Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Lipid Res ; 53(12): 2610-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23028113

RESUMO

Mitochondrial dysfunction has been implicated in the pathogenesis of type 2 diabetes. Identifying novel regulators of mitochondrial bioenergetics will broaden our understanding of regulatory checkpoints that coordinate complex metabolic pathways. We previously showed that Nur77, an orphan nuclear receptor of the NR4A family, regulates the expression of genes linked to glucose utilization. Here we demonstrate that expression of Nur77 in skeletal muscle also enhances mitochondrial function. We generated MCK-Nur77 transgenic mice that express wild-type Nur77 specifically in skeletal muscle. Nur77-overexpressing muscle had increased abundance of oxidative muscle fibers and mitochondrial DNA content. Transgenic muscle also exhibited enhanced oxidative metabolism, suggestive of increased mitochondrial activity. Metabolomic analysis confirmed that Nur77 transgenic muscle favored fatty acid oxidation over glucose oxidation, mimicking the metabolic profile of fasting. Nur77 expression also improved the intrinsic respiratory capacity of isolated mitochondria, likely due to the increased abundance of complex I of the electron transport chain. These changes in mitochondrial metabolism translated to improved muscle contractile function ex vivo and improved cold tolerance in vivo. Our studies outline a novel role for Nur77 in the regulation of oxidative metabolism and mitochondrial activity in skeletal muscle.


Assuntos
Músculo Esquelético/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Animais , Creatina Quinase/genética , Creatina Quinase/metabolismo , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/química , Músculo Esquelético/enzimologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Oxirredução , Regiões Promotoras Genéticas/genética
2.
J Lipid Res ; 53(6): 1126-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22454476

RESUMO

The liver X receptor (LXR) signaling pathway is an important modulator of atherosclerosis, but the relative importance of the two LXRs in atheroprotection is incompletely understood. We show here that LXRα, the dominant LXR isotype expressed in liver, plays a particularly important role in whole-body sterol homeostasis. In the context of the ApoE(-/-) background, deletion of LXRα, but not LXRß, led to prominent increases in atherosclerosis and peripheral cholesterol accumulation. However, combined loss of LXRα and LXRß on the ApoE(-/-) background led to an even more severe cholesterol accumulation phenotype compared to LXRα(-/-)ApoE(-/-) mice, indicating that LXRß does contribute to reverse cholesterol transport (RCT) but that this contribution is quantitatively less important than that of LXRα. Unexpectedly, macrophages did not appear to underlie the differential phenotype of LXRα(-/-)ApoE(-/-) and LXRß(-/-)ApoE(-/-) mice, as in vitro assays revealed no difference in the efficiency of cholesterol efflux from isolated macrophages. By contrast, in vivo assays of RCT using exogenously labeled macrophages revealed a marked defect in fecal sterol efflux in LXRα(-/-)ApoE(-/-) mice. Mechanistically, this defect was linked to a specific requirement for LXRα(-/-) in the expression of hepatic LXR target genes involved in sterol transport and metabolism. These studies reveal a previously unrecognized requirement for hepatic LXRα for optimal reverse cholesterol transport in mice.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Colesterol/metabolismo , Receptores Nucleares Órfãos/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Fígado/metabolismo , Receptores X do Fígado , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
3.
J Lipid Res ; 52(3): 531-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21187453

RESUMO

Ligand activation of liver X receptors (LXRs) has been shown to impact both lipid metabolism and inflammation. One complicating factor in studies utilizing synthetic LXR agonists is the potential for pharmacologic and receptor-independent effects. Here, we describe an LXR gain-of-function system that does not depend on the addition of exogenous ligand. We generated transgenic mice expressing a constitutively active VP16-LXRα protein from the aP2 promoter. These mice exhibit increased LXR signaling selectively in adipose and macrophages. Analysis of gene expression in primary macrophages derived from two independent VP16-LXRα transgenic lines confirmed the ability of LXR to drive expression of genes involved in cholesterol efflux and fatty acid synthesis. Moreover, VP16-LXRα expression also suppressed the induction of inflammatory genes by lipopolysaccharide to a comparable degree as synthetic agonist. We further utilized VP16-LXRα-expressing macrophages to identify and validate new targets for LXRs, including the gene encoding ADP-ribosylation factor-like 7 (ARL7). ARL7 has previously been shown to transport cholesterol to the membrane for ABCA1-associated removal and thus may be integral to the LXR-dependent efflux pathway. We show that the ARL7 promoter contains a functional LXRE and can be transactivated by LXRs in a sequence-specific manner, indicating that ARL7 is a direct target of LXR. These findings provide further support for an important role of LXRs in the coordinated regulation of lipid metabolic and inflammatory gene programs in macrophages.


Assuntos
Fatores de Ribosilação do ADP/genética , Regulação da Expressão Gênica , Macrófagos/metabolismo , Receptores Nucleares Órfãos/metabolismo , Tecido Adiposo/metabolismo , Animais , Linhagem Celular , Colesterol/metabolismo , Humanos , Inflamação/genética , Ligantes , Metabolismo dos Lipídeos/genética , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Nucleares Órfãos/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética
4.
J Biol Chem ; 285(26): 19720-6, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20427281

RESUMO

We have previously identified the E3 ubiquitin ligase-inducible degrader of the low density lipoprotein receptor (LDLR) (Idol) as a post-translational modulator of LDLR levels. Idol is a direct target for regulation by liver X receptors (LXRs), and its expression is responsive to cellular sterol status independent of the sterol-response element-binding proteins. Here we demonstrate that Idol also targets two closely related LDLR family members, VLDLR and ApoE receptor 2 (ApoER2), proteins implicated in both neuronal development and lipid metabolism. Idol triggers ubiquitination of the VLDLR and ApoER2 on their cytoplasmic tails, leading to their degradation. We further show that the level of endogenous VLDLR is sensitive to cellular sterol content, Idol expression, and activation of the LXR pathway. Pharmacological activation of the LXR pathway in mice leads to increased Idol expression and to decreased Vldlr levels in vivo. Finally, we establish an unexpected functional link between LXR and Reelin signaling. We demonstrate that LXR activation results in decreased Reelin binding to VLDLR and reduced Dab1 phosphorylation. The identification of VLDLR and ApoER2 as Idol targets suggests potential roles for this LXR-inducible E3 ligase in the central nervous system in addition to lipid metabolism.


Assuntos
Receptores de LDL/metabolismo , Receptores de Lipoproteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Humanos , Hidrocarbonetos Fluorados/farmacologia , Immunoblotting , Proteínas Relacionadas a Receptor de LDL , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Fosforilação , Ligação Proteica , Receptores de LDL/genética , Receptores de Lipoproteínas/genética , Proteína Reelina , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
5.
Circ Res ; 104(6): e42-54, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19265038

RESUMO

Excess food intake leads to obesity and diabetes, both of which are well-known independent risk factors for atherosclerosis, and both of which are growing epidemics in an aging population. We hypothesized that aging enhances the metabolic and vascular effects of high fat diet (HFD) and therefore examined the effect of age on atherosclerosis and insulin resistance in lipoprotein receptor knockout (LDLR(-/-)) mice. We found that 12-month-old (middle-aged) LDLR(-/-) mice developed substantially worse metabolic syndrome, diabetes, and atherosclerosis than 3-month-old (young) LDLR(-/-) mice when both were fed HFD for 3 months, despite similar elevations in total cholesterol levels. Microarray analyses were performed to analyze the mechanism responsible for the marked acceleration of atherosclerosis in middle-aged mice. Chow-fed middle-aged mice had greater aortic expression of multiple antioxidant genes than chow-fed young mice, including glutathione peroxidase-1 and -4, catalase, superoxide dismutase-2, and uncoupling protein-2. Aortic expression of these enzymes markedly increased in young mice fed HFD but decreased or only modestly increased in middle-aged mice fed HFD, despite the fact that systemic oxidative stress and vascular reactive oxygen species generation, measured by plasma F2alpha isoprostane concentration (systemic) and dihydroethidium conversion and p47phox expression (vascular), were greater in middle-aged mice fed HFD. Thus, the mechanism for the accelerated vascular injury in older LDLR(-/-) mice was likely the profound inability to mount an antioxidant response. This effect was related to a decrease in vascular expression of 2 key transcriptional pathways regulating the antioxidant response, DJ-1 and forkhead box, subgroup O family (FOXOs). Treatment of middle-aged mice fed HFD with the antioxidant apocynin attenuated atherosclerosis, whereas treatment with the insulin sensitizer rosiglitazone attenuated both metabolic syndrome and atherosclerosis. Both treatments decreased oxidative stress. A novel effect of rosiglitazone was to increase expression of Nrf2 (nuclear factor [erythroid-derived 2]-like 2), a downstream target of DJ-1 contributing to enhanced expression of vascular antioxidant enzymes. This investigation underscores the role of oxidative stress when multiple atherosclerotic risk factors, particularly aging, converge on the vessel wall and emphasizes the need to develop effective strategies to inhibit oxidative stress to protect aging vasculature.


Assuntos
Envelhecimento/metabolismo , Antioxidantes/metabolismo , Aterosclerose/metabolismo , Dieta Aterogênica , Regulação da Expressão Gênica , Acetofenonas/farmacologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Aterosclerose/genética , Aterosclerose/patologia , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Ingestão de Alimentos , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Resistência à Insulina/genética , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Receptores de LDL/genética
6.
J Clin Endocrinol Metab ; 87(8): 3997-4000, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12161550

RESUMO

Ghrelin is a novel gastric peptide which stimulates growth hormone and has orexigenic and adipogenic properties. Plasma ghrelin is influenced by nutritional status and is thought to play a role in regulating food intake and body weight. We examined the effect of infusing insulin (40 mU/m(2)/min) for 2 hours while maintaining euglycemia on plasma ghrelin in 8 subjects (5 M, 3 F) aged 46 +/- 4 yrs (mean +/- SEM). Plasma insulin increased from 78 +/- 9 to 564 +/- 23 pmol/L during and returned rapidly to basal values after stopping the insulin infusion. Plasma ghrelin decreased from 85 +/- 28 to 61 +/- 18 pmol/L (p < 0.01) by 90 minutes of and continued to be suppressed for 15 minutes after the insulin infusion was discontinued. Subsequently, plasma ghrelin rose rapidly to near-basal values (81 +/- 23 pmol/L) within 60 minutes. The reciprocal relation between insulin and ghrelin was observed consistently in all subjects with the maximum insulin-induced suppression of ghrelin ranging from 19 to 64% (mean 32 +/- 5) and occurring 90-135 minutes after starting the insulin infusion (median 120). These findings indicate that insulin is a physiological and dynamic modulator of plasma ghrelin and that insulinemia possibly mediates the effect of nutritional status on its concentration.


Assuntos
Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Hormônios Peptídicos , Peptídeos/sangue , Adulto , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Feminino , Grelina , Humanos , Hipoglicemiantes/sangue , Insulina/sangue , Masculino , Pessoa de Meia-Idade
7.
Cell Metab ; 20(5): 910-918, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25440061

RESUMO

The LXR-regulated E3 ubiquitin ligase IDOL controls LDLR receptor stability independent of SREBP and PCSK9, but its relevance to plasma lipid levels is unknown. Here we demonstrate that the effects of the LXR-IDOL axis are both tissue and species specific. In mice, LXR agonist induces Idol transcript levels in peripheral tissues but not in liver, and does not change plasma LDL levels. Accordingly, Idol-deficient mice exhibit elevated LDLR protein levels in peripheral tissues, but not in the liver. By contrast, LXR activation in cynomolgus monkeys induces hepatic IDOL expression, reduces LDLR protein levels, and raises plasma LDL levels. Knockdown of IDOL in monkeys with an antisense oligonucleotide blunts the effect of LXR agonist on LDL levels. These results implicate IDOL as a modulator of plasma lipid levels in primates and support further investigation into IDOL inhibition as a potential strategy for LDL lowering in humans.


Assuntos
LDL-Colesterol/sangue , Fígado/metabolismo , Receptores Nucleares Órfãos/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células Cultivadas , LDL-Colesterol/metabolismo , Haplorrinos , Humanos , Receptores X do Fígado , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/metabolismo , Especificidade da Espécie
8.
Mol Cell Biol ; 31(9): 1885-93, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21343340

RESUMO

Previously, we identified the E3 ubiquitin ligase Idol (inducible degrader of the low-density lipoprotein [LDL] receptor [LDLR]) as a posttranscriptional regulator of the LDLR pathway. Idol stimulates LDLR degradation through ubiquitination of its C-terminal domain, thereby limiting cholesterol uptake. Here we report the generation and characterization of mouse embryonic stem cells homozygous for a null mutation in the Idol gene. Cells lacking Idol exhibit markedly elevated levels of the LDLR protein and increased rates of LDL uptake. Furthermore, despite an intact sterol responsive element-binding protein (SREBP) pathway, Idol-null cells exhibit an altered response to multiple regulators of sterol metabolism, including serum, oxysterols, and synthetic liver X receptor (LXR) agonists. The ability of oxysterols and lipoprotein-containing serum to suppress LDLR protein levels is reduced, and the time course of suppression is delayed, in cells lacking Idol. LXR ligands have no effect on LDLR levels in Idol-null cells, indicating that Idol is required for LXR-dependent inhibition of the LDLR pathway. In line with these results, the half-life of the LDLR protein is prolonged in the absence of Idol. Finally, the ability of statins and PCSK9 to alter LDLR levels is independent of, and additive with, the LXR-Idol pathway. These results demonstrate that the LXR-Idol pathway is an important contributor to feedback inhibition of the LDLR by sterols and a biological determinant of cellular LDL uptake.


Assuntos
Células-Tronco Embrionárias/metabolismo , Receptores Nucleares Órfãos/metabolismo , Receptores de LDL/metabolismo , Esteróis/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células Cultivadas , Células-Tronco Embrionárias/citologia , Expressão Gênica , Técnicas de Inativação de Genes , Lipoproteínas LDL/metabolismo , Fígado/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Serina Endopeptidases/metabolismo , Ubiquitina-Proteína Ligases/genética
9.
Cell Metab ; 13(4): 413-427, 2011 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-21459326

RESUMO

PPARγ and Wnt signaling are central positive and negative regulators of adipogenesis, respectively. Here we identify the groucho family member TLE3 as a transcriptional integrator of the PPARγ and Wnt pathways. TLE3 is a direct target of PPARγ that participates in a feed-forward loop during adipocyte differentiation. TLE3 enhances PPARγ activity and functions synergistically with PPARγ on its target promoters to stimulate adipogenesis. At the same time, induction of TLE3 during differentiation provides a mechanism for termination of Wnt signaling. TLE3 antagonizes TCF4 activation by ß-catenin in preadipocytes, thereby inhibiting Wnt target gene expression and reversing ß-catenin-dependent repression of adipocyte gene expression. Transgenic expression of TLE3 in adipose tissue in vivo mimics the effects of PPARγ agonist and ameliorates high-fat-diet-induced insulin resistance. Our data suggest that TLE3 acts as a dual-function switch, driving the formation of both active and repressive transcriptional complexes that facilitate the adipogenic program.


Assuntos
Adipogenia/genética , Proteínas/metabolismo , Adipócitos/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linhagem Celular , Proteínas Correpressoras , Gorduras na Dieta/farmacologia , Regulação da Expressão Gênica , Resistência à Insulina , Camundongos , PPAR gama/agonistas , PPAR gama/genética , PPAR gama/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Transcrição 4 , Transcrição Gênica , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
10.
Science ; 325(5936): 100-4, 2009 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-19520913

RESUMO

Cellular cholesterol levels reflect a balance between uptake, efflux, and endogenous synthesis. Here we show that the sterol-responsive nuclear liver X receptor (LXR) helps maintain cholesterol homeostasis, not only through promotion of cholesterol efflux but also through suppression of low-density lipoprotein (LDL) uptake. LXR inhibits the LDL receptor (LDLR) pathway through transcriptional induction of Idol (inducible degrader of the LDLR), an E3 ubiquitin ligase that triggers ubiquitination of the LDLR on its cytoplasmic domain, thereby targeting it for degradation. LXR ligand reduces, whereas LXR knockout increases, LDLR protein levels in vivo in a tissue-selective manner. Idol knockdown in hepatocytes increases LDLR protein levels and promotes LDL uptake. Conversely, adenovirus-mediated expression of Idol in mouse liver promotes LDLR degradation and elevates plasma LDL levels. The LXR-Idol-LDLR axis defines a complementary pathway to sterol response element-binding proteins for sterol regulation of cholesterol uptake.


Assuntos
Colesterol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de LDL/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/agonistas , Homeostase , Humanos , Ligantes , Lipoproteínas LDL/sangue , Lipoproteínas LDL/metabolismo , Fígado/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Receptores Nucleares Órfãos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores de LDL/genética , Transcrição Gênica , Ubiquitina-Proteína Ligases , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa