , Gravidez
8.
J Biol Chem ; 293(37): 14249-14259, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30006347

RESUMO

In adipose tissue, resistance to insulin's ability to increase glucose uptake can be induced by multiple factors, including obesity. Impaired insulin action may take place at different spatial loci at the cellular or subcellular level. To begin to understand the spatial response to insulin in human subcutaneous adipose tissue (hSAT), we developed a quantitative imaging method for activation of a major signaling node in the glucoregulatory insulin signaling pathway. After treatment with insulin or control media, biopsied tissues were immunostained for Akt phosphorylation at Thr-308/9 (pAkt) and then imaged by confocal fluorescence microscopy automated to collect a large grid of high resolution fields. In hSAT from 40 men and women with obesity, substantial heterogeneity of pAkt densities in adipocyte membranes were quantified in each image mosaic using a spatial unit of at least twice the size of the point spread function. Statistical analysis of the distribution of pAkt spatial units was best fit as the weighted sum of two separate distributions, corresponding to either a low or high pAkt density. A "high pAkt fraction" metric was calculated from the fraction of high pAkt distributed units over the total units. Importantly, upon insulin stimulation, tissues from the same biopsy showed either a minimal or a substantial change in the high pAkt fraction. Further supporting a two-state response to insulin stimulation, subjects with similar insulin sensitivity indices are also segregated into either of two clusters identified by the amount of membrane-localized pAkt.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Obesidade/metabolismo , Gordura Subcutânea/metabolismo , Adipócitos/enzimologia , Adulto , Idoso , Membrana Celular/metabolismo , Estudos de Coortes , Ativação Enzimática , Feminino , Transportador de Glucose Tipo 4/metabolismo , Humanos , Resistência à Insulina , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Pessoa de Meia-Idade , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Gordura Subcutânea/enzimologia , Adulto Jovem
9.
J Biol Chem ; 292(11): 4469-4483, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28119455

RESUMO

Obesity is closely associated with various metabolic disorders. However, little is known about abnormalities in the metabolic change of obese adipose tissue. Here we use static metabolic analysis and in vivo metabolic turnover analysis to assess metabolic dynamics in obese mice. The static metabolic analyses showed that glutamate and constitutive metabolites of the TCA cycle were increased in the white adipose tissue (WAT) of ob/ob and diet-induced obesity mice but not in the liver or skeletal muscle of these obese mice. Moreover, in vivo metabolic turnover analyses demonstrated that these glucose-derived metabolites were dynamically and specifically produced in obese WAT compared with lean WAT. Glutamate rise in obese WAT was associated with down-regulation of glutamate aspartate transporter (GLAST), a major glutamate transporter for adipocytes, and low uptake of glutamate into adipose tissue. In adipocytes, glutamate treatment reduced adiponectin secretion and insulin-mediated glucose uptake and phosphorylation of Akt. These data suggest that a high intra-adipocyte glutamate level potentially relates to adipocyte dysfunction in obesity. This study provides novel insights into metabolic dysfunction in obesity through comprehensive application of in vivo metabolic turnover analysis in two obese animal models.


Assuntos
Tecido Adiposo Branco/metabolismo , Ciclo do Ácido Cítrico , Glutamatos/metabolismo , Metaboloma , Obesidade/metabolismo , Células 3T3-L1 , Animais , Dieta Hiperlipídica/efeitos adversos , Glucose/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Músculo Esquelético/metabolismo , Obesidade/etiologia
10.
J Biol Chem ; 292(41): 16810-16816, 2017 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-28842500

RESUMO

Brown and beige adipose tissues can catabolize stored energy to generate heat, relying on the principal effector of thermogenesis: uncoupling protein 1 (UCP1). This unique capability could be leveraged as a therapy for metabolic disease. Numerous animal and cellular models have now demonstrated that mitochondrial reactive oxygen species (ROS) signal to support adipocyte thermogenic identity and function. Herein, we contextualize these findings within the established principles of redox signaling and mechanistic studies of UCP1 function. We provide a framework for understanding the role of mitochondrial ROS signaling in thermogenesis together with testable hypotheses for understanding mechanisms and developing therapies.


Assuntos
Tecido Adiposo Marrom/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Termogênese , Proteína Desacopladora 1/metabolismo , Adipócitos/metabolismo , Animais , Humanos , Oxirredução
11.
J Biol Chem ; 292(24): 9958-9966, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28473468

RESUMO

Brown adipose tissue dissipates energy as heat, a process that relies on a high abundance of mitochondria and high levels of electron transport chain (ETC) complexes within these mitochondria. Two regulators of mitochondrial respiration and heat production in brown adipocytes are the transcriptional coactivator PGC-1α and its splicing isoform NT-PGC-1α, which control mitochondrial gene expression in the nucleus. Surprisingly, we found that, in brown adipocytes, some NT-PGC-1α localizes to mitochondria, whereas PGC-1α resides in the nucleus. Here we sought to investigate the role of NT-PGC-1α in brown adipocyte mitochondria. Immunocytochemistry, immunotransmission electron microscopy, and biochemical analyses indicated that NT-PGC-1α was located in the mitochondrial matrix in brown adipocytes. NT-PGC-1α was specifically enriched at the D-loop region of the mtDNA, which contains the promoters for several essential ETC complex genes, and was associated with LRP130, an activator of mitochondrial transcription. Selective expression of NT-PGC-1α and PGC-1α in PGC-1α-/- brown adipocytes similarly induced expression of nuclear DNA-encoded mitochondrial ETC genes, including the key mitochondrial transcription factor A (TFAM). Despite having comparable levels of TFAM expression, PGC-1α-/- brown adipocytes expressing NT-PGC-1α had higher expression of mtDNA-encoded ETC genes than PGC-1α-/- brown adipocytes expressing PGC-1α, suggesting a direct effect of NT-PGC-1α on mtDNA transcription. Moreover, this increase in mtDNA-encoded ETC gene expression was associated with enhanced respiration in NT-PGC-1α-expressing PGC-1α-/- brown adipocytes. Our findings reveal a previously unappreciated and isoform-specific role for NT-PGC-1α in the regulation of mitochondrial transcription in brown adipocytes and provide new insight into the transcriptional control of mitochondrial respiration.


Assuntos
Adipócitos Marrons/metabolismo , DNA Mitocondrial/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/agonistas , Mitocôndrias/metabolismo , Proteínas de Neoplasias/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Regiões Promotoras Genéticas , Adipócitos Marrons/citologia , Adipócitos Marrons/ultraestrutura , Adipogenia , Processamento Alternativo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Camundongos Endogâmicos C57BL , Mitocôndrias/ultraestrutura , Proteínas de Neoplasias/genética , Fosforilação Oxidativa , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta
12.
J Biol Chem ; 291(8): 4185-96, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26706316

RESUMO

Phospholipase C-related catalytically inactive protein (PRIP) was first identified as an inositol 1,4,5-trisphosphate-binding protein, and was later found to be involved in a variety of cellular events, particularly those related to protein phosphatases. We previously reported that Prip knock-out (KO) mice exhibit a lean phenotype with a small amount of white adipose tissue. In the present study, we examined whether PRIP is involved in energy metabolism, which could explain the lean phenotype, using high-fat diet (HFD)-fed mice. Prip-KO mice showed resistance to HFD-induced obesity, resulting in protection from glucose metabolism dysfunction and insulin resistance. Energy expenditure and body temperature at night were significantly higher in Prip-KO mice than in wild-type mice. Gene and protein expression of uncoupling protein 1 (UCP1), a thermogenic protein, was up-regulated in Prip-KO brown adipocytes in thermoneutral or cold environments. These phenotypes were caused by the promotion of lipolysis in Prip-KO brown adipocytes, which is triggered by up-regulation of phosphorylation of the lipolysis-related proteins hormone-sensitive lipase and perilipin, followed by activation of UCP1 and/or up-regulation of thermogenesis-related genes (e.g. peroxisome proliferator-activated receptor-γ coactivator-1α). The results indicate that PRIP negatively regulates UCP1-mediated thermogenesis in brown adipocytes.


Assuntos
Adipócitos Marrons/metabolismo , Canais Iônicos/metabolismo , Lipólise , Proteínas Mitocondriais/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Obesidade/metabolismo , Termogênese , Adipócitos Marrons/patologia , Animais , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Coativadores de Receptor Nuclear/genética , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Proteína Desacopladora 1
13.
J Biol Chem ; 291(13): 6989-7003, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26853464

RESUMO

Ceramides are bioactive sphingolipids, which are composed of sphingoid bases carrying acyl chains of various lengths. Ceramides are synthesized by a family of six ceramide synthases (CerS) in mammals, which produce ceramides with differentN-linked acyl chains. Increased ceramide levels are known to contribute to the development of obesity and insulin resistance. Recently, it has been demonstrated that the ceramide acylation pattern is of particular importance for an organism to maintain energy homeostasis. However, which of theCerSfamily members are involved in this process is not yet completely known. Using newly developedCerS5knock-out mice, we show here thatCerS5is essential to maintain cellular C16:0sphingolipid pools in lung, spleen, muscle, liver, and white adipose tissue. Glycerophospholipid levels inCerS5-deficient mice were not altered. We found a strong impact of CerS5-dependent ceramide synthesis in white adipose tissue after high fat diet feeding. In skeletal muscle, liver, and spleen, C16:0-ceramide levels were altered independent of feeding conditions. The loss ofCerS5is associated with reduced weight gain and improved systemic health, including maintenance of glucose homeostasis and reduced white adipose tissue inflammation after high fat diet challenge. Our findings indicate that reduction of endogenous C16:0-ceramide by genetic inhibition ofCerS5is sufficient to ameliorate obesity and its comorbidities.


Assuntos
Ceramidas/biossíntese , Dieta Hiperlipídica , Gorduras na Dieta/efeitos adversos , Obesidade/enzimologia , Esfingosina N-Aciltransferase/genética , Tecido Adiposo Branco/enzimologia , Tecido Adiposo Branco/patologia , Animais , Glicemia/metabolismo , Expressão Gênica , Teste de Tolerância a Glucose , Resistência à Insulina/genética , Isoenzimas/deficiência , Isoenzimas/genética , Fígado/enzimologia , Fígado/patologia , Pulmão/enzimologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Esfingosina N-Aciltransferase/deficiência , Baço/enzimologia , Baço/patologia
14.
J Biol Chem ; 291(20): 10867-75, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-27002153

RESUMO

FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified ßKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of ßKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais/fisiologia , Adipócitos/citologia , Tecido Adiposo/citologia , Animais , Feminino , Fatores de Crescimento de Fibroblastos/genética , Proteínas Klotho , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , PPAR gama/genética , PPAR gama/metabolismo
15.
J Biol Chem ; 291(47): 24747-24755, 2016 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-27758866

RESUMO

The acquisition of beige adipocyte features by white fat cells corresponds to protection against obesity-induced metabolic diseases in humans and animal models of type 2 diabetes. In adipose tissue, expression of the E2 small ubiquitin-like modifier ligase ubiquitin carrier protein 9 (Ubc9) is positively correlated with markers of insulin resistance and corresponds with impaired browning of human white adipocytes. However, the molecular regulation of Ubc9 expression in adipocytes and other cells remains unclear. In this study, we demonstrate that the mRNA and protein expression of Ubc9 are regulated by the microRNA miRNA-30a (miR-30a) in human subcutaneous adipocytes. Ubc9 and miR-30a exhibit inverse expression in adipose tissue, with miR-30a robustly elevated in brown fat. Depletion of Ubc9 by siRNA or enforced expression of a miR-30a mimic augments mitochondrial volume and respiration in human white adipocytes, reflecting features of brown fat cells. Furthermore, Ubc9 depletion induces a brown fat gene program in human subcutaneous adipocytes. Induction of the beige-selective gene program corresponds to stabilization of the PR domain-containing 16 (PRDM16) protein, an obligate transcriptional regulator of the brown/beige fat metabolic program in white adipocytes that interacts with Ubc9. Taken together, our data demonstrate a previously unappreciated molecular axis that controls browning of human white adipocytes.


Assuntos
Adipócitos Brancos/metabolismo , Regulação da Expressão Gênica/fisiologia , MicroRNAs/biossíntese , Mitocôndrias/metabolismo , Enzimas de Conjugação de Ubiquitina/biossíntese , Adipócitos Brancos/citologia , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Masculino , Camundongos , Fatores de Transcrição/metabolismo
16.
J Biol Chem ; 291(39): 20551-62, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27489163

RESUMO

Emerging evidence suggests that n-3 polyunsaturated fatty acids (PUFA) promote brown adipose tissue thermogenesis. However, the underlying mechanisms remain elusive. Here, we hypothesize that n-3 PUFA promotes brown adipogenesis by modulating miRNAs. To test this hypothesis, murine brown preadipocytes were induced to differentiate the fatty acids of palmitic, oleate, or eicosapentaenoic acid (EPA). The increases of brown-specific signature genes and oxygen consumption rate by EPA were concurrent with up-regulation of miR-30b and 378 but not by oleate or palmitic acid. Next, we hypothesize that free fatty acid receptor 4 (Ffar4), a functional receptor for n-3 PUFA, modulates miR-30b and 378. Treatment of Ffar4 agonist (GW9508) recapitulated the thermogenic activation of EPA by increasing oxygen consumption rate, brown-specific marker genes, and miR-30b and 378, which were abrogated in Ffar4-silenced cells. Intriguingly, addition of the miR-30b mimic was unable to restore EPA-induced Ucp1 expression in Ffar4-depleted cells, implicating that Ffar4 signaling activity is required for up-regulating the brown adipogenic program. Moreover, blockage of miR-30b or 378 by locked nucleic acid inhibitors significantly attenuated Ffar4 as well as brown-specific signature gene expression, suggesting the signaling interplay between Ffar4 and miR-30b/378. The association between miR-30b/378 and brown thermogenesis was also confirmed in fish oil-fed C57/BL6 mice. Interestingly, the Ffar4 agonism-mediated signaling axis of Ffar4-miR-30b/378-Ucp1 was linked with an elevation of cAMP in brown adipocytes, similar to cold-exposed or fish oil-fed brown fat. Taken together, our work identifies a novel function of Ffar4 in modulating brown adipogenesis partly through a mechanism involving cAMP activation and up-regulation of miR-30b and miR-378.


Assuntos
Tecido Adiposo Marrom/metabolismo , Ácido Eicosapentaenoico/farmacologia , MicroRNAs/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Termogênese/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Temperatura Baixa , AMP Cíclico/metabolismo , Feminino , Masculino , Metilaminas/farmacologia , Camundongos , Propionatos/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Termogênese/fisiologia , Proteína Desacopladora 1/metabolismo , Regulação para Cima/fisiologia
17.
J Biol Chem ; 290(39): 23603-15, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26260790

RESUMO

Endurance exercise training induces substantial adaptive cardiac modifications such as left ventricular hypertrophy (LVH). Simultaneously to the development of LVH, adipose tissue (AT) lipolysis becomes elevated upon endurance training to cope with enhanced energy demands. In this study, we investigated the impact of adipose tissue lipolysis on the development of exercise-induced cardiac hypertrophy. Mice deficient for adipose triglyceride lipase (Atgl) in AT (atATGL-KO) were challenged with chronic treadmill running. Exercise-induced AT lipolytic activity was significantly reduced in atATGL-KO mice accompanied by the absence of a plasma fatty acid (FA) increase. These processes were directly associated with a prominent attenuation of myocardial FA uptake in atATGL-KO and a significant reduction of the cardiac hypertrophic response to exercise. FA serum profiling revealed palmitoleic acid (C16:1n7) as a new molecular co-mediator of exercise-induced cardiac hypertrophy by inducing nonproliferative cardiomyocyte growth. In parallel, serum FA analysis and echocardiography were performed in 25 endurance athletes. In consonance, the serum C16:1n7 palmitoleate level exhibited a significantly positive correlation with diastolic interventricular septum thickness in those athletes. No correlation existed between linoleic acid (18:2n6) and diastolic interventricular septum thickness. Collectively, our data provide the first evidence that adipose tissue lipolysis directly promotes the development of exercise-induced cardiac hypertrophy involving the lipokine C16:1n7 palmitoleate as a molecular co-mediator. The identification of a lipokine involved in physiological cardiac growth may help to develop future lipid-based therapies for pathological LVH or heart failure.


Assuntos
Tecido Adiposo/metabolismo , Cardiomegalia/etiologia , Ácidos Graxos Monoinsaturados/metabolismo , Lipólise , Condicionamento Físico Animal , Animais , Cardiomegalia/metabolismo , Linhagem Celular , Camundongos , Camundongos Knockout
18.
J Biol Chem ; 290(44): 26476-90, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26370079

RESUMO

Adaptive thermogenesis is the cellular process transforming chemical energy into heat in response to cold. A decrease in adaptive thermogenesis is a contributing factor to obesity. However, the molecular mechanisms responsible for the compromised adaptive thermogenesis in obese subjects have not yet been elucidated. In this study we hypothesized that Toll-like receptor 4 (TLR4) activation and subsequent inflammatory responses are key regulators to suppress adaptive thermogenesis. To test this hypothesis, C57BL/6 mice were either fed a palmitate-enriched high fat diet or administered with chronic low-dose LPS before cold acclimation. TLR4 stimulation by a high fat diet or LPS were both associated with reduced core body temperature and heat release. Impairment of thermogenic activation was correlated with diminished expression of brown-specific markers and mitochondrial dysfunction in subcutaneous white adipose tissue (sWAT). Defective sWAT browning was concomitant with elevated levels of endoplasmic reticulum (ER) stress and autophagy. Consistently, TLR4 activation by LPS abolished cAMP-induced up-regulation of uncoupling protein 1 (UCP1) in primary human adipocytes, which was reversed by silencing of C/EBP homologous protein (CHOP). Moreover, the inactivation of ER stress by genetic deletion of CHOP or chemical chaperone conferred a resistance to the LPS-induced suppression of adaptive thermogenesis. Collectively, our data indicate the existence of a novel signaling network that links TLR4 activation, ER stress, and mitochondrial dysfunction, thereby antagonizing thermogenic activation of sWAT. Our results also suggest that TLR4/ER stress axis activation may be a responsible mechanism for obesity-mediated defective brown adipose tissue activation.


Assuntos
Tecido Adiposo/metabolismo , Estresse do Retículo Endoplasmático , Mitocôndrias/metabolismo , Termogênese , Receptor 4 Toll-Like/metabolismo , Animais , Deleção de Genes , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Receptor 4 Toll-Like/genética , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Proteína Desacopladora 1
19.
J Transl Med ; 14(1): 212, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27421877

RESUMO

BACKGROUND: Adipose tissue regulates postprandial lipid metabolism by storing dietary fat through lipoprotein lipase-mediated hydrolysis of exogenous triglycerides, and by inhibiting delivery of endogenous non-esterified fatty acid to nonadipose tissues. Animal studies show that acute hypoxia, a model of obstructive sleep apnea, reduces adipose tissue lipoprotein lipase activity and increases non-esterified fatty acid release, adversely affecting postprandial lipemia. These observations remain to be tested in humans. METHODS: We used differentiated human preadipocytes exposed to acute hypoxia as well as adipose tissue biopsies obtained from 10 healthy men exposed for 6 h to either normoxia or intermittent hypoxia following an isocaloric high-fat meal. RESULTS: In differentiated preadipocytes, acute hypoxia induced a 6-fold reduction in lipoprotein lipase activity. In humans, the rise in postprandial triglyceride levels did not differ between normoxia and intermittent hypoxia. Non-esterified fatty acid levels were higher during intermittent hypoxia session. Intermittent hypoxia did not affect subcutaneous abdominal adipose tissue lipoprotein lipase activity. No differences were observed in lipolytic responses of isolated subcutaneous abdominal adipocytes between normoxia and intermittent hypoxia sessions. CONCLUSIONS: Acute hypoxia strongly inhibits lipoprotein lipase activity in differentiated human preadipocytes. Acute intermittent hypoxia increases circulating plasma non-esterified fatty acid in young healthy men, but does not seem to affect postprandial triglyceride levels, nor subcutaneous abdominal adipose tissue lipoprotein lipase activity and adipocyte lipolysis.


Assuntos
Tecido Adiposo/enzimologia , Tecido Adiposo/patologia , Hipóxia/enzimologia , Hipóxia/patologia , Lipólise , Lipase Lipoproteica/metabolismo , Adipócitos/metabolismo , Adulto , Diferenciação Celular , Feminino , Frequência Cardíaca , Humanos , Hipóxia/sangue , Masculino , Oxiemoglobinas/metabolismo , Adulto Jovem
20.
J Biol Chem ; 289(20): 14045-55, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24662293

RESUMO

Successful adaptation to periods of chronic caloric excess is a highly coordinated event that is critical to the survival and propagation of species. Transcription factor C/ebp homologous protein (Chop) is thought to be an important molecular mediator that integrates nutrient signals to endoplasmic reticulum (ER) stress and innate immune activation. Given that aberrant ER stress response is implicated in inducing metabolic inflammation and insulin resistance, we hypothesized that ER stress target gene Chop integrates immune and metabolic systems to adapt to chronic positive energy balance. Here we report that inactivation of Chop in mice fed a high fat diet led to significant increase in obesity caused by a reduction in energy expenditure without any change in food intake. Importantly, ablation of Chop does not induce metabolically healthy obesity, because Chop-deficient mice fed a high fat diet had increased hepatic steatosis with significantly higher insulin resistance. Quantification of adipose tissue leukocytosis revealed that elimination of Chop during obesity led to substantial increase in number of adipose tissue T and B lymphocytes. In addition, deficiency of Chop led to increase in total number of myeloid subpopulations like neutrophils and F4/80(+) adipose tissue macrophages without any alterations in the frequency of M1- or M2-like adipose tissue macrophages. Further investigation of inflammatory mechanisms revealed that ablation of Chop increases the sensitivity of macrophages to inflammasome-induced activation of IL-ß in macrophages. Our findings indicate that regulated expression of Chop during obesity is critical for adaptation to chronic caloric excess and maintenance of energy homeostasis via integration of metabolic and immune systems.


Assuntos
Tecido Adiposo/imunologia , Leucocitose/imunologia , Leucocitose/metabolismo , Obesidade/imunologia , Obesidade/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Metabolismo Energético , Deleção de Genes , Inflamassomos/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Linfócitos T/imunologia , Fator de Transcrição CHOP/deficiência , Fator de Transcrição CHOP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA