Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Bioorg Med Chem ; 27(18): 4059-4068, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31351846

ABSTRACT

The peroxisome proliferator activated receptors (PPARs) are important drug targets in treatment of metabolic and inflammatory disorders. Fibrates, acting as PPARα agonists, have been widely used lipid-lowering agents for decades. However, the currently available PPARα targeting agents show low subtype-specificity and consequently a search for more potent agonists have emerged. In this study, previously isolated oxohexadecenoic acids from the marine algae Chaetoceros karianus were used to design a PPARα-specific analogue. Herein we report the design, synthesis, molecular modelling studies and biological evaluations of the novel 3,5-disubstituted isoxazole analogue 6-(5-heptyl-1,2-oxazol-3-yl)hexanoic acid (1), named ADAM. ADAM shows a clear receptor preference and significant dose-dependent activation of PPARα (EC50 = 47 µM) through its ligand-binding domain (LBD). Moreover, ADAM induces expression of important PPARα target genes, such as CPT1A, in the Huh7 cell line and primary mouse hepatocytes. In addition, ADAM exhibits a moderate ability to regulate PPARγ target genes and drive adipogenesis. Molecular modelling studies indicated that ADAM docks its carboxyl group into opposite ends of the PPARα and -γ LBD. ADAM interacts with the receptor-activating polar network of amino acids (Tyr501, His447 and Ser317) in PPARα, but not in PPARγ LBD. This may explain the lack of PPARγ agonism, and argues for a PPARα-dependent adipogenic function. Such compounds are of interest towards developing new lipid-lowering remedies.


Subject(s)
Fatty Acids/metabolism , Isoxazoles/metabolism , PPAR alpha/agonists , Humans , Models, Molecular
2.
Biochem J ; 475(23): 3827-3846, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30373764

ABSTRACT

Here, we report the biochemical characterization of the mono-ADP-ribosyltransferase 2,3,7,8-tetrachlorodibenzo-p-dioxin poly-ADP-ribose polymerase (TIPARP/ARTD14/PARP7), which is known to repress aryl hydrocarbon receptor (AHR)-dependent transcription. We found that the nuclear localization of TIPARP was dependent on a short N-terminal sequence and its zinc finger domain. Deletion and in vitro ADP-ribosylation studies identified amino acids 400-657 as the minimum catalytically active region, which retained its ability to mono-ADP-ribosylate AHR. However, the ability of TIPARP to ADP-ribosylate and repress AHR in cells was dependent on both its catalytic activity and zinc finger domain. The catalytic activity of TIPARP was resistant to meta-iodobenzylguanidine but sensitive to iodoacetamide and hydroxylamine, implicating cysteines and acidic side chains as ADP-ribosylated target residues. Mass spectrometry identified multiple ADP-ribosylated peptides in TIPARP and AHR. Electron transfer dissociation analysis of the TIPARP peptide 33ITPLKTCFK41 revealed cysteine 39 as a site for mono-ADP-ribosylation. Mutation of cysteine 39 to alanine resulted in a small, but significant, reduction in TIPARP autoribosylation activity, suggesting that additional amino acid residues are modified, but loss of cysteine 39 did not prevent its ability to repress AHR. Our findings characterize the subcellular localization and mono-ADP-ribosyltransferase activity of TIPARP, identify cysteine as a mono-ADP-ribosylated residue targeted by this enzyme, and confirm the TIPARP-dependent mono-ADP-ribosylation of other protein targets, such as AHR.


Subject(s)
ADP Ribose Transferases/genetics , Cysteine/genetics , Mutation, Missense , Poly(ADP-ribose) Polymerases/genetics , ADP Ribose Transferases/metabolism , ADP-Ribosylation/drug effects , Animals , Biocatalysis/drug effects , COS Cells , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Chlorocebus aethiops , Cysteine/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , MCF-7 Cells , Nucleoside Transport Proteins , Poly(ADP-ribose) Polymerases/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Zinc Fingers/genetics
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(6): 623-635, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28219719

ABSTRACT

Plasma cysteine is strongly associated with body fat mass in human cohorts and diets low in cysteine prevents fat accumulation in mice. It is unclear if plasma cysteine affects fat development or if fat accumulation raises plasma cysteine. To determine if cysteine affects adipogenesis, we differentiated 3T3-L1 preadipocytes in medium with reduced cysteine. Cells incubated in media with 10-20µM cysteine exhibited reduced capacity to differentiate into triacylglycerol-storing mature adipocytes compared with cells incubated with 50µM cysteine. Low cysteine severely reduced expression of peroxisome proliferator-activated receptor gamma2 (Pparγ2) and its target genes perlipin1 (Plin1) and fatty acid binding protein-4 (Fabp4). Expression of stearoyl-CoA desaturase-1 (Scd1), known to be repressed with cysteine depletion, was also reduced with low cysteine. Medium depletion of the essential amino acids leucine, valine, and isoleucine had only a modest effect on adipocyte specific gene expression and differentiation. Stimulation with the PPARγ agonist BRL-49653 or addition of a hydrogen sulfide donor enhanced differentiation of 3T3-L1 cells cultured in low cysteine. This demonstrates that the ability to induce PPARγ expression is preserved when cells are cultured in low cysteine. It therefore appears that cysteine depletion inhibits adipogenesis by specifically affecting molecular pathways required for induction of PPARγ expression, rather than through a general reduction of global protein synthesis. In conclusion, we show that low extracellular cysteine reduces adipocyte differentiation by interfering with PPARγ2 and PPARγ target gene expression. Our results provide further evidence for the hypothesis that plasma cysteine is a casual determinant for body fat mass.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/physiology , Cysteine/metabolism , Fatty Acid-Binding Proteins/metabolism , PPAR gamma/metabolism , Perilipin-1/metabolism , Stearoyl-CoA Desaturase/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cysteine/pharmacology , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation/physiology , Mice , PPAR gamma/genetics , Perilipin-1/genetics , Stearoyl-CoA Desaturase/genetics
4.
Biochem J ; 473(7): 899-910, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26814197

ABSTRACT

Members of the poly-ADP-ribose polymerase (PARP) family catalyse the ADP-ribosylation of target proteins and are known to play important roles in many cellular processes, including DNA repair, differentiation and transcription. The majority of PARPs exhibit mono-ADP-ribosyltransferase activity rather than PARP activity; however, little is known about their biological activity. In the present study, we report that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly-ADP-ribose polymerase (TIPARP), mono-ADP-ribosylates and positively regulates liver X receptor α (LXRα) and LXRß activity. Overexpression of TIPARP enhanced LXR-reporter gene activity. TIPARP knockdown or deletion reduced LXR regulated target gene expression levels in HepG2 cells and in Tiparp(-/-)mouse embryonic fibroblasts (MEFs) respectively. Deletion and mutagenesis studies showed that TIPARP's zinc-finger and catalytic domains were required to enhance LXR activity. Protein interaction studies using TIPARP and LXRα/ß peptide arrays revealed that LXRs interacted with an N-terminal sequence (a.a. 209-236) of TIPARP, which also overlapped with a putative co-activator domain of TIPARP (a.a. 200-225). Immunofluorescence studies showed that TIPARP and LXRα or LXRß co-localized in the nucleus.In vitroribosylation assays provided evidence that TIPARP mono-ADP-ribosylated both LXRα and LXRß. Co-immunoprecipitation (co-IP) studies revealed that ADP-ribosylase macrodomain 1 (MACROD1), but not MACROD2, interacted with LXRs in a TIPARP-dependent manner. This was complemented by reporter gene studies showing that MACROD1, but not MACROD2, prevented the TIPARP-dependent increase in LXR activity. GW3965-dependent increases in hepatic Srebp1 mRNA and protein expression levels were reduced in Tiparp(-/-)mice compared with Tiparp(+/+)mice. Taken together, these data identify a new mechanism of LXR regulation that involves TIPARP, ADP-ribosylation and MACROD1.


Subject(s)
ADP Ribose Transferases/metabolism , Cell Nucleus/metabolism , Orphan Nuclear Receptors/metabolism , Poly(ADP-ribose) Polymerases/metabolism , ADP Ribose Transferases/genetics , Adenosine Diphosphate Ribose/genetics , Adenosine Diphosphate Ribose/metabolism , Animals , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Hep G2 Cells , Humans , Hydrolases/genetics , Hydrolases/metabolism , Liver X Receptors , Mice , Mice, Knockout , Nucleoside Transport Proteins , Orphan Nuclear Receptors/genetics , Poly(ADP-ribose) Polymerases/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
5.
Mar Drugs ; 15(6)2017 May 25.
Article in English | MEDLINE | ID: mdl-28587091

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) function as ligand-activated transcription factors that convert signals in the form of lipids to physiological responses through the activation of metabolic target genes. Due to their key roles in lipid and carbohydrate metabolism, the PPARs are important drug targets. However, for several of the PPAR drugs currently in use, adverse side effects have been reported. In an effort to identify compounds from marine organisms that may serve as molecular scaffolds for the development of novel and safer PPAR-targeting drugs, we performed a bioassay-guided screening of organic extracts made from organisms supplied by the Norwegian Biobank of Arctic Marine Organisms (Marbank). Among several interesting hits, we identified two poorly described isomeric oxo-fatty acids from the microalgae Chaetoceros karianus for which we provide the first evidence that they might display dual specificity towards human PPARα and PPARγ. Principal component analysis showed that C. karianus stood out from other Chaetoceros species, both with respect to the metabolic profile and the PPAR activity. The isolation of these compounds holds the potential of uncovering a PPAR pharmacophore with tunable activity and specificity.


Subject(s)
Diatoms/chemistry , Fatty Acids/chemistry , Fatty Acids/pharmacology , PPAR alpha/agonists , PPAR alpha/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Isomerism , Ligands , Metabolome/drug effects , Microalgae/chemistry
6.
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
7.
Biochem Biophys Res Commun ; 450(4): 1525-30, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25035925

ABSTRACT

Interleukin (IL)-10 is a prototypical anti-inflammatory cytokine that has been shown to attenuate atherosclerosis development. In addition to its anti-inflammatory properties, the anti-atherogenic effect of IL-10 has recently also been suggested to reflect a complex effect of IL-10 on lipid metabolism in macrophages. In the present study we examined the effects of IL-10 on cholesterol efflux mechanism in lipid-loaded THP-1 macrophages. Our main findings were: (i) IL-10 significantly enhanced cholesterol efflux induced by fetal-calf serum, high-density lipoprotein (HDL)2 and apolipoprotein A-1. (ii) The IL-10-mediated effects on cholesterol efflux were accompanied by an increased IL-10-mediated expression of the ATP-binding cassette transporters ABCA1 and ABCG1, that was further enhanced when the cells were co-activated with the liver X receptor (LXR)α agonist (22R)-hydroxycholesterol. (iii) The effect of LXRα activation on the IL-10-mediated effects on the ATP-binding cassette transporters seems to include enhancing effects on the IL-10 receptor 1 (IL10R1) expression and interaction with STAT-3 signaling. (iv) These enhancing effects on ABCA1 and ABCG1 was not seen when the cells were stimulated with the IL-10 family members IL-22 and IL-24. This study suggests that the anti-atherogenic properties of IL-10 may include enhancing effects on cholesterol efflux mechanism that involves cross-talk with LXRα activation.


Subject(s)
Cholesterol/metabolism , Interleukin-10/physiology , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , Biological Transport , Cell Line , Humans , Liver X Receptors , Protein Binding , Real-Time Polymerase Chain Reaction
8.
J Lipid Res ; 54(7): 1949-63, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23606724

ABSTRACT

The surface of lipid droplets (LDs) in various cell types is coated with perilipin proteins encoded by the Plin genes. Perilipins regulate LD metabolism by selectively recruiting lipases and other proteins to LDs. We have studied the expression of perilipins in mouse muscle. The glycolytic fiber-enriched gastrocnemius muscle expresses predominantly Plin2-4. The oxidative fiber-enriched soleus muscle expresses Plin2-5. Expression of Plin2 and Plin4-5 is elevated in gastrocnemius and soleus muscles from mice fed a high-fat diet. This effect is preserved in peroxisome proliferator-activated receptor (PPAR)α-deficient mice. Mouse muscle derived C2C12 cells differentiated into glycolytic fibers increase transcription of these Plins when exposed to various long chain fatty acids (FAs). To understand how FAs regulate Plin genes, we used specific activators and antagonists against PPARs, Plin promoter reporter assays, chromatin immunoprecipitation, siRNA, and animal models. Our analyses demonstrate that FAs require PPARδ to induce transcription of Plin4 and Plin5. We further identify a functional PPAR binding site in the Plin5 gene and establish Plin5 as a novel direct PPARδ target in muscle. Our study reveals that muscle cells respond to elevated FAs by increasing transcription of several perilipin LD-coating proteins. This induction renders the muscle better equipped to sequester incoming FAs into cytosolic LDs.


Subject(s)
Fatty Acids/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , PPAR delta/metabolism , Animals , Binding Sites/drug effects , Cells, Cultured , Fatty Acids/administration & dosage , Gene Silencing/drug effects , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , PPAR delta/chemistry , PPAR delta/deficiency , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Basic Res Cardiol ; 108(1): 323, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23266787

ABSTRACT

Liver X receptor (LXR)-α and -ß play a major role in lipid and glucose homeostasis. Their expression and function in the heart is not well characterized. Our aim was to describe the expression of LXRs in the murine heart, and to determine effects of cardiac LXR activation on target gene expression, lipid homeostasis and ischemia. Both LXRα and -ß were expressed in heart tissues, HL-1 cells and isolated cardiomyocytes as determined by qRT-PCR. Elevated cardiac expression of LXR target genes and LXRß was observed 24 h after in vivo permanent coronary artery ligation. The synthetic LXR agonist GW3965 induced mRNA expression of the LXR target genes in HL-1 cells and isolated cardiomyocytes. This was associated with a buildup of intracellular triglycerides and expanding lipid droplets as quantified by confocal microscopy. Mice injected with GW3965 had cardiac LXR activation as judged by increased target gene expression and lipid droplet accumulation. GW3965 in vivo and in vitro increased expression of genes inducing triglyceride synthesis, and altered expression of lipid droplet-binding protein genes. GW3965 protected HL-1 cells against hypoxia-reoxygenation induced apoptosis. LXR activation by GW3965 in vivo prior to heart isolation and perfusion with induced global ischemia and reperfusion improved left ventricular contractile function and decreased infarct size. In conclusion, LXRs are expressed in the murine heart in the basal state, and are activated by myocardial infarction. Activation of LXR by the synthetic agonist GW3965 is associated with intracardiac accumulation of lipid droplets and protection against myocardial ischemia-reperfusion injury.


Subject(s)
Lipid Metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Orphan Nuclear Receptors/physiology , Animals , Apoptosis/drug effects , Benzoates/pharmacology , Benzylamines/pharmacology , Cells, Cultured , Intracellular Space/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Triglycerides/metabolism
10.
Tidsskr Nor Laegeforen ; 138(3)2018 02 06.
Article in Norwegian | MEDLINE | ID: mdl-29411567
11.
J Biol Chem ; 285(3): 1607-15, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19933273

ABSTRACT

Post-translational modification of nucleocytoplasmic proteins by O-linked beta-N-acetylglucosamine (O-GlcNAc) has for the last 25 years emerged as an essential glucose-sensing mechanism. The liver X receptors (LXRs) function as nutritional sensors for cholesterol-regulating lipid metabolism, glucose homeostasis, and inflammation. LXRs are shown to be post-translationally modified by phosphorylation, acetylation, and sumoylation, affecting their target gene specificity, stability, and transactivating and transrepressional activity, respectively. In the present study, we show for the first time that LXRalpha and LXRbeta are targets for glucose-hexosamine-derived O-GlcNAc modification in human Huh7 cells. Furthermore, we observed increased hepatic LXRalpha O-GlcNAcylation in vivo in refed mice and in streptozotocin-induced refed diabetic mice. Importantly, induction of LXRalpha O-GlcNAcylation in both mouse models was concomitant with increased expression of the lipogenic gene SREBP-1c (sterol regulatory element-binding protein 1c). Furthermore, glucose increased LXR/retinoic acid receptor-dependent activation of luciferase reporter activity driven by the mouse SREBP-1c promoter via the hexosamine biosynthetic pathway in Huh7 cells. Altogether, our results suggest that O-GlcNAcylation of LXR is a novel mechanism by which LXR acts as a glucose sensor affecting LXR-dependent gene expression, substantiating the crucial role of LXR as a nutritional sensor in lipid and glucose metabolism.


Subject(s)
Acetylglucosamine/metabolism , Glucose/pharmacology , Orphan Nuclear Receptors/metabolism , Animals , Cell Line, Tumor , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Glycosylation , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Mice , Promoter Regions, Genetic/genetics , Retinoid X Receptors/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Streptozocin/pharmacology , Transcriptional Activation/drug effects
12.
J Lipid Res ; 51(7): 1886-96, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20219900

ABSTRACT

Placental fatty acid transport and metabolism are important for proper growth and development of the feto-placental unit. The nuclear receptors, liver X receptors alpha and beta (LXRalpha and LXRbeta), are key regulators of lipid metabolism in many tissues, but little is known about their role in fatty acid transport and metabolism in placenta. The current study investigates the LXR-mediated regulation of long-chain acyl-CoA synthetase 3 (ACSL3) and its functions in human placental trophoblast cells. We demonstrate that activation of LXR increases ACSL3 expression, acyl-CoA synthetase activity, and fatty acid uptake in human tropholast cells. Silencing of ACSL3 in these cells attenuates the LXR-mediated increase in acyl-CoA synthetase activity. Furthermore, we show that ACSL3 is directly regulated by LXR through a conserved LXR responsive element in the ACSL3 promoter. Our results suggest that LXR plays a regulatory role in fatty acid metabolism by direct regulation of ACSL3 in human placental trophoblast cells.


Subject(s)
Coenzyme A Ligases/metabolism , Orphan Nuclear Receptors/metabolism , Placenta/metabolism , Trophoblasts/metabolism , Animals , Base Sequence , Cell Line , Coenzyme A Ligases/genetics , Fatty Acids/metabolism , Female , Humans , Liver X Receptors , Microarray Analysis , Molecular Sequence Data , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Placenta/cytology , Pregnancy , Sequence Alignment , Trophoblasts/cytology
13.
Biochim Biophys Acta ; 1791(2): 140-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19136075

ABSTRACT

Dietary essential fatty acids linoleic acid and alpha-linolenic acid are converted to arachidonic-, eicosapentaenoic-, and docosahexaenoic acid under tight regulation by nutritional status and hormones. Hepatic fatty acid elongase 5 (Elovl5) elongates C18-20 polyunsaturated fatty acids (PUFAs) and is important for biosynthesis of C20-22 PUFAs. We demonstrate that Liver X Receptor alpha (LXRalpha) and sterol regulatory binding protein-1c (SREBP-1c) regulate hepatic Elovl5 expression. LXRalpha and LXRbeta play different roles in maintenance of basal expression of Elovl5. LXRalpha is necessary for basal as well as LXR agonist induced Elovl5 transcription. Promoter studies revealed that the mouse Elovl5 gene is a direct SREBP-1c target. The up-regulation of Elovl5 expression by LXR agonist is likely secondary to the induction of SREBP-1c. PUFAs repress expression of SREBP-1c and Elovl5, but when combined with LXR ligand stimulation, which increases SREBP-1c mRNA and nuclear SREBP-1c, Elovl5 mRNA levels are restored to normal. Our studies suggest that an LXRalpha-SREBP-1c pathway plays a regulatory role in hepatic biosynthesis of PUFAs through transcriptional activation of Elovl5 as well as other desaturases. The stimulatory role of LXRalpha-SREBP-1c in the production of PUFAs enables the possibility for a feedback regulation of hepatic lipogenesis through PUFA mediated repression of SREBP-1c expression.


Subject(s)
Acetyltransferases/genetics , DNA-Binding Proteins/physiology , Liver/enzymology , Receptors, Cytoplasmic and Nuclear/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Acetyltransferases/metabolism , Animals , Blotting, Western , COS Cells , Cells, Cultured , Chlorocebus aethiops , Electrophoretic Mobility Shift Assay , Fatty Acid Elongases , Fatty Acids, Unsaturated/metabolism , Hepatoblastoma/genetics , Hepatoblastoma/metabolism , Hepatoblastoma/pathology , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver X Receptors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Orphan Nuclear Receptors , Promoter Regions, Genetic , RNA, Messenger , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Sulfonamides/pharmacology
14.
Biochem Biophys Res Commun ; 392(3): 436-41, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20079709

ABSTRACT

The present study addresses the insulin sensitivity in mice deficient in LXRbeta (LXRbeta(-/-)) as well as in wild type (wt) mice assessed by hyperinsulinemic euglycemic clamp. Wt and LXRbeta(-/-) mice were fed either a normal chow diet or a high fat and high cholesterol diet (HFCD), and insulin sensitivity was assessed by hyperinsulinemic euglycemic clamps. We show that LXRbeta(-/-) mice have reduced insulin clearance during hyperinsulinemic clamps upon feeding both HFCD and a regular chow diet. Moreover we also observed reduced hepatic inflammation in LXRbeta(-/-) mice compared to wt mice upon feeding an HFCD, despite equal levels of hepatic steatosis. In summary, our results indicate that LXRbeta(-/-) mice have reduced insulin clearance during hyperinsulinemic euglycemic clamps and also reduced hepatic inflammation upon feeding an HFCD for 26weeks.


Subject(s)
Hepatitis/genetics , Insulin Resistance/genetics , Insulin/metabolism , Liver/metabolism , Orphan Nuclear Receptors/physiology , Animals , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Diet/adverse effects , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Fatty Liver/genetics , Glucose Clamp Technique , Insulin/pharmacology , Liver/pathology , Liver X Receptors , Male , Mice , Mice, Knockout , Orphan Nuclear Receptors/genetics , Triglycerides/metabolism
15.
BMC Med Genet ; 11: 144, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20939869

ABSTRACT

BACKGROUND: The liver X receptors (LXR) α and ß regulate lipid and carbohydrate homeostasis and inflammation. Lxrß⁻/⁻ mice are glucose intolerant and at the same time lean. We aimed to assess the associations between single nucleotide polymorphisms (SNPs) in LXRß and risk of type 2 diabetes mellitus (T2DM), obesity and related traits in 3 separate cohort studies. METHODS: Twenty LXRß SNPs were identified by sequencing and genotyped in the HUNT2 adult nested case-control study for T2DM (n = 835 cases/1986 controls). Five tag-SNPs (rs17373080, rs2695121, rs56151148, rs2303044 and rs3219281), covering 99.3% of the entire common genetic variability of the LXRß gene were identified and genotyped in the French MONICA adult study (n = 2318) and the European adolescent HELENA cross-sectional study (n = 1144). In silico and in vitro functionality studies were performed. RESULTS: We identified suggestive or significant associations between rs17373080 and the risk of (i) T2DM in HUNT2 (OR = 0.82, p = 0.03), (ii) obesity in MONICA (OR = 1.26, p = 0.05) and (iii) overweight/obesity in HELENA (OR = 1.59, p = 0.002). An intron 4 SNP (rs28514894, a perfect proxy for rs17373080) could potentially create binding sites for hepatic nuclear factor 4 alpha (HNF4α) and nuclear factor 1 (NF1). The C allele of rs28514894 was associated with ~1.25-fold higher human LXRß basal promoter activity in vitro. However, no differences between alleles in terms of DNA binding and reporter gene transactivation by HNF4α or NF1 were observed. CONCLUSIONS: Our results suggest that rs17373080 in LXRß is associated with T2DM and obesity, maybe via altered LXRß expression.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Obesity/genetics , Orphan Nuclear Receptors/genetics , Adolescent , Adult , Aged , Alleles , Binding Sites , Cohort Studies , Europe , Female , France , Genetic Predisposition to Disease , Genotype , Hepatocyte Nuclear Factor 4/metabolism , Humans , Introns , Liver X Receptors , Male , Middle Aged , NFI Transcription Factors/metabolism , Norway , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
16.
Cells ; 9(5)2020 05 13.
Article in English | MEDLINE | ID: mdl-32414201

ABSTRACT

The cholesterol-sensing nuclear receptor liver X receptor (LXR) and the glucose-sensing transcription factor carbohydrate responsive element-binding protein (ChREBP) are central players in regulating glucose and lipid metabolism in the liver. More knowledge of their mechanistic interplay is needed to understand their role in pathological conditions like fatty liver disease and insulin resistance. In the current study, LXR and ChREBP co-occupancy was examined by analyzing ChIP-seq datasets from mice livers. LXR and ChREBP interaction was determined by Co-immunoprecipitation (CoIP) and their transactivity was assessed by real-time quantitative polymerase chain reaction (qPCR) of target genes and gene reporter assays. Chromatin binding capacity was determined by ChIP-qPCR assays. Our data show that LXRα and ChREBPα interact physically and show a high co-occupancy at regulatory regions in the mouse genome. LXRα co-activates ChREBPα and regulates ChREBP-specific target genes in vitro and in vivo. This co-activation is dependent on functional recognition elements for ChREBP but not for LXR, indicating that ChREBPα recruits LXRα to chromatin in trans. The two factors interact via their key activation domains; the low glucose inhibitory domain (LID) of ChREBPα and the ligand-binding domain (LBD) of LXRα. While unliganded LXRα co-activates ChREBPα, ligand-bound LXRα surprisingly represses ChREBPα activity on ChREBP-specific target genes. Mechanistically, this is due to a destabilized LXRα:ChREBPα interaction, leading to reduced ChREBP-binding to chromatin and restricted activation of glycolytic and lipogenic target genes. This ligand-driven molecular switch highlights an unappreciated role of LXRα in responding to nutritional cues that was overlooked due to LXR lipogenesis-promoting function.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/agonists , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Liver X Receptors/agonists , Liver X Receptors/metabolism , Transcriptional Activation/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Cell Line, Tumor , Chromatin/metabolism , Female , Genome , Humans , Ligands , Liver/metabolism , Liver X Receptors/chemistry , Male , Mice, Inbred C57BL , Models, Biological , Protein Binding , Protein Domains , Response Elements/genetics
17.
Biochim Biophys Acta ; 1771(2): 210-27, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17234449

ABSTRACT

The PAT family (originally named for Perilipin, ADFP and TIP47) now includes four members: Perilipins, ADFP, TIP47 and S3-12. Significant primary sequence homology and the ability to associate with lipid storage droplets (LSDs) are well conserved within this family and across species. In this study, we have characterized a novel PAT protein, lipid storage droplet protein 5 (LSDP5) of 463 residues. A detailed sequence analysis of all murine PAT proteins reveals that LSDP5, TIP47 and ADFP share the highest order of sequence similarity, whereas perilipin and S3-12 have more divergent carboxyl- and amino-termini, respectively. Ectopically-expressed YFP-LSDP5 or flag-LSDP5 fusion proteins associate with LSDs. In accord with recent published data for perilipin, forced expression of LSDP5 in CHO cells inhibits lipolysis of intracellular LSDs. The LSDP5 gene is primarily transcribed in cells that actively oxidize fatty acids, such as heart, red muscle and liver. Expression of LSDP5 is stimulated by ligand activation of peroxisomal proliferator-activated receptor alpha (PPARalpha), and significantly reduced in liver and heart in the absence of this transcription factor. PPARalpha is generally required for regulation of fatty acid metabolism during fasting, but fasting induces LSDP5 mRNA in liver even in the absence of PPARalpha.


Subject(s)
Fatty Acids/metabolism , Phosphoproteins/metabolism , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins , Chlorocebus aethiops , Chromosomes, Human, Pair 17 , Exons , Fasting/metabolism , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidation-Reduction , PPAR alpha/metabolism , Perilipin-1 , Perilipin-5 , Phosphoproteins/genetics , Sequence Homology, Amino Acid , Tissue Distribution
18.
Eur J Med Chem ; 155: 736-753, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29940464

ABSTRACT

Obesity and associated disorders such as metabolic syndrome and type 2 diabetes (T2D) have reached epidemic proportions. Several natural products have been reported as Peroxisome Proliferator-Activated Receptor (PPAR) agonists, functioning as lead compounds towards developing new anti-diabetic drugs due to adverse side effects of existing PPAR drugs. We recently isolated and identified (7E)-9-oxohexadec-7-enoic acid (1) and (10E)-9-oxohexadec-10-enoic acid (2) from the marine algae Chaetoceros karianus. Herein we report the total synthesis, pharmacological characterization, and biological evaluations of these naturally occurring oxo-fatty acids (oFAs). The syntheses of 1 and 2 afforded sufficient material for extensive biological evaluations. Both oFAs show an appreciable dose-dependent activation of PPARα and -γ, with EC50 values in the micromolar range, and an ability to regulate important PPAR target genes in hepatocytes and adipocytes. Moreover, both 1 and 2 are able to drive adipogenesis when evaluated in the Simpson-Golabi-Behmel syndrome (SGBS) pre-adipocyte cell model, but with lowered expression of adipocyte markers and reduced lipid accumulation compared to the drug rosiglitazone. This seems to be caused by a transient upregulation of PPARγ and C/EBPα expression. Importantly, whole transcriptome analysis shows that both compounds induce anti-diabetic gene programs in adipocytes by upregulating insulin-sensitizing adipokines and repressing pro-inflammatory cytokines.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Keto Acids/pharmacology , Microalgae/chemistry , PPAR alpha/agonists , PPAR gamma/agonists , Palmitic Acids/pharmacology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Diabetes Mellitus, Type 2/genetics , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Keto Acids/chemical synthesis , Keto Acids/chemistry , Molecular Structure , PPAR alpha/genetics , PPAR gamma/genetics , Palmitic Acids/chemical synthesis , Palmitic Acids/chemistry , Structure-Activity Relationship
19.
Biochim Biophys Acta ; 1761(12): 1515-22, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17055780

ABSTRACT

The nuclear liver X receptors (LXRalpha and beta) are regulators of lipid and cholesterol metabolism. Oxysterols are known LXR ligands, but the functional role of hydroxycholesterols is at present unknown. In human myotubes, chronic exposure to the LXR ligand T0901317 promoted formation of diacylglycerol (DAG) and triacylglycerol (TAG), 22-R-hydroxycholesterol (22-R-HC) had no effect, and 22-S-hydroxycholesterol (22-S-HC) reduced the formation. In accordance with this, 22-HC and T0901317 regulated the expression of fatty acid transporter CD36, stearoyl-CoA desaturase-1, acyl-CoA synthetase long chain family member 1 and fatty acid synthase (FAS) differently; all genes were increased by T0901317, 22-R-HC did not change their expression level, while 22-S-HC reduced it. Transfection studies confirmed that the FAS promoter was activated by T0901317 and repressed by 22-S-HC through an LXR response element in the promoter. Both 22-R-HC and T0901317 increased gene expression of LXRalpha, sterol regulatory element-binding protein 1c and ATP-binding cassette transporter A1, while 22-S-HC had little effect. In summary, 22-R-HC regulated lipid metabolism and mRNA expression of some LXR target genes in human myotubes differently than T0901317. Moreover, 22-S-HC did not behave like an inactive ligand; it reduced synthesis of complex lipids and repressed certain genes involved in lipogenesis and lipid handling.


Subject(s)
Hydroxycholesterols/pharmacology , Lipid Metabolism/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Sulfonamides/pharmacology , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Humans , Hydrocarbons, Fluorinated , Hydroxycholesterols/metabolism , Ligands , Liver X Receptors , Orphan Nuclear Receptors , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Sulfonamides/metabolism , Transfection , Triglycerides/biosynthesis , fas Receptor/genetics
20.
Mol Cell Biol ; 22(16): 5989-99, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12138207

ABSTRACT

Wnt signaling maintains preadipocytes in an undifferentiated state. When Wnt signaling is enforced, 3T3-L1 preadipocytes no longer undergo adipocyte conversion in response to adipogenic medium. Here we used microarray analyses to identify subsets of genes whose expression is aberrant when differentiation is blocked through enforced Wnt signaling. Furthermore, we used the microarray data to identify potentially important adipocyte genes and chose one of these, the liver X receptor alpha (LXR alpha), for further analyses. Our studies indicate that enforced Wnt signaling blunts the changes in gene expression that correspond to mitotic clonal expansion, suggesting that Wnt signaling inhibits adipogenesis in part through dysregulation of the cell cycle. Experiments designed to uncover the potential role of LXR alpha in adipogenesis revealed that this transcription factor, unlike CCAAT/enhancer binding protein alpha and peroxisome proliferator-activated receptor gamma, is not adipogenic but rather inhibits adipogenesis if inappropriately expressed and activated. However, LXR alpha has several important roles in adipocyte function. Our studies show that this nuclear receptor increases basal glucose uptake and glycogen synthesis in 3T3-L1 adipocytes. In addition, LXR alpha increases cholesterol synthesis and release of nonesterified fatty acids. Finally, treatment of mice with an LXR alpha agonist results in increased serum levels of glycerol and nonesterified fatty acids, consistent with increased lipolysis within adipose tissue. These findings demonstrate new metabolic roles for LXR alpha and increase our understanding of adipogenesis.


Subject(s)
Adipocytes/physiology , Cell Differentiation/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Zebrafish Proteins , Adipocytes/drug effects , Adipose Tissue/cytology , Animals , Anticholesteremic Agents/pharmacology , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fatty Acids, Nonesterified/blood , Female , Gene Expression Profiling , Glycerol/blood , Humans , Hydrocarbons, Fluorinated , Ligands , Lipid Metabolism , Liver X Receptors , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors , Phenotype , Proto-Oncogene Proteins/genetics , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/physiology , Sulfonamides , Wnt Proteins
SELECTION OF CITATIONS
SEARCH DETAIL