Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 14(668): eabh1316, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36288279

RESUMO

Circadian rhythms play a critical role in regulating metabolism, including daily cycles of feeding/fasting. Glucokinase (GCK) is central for whole-body glucose homeostasis and oscillates according to a circadian clock. GCK activators (GKAs) effectively reduce hyperglycemia, but their use is also associated with hypoglycemia, hyperlipidemia, and hepatic steatosis. Given the circadian rhythmicity and natural postprandial activation of GCK, we hypothesized that GKA treatment would benefit from being timed specifically during feeding periods. Acute treatment of obese Zucker rats with the GKA AZD1656 robustly increased flux into all major metabolic pathways of glucose disposal, enhancing glucose elimination. Four weeks of continuous AZD1656 treatment of obese Zucker rats improved glycemic control; however, hepatic steatosis and inflammation manifested. In contrast, timing AZD1656 to feeding periods robustly reduced hepatic steatosis and inflammation in addition to improving glycemia, whereas treatment timed to fasting periods caused overall detrimental metabolic effects. Mechanistically, timing AZD1656 to feeding periods diverted newly synthesized lipid toward direct VLDL secretion rather than intrahepatic storage. In line with increased hepatic insulin signaling, timing AZD1656 to feeding resulted in robust activation of AKT, mTOR, and SREBP-1C after glucose loading, pathways known to regulate VLDL secretion and hepatic de novo lipogenesis. In conclusion, intermittent AZD1656 treatment timed to feeding periods promotes glucose disposal when needed the most, restores metabolic flexibility and hepatic insulin sensitivity, and thereby avoids hepatic steatosis. Thus, chronotherapeutic approaches may benefit the development of GKAs and other drugs acting on metabolic targets.


Assuntos
Fígado Gorduroso , Glucoquinase , Ratos , Animais , Ratos Zucker , Glucoquinase/metabolismo , Hipoglicemiantes/uso terapêutico , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Insulina/farmacologia , Glucose/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Fígado/metabolismo , Cronoterapia , Inflamação/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Lipídeos
2.
Nat Med ; 27(11): 1941-1953, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34608330

RESUMO

Obesity is considered an important factor for many chronic diseases, including diabetes, cardiovascular disease and cancer. The expansion of adipose tissue in obesity is due to an increase in both adipocyte progenitor differentiation and mature adipocyte cell size. Adipocytes, however, are thought to be unable to divide or enter the cell cycle. We demonstrate that mature human adipocytes unexpectedly display a gene and protein signature indicative of an active cell cycle program. Adipocyte cell cycle progression associates with obesity and hyperinsulinemia, with a concomitant increase in cell size, nuclear size and nuclear DNA content. Chronic hyperinsulinemia in vitro or in humans, however, is associated with subsequent cell cycle exit, leading to a premature senescent transcriptomic and secretory profile in adipocytes. Premature senescence is rapidly becoming recognized as an important mediator of stress-induced tissue dysfunction. By demonstrating that adipocytes can activate a cell cycle program, we define a mechanism whereby mature human adipocytes senesce. We further show that by targeting the adipocyte cell cycle program using metformin, it is possible to influence adipocyte senescence and obesity-associated adipose tissue inflammation.


Assuntos
Adipócitos/metabolismo , Ciclo Celular/fisiologia , Senescência Celular/fisiologia , Hiperinsulinismo/patologia , Obesidade/patologia , Tecido Adiposo/metabolismo , Diferenciação Celular/fisiologia , Ciclina D1/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Metformina/farmacologia
3.
PLoS One ; 16(2): e0247300, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606810

RESUMO

OBJECTIVE: Gremlin 1 (GREM1) is a secreted BMP2/4 inhibitor which regulates commitment and differentiation of human adipose precursor cells and prevents the browning effect of BMP4. GREM1 is an insulin antagonist and serum levels are high in type 2 diabetes (T2D). We here examined in vivo effects of AAV8 (Adeno-Associated Viral vectors of serotype eight) GREM 1 targeting the liver in mature mice to increase its systemic secretion and also, in a separate study, injected recombinant GREM 1 intraperitoneally. The objective was to characterize systemic effects of GREM 1 on insulin sensitivity, glucose tolerance, body weight, adipose cell browning and other local tissue effects. METHODS: Adult mice were injected with AAV8 vectors expressing GREM1 in the liver or receiving regular intra-peritoneal injections of recombinant GREM1 protein. The mice were fed with a low fat or high fat diet (HFD) and followed over time. RESULTS: Liver-targeted AAV8-GREM1 did not alter body weight, whole-body glucose and insulin tolerance, or adipose tissue gene expression. Although GREM1 protein accumulated in liver cells, GREM1 serum levels were not increased suggesting that it may not have been normally processed for secretion. Hepatic lipid accumulation, inflammation and fibrosis were also not changed. Repeated intraperitoneal rec-GREM1 injections for 5 weeks were also without effects on body weight and insulin sensitivity. UCP1 was slightly but significantly reduced in both white and brown adipose tissue but this was not of sufficient magnitude to alter body weight. We validated that recombinant GREM1 inhibited BMP4-induced pSMAD1/5/9 in murine cells in vitro, but saw no direct inhibitory effect on insulin signalling and pAkt (ser 473 and thr 308) activation. CONCLUSION: GREM1 accumulates intracellularly when overexpressed in the liver cells of mature mice and is apparently not normally processed/secreted. However, also repeated intraperitoneal injections were without effects on body weight and insulin sensitivity and adipose tissue UCP1 levels were only marginally reduced. These results suggest that mature mice do not readily respond to GREMLIN 1 but treatment of murine cells with GREMLIN 1 protein in vitro validated its inhibitory effect on BMP4 signalling while insulin signalling was not altered.


Assuntos
Dependovirus/genética , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fígado/metabolismo , Células 3T3-L1 , Animais , Peso Corporal , Linhagem Celular , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Teste de Tolerância a Glucose , Humanos , Injeções Intraperitoneais , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Masculino , Camundongos , Proteínas Recombinantes/administração & dosagem
4.
Nat Rev Endocrinol ; 17(5): 276-295, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33627836

RESUMO

In mammals, the white adipocyte is a cell type that is specialized for storage of energy (in the form of triacylglycerols) and for energy mobilization (as fatty acids). White adipocyte metabolism confers an essential role to adipose tissue in whole-body homeostasis. Dysfunction in white adipocyte metabolism is a cardinal event in the development of insulin resistance and associated disorders. This Review focuses on our current understanding of lipid and glucose metabolic pathways in the white adipocyte. We survey recent advances in humans on the importance of adipocyte hypertrophy and on the in vivo turnover of adipocytes and stored lipids. At the molecular level, the identification of novel regulators and of the interplay between metabolic pathways explains the fine-tuning between the anabolic and catabolic fates of fatty acids and glucose in different physiological states. We also examine the metabolic alterations involved in the genesis of obesity-associated metabolic disorders, lipodystrophic states, cancers and cancer-associated cachexia. New challenges include defining the heterogeneity of white adipocytes in different anatomical locations throughout the lifespan and investigating the importance of rhythmic processes. Targeting white fat metabolism offers opportunities for improved patient stratification and a wide, yet unexploited, range of therapeutic opportunities.


Assuntos
Adipócitos Brancos/metabolismo , Gerenciamento Clínico , Metabolismo dos Lipídeos/fisiologia , Obesidade/metabolismo , Animais , Homeostase , Humanos , Obesidade/terapia
5.
Methods Mol Biol ; 2164: 121-127, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32607889

RESUMO

Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease (NAFLD), characterized by steatosis (fat within the liver), inflammation, and fibrosis, which may progress to cirrhosis and hepatocellular carcinoma. Despite the high prevalence, there are currently no approved NASH drug treatments, which urges a faster development of new therapies to address this high unmet medical need. Drug development is facilitated by having reliable and translatable preclinical NASH models. Obesogenic dietary models recapitulate better the natural progression of NASH, with overnutrition and sedentary lifestyle being the main causes. Here we describe the use of a modified version of a diet-induced NASH model, known as the Amylin NASH diet model (AMLN-diet), particularly in the leptin-deficient Lepob/Lepob (ob/ob) mice.


Assuntos
Cirrose Hepática/patologia , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/patologia , Animais , Carcinoma Hepatocelular/patologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
6.
Mol Metab ; 32: 15-26, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32029225

RESUMO

OBJECTIVE: Bone morphogenetic protein 4 (BMP4) adeno-associated viral vectors of serotype 8 (AAV8) gene therapy targeting the liver prevents the development of obesity in initially lean mice by browning the large subcutaneous white adipose tissue (WAT) and enhancing energy expenditure. Here, we examine whether this approach could also reduce established obesity. METHODS: Dietary-induced obese C57BL6/N mice received AAV8 BMP4 gene therapy at 17-18 weeks of age. They were kept on a high-fat diet and phenotypically characterized for an additional 10-12 weeks. Following termination, the mice underwent additional characterization in vitro. RESULTS: Surprisingly, we observed no effect on body weight, browning of WAT, or energy expenditure in these obese mice, but whole-body insulin sensitivity and glucose tolerance were robustly improved. Insulin signaling and insulin-stimulated glucose uptake were increased in both adipose cells and skeletal muscle. BMP4 also decreased hepatic glucose production and reduced gluconeogenic enzymes in the liver, but not in the kidney, in addition to enhancing insulin action in the liver. CONCLUSIONS: Our findings show that BMP4 prevents, but does not reverse, established obesity in adult mice, while it improves insulin sensitivity independent of weight reduction. The BMP antagonist Noggin was increased in WAT in obesity, which may account for the lack of browning.


Assuntos
Tecido Adiposo Marrom , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/uso terapêutico , Terapia Genética , Insulina/metabolismo , Obesidade/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/induzido quimicamente , Transdução de Sinais
7.
Theranostics ; 10(2): 585-601, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31903139

RESUMO

Macrophages are important regulators of obesity-associated inflammation and PPARα and -γ agonism in macrophages has anti-inflammatory effects. In this study, we tested the efficacy with which liposomal delivery could target the PPARα/γ dual agonist tesaglitazar to macrophages while reducing drug action in common sites of drug toxicity: the liver and kidney, and whether tesaglitazar had anti-inflammatory effects in an in vivo model of obesity-associated dysmetabolism. Methods: Male leptin-deficient (ob/ob) mice were administered tesaglitazar or vehicle for one week in a standard oral formulation or encapsulated in liposomes. Following the end of treatment, circulating metabolic parameters were measured and pro-inflammatory adipose tissue macrophage populations were quantified by flow cytometry. Cellular uptake of liposomes in tissues was assessed using immunofluorescence and a broad panel of cell subset markers by flow cytometry. Finally, PPARα/γ gene target expression levels in the liver, kidney, and sorted macrophages were quantified to determine levels of drug targeting to and drug action in these tissues and cells. Results: Administration of a standard oral formulation of tesaglitazar effectively treated symptoms of obesity-associated dysmetabolism and reduced the number of pro-inflammatory adipose tissue macrophages. Macrophages are the major cell type that took up liposomes with many other immune and stromal cell types taking up liposomes to a lesser extent. Liposome delivery of tesaglitazar did not have effects on inflammatory macrophages nor did it improve metabolic parameters to the extent of a standard oral formulation. Liposomal delivery did, however, attenuate effects on liver weight and liver and kidney expression of PPARα and -γ gene targets compared to oral delivery. Conclusions: These findings reveal for the first time that tesaglitazar has anti-inflammatory effects on adipose tissue macrophage populations in vivo. These data also suggest that while nanoparticle delivery reduced off-target effects, yet the lack of tesaglitazar actions in non-targeted cells such (as hepatocytes and adipocytes) and the uptake of drug-loaded liposomes in many other cell types, albeit to a lesser extent, may have impacted overall therapeutic efficacy. This fulsome analysis of cellular uptake of tesaglitazar-loaded liposomes provides important lessons for future studies of liposome drug delivery.


Assuntos
Alcanossulfonatos/farmacologia , Rim/efeitos dos fármacos , Lipossomos/administração & dosagem , Fígado/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Obesidade/tratamento farmacológico , PPAR alfa/agonistas , PPAR gama/agonistas , Fenilpropionatos/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Inflamação/metabolismo , Rim/metabolismo , Lipossomos/química , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia
8.
PLoS One ; 14(11): e0224917, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31725756

RESUMO

Targeted nanoparticle delivery is a promising strategy for increasing efficacy and limiting side effects of therapeutics. When designing a targeted liposomal formulation, the in vivo biodistribution of the particles must be characterized to determine the value of the targeting approach. Peroxisome proliferator-activated receptor (PPAR) agonists effectively treat metabolic syndrome by decreasing dyslipidemia and insulin resistance but side effects have limited their use, making them a class of compounds that could benefit from targeted liposomal delivery. The adipose targeting sequence peptide (ATS) could fit this role, as it has been shown to bind to adipose tissue endothelium and induce weight loss when delivered conjugated to a pro-apoptotic peptide. To date, however, a full assessment of ATS in vivo biodistribution has not been reported, leaving important unanswered questions regarding the exact mechanisms whereby ATS targeting enhances therapeutic efficacy. We designed this study to evaluate the biodistribution of ATS-conjugated liposomes loaded with the PPARα/γ dual agonist tesaglitazar in leptin-deficient ob/ob mice. The ATS-liposome biodistribution in adipose tissue and other organs was examined at the cellular and tissue level using microscopy, flow cytometry, and fluorescent molecular tomography. Changes in metabolic parameters and gene expression were measured by target and off-target tissue responses to the treatment. Unexpectedly, ATS targeting did not increase liposomal uptake in adipose relative to other tissues, but did increase uptake in the kidneys. Targeting also did not significantly alter metabolic parameters. Analysis of the liposome cellular distribution in the stromal vascular fraction with flow cytometry revealed high uptake by multiple cell types. Our findings highlight the need for thorough study of in vivo biodistribution when evaluating a targeted therapy.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Sistemas de Liberação de Medicamentos , Tecido Adiposo/efeitos dos fármacos , Alcanossulfonatos/farmacologia , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Lipossomos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/efeitos dos fármacos , Peptídeos/farmacologia , Fenilpropionatos/farmacologia
9.
Nat Metab ; 1(4): 445-459, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-32694874

RESUMO

Liver macrophages (LMs) have been proposed to contribute to metabolic disease through secretion of inflammatory cytokines. However, anti-inflammatory drugs lead to only modest improvements in systemic metabolism. Here we show that LMs do not undergo a proinflammatory phenotypic switch in obesity-induced insulin resistance in flies, mice and humans. Instead, we find that LMs produce non-inflammatory factors, such as insulin-like growth factor-binding protein 7 (IGFBP7), that directly regulate liver metabolism. IGFBP7 binds to the insulin receptor and induces lipogenesis and gluconeogenesis via activation of extracellular-signal-regulated kinase (ERK) signalling. We further show that IGFBP7 is subject to RNA editing at a higher frequency in insulin-resistant than in insulin-sensitive obese patients (90% versus 30%, respectively), resulting in an IGFBP7 isoform with potentially higher capacity to bind to the insulin receptor. Our study demonstrates that LMs can contribute to insulin resistance independently of their inflammatory status and indicates that non-inflammatory factors produced by macrophages might represent new drug targets for the treatment of metabolic diseases.


Assuntos
Fígado/metabolismo , Macrófagos/metabolismo , Animais , Humanos , Inflamação/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Camundongos , Obesidade/metabolismo
10.
Diabetes ; 65(8): 2187-200, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27207510

RESUMO

Ectopic lipid accumulation in the liver is an almost universal feature of human and rodent models of generalized lipodystrophy and is also a common feature of type 2 diabetes, obesity, and metabolic syndrome. Here we explore the progression of fatty liver disease using a mouse model of lipodystrophy created by a fat-specific knockout of the insulin receptor (F-IRKO) or both IR and insulin-like growth factor 1 receptor (F-IR/IGFRKO). These mice develop severe lipodystrophy, diabetes, hyperlipidemia, and fatty liver disease within the first weeks of life. By 12 weeks of age, liver demonstrated increased reactive oxygen species, lipid peroxidation, histological evidence of balloon degeneration, and elevated serum alanine aminotransferase and aspartate aminotransferase levels. In these lipodystrophic mice, stored liver lipids can be used for energy production, as indicated by a marked decrease in liver weight with fasting and increased liver fibroblast growth factor 21 expression and intact ketogenesis. By 52 weeks of age, liver accounted for 25% of body weight and showed continued balloon degeneration in addition to inflammation, fibrosis, and highly dysplastic liver nodules. Progression of liver disease was associated with improvement in blood glucose levels, with evidence of altered expression of gluconeogenic and glycolytic enzymes. However, these mice were able to mobilize stored glycogen in response to glucagon. Feeding F-IRKO and F-IR/IGFRKO mice a high-fat diet for 12 weeks accelerated the liver injury and normalization of blood glucose levels. Thus, severe fatty liver disease develops early in lipodystrophic mice and progresses to advanced nonalcoholic steatohepatitis with highly dysplastic liver nodules. The liver injury is propagated by lipotoxicity and is associated with improved blood glucose levels.


Assuntos
Tecido Adiposo/metabolismo , Lipodistrofia/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptor de Insulina/metabolismo , Alanina Transaminase/metabolismo , Animais , Glicemia/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Teste de Tolerância a Glucose , Glicogênio/metabolismo , Immunoblotting , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/metabolismo , Lipodistrofia/genética , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Receptor de Insulina/genética
11.
FASEB J ; 27(8): 3257-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23682123

RESUMO

Vascular endothelial growth factor (VEGF) is critical for angiogenesis, but also has pleiotropic effects on several nonvascular cells. Our aim was to investigate the role of VEGF in brown adipose tissue (BAT). We show that VEGF expression increases 2.5-fold during differentiation of cultured murine brown adipocytes and that VEGF receptor-2 is phosphorylated, indicating VEGF signaling. VEGF increased proliferation in brown preadipocytes in vitro by 70%, and blockade of VEGF signaling using anti-VEGFR2 antibody DC101 increased brown adipocyte apoptosis, as determined by cell number and activation of caspase 3. Systemic VEGF neutralization in mice, accomplished by adenoviral expression of soluble Flt1, resulted in 7-fold increase in brown adipocyte apoptosis, mitochondrial degeneration, and increased mitophagy compared to control mice expressing a null adenovirus. Absence of the heparan sulfate-binding VEGF isoforms, VEGF164 and VEGF188, resulted in abnormal BAT development in mice at E15.5, with fewer brown adipocytes and lower mitochondrial protein compared to wild-type littermates. These results suggest a role for VEGF in brown adipocytes and preadipocytes to promote survival, proliferation, and normal mitochondria and development.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/embriologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Caspase 3/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
12.
Sci Signal ; 3(151): ra87, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21139139

RESUMO

Insulin and insulin-like growth factor 1 (IGF-1) act as antiapoptotic hormones. We found that, unexpectedly, double-knockout (DKO) cells that lacked both insulin and IGF-1 receptors (IR and IGF1R, respectively) were resistant to apoptosis induced through either the intrinsic or the extrinsic pathway. This resistance to apoptosis was associated with decreased abundance of the proapoptotic protein Bax and increases in abundance of the antiapoptotic proteins Bcl-2, Bcl-xL, XIAP, and Flip. These changes in protein abundance involved primarily posttranscriptional mechanisms. Restoration of IR or IGF1R to DKO cells also restored their sensitivity to apoptosis. Notably, expression of a catalytically inactive mutant form of the IR also restored susceptibility to apoptosis. Thus, IR and IGF1R have bidirectional roles in the control of cell survival and can be viewed as dependence receptors. Insulin and IGF-1 binding stimulates receptor tyrosine kinase activity and blocks apoptosis, whereas unliganded IR and IGF1R, acting through a mechanism independent of their catalytic activity, exert a permissive effect on cell death.


Assuntos
Apoptose/fisiologia , Modelos Biológicos , Receptor IGF Tipo 1/fisiologia , Receptor de Insulina/fisiologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Linhagem Celular Transformada , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxidantes/farmacologia , Fosfotransferases/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia
13.
Diabetes ; 58(4): 803-12, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19136652

RESUMO

OBJECTIVE: To investigate how insulin sensitivity and glucose metabolism differ in adipocytes between different fat depots of male and female mice and how sex steroids contribute to these differences. RESEARCH DESIGN AND METHODS: Adipocytes from intra-abdominal/perigonadal (PG) and subcutaneous (SC) adipose tissue from normal, castrated, or steroid-implanted animals were isolated and analyzed for differences in insulin sensitivity and glucose metabolism. RESULTS: Adipocytes from both PG and SC depots of females have increased lipogenic rates compared with those from males. In females, intra-abdominal PG adipocytes are more insulin-sensitive than SC adipocytes and more insulin-sensitive than male adipocytes from either depot. When stimulated by low physiological concentrations of insulin, female PG adipocytes show a robust increase in Akt and extracellular signal-related kinase (ERK) phosphorylation and lipogenesis, whereas male adipocytes show activation only at higher insulin concentrations. Adipocytes from females have higher mRNA/protein levels of several genes involved in glucose and lipid metabolism. After castration, adipocytes of male mice showed increased insulin sensitivity and increased lipogenic rates, whereas adipocytes of females demonstrate decreased lipid production. Increasing estrogen above physiological levels, however, also reduced lipid synthesis in females, whereas increasing dihydrotestosterone in males had no effect. CONCLUSIONS: There are major sex differences in insulin sensitivity in adipose tissue, particularly in the intra-abdominal depot, that are regulated by physiological levels of sex steroids. The increased sensitivity to insulin and lipogenesis observed in adipocytes from females may account for their lower level of insulin resistance and diabetes risk despite similar or higher fat content than in males.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Glucose/metabolismo , Caracteres Sexuais , Abdome , Adipócitos/citologia , Tecido Adiposo/citologia , Animais , Di-Hidrotestosterona/farmacologia , Estradiol/farmacologia , Feminino , Teste de Tolerância a Glucose , Proteínas Facilitadoras de Transporte de Glucose/genética , Insulina/metabolismo , Insulina/farmacologia , Insulina/fisiologia , Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Ovário/citologia , Ovário/fisiologia , RNA Mensageiro/genética , Fenômenos Fisiológicos da Pele , Testículo/citologia , Testículo/fisiologia
14.
Cell Metab ; 8(5): 437-45, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19046574

RESUMO

Adipose tissue (AT) secretes several adipokines that influence insulin sensitivity and potentially link obesity to insulin resistance. Apelin, a peptide present in different tissues, is also secreted by adipocytes. Apelin is upregulated in obese and hyperinsulinemic humans and mice. Although a tight relation exists between the regulation of apelin and insulin, it remains largely unknown whether apelin affects whole-body glucose utilization. Herein, we show that in chow-fed mice, acute intravenous injection of apelin has a powerful glucose-lowering effect associated with enhanced glucose utilization in skeletal muscle and AT. Through in vivo and in vitro pharmacological and genetic approaches, we demonstrate the involvement of endothelial NO synthase, AMP-activated protein kinase, and Akt in apelin-stimulated glucose uptake in soleus muscle. Remarkably, in obese and insulin-resistant mice, apelin restored glucose tolerance and increased glucose utilization. Apelin could thus represent a promising target in the management of insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Proteínas de Transporte/fisiologia , Glucose/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adipocinas , Animais , Apelina , Proteínas de Transporte/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína Oncogênica v-akt/metabolismo
15.
Obesity (Silver Spring) ; 14(11): 1890-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17135603

RESUMO

OBJECTIVE: To explore the regulation of secreted protein acidic and rich in cysteine (SPARC) expression and its role in adipose tissue. RESEARCH METHODS AND PROCEDURES: We studied the regulation of SPARC expression in transgenic mice expressing the human beta3 and alpha2 adrenergic receptors on a murine beta3 adrenergic receptor null background that became obese under a high-fat diet mainly as a result of adipose tissue hyperplasia. Furthermore, we analyzed its expression in human adipose tissue and its regulation during adipocyte differentiation. RESULTS: SPARC protein in adipose tissue was increased in obese transgenic mice compared with control mice, indicating that SPARC expression was associated with adipose tissue hyperplasia. Both SPARC mRNA and protein were detected in human adipose tissue. Comparing adipocytes and vascular stroma, we found that SPARC expression was mainly associated with the adipocyte fraction. Consistent with this, SPARC transcript increased during differentiation of human primary preadipocytes. 3T3-L1 preadipocytes showed an increase in SPARC expression in differentiated cells but with biphasic expression during the process. After induction in committed cells, SPARC mRNA and protein levels declined as differentiation began and returned to elevated levels in fully differentiated adipocytes. DISCUSSION: SPARC expression correlated with adipose tissue hyperplasia and adipogenesis. Therefore, SPARC seems to play a role in adipose tissue physiology as it is involved in growth and differentiation.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Regulação da Expressão Gênica , Obesidade/metabolismo , Osteonectina/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Humanos , Camundongos , Camundongos Transgênicos , Osteonectina/genética , RNA Mensageiro/metabolismo
16.
Metabolism ; 55(10): 1397-405, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16979412

RESUMO

Semicarbazide-sensitive amine oxidase (SSAO) is known to increase during in vitro adipogenesis and to be one of the most highly expressed membrane proteins of white adipocytes. Although less well documented, mitochondrial monoamine oxidases (MAOs) are also present in adipocytes and share with SSAO the capacity to generate hydrogen peroxide. This work therefore aimed to compare several biologic effects of MAO and SSAO substrates in 3T3-F442A adipocytes. In differentiated cells, tyramine oxidation was predominantly MAO dependent, whereas benzylamine oxidation was SSAO dependent. Both amines partially mimicked insulin actions, including stimulation of Akt phosphorylation and glucose uptake. In addition, tyramine and benzylamine impaired tumor necrosis factor alpha-dependent nitric oxide formation in a pargyline- and semicarbazide-sensitive manner, respectively. Various biogenic amines were tested in competition for tyramine or benzylamine oxidation and classified as MAO-preferring (methoxytyramine, tryptamine) or SSAO-preferring substrates (methylamine, octopamine). Short-term incubation with 1 mmol/L of all amines except histamine stimulated glucose uptake up to 20% to 50% of maximal insulin activation. One-week treatment with either MAO or SSAO substrates alone allowed postconfluent cells to differentiate into adipocytes, reproducing 60% of insulin-promoted lipid accumulation. All amines also exerted a slight improvement in the adipogenic action of insulin. Therefore, like SSAO, substrate activation of MAO can interact with adipocyte metabolism by mimicking diverse effects of insulin in addition to preventing tumor necrosis factor alpha-dependent responses.


Assuntos
Adipócitos/metabolismo , Amina Oxidase (contendo Cobre)/metabolismo , Insulina/fisiologia , Monoaminoxidase/metabolismo , Células 3T3 , Animais , Benzamidas/metabolismo , Transporte Biológico Ativo , Diferenciação Celular/fisiologia , Células Cultivadas , Glucose/metabolismo , Hexoses/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipólise/fisiologia , Camundongos , Nitritos/metabolismo , Fosforilação , Fatores de Necrose Tumoral/metabolismo , Tiramina/metabolismo
17.
FASEB J ; 20(9): 1528-30, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16723381

RESUMO

We have recently identified apelin as a novel adipokine up-regulated by insulin and obesity. Since obesity and insulin resistance are associated with chronically elevated levels of both insulin and TNFalpha, the present study was performed to investigate a putative regulation of apelin expression in adipocytes by TNFalpha. Herein, we report a tight correlation between apelin and TNFalpha expression in adipose tissue of lean and obese humans. Apelin regulation by TNFalpha was further studied in cultured explants of human adipose tissue. The endogenous expression of TNFalpha in adipocytes isolated from the explants was accompanied by a 6-9 h subsequent increase of apelin expression in adipocytes. This increase was reversed by inhibiting TNFalpha expression with 100 microM isobutylmethylxanthine. In different mouse models of obesity, expression of both TNFalpha and apelin was also significantly increased in adipocytes of obese mice. Furthermore, short-term exposure to an i.p. injection of TNFalpha in C57Bl6/J mice induced an increase of apelin expression in adipose tissue as well as apelin plasma levels. Finally, a direct positive effect of TNFalpha has been shown in differentiated 3T3F442A adipocytes on apelin expression and secretion. The signaling pathways of TNFalpha for the induction of apelin were dependent of PI3-kinase, c-Jun NH2-terminal kinase (JNK), and MAPK but not PKC activation. All together, these findings suggest that apelin might be a candidate to better understand potential links between obesity and associated disorders such as inflammation and insulin resistance.


Assuntos
Tecido Adiposo/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Células 3T3 , Abdome , Adipócitos/citologia , Adipócitos/fisiologia , Adipocinas , Tecido Adiposo/efeitos dos fármacos , Adulto , Animais , Apelina , Proteínas de Transporte , Diferenciação Celular , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/fisiopatologia , Resistência à Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/fisiopatologia
18.
J Biol Chem ; 278(20): 18162-9, 2003 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-12642576

RESUMO

Our group has recently demonstrated (Gesta, S., Simon, M., Rey, A., Sibrac, D., Girard, A., Lafontan, M., Valet, P., and Saulnier-Blache, J. S. (2002) J. Lipid Res. 43, 904-910) the presence, in adipocyte conditioned-medium, of a soluble lysophospholipase d-activity (LPLDact) involved in synthesis of the bioactive phospholipid lysophosphatidic acid (LPA). In the present report, LPLDact was purified from 3T3F442A adipocyte-conditioned medium and identified as the type II ecto-nucleotide pyrophosphatase phosphodiesterase, autotaxin (ATX). A unique ATX cDNA was cloned from 3T3F442A adipocytes, and its recombinant expression in COS-7 cells led to extracellular release of LPLDact. ATX mRNA expression was highly up-regulated during adipocyte differentiation of 3T3F442A-preadipocytes. This up-regulation was paralleled by the ability of newly differentiated adipocytes to release LPLDact and LPA. Differentiation-dependent up-regulation of ATX expression was also observed in a primary culture of mouse preadipocytes. Treatment of 3T3F442A-preadipocytes with concentrated conditioned medium from ATX-expressing COS-7 cells led to an increase in cell number as compared with concentrated conditioned medium from ATX non-expressing COS-7 cells. The specific effect of ATX on preadipocyte proliferation was completely suppressed by co-treatment with a LPA-hydrolyzing phospholipase, phospholipase B. Finally, ATX expression was found in mature adipocytes isolated from mouse adipose tissue and was substantially increased in genetically obese-diabetic db/db mice when compared with their lean siblings. In conclusion, the present work shows that ATX is responsible for the LPLDact released by adipocytes and exerts a paracrine control on preadipocyte growth via an LPA-dependent mechanism. Up-regulations of ATX expression with adipocyte differentiation and genetic obesity suggest a possible involvement of this released protein in the development of adipose tissue and obesity-associated pathologies.


Assuntos
Adipócitos/metabolismo , Glucose-6-Fosfato Isomerase/química , Glicoproteínas/química , Lisofosfolipídeos/metabolismo , Complexos Multienzimáticos , Sequência de Aminoácidos , Animais , Células COS , Diferenciação Celular , Divisão Celular , Clonagem Molecular , Meios de Cultivo Condicionados/farmacologia , DNA Complementar/metabolismo , Bases de Dados como Assunto , Eletroforese em Gel de Poliacrilamida , Glucose-6-Fosfato Isomerase/metabolismo , Glicoproteínas/metabolismo , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fosfodiesterase I , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA