Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Genomics ; 12: 96, 2011 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-21291556

RESUMO

BACKGROUND: Divergently selected Lean and Fat mouse lines represent unique models for a polygenic form of resistance and susceptibility to obesity development. Previous research on these lines focused mainly on obesity-susceptible factors in the Fat line. This study aimed to examine the molecular basis of obesity-resistant mechanisms in the Lean line by analyzing various fat depots and organs, the liver transcriptome of selected metabolic pathways, plasma and lipid homeostasis and expression of selected skeletal muscle genes. RESULTS: Expression profiling using our custom Steroltalk v2 microarray demonstrated that Lean mice exhibit a higher hepatic expression of cholesterol biosynthesis genes compared to the Fat line, although this was not reflected in elevation of total plasma or liver cholesterol. However, FPLC analysis showed that protective HDL cholesterol was elevated in Lean mice. A significant difference between the strains was also found in bile acid metabolism. Lean mice had a higher expression of Cyp8b1, a regulatory enzyme of bile acid synthesis, and the Abcb11 bile acid transporter gene responsible for export of acids to the bile. Additionally, a higher content of blood circulating bile acids was observed in Lean mice. Elevated HDL and upregulation of some bile acids synthesis and transport genes suggests enhanced reverse cholesterol transport in the Lean line--the flux of cholesterol out of the body is higher which is compensated by upregulation of endogenous cholesterol biosynthesis. Increased skeletal muscle Il6 and Dio2 mRNA levels as well as increased activity of muscle succinic acid dehydrogenase (SDH) in the Lean mice demonstrates for the first time that changes in muscle energy metabolism play important role in the Lean line phenotype determination and corroborate our previous findings of increased physical activity and thermogenesis in this line. Finally, differential expression of Abcb11 and Dio2 identifies novel strong positional candidate genes as they map within the quantitative trait loci (QTL) regions detected previously in crosses between the Lean and Fat mice. CONCLUSION: We identified novel candidate molecular targets and metabolic changes which can at least in part explain resistance to obesity development in the Lean line. The major difference between the Lean and Fat mice was in increased liver cholesterol biosynthesis gene mRNA expression, bile acid metabolism and changes in selected muscle genes' expression in the Lean line. The liver Abcb11 and muscle Dio2 were identified as novel positional candidate genes to explain part of the phenotypic difference between the Lean and Fat lines.


Assuntos
Fígado/metabolismo , Músculo Esquelético/metabolismo , Obesidade/genética , Magreza/genética , Animais , Perfilação da Expressão Gênica , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Locos de Características Quantitativas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
J Biol Chem ; 285(47): 36759-67, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20851881

RESUMO

The bile acid receptor farnesoid X receptor (FXR) is expressed in adipose tissue, but its function remains poorly defined. Peroxisome proliferator-activated receptor-γ (PPARγ) is a master regulator of adipocyte differentiation and function. The aim of this study was to analyze the role of FXR in adipocyte function and to assess whether it modulates PPARγ action. Therefore, we tested the responsiveness of FXR-deficient mice (FXR(-/-)) and cells to the PPARγ activator rosiglitazone. Our results show that genetically obese FXR(-/-)/ob/ob mice displayed a resistance to rosiglitazone treatment. In vitro, rosiglitazone treatment did not induce normal adipocyte differentiation and lipid droplet formation in FXR(-/-) mouse embryonic fibroblasts (MEFs) and preadipocytes. Moreover, FXR(-/-) MEFs displayed both an increased lipolysis and a decreased de novo lipogenesis, resulting in reduced intracellular triglyceride content, even upon PPARγ activation. Retroviral-mediated FXR re-expression in FXR(-/-) MEFs restored the induction of adipogenic marker genes during rosiglitazone-forced adipocyte differentiation. The expression of Wnt/ß-catenin pathway and target genes was increased in FXR(-/-) adipose tissue and MEFs. Moreover, the expression of several endogenous inhibitors of this pathway was decreased early during the adipocyte differentiation of FXR(-/-) MEFs. These findings demonstrate that FXR regulates adipocyte differentiation and function by regulating two counteracting pathways of adipocyte differentiation, the PPARγ and Wnt/ß-catenin pathways.


Assuntos
Adipócitos/citologia , Diferenciação Celular , PPAR gama/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Resistência a Medicamentos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteína-1 Reguladora de Fusão , Perfilação da Expressão Gênica , Humanos , Hipoglicemiantes/farmacologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Lipólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Análise de Sequência com Séries de Oligonucleotídeos , PPAR gama/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Transdução de Sinais , Tiazolidinedionas/farmacologia , Proteínas Wnt/genética , beta Catenina/genética
3.
Mol Cell Biol ; 30(12): 3059-70, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20385766

RESUMO

The circadian timing system coordinates many aspects of mammalian physiology and behavior in synchrony with the external light/dark cycle. These rhythms are driven by endogenous molecular clocks present in most body cells. Many clock outputs are transcriptional regulators, suggesting that clock genes primarily control physiology through indirect pathways. Here, we show that Krüppel-like factor 10 (KLF10) displays a robust circadian expression pattern in wild-type mouse liver but not in clock-deficient Bmal1 knockout mice. Consistently, the Klf10 promoter recruited the BMAL1 core clock protein and was transactivated by the CLOCK-BMAL1 heterodimer through a conserved E-box response element. Profiling the liver transcriptome from Klf10(-/-) mice identified 158 regulated genes with significant enrichment for transcripts involved in lipid and carbohydrate metabolism. Importantly, approximately 56% of these metabolic genes are clock controlled. Male Klf10(-/-) mice displayed postprandial and fasting hyperglycemia, a phenotype accompanied by a significant time-of-day-dependent upregulation of the gluconeogenic gene Pepck and increased hepatic glucose production. Consistently, functional data showed that the proximal Pepck promoter is repressed directly by KLF10. Klf10(-/-) females were normoglycemic but displayed higher plasma triglycerides. Correspondingly, rhythmic gene expression of components of the lipogenic pathway, including Srebp1c, Fas, and Elovl6, was altered in females. Collectively, these data establish KLF10 as a required circadian transcriptional regulator that links the molecular clock to energy metabolism in the liver.


Assuntos
Relógios Biológicos/genética , Ritmo Circadiano/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Animais , Linhagem Celular , Jejum , Comportamento Alimentar , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genoma/genética , Glucose/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Lipogênese/genética , Masculino , Metaboloma/genética , Camundongos , Regiões Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
PLoS One ; 5(1): e8722, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20090943

RESUMO

BACKGROUND: Liver-selective thyromimetics have been reported to efficiently reduce plasma cholesterol through the hepatic induction of both, the low-density lipoprotein receptor (LDLr) and the high-density lipoprotein (HDL) receptor; the scavenger receptor class B type I (SR-BI). Here, we investigated the effect of the thyromimetic T-0681 on reverse cholesterol transport (RCT) and atherosclerosis, and studied the underlying mechanisms using different mouse models, including mice lacking LDLr, SR-BI, and apoE, as well as CETP transgenic mice. METHODOLOGY/PRINCIPAL FINDINGS: T-0681 treatment promoted bile acid production and biliary sterol secretion consistently in the majority of the studied mouse models, which was associated with a marked reduction of plasma cholesterol. Using an assay of macrophage RCT in mice, we found T-0681 to significantly increase fecal excretion of macrophage-derived neutral and acidic sterols. No positive effect on RCT was found in CETP transgenic mice, most likely due to the observed decrease in plasma CETP mass. Studies in SR-BI KO and LDLr KO mice suggested hepatic LDLr to be necessary for the action of T-0681 on lipid metabolism, as the compound did not have any influence on plasma cholesterol levels in mice lacking this receptor. Finally, prolonged treatment with T-0681 reduced the development of atherosclerosis by 60% in apoE KOs on Western type diet. In contrast, at an earlier time-point T-0681 slightly increased small fatty streak lesions, in part due to an impaired macrophage cholesterol efflux capacity, when compared to controls. CONCLUSIONS/SIGNIFICANCE: The present results show that liver-selective thyromimetics can promote RCT and that such compounds may protect from atherosclerosis partly through induction of bile acid metabolism and biliary sterol secretion. On-going clinical trials will show whether selective thyromimetics do prevent atherosclerosis also in humans.


Assuntos
Aterosclerose/prevenção & controle , Colesterol/metabolismo , Fígado/efeitos dos fármacos , Malonatos/farmacologia , Éteres Fenílicos/farmacologia , Animais , Transporte Biológico , Modelos Animais de Doenças , Fígado/metabolismo , Camundongos , Camundongos Transgênicos
5.
Arterioscler Thromb Vasc Biol ; 30(2): 232-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19910634

RESUMO

INTRODUCTION: Apolipoprotein (apo) A-II is the second most abundant high-density lipoprotein (HDL) apolipoprotein. We assessed the mechanism involved in the altered postprandial triglyceride-rich lipoprotein metabolism of female human apoA-II-transgenic mice (hapoA-II-Tg mice), which results in up to an 11-fold increase in plasma triglyceride concentration. The relationships between apoA-II, HDL composition, and lipoprotein lipase (LPL) activity were also analyzed in a group of normolipidemic women. METHODS AND RESULTS: Triglyceride-rich lipoprotein catabolism was decreased in hapoA-II-Tg mice compared to control mice. This suggests that hapoA-II, which was mainly associated with HDL during fasting and postprandially, impairs triglyceride-rich lipoprotein lipolysis. HDL isolated from hapoA-II-Tg mice impaired bovine LPL activity. Two-dimensional gel electrophoresis, mass spectrometry, and immunonephelometry identified a marked deficiency in the HDL content of apoA-I, apoC-III, and apoE in these mice. In normolipidemic women, apoA-II concentration was directly correlated with plasma triglyceride and inversely correlated with the HDL-apoC-II+apoE/apoC-III ratio [corrected]. HDL-mediated induction of LPL activity was inversely correlated with apoA-II and directly correlated with the HDL-apoC-II+apoE/apoC-III ratio [corrected]. Purified hapoA-II displaced apoC-II, apoC-III, and apoE from human HDL2. Human HDL3 was, compared to HDL2, enriched in apoA-II but poorer in apoC-II, apoC-III, and apoE. CONCLUSIONS: ApoA-II plays a crucial role in triglyceride catabolism by regulating LPL activity, at least in part, through HDL proteome modulation.


Assuntos
Apolipoproteína A-II/sangue , Lipólise , Lipase Lipoproteica/sangue , Lipoproteínas HDL2/sangue , Lipoproteínas HDL3/sangue , Triglicerídeos/sangue , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-II/genética , Apolipoproteína C-II/sangue , Apolipoproteína C-III/sangue , Apolipoproteínas E/sangue , Biomarcadores/sangue , Gorduras na Dieta/metabolismo , Eletroforese em Gel Bidimensional , Feminino , Humanos , Intestinos/enzimologia , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nefelometria e Turbidimetria , Período Pós-Prandial , Proteoma , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fatores de Tempo , Regulação para Cima
6.
Curr Opin Lipidol ; 20(6): 505-11, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19829109

RESUMO

PURPOSE OF REVIEW: Despite the fact that statin treatment substantially reduces cardiovascular morbidity and mortality, many treated patients still experience a high residual risk. Statins lower LDL cholesterol (LDL-C), with limited effects on other lipid parameters. Fibrates improve atherogenic dyslipidemia characterized by high triglyceride and/or low HDL cholesterol levels and elevated concentrations of small dense LDL particles, with or without high LDL-C levels. Fibrates decrease cardiovascular morbidity, especially in patients with the metabolic syndrome. The purpose of this review is to provide a rationale for the combined use of statins and fibrates in the management of patients with high residual cardiovascular risk related to atherogenic dyslipidemia and persisting after single therapy. RECENT FINDINGS: A meta-analysis from 14 randomized trials conducted in high-risk patients reported that statin therapy is effective in reducing the proportional risk for major vascular events by 21% for each mmol/l lowering of LDL-C. However, on an average, 14% of patients still experienced an event despite being allocated to statin. Beyond LDL-C, other factors, including triglycerides, non-HDL cholesterol, HDL cholesterol, and apolipoprotein B, have been identified as factors determining residual risk, and normalization of these parameters may further decrease cardiovascular disease in patients treated with statins. Data from fibrate trials indicate that these drugs are particularly effective in reducing cardiovascular morbidity in patients with atherogenic dyslipidemia. SUMMARY: Reducing the residual cardiovascular risk in patients treated with statins requires addressing multiple lipid goals. In this context, future therapeutic interventions based on combination therapy, such as statins and fibrates, appear particularly promising.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Ácido Clofíbrico/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Doenças Cardiovasculares/sangue , LDL-Colesterol/sangue , Quimioterapia Combinada , Humanos , Hipolipemiantes/uso terapêutico , Metanálise como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
7.
Arterioscler Thromb Vasc Biol ; 29(10): 1488-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19592467

RESUMO

OBJECTIVE: Bexarotene (Targretin) is a clinically used antitumoral agent which exerts its action through binding to and activation of the retinoid-X-receptor (RXR). The most frequent side-effect of bexarotene administration is an increase in plasma triglycerides, an independent risk factor of cardiovascular disease. The molecular mechanism behind this hypertriglyceridemia remains poorly understood. METHODS AND RESULTS: Using wild-type and LXR alpha/beta-deficient mice, we show here that bexarotene induces hypertriglyceridemia and activates hepatic LXR-target genes of lipogenesis in an LXR-dependent manner, hence exerting a permissive effect on RXR/LXR heterodimers. Interestingly, RNA analysis and Chromatin Immunoprecipitation assays performed in the liver reveal that the in vivo permissive effect of bexarotene on the RXR/LXR heterodimer is restricted to lipogenic genes without modulation of genes controlling cholesterol homeostasis. CONCLUSIONS: These findings demonstrate that the hypertriglyceridemic action of bexarotene occurs via the RXR/LXR heterodimer and show that RXR heterodimers can act with a selective permissivity on target genes of specific metabolic pathways in the liver.


Assuntos
Colesterol/metabolismo , Proteínas de Ligação a DNA/fisiologia , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores X de Retinoides/fisiologia , Tetra-Hidronaftalenos/farmacologia , Triglicerídeos/metabolismo , Animais , Bexaroteno , Proteínas de Ligação a DNA/química , Dimerização , Feminino , Homeostase , Lipogênese , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Receptores Nucleares Órfãos , Receptores Citoplasmáticos e Nucleares/química , Receptores X de Retinoides/química
8.
Curr Atheroscler Rep ; 11(4): 281-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19500491

RESUMO

The greatest clinical challenge in type 2 diabetes mellitus is the prevention of its long-term complications, many of which are of a cardiovascular nature. Despite the progress in cardiovascular risk management of diabetes patients using lipid-lowering and antihypertensive drugs, a substantial residual risk persists. Indeed, treated diabetes patients have a similar risk as untreated nondiabetic individuals. Although glycemic control through the use of antihyperglycemic agents improves microvascular complications, macrovascular disease risk is not reduced. These observations point to the need for additional therapeutic approaches in order to better control global cardiovascular risk. The peroxisome proliferator-activated receptor (PPAR) family members play major roles in the regulation of lipid and glucose metabolism and immune-inflammatory processes, making these transcription factors ideal targets for such therapeutic strategies. This review discusses our current knowledge of the effectiveness of PPAR-based therapeutics, focusing exclusively on cardiovascular disease in type 2 diabetes mellitus and the future prospects for novel generation of PPAR agonists.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Diabetes Mellitus Tipo 2/complicações , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipoglicemiantes/uso terapêutico , Hipolipemiantes/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/fisiologia
9.
Arterioscler Thromb Vasc Biol ; 29(4): 548-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19201688

RESUMO

OBJECTIVE: The ATP-binding cassette transporter, subfamily A, member 1 (ABCA1) plays a key role in HDL cholesterol metabolism. However, the role of ABCA1 in modulating susceptibility to atherosclerosis is controversial. METHODS AND RESULTS: We investigated the role of ABCA1 in atherosclerosis using a combination of overexpression and selective deletion models. First, we examined the effect of transgenic overexpression of a full-length human ABCA1-containing bacterial artificial chromosome (BAC) in the presence or absence of the endogenous mouse Abca1 gene. ABCA1 overexpression in the atherosclerosis-susceptible Ldlr(-/-) background significantly reduced the development of atherosclerosis in both the presence and absence of mouse Abca1. Next, we used mice with tissue-specific inactivation of Abca1 to dissect the discrete roles of Abca1 in different tissues on susceptibility to atherosclerosis. On the Apoe(-/-) background, mice lacking hepatic Abca1 had significantly reduced HDL cholesterol and accelerated atherosclerosis, indicating that the liver is an important site at which Abca1 plays an antiatherogenic role. In contrast, mice with macrophage-specific inactivation of Abca1 on the Ldlr(-/-) background displayed no change in atherosclerotic lesion area. CONCLUSIONS: These data indicate that physiological expression of Abca1 modulates the susceptibility to atherosclerosis and establish hepatic Abca1 expression as an important site of atheroprotection. In contrast, we show that selective deletion of macrophage Abca1 does not significantly modulate atherogenesis.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Aterosclerose/metabolismo , HDL-Colesterol/metabolismo , Fígado/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , HDL-Colesterol/sangue , LDL-Colesterol/metabolismo , VLDL-Colesterol/metabolismo , Cromossomos Artificiais Bacterianos , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Receptores de LDL/deficiência , Receptores de LDL/genética
10.
Arterioscler Thromb Vasc Biol ; 29(3): 341-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19095997

RESUMO

OBJECTIVE: Absence of stearoyl-CoA desaturase-1 (SCD1) in mice reduces plasma triglycerides and provides protection from obesity and insulin resistance, which would be predicted to be associated with reduced susceptibility to atherosclerosis. The aim of this study was to determine the effect of SCD1 deficiency on atherosclerosis. METHODS AND RESULTS: Despite an antiatherogenic metabolic profile, SCD1 deficiency increases atherosclerosis in hyperlipidemic low-density lipoprotein receptor (LDLR)-deficient mice challenged with a Western diet. Lesion area at the aortic root is significantly increased in males and females in two models of SCD1 deficiency. Inflammatory changes are evident in the skin of these mice, including increased intercellular adhesion molecule (ICAM)-1 and ulcerative dermatitis. Increases in ICAM-1 and interleukin-6 are also evident in plasma of SCD1-deficient mice. HDL particles demonstrate changes associated with inflammation, including decreased plasma apoA-II and apoA-I and paraoxonase-1 and increased plasma serum amyloid A. Lipopolysaccharide-induced inflammatory response and cholesterol efflux are not altered in SCD1-deficient macrophages. In addition, when SCD1 deficiency is limited to bone marrow-derived cells, lesion size is not altered in LDLR-deficient mice. CONCLUSIONS: These studies reinforce the crucial role of chronic inflammation in promoting atherosclerosis, even in the presence of antiatherogenic biochemical and metabolic characteristics.


Assuntos
Aterosclerose/enzimologia , Hiperlipidemias/enzimologia , Inflamação/enzimologia , Estearoil-CoA Dessaturase/deficiência , Animais , Apolipoproteínas/sangue , Arildialquilfosfatase/sangue , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Hiperlipidemias/genética , Hiperlipidemias/imunologia , Hiperlipidemias/patologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/sangue , Molécula 1 de Adesão Intercelular/sangue , Interleucina-6/sangue , Lipoproteínas HDL/sangue , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética , Proteína Amiloide A Sérica/metabolismo , Úlcera Cutânea/enzimologia , Úlcera Cutânea/patologia , Estearoil-CoA Dessaturase/genética , Fatores de Tempo
11.
Clin Chim Acta ; 401(1-2): 37-41, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19036342

RESUMO

BACKGROUND: Rosiglitazone is an agonist of the peroxisome proliferator-activated receptor (PPAR) gamma that may modify HDL metabolism in humans, but this effect has not been completely elucidated. Therefore, we determined the effect of rosiglitazone on apo AI turnover, HDL structure, and PON1 plasma activity. METHODS: Kinetic studies of HDL-apo AI radiolabeled with (125)I were performed in 7 chow-fed, male, New Zealand white rabbits after 6 weeks of 0.32 mg/kg/d rosiglitazone-treatment vs. vehicle-treated rabbits (n=11). HDL size distribution was determined by polyacrylamide gradient electrophoresis and paraoxonase-1 (PON1); plasma activity was assessed spectrophotometrically using phenylacetate as substrate. RESULTS: Fractional catabolic rate (FCR) of HDL apo AI was higher in the rosiglitazone-treated group than in the control group (0.031+/-0.004 vs. 0.025+/-0.006 pools/h, respectively, p<0.05). The mean apo AI production rate (PR) was 62% higher in the rosiglitazone group as compared to controls (0.918+/-0.238 vs. 0.564+/-0.160 mg/kg/h, p<0.01). Accordingly, apo AI plasma levels in rosiglitazone-treated animals were about 37% higher than in the control group. Rosiglitazone-induced changes in apo AI turnover appeared concomitantly with a significant increase of phospholipids and a decrease in colesteryl esters content of the HDL. Compositional changes resulted in a relative increase of the HDL3b and HDL3c subfractions and a significant enhancement of the plasma PON1 activity (488.5+/-138.2 vs. 595.2+/-179.4 micromol/min/ml, p<0.05). CONCLUSIONS: Rosiglitazone increased apo AI plasma concentrations, resulting from an enhancement of apo AI synthesis, and induced the synthesis of smaller HDL particles with a concomitant increase of plasma PON1 activity. These modifications may contribute to the anti-atherogenic potential of rosiglitazone.


Assuntos
Apolipoproteína A-I/metabolismo , HDL-Colesterol/metabolismo , Hipoglicemiantes/farmacologia , Tiazolidinedionas/farmacologia , Animais , Apolipoproteína A-I/biossíntese , Apolipoproteína A-I/sangue , Arildialquilfosfatase/sangue , Glicemia/análise , HDL-Colesterol/biossíntese , HDL-Colesterol/sangue , Masculino , PPAR gama/agonistas , Coelhos , Rosiglitazona , Triglicerídeos/sangue
12.
J Lipid Res ; 50(5): 938-44, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19106072

RESUMO

This report describes studies in hyperlipidemic New Zealand White (NZW) rabbits investigating the impact of the liver-selective thyromimetic T-0681 on lipoprotein metabolism and the development of atherosclerosis. Prolonged treatment with T-0681 increased the hepatic expression of both LDL receptor and scavenger receptor class B, type I without affecting cholesteryl ester transfer protein activity. Upregulation of hepatic lipoprotein receptors was accompanied by a marked decrease of apolipoprotein B-containing lipoproteins, reflected by a 60% reduction of plasma cholesterol and a >70% reduction of plasma triglyceride levels. Most importantly, T-0681 reduced the development of atherosclerosis by 80% in NZW rabbits on high-cholesterol chow. Our data suggest that liver-selective thyromimetics, such as T-0681, may prove to be useful therapeutic agents against the development of atherosclerosis in humans.


Assuntos
Anticolesterolemiantes/uso terapêutico , Aterosclerose/prevenção & controle , Malonatos/uso terapêutico , Éteres Fenílicos/uso terapêutico , Animais , Aorta/anatomia & histologia , Aorta/patologia , Antígenos CD36/metabolismo , Linhagem Celular , Colesterol na Dieta , Dieta , Modelos Animais de Doenças , Humanos , Hiperlipidemias/tratamento farmacológico , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Coelhos
13.
Circ Res ; 103(5): 450-3, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18658049

RESUMO

Hypertriglyceridemia is an independent risk factor for coronary artery disease. Because apolipoprotein (Apo)A5 regulates plasma triglyceride levels, we investigated the impact of human (h)ApoA5 on atherogenesis. The influence of hApoA5 transgenic expression was studied in the ApoE2 knock-in mouse model of mixed dyslipidemia. Our results demonstrate that hApoA5 lowers plasma triglyceride levels in Western diet-fed ApoE2 knock-in mice. Moreover, atherosclerotic lesion development was significantly decreased in the hApoA5 transgenic mice. Finally, pharmacologic activation of hApoA5 expression by the peroxisome proliferator-activated receptor-alpha agonist fenofibrate resulted in an enhanced atheroprotection. These results identify an atheroprotective role of hApoA5 in a mouse model of mixed dyslipidemia.


Assuntos
Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Aterosclerose/fisiopatologia , Dislipidemias/fisiopatologia , Triglicerídeos/sangue , Ração Animal , Animais , Apolipoproteína A-V , Apolipoproteína E2/sangue , Apolipoproteína E2/genética , Aterosclerose/metabolismo , Colesterol/sangue , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Dislipidemias/tratamento farmacológico , Dislipidemias/metabolismo , Feminino , Fenofibrato/farmacologia , Homeostase/fisiologia , Humanos , Hipolipemiantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
14.
J Lipid Res ; 49(9): 1904-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18509196

RESUMO

Liver X receptor (LXR) agonists increase both total fecal sterol excretion and macrophage-specific reverse cholesterol transport (RCT) in vivo. In this study, we assessed the effects of ABCG5/G8 deficiency as well as those of LXR agonist-induction of RCT from macrophages to feces in vivo. A [(3)H]cholesterol-labeled macrophage cell line was injected intraperitoneally into ABCG5/G8-deficient (G5/G8(-/-)), heterozygous (G5G8(+/-)), and wild-type G5/G8(+/+) mice. G5/G8(-/-)mice presented increased radiolabeled HDL-bound [(3)H]cholesterol 24 h after the label injection. However, the magnitude of macrophage-derived [(3)H]cholesterol in liver and feces did not differ between groups. A separate experiment was conducted in G5G8(+/+) and G5G8(-/-) mice treated with or without the LXR agonist T0901317. Treatment with T0901317 increased liver ABCG5/G8 expression, which was associated with a 2-fold increase in macrophage-derived [(3)H]cholesterol in feces of G5/G8(+/+) mice. However, T0901317 treatment had no effect on fecal [(3)H]cholesterol excretion in G5G8(-/-) mice. Additionally, LXR activation stimulated the fecal excretion of labeled cholesterol after an intravenous injection of HDL-[(3)H]cholesteryl oleate in G5/G8(+/+) mice, but failed to enhance fecal [(3)H]cholesterol in G5/G8(-/-) mice. Our data provide direct in vivo evidence of the crucial role of ABCG5 and ABCG8 in LXR-mediated induction of macrophage-specific RCT.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Colesterol/metabolismo , Proteínas de Ligação a DNA/fisiologia , Fezes/química , Lipoproteínas/fisiologia , Macrófagos/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Transporte Biológico , Feminino , Lipoproteínas/deficiência , Receptores X do Fígado , Masculino , Camundongos , Receptores Nucleares Órfãos
15.
Endocrinology ; 149(7): 3708-12, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18388200

RESUMO

The aim of the study was to investigate the influence of severe hyperthyroidism on plasma high-density lipoprotein cholesterol (HDL-C). Recently, it was shown in mice that increasing doses of T(3) up-regulate hepatic expression of scavenger receptor class B, type I, resulting in increased clearance of plasma HDL-C. Here, we show that severe hyperthyroidism in mice did not affect hepatic expression of scavenger receptor class B, type I, but reduced hepatic expression of ATP-binding cassette transporter 1, accompanied by a 40% reduction of HDL-C. The sterol content of bile, liver, and feces was markedly increased, accompanied by up-regulation of hepatic cholesterol 7alpha-hydroxylase, and ATP-binding cassette transporter 5, which is known to promote biliary sterol secretion upon dimerization with ATP-binding cassette transporter 8. Both control and hyperthyroid mice exerted identical plasma clearance of iv injected [(3)H]HDL-C, supporting the view that severe hyperthyroidism does not affect HDL-C clearance but, rather, its formation via hepatic ATP-binding cassette transporter 1.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , HDL-Colesterol/sangue , Hipertireoidismo/fisiopatologia , Fígado/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Bile/química , Western Blotting , Colesterol/metabolismo , Fezes/química , Expressão Gênica , Hipertireoidismo/sangue , Hipertireoidismo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
J Lipid Res ; 49(1): 217-29, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17960025

RESUMO

A combination of the interrelated metabolic risk factors obesity, insulin resistance, dyslipidemia, and hypertension, often described as the "metabolic syndrome," is known to increase the risk of developing cardiovascular disease and diabetes. Stearoyl-coenzyme A desaturase (SCD) activity has been implicated in the metabolic syndrome, but detailed studies of the beneficial metabolic effects of SCD deficiency have been limited. Here, we show that absence of the Scd1 gene product reduces plasma triglycerides and reduces weight gain in severely hyperlipidemic low density lipoprotein receptor (LDLR)-deficient mice challenged with a Western diet. Absence of SCD1 also increases insulin sensitivity, as measured by intraperitoneal glucose and insulin tolerance testing. SCD1 deficiency dramatically reduces hepatic lipid accumulation while causing more modest reductions in plasma apolipoproteins, suggesting that in conditions of sustained hyperlipidemia, SCD1 functions primarily to mediate lipid stores. In addition, absence of SCD1 partially ameliorates the undesirable hypertriglyceridemic effect of antiatherogenic liver X receptor agonists. Our results demonstrate that constitutive reduction of SCD activity improves the metabolic phenotype of LDLR-deficient mice on a Western diet.


Assuntos
Hepatócitos/metabolismo , Hiperlipidemias/metabolismo , Lipídeos/sangue , Síndrome Metabólica/metabolismo , Receptores de LDL/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Adiposidade/fisiologia , Animais , Peso Corporal , Células Cultivadas , Dieta/efeitos adversos , Fígado Gorduroso/metabolismo , Hiperlipidemias/sangue , Resistência à Insulina , Metabolismo dos Lipídeos , Camundongos , Camundongos Mutantes , Receptores de LDL/deficiência , Receptores de LDL/genética , Estearoil-CoA Dessaturase/deficiência , Estearoil-CoA Dessaturase/genética
17.
Diabetes ; 56(11): 2797-808, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17704298

RESUMO

OBJECTIVE: Rosiglitazone displays powerful antidiabetes benefits but is associated with increased body weight and adipogenesis. Keeping in mind the concept of selective peroxisome proliferator-activated receptor (PPAR)gamma modulator, the aim of this study was to characterize the properties of a new PPARgamma ligand, S 26948, with special attention in body-weight gain. RESEARCH DESIGN AND METHODS: We used transient transfection and binding assays to characterized the binding characteristics of S 26948 and GST pull-down experiments to investigate its pattern of coactivator recruitment compared with rosiglitazone. We also assessed its adipogenic capacity in vitro using the 3T3-F442A cell line and its in vivo effects in ob/ob mice (for antidiabetes and antiobesity properties), as well as the homozygous human apolipoprotein E2 knocking mice (E2-KI) (for antiatherogenic capacity). RESULTS: S 26948 displayed pharmacological features of a high selective ligand for PPARgamma with low potency in promoting adipocyte differentiation. It also displayed a different coactivator recruitment profile compared with rosiglitazone, being unable to recruit DRIP205 or PPARgamma coactivator-1 alpha. In vivo experiments showed that S 26948 was as efficient in ameliorating glucose and lipid homeostasis as rosiglitazone, but it did not increase body and white adipose tissue weights and improved lipid oxidation in liver. In addition, S 26948 represented one of the few molecules of the PPARgamma ligand class able to decrease atherosclerotic lesions. CONCLUSIONS: These findings establish S 26948 as a selective PPARgamma ligand with distinctive coactivator recruitment and gene expression profile, reduced adipogenic effect, and improved biological responses in vivo.


Assuntos
Aterosclerose/prevenção & controle , Angiopatias Diabéticas/prevenção & controle , Hipoglicemiantes/farmacologia , PPAR gama/fisiologia , Animais , Células COS , Membrana Celular/fisiologia , Chlorocebus aethiops , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Haplorrinos , Humanos , Ligantes , PPAR gama/efeitos dos fármacos , PPAR gama/genética , Receptor X Retinoide alfa/efeitos dos fármacos , Receptor X Retinoide alfa/fisiologia , Transfecção
18.
Front Biosci ; 12: 4132-56, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17485363

RESUMO

Experimental approaches to understand the pathogenesis and to develop treatments of atherosclerosis involve studies in animal and cellular models. However, relevant animal models are rare since atherosclerosis is a disease that naturally affects only humans and one or two other species (pigs and certain primates). For a long time, atherosclerosis studies were carried out using diet-induced atherosclerosis models, even though the diets were unphysiological and the arterial lesions that developed were often limited in size, composition and location. During the last decade, with the advent of molecular genetics and genetic manipulation techniques, the development of genetically-engineered animals, mainly mice, allowed an explosion in the number of models resulting in a tremendous progress in atherosclerosis research and enhancement of our understanding of the disease. Atherosclerosis is a multifactorial disease which normally develops very slowly and asymptomatically during several decades, leading to atheromatous plaque formation. Once the plaque is weakened, its rupture or erosion induces severe clinical complications, such as myocardial infarction or cerebrovascular accidents. Several risk factors predispose to atherosclerosis including hypertension and abnormalities in lipoprotein metabolism and glucose homeostasis. The formation of the atherosclerotic lesion is a complex process, characterized by the presence of lipid-laden monocyte-derived macrophages (called foam cells), establishing therefore a status of chronic inflammation. The dysregulated expression of genes encoding proteins involved in the control of metabolic pathways contributes to vascular inflammation and the development of atherosclerosis. The expression of these genes is controlled by different transcription factors amongst which are the Peroxisome Proliferator-Activated Receptor (PPAR) family of nuclear receptors. This review focuses on the use of genetically-engineered animals as models for experimental atherosclerosis research, pointing out their contribution to investigate the implication of PPARs and their ligands in regulating metabolic and inflammatory abnormalities predisposing to atherosclerosis development.


Assuntos
Animais Geneticamente Modificados , Aterosclerose/fisiopatologia , Modelos Animais de Doenças , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Expressão Gênica , Camundongos
19.
Arterioscler Thromb Vasc Biol ; 26(12): 2731-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17008586

RESUMO

OBJECTIVE: The activity of the antitumoral agent bexarotene (Targretin, Bexarotene) depends on its binding to the nuclear retinoid-X receptor (RXR) and subsequent transcriptional regulation of target genes. Through RXR activation, bexarotene may modulate numerous metabolic pathways involved in atherosclerosis. Here, we investigated the effect of bexarotene on atherosclerosis progression in a dyslipidemic murine model, the human apolipoprotein E2 knockin mouse, that develops essentially macrophage-laden lesions. METHODS AND RESULTS: Atherosclerotic lesions together with different metabolic pathways involved in atherosclerosis were investigated in mice treated or not with bexarotene. Bexarotene protects from atherosclerosis development in mice, at least in part by improving the circulating cholesterol distribution profile likely via a marked decrease of dietary cholesterol absorption caused by modulation of intestinal expression of genes recently identified as major players in this process, Niemann-Pick-C1-Like1 (NPC1L1) and CD13. This atheroprotection appears despite a strong hypertriglyceridemia. Moreover, bexarotene treatment only modestly modulates inflammatory gene expression in the vascular wall, but markedly enhanced the capacity of macrophages to efflux cellular lipids. CONCLUSIONS: These data provide evidence of a favorable pharmacological effect of bexarotene on atherosclerosis despite the induction of hypertriglyceridemia, likely via a beneficial action on intestinal absorption and macrophage efflux.


Assuntos
Aterosclerose/tratamento farmacológico , Aterosclerose/etiologia , Colesterol/metabolismo , Dislipidemias/complicações , Homeostase/efeitos dos fármacos , Receptores X de Retinoides/agonistas , Tetra-Hidronaftalenos/farmacologia , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Aterosclerose/metabolismo , Bexaroteno , Antígenos CD13/genética , Antígenos CD13/metabolismo , Modelos Animais de Doenças , Dislipidemias/tratamento farmacológico , Dislipidemias/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/fisiologia , Absorção Intestinal/efeitos dos fármacos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Tetra-Hidronaftalenos/uso terapêutico , Triglicerídeos/sangue
20.
Arterioscler Thromb Vasc Biol ; 26(12): 2724-30, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17008588

RESUMO

OBJECTIVE: High-density lipoprotein (HDL) plays a key role in protection against development of atherosclerosis by reducing inflammation, protecting against LDL oxidation, and promoting reverse cholesterol transport from peripheral tissues to the liver for secretion into bile. Cholesterol 7alpha-hydroxylase (Cyp7a1) catalyzes the rate-limiting step in the intrahepatic conversion of cholesterol to bile acids that may have a role in HDL metabolism. We investigated the effect of Cyp7a1 deficiency on HDL metabolism in APOE*3-Leiden transgenic mice. METHODS AND RESULTS: Reduced bile acid biosynthesis in Cyp7a1-/-.APOE*3-Leiden mice versus APOE*3-Leiden mice did not affect total plasma cholesterol levels, but the distribution of cholesterol over various lipoproteins was different. Cholesterol was decreased in apoB-containing lipoproteins (ie, VLDL and IDL/LDL), whereas cholesterol was increased in HDL. The activity of PLTP and LCAT, which play a role in HDL catabolism, were not changed, and neither was HDL clearance. However, the hepatic cholesterol content was 2-fold increased, which was accompanied by a 2-fold elevated expression of hepatic ABCA1 and increased rate of cholesterol efflux from the liver to HDL. CONCLUSIONS: Strongly reduced bile acid synthesis in Cyp7a1-/-.APOE*3-Leiden mice leads to increased plasma HDL-cholesterol levels, as related to an increased hepatic expression of ABCA1.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Apolipoproteína E3/metabolismo , Colesterol 7-alfa-Hidroxilase/metabolismo , HDL-Colesterol/metabolismo , RNA Mensageiro/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apolipoproteína E3/genética , Ácidos e Sais Biliares/metabolismo , Colesterol 7-alfa-Hidroxilase/genética , HDL-Colesterol/genética , Feminino , Regulação da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilcolina-Esterol O-Aciltransferase/genética , Fosfatidilcolina-Esterol O-Aciltransferase/metabolismo , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Transferência de Fosfolipídeos/metabolismo , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...