Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Intern Med ; 286(6): 660-675, 2019 12.
Article in English | MEDLINE | ID: mdl-31251843

ABSTRACT

BACKGROUND: Hyperlipidaemia is a major risk factor for cardiovascular disease, and atherosclerosis is the underlying cause of both myocardial infarction and stroke. We have previously shown that the Pro251 variant of perilipin-2 reduces plasma triglycerides and may therefore be beneficial to reduce atherosclerosis development. OBJECTIVE: We sought to delineate putative beneficial effects of the Pro251 variant of perlipin-2 on subclinical atherosclerosis and the mechanism by which it acts. METHODS: A pan-European cohort of high-risk individuals where carotid intima-media thickness has been assessed was adopted. Human primary monocyte-derived macrophages were prepared from whole blood from individuals recruited by perilipin-2 genotype or from buffy coats from the Karolinska University hospital blood central. RESULTS: The Pro251 variant of perilipin-2 is associated with decreased intima-media thickness at baseline and over 30 months of follow-up. Using human primary monocyte-derived macrophages from carriers of the beneficial Pro251 variant, we show that this variant increases autophagy activity, cholesterol efflux and a controlled inflammatory response. Through extensive mechanistic studies, we demonstrate that increase in autophagy activity is accompanied with an increase in liver-X-receptor (LXR) activity and that LXR and autophagy reciprocally activate each other in a feed-forward loop, regulated by CYP27A1 and 27OH-cholesterol. CONCLUSIONS: For the first time, we show that perilipin-2 affects susceptibility to human atherosclerosis through activation of autophagy and stimulation of cholesterol efflux. We demonstrate that perilipin-2 modulates levels of the LXR ligand 27OH-cholesterol and initiates a feed-forward loop where LXR and autophagy reciprocally activate each other; the mechanism by which perilipin-2 exerts its beneficial effects on subclinical atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Autophagy , Carotid Intima-Media Thickness , Liver X Receptors/metabolism , Macrophages/metabolism , Perilipin-2/metabolism , Aged , Disease Progression , Europe , Female , Foam Cells/metabolism , Humans , Lipoproteins/metabolism , Longitudinal Studies , Male , Middle Aged
2.
Dig Liver Dis ; 51(7): 1016-1022, 2019 07.
Article in English | MEDLINE | ID: mdl-30630736

ABSTRACT

Depletion of the cholesterol esterifying enzyme acyl-Coenzyme A: cholesterol acyltransferase 2 (ACAT2, encoded by Soat2) protects mice from atherosclerosis, diet-induced hypercholesterolemia, and hepatic steatosis when fed high-cholesterol diet. The glucose transporter 2 (GLUT2) represents the main gate of glucose uptake by the liver. Lipid synthesis from glucose (de novo lipogenesis; DNL) plays a pivotal role in the development of hepatic steatosis. Inhibition of DNL is a successful approach to reverse hepatic steatosis, as shown by different studies in mice and humans. Here we aimed to investigate whether depletion of Soat2 per se can reduce hepatic steatosis, also in the presence of very low levels of cholesterol in the diet, and the underlying mechanisms. Female Soat2-/- and wild type mice were either fed high-fat or high-carbohydrate diet and both contained <0.05% (w/w) cholesterol. Analysis in serum, liver, muscles and adipose tissues were performed. We found Soat2-/- mice fed high-fat, low-cholesterol diet to have less hepatic steatosis, decreased expression of genes involved in DNL and lower hepatic GLUT2. Similar findings were found in Soat2-/- mice fed high-carbohydrate, low-cholesterol diet. CONCLUSION: Depletion of Soat2 reduces hepatic steatosis independently of the presence of high levels of cholesterol in the diet. Our study provides a link between hepatic cholesterol esterification, DNL, and GLUT2.


Subject(s)
Glucose Transporter Type 2/genetics , Hyperlipidemias/genetics , Lipogenesis/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Sterol O-Acyltransferase/genetics , Animals , Cholesterol/metabolism , Diet, High-Fat , Female , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Sterol O-Acyltransferase 2
3.
J Intern Med ; 281(6): 534-553, 2017 06.
Article in English | MEDLINE | ID: mdl-28295777

ABSTRACT

Following the continuous accumulation of evidence supporting the beneficial role of reducing low-density lipoprotein cholesterol (LDL-C) levels in the treatment and prevention of atherosclerotic cardiovascular disease and its complications, therapeutic possibilities now exist to lower LDL-C to very low levels, similar to or even lower than those seen in newborns and nonhuman species. In addition to the important task of evaluating potential side effects of such treatments, the question arises whether extremely low LDL-C levels per se may provoke adverse effects in humans. In this review, we summarize information from studies of human cellular and organ physiology, phenotypic characterization of rare genetic diseases of lipid metabolism, and experience from clinical trials. Specifically, we emphasize the importance of the robustness of the regulatory systems that maintain balanced fluxes and levels of cholesterol at both cellular and organismal levels. Even at extremely low LDL-C levels, critical capacities of steroid hormone and bile acid production are preserved, and the presence of a cholesterol blood-brain barrier protects cells in the central nervous system. Apparent relationships sometimes reported between less pronounced low LDL-C levels and disease states such as cancer, depression, infectious disease and others can generally be explained as secondary phenomena. Drug-related side effects including an increased propensity for development of type 2 diabetes occur during statin treatment, whilst further evaluation of more potent LDL-lowering treatments such as PCSK9 inhibitors is needed. Experience from the recently reported and ongoing large event-driven trials are of great interest, and further evaluation including careful analysis of cognitive functions will be important.


Subject(s)
Cholesterol, LDL/blood , Bone and Bones/metabolism , Brain/physiology , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Immune System Phenomena , Lipoproteins, LDL/blood , Mutation , Neoplasms/blood , Proprotein Convertase 9/genetics , Risk Factors
4.
Acta Physiol (Oxf) ; 217(3): 184-216, 2016 07.
Article in English | MEDLINE | ID: mdl-26748938

ABSTRACT

Thyroid hormones (THs) are essential for the regulation of development and metabolism in key organs. THs produce biological effects both by directly affecting gene expression through the interaction with nuclear receptors (genomic effects) and by activating protein kinases and/or ion channels (short-term effects). Such activations can be either direct, in the case of ion channels, or mediated by membrane or cytoplasmic receptors. Short-term-activated signalling pathways often play a role in the regulation of genomic effects. Several TH intermediate metabolites, which were previously considered without biological activity, have now been associated with a broad range of actions, mostly attributable to short-term effects. Here, we give an overview of the physiological roles and mechanisms of action of THs, focusing on the emerging position that TH metabolites are acquiring as important regulators of physiology and metabolism.


Subject(s)
Thyroid Hormones/metabolism , Thyroid Hormones/physiology , Animals , Antithyroid Agents/pharmacology , Humans , Hypothalamo-Hypophyseal System/metabolism , Signal Transduction , Thyroid Gland/metabolism , Thyroid Hormones/biosynthesis
5.
Atherosclerosis ; 237(2): 544-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25463087

ABSTRACT

Thyroid hormone reduces plasma cholesterol and increases expression of low-density lipoprotein receptor (LDL-R) in liver, an effect mediated by thyroid receptor ß (TRß). The selective TRß modulator GC-1 also enhances several steps in reverse cholesterol transport and can decrease serum cholesterol independently of LDL-R. To test whether GC-1 reduces atherosclerosis and to determine which mechanisms are active, we treated ApoE deficient mice with atherogenic diet ± GC-1. GC-1 reduced cholesteryl esters in aorta after 20 weeks. Serum free and esterified cholesterol were reduced after 1 and 10 weeks, but not 20 weeks. Hepatic bile acid synthesis and LDL-R expression was elevated after 1, 10 and 20 weeks, without changes in hepatic de novo cholesterol synthesis. GC-1 increased faecal neutral sterols and reduced serum campesterol after 1 week, indicating reduced intestinal cholesterol absorption. After 20 weeks, GC-1 increased faecal bile acids, but not faecal neutral sterols. Hepatic scavenger receptor B1 (SR-B1) expression was decreased by GC-1. We conclude that GC-1 delays the onset of atherosclerosis in ApoE deficient mice. Since ApoE is needed for hepatic cholesterol reabsorption by LDL-R, this supports the idea that GC-1 reduces serum cholesterol independently of LDL-R by increasing hepatic bile acid synthesis. GC-1 lipid-lowering effects in ApoE deficient mice may also be partly due to reduced intestinal cholesterol absorption. Since reductions in serum cholesterol are reversed at longer times, these GC-1 dependent effects may not be enough for sustained cholesterol reduction in long term treatments.


Subject(s)
Acetates/pharmacology , Atherosclerosis/drug therapy , Cholesterol/blood , Phenols/pharmacology , Thyroid Hormone Receptors beta/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Bile Acids and Salts/chemistry , Biological Transport , Cholesterol/analogs & derivatives , Cholesterol/chemistry , Cholesterol/metabolism , Disease Models, Animal , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phytosterols/blood , Receptors, LDL/metabolism , Sterols/chemistry , Time Factors
6.
Transplant Proc ; 45(5): 1780-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23769043

ABSTRACT

INTRODUCTION: Hepatocyte transplantation, a promising treatment for patients with acute hepatic failure or metabolic liver diseases, requires improvement in engraftment as well as long-term function of the liver cells. We established a hepatocyte transplantation model in apolipoprotein E (ApoE) knockout mice, evaluating serum ApoE and lipoprotein profiles as markers of engraftment of transplanted wild-type hepatocytes. Herein we have described a method to monitor the function of transplanted hepatocytes at low levels of engraftment, corresponding to those reported in clinical cases. We also investigated whether pretreatment with anakinra, an anti-interleukin-1 antagonist, methylprednisolone, or a combination of the two agents improved engraftment. METHODS: ApoE (-/-) mice were transplanted with hepatocytes isolated from wild-type C57/bl6 mice. A total of 6 × 10(6) hepatocytes were transplanted by 3 separate intrasplenic injections. Animals were treated before transplantation and daily thereafter for 7 days with anakinra, methylprednisolone, or a combination of both. Graft function was monitored by lipoprotein analysis and quantification of ApoE by enzyme-linked immunosorbent assay. Expression of hepatic ApoE mRNA was quantitated by reverse-transcriptase polymerase chain reaction. RESULTS: Treatment with anakinra with or without methylprednisolone did not significantly increase serum or hepatic mRNA ApoE expression. The low level of hepatocyte engraftment did not normalize lipoprotein profiles, but produced a significant decline in very low-density lipoprotein and total cholesterol. Repeated transplantations significantly enhanced liver repopulation; serum ApoE levels increased with each infusion, correlating well with hepatic mRNA expression. CONCLUSIONS: The model of serum ApoE, a sensitive marker of engraftment and transplanted hepatocyte function, allowed us to study hepatocyte transplantation in a clinically relevant manner, that is, without pretreatments such as retrorsine or carbon tetrachloride.


Subject(s)
Apolipoproteins E/blood , Biomarkers/blood , Hepatocytes/transplantation , Models, Animal , Animals , Apolipoproteins E/genetics , Cholesterol/blood , Enzyme-Linked Immunosorbent Assay , Female , Liver/metabolism , Mice , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
J Intern Med ; 272(5): 452-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22329358

ABSTRACT

OBJECTIVES: Liver X receptors (LXRs) are essential for the regulation of intestinal cholesterol absorption. Because two isoforms exist, LXRα and LXRß, with overlapping but not identical functions, we investigated whether LXRα and LXRß exert different effects on intestinal cholesterol absorption. DESIGN: Wild-type (WT), LXRα(-/-) and LXRß(-/-) mice were fed control diet, 0.2% cholesterol-enriched diet or 0.2% cholesterol-enriched diet plus the LXR agonist GW3965. RESULTS: When fed a control diet, all three genotypes showed similar levels of cholesterol absorption. Of interest, a significant increase in cholesterol absorption was found in the LXRα(-/-) mice, but not in the WT or LXRß(-/-) animals, when fed a diet enriched with 0.2% cholesterol or 0.2% cholesterol + GW3965. Reduced faecal neutral sterol excretion and a hydrophobic bile acid profile were also observed in LXRα(-/-) mice. Greater increases in the apolipoprotein (apo)B-containing lipoproteins in serum were seen in the LXRα(-/-) mice. A 0.2% cholesterol +GW3965 diet suppressed intestinal Npc1l1 protein expression to the same extent for all genotypes, while Abca1 and Abcg5 were elevated to the same degree. CONCLUSIONS: In the intestine, LXRα and LXRß seem to exert similar effects on expression of cholesterol-transporting proteins such as Npc1l1. Selective activation of LXRß may generate effects such as increased cholesterol absorption and elevated serum levels of apoB-containing lipoproteins, which seem to be counteracted by LXRα. Therefore, an intestinal LXRß-specific pathway might exist in terms of cholesterol transportation in addition to the main pathway.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/blood , Intestinal Absorption , Lipoproteins/metabolism , Liver/metabolism , Orphan Nuclear Receptors/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/genetics , Analysis of Variance , Animals , Benzoates/administration & dosage , Benzylamines/administration & dosage , Bile/metabolism , Cholesterol, Dietary/administration & dosage , Intestine, Small/metabolism , Lipid Metabolism , Lipoproteins/genetics , Liver X Receptors , Male , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Models, Animal , Protein Isoforms , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/methods
8.
J Intern Med ; 270(4): 377-87, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21486371

ABSTRACT

BACKGROUND: Oxysterols such as 24S-hydroxycholesterol (OHC) and 27-OHC are intermediates of cholesterol excretion pathways. In addition, they are putative endogenous agonists of the liver X receptor (LXR) class of nuclear hormone receptors and are thought to be important mediators of cholesterol-dependent gene regulation. 24S-OHC is one of the most efficient endogenous LXR agonists known and is present in the brain and in the circulation at relatively high levels. OBJECTIVES: To explore the regulatory importance of 24S-OHC in vivo. DESIGN: We developed a transgenic mouse model in which human cholesterol 24-hydroxylase, the enzyme responsible for the formation of 24S-OHC, was expressed under the control of a promoter derived from the ß-actin gene. RESULTS: Both male and female transgenic mice had elevated levels of cerebral, plasma, biliary and faecal 24S-OHC. According to the faecal excretion results, production of 24S-OHC was increased four- to sevenfold. Gene expression profiling revealed that the elevated production of 24S-OHC did not result in the anticipated activation of LXR target genes in the brain or liver. CONCLUSION: In spite of the fact that 24S-OHC is a highly effective agonist of LXRs in vitro, it is not a critical activator of target genes to this nuclear receptor in vivo, either in the brain or in the liver.


Subject(s)
Brain/metabolism , Hydroxycholesterols/metabolism , Liver/metabolism , Orphan Nuclear Receptors/genetics , Animals , Cholesterol 24-Hydroxylase , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/physiology , Humans , Liver X Receptors , Male , Mice , Mice, Transgenic , Polymerase Chain Reaction/methods , Steroid Hydroxylases/genetics
9.
J Intern Med ; 269(3): 333-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21083855

ABSTRACT

OBJECTIVES: Atorvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, the rate-limiting enzyme in cholesterol synthesis, lowers plasma cholesterol and triglyceride (TG) levels dose dependently. The aim of this study was to investigate the molecular mechanism(s) of this decrease in plasma TG levels in atorvastatin-treated subjects. RESEARCH DESIGN AND METHODS: Lipoprotein separation and plasma analysis of lipids, glucose and insulin were performed in subjects randomly assigned to placebo (n = 9) or atorvastatin (80 mg per day) (n = 10) for 4 weeks. Liver TG mass was determined in pooled samples. Hepatic expression of several genes involved in carbohydrate and TG metabolism was determined. RESULTS: Atorvastatin lowered plasma levels of very low-density lipoprotein (VLDL) TG (∼50%, P < 0.05) and liver TG mass compared to placebo. Except for cholesterol changes, there were no other significant differences in plasma lipids, glucose or insulin. However, atorvastatin reduced mRNA expression of sterol regulatory element-binding protein 1c (SREBP1c) (>30%, P < 0.05), glucokinase (∼50%, P < 0.05) and angiopoietin-like protein 3 (ANGPTL3) (∼25%, P < 0.01), and induced mRNA expression of acetyl-coenzyme A carboxylase 1 (∼45%, P < 0.05) and glucose-6-phosphatase (∼90%, P < 0.05) compared to placebo. CONCLUSIONS: Following treatment with atorvastatin, reduced ANGPTL3 mRNA expression may contribute to the reduced plasma levels of VLDL TG. The reduced liver TG mass induced by a high dosage of atorvastatin may be important for the treatment of patients with fatty liver.


Subject(s)
Carbohydrates/blood , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/drug effects , Pyrroles/pharmacology , Triglycerides/blood , Adult , Aged , Atorvastatin , Blood Glucose/metabolism , C-Peptide/blood , Drug Administration Schedule , Female , Gene Expression Regulation/drug effects , Humans , Insulin/blood , Lipids/blood , Liver/metabolism , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction/methods
10.
Arterioscler Thromb Vasc Biol ; 29(8): 1235-41, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19478207

ABSTRACT

OBJECTIVE: ACAT2 is thought to be responsible for cholesteryl ester production in chylomicron and VLDL assembly. Recently, we identified HNF1alpha as an important regulator of the human ACAT2 promoter. Thus, we hypothesized that MODY3 (HNF1alpha gene mutations) and possibly MODY1 (HNF4alpha, upstream regulator of HNF1alpha, gene mutations) subjects may have lower VLDL esterified cholesterol. METHODS AND RESULTS: Serum analysis and lipoprotein separation using size-exclusion chromatography were performed in controls and MODY1 and MODY3 subjects. In vitro analyses included mutagenesis and cotransfections in HuH7 cells. Finally, the relevance in vivo of these findings was tested by ChIP assays in human liver. Whereas patients with MODY3 had normal lipoprotein composition, those with MODY1 had lower levels of VLDL and LDL esterified cholesterol, as well as of VLDL triglyceride. Mutagenesis revealed one important HNF4 binding site in the human ACAT2 promoter. ChIP assays and protein-to-protein interaction studies showed that HNF4alpha, directly or indirectly (via HNF1alpha), can bind to the ACAT2 promoter. CONCLUSIONS: We identified HNF4alpha as an important regulator of the hepatocyte-specific expression of the human ACAT2 promoter. Our results suggest that the lower levels of esterified cholesterol in VLDL- and LDL-particles in patients with MODY1 may-at least in part-be attributable to lower ACAT2 activity in these patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocyte Nuclear Factor 4/biosynthesis , Liver Neoplasms/metabolism , Liver/metabolism , Sterol O-Acyltransferase/biosynthesis , Adult , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Female , Humans , Immunoblotting , Liver/pathology , Liver Neoplasms/pathology , Male , Middle Aged , Sterol O-Acyltransferase 2
11.
Biochem Biophys Res Commun ; 357(3): 707-11, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17448444

ABSTRACT

Modulation of bile acid synthesis in human by cholestyramine or by chenodeoxycholic acid (CDCA) treatment affects lipoprotein metabolism leading to altered plasma lipid levels. The molecular changes caused by these treatments, which in turn influence lipoprotein metabolism, are still not entirely known in humans. In this study, mRNA levels were analyzed using real time RT-PCR in liver tissue from patients undergoing cholecystectomy due to gallstone disease. The patients were treated with either CDCA (n=6) or cholestyramine (n=5) for three weeks prior to surgery, six patients received no treatment and served as controls. Cholestyramine increased the expression of the LDL receptor (LDLR) by about 65% and that of proprotein convertase subtilisin kexin 9 (PCSK9) by 70%. After CDCA the levels of both LDLR and hydroxy-methyl-glutaryl coenzyme A reductase mRNA decreased approximately by 50%. The expression of PCSK9 was not changed. The mRNA levels of PCSK9, LDLR, and HMGCoAR were significantly correlated to those of sterol regulatory element binding protein 2 (SREBP2), indicating that SREBP2 is of importance in the regulation of the expression of these genes also in human liver.


Subject(s)
Bile Acids and Salts/metabolism , Chenodeoxycholic Acid/pharmacology , Cholestyramine Resin/pharmacology , Lipoproteins/metabolism , Liver/drug effects , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Apolipoprotein A-I/genetics , Chenodeoxycholic Acid/therapeutic use , Cholecystectomy , Cholestyramine Resin/therapeutic use , Female , Gallstones/drug therapy , Gallstones/genetics , Gallstones/metabolism , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Gene Expression Regulation/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Liver/metabolism , Male , Middle Aged , Proprotein Convertase 9 , Proprotein Convertases , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Sterol Regulatory Element Binding Protein 2/genetics
12.
Eur J Clin Invest ; 36(2): 98-104, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436091

ABSTRACT

BACKGROUND: High-quality methods for lipoprotein characterization are warranted in studies on various metabolic diseases. MATERIALS AND METHODS: An automated system for size-exclusion chromatography (SEC) of lipoproteins using commercially available components is described. Cholesterol or triglyceride content in separated lipoproteins from plasma and interstitial fluid (IF) was continuously determined on-line using microlitre sample volumes. RESULTS: The lipoprotein assay showed a good concordance with the classic ultra-centrifugation/precipitation technique using fresh or frozen samples. Determination of lipoproteins in IF obtained from vacuum-induced skin blisters from 18 healthy subjects revealed that very low density lipoprotein (VLDL), low density lipoprotein (LDL) and high density lipoprotein (HDL) cholesterol levels were 18%, 19% and 25%, respectively, of concomitant plasma concentrations. The size-exclusion chromatography (SEC) system also allows for triglyceride determination on-line and it could be shown that the system is advantageous for an accurate determination of triglycerides in conditions when there are high levels of glycerol, e.g. in mice and in patients with hyperglycerolaemia (pseudo-hypertriglyceridaemia). CONCLUSIONS: The described system should be of value in studies where detailed lipoprotein analysis is warranted and particularly when significant sample series with small volumes are available. Our data also suggest that there is a 4-5.5-fold concentration gradient between plasma and IF for the three major plasma lipoproteins.


Subject(s)
Chromatography, Gel/methods , Extracellular Fluid/chemistry , Lipoproteins/analysis , Animals , Cholesterol, HDL/analysis , Cholesterol, HDL/blood , Cholesterol, LDL/analysis , Cholesterol, LDL/blood , Cholesterol, VLDL/analysis , Cholesterol, VLDL/blood , Fasting , Glycerol/blood , Humans , Lipoproteins/blood , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Triglycerides/analysis , Triglycerides/blood
13.
J Lipid Res ; 47(2): 421-30, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16264196

ABSTRACT

As previously reported by us, mice with targeted disruption of the CYP8B1 gene (CYP8B1-/-) fail to produce cholic acid (CA), upregulate their bile acid synthesis, reduce the absorption of dietary cholesterol and, after cholesterol feeding, accumulate less liver cholesterol than wild-type (CYP8B1+/+) mice. In the present study, cholesterol-enriched diet (0.5%) or administration of a synthetic liver X receptor (LXR) agonist strongly upregulated CYP7A1 expression in CYP8B1-/- mice, compared to CYP8B1+/+ mice. Cholesterol-fed CYP8B1-/- mice also showed a significant rise in HDL cholesterol and increased levels of liver ABCA1 mRNA. A combined CA (0.25%)/cholesterol (0.5%) diet enhanced absorption of intestinal cholesterol in both groups of mice, increased their liver cholesterol content, and reduced their expression of CYP7A1 mRNA. The ABCG5/G8 liver mRNA was increased in both groups of mice, but cholesterol crystals were only observed in bile from the CYP8B1+/+ mice. The results demonstrate the cholesterol-sparing effects of CA: enhanced absorption and reduced conversion into bile acids. Farnesoid X receptor (FXR)-mediated suppression of CYP7A1 in mice seems to be a predominant mechanism for regulation of bile acid synthesis under normal conditions and, as confirmed, able to override LXR-mediated mechanisms. Interaction between FXR- and LXR-mediated stimuli might also regulate expression of liver ABCG5/G8.


Subject(s)
Cholesterol/metabolism , Cholic Acid/deficiency , DNA-Binding Proteins/physiology , Homeostasis/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoprotein A-I/genetics , Bile/chemistry , Bile Acids and Salts/analysis , Bile Acids and Salts/metabolism , Cholesterol/biosynthesis , Cholesterol/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/pharmacology , Cholic Acid/pharmacology , DNA-Binding Proteins/agonists , Feces/chemistry , Female , Gene Expression/drug effects , Gene Expression/genetics , Hydroxymethylglutaryl CoA Reductases/genetics , Isoxazoles/pharmacology , Lipids/analysis , Lipoproteins/blood , Lipoproteins/chemistry , Lipoproteins/genetics , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/agonists , Steroid 12-alpha-Hydroxylase/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Transcription Factors/agonists , ATP-Binding Cassette Sub-Family B Member 4
14.
J Steroid Biochem Mol Biol ; 78(1): 83-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11530288

ABSTRACT

The majority of data regarding biological effects of estrogens is based on studies in male rats or ovariectomized (Ovx) female rats. Therefore, in this study, the effects of estradiol treatment on the regulation of the hepatic estrogen receptor and the level of circulating angiotensinogen were examined in the intact female rat. The data were compared with that of the hypophysectomized (Hx) rat. Animals were treated with either low (physiological) or high (pharmacological) doses of estrogen. In intact rats, the hepatic estrogen receptor (ER) level increased with increasing doses of estrogen. This was in contrast to the Hx rats where growth hormone (GH) and dexametasone (Dex) in combination were the sole modulators of the estrogen receptor. The angiotensinogen level increased in normal rats after estrogen administration in a dose dependent manner, regardless of the mode of administration. The pure antiestrogen ICI 182 780 efficiently blocked the increase in circulating angiotensinogen. The conclusion is that in the normal female, estrogens are important modulators of the serum angiotensinogen level.


Subject(s)
Angiotensins/blood , Estrogens/pharmacology , Liver/metabolism , Receptors, Estrogen/biosynthesis , Angiotensinogen/blood , Animals , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Female , Fulvestrant , Glucocorticoids/pharmacology , Growth Hormone/pharmacology , Immunoenzyme Techniques , In Situ Hybridization , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism
15.
J Clin Invest ; 107(5): 565-73, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238557

ABSTRACT

The nuclear oxysterol-receptor paralogues LXRalpha and LXRbeta share a high degree of amino acid identity and bind endogenous oxysterol ligands with similar affinities. While LXRalpha has been established as an important regulator of cholesterol catabolism in cholesterol-fed mice, little is known about the function of LXRbeta in vivo. We have generated mouse lines with targeted disruptions of each of these LXR receptors and have compared their responses to dietary cholesterol. Serum and hepatic cholesterol levels and lipoprotein profiles of cholesterol-fed animals revealed no significant differences between LXRbeta(-/-) and wild-type mice. Steady-state mRNA levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase, farnesyl diphosphate synthase, and squalene synthase were increased in LXRbeta(-/-) mice compared with LXRbeta(+/+) mice, when fed standard chow. The mRNA levels for cholesterol 7alpha-hydroxylase, oxysterol 7alpha-hydroxylase, sterol 12alpha-hydroxylase, and sterol 27-hydroxylase, respectively, were comparable in these strains, both on standard and 2% cholesterol chow. Our results indicate that LXRbeta(-/-) mice - in contrast to LXRalpha(-/-) mice - maintain their resistance to dietary cholesterol, despite subtle effects on the expression of genes coding for enzymes involved in lipid metabolism. Thus, our data indicate that LXRbeta has no complete overlapping function compared with LXRalpha in the liver.


Subject(s)
Cholesterol/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Alanine Transaminase/blood , Animals , Bile Acids and Salts/metabolism , Cholesterol, Dietary/metabolism , DNA-Binding Proteins , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Female , Gene Expression Regulation, Enzymologic , Hydroxycholesterols/blood , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipid Metabolism , Lipoproteins/blood , Liver/anatomy & histology , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics , Transcription, Genetic
16.
Biochem Biophys Res Commun ; 278(3): 640-5, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11095962

ABSTRACT

Clinical case reports have documented disturbances of carbohydrate and lipid metabolism in aromatase deficient and estrogen resistant males. The aim of the present study was to explore the metabolic functions of estrogens in male mice and to dissect the estrogen receptor (ER) specificity of such effects. Total body fat content and serum levels of leptin were followed in ERalpha knockout (ERKO), ERbeta knockout (BERKO), and ERalpha/beta double knockout (DERKO) mice. Neither the total body fat nor serum leptin levels were altered in any group before or during sexual maturation. However, after sexual maturation ERKO and DERKO, but not BERKO, demonstrated a clear increase in total body fat and enhanced serum leptin levels. Serum cholesterol was increased and a qualitative change in the lipoprotein profile, including smaller LDL particles, was observed in ERKO and DERKO mice. In conclusion, ERalpha but not ERbeta-inactivated male mice develop obesity after sexual maturation.


Subject(s)
Leptin/blood , Lipoproteins/blood , Obesity/genetics , Obesity/physiopathology , Receptors, Estrogen/physiology , Adipose Tissue/anatomy & histology , Adipose Tissue/physiopathology , Animals , Cholesterol/blood , Crosses, Genetic , Estrogen Receptor alpha , Estrogen Receptor beta , Fatty Acids, Nonesterified/blood , Female , Heterozygote , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Sexual Maturation , Triglycerides/blood
17.
Arterioscler Thromb Vasc Biol ; 20(7): 1817-23, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10894823

ABSTRACT

The protective influence of estrogens in cardiovascular disease is believed to be partly due to beneficial effects on cholesterol metabolism. Much of the experimental data are based on models in which synthetic estrogens have been used in pharmacological doses, and therefore, the physiological role of estrogens in cholesterol metabolism is uncertain. To evaluate this important issue, we performed experiments in intact female rats with use of the natural estrogen 17beta-estradiol (E2) administered either subcutaneously or orally. After physiological doses of E2 (< or =0.04 mg. kg(-1). d(-1)) were administered, plasma levels of high density lipoprotein (HDL) cholesterol and apolipoprotein (apo) A-I were increased. In the liver, 3-hydroxy-3-methylglutaryl coenzyme A reductase and cholesterol 7alpha-hydroxylase activities were increased, as well as cholesterol 7alpha-hydroxylase mRNA levels. These effects were abolished during treatment with higher doses of E2, whereas apo A-I mRNA increased in a dose-dependent way. After treatment with pharmacological doses of E2 (> or =0.2 mg. kg(-1). d(-1)), the number of hepatic low density lipoprotein receptors increased and plasma cholesterol was reduced. These effects were similar after both oral and subcutaneous administration of E2. Our results show that the responses to E2 are biphasic: plasma HDL, apo A-I, and hepatic enzyme activities governing bile acid and cholesterol synthesis increased only at physiological doses of E2. At pharmacological doses of E2, hepatic low density lipoprotein receptors are stimulated and plasma cholesterol is reduced. Therefore, under physiological conditions, E2 exerts its major effects on hepatic cholesterol metabolism through mechanisms other than stimulation of low density lipoprotein receptor expression.


Subject(s)
Cholesterol/metabolism , Estradiol/pharmacology , Liver/drug effects , Liver/enzymology , Administration, Oral , Animals , Apolipoprotein A-I/metabolism , Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Enzymologic/drug effects , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Injections, Subcutaneous , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Receptors, LDL/metabolism
19.
Arterioscler Thromb Vasc Biol ; 19(4): 832-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195906

ABSTRACT

Plasma cholesterol levels increase with age, as does the incidence of coronary heart disease. The mechanisms responsible for the age-related hypercholesterolemia are not well understood. An interesting hypothesis suggests that the relative deficiency in growth hormone (GH), which occurs with aging, contributes to the development of the age-related hypercholesterolemia, because GH has beneficial effects on cholesterol metabolism. In the present work, we tested this hypothesis by the administration of GH to normal rats of varying ages. Plasma lipids and hepatic cholesterol metabolism were characterized in 2-, 12-, and 18-month-old male Sprague-Dawley rats. In 2-month-old rats, GH specifically stimulated the hepatic low density lipoprotein (LDL) receptor expression in a dose-dependent way, both at the protein level and at the mRNA level. Concomitantly, plasma cholesterol increased by approximately 30% within the large high density lipoprotein and LDL fractions. In 12-month-old animals, cholesterol 7alpha-hydroxylase (C7alphaOH) activity was reduced, whereas hepatic LDL receptors and plasma total cholesterol were unchanged. GH treatment (1 mg. kg-1. d-1) normalized the activity of C7alphaOH and had effects on plasma cholesterol and LDL receptors similar to those seen in 2-month-old animals. In 18-month-old rats, plasma cholesterol was increased 2-fold, whereas hepatic LDL receptor expression and C7alphaOH activity were similar to those of the 12-month-old animals. Infusion of GH to 18-month-old rats had similar effects on hepatic C7alphaOH and LDL receptors as seen in 12-month-old rats. However, GH treatment strongly reduced the hypercholesterolemia in 18-month-old animals. We conclude that the age-dependent increase of plasma cholesterol in rats can be reversed by the administration of GH, presumably through the pleiotropic effects of this hormone on lipoprotein metabolism.


Subject(s)
Aging/metabolism , Cholesterol/metabolism , Human Growth Hormone/therapeutic use , Hypercholesterolemia/drug therapy , Lipoproteins/metabolism , Animals , Cholesterol/blood , Infusion Pumps, Implantable , Lipoproteins/blood , Male , Rats , Rats, Sprague-Dawley , Receptors, LDL/biosynthesis , Receptors, LDL/genetics
20.
Am J Pathol ; 153(6): 1923-35, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9846982

ABSTRACT

The high-affinity degradation of low-density lipoprotein (LDL) is enhanced 3- to 100-fold in leukemic blood cells from patients with acute myelogenous leukemia (AML), suggesting an increased cellular LDL receptor expression. There are, however, inconsistencies regarding the published properties of LDL receptor regulation in AML cells, and previous data on this are indirect. In the present study the aim was to determine whether the LDL receptor number is increased in AML cells. The LDL receptor number was assayed by ligand blot with rabbit 125I-labeled beta-very-low-density lipoprotein (beta-VLDL) of transferred, SDS-polyacrylamide-gel-electrophoresis-separated AML cell membranes. Samples from 10 patients, six with AML, one with chronic myelogenous leukemia in blast crisis, and three with acute lymphoblastic leukemia, were investigated. The LDL receptor expression was strongly suppressed in all samples to levels lower than that of normal mononuclear cells. This was despite the fact that cells from one patient with AML of M4 subtype had a 50- to 100-fold higher 125I labeled LDL degradation compared with normal cells. Immunoblots with antibodies against gp330/megalin and the LDL-receptor-related protein (LRP) and ligand blot using 125I-labeled 39-kd receptor-associated protein (RAP) could not detect gp330/megalin or VLDL receptors. The LRP was abundant in AML samples of M4 and M5b subtype, as determined from both RAP ligand blot and immunoblot using an LRP-specific antibody. It is concluded that LDL receptors are suppressed in AML cells. It is possible that the high degradation of 125I-labeled LDL present in type M4 and M5 AML cells may involve another lipoprotein receptor.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukocytes, Mononuclear/metabolism , Receptors, LDL/metabolism , Adolescent , Adrenal Glands/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cattle , Female , Heymann Nephritis Antigenic Complex , Humans , Immunoblotting , Kidney/metabolism , Male , Membrane Glycoproteins/metabolism , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...