Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Publication year range
1.
J Lipid Res ; 63(7): 100232, 2022 07.
Article in English | MEDLINE | ID: mdl-35598637

ABSTRACT

Mutations in the LCAT gene cause familial LCAT deficiency (Online Mendelian Inheritance in Man ID: #245900), a very rare metabolic disorder. LCAT is the only enzyme able to esterify cholesterol in plasma, whereas sterol O-acyltransferases 1 and 2 are the enzymes esterifying cellular cholesterol in cells. Despite the complete lack of LCAT activity, patients with familial LCAT deficiency exhibit circulating cholesteryl esters (CEs) in apoB-containing lipoproteins. To analyze the origin of these CEs, we investigated 24 carriers of LCAT deficiency in this observational study. We found that CE plasma levels were significantly reduced and highly variable among carriers of two mutant LCAT alleles (22.5 [4.0-37.8] mg/dl) and slightly reduced in heterozygotes (218 [153-234] mg/dl). FA distribution in CE (CEFA) was evaluated in whole plasma and VLDL in a subgroup of the enrolled subjects. We found enrichment of C16:0, C18:0, and C18:1 species and a depletion in C18:2 and C20:4 species in the plasma of carriers of two mutant LCAT alleles. No changes were observed in heterozygotes. Furthermore, plasma triglyceride-FA distribution was remarkably similar between carriers of LCAT deficiency and controls. CEFA distribution in VLDL essentially recapitulated that of plasma, being mainly enriched in C16:0 and C18:1, while depleted in C18:2 and C20:4. Finally, after fat loading, chylomicrons of carriers of two mutant LCAT alleles showed CEs containing mainly saturated FAs. This study of CEFA composition in a large cohort of carriers of LCAT deficiency shows that in the absence of LCAT-derived CEs, CEs present in apoB-containing lipoproteins are derived from hepatic and intestinal sterol O-acyltransferase 2.


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/metabolism , Apolipoproteins B , Cholesterol/metabolism , Cholesterol Esters , Humans , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lipoproteins , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase 2
2.
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
3.
Rev Endocr Metab Disord ; 23(5): 919-930, 2022 10.
Article in English | MEDLINE | ID: mdl-35687272

ABSTRACT

The growth hormone/insulin growth factor-1 axis is a key endocrine system that exerts profound effects on metabolism by its actions on different peripheral tissues but also in the brain. Growth hormone together with insulin growth factor-1 perform metabolic adjustments, including regulation of food intake, energy expenditure, and glycemia. The dysregulation of this hepatic axis leads to different metabolic disorders including obesity, type 2 diabetes or liver disease. In this review, we discuss how the growth hormone/insulin growth factor-1 axis regulates metabolism and its interactions with the central nervous system. Finally, we state our vision for possible therapeutic uses of compounds based in the components of this hepatic axis.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin , Central Nervous System/metabolism , Growth Hormone/metabolism , Humans , Insulin-Like Growth Factor I/metabolism
4.
J Lipid Res ; 62: 100065, 2021.
Article in English | MEDLINE | ID: mdl-33713671

ABSTRACT

Plasma cholesterol and triglyceride (TG) levels are twice as high in hibernating brown bears (Ursus arctos) than healthy humans. Yet, bears display no signs of early stage atherosclerosis development when adult. To explore this apparent paradox, we analyzed plasma lipoproteins from the same 10 bears in winter (hibernation) and summer using size exclusion chromatography, ultracentrifugation, and electrophoresis. LDL binding to arterial proteoglycans (PGs) and plasma cholesterol efflux capacity (CEC) were also evaluated. The data collected and analyzed from bears were also compared with those from healthy humans. In bears, the cholesterol ester, unesterified cholesterol, TG, and phospholipid contents of VLDL and LDL were higher in winter than in summer. The percentage lipid composition of LDL differed between bears and humans but did not change seasonally in bears. Bear LDL was larger, richer in TGs, showed prebeta electrophoretic mobility, and had 5-10 times lower binding to arterial PGs than human LDL. Finally, plasma CEC was higher in bears than in humans, especially the HDL fraction when mediated by ABCA1. These results suggest that in brown bears the absence of early atherogenesis is likely associated with a lower affinity of LDL for arterial PGs and an elevated CEC of bear plasma.


Subject(s)
Hibernation , Lipoproteins , Ursidae , Animals , Cholesterol/blood , Lipoproteins/blood , Seasons , Ursidae/physiology
5.
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
6.
Vasa ; 50(6): 401-411, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34743585

ABSTRACT

Patients with peripheral arterial disease (PAD) are at very high risk of cardiovascular events, but risk factor management is usually suboptimal. This Joint Task Force from the European Atherosclerosis Society and the European Society of Vascular Medicine has updated evidence on the management on dyslipidaemia and thrombotic factors in patients with PAD. Guidelines recommend a low-density lipoprotein cholesterol (LDLC) goal of more than 50% reduction from baseline and <1.4 mmol/L (<55 mg/dL) in PAD patients. As demonstrated by randomized controlled trials, lowering LDL-C not only reduces cardiovascular events but also major adverse limb events (MALE), including amputations, of the order of 25%. Addition of ezetimibe or a PCSK9 inhibitor further decreases the risk of cardiovascular events, and PCSK9 inhibition has also been associated with reduction in the risk of MALE by up to 40%. Furthermore, statin- based treatment improved walking performance, including maximum walking distance, and pain-free walking distance and duration. This Task Force recommends strategies for managing statin-associated muscle symptoms to ensure that PAD patients benefit from lipid-lowering therapy. Antiplatelet therapy, either daily clopidogrel 75 mg or the combination of aspirin 100 mg and rivaroxaban (2×2.5 mg) is also indicated to prevent cardiovascular events. Dual pathway inhibition (aspirin and rivaroxaban) may be considered following revascularization, taking into account bleeding risk. This Joint Task Force believes that adherence with these recommendations for lipid-lowering and antithrombotic therapy will improve the morbidity and mortality in patients with PAD.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Peripheral Arterial Disease , Cholesterol, LDL , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/drug therapy , Proprotein Convertase 9 , Treatment Outcome
7.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34576136

ABSTRACT

Men with nonalcoholic fatty liver disease (NAFLD) are more exposed to nonalcoholic steatohepatitis (NASH) and liver fibrosis than women. However, the underlying molecular mechanisms of NALFD sex dimorphism are unclear. We combined gene expression, histological and lipidomic analyses to systematically compare male and female liver steatosis. We characterized hepatosteatosis in three independent mouse models of NAFLD, ob/ob and lipodystrophic fat-specific (PpargFΔ/Δ) and whole-body PPARγ-null (PpargΔ/Δ) mice. We identified a clear sex dimorphism occurring only in PpargΔ/Δ mice, with females showing macro- and microvesicular hepatosteatosis throughout their entire life, while males had fewer lipid droplets starting from 20 weeks. This sex dimorphism in hepatosteatosis was lost in gonadectomized PpargΔ/Δ mice. Lipidomics revealed hepatic accumulation of short and highly saturated TGs in females, while TGs were enriched in long and unsaturated hydrocarbon chains in males. Strikingly, sex-biased genes were particularly perturbed in both sexes, affecting lipid metabolism, drug metabolism, inflammatory and cellular stress response pathways. Most importantly, we found that the expression of key sex-biased genes was severely affected in all the NAFLD models we tested. Thus, hepatosteatosis strongly affects hepatic sex-biased gene expression. With NAFLD increasing in prevalence, this emphasizes the urgent need to specifically address the consequences of this deregulation in humans.


Subject(s)
Non-alcoholic Fatty Liver Disease/pathology , PPAR gamma/deficiency , Sex Characteristics , Animals , Disease Models, Animal , Fatty Acids/metabolism , Female , Gene Expression Regulation , Gonadal Steroid Hormones/metabolism , Inflammation/pathology , Lipid Droplets/metabolism , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , PPAR gamma/metabolism , Phenotype , Signal Transduction , Triglycerides/metabolism
8.
Arterioscler Thromb Vasc Biol ; 38(8): 1723-1737, 2018 08.
Article in English | MEDLINE | ID: mdl-29930001

ABSTRACT

Objective- Recent cohort studies have shown that nonalcoholic fatty liver disease (NAFLD), and especially nonalcoholic steatohepatitis (NASH), associate with atherosclerosis and cardiovascular disease, independently of conventional cardiometabolic risk factors. However, the mechanisms underlying the pathophysiological link between NAFLD/NASH and cardiovascular disease still remain unclear. Our previous studies have identified STK25 (serine/threonine protein kinase 25) as a critical determinant in ectopic lipid storage, meta-inflammation, and progression of NAFLD/NASH. The aim of this study was to assess whether STK25 is also one of the mediators in the complex molecular network controlling the cardiovascular disease risk. Approach and Results- Atherosclerosis was induced in Stk25 knockout and transgenic mice, and their wild-type littermates, by gene transfer of gain-of-function mutant of PCSK9 (proprotein convertase subtilisin/kexin type 9), which induces the downregulation of hepatic LDLR (low-density lipoprotein receptor), combined with an atherogenic western-type diet. We found that Stk25-/- mice displayed reduced atherosclerosis lesion area as well as decreased lipid accumulation, macrophage infiltration, collagen formation, and oxidative stress in aortic lesions compared with wild-type littermates, independently from alterations in dyslipidemia. Reciprocally, Stk25 transgenic mice presented aggravated plaque formation and maturation compared with wild-type littermates despite similar levels of fasting plasma cholesterol. We also found that STK25 protein was expressed in all layers of the aorta, suggesting a possible direct role in cardiovascular disease. Conclusions- This study provides the first evidence that STK25 plays a critical role in regulation of cardiovascular disease risk and suggests that pharmacological inhibition of STK25 function may provide new possibilities for prevention/treatment of atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Hypercholesterolemia/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Plaque, Atherosclerotic , Protein Serine-Threonine Kinases/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Diet, High-Fat , Disease Models, Animal , Disease Progression , Gene Transfer Techniques , Hypercholesterolemia/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/enzymology , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction
9.
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
10.
J Lipid Res ; 58(2): 420-432, 2017 02.
Article in English | MEDLINE | ID: mdl-27941027

ABSTRACT

Hepatitis C virus (HCV) is an enveloped RNA virus responsible for 170 million cases of viral hepatitis worldwide. Over 50% of chronically infected HCV patients develop hepatic steatosis, and steatosis can be induced by expression of HCV core protein (core) alone. Additionally, core must associate with cytoplasmic lipid droplets (LDs) for steatosis development and viral particle assembly. Due to the importance of the LD as a key component of hepatic lipid storage and as a platform for HCV particle assembly, it seems this dynamic subcellular organelle is a gatekeeper in the pathogenesis of viral hepatitis. Here, we hypothesized that core requires the host LD scaffold protein, perilipin (PLIN)3, to induce hepatic steatosis. To test our hypothesis in vivo, we have studied core-induced hepatic steatosis in the absence or presence of antisense oligonucleotide-mediated knockdown of PLIN3. PLIN3 knockdown blunted HCV core-induced steatosis in transgenic mice fed either chow or a moderate fat diet. Collectively, our studies demonstrate that the LD scaffold protein, PLIN3, is essential for HCV core-induced hepatic steatosis and provide new insights into the pathogenesis of HCV.


Subject(s)
Fatty Liver/genetics , Hepatitis C/metabolism , Liver/metabolism , Perilipin-3/genetics , Animals , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Knockdown Techniques , Genotype , Hepacivirus , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis C/virology , Humans , Lipid Droplets/metabolism , Lipid Droplets/pathology , Lipid Droplets/virology , Lipid Metabolism/genetics , Liver/pathology , Liver/virology , Mice , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Perilipin-3/antagonists & inhibitors
11.
Biochim Biophys Acta ; 1861(1): 51-59, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26515253

ABSTRACT

Primary human hepatocytes are considered to be the "gold standard" in studies of lipid metabolism despite a number of disadvantages like large inter-donor differences and inability to proliferate. Human hepatoma HepG2 cells retain many hepatocyte-specific functions but do also exhibit disadvantages like secretion of lipoproteins and bile acids that do not emulate human hepatocytes in vivo. The aim of this study was to investigate whether supplementation of the culturing media with human serum could improve the functionality of HepG2 cells and thereby make them more apposite in studies of lipid metabolism. The cells were cultured with human sera (2%) from three healthy individuals or with fetal bovine serum (10%). Lipoprotein, apolipoprotein, bile acid, albumin, and proprotein subtilisin/kexin type 9 (Pcsk9) concentrations in the cell media, as well as gene and protein expressions were then measured. We found apoB-containing LDL-sized but also apoA1-containing HDL-sized particles, increased bile acid and Pcsk9 concentrations in the cell media, as well as increased expression of genes involved in lipid metabolism and differentiation in HepG2 cells cultured with human sera. Thus, supplementation of the culturing media with human serum improves the functionality of HepG2 cells and makes them more apposite in studies of lipid metabolism.


Subject(s)
Hepatocytes/metabolism , Lipid Metabolism , Culture Media , Hep G2 Cells , Humans , Lipoproteins, LDL/metabolism , Serum
12.
Eur J Clin Invest ; 47(2): 137-148, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28036114

ABSTRACT

BACKGROUND: In end-stage renal disease (ESRD), coronary artery calcification (CAC) and inflammation contribute to cardiovascular disease (CVD). Statins do not improve survival in patients with ESRD, and their effect on vascular calcification is unclear. We explored associations between CAC, inflammatory biomarkers, statins and mortality in ESRD. MATERIALS AND METHODS: In 240 patients with ESRD (63% males; median age 56 years) from cohorts including 86 recipients of living donor kidney transplant (LD-Rtx), 96 incident dialysis patients and 58 prevalent peritoneal dialysis patients, associations of CAC score (Agatston Units, AUs), interleukin-6 (IL-6) with high-sensitivity C-reactive protein (hsCRP), tumour necrosis factor (TNF), use of statins and all-cause mortality were analysed. Cardiac CT was repeated in 35 patients after 1·5 years of renal replacement therapy. In vitro, human vascular smooth muscle cells (hVSMCs) were used to measure vitamin K metabolism. RESULTS: Among 240 patients, 129 (53%) had a CAC score > 100 AUs. Multivariate analysis revealed that independent predictors of 1-SD higher CAC score were age, male gender, diabetes and use of statins. The association between CAC score and mortality remained significant after adjustment for age, gender, diabetes, CVD, use of statins, protein-energy wasting and inflammation. Repeated CAC imaging in 35 patients showed that statin therapy was associated with greater progression of CAC. In vitro synthesis of menaquinone-4 by hVSMCs was significantly impaired by statins. CONCLUSION: Elevated CAC score is a mortality risk factor in ESRD independent of inflammation. Future studies should resolve if statins promote vascular calcification and inhibition of vitamin K synthesis in the uremic milieu.


Subject(s)
Coronary Artery Disease/chemically induced , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Kidney Failure, Chronic/complications , Vascular Calcification/chemically induced , Adult , Aged , Biomarkers/metabolism , Coronary Artery Disease/mortality , Female , Humans , Kaplan-Meier Estimate , Kidney Failure, Chronic/mortality , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Vascular Calcification/mortality , Vitamin K/metabolism
13.
Arterioscler Thromb Vasc Biol ; 36(5): 787-91, 2016 05.
Article in English | MEDLINE | ID: mdl-27034474

ABSTRACT

OBJECTIVE: Patients with type 2 diabetes mellitus (T2D) have an increased risk of cardiovascular disease, the mechanism of which is incompletely understood. Their high-density lipoprotein (HDL) particles in plasma have been reported to have impaired cholesterol efflux capacity. However, the efflux capacity of HDL from interstitial fluid (IF), the starting point for reverse cholesterol transport, has not been studied. We here investigated the cholesterol efflux capacity of HDL from IF and plasma from T2D patients and healthy controls. APPROACH AND RESULTS: HDL was isolated from IF and peripheral plasma from 35 T2D patients and 35 age- and sex-matched healthy controls. Cholesterol efflux to HDL was determined in vitro, normalized for HDL cholesterol, using cholesterol-loaded macrophages. Efflux capacity of plasma HDL was 10% lower in T2D patients than in healthy controls, in line with previous observations. This difference was much more pronounced for HDL from IF, where efflux capacity was reduced by 28% in T2D. Somewhat surprisingly, the efflux capacity of HDL from IF was lower than that of plasma HDL, by 15% and 32% in controls and T2D patients, respectively. CONCLUSION: These data demonstrate that (1) HDL from IF has a lower cholesterol efflux capacity than plasma HDL and (2) the efflux capacity of HDL from IF is severely impaired in T2D when compared with controls. Because IF comprises the compartment where reverse cholesterol transport is initiated, the marked reduction in cholesterol efflux capacity of IF-HDL from T2D patients may play an important role for their increased risk to develop atherosclerosis.


Subject(s)
Cholesterol, HDL/metabolism , Diabetes Mellitus, Type 2/metabolism , Extracellular Fluid/metabolism , Biological Transport , Case-Control Studies , Cholesterol, HDL/blood , Cholesterol, HDL/isolation & purification , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Humans , Time Factors
14.
Biochim Biophys Acta ; 1851(1): 9-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25088698

ABSTRACT

TG interacting factors (Tgifs) 1 and 2 are members of the TALE (three-amino-acid loop extension) superfamily of homeodomain proteins. These two proteins bind to the same DNA sequence and share a conserved C-terminal repression domain. Mutations in TGIF1 have been linked to holoprosencephaly, which is a human genetic disease that affects craniofacial development. As these proteins can interact with the ligand binding domain of retinoid X receptor α, a common heterodimeric partner of several nuclear receptors [e.g., liver X receptors (LXRs) and peroxisome proliferator-activated receptors (PPARs)], Tgif1 and Tgif2 might repress other transcriptional pathways activated by lipids. In line with this, Tgif1 interacts with LXRα and Tgif1 null mice have increased expression of the two Lxrα target genes apolipoproteins (Apo) c2 and a4. Also, we have recently identified Tgif1 to function as a transcriptional repressor of the cholesterol esterifying enzyme acyl-coenzyme A:cholesterol acyltransferase 2 (gene name SOAT2). As no studies yet have shown involvement of Tgif2 in the lipid metabolism, this review will focus on the role of Tgif1 in lipid and cholesterol metabolism. This article is part of a Special Issue entitled: Linking transcription to physiology in lipodomics.


Subject(s)
Homeodomain Proteins/metabolism , Lipid Metabolism/physiology , Repressor Proteins/metabolism , Animals , Cholesterol/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Humans , Repressor Proteins/genetics , Transcription, Genetic
15.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G952-61, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27056725

ABSTRACT

It is well known that reduced glomerular filtration rate (GFR) leads to an increased risk of dyslipidemia, insulin resistance, and cardiovascular mortality. The liver is a central organ for metabolism, but its function in the uremic setting is still poorly characterized. We used human primary hepatocytes isolated from livers of nine donors with normal renal function to investigate perturbations in key metabolic pathways following exposure to uremic (n = 8) or healthy (n = 8) sera, and to serum-free control medium. Both uremic and healthy elicited consistent responses from hepatocytes from multiple donors and compared with serum-free control. However, at physiological insulin concentrations, uremic cells accumulated 56% more intracellular lipids. Also, when comparing uremic with healthy medium after culture, it contained more very-low-density lipoprotein-triglyceride and glucose. These changes were accompanied by decreased phosphorylation of AktS473 mRNA levels of key regulators of gluconeogenesis in uremic sera-treated hepatocytes such as phosphoenolpyruvate carboxykinase 1 and glucose 6-phosphate were elevated. We also found increased expression of 11ß-hydroxysteroid dehydrogenase mRNA in uremic cells, along with high phosphorylation of downstream p53 and phospholipase C-γ1Y783 Thus our ex vivo data suggest that the uremic hepatocytes rapidly develop a glycogenic and lipogenic condition accompanied by perturbations in a large number of signaling networks.


Subject(s)
Gluconeogenesis , Hepatocytes/metabolism , Lipid Metabolism , Uremia/metabolism , Aged , Animals , Case-Control Studies , Cells, Cultured , Female , Glucose-6-Phosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Rats , Triglycerides/metabolism , Uremia/blood
16.
Hepatology ; 61(1): 161-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24954587

ABSTRACT

UNLABELLED: Farnesoid X receptor (FXR) is the master regulator of bile acid (BA) homeostasis because it controls BA synthesis, influx, efflux, and detoxification in the gut/liver axis. Deregulation of BA homeostasis has been linked to hepatocellular carcinoma (HCC), and spontaneous hepatocarcinogenesis has been observed in FXR-null mice. This dreaded liver neoplasm has been associated with both FXR gene deletion and BA-mediated metabolic abnormalities after inactivation of FXR transcriptional activity. In the present study, we addressed the hypothesis that intestinal selective FXR reactivation would be sufficient to restore the fibroblast growth factor 15 (FGF15)/cholesterol-7alpha-hydroxylase (Cyp7a1) enterohepatic axis and eventually provide protection against HCC. To this end, we generated FXR-null mice with re-expression of constitutively active FXR in enterocytes (FXR(-/-)iVP16FXR) and corresponding control mice (FXR(-/-)iVP16). In FXR-null mice, intestinal selective FXR reactivation normalized BA enterohepatic circulation along with up-regulation of intestinal FXR transcriptome and reduction of hepatic BA synthesis. At 16 months of age, intestinal FXR reactivation protected FXR-null mice from spontaneous HCC development that occurred in otherwise FXR-null mice. Activation of intestinal FXR conferred hepatoprotection by restoring hepatic homeostasis, limiting cellular proliferation through reduced cyclinD1 expression, decreasing hepatic inflammation and fibrosis (decreased signal transducer and activator of transcription 3 activation and curtailed collagen deposition). CONCLUSION: Intestinal FXR is sufficient to restore BA homeostasis through the FGF15 axis and prevent progression of liver damage to HCC even in the absence of hepatic FXR. Intestinal-selective FXR modulators could stand as potential therapeutic intervention to prevent this devastating hepatic malignancy, even if carrying a somatic FXR mutation.


Subject(s)
Bile Acids and Salts/metabolism , Carcinoma, Hepatocellular/etiology , Intestinal Mucosa/metabolism , Liver Neoplasms/etiology , Receptors, Cytoplasmic and Nuclear/metabolism , Aging/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Female , Fibroblast Growth Factors/metabolism , Genes, cdc , Homeostasis , Male , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Aryl Hydrocarbon/metabolism
17.
J Mol Cell Cardiol ; 82: 186-93, 2015 May.
Article in English | MEDLINE | ID: mdl-25791168

ABSTRACT

Hypercholesterolemia is a medical condition often characterized by high levels of low-density lipoprotein cholesterol (LDL-C) in the blood. Despite the available therapies, not all patients show sufficient responses, especially those with very high levels of LDL-C or those with familial hypercholesterolemia. Regulation of plasma cholesterol levels is very complex and several proteins are involved (both receptors and enzymes). From these, the proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a promising pharmacologic target. The objective of this work is to develop a new approach to inactivate PCSK9 by splice-switching oligonucleotides (SSOs), converting the normal splice form to a natural, less abundant and inactive, splice variant. For this purpose, a new RNA therapeutic approach for hypercholesterolemia based on SSOs was developed for modulation of the splice pattern of human PCSK9 pre-mRNA. Our results show an increase of the selected splice form at both the mRNA and protein level when compared to non-treated Huh7 and HepG2 cell lines, with concomitant increase of the protein level of the low-density lipoprotein receptor (LDLR) demonstrating the specificity and efficiency of the system. In vivo, full conversion to the splice form was achieved in a reporter system when mice were treated with the specific oligonucleotide, thus further indicating the therapeutic potential of the approach. In conclusion, PCSK9 activity can be modulated by splice-switching through an RNA therapeutic approach. The tuning of the natural active to non-active isoforms represents a physiological way of regulating the cholesterol metabolism, by controlling the amount of LDL receptor available and the rate of LDL-cholesterol clearance.


Subject(s)
Gene Silencing , Oligonucleotides/genetics , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , RNA/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Animals , Cell Line , Cell Survival/genetics , Gene Expression , Genes, Reporter , Hepatocytes/metabolism , Humans , Intracellular Space/metabolism , Mice , Proprotein Convertase 9 , Protein Transport , RNA Splicing , Receptors, LDL/metabolism , Transfection
18.
J Lipid Res ; 56(8): 1633-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26092865

ABSTRACT

At a given level of serum cholesterol, patients with T2D have an increased risk of developing atherosclerosis compared with nondiabetic subjects. We hypothesized that T2D patients have an increased interstitial fluid (IF)-to-serum gradient ratio for LDL, due to leakage over the vascular wall. Therefore, lipoprotein profiles in serum and IF from 35 T2D patients and 35 healthy controls were assayed using fast performance liquid chromatography. The IF-to-serum gradients for VLDL and LDL cholesterol, as well as for apoB, were clearly reduced in T2D patients compared with healthy controls. No such differences were observed for HDL cholesterol. Contrary to our hypothesis, the atherogenic VLDL and LDL particles were not increased in IF from diabetic patients. Instead, they were relatively sparser than in healthy controls. The most probable explanation to our unexpected finding is that these lipoproteins are more susceptible to retainment in the extravascular space of these patients, reflecting a more active uptake by, or adhesion to, tissue cells, including macrophages in the vascular wall. Further studies are warranted to further characterize the mechanisms underlying these observations, which may be highly relevant for the understanding of why the propensity to develop atherosclerosis is increased in T2D.


Subject(s)
Atherosclerosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Extracellular Fluid/metabolism , Lipoproteins/metabolism , Atherosclerosis/complications , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Extracellular Fluid/drug effects , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins/blood , Male , Middle Aged
19.
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
20.
J Lipid Res ; 55(4): 709-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478032

ABSTRACT

Acat2 [gene name: sterol O-acyltransferase 2 (SOAT2)] esterifies cholesterol in enterocytes and hepatocytes. This study aims to identify repressor elements in the human SOAT2 promoter and evaluate their in vivo relevance. We identified TG-interacting factor 1 (Tgif1) to function as an important repressor of SOAT2. Tgif1 could also block the induction of the SOAT2 promoter activity by hepatocyte nuclear factor 1α and 4α. Women have ∼ 30% higher hepatic TGIF1 mRNA compared with men. Depletion of Tgif1 in mice increased the hepatic Soat2 expression and resulted in higher hepatic lipid accumulation and plasma cholesterol levels. Tgif1 is a new player in human cholesterol metabolism.


Subject(s)
Gene Silencing , Homeodomain Proteins/physiology , Repressor Proteins/physiology , Sterol O-Acyltransferase/genetics , Animals , Binding Sites , Cell Line, Tumor , Enzyme Repression , Female , Gallstones/enzymology , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 4/physiology , Homeodomain Proteins/metabolism , Humans , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Sex Characteristics , Sterol O-Acyltransferase/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Sterol O-Acyltransferase 2
SELECTION OF CITATIONS
SEARCH DETAIL