Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Intern Med ; 292(2): 296-307, 2022 08.
Article in English | MEDLINE | ID: mdl-34982494

ABSTRACT

BACKGROUND: Sterol O-acyltransferase 2 (Soat2) encodes acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2), which synthesizes cholesteryl esters in hepatocytes and enterocytes fated either to storage or to secretion into nascent triglyceride-rich lipoproteins. OBJECTIVES: We aimed to unravel the molecular mechanisms leading to reduced hepatic steatosis when Soat2 is depleted in mice. METHODS: Soat2-/- and wild-type mice were fed a high-fat, a high-carbohydrate, or a chow diet, and parameters of lipid and glucose metabolism were assessed. RESULTS: Glucose, insulin, homeostatic model assessment for insulin resistance (HOMA-IR), oral glucose tolerance (OGTT), and insulin tolerance tests significantly improved in Soat2-/- mice, irrespective of the dietary regimes (2-way ANOVA). The significant positive correlations between area under the curve (AUC) OGTT (r = 0.66, p < 0.05), serum fasting insulin (r = 0.86, p < 0.05), HOMA-IR (r = 0.86, p < 0.05), Adipo-IR (0.87, p < 0.05), hepatic triglycerides (TGs) (r = 0.89, p < 0.05), very-low-density lipoprotein (VLDL)-TG (r = 0.87, p < 0.05) and the hepatic cholesteryl esters in wild-type mice disappeared in Soat2-/- mice. Genetic depletion of Soat2 also increased whole-body oxidation by 30% (p < 0.05) compared to wild-type mice. CONCLUSION: Our data demonstrate that ACAT2-generated cholesteryl esters negatively affect the metabolic control by retaining TG in the liver and that genetic inhibition of Soat2 improves liver steatosis via partitioning of lipids into secretory (VLDL-TG) and oxidative (fatty acids) pathways.


Subject(s)
Fatty Liver , Insulins , Sterol O-Acyltransferase , Animals , Cholesterol Esters/metabolism , Fatty Liver/metabolism , Glucose/metabolism , Insulins/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Triglycerides , Sterol O-Acyltransferase 2
2.
J Autoimmun ; 124: 102723, 2021 11.
Article in English | MEDLINE | ID: mdl-34481107

ABSTRACT

The initiation and progression of autoimmune disorders such as multiple sclerosis (MS) is linked to aberrant cholesterol metabolism and overt inflammation. Liver X receptors (LXR) are nuclear receptors that function at the crossroads of cholesterol metabolism and immunity, and their activation is considered a promising therapeutic strategy to attenuate autoimmunity. However, despite clear functional heterogeneity and cell-specific expression profiles, the impact of the individual LXR isoforms on autoimmunity remains poorly understood. Here, we show that LXRα and LXRß have an opposite impact on immune cell function and disease severity in the experimental autoimmune encephalomyelitis model, an experimental MS model. While Lxrα deficiency aggravated disease pathology and severity, absence of Lxrß was protective. Guided by flow cytometry and by using cell-specific knockout models, reduced disease severity in Lxrß-deficient mice was primarily attributed to changes in peripheral T cell physiology and occurred independent from alterations in microglia function. Collectively, our findings indicate that LXR isoforms play functionally non-redundant roles in autoimmunity, potentially having broad implications for the development of LXR-based therapeutic strategies aimed at dampening autoimmunity and neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Liver X Receptors/metabolism , Microglia/pathology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity , Cholesterol/metabolism , Disease Models, Animal , Humans , Liver X Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation
3.
Hepatology ; 72(2): 656-670, 2020 08.
Article in English | MEDLINE | ID: mdl-31785104

ABSTRACT

BACKGROUND AND AIMS: Genetically modified mice have been used extensively to study human disease. However, the data gained are not always translatable to humans because of major species differences. Liver-humanized mice (LHM) are considered a promising model to study human hepatic and systemic metabolism. Therefore, we aimed to further explore their lipoprotein metabolism and to characterize key hepatic species-related, physiological differences. APPROACH AND RESULTS: Fah-/- , Rag2-/- , and Il2rg-/- knockout mice on the nonobese diabetic (FRGN) background were repopulated with primary human hepatocytes from different donors. Cholesterol lipoprotein profiles of LHM showed a human-like pattern, characterized by a high ratio of low-density lipoprotein to high-density lipoprotein, and dependency on the human donor. This pattern was determined by a higher level of apolipoprotein B100 in circulation, as a result of lower hepatic mRNA editing and low-density lipoprotein receptor expression, and higher levels of circulating proprotein convertase subtilisin/kexin type 9. As a consequence, LHM lipoproteins bind to human aortic proteoglycans in a pattern similar to human lipoproteins. Unexpectedly, cholesteryl ester transfer protein was not required to determine the human-like cholesterol lipoprotein profile. Moreover, LHM treated with GW3965 mimicked the negative lipid outcomes of the first human trial of liver X receptor stimulation (i.e., a dramatic increase of cholesterol and triglycerides in circulation). Innovatively, LHM allowed the characterization of these effects at a molecular level. CONCLUSIONS: LHM represent an interesting translatable model of human hepatic and lipoprotein metabolism. Because several metabolic parameters displayed donor dependency, LHM may also be used in studies for personalized medicine.


Subject(s)
Benzoates/pharmacokinetics , Benzylamines/pharmacokinetics , Cholesterol/metabolism , Hepatocytes/metabolism , Lipoproteins/metabolism , Liver X Receptors/agonists , Liver/metabolism , Animals , Hepatocytes/transplantation , Humans , Liver/surgery , Male , Mice , Mice, Knockout
4.
Genes Dev ; 24(4): 381-95, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20159957

ABSTRACT

The orphan receptor LRH-1 and the oxysterol receptors LXRalpha and LXRbeta are established transcriptional regulators of lipid metabolism that appear to control inflammatory processes. Here, we investigate the anti-inflammatory actions of these nuclear receptors in the hepatic acute phase response (APR). We report that selective synthetic agonists induce SUMOylation-dependent recruitment of either LRH-1 or LXR to hepatic APR promoters and prevent the clearance of the N-CoR corepressor complex upon cytokine stimulation. Investigations of the APR in vivo, using LXR knockout mice, indicate that the anti-inflammatory actions of LXR agonists are triggered selectively by the LXRbeta subtype. We further find that hepatic APR responses in small ubiquitin-like modifier-1 (SUMO-1) knockout mice are increased, which is due in part to diminished LRH-1 action at APR promoters. Finally, we provide evidence that the metabolically important coregulator GPS2 functions as a hitherto unrecognized transrepression mediator of interactions between SUMOylated nuclear receptors and the N-CoR corepressor complex. Our study extends the knowledge of anti-inflammatory mechanisms and pathways directed by metabolic nuclear receptor-corepressor networks to the control of the hepatic APR, and implies alternative pharmacological strategies for the treatment of human metabolic diseases associated with inflammation.


Subject(s)
Acute-Phase Reaction/immunology , Intracellular Signaling Peptides and Proteins/immunology , Liver/immunology , Orphan Nuclear Receptors/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Small Ubiquitin-Related Modifier Proteins/immunology , Animals , Anti-Inflammatory Agents/immunology , COS Cells , Chlorocebus aethiops , Female , Gene Expression Regulation , HeLa Cells , Humans , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Mol Cell ; 34(4): 510-8, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19481530

ABSTRACT

Transcriptional coregulators, rather than ligand signals, are suspected to confer context and pathway specificity to nuclear receptor signaling, but the identity of such specifying coregulators and the underlying molecular mechanisms remain largely enigmatic. Here we address this issue in metabolic oxysterol receptor LXR pathways and describe the selective requirement of GPS2 for ABCG1 cholesterol transporter gene transcription and cholesterol efflux from macrophages. We implicate GPS2 in facilitating LXR recruitment to an ABCG1-specific promoter/enhancer unit upon ligand activation and identify functional links to histone H3K9 demethylation. We further describe fundamental differences between ABCG1 and ABCA1 with regard to GPS2 in relation to other coregulators, which are likely to apply to additional LXR-regulated genes. Our work identifies a coregulator-dependent epigenetic mechanism governing the access of a nuclear receptor to communicating regulatory regions in the genome. The pathway and coregulator selectivity of this mechanism implies pharmacological possibilities for the development of selective LXR agonists.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Epistasis, Genetic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver X Receptors , Macrophages/cytology , Macrophages/metabolism , Orphan Nuclear Receptors , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
6.
Diabetologia ; 59(3): 634-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26684450

ABSTRACT

AIMS/HYPOTHESIS: Diabetic cardiomyopathy is a myocardial disease triggered by impaired insulin signalling, increased fatty acid uptake and diminished glucose utilisation. Liver X receptors (LXRs) are key transcriptional regulators of metabolic homeostasis. However, their effect in the diabetic heart is largely unknown. METHODS: We cloned murine Lxrα (also known as Nr1h3) behind the α-myosin heavy chain (αMhc; also known as Myh6) promoter to create transgenic (Lxrα-Tg) mice and transgene-negative littermates (wild-type [WT]). A mouse model of type 2 diabetes was induced by a high-fat diet (HFD, 60% energy from fat) over 16 weeks and compared with a low-fat diet (10% energy from fat). A mouse model of type 1 diabetes was induced via streptozotocin injection over 12 weeks. RESULTS: HFD manifested comparable increases in body weight, plasma triacylglycerol and insulin resistance per OGTT in Lxrα-Tg and WT mice. HFD significantly increased left ventricular weight by 21% in WT hearts, but only by 5% in Lxrα-Tg. To elucidate metabolic effects in the heart, microPET (positron emission tomography) imaging revealed that cardiac glucose uptake was increased by 1.4-fold in WT mice on an HFD, but further augmented by 1.7-fold in Lxrα-Tg hearts, in part through 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and restoration of glucose transporter 4 (GLUT4). By contrast, streptozotocin-induced ablation of insulin signalling diminished cardiac glucose uptake levels and caused cardiac dysfunction, indicating that insulin may be important in LXRα-mediated glucose uptake. Chromatin immunoprecipitation assays identified natriuretic peptides, atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP), as potential direct targets of cardiac LXRα overexpression. CONCLUSIONS/INTERPRETATION: Cardiac-specific LXRα overexpression ameliorates the progression of HFD-induced left ventricular hypertrophy in association with increased glucose reliance and natriuretic peptide signalling during the early phase of diabetic cardiomyopathy. These findings implicate a potential protective role for LXR in targeting metabolic disturbances underlying diabetes.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Liver X Receptors/physiology , Obesity/complications , Animals , Chromatin Immunoprecipitation , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat , Disease Models, Animal , Liver X Receptors/genetics , Mice , Mice, Transgenic , Myocardium/metabolism
7.
Lab Invest ; 96(2): 230-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595172

ABSTRACT

Nuclear hormone receptor liver X receptor-alpha (LXRα) has a vital role in cholesterol homeostasis and is reported to have a role in adipose function and obesity although this is controversial. Conversely, mesenchymal stem cells (MSCs) are suggested to be a major source of adipocyte generation. Accordingly, we examined the role of LXRα in adipogenesis of MSCs. Adult murine MSCs (mMSCs) were isolated from wild-type (WT) and LXR-null mice. Using WT mMSCs, we further generated cell lines stably overexpressing GFP-LXRα (mMSC/LXRα/GFP) or GFP alone (mMSC/GFP) by retroviral infection. Confluent mMSCs were differentiated into adipocytes by the established protocol. Compared with MSCs isolated from WT mice, MSCs from LXR-null mice showed significantly increased adipogenesis, as determined by lipid droplet accumulation and adipogenesis-related gene expression. Moreover, mMSCs stably overexpressing GFP-LXRα (mMSC/LXRα/GFP) exhibited significantly decreased adipogenesis compared with mMSCs overexpressing GFP alone (mMSC/GFP). Since Wnt/beta-catenin signaling is reported to inhibit adipogenesis, we further examined it. The LXR-null group showed significantly decreased Wnt expression accompanied by a decrease of cellular beta-catenin (vs WT). The mMSC/LXRα/GFP group exhibited significantly increased Wnt expression accompanied by an increase of cellular beta-catenin (vs mMSC/GFP). These data demonstrate that LXRα has an inhibitory effect on adipogenic differentiation in mMSCs with Wnt/beta-catenin signaling. These results provide important insights into the pathophysiology of obesity and obesity-related consequences such as metabolic syndrome and may identify potential therapeutic targets.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Cell Differentiation/physiology , Orphan Nuclear Receptors/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Humans , Liver X Receptors , Mesenchymal Stem Cells/cytology , Mice , Orphan Nuclear Receptors/genetics , Wnt Signaling Pathway/physiology
8.
J Lipid Res ; 56(4): 771-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724563

ABSTRACT

Liver X receptor (LXR)α and LXRß play key roles in hepatic de novo lipogenesis through their regulation of lipogenic genes, including sterol regulatory element-binding protein (SREBP)-1c and carbohydrate responsive element-binding protein (ChREBP). LXRs activate lipogenic gene transcription in response to feeding, which is believed to be mediated by insulin. We have previously shown that LXRs are targets for glucose-hexosamine-derived O-linked ß-N-acetylglucosamine (O-GlcNAc) modification enhancing their ability to regulate SREBP-1c promoter activity in vitro. To elucidate insulin-independent effects of feeding on LXR-mediated lipogenic gene expression in vivo, we subjected control and streptozotocin-treated LXRα/ß(+/+) and LXRα/ß(-/-) mice to a fasting-refeeding regime. We show that under hyperglycemic and hypoinsulinemic conditions, LXRs maintain their ability to upregulate the expression of glycolytic and lipogenic enzymes, including glucokinase (GK), SREBP-1c, ChREBPα, and the newly identified shorter isoform ChREBPß. Furthermore, glucose-dependent increases in LXR/retinoid X receptor-regulated luciferase activity driven by the ChREBPα promoter was mediated, at least in part, by O-GlcNAc transferase (OGT) signaling in Huh7 cells. Moreover, we show that LXR and OGT interact and colocalize in the nucleus and that loss of LXRs profoundly reduced nuclear O-GlcNAc signaling and ChREBPα promoter binding activity in vivo. In summary, our study provides evidence that LXRs act as nutrient and glucose metabolic sensors upstream of ChREBP by modulating GK expression, nuclear O-GlcNAc signaling, and ChREBP expression and activity.


Subject(s)
Acetylglucosamine/metabolism , Cell Nucleus/metabolism , Liver/cytology , Liver/metabolism , Nuclear Proteins/metabolism , Orphan Nuclear Receptors/metabolism , Signal Transduction , Transcription Factors/metabolism , Acylation/drug effects , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Line , Cell Nucleus/drug effects , Eating , Fasting , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Lipogenesis/drug effects , Liver/drug effects , Liver/enzymology , Liver X Receptors , Male , Mice , Nuclear Proteins/genetics , Orphan Nuclear Receptors/deficiency , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , Pyruvate Kinase/metabolism , Signal Transduction/drug effects , Streptozocin/adverse effects , Transcription Factors/genetics , Transcriptional Activation/drug effects , Triglycerides/biosynthesis , Triglycerides/blood
9.
Eur J Immunol ; 44(7): 1896-903, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777958

ABSTRACT

Oxysterols are involved in maintaining cellular cholesterol levels. Recently, oxysterols have been demonstrated to modulate the function of immune cells and tumor growth. These effects can be dependent on the activation of the oxysterol-binding liver X receptors (LXRs) or, as recently demonstrated for T and B cells, DCs and neutrophils, can be independent of LXR activation. LXR-dependent oxysterol effects can be ascribed to the activation of LXRα, LXRß or LXRαß isoforms, which induces transcriptional activation or trans-repression of target genes. The prevalent activation of one isoform seems to be cell-, tissue-, or context-specific, as shown in some pathologic processes, i.e., infectious diseases, atherosclerosis, and autoimmunity. Oxysterol-LXR signaling has recently been shown to inhibit antitumor immune responses, as well as to modulate tumor cell growth. Here, we review the mechanisms that link oxysterols to tumor growth, and discuss possible networks at the basis of LXR-dependent and -independent oxysterol effects on immune cells and tumor development.


Subject(s)
Cholesterol/metabolism , Hydroxycholesterols/metabolism , Immunity , Neoplasms/pathology , Orphan Nuclear Receptors/physiology , Animals , Dendritic Cells/immunology , Humans , Liver X Receptors , Lymphocytes/immunology , Macrophages/physiology , Monocytes/physiology , Receptors, CCR7/physiology , Tumor Microenvironment
10.
Hepatology ; 59(5): 1791-802, 2014 May.
Article in English | MEDLINE | ID: mdl-24277692

ABSTRACT

UNLABELLED: Liver X receptor (LXR) activation stimulates triglyceride (TG) accumulation in the liver. Several lines of evidence indicate that estradiol-17ß (E2) reduces TG levels in the liver; however, the molecular mechanism underlying the E2 effect remains unclear. Here, we show that administration of E2 attenuated sterol regulatory element-binding protein (SREBP)-1 expression and TG accumulation induced by LXR activation in mouse liver. In estrogen receptor alpha (ERα) knockout (KO) and liver-specific ERα KO mice, E2 did not affect SREBP-1 expression or TG levels. Molecular analysis revealed that ERα is recruited to the SREBP-1c promoter through direct binding to LXR and inhibits coactivator recruitment to LXR in an E2-dependent manner. Our findings demonstrate the existence of a novel liver-dependent mechanism controlling TG accumulation through the nonclassical ER/LXR pathway. To confirm that a nonclassical ER/LXR pathway regulates ERα-dependent inhibition of LXR activation, we screened ERα ligands that were able to repress LXR activation without enhancing ERα transcriptional activity, and, as a result, we identified the phytoestrogen, phloretin. In mice, phloretin showed no estrogenic activity; however, it did reduce SREBP-1 expression and TG levels in liver of mice fed a high-fat diet to an extent similar to that of E2. CONCLUSION: We propose that ER ligands reduce TG levels in the liver by inhibiting LXR activation through a nonclassical pathway. Our results also indicate that the effects of ER on TG accumulation can be distinguished from its estrogenic effects by a specific ER ligand.


Subject(s)
Fatty Liver/prevention & control , Orphan Nuclear Receptors/physiology , Receptors, Estrogen/physiology , Animals , Diet, High-Fat , Estradiol/pharmacology , Female , Ligands , Liver X Receptors , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/antagonists & inhibitors , Phloretin/pharmacology , Promoter Regions, Genetic , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation , Triglycerides/metabolism
11.
Nat Chem Biol ; 9(2): 126-33, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292650

ABSTRACT

Liver X receptors (Lxrα and Lxrß) are ligand-dependent nuclear receptors critical for ventral midbrain neurogenesis in vivo. However, no endogenous midbrain Lxr ligand has so far been identified. Here we used LC/MS and functional assays to identify cholic acid as a new Lxr ligand. Moreover, 24(S),25-epoxycholesterol (24,25-EC) was found to be the most potent and abundant Lxr ligand in the developing mouse midbrain. Both Lxr ligands promoted neural development in an Lxr-dependent manner in zebrafish in vivo. Notably, each ligand selectively regulated the development of distinct midbrain neuronal populations. Whereas cholic acid increased survival and neurogenesis of Brn3a-positive red nucleus neurons, 24,25-EC promoted dopaminergic neurogenesis. These results identify an entirely new class of highly selective and cell type-specific regulators of neurogenesis and neuronal survival. Moreover, 24,25-EC promoted dopaminergic differentiation of embryonic stem cells, suggesting that Lxr ligands may thus contribute to the development of cell replacement and regenerative therapies for Parkinson's disease.


Subject(s)
Mesencephalon/metabolism , Neurogenesis , Orphan Nuclear Receptors/metabolism , Animals , Brain Mapping/methods , Cell Differentiation , Cell Nucleus/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Cholic Acid/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Embryonic Stem Cells/cytology , Ligands , Liver X Receptors , Mice , Models, Biological , Time Factors , Transfection , Zebrafish
12.
J Immunol ; 190(12): 6520-32, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23686490

ABSTRACT

Liver X receptors (LXRs) exert key functions in lipid homeostasis and in control of inflammation. In this study we have explored the impact of LXR activation on the macrophage response to the endogenous inflammatory cytokine IFN-γ. Transcriptional profiling studies demonstrate that ∼38% of the IFN-γ-induced transcriptional response is repressed by LXR activation in macrophages. LXRs also mediated inhibitory effects on selected IFN-γ-induced genes in primary microglia and in a model of IFN-γ-induced neuroinflammation in vivo. LXR activation resulted in reduced STAT1 recruitment to the promoters tested in this study without affecting STAT1 phosphorylation. A closer look into the mechanism revealed that SUMOylation of LXRs, but not the presence of nuclear receptor corepressor 1, was required for repression of the NO synthase 2 promoter. We have also analyzed whether IFN-γ signaling exerts reciprocal effects on LXR targets. Treatment with IFN-γ inhibited, in a STAT1-dependent manner, the LXR-dependent upregulation of selective targets, including ATP-binding cassette A1 (ABCA1) and sterol response element binding protein 1c. Downregulation of ABCA1 expression correlated with decreased cholesterol efflux to apolipoprotein A1 in macrophages stimulated with IFN-γ. The inhibitory effects of IFN-γ on LXR signaling did not involve reduced binding of LXR/retinoid X receptor heterodimers to target gene promoters. However, overexpression of the coactivator CREB-binding protein/p300 reduced the inhibitory actions of IFN-γ on the Abca1 promoter, suggesting that competition for CREB-binding protein may contribute to STAT1-dependent downregulation of LXR targets. The results from this study suggest an important level of bidirectional negative cross-talk between IFN-γ/STAT1 and LXRs with implications both in the control of IFN-γ-mediated immune responses and in the regulation of lipid metabolism.


Subject(s)
Interferon-gamma/immunology , Macrophages/immunology , Orphan Nuclear Receptors/immunology , Receptor Cross-Talk/immunology , STAT1 Transcription Factor/immunology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Gene Expression Regulation/immunology , Inflammation/immunology , Lipid Metabolism/physiology , Liver X Receptors , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors/metabolism , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Transcriptome
13.
Am J Physiol Endocrinol Metab ; 306(5): E494-502, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24368671

ABSTRACT

The liver X receptors (LXR)α and LXRß are transcription factors belonging to the nuclear receptor family, which play a central role in metabolic homeostasis, being master regulators of key target genes in the glucose and lipid pathways. Wild-type (WT), LXRα(-/-), and LXRß(-/-) mice were fed a chow diet with (treated) or without (control) the synthetic dual LXR agonist GW3965 for 5 wk. GW3965 raised intrahepatic triglyceride (TG) level but, surprisingly, reduced serum TG level through the activation of serum lipase activity. The serum TG reduction was associated with a repression of both catecholamine-stimulated lipolysis and relative glucose incorporation into lipid in isolated adipocytes through activation of LXRß. We also demonstrated that LXRα is required for basal (nonstimulated) adipocyte metabolism, whereas LXRß acts as a repressor of lipolysis. On the contrary, in skeletal muscle (SM), the lipogenic and cholesterol transporter LXR target genes were markedly induced in WT and LXRα(-/-) mice and to a lesser extent in LXRß(-/-) mice following treatment with GW3965. Moreover, TG content was reduced in SM of LXRß(-/-) mice, associated with increased expression of the main TG-lipase genes Hsl and Atgl. Energy expenditure was increased, and a switch from glucose to lipid oxidation was observed. In conclusion, we provide evidence that LXR might be an essential regulator of the lipid balance between tissues to ensure appropriate control of the flux of fuel. Importantly, we show that, after chronic treatment with GW3965, SM becomes the target tissue for LXR activation, as opposed to liver, in acute treatment.


Subject(s)
Adipocytes/drug effects , Homeostasis/drug effects , Lipid Metabolism/drug effects , Muscle, Skeletal/drug effects , Orphan Nuclear Receptors/agonists , Adipocytes/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Cholesterol/metabolism , Female , Homeostasis/physiology , Lipid Metabolism/physiology , Lipolysis/drug effects , Lipolysis/physiology , Liver X Receptors , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Triglycerides/blood
14.
Adv Sci (Weinh) ; 11(20): e2307201, 2024 May.
Article in English | MEDLINE | ID: mdl-38549193

ABSTRACT

Macrophages regulate essential aspects of innate immunity against pathogens. In response to microbial components, macrophages activate primary and secondary inflammatory gene programs crucial for host defense. The liver X receptors (LXRα, LXRß) are ligand-dependent nuclear receptors that direct gene expression important for cholesterol metabolism and inflammation, but little is known about the individual roles of LXRα and LXRß in antimicrobial responses. Here, the results demonstrate that induction of LXRα transcription by prolonged exposure to lipopolysaccharide (LPS) supports inflammatory gene expression in macrophages. LXRα transcription is induced by NF-κB and type-I interferon downstream of TLR4 activation. Moreover, LPS triggers a reprogramming of the LXRα cistrome that promotes cytokine and chemokine gene expression through direct LXRα binding to DNA consensus sequences within cis-regulatory regions including enhancers. LXRα-deficient macrophages present fewer binding of p65 NF-κB and reduced histone H3K27 acetylation at enhancers of secondary inflammatory response genes. Mice lacking LXRα in the hematopoietic compartment show impaired responses to bacterial endotoxin in peritonitis models, exhibiting reduced neutrophil infiltration and decreased expansion and inflammatory activation of recruited F4/80lo-MHC-IIhi peritoneal macrophages. Together, these results uncover a previously unrecognized function for LXRα-dependent transcriptional cis-activation of secondary inflammatory gene expression in macrophages and the host response to microbial ligands.


Subject(s)
Inflammation , Liver X Receptors , Macrophages , Transcriptome , Animals , Mice , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL
15.
Breast Cancer Res ; 15(3): R51, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23809258

ABSTRACT

INTRODUCTION: Liver × receptors (LXRs) are members of the nuclear receptor family of ligand-dependent transcription factors and have established functions as regulators of cholesterol, glucose, and fatty acid metabolism and inflammatory responses. Published reports of anti-proliferative effects of synthetic LXR ligands on breast, prostate, ovarian, lung, skin, and colorectal cancer cells suggest that LXRs are potential targets in cancer prevention and treatment. METHODS: To further determine the effects of LXR ligands and identify their potential mechanisms of action in breast cancer cells, we carried out microarray analysis of gene expression in four breast cancer cell lines following treatments with the synthetic LXR ligand GW3965. Differentially expressed genes were further subjected to gene ontology and pathway analyses, and their expression profiles and associations with disease parameters and outcomes were examined in clinical samples. Response of E2F target genes were validated by real-time PCR, and the posited role of E2F2 in breast cancer cell proliferation was tested by RNA interference experiments. RESULTS: We observed cell line-specific transcriptional responses as well as a set of common responsive genes. In the common responsive gene set, upregulated genes tend to function in the known metabolic effects of LXR ligands and LXRs whereas the downregulated genes mostly include those which function in cell cycle regulation, DNA replication, and other cell proliferation-related processes. Transcription factor binding site analysis of the downregulated genes revealed an enrichment of E2F binding site sequence motifs. Correspondingly, E2F2 transcript levels are downregulated following LXR ligand treatment. Knockdown of E2F2 expression, similar to LXR ligand treatment, resulted in a significant disruption of estrogen receptor positive breast cancer cell proliferation. Ligand treatment also decreased E2F2 binding to cis-regulatory regions of target genes. Hierarchical clustering of breast cancer patients based on the expression profiles of the commonly downregulated LXR ligand-responsive genes showed a strong association of these genes with patient survival. CONCLUSIONS: Taken together, these results indicate that LXR ligands target gene networks, including those regulated by E2F family members, are critical for tumor biology and disease progression and merit further consideration as potential agents in the prevention and treatment of breast cancers.


Subject(s)
Benzoates/metabolism , Benzylamines/metabolism , Breast Neoplasms/genetics , E2F2 Transcription Factor/biosynthesis , Orphan Nuclear Receptors/metabolism , Benzoates/administration & dosage , Benzylamines/administration & dosage , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , E2F2 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Ligands , Liver X Receptors , Promoter Regions, Genetic , Transcription, Genetic/drug effects
16.
Mol Carcinog ; 52(11): 835-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22610535

ABSTRACT

The oxysterol receptors LXRα and LXRß are members of the nuclear receptor family and established transcriptional regulators of lipid metabolism with additional anti-inflammatory functions. Recent investigations have indicated an important role of LXRs in the control of proliferation. Here we further extend this knowledge to human colon cancer cells and proliferation in mouse colon. We show that activation of LXRs leads to a robust cell cycle arrest in colorectal adenocarcinoma cell lines. At the molecular level LXRs control expression of several cell cycle genes including Skp2, c-Myc, CDKs, cyclins, and p15. Furthermore, activation of LXRs causes hypo-phosphorylation of the retinoblastoma (Rb) tumor suppressor protein. Experiments performed in vivo show that the colon structure appears to be intact in LXR null mice. However, LXRαß(-/-) mice show a significant increase of proliferation markers in colon compared to wild type mice and administration of the LXR specific agonist, GW3965 significantly reduced expression of proliferation in mouse colon. Taken together, these findings point toward a strong anti-proliferative effect of LXRs in colon revealing the potential of LXR ligands as possible anti cancer agents.


Subject(s)
Cell Proliferation , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Orphan Nuclear Receptors/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Colon/cytology , Gene Expression Regulation , Humans , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/analysis , Orphan Nuclear Receptors/genetics
17.
Mol Membr Biol ; 29(7): 299-308, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22694168

ABSTRACT

The organ content of the mevalonate pathway lipids was investigated in liver-X-receptor (LXR) α, ß and double knock-out mice. An extensive or moderate increase of total cholesterol in the double KO mice was found in all organs elicited by the increase of the esterified form. In LXRα and double KO mice, coenzyme Q (CoQ) was decreased in liver and increased in spleen, thymus and lung, while dolichol was increased in all organs investigated. This effect was confirmed using LXR- agonist GW 3965. Analysis of CoQ distribution in organelles showed that the modifications are present in all cellular compartments and that the increase of the lipid in mitochondria was the result of a net increase of CoQ without changing the number of mitochondria. It appears that LXR influences not only cellular cholesterol homeostasis but also the metabolism of CoQ and dolichol, in an indirect manner.


Subject(s)
Cholesterol/metabolism , Dolichols/metabolism , Orphan Nuclear Receptors/metabolism , Ubiquinone/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Cholesterol/genetics , Dolichols/genetics , Female , Liver/metabolism , Liver X Receptors , Lung/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Organ Specificity/physiology , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Spleen/metabolism , Thymus Gland/metabolism , Ubiquinone/genetics
18.
J Neurosci ; 31(19): 7049-59, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21562267

ABSTRACT

Liver X receptors (LXRs) regulate immune cell function and cholesterol metabolism, both factors that are critically involved in Alzheimer's disease (AD). To investigate the therapeutic potential of long-term LXR activation in amyloid-ß (Aß) peptide deposition in an AD model, 13-month-old, amyloid plaque-bearing APP23 mice were treated with the LXR agonist TO901317. Postmortem analysis demonstrated that TO901317 efficiently crossed the blood-brain barrier. Insoluble and soluble Aß levels in the treated APP23 mice were reduced by 80% and 40%, respectively, compared with untreated animals. Amyloid precursor protein (APP) processing, however, was hardly changed by the compound, suggesting that the observed effects were instead mediated by Aß disposal. Despite the profound effect on Aß levels, spatial learning in the Morris water maze was only slightly improved by the treatment. ABCA1 (ATP-binding cassette transporter 1) and apolipoprotein E (ApoE) protein levels were increased and found to be primarily localized in astrocytes. Experiments using primary microglia demonstrated that medium derived from primary astrocytes exposed to TO901317 stimulated phagocytosis of fibrillar Aß. Conditioned medium from TO901317-treated ApoE(-/-) or LXRα(-/-) astrocytes did not increase phagocytosis of Aß. In APP23 mice, long-term treatment with TO901317 strongly increased the association of microglia and Aß plaques. Short-term treatment of APP/PS1 mice with TO901317 also increased this association, which was dependent on the presence of LXRα and was accompanied by increased ApoE lipidation. Together, these data suggest that astrocytic LXRα activation and subsequent release of ApoE by astrocytes is critical for the ability of microglia to remove fibrillar Aß in response to treatment with TO901317.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Astrocytes/metabolism , Microglia/metabolism , Orphan Nuclear Receptors/metabolism , Phagocytosis/physiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Apolipoproteins E/genetics , Astrocytes/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blotting, Western , Brain/drug effects , Brain/metabolism , Culture Media, Conditioned , Enzyme-Linked Immunosorbent Assay , Hydrocarbons, Fluorinated/pharmacology , Immunoassay , Immunohistochemistry , Liver X Receptors , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Microglia/drug effects , Orphan Nuclear Receptors/genetics , Sulfonamides/pharmacology
19.
BMC Genomics ; 13: 50, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22292898

ABSTRACT

BACKGROUND: The liver X receptors (LXRs) are oxysterol sensing nuclear receptors with multiple effects on metabolism and immune cells. However, the complete genome-wide cistrome of LXR in cells of human origin has not yet been provided. RESULTS: We performed ChIP-seq in phorbol myristate acetate-differentiated THP-1 cells (macrophage-type) after stimulation with the potent synthetic LXR ligand T0901317 (T09). Microarray gene expression analysis was performed in the same cellular model. We identified 1357 genome-wide LXR locations (FDR < 1%), of which 526 were observed after T09 treatment. De novo analysis of LXR binding sequences identified a DR4-type element as the major motif. On mRNA level T09 up-regulated 1258 genes and repressed 455 genes. Our results show that LXR actions are focused on 112 genomic regions that contain up to 11 T09 target genes per region under the control of highly stringent LXR binding sites with individual constellations for each region. We could confirm that LXR controls lipid metabolism and transport and observed a strong association with apoptosis-related functions. CONCLUSIONS: This first report on genome-wide binding of LXR in a human cell line provides new insights into the transcriptional network of LXR and its target genes with their link to physiological processes, such as apoptosis.The gene expression microarray and sequence data have been submitted collectively to the NCBI Gene Expression Omnibus http://www.ncbi.nlm.nih.gov/geo under accession number GSE28319.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , Apoptosis , Binding Sites , Cell Line , Down-Regulation , Humans , Lipid Metabolism , Liver X Receptors , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors/antagonists & inhibitors , Protein Binding , Up-Regulation
20.
Biochim Biophys Acta ; 1801(4): 421-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19962449

ABSTRACT

Cold adaptation elicits a paradoxical simultaneous induction of fatty acid synthesis and beta-oxidation in brown adipose tissue. We show here that cold exposure coordinately induced liver X receptor alpha (LXRalpha), adipocyte determination and differentiation-dependent factor 1 (ADD1)/sterol regulatory element-binding protein-1c (SREBP1c) and peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC1alpha) in brown and inguinal white adipose tissues, but not in epididymal white adipose tissue. Using in vitro models of white and brown adipocytes we demonstrate that beta-adrenergic stimulation induced expression of LXRalpha, ADD1/SREBP1c and PGC1alpha in cells with a brown-like adipose phenotype. We demonstrate that ADD1/SREBP1c is a powerful inducer of PGC1alpha expression via a conserved E box in the proximal promoter and that beta-adrenergic stimulation led to recruitment of ADD1/SREBP1c to this E box. The ability of ADD1/SREBP1c to activate the PGC1alpha promoter exhibited a striking cell type dependency, suggesting that additional cell type-restricted factors contribute to ADD1/SREBP1c-mediated activation. In conclusion, our data demonstrate a novel role of ADD1/SREBP1c as a regulator of PGC1alpha expression in brown adipose tissue.


Subject(s)
Adipocytes, Brown/metabolism , Promoter Regions, Genetic/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Electroporation , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Trans-Activators/genetics , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL