Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 31(12): 2920-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21940942

RESUMO

OBJECTIVE: Insulin resistance renders macrophages more prone to cholesterol-induced apoptosis by promoting nuclear localization of transcription factor forkhead box transcription factor (Fox) O1. However, FoxO1 also decreases macrophage inflammation, raising the question of how the balance between proapoptotic and antiinflammatory effects is determined. We sought to identify the mechanism whereby FoxO1 dampens inflammation without promoting apoptosis. We hypothesized that nutrient-dependent FoxO1 acetylation plays a role in this process. METHODS AND RESULTS: We generated knock-in mice bearing alleles that encode constitutively deacetylated FoxO1 and studied the ex vivo response of primary peritoneal macrophages. We show that macrophages derived from mice homozygous for constitutively deacetylated FoxO1 alleles retain antiinflammatory properties in response to free cholesterol loading, without increasing apoptosis. Deacetylated FoxO1 inhibits free cholesterol-induced Akt phosphorylation and increases levels of the nuclear factor-κB precursor p105, decreasing nuclear translocation of nuclear factor-κB p65 subunit and dampening mitogen-activated protein/extracellular signal-regulated kinase activation to prevent inflammation. CONCLUSIONS: Deacetylated FoxO1 regulates p105 to prevent macrophage inflammation without causing apoptosis, suggesting a potential novel therapeutic approach to atherosclerosis through FoxO1 deacetylation.


Assuntos
Alelos , Apoptose/fisiologia , Colesterol/efeitos adversos , Fatores de Transcrição Forkhead/genética , Homozigoto , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Macrófagos Peritoneais/metabolismo , Acetilação , Animais , Células Cultivadas , Colesterol/farmacologia , Citocinas/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Inflamação/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Camundongos , Modelos Animais , NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
2.
Cell Metab ; 3(4): 257-66, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16581003

RESUMO

Insulin resistance in diabetes and metabolic syndrome is thought to increase susceptibility to atherosclerotic cardiovascular disease, but the underlying mechanisms are poorly understood. To evaluate the possibility that decreased insulin signaling in macrophage foam cells might worsen atherosclerosis, Ldlr(-/-) mice were transplanted with insulin receptor Insr(+/+) or Insr(-/-) bone marrow. Western diet-fed Insr(-/-) recipients developed larger, more complex lesions with increased necrotic cores and increased numbers of apoptotic cells. Insr(-/-) macrophages showed diminished Akt phosphorylation and an augmented ER stress response, leading to induction of scavenger receptor A and increased apoptosis when challenged with cholesterol loading or nutrient deprivation. These studies suggest that defective insulin signaling and reduced Akt activity impair the ability of macrophages to deal with ER stress-induced apoptosis within atherosclerotic plaques.


Assuntos
Apoptose , Aterosclerose/fisiopatologia , Retículo Endoplasmático/fisiologia , Insulina/fisiologia , Macrófagos/química , Receptor de Insulina/deficiência , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/patologia , Transplante de Medula Óssea , Colesterol/farmacologia , Feminino , Células Espumosas/patologia , Células Espumosas/fisiologia , Expressão Gênica , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Insulina/genética , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Necrose , Proteína Oncogênica v-akt/fisiologia , Fosforilação , Receptor de Insulina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe A/genética , Receptores Depuradores Classe A/fisiologia , Transdução de Sinais
3.
Circ Res ; 105(11): 1072-82, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19834009

RESUMO

RATIONALE: The complications of atherosclerosis are a major cause of death and disability in type 2 diabetes. Defective clearance of apoptotic cells by macrophages (efferocytosis) is thought to lead to increased necrotic core formation and inflammation in atherosclerotic lesions. OBJECTIVE: To determine whether there is defective efferocytosis in a mouse model of obesity and atherosclerosis. METHODS AND RESULTS: We quantified efferocytosis in peritoneal macrophages and in atherosclerotic lesions of obese ob/ob or ob/ob;Ldlr(-/-) mice and littermate controls. Peritoneal macrophages from ob/ob and ob/ob;Ldlr(-/-) mice showed impaired efferocytosis, reflecting defective phosphatidylinositol 3-kinase activation during uptake of apoptotic cells. Membrane lipid composition of ob/ob and ob/ob;Ldlr(-/-) macrophages showed an increased content of saturated fatty acids (FAs) and decreased omega-3 FAs (eicosapentaenoic acid and docosahexaenoic acid) compared to controls. A similar defect in efferocytosis was induced by treating control macrophages with saturated free FA/BSA complexes, whereas the defect in ob/ob macrophages was reversed by treatment with eicosapentaenoic acid/BSA or by feeding ob/ob mice a fish oil diet rich in omega-3 FAs. There was also defective macrophage efferocytosis in atherosclerotic lesions of ob/ob;Ldlr(-/-) mice and this was reversed by a fish oil-rich diet. CONCLUSIONS: The findings suggest that in obesity and type 2 diabetes elevated levels of saturated FAs and/or decreased levels of omega-3 FAs contribute to decreased macrophage efferocytosis. Beneficial effects of fish oil diets in atherosclerotic cardiovascular disease may involve improvements in macrophage function related to reversal of defective efferocytosis and could be particularly important in type 2 diabetes and obesity.


Assuntos
Apoptose/fisiologia , Óleos de Peixe/farmacologia , Macrófagos Peritoneais/fisiologia , Obesidade/dietoterapia , Obesidade/patologia , Fagocitose/fisiologia , Adipocinas/metabolismo , Ração Animal , Animais , Aterosclerose/dietoterapia , Aterosclerose/patologia , Células Cultivadas , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/patologia , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Macrófagos Peritoneais/citologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de LDL/genética
4.
Circ Res ; 100(11): 1546-55, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17556668

RESUMO

The macrophage has emerged as an important player in the pathogenesis of both atherosclerosis and insulin resistance. Cross-talk between inflammatory macrophages and adipocytes may be involved in insulin resistance in peripheral tissues. Defective insulin signaling in cells of the arterial wall including macrophages may promote the development of atherosclerosis. Insulin resistant macrophages are more susceptible to endoplasmic reticulum stress and apoptosis in response to various stimuli such as nutrient deprivation, free cholesterol loading, and oxidized LDL. Increased apoptosis of insulin resistant macrophages and impaired phagocytic clearance of apoptotic cells by insulin resistant macrophages in atherosclerotic lesions may lead to enhanced postapoptotic necrosis, larger lipid-rich cores, increased inflammation, and more complex vulnerable plaques.


Assuntos
Aterosclerose/fisiopatologia , Diabetes Mellitus/fisiopatologia , Resistência à Insulina , Macrófagos/metabolismo , Transdução de Sinais , Animais , Apoptose , Progressão da Doença , Retículo Endoplasmático/metabolismo , Humanos , Inflamação/fisiopatologia , Monócitos/metabolismo
5.
J Clin Invest ; 115(9): 2434-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127462

RESUMO

Direct evidence that hyperglycemia, rather than concomitant increases in known risk factors, induces atherosclerosis is lacking. Most diabetic mice do not exhibit a higher degree of atherosclerosis unless the development of diabetes is associated with more severe hyperlipidemia. We hypothesized that normal mice were deficient in a gene that accelerated atherosclerosis with diabetes. The gene encoding aldose reductase (AR), an enzyme that mediates the generation of toxic products from glucose, is expressed at low levels in murine compared with human tissues. Mice in which diabetes was induced through streptozotocin (STZ) treatment, but not nondiabetic mice, expressing human AR (hAR) crossed with LDL receptor-deficient (Ldlr-/-) C57BL/6 male mice had increased aortic atherosclerosis. Diabetic hAR-expressing heterozygous LDL receptor-knockout mice (Ldlr+/-) fed a cholesterol/cholic acid-containing diet also had increased aortic lesion size. Lesion area at the aortic root was increased by STZ treatment alone but was further increased by hAR expression. Macrophages from hAR-transgenic mice expressed more scavenger receptors and had greater accumulation of modified lipoproteins than macrophages from nontransgenic mice. Expression of genes that regulate regeneration of glutathione was reduced in the hAR-expressing aortas. Thus, hAR increases atherosclerosis in diabetic mice. Inhibitors of AR or other enzymes that mediate glucose toxicity could be useful in the treatment of diabetic atherosclerosis.


Assuntos
Aldeído Redutase/metabolismo , Aterosclerose , Complicações do Diabetes/fisiopatologia , Diabetes Mellitus Experimental , Aldeído Redutase/genética , Animais , Aorta/patologia , Aterosclerose/etiologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Glicemia/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Dieta Aterogênica , Humanos , Lipídeos/sangue , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Fatores de Risco
6.
J Clin Invest ; 113(5): 764-73, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14991075

RESUMO

Accelerated atherosclerosis is a major cause of morbidity and death in insulin-resistant states such as obesity and the metabolic syndrome, but the underlying mechanisms are poorly understood. We show that macrophages from obese (ob/ob) mice have increased binding and uptake of oxidized LDL, in part due to a post-transcriptional increase in CD36 protein. Macrophages from ob/ob mice are also insulin resistant, as shown by reduced expression and signaling of insulin receptors. Three lines of evidence indicate that the increase in CD36 is caused by defective insulin signaling: (a) Treatment of wild-type macrophages with LY294002, an inhibitor of insulin signaling via PI3K, results in an increase in CD36; (b) insulin receptor knockout macrophages show a post-transcriptional increase in CD36 protein; and (c) administration of thiazolidinediones to intact ob/ob mice and ob/ob, LDL receptor-deficient mice results in a reversal of macrophage insulin receptor defects and decreases CD36 protein. The last finding contrasts with the increase in CD36 that results from treatment of macrophages with these drugs ex vivo. The results suggest that defective macrophage insulin signaling predisposes to foam cell formation and atherosclerosis in insulin-resistant states and that this is reversed in vivo by treatment with PPAR-gamma activators.


Assuntos
Antígenos CD36/biossíntese , Insulina/metabolismo , Macrófagos/imunologia , Transdução de Sinais , Animais , Northern Blotting , Western Blotting , Cromonas/farmacologia , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Células Espumosas/metabolismo , Glutationa/metabolismo , Hipoglicemiantes/farmacologia , Resistência à Insulina , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Morfolinas/farmacologia , Oxigênio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Tiazolidinedionas/farmacologia , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Tirosina/metabolismo
7.
Novartis Found Symp ; 286: 99-109; discussion 109-12, 162-3, 196-203, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18269177

RESUMO

Macrophage death in advanced atherosclerosis causes plaque necrosis, which promotes plaque rupture and acute atherothrombotic vascular events. Of interest, plaque necrosis and atherothrombotic disease are markedly increased in diabetes and metabolic syndrome. We discovered a novel 'multi-hit' macrophage apoptosis pathway that appears to be highly relevant to advanced atherosclerosis. The elements of the pathway include: (a) activation of the unfolded protein response (UPR) by cholesterol overloading of the endoplasmic reticulum or by other UPR activators known to exist in atheromata; and (b) pro-apoptotic signalling involving the type A scavenger receptor (SRA). The downstream apoptosis effectors include CHOP (GADD153) for the UPR and JNK for SRA signalling. Remarkably, components of this pathway are enhanced in macrophages with defective insulin signalling, including UPR activation and SRA expression. As a result, insulin-resistant macrophages show increased susceptibility to apoptosis when exposed to UPR activators and SRA ligands. Moreover, the advanced lesions of atherosclerosis-prone mice reconstituted with insulin-resistant macrophages show increased macrophage apoptosis and plaque necrosis. Based on these findings, we propose that one mechanism of increased plaque necrosis and atherothrombotic vascular disease in insulin resistant syndromes is up-regulation of a two-hit signal transduction pathway involved in advanced lesional macrophage death.


Assuntos
Aterosclerose/metabolismo , Resistência à Insulina , Macrófagos/metabolismo , Transdução de Sinais , Animais , Apoptose , Humanos , Macrófagos/citologia , Modelos Biológicos , Receptores Depuradores Classe A/metabolismo
8.
Diabetes ; 61(10): 2609-20, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22751695

RESUMO

Accumulation of toxic lipids evokes the unfolded protein response (UPR) and apoptotic death of macrophages and vascular cells in atherosclerotic plaques. Primary macrophages from insulin-resistant ob/ob and insulin receptor (Insr)(-/-) mice display increased apoptosis in response to loading with free cholesterol or oxysterol, but underlying mechanisms have not been elucidated. We show increased activation of all three major branches of the UPR in response to free cholesterol or oxysterol loading in insulin-resistant macrophages. Inhibition and rescue experiments revealed that defective MEK/extracellular signal\x{2013}related kinase (ERK)/cAMP-responsive element-binding protein (CREBP) signaling in insulin-resistant macrophages leads to decreased expression of sarcoplasmic endoplasmic reticulum (ER) Ca(2+)-ATPase, depletion of ER calcium stores, PKR-like ER kinase activation, and ER stress-associated apoptosis. Activation of macrophage glucagon-like peptide 1 (GLP-1) receptor via the antidiabetic drug exenatide led to improvements in both ERK and AKT signaling and reversed the increase in UPR and apoptosis of insulin-resistant macrophages in atherosclerotic lesions of ob/ob.Ldlr(-/-) and Insr(-/-).Ldlr(-/-) mice. Increased signaling via GLP-1 receptor or the CREBP activator protein kinase A thus offers a way to rescue insulin-resistant macrophages from excessive ER stress responses and apoptosis in insulin resistance and type 2 diabetes.


Assuntos
Apoptose/genética , Estresse do Retículo Endoplasmático/genética , Hipoglicemiantes/farmacologia , Resistência à Insulina/genética , Macrófagos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Peptídeos/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Peçonhas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Exenatida , Insulina/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Camundongos Obesos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Resposta a Proteínas não Dobradas/genética
9.
Mol Cell Biol ; 31(20): 4205-18, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21844222

RESUMO

It has been shown that inhibition of de novo sphingolipid synthesis increases insulin sensitivity. For further exploration of the mechanism involved, we utilized two models: heterozygous serine palmitoyltransferase (SPT) subunit 2 (Sptlc2) gene knockout mice and sphingomyelin synthase 2 (Sms2) gene knockout mice. SPT is the key enzyme in sphingolipid biosynthesis, and Sptlc2 is one of its subunits. Homozygous Sptlc2-deficient mice are embryonic lethal. However, heterozygous Sptlc2-deficient mice that were viable and without major developmental defects demonstrated decreased ceramide and sphingomyelin levels in the cell plasma membranes, as well as heightened sensitivity to insulin. Moreover, these mutant mice were protected from high-fat diet-induced obesity and insulin resistance. SMS is the last enzyme for sphingomyelin biosynthesis, and SMS2 is one of its isoforms. Sms2 deficiency increased cell membrane ceramide but decreased SM levels. Sms2 deficiency also increased insulin sensitivity and ameliorated high-fat diet-induced obesity. We have concluded that Sptlc2 heterozygous deficiency- or Sms2 deficiency-mediated reduction of SM in the plasma membranes leads to an improvement in tissue and whole-body insulin sensitivity.


Assuntos
Membrana Celular/metabolismo , Resistência à Insulina , Insulina/metabolismo , Esfingomielinas/metabolismo , Animais , Teste de Tolerância a Glucose , Células Hep G2 , Heterozigoto , Humanos , Insulina/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptor de Insulina/metabolismo , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/metabolismo , Esfingomielinas/análise , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo
10.
J Clin Invest ; 119(4): 1029-41, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19273907

RESUMO

Type 2 diabetes is associated with accelerated atherogenesis, which may result from a combination of factors, including dyslipidemia characterized by increased VLDL secretion, and insulin resistance. To assess the hypothesis that both hepatic and peripheral insulin resistance contribute to atherogenesis, we crossed mice deficient for the LDL receptor (Ldlr-/- mice) with mice that express low levels of IR in the liver and lack IR in peripheral tissues (the L1B6 mouse strain). Unexpectedly, compared with Ldlr-/- controls, L1B6Ldlr-/- mice fed a Western diet showed reduced VLDL and LDL levels, reduced atherosclerosis, decreased hepatic AKT signaling, decreased expression of genes associated with lipogenesis, and diminished VLDL apoB and lipid secretion. Adenovirus-mediated hepatic expression of either constitutively active AKT or dominant negative glycogen synthase kinase (GSK) markedly increased VLDL and LDL levels such that they were similar in both Ldlr-/- and L1B6Ldlr-/- mice. Knocking down expression of hepatic IR by adenovirus-mediated shRNA decreased VLDL triglyceride and apoB secretion in Ldlr-/- mice. Furthermore, knocking down hepatic IR expression in either WT or ob/ob mice reduced VLDL secretion but also resulted in decreased hepatic Ldlr protein. These findings suggest a dual action of hepatic IR on lipoprotein levels, in which the ability to increase VLDL apoB and lipid secretion via AKT/GSK is offset by upregulation of Ldlr.


Assuntos
Aterosclerose/etiologia , Insulina/metabolismo , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Animais , Apolipoproteínas B/sangue , Apolipoproteínas B/metabolismo , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/metabolismo , Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Lipídeos/sangue , Lipogênese , Lipoproteínas VLDL/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Obesos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
11.
Diabetes ; 57(11): 2967-76, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18728232

RESUMO

OBJECTIVE: Endoplasmic reticulum stress increases macrophage apoptosis, contributing to the complications of atherosclerosis. Insulin-resistant macrophages are more susceptible to endoplasmic reticulum stress-associated apoptosis probably contributing to macrophage death and necrotic core formation in atherosclerotic plaques in type 2 diabetes. However, the molecular mechanisms of increased apoptosis in insulin-resistant macrophages remain unclear. RESEARCH DESIGN AND METHODS: The studies were performed in insulin-resistant macrophages isolated from insulin receptor knockout or ob/ob mice. Gain- or loss-of-function approaches were used to evaluate the roles of forkhead transcription factors (FoxOs) in endoplasmic reticulum stress-associated macrophage apoptosis. RESULTS: Insulin-resistant macrophages showed attenuated Akt activation and increased nuclear localization of FoxO1 during endoplasmic reticulum stress induced by free cholesterol loading. Overexpression of active FoxO1 or FoxO3 failed to induce apoptosis in unchallenged macrophages but exacerbated apoptosis in macrophages with an active endoplasmic reticulum stress response. Conversely, macrophages with genetic knockouts of FoxO1, -3, and -4 were resistant to apoptosis in response to endoplasmic reticulum stress. FoxO1 was shown by chromatin immunoprecipitation and promoter expression analysis to induce inhibitor of kappaBepsilon gene expression and thereby to attenuate the increase of nuclear p65 and nuclear factor-kappaB activity during endoplasmic reticulum stress, with proapoptotic and anti-inflammatory consequences. CONCLUSIONS: Decreased Akt and increased FoxO transcription factor activity during the endoplasmic reticulum stress response leads to increased apoptosis of insulin-resistant macrophages. FoxOs may have a dual cellular function, resulting in either proapoptotic or anti-inflammatory effects in an endoplasmic reticulum stress-modulated manner. In the complex plaque milieu, the ultimate effect is likely to be an increase in macrophage apoptosis, plaque inflammation, and destabilization.


Assuntos
Apoptose/fisiologia , Retículo Endoplasmático/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Macrófagos Peritoneais/metabolismo , Adenoviridae/genética , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Colesterol/farmacologia , Imunoprecipitação da Cromatina , Imunofluorescência , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Resistência à Insulina/genética , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Transfecção
12.
J Biol Chem ; 278(8): 5813-20, 2003 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-12482877

RESUMO

Liver X receptor/retinoid X receptor (LXR/RXR) transcription factors have been found to induce a number of genes involved in the regulation of cellular cholesterol efflux, including the ATP-binding cassette transporter A1 (ABCA1), which mediates the active efflux of cellular cholesterol and phospholipids to extracellular acceptors, such as apolipoprotein A-I (apoA-I). In a screen for macrophage LXR/RXR target genes, we identified stearoyl-CoA desaturases 1 and 2 (Scd1 and Scd2), and subsequently tested the hypothesis that SCD activity might modulate cellular cholesterol efflux. In HEK 293 cells co-transfection of ABCA1 with either SCD1 or SCD2 inhibited ABCA1-mediated cholesterol efflux but not phospholipid efflux. In Chinese hamster ovary (CHO) cells with moderate stable overexpression of SCD1, cholesterol efflux to apoA-I was inhibited by 73%, whereas phospholipid efflux and ABCA1 protein levels were unchanged. In contrast, cholesterol efflux to HDL(2), which is not dependent on ABCA1, was increased 2-fold in CHO-SCD1 cells. The effect of SCD on cholesterol efflux to apoA-I was independent of acyl-CoA:cholesterol acyltransferase (ACAT) activity. SCD activity led to an increased content of plasma membrane monounsaturated fatty acids (18:1) at the expense of saturated fatty acids (18:0). As shown by confocal microscopy, SCD overexpression led to a decrease of Triton X-100-resistant domains in the plasma membrane, indicating a decrease in membrane-ordered regions. The data suggest that SCD changes membrane organization and depletes a specific pool of membrane cholesterol supporting ABCA1-mediated efflux, whereas increasing availability of cholesterol for passive efflux by HDL(2). ABCA1-mediated cholesterol and phospholipid efflux may be uncoupled in pathological states associated with high SCD activity, as in hyperinsulinemic obese mice, or in animals treated with LXR activators.


Assuntos
Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Membrana Celular/ultraestrutura , Colesterol/metabolismo , Receptores do Ácido Retinoico/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Células CHO , Linhagem Celular , Células Cultivadas , Cricetinae , Regulação da Expressão Gênica , Humanos , Isoenzimas/metabolismo , Cinética , Fosfolipídeos/metabolismo , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Receptores X de Retinoides , Estearoil-CoA Dessaturase/genética , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa