Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33058756

RESUMO

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Assuntos
Doenças Metabólicas/genética , MicroRNAs/genética , Adipócitos Marrons/patologia , Adiposidade , Alelos , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Dieta Hiperlipídica , Metabolismo Energético , Epigênese Genética , Loci Gênicos , Glucose/metabolismo , Homeostase , Humanos , Hipertrofia , Resistência à Insulina , Leptina/deficiência , Leptina/metabolismo , Masculino , Mamíferos/genética , Camundongos Endogâmicos C57BL , Camundongos Obesos , MicroRNAs/metabolismo , Obesidade/genética , Oligonucleotídeos/metabolismo , Especificidade da Espécie
2.
J Lipid Res ; 60(4): 856-868, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30782959

RESUMO

Treatment with PPARγ agonists in vivo improves human adipocyte metabolism, but the cellular mechanisms and possible depot differences in responsiveness to their effects are poorly understood. To examine the ex vivo metabolic effects of rosiglitazone (Rosi), we cultured explants of human visceral (omental) and abdominal subcutaneous adipose tissues for 7 days. Rosi increased mRNA levels of transcriptional regulators of brite/beige adipocytes (PGC1α, PRDM16), triglyceride synthesis (GPAT3, DGAT1), and lipolysis (ATGL) similarly in adipose tissues from both depots. In parallel, Rosi increased key modulators of FA oxidation (UCP1, FABP3, PLIN5 protein), rates of FA oxidation, and protein levels of electron transport complexes, suggesting an enhanced respiratory capacity as confirmed in newly differentiated adipocytes. Rosi led to the formation of small lipid droplets (SLDs) around the adipocyte central lipid droplet; each SLD was decorated with redistributed mitochondria that colocalized with PLIN5. SLD maintenance required lipolysis and FA reesterification. Rosi thus coordinated a structural and metabolic remodeling in adipocytes from both visceral and subcutaneous depots that enhanced oxidative capacity. Selective targeting of these cellular mechanisms to improve adipocyte FA handling may provide a new approach to treat metabolic complications of obesity and diabetes.


Assuntos
Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Gotículas Lipídicas/efeitos dos fármacos , Rosiglitazona/farmacologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Adulto , Idoso , Feminino , Humanos , Gotículas Lipídicas/metabolismo , Masculino , Pessoa de Meia-Idade , Oxirredução , Fenótipo
3.
Arterioscler Thromb Vasc Biol ; 38(4): 733-743, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29348118

RESUMO

OBJECTIVE: Adipose tissue cholesterol increases with adipocyte triglyceride content and size during development of obesity. However, how adipocyte cholesterol affects adipocyte function is poorly understood. The aim of this study was to evaluate the role of the cellular cholesterol exporter, Abca1 (ATP-binding cassette transporter A1), on adipose tissue function during diet-induced obesity. APPROACH AND RESULTS: Adiponectin Cre recombinase transgenic mice were crossed with Abca1flox/flox mice to generate ASKO (adipocyte-specific Abca1 knockout) mice. Control and ASKO mice were then fed a high-fat, high-cholesterol (45% calories as fat and 0.2% cholesterol) diet for 16 weeks. Compared with control mice, ASKO mice had a 2-fold increase in adipocyte plasma membrane cholesterol content and significantly lower body weight, epididymal fat pad weight, and adipocyte size. ASKO versus control adipose tissue had decreased PPARγ (peroxisome proliferator-activated receptor γ) and CCAAT/enhancer-binding protein expression, nuclear SREBP1 (sterol regulatory element-binding protein 1) protein, lipogenesis, and triglyceride accretion but similar Akt activation after acute insulin stimulation. Acute siRNA-mediated Abca1 silencing during 3T3L1 adipocyte differentiation reduced adipocyte Abca1 and PPARγ protein expression and triglyceride content. Systemic stimulated triglyceride lipolysis and glucose homeostasis were similar between control and ASKO mice. CONCLUSIONS: Adipocyte Abca1 is a key regulator of adipocyte lipogenesis and lipid accretion, likely because of increased adipose tissue membrane cholesterol, resulting in decreased activation of lipogenic transcription factors PPARγ and SREBP1.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/deficiência , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Colesterol na Dieta , Dieta Hiperlipídica , Deleção de Genes , Lipólise , Obesidade/prevenção & controle , Células 3T3-L1 , Transportador 1 de Cassete de Ligação de ATP/genética , Adipócitos/patologia , Adipogenia , Tecido Adiposo/patologia , Tecido Adiposo/fisiopatologia , Animais , Colesterol na Dieta/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , PPAR gama/metabolismo , Fenótipo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo , Aumento de Peso
4.
Am J Physiol Endocrinol Metab ; 312(1): E58-E71, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27879248

RESUMO

Increased adipocyte size is hypothesized to signal the recruitment of adipose progenitor cells (APCs) to expand tissue storage capacity. To investigate depot and sex differences in adipose growth, male and female C57BL/6J mice (10 wk-old) were challenged with high-fat (HF) or low-fat (LF) diets (D) for 14 wk. The HFD increased gonadal (GON) depot weight by adipocyte hypertrophy and hyperplasia in females but hypertrophy alone in males. In both sexes, inguinal (ING) adipocytes were smaller than GON, and depot expansion was due to hypertrophy. Matrix metalloproteinase 3 (Mmp3), an antiadipogenic factor, and its inhibitor Timps modulate the extracellular matrix remodeling needed for depot expansion. Mmp3 mRNA was depot different (ING > GON), higher in females than males and mainly expressed in APCs. In males, HFD-induced obesity increased tissue and APC Mmp3 mRNA levels and MMP3 protein and enzymatic activity. In females however, HFD significantly decreased MMP3 protein without affecting its mRNA levels. MMP3 activity also decreased (significant in ING). Timp4 mRNA was expressed mainly in adipocytes, and HFD-induced obesity tended to increase the ratio of TIMP4 to MMP3 protein in females, whereas it decreased it in males. Overexpression of Mmp3 in 3T3-L1 preadipocytes or rhMMP3 protein added to primary human preadipocytes inhibited differentiation, whereas rhTIMP4 improved adipogenesis and attenuated the inhibitory effect of rhMMP3. These data suggest that HFD-induced obesity downregulates APC MMP3 expression to trigger adipogenesis, and adipocyte TIMP4 may modulate this process to regulate hyperplastic vs. hypertrophic adipose tissue expansion, fat distribution, and metabolic health in a sex- and depot-dependent manner.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/crescimento & desenvolvimento , Dieta Hiperlipídica , Metaloproteinase 3 da Matriz/genética , Obesidade/genética , RNA Mensageiro/metabolismo , Inibidores Teciduais de Metaloproteinases/genética , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Adipogenia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Dieta com Restrição de Gorduras , Feminino , Humanos , Hiperplasia , Hipertrofia , Masculino , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Proteínas Recombinantes/farmacologia , Inibidores Teciduais de Metaloproteinases/metabolismo , Inibidores Teciduais de Metaloproteinases/farmacologia , Inibidor Tecidual 4 de Metaloproteinase
5.
J Immunol ; 194(4): 1467-79, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595782

RESUMO

Premature atherosclerosis is a severe complication of lupus and other systemic autoimmune disorders. Gain-of-function polymorphisms in IFN regulatory factor 5 (IRF5) are associated with an increased risk of developing lupus, and IRF5 deficiency in lupus mouse models ameliorates disease. However, whether IRF5 deficiency also protects against atherosclerosis development in lupus is not known. In this study, we addressed this question using the gld.apoE(-/-) mouse model. IRF5 deficiency markedly reduced lupus disease severity. Unexpectedly, despite the reduction in systemic immune activation, IRF5-deficient mice developed increased atherosclerosis and also exhibited metabolic dysregulation characterized by hyperlipidemia, increased adiposity, and insulin resistance. Levels of the atheroprotective cytokine IL-10 were reduced in aortae of IRF5-deficient mice, and in vitro studies demonstrated that IRF5 is required for IL-10 production downstream of TLR7 and TLR9 signaling in multiple immune cell types. Chimera studies showed that IRF5 deficiency in bone marrow-derived cells prevents lupus development and contributes in part to the increased atherosclerosis. Notably, IRF5 deficiency in non-bone marrow-derived cells also contributes to the increased atherosclerosis through the generation of hyperlipidemia and increased adiposity. Together, our results reveal a protective role for IRF5 in lupus-associated atherosclerosis that is mediated through the effects of IRF5 in both immune and nonimmune cells. These findings have implications for the proposed targeting of IRF5 in the treatment of autoimmune disease as global IRF5 inhibition may exacerbate cardiovascular disease in these patients.


Assuntos
Aterosclerose/etiologia , Fatores Reguladores de Interferon/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Síndrome Metabólica/etiologia , Animais , Aterosclerose/imunologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Fatores Reguladores de Interferon/deficiência , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/patologia , Masculino , Síndrome Metabólica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Adv Exp Med Biol ; 1043: 29-51, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224089

RESUMO

Sex differences in adipose tissue distribution and the metabolic, endocrine, and immune functions of different anatomical fat depots have been described, but they are incompletely documented in the literature. It is becoming increasingly clear that adipose depots serve distinct functions in males and females and have specific physiological roles. However, the mechanisms that regulate the size and function of specific adipose tissues in men and women remain poorly understood. New insights from mouse models have advanced our understanding of depot differences in adipose growth and remodeling via the proliferation and differentiation of adipose progenitors that can expand adipocyte number in the tissue or simply replace dysfunctional older and larger adipocytes. A limited ability of a depot to expand or remodel can lead to excessive adipocyte hypertrophy, which is often correlated with metabolic dysfunction. However, the relationship of adipocyte size and function varies by depot and sex. For example, femoral adipose tissues of premenopausal women appear to have a greater capacity for adipose expansion via hyperplasia and hypertrophy; although larger, these gluteal-femoral adipocytes remain insulin sensitive. The microenvironment of specific depots, including the composition of the extracellular matrix and cellular composition, as well as cell-autonomous genetic differences, influences sex- and depot-dependent metabolic and growth properties. Although there are some species differences, studies of the molecular and physiological determinants of sex differences in adipocyte growth and function in humans and rodents are both needed for understanding sex differences in health and disease.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Adiposidade , Adipócitos/patologia , Adipogenia , Tecido Adiposo/fisiopatologia , Adiposidade/genética , Animais , Proliferação de Células , Microambiente Celular , Feminino , Disparidades nos Níveis de Saúde , Humanos , Masculino , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Doenças Metabólicas/fisiopatologia , Modelos Animais , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Fatores de Risco , Caracteres Sexuais , Fatores Sexuais , Transdução de Sinais
7.
J Lipid Res ; 57(7): 1256-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27178044

RESUMO

The glucocorticoid-induced leucine zipper (GILZ), a primary target of glucocorticoids, is expressed in human adipocytes, but its importance in adipocyte function is unknown. Because TNFα is increased in obese adipose tissue and antagonizes a number of glucocorticoid actions, we investigated the interplay of these pathways. GILZ knockdown increased and GILZ overexpression decreased interleukin-6 (IL-6) and leptin mRNA and protein secretion. GILZ knockdown increased the magnitude of the glucocorticoid effect on leptin secretion, but did not affect the glucocorticoid suppression of IL-6. Although GILZ silencing decreased adiponectin mRNA levels, it did not affect the amount of adiponectin secreted. GILZ negatively modulated pro-inflammatory signaling pathways, blocking basal and TNFα-stimulated (1 h) p65 nuclear factor κB nuclear translocation and transcriptional activity by binding to p65 in the cytoplasm. GILZ silencing increased basal ERK1/2 and JNK phosphorylation, and decreased MAPK phosphatase-1 protein levels. Longer term TNFα (4 h or 24 h) treatment decreased GILZ expression in human adipocytes. Furthermore, adipose tissue GILZ mRNA levels were reduced in proportion to the degree of obesity and expression of inflammatory markers. Overall, these results suggest that GILZ antagonizes the pro-inflammatory effects of TNFα in human adipocytes, and its downregulation in obesity may contribute to adipose inflammation and dysregulated adipokine production, and thereby systemic metabolism.


Assuntos
Inflamação/genética , Obesidade/genética , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Adulto , Biópsia , Fosfatase 1 de Especificidade Dupla/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , Inflamação/patologia , Interleucina-6/metabolismo , Leptina/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Obesidade/metabolismo , Obesidade/patologia , RNA Mensageiro/biossíntese , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/biossíntese , Fator de Necrose Tumoral alfa/genética
8.
J Biol Chem ; 289(12): 8473-83, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24509860

RESUMO

Mice and humans lacking caveolae due to gene knock-out or inactivating mutations of cavin-1/PTRF have numerous pathologies including markedly aberrant fuel metabolism, lipodystrophy, and muscular dystrophy. We characterized the physiologic/metabolic profile of cavin-1 knock-out mice and determined that they were lean because of reduced white adipose depots. The knock-out mice were resistant to diet-induced obesity and had abnormal lipid metabolism in the major metabolic organs of white and brown fat and liver. Epididymal white fat cells from cavin-1-null mice were small and insensitive to insulin and ß-adrenergic agonists resulting in reduced adipocyte lipid storage and impaired lipid tolerance. At the molecular level, the lipolytic defects in white fat were caused by impaired perilipin phosphorylation, and the reduced triglyceride accumulation was caused by decreased fatty acid uptake and incorporation as well as the virtual absence of insulin-stimulated glucose transport. The livers of cavin-1-null mice were mildly steatotic and did not accumulate more lipid after high-fat feeding. The brown adipose tissues of cavin-1-null mice exhibited decreased mitochondria protein expression, which was restored upon high fat feeding. Taken together, these data suggest that dysfunction in fat, muscle, and liver metabolism in cavin-1-null mice causes a pleiotropic phenotype, one apparently identical to that of humans lacking caveolae in all tissues.


Assuntos
Adipócitos/metabolismo , Adipócitos/patologia , Deleção de Genes , Metabolismo dos Lipídeos , Proteínas de Membrana/genética , Animais , Cavéolas/metabolismo , Cavéolas/patologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA , Triglicerídeos/metabolismo
9.
Biochim Biophys Acta ; 1842(3): 473-81, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23735216

RESUMO

Central obesity is associated with insulin resistance and dyslipidemia. Thus, the mechanisms that control fat distribution and its impact on systemic metabolism have importance for understanding the risk for diabetes and cardiovascular disease. Hypercortisolemia at the systemic (Cushing's syndrome) or local levels (due to adipose-specific overproduction via 11ß-hydroxysteroid dehydrogenase 1) results in the preferential expansion of central, especially visceral fat depots. At the same time, peripheral subcutaneous depots can become depleted. The biochemical and molecular mechanisms underlying the depot-specific actions of glucocorticoids (GCs) on adipose tissue function remain poorly understood. GCs exert pleiotropic effects on adipocyte metabolic, endocrine and immune functions, and dampen adipose tissue inflammation. GCs also regulate multiple steps in the process of adipogenesis. Acting synergistically with insulin, GCs increase the expression of numerous genes involved in fat deposition. Variable effects of GC on lipolysis are reported, and GC can improve or impair insulin action depending on the experimental conditions. Thus, the net effect of GC on fat storage appears to depend on the physiologic context. The preferential effects of GC on visceral adipose tissue have been linked to higher cortisol production and glucocorticoid receptor expression, but the molecular details of the depot-dependent actions of GCs are only beginning to be understood. In addition, increasing evidence underlines the importance of circadian variations in GCs in relationship to the timing of meals for determining their anabolic actions on the adipocyte. In summary, although the molecular mechanisms remain to be fully elucidated, there is increasing evidence that GCs have multiple, depot-dependent effects on adipocyte gene expression and metabolism that promote central fat deposition. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/patologia , Síndrome de Cushing/patologia , Obesidade Abdominal/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Adipócitos/patologia , Tecido Adiposo/metabolismo , Síndrome de Cushing/metabolismo , Glucocorticoides/metabolismo , Humanos , Resistência à Insulina/genética , Lipólise/genética , Obesidade Abdominal/genética , Obesidade Abdominal/patologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
10.
J Biol Chem ; 288(42): 30330-30344, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23990461

RESUMO

The enterocyte expresses two fatty acid-binding proteins (FABP), intestinal FABP (IFABP; FABP2) and liver FABP (LFABP; FABP1). LFABP is also expressed in liver. Despite ligand transport and binding differences, it has remained uncertain whether these intestinally coexpressed proteins, which both bind long chain fatty acids (FA), are functionally distinct. Here, we directly compared IFABP(-/-) and LFABP(-/-) mice fed high fat diets containing long chain saturated or unsaturated fatty acids, reasoning that providing an abundance of dietary lipid would reveal unique functional properties. The results showed that mucosal lipid metabolism was indeed differentially modified, with significant decreases in FA incorporation into triacylglycerol (TG) relative to phospholipid (PL) in IFABP(-/-) mice, whereas LFABP(-/-) mice had reduced monoacylglycerol incorporation in TG relative to PL, as well as reduced FA oxidation. Interestingly, striking differences were found in whole body energy homeostasis; LFABP(-/-) mice fed high fat diets became obese relative to WT, whereas IFABP(-/-) mice displayed an opposite, lean phenotype. Fuel utilization followed adiposity, with LFABP(-/-) mice preferentially utilizing lipids, and IFABP(-/-) mice preferentially metabolizing carbohydrate for energy production. Changes in body weight and fat may arise, in part, from altered food intake; mucosal levels of the endocannabinoids 2-arachidonoylglycerol and arachidonoylethanolamine were elevated in LFABP(-/-), perhaps contributing to increased energy intake. This direct comparison provides evidence that LFABP and IFABP have distinct roles in intestinal lipid metabolism; differential intracellular functions in intestine and in liver, for LFABP(-/-) mice, result in divergent downstream effects at the systemic level.


Assuntos
Gorduras na Dieta/farmacologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Ácidos Graxos/farmacologia , Fosfolipídeos/metabolismo , Triglicerídeos/metabolismo , Animais , Gorduras na Dieta/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Fosfolipídeos/genética , Triglicerídeos/genética
11.
Obesity (Silver Spring) ; 32(1): 70-79, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37929774

RESUMO

OBJECTIVE: This study investigated remodeling of cellular metabolism and structures during browning of primary human adipocytes derived from both visceral and subcutaneous adipose tissues. Effects of glucocorticoids on the browning were also assessed. METHODS: Differentiated omental and subcutaneous human adipocytes were treated with rosiglitazone, with or without dexamethasone, and expression levels of brite adipocyte markers, lipolysis, and lipid droplet and mitochondrial structures were examined. RESULTS: Both omental and subcutaneous adipocytes acquired brite phenotypes upon peroxisome proliferator-activated receptor-γ agonist treatment, and dexamethasone tended to enhance the remodeling. Although rosiglitazone increased lipolysis during treatment, brite adipocytes exhibited lower basal lipolytic rates and enhanced responses to ß-adrenergic agonists or atrial natriuretic peptide. Transcriptome analysis identified induction of both breakdown and biosynthesis of lipids in brite adipocytes. After 60+ days in culture, lipid droplet size increased to ~50 microns, becoming almost unilocular in control adipocytes, and after browning, they acquired paucilocular morphology, clusters of small lipid droplets (1-2 micron) surrounded by mitochondria appearing on the periphery of the central large one. CONCLUSIONS: Metabolic and structural remodeling during browning of primary human adipocytes is similar to previous findings in human adipocytes in vivo, supporting their uses for mechanical studies investigating browning with translational relevance.


Assuntos
Adipócitos , Gordura Subcutânea , Humanos , Rosiglitazona/farmacologia , Rosiglitazona/metabolismo , Adipócitos/metabolismo , Gordura Subcutânea/metabolismo , Lipólise , Dexametasona
12.
Cell Rep ; 43(5): 114240, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38753486

RESUMO

Adipose tissue remodeling and dysfunction, characterized by elevated inflammation and insulin resistance, play a central role in obesity-related development of type 2 diabetes (T2D) and cardiovascular diseases. Long intergenic non-coding RNAs (lincRNAs) are important regulators of cellular functions. Here, we describe the functions of linc-ADAIN (adipose anti-inflammatory), an adipose lincRNA that is downregulated in white adipose tissue of obese humans. We demonstrate that linc-ADAIN knockdown (KD) increases KLF5 and interleukin-8 (IL-8) mRNA stability and translation by interacting with IGF2BP2. Upregulation of KLF5 and IL-8, via linc-ADAIN KD, leads to an enhanced adipogenic program and adipose tissue inflammation, mirroring the obese state, in vitro and in vivo. KD of linc-ADAIN in human adipose stromal cell (ASC) hTERT adipocytes implanted into mice increases adipocyte size and macrophage infiltration compared to implanted control adipocytes, mimicking hallmark features of obesity-induced adipose tissue remodeling. linc-ADAIN is an anti-inflammatory lincRNA that limits adipose tissue expansion and lipid storage.


Assuntos
Adipogenia , Interleucina-8 , Fatores de Transcrição Kruppel-Like , Estabilidade de RNA , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Adipogenia/genética , Animais , Estabilidade de RNA/genética , Interleucina-8/metabolismo , Interleucina-8/genética , Camundongos , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Obesidade/genética , Obesidade/patologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Masculino , Inflamação/patologia , Inflamação/genética , Inflamação/metabolismo
13.
Biochem Biophys Res Commun ; 432(2): 296-301, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23399566

RESUMO

Human adipocytes express high levels of two distinct lipid droplet proteins, fat specific protein 27 (FSP27; also called CIDEC), a member of the CIDE family, and perilipin1 (PLIN1), a member of the PAT family. Both proteins play a role in fat metabolism in adipocytes, but how they interact is not known. Our present study demonstrates that FSP27 and PLIN1 co-localize and interact in cultured human primary adipocytes. We also found that the C-terminal domain of FSP27, aa 120-220, interacts with PLIN1. Individual expression of exogenous FSP27 or PLIN1 increased triglyceride content and decreased glycerol release (a measure of lipolysis), but co-expression of both proteins did not further increase triglyceride content or decrease lipolysis in human adipocytes. However, the combination of PLIN1 and FSP27 increased the average size of lipid droplets or caused the formation of unilocular adipocytes. Our data suggest that FSP27 interacts with PLIN1 to regulate lipid droplet size in human adipocytes in a concerted manner.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte/metabolismo , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Triglicerídeos/metabolismo , Proteínas Reguladoras de Apoptose , Células Cultivadas , Humanos , Lipólise , Perilipina-1 , Proteínas/genética
14.
Curr Atheroscler Rep ; 15(10): 361, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23982264

RESUMO

Adipose tissue modifies the development of cardiovascular disease in a complex manner: obesity is a major risk factor, especially when accompanied by a central fat distribution. For that reason the characteristics of visceral adipose tissue have attracted most of the research interest thus far, and measurement of waist circumference is now recommended for everyday clinical practice. However, the direct, causative role of visceral fat in cardiometabolic disease remains to be established. Epidemiological and clinical studies show that accumulation of fat subcutaneously, in the gluteofemoral area, is protective against cardiovascular disease, but the exact molecular mechanisms remain unclear. In the last few years, imaging has allowed the study of smaller fat depots that may interact locally with important tissues: epicardial fat with the myocardium, perivascular fat with the vessel wall and the developing atherosclerotic plaque, and renal sinus fat with the renal artery. Unraveling the heterogeneous fat distribution and metabolic phenotypes in human obesity will facilitate optimal assessment of cardiovascular risk in overweight and obese individuals.


Assuntos
Doenças Cardiovasculares/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Doenças Cardiovasculares/etiologia , Modelos Animais de Doenças , Humanos , Obesidade/complicações , Fatores de Risco
15.
Curr Opin Genet Dev ; 81: 102079, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37406429

RESUMO

Body fat is stored in anatomically distinct adipose depots that vary in their cell composition and play specialized roles in systemic metabolic homeostasis via secreted products. Their local effects on nearby tissues (e.g. the gut and visceral adipose tissues) are increasingly recognized and this local crosstalk is being elucidated. The major subcutaneous fat depots, abdominal and gluteal-femoral, exert opposite effects on the risk of metabolic disease. The pace of research into developmental, sex, and genetic determinants of human adipose depot growth and function is rapidly accelerating, providing insight into the pathogenesis of metabolic dysfunction in persons with obesity.


Assuntos
Tecido Adiposo , Obesidade , Humanos , Obesidade/metabolismo , Tecido Adiposo/patologia , Adiposidade , Gordura Subcutânea/metabolismo , Gordura Subcutânea/patologia , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia
16.
Exp Biol Med (Maywood) ; 248(3): 209-216, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36544403

RESUMO

Dietary supplementation with l-arginine has been reported to reduce white fat mass in diet-induced obese rats and in obese humans. This study was conducted to test the hypothesis that the arginine treatment regulates glucose and fatty acid metabolism in insulin-sensitive tissues. Male Sprague-Dawley rats (4-week-old) were fed either low- or high-fat diets for 15 weeks (n = 16/diet). Thereafter, lean or obese rats were fed their respective diets and received drinking water containing either 1.51% l-arginine-HCl or 2.55% alanine (isonitrogenous control) (n = 8/treatment group). After 12 weeks of treatment, rats were euthanized and tissue samples were collected for biochemical assays. High-fat feeding increased the size of adipocytes isolated from retroperitoneal (RP) adipose tissue, while arginine treatment reduced their size. The total number of adipocytes in the adipose tissue did not differ among the four groups of rats. Glucose oxidation in extensor digitorum longus (EDL) muscle, soleus muscle, and RP adipose tissue were reduced in response to high-fat feeding. On the contrary, oleic acid oxidation in RP adipose tissue was enhanced in rats fed the high-fat diet. Arginine treatment stimulated both glucose and oleic acid oxidation in EDL and soleus muscles, while having no effect on glucose oxidation, oleic acid oxidation, or basal lipolysis per 106 adipocytes in RP adipose tissue. Collectively, these results indicate that oral supplementation with arginine to diet-induced obese rats promoted the oxidation of energy substrates in skeletal muscle, thereby reducing white fat in the body.


Assuntos
Tecido Adiposo , Ácido Oleico , Humanos , Ratos , Masculino , Animais , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ratos Sprague-Dawley , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Músculo Esquelético/metabolismo , Arginina/metabolismo , Glucose/metabolismo , Dieta Hiperlipídica , Suplementos Nutricionais
17.
Cell Metab ; 6(3): 155-6, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17767901

RESUMO

The essential amino acid leucine serves as a signal that activates protein synthesis. A new study by She et al. (2007) in this issue of Cell Metabolism shows that raising circulating leucine by blocking leucine breakdown drives a futile cycle of protein synthesis and degradation that contributes to higher-energy expenditure, resistance to dietary obesity, and improved insulin sensitivity.


Assuntos
Leucina/metabolismo , Ciclização de Substratos , Redução de Peso , Animais , Metabolismo Energético , Humanos , Obesidade/prevenção & controle , Proteínas/metabolismo
18.
Am J Physiol Endocrinol Metab ; 303(9): E1126-33, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22949029

RESUMO

High concentrations of TNF within obese adipose tissue increase basal lipolysis and antagonize insulin signaling. Adipocytes of the obese are also exposed to elevated levels of glucocorticoids (GCs), which antagonize TNF actions in many cell types. We tested the hypothesis that TNF decreases sensitivity to the antilipolytic effect of insulin and that GCs antagonize this effect in differentiated human adipocytes. Lipolysis and expression levels of lipolytic proteins were measured after treating adipocytes with TNF, dexamethasone (DEX), or DEX + TNF for up to 48 h. TNF not only increased basal lipolysis, it caused resistance to the antilipolytic effects of insulin in human adipocytes. DEX alone did not significantly affect lipolysis. Cotreatment with DEX blocked TNF induction of basal lipolysis and insulin resistance by antagonizing TNF stimulation of PKA-mediated phosphorylation of hormone-sensitive lipase (HSL) at Ser56³ and Ser66° and perilipin. TNF did not affect perilipin, HSL, or phosphodiesterase-3B mass but paradoxically suppressed adipose tissue triglyceride lipase expression, and this effect was blocked by DEX. The extent to which GCs can restrain the lipolytic actions of TNF may both diminish the potentially deleterious effects of excess lipolysis and contribute to fat accumulation in obesity.


Assuntos
Glucocorticoides/farmacologia , Resistência à Insulina , Insulina/metabolismo , Lipólise/efeitos dos fármacos , Gordura Subcutânea Abdominal/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Dexametasona/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipase/genética , Lipase/metabolismo , Masculino , Pessoa de Meia-Idade , Perilipina-1 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Esterol Esterase/genética , Esterol Esterase/metabolismo , Gordura Subcutânea Abdominal/citologia , Gordura Subcutânea Abdominal/metabolismo , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/antagonistas & inibidores
19.
Metabolism ; 137: 155331, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36228741

RESUMO

BACKGROUND: The triglyceride (TG) transfer activity of microsomal triglyceride transfer protein (MTP) is essential for lipoprotein assembly in the liver and intestine; however, its function in adipose tissue, which does not assemble lipoproteins, is unknown. Here we have elucidated the function of MTP in adipocytes. APPROACH AND RESULTS: We demonstrated that MTP is present on lipid droplets in human adipocytes. Adipose-specific MTP deficient (A-Mttp-/-) male and female mice fed an obesogenic diet gained less weight than Mttpf/f mice, had less fat mass, smaller adipocytes and were insulin sensitive. A-Mttp-/- mice showed higher energy expenditure than Mttpf/f mice. During a cold challenge, A-Mttp-/- mice maintained higher body temperature by mobilizing more fatty acids. Biochemical studies indicated that MTP deficiency de-repressed adipose triglyceride lipase (ATGL) activity and increased TG lipolysis. Both wild type MTP and mutant MTP deficient in TG transfer activity interacted with and inhibited ATGL activity. Thus, the TG transfer activity of MTP is not required for ATGL inhibition. C-terminally truncated ATGL that retains its lipase activity interacted less efficiently than full-length ATGL. CONCLUSION: Our findings demonstrate that adipose-specific MTP deficiency increases ATGL-mediated TG lipolysis and enhances energy expenditure, thereby resisting diet-induced obesity. We speculate that the regulatory function of MTP involving protein-protein interactions might have evolved before the acquisition of TG transfer activity in vertebrates. Adipose-specific inhibition of MTP-ATGL interactions may ameliorate obesity while avoiding the adverse effects associated with inhibition of the lipid transfer activity of MTP.


Assuntos
Lipase , Lipólise , Animais , Feminino , Humanos , Masculino , Camundongos , Adipócitos/metabolismo , Lipase/metabolismo , Lipídeos/farmacologia , Obesidade/metabolismo
20.
Am J Physiol Endocrinol Metab ; 300(3): E571-80, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21189358

RESUMO

Glucocorticoids (GC) are powerful regulators of adipocyte differentiation, metabolism, and endocrine function and promote the development of upper body obesity, especially visceral fat stores. To provide a comprehensive understanding of how GC affect adipose tissue and adipocyte function, we analyzed patterns of gene expression (HG U95 Affymetrix arrays) after culture of abdominal subcutaneous (Abd sc) and omental (Om) adipose tissues from severely obese subjects (3 F, 1 M) in the presence of insulin or insulin (7 nM) plus dexamethasone (Dex, 25 nM) for 7 days. About 20% (561 genes in Om and 569 genes in sc) of 2,803 adipose expressed genes were affected by long-term GC. While most of the genes (90%) were commonly regulated by Dex in both depots, 26 in Om and 34 in Abd sc were affected by Dex in only one depot. 60% of the commonly upregulated genes were involved in metabolic pathways and were expressed mainly in adipocytes. Dex suppressed genes in immune/inflammatory (IL-6, IL-8, and MCP-1, expressed in nonadipocytes) and proapoptotic pathways, yet induced genes related to the acute-phase response (SAA, factor D, haptoglobin, and RBP4, expressed in adipocytes) and stress/defense response. Functional classification analysis showed that Dex also induced expression levels of 22 transcription factors related to insulin action and lipogenesis (LXRα, STAT5α, SREBP1, and FoxO1) and immunity/adipogenesis (TSC22D3) while suppressing 17 transcription factors in both depots. Overall, these studies reveal the powerful effects of GC on gene networks that regulate many key functions in human adipose tissue.


Assuntos
Tecido Adiposo/metabolismo , Glucocorticoides/farmacologia , Omento/metabolismo , Gordura Subcutânea/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/efeitos dos fármacos , Adulto , Idoso , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Metabolismo dos Carboidratos/efeitos dos fármacos , Metabolismo dos Carboidratos/genética , Dexametasona/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , RNA/biossíntese , RNA/genética , Transdução de Sinais/efeitos dos fármacos , Células Estromais/metabolismo , Gordura Subcutânea/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA