Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Int J Obes (Lond) ; 41(4): 664-668, 2017 04.
Article in English | MEDLINE | ID: mdl-28093571

ABSTRACT

Roux-en-Y gastric bypass (RYGB) surgery is widely used in the management of morbid obesity. RYGB improves metabolism independently of weight loss by still unknown mechanisms. Bile acids (BAs) are good candidates to explain this benefit, since they regulate metabolic homeostasis and their systemic concentrations increase upon RYGB. Here we analyzed the mechanisms underlying the increase in systemic BA concentrations after RYGB and the role of the liver therein. To this aim, we used the Göttingen-like minipig, a human-size mammalian model, which allows continuous sampling and simultaneous analysis of pre-hepatic portal and systemic venous blood. BA concentrations and pool composition were measured in portal blood, containing intestinal reabsorbed BAs and compared to systemic blood during a standardized meal test before and after RYGB. Systemic total BA concentrations increased after RYGB, due to an increase in conjugated BAs. Interestingly, the ratio of portal:systemic conjugated BAs decreased after RYGB, indicating a role for the liver in systemic BA concentrations changes. In line, hepatic expression of BA transporter genes decreased after RYGB. Our results show that the increase in systemic BAs after surgery is due to decreased selective hepatic recapture. Thus, alterations in hepatic function contribute to the increase in systemic BAs after RYGB.


Subject(s)
Bile Acids and Salts/metabolism , Gastric Bypass , Liver/metabolism , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Swine, Miniature/metabolism , Animals , Disease Models, Animal , Male , Swine , Weight Loss/physiology
2.
Int J Obes (Lond) ; 40(8): 1260-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27089995

ABSTRACT

BACKGROUND: Roux-en-Y gastric bypass (RYGBP) is the most widely used bariatric surgery procedure, which induces profound metabolic and physiological effects, such as substantial improvements in obesity, type 2 diabetes and their comorbidities. Increasing evidence identifies bile acids (BAs) as signaling molecules that contribute to the metabolic improvement after RYGBP. However, how and to what extent BAs mediate the metabolic effects of RYGBP still remains unclear and requires mechanism of action studies using preclinical models. In this study, we compared plasma BA profiles before and after RYGBP in two animal models, rats and pigs, with humans to evaluate their translational potential. METHODS: Plasma BAs were profiled in rats, pigs and humans by liquid chromatography coupled with tandem mass spectrometry before and after RYGBP. RESULTS: RYGBP increased baseline plasma total BA concentrations in humans and in the two animal models to a similar extent (∼3-fold increase), despite differences in presurgery BA levels and profiles between the models. However, qualitatively, RYGBP differently affected individual plasma BA species, with similar increases in some free species (cholic acid (CA), chenodeoxycholic acid (CDCA) and deoxycholic acid (DCA)), different increases in glyco-conjugated species depending on the model and globally no increase in tauro-conjugated species whatever the model. CONCLUSIONS: The tested animal models share similar quantitative RYGBP-induced increases in peripheral blood BAs as humans, which render them useful for mechanistic studies. However, they also present qualitative differences in BA profiles, which may result in different signaling responses. Such differences need to be taken into account when translating results to humans.


Subject(s)
Bile Acids and Salts/blood , Gastric Bypass , Obesity/blood , Obesity/surgery , Adult , Animals , Blood Glucose/metabolism , Disease Models, Animal , Female , Humans , Male , Middle Aged , Obesity/physiopathology , Rats , Signal Transduction , Swine , Swine, Miniature , Treatment Outcome , Weight Loss
3.
Clin Chim Acta ; 392(1-2): 25-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18355452

ABSTRACT

BACKGROUND: Familial hypercholesterolemia (FH) is an autosomal dominant inherited disease caused by mutations in either the low-density lipoprotein receptor, the apolipoprotein B or the proprotein convertase subtilisin/kexin type 9 genes. It is characterized by a high concentration of low-density lipoprotein (LDL), which frequently gives rise to premature coronary disease. In this study, we report a novel splice site mutation of the LDL receptor gene in a Tunisian family. METHODS: Seven patients from the family were screened for mutations in the LDLR gene and the apoB gene, using direct sequencing. RT-PCR and study on cultured skin fibroblast were realised to characterize the effect of novel mutation. RESULTS: Direct sequencing of the promoter and 18 exons reveals a G>A substitution in the splice site junction of intron 8 (c.1186+1 G>A). Study on cultured skin fibroblasts showed a residual activity of 10% of the LDL receptor. Reverse transcription, amplification and direct sequencing of RNA from patient's lymphocytes reveal a deletion of the final 51 bp of exon 8 preserving the reading frame. CONCLUSIONS: The study identified a novel splice mutation c.1186+1 G>A in the LDL receptor gene. It causes the utilization of a new cryptic donor splice site 51 bp downstream from the normal site.


Subject(s)
Apolipoproteins B/genetics , Fibroblasts/metabolism , Hypercholesterolemia/genetics , Mutation , RNA Splice Sites , Receptors, LDL/genetics , Adult , Aged , Apolipoproteins B/metabolism , Cells, Cultured , Family , Female , Humans , Hypercholesterolemia/blood , Introns , Male , Middle Aged , Receptors, LDL/metabolism , Tunisia
4.
Circ Res ; 97(7): 682-9, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16141411

ABSTRACT

Liver X receptors (LXRs) are nuclear receptors that regulate macrophage cholesterol efflux by inducing ATP-binding cassette transporter A1 (ABCA1) and ABCG1/ABCG4 gene expression. The Niemann-Pick C (NPC) proteins NPC1 and NPC2 are located in the late endosome, where they control cholesterol trafficking to the plasma membrane. The mobilization of cholesterol from intracellular pools to the plasma membrane is a determinant governing its availability for efflux to extracellular acceptors. Here we investigated the influence of LXR activation on intracellular cholesterol trafficking in primary human macrophages. Synthetic LXR activators increase the amount of free cholesterol in the plasma membrane by inducing NPC1 and NPC2 gene expression. Moreover, ABCA1-dependent cholesterol efflux induced by LXR activators was drastically decreased in the presence of progesterone, which blocks postlysosomal cholesterol trafficking, and reduced when NPC1 and NPC2 mRNA expression was depleted using small interfering RNA. The stimulation of cholesterol mobilization to the plasma membrane by LXRs led to a decrease in cholesteryl ester formation and Acyl-coenzyme A cholesterol acyltransferase-1 activity. These data indicate that LXR activation enhances cholesterol trafficking to the plasma membrane, where it becomes available for efflux, at the expense of esterification, thus contributing to the overall effects of LXR agonists in the control of macrophage cholesterol homeostasis.


Subject(s)
Cholesterol Esters/metabolism , Cholesterol/metabolism , DNA-Binding Proteins/physiology , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/physiology , Animals , Biological Transport , Carrier Proteins/physiology , Cell Membrane/metabolism , Cells, Cultured , Cholesterol Esters/analysis , Foam Cells/metabolism , Glycoproteins/physiology , Humans , Intracellular Signaling Peptides and Proteins , Liver X Receptors , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Niemann-Pick C1 Protein , Orphan Nuclear Receptors , Progesterone/pharmacology , RNA, Small Interfering/pharmacology , Vesicular Transport Proteins
5.
J Lipid Res ; 46(12): 2717-25, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16162941

ABSTRACT

The mobilization of cholesterol from intracellular pools to the plasma membrane is a determinant that governs its availability for efflux to extracellular acceptors. NPC1 and NPC2 are proteins localized in the late endosome and control cholesterol transport from the lysosome to the plasma membrane. Here, we report that NPC1 and NPC2 gene expression is induced by oxidized LDL (OxLDL) in human macrophages. Because OxLDLs contain natural activators of peroxisome proliferator-activated receptor alpha (PPARalpha), a fatty acid-activated nuclear receptor, the regulation of NPC1 and NPC2 by PPARalpha and the consequences on cholesterol trafficking were further studied. NPC1 and NPC2 expression is induced by synthetic PPARalpha ligands in human macrophages. Furthermore, PPARalpha activation leads to an enrichment of cholesterol in the plasma membrane. By contrast, incubation with progesterone, which blocks postlysosomal cholesterol trafficking, as well as NPC1 and NPC2 mRNA depletion using small interfering RNA, abolished ABCA1-dependent cholesterol efflux induced by PPARalpha activators. These observations identify a novel regulatory role for PPARalpha in the control of cholesterol availability for efflux that, associated with its ability to inhibit cholesterol esterification and to stimulate ABCA1 and scavenger receptor class B type I expression, may contribute to the stimulation of reverse cholesterol transport.


Subject(s)
Cholesterol/metabolism , Macrophages/metabolism , PPAR alpha/metabolism , Biological Transport , Carrier Proteins/genetics , Cell Differentiation , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Gene Expression Regulation , Glycoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lipoproteins, LDL/pharmacology , Macrophages/cytology , Membrane Glycoproteins/genetics , Niemann-Pick C1 Protein , PPAR alpha/genetics , Progesterone/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Vesicular Transport Proteins
6.
Circ Res ; 92(2): 212-7, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12574149

ABSTRACT

Peroxisome proliferator-activated receptor alpha (PPARalpha) is a nuclear receptor activated by fatty acid derivatives and hypolipidemic drugs of the fibrate class. PPARalpha is expressed in monocytes, macrophages, and foam cells, suggesting a role for this receptor in macrophage lipid homeostasis with consequences for atherosclerosis development. Recently, it was shown that PPARalpha activation promotes cholesterol efflux from macrophages via induction of the ABCA1 pathway. In the present study, the influence of PPARalpha activators on intracellular cholesterol homeostasis was investigated. In human macrophages and foam cells, treatment with fibrates, synthetic PPARalpha activators, led to a decrease in the cholesteryl ester (CE):free cholesterol (FC) ratio. In these cells, PPARalpha activation reduced cholesterol esterification rates and Acyl-CoA:cholesterol acyltransferase-1 (ACAT1) activity. However, PPARalpha activation did not alter ACAT1 gene expression, whereas mRNA levels of carnitine palmitoyltransferase type 1 (CPT-1), a key enzyme in mitochondrial fatty acid catabolism, were induced. Finally, PPARalpha activation blocked CE formation induced by TNF-alpha, possibly due to the inhibition of neutral sphingomyelinase activation by TNF-alpha. In conclusion, our results identify a role for PPARalpha in the control of cholesterol esterification in macrophages, resulting in an enhanced availability of FC for efflux through the ABCA1 pathway.


Subject(s)
Cholesterol/metabolism , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Cholesterol Esters/metabolism , Enzyme Activation/drug effects , Esterification/drug effects , Foam Cells/cytology , Foam Cells/drug effects , Foam Cells/metabolism , Gene Expression/drug effects , Homeostasis/drug effects , Humans , Ligands , Macrophages/cytology , Macrophages/drug effects , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Sphingomyelin Phosphodiesterase/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Transcription Factors/drug effects , Tumor Necrosis Factor-alpha/pharmacology
8.
Atherosclerosis ; 154(3): 557-65, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11257256

ABSTRACT

Familial hypercholesterolemia (FH) has a higher prevalence in central Tunisia together with a milder clinical expression than in western countries. The molecular basis of FH in Tunisia remains unknown. Our aim was to identify FH-causing mutations in three unrelated families (21 subjects) from the area of Souassi (central Tunisia). In probands with a presentation of homozygous FH, the promoter and 18 exons of the low density lipoprotein (LDL)-receptor gene were sequenced in both orientations. A novel complex frameshift mutation was identified in exon 10, nucleotides 1477-1479 (TCT) at Serine 472 were replaced by an insertion of seven nucleotides (AGAGACA), producing a premature termination codon 43 amino acids downstream. Binding of 125I-labelled LDL at 4 degrees C to cultured fibroblasts from two probands showed <2% normal LDL-receptor activity. AvaII digestion of PCR amplified genomic DNA identified this unique mutation in all families; homozygotes n=11, heterozygotes n=10. All mutation carriers shared the same haplotype (7 RFLPs), suggesting that they had a common ancestor. Despite high plasma LDL levels (m=16.0+/-3.0 mmol/l) and extravascular cholesterol deposits, most homozygotes were diagnosed after puberty and had a delayed onset of cardiovascular complications. Moreover, most heterozygotes were free of clinical signs and had plasma LDL cholesterol in the normal range (4.7+/-1.3 mmol/l) without taking any lipid-lowering medication. This mild clinical phenotype which contrasted with the severity of the mutation, could not be explained by specific apolipoprotein E or lipoprotein lipase alleles.


Subject(s)
Exons/genetics , Frameshift Mutation , Hyperlipoproteinemia Type II/genetics , Receptors, LDL/genetics , Adolescent , Adult , Amino Acid Sequence/genetics , Base Sequence/genetics , Child , Cholesterol, LDL/blood , Female , Frameshift Mutation/genetics , Haplotypes , Homozygote , Humans , Male , Middle Aged , Pedigree , Phenotype , Promoter Regions, Genetic/genetics , Tunisia
9.
Nat Med ; 7(1): 53-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135616

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that regulate lipid and glucose metabolism and cellular differentiation. PPAR-alpha and PPAR-gamma are both expressed in human macrophages where they exert anti-inflammatory effects. The activation of PPAR-alpha may promote foam-cell formation by inducing expression of the macrophage scavenger receptor CD36. This prompted us to investigate the influence of different PPAR-activators on cholesterol metabolism and foam-cell formation of human primary and THP-1 macrophages. Here we show that PPAR-alpha and PPAR-gamma activators do not influence acetylated low density lipoprotein-induced foam-cell formation of human macrophages. In contrast, PPAR-alpha and PPAR-gamma activators induce the expression of the gene encoding ABCA1, a transporter that controls apoAI-mediated cholesterol efflux from macrophages. These effects are likely due to enhanced expression of liver-x-receptor alpha, an oxysterol-activated nuclear receptor which induces ABCA1-promoter transcription. Moreover, PPAR-alpha and PPAR-gamma activators increase apoAI-induced cholesterol efflux from normal macrophages. In contrast, PPAR-alpha or PPAR-gamma activation does not influence cholesterol efflux from macrophages isolated from patients with Tangier disease, which is due to a genetic defect in ABCA1. Here we identify a regulatory role for PPAR-alpha and PPAR-gamma in the first steps of the reverse-cholesterol-transport pathway through the activation of ABCA1-mediated cholesterol efflux in human macrophages.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , Foam Cells/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , ATP Binding Cassette Transporter 1 , Base Sequence , Biological Transport , Cells, Cultured , DNA Primers , Humans
10.
Arterioscler Thromb Vasc Biol ; 20(4): 1074-81, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764676

ABSTRACT

Apolipoproteins of high density lipoprotein (HDL) and especially apolipoprotein (apo)AI and apoAII have been demonstrated as binding directly to the class B type I scavenger receptor (SR-BI), the HDL receptor that mediates selective cholesteryl ester uptake. However, the functional relevance of the binding capacity of each apolipoprotein is still unknown. The human adrenal cell line, NCI-H295R, spontaneously expresses a high level of SR-BI, the major apoAI binding protein in these cells. As previously described for murine SR-BI, free apoAI, palmitoyl-oleoyl-phosphatidylcholine (POPC)-AI, and HDL are good ligands for human SR-BI. In vitro displacement of apoAI by apoAII in HDLs or in Lp AI purified from HDL by immunoaffinity enhances their ability to compete with POPC-AI to bind to SR-BI and also enhances their direct binding capacity. The next step was to determine whether the higher affinity of apoAII for SR-BI correlated with the specific uptake of cholesteryl esters from these HDLs. Free apoAII and, to a lesser extent, free apoAI that were added to the cell medium during uptake experiments inhibited the specific uptake of [(3)H]cholesteryl esters from HDL, indicating that binding sites on cells were the same as cholesteryl ester uptake sites. In direct experiments, the uptake of [(3)H]cholesteryl esters from apoAII-enriched HDL was highly reduced compared with the uptake from native HDL. These results demonstrate that in the human adrenal cell line expressing SR-BI as the major HDL binding protein, efficient apoAII binding has an inhibitory effect on the delivery of cholesteryl esters to cells.


Subject(s)
Apolipoprotein A-II/metabolism , Cholesterol Esters/metabolism , Lipoproteins, HDL/metabolism , Membrane Proteins , Receptors, Immunologic/metabolism , Receptors, Lipoprotein , Adrenal Cortex Neoplasms/metabolism , Animals , Apolipoprotein A-I/pharmacology , Apolipoprotein A-II/pharmacology , Binding, Competitive , CD36 Antigens , Humans , Lipoproteins/chemistry , Lipoproteins/metabolism , Lipoproteins, HDL/chemistry , Mice , Phosphatidylcholines/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Tritium , Tumor Cells, Cultured
11.
Arterioscler Thromb Vasc Biol ; 20(3): 763-72, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712402

ABSTRACT

The serum amyloid A (SAA) family of proteins is encoded by multiple genes that display allelic variation and a high degree of homology in mammals. Triggered by inflammation after stimulation of hepatocytes by lymphokine-mediated processes, the concentrations of SAA may increase during the acute-phase reaction to levels 1000-fold greater than those found in the noninflammatory state. In addition to its role as an acute-phase reactant, SAA (104 amino acids, 12 kDa) is considered to be the precursor protein of secondary reactive amyloidosis, in which the N-terminal portion is incorporated into the bulk of amyloid fibrils. However, the association with lipoproteins of the high-density range and subsequent modulation of the metabolic properties of its physiological carrier appear to be the principal role of SAA. Because SAA may displace apolipoprotein A-I, the major protein component of native high density lipoprotein (HDL), during the acute-phase reaction, the present study was aimed at (1) investigating binding properties of native and acute-phase (SAA-enriched) HDL by J774 macrophages, (2) elucidating whether the presence of SAA on HDL particles affects selective uptake of HDL-associated cholesteryl esters, and (3) comparing cellular cholesterol efflux mediated by native and acute-phase HDL. Both the total and the specific binding at 4 degrees C of rabbit acute-phase HDL were approximately 2-fold higher than for native HDL. Nonlinear regression analysis revealed K(d) values of 7.0 x 10(-7) mol/L (native HDL) and 3.1 x 10(-7) mol/L (acute-phase HDL), respectively. The corresponding B(max) values were 203 ng of total lipoprotein per milligram of cell protein (native HDL) and 250 ng of total lipoprotein per milligram of cell protein (acute-phase HDL). At 37 degrees C, holoparticle turnover was slightly enhanced for acute-phase HDL, a fact reflected by 2-fold higher degradation rates. In contrast, the presence of SAA on HDL specifically increased (1. 7-fold) the selective uptake of HDL cholesteryl esters from acute-phase HDL by J774 macrophages, a widely used in vitro model to study foam cell formation and cholesterol efflux properties. Although ligand blotting experiments with solubilized J774 membrane proteins failed to identify the scavenger receptor-BI as a binding protein for both native and acute-phase HDL, 2 binding proteins with molecular masses of 100 and 72 kDa, the latter comigrating with CD55 (also termed decay-accelerating factor), were identified. During cholesterol efflux studies, it became apparent that the ability of acute-phase HDL with regard to cellular cholesterol removal was considerably lower than that for native HDL. This was reflected by a 1.7-fold increase in tau/2 values (22 versus 36 hours; native versus acute-phase HDL). Our observations of increased HDL cholesteryl ester uptake and reduced cellular cholesterol efflux (acute-phase versus native HDL) suggest that displacement of apolipoprotein A-I by SAA results in considerable altered metabolic properties of its main physiological carrier. These changes in the apolipoprotein moieties appear (at least in the in vitro system tested) to transform an originally antiatherogenic into a proatherogenic lipoprotein particle.


Subject(s)
Acute-Phase Reaction/immunology , Apolipoproteins/metabolism , Cholesterol, HDL/metabolism , Macrophages/immunology , Macrophages/metabolism , Serum Amyloid A Protein/metabolism , Animals , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/pharmacokinetics , Epitopes/immunology , Ligands , Macrophages/cytology , Mice , Phosphoproteins/blood , Protein Binding/immunology , Rabbits , Triglycerides/blood , Tritium
12.
J Cardiovasc Pharmacol ; 32(4): 582-92, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9781926

ABSTRACT

The pineal hormone, melatonin, was recently found to be a potent free scavenger for hydroxyl and peroxyl radicals. Melatonin also inhibits neuronal and thymocyte damage due to oxidative stress. Atherosclerosis development is mediated by low-density lipoprotein (LDL) oxidation and the endocytosis of oxidized LDL by resident macrophages in the subendothelial vascular wall. Furthermore, the cytotoxic effect of oxidized LDL increases atherogenicity. The goal of this study was to compare the antioxidant activities of melatonin and vitamin E against in vitro LDL oxidation and their cytoprotective actions against oxidized LDL-induced endothelial cell toxicity. An attempt at loading LDL with melatonin by incubating human plasma with an ethanolic melatonin solution gave only low protection against Cu2+-induced LDL oxidation in comparison with vitamin E and gave no detectable incorporation of melatonin into LDL, measured by high-performance liquid chromatography (HPLC) coupled to UV detection. High concentrations of melatonin (10-100 microM) added to the oxidative medium induced a clear inhibition of Cu2+-induced LDL oxidation, characterized as an increase in the lag-phase duration of conjugated diene formation and decreases in the maximal rate of the propagation phase and in the maximal amount of conjugated diene formation. Determination of the median efficacious dose (ED50) of melatonin and vitamin E by their ability to increase lag-phase duration showed that melatonin was less active than vitamin E (ED50, 79 vs. 10 microM, respectively). Melatonin was also less active than vitamin E in limiting the formation of thiobarbituric acid-reactive substances (TBARS) and LDL fluorescence intensity increase in the medium during Cu2+-induced LDL oxidation. Cu2+-induced LDL oxidation in the presence of 100 microM melatonin produced oxidized LDLs that were less recognizable for the scavenger receptors of J774 macrophages than were untreated LDLs. Vitamin E, 10 microM, was more active than 100 microM melatonin in inhibiting LDL oxidation and the resulting lipoprotein alterations leading to binding internalization and degradation by the J774 macrophages. Vitamin E, 100 microM, inhibited the pursuit of the oxidation of oxidized LDL mediated by bovine aortic endothelial cells (BAECs) in a culture medium containing Cu2+, whereas 100 microM melatonin had no antioxidant effect. Melatonin, 100 microM, as well as 100 microM vitamin E inhibited intracellular TBARS formation during the incubation of BAECs with highly oxidized LDL but had no influence on the increase in glutathione (GSH) concentration during this lengthy exposure (16 h) of BAECs to highly oxidized LDL. During this period, the same dose of vitamin E but not of melatonin tended to limit the decrease in adenosine triphosphate (ATP) concentration. Vitamin E, 100 microM, did not significantly reduce cellular lactate dehydrogenase (LDH) release in the culture medium during the incubation of oxidized LDL with BAECs, whereas 100 microM melatonin dramatically increased this release. These data show that melatonin is less active than vitamin E in inhibiting in vitro LDL oxidation and does not inhibit the cytotoxicity of oxidized LDL toward cultured endothelial cells. The concentrations necessary to inhibit LDL oxidation are far beyond those found in human plasma (100 microM vs. 100 pM). Therefore our results indicate that the pineal hormone melatonin per se appears to have little antiatherogenic property in the in vitro oxidation of LDL and the cytoprotective action against the toxicity of oxidized LDL. Nevertheless, in vivo LDL oxidation takes place in the subendothelium of the artery wall, and nothing is known about the concentration of melatonin or its catabolites in this environment.


Subject(s)
Antioxidants/pharmacology , Lipid Peroxidation/drug effects , Lipoproteins, LDL/drug effects , Melatonin/pharmacology , Vitamin E/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Copper/pharmacology , Drug Interactions , Endothelium/drug effects , Humans , Lipoproteins, LDL/adverse effects , Lipoproteins, LDL/metabolism , Thiobarbituric Acid Reactive Substances/pharmacokinetics
13.
Clin Genet ; 54(1): 79-82, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9727746

ABSTRACT

Mutations in the LDL receptor (LDLR) or the apolipoprotein B-100 genes causing familial hypercholesterolemia (FH) and familial defective apolipoprotein B-100 (FDB), two of the most frequent inherited diseases, are the underlying genetic defects in a small proportion of patients suffering from premature atherosclerotic heart disease. Consequently, secure diagnostic tools for these conditions allowing early preventive measures are needed. Since clinical and biochemical diagnosis often is inaccurate, assays analyzing patient LDLR function and LDL affinity have been established. These assays are, however, not able clearly to differentiate between suspected FH/FDB samples and normal controls. To evaluate if this may be caused by other hitherto undescribed genetic defects or to failure of the functional assays, we undertook denaturing gradient gel electrophoresis based mutation screening of the LDLR gene and the codon 3456 3553 region of the apolipoprotein B gene in six French FH/FDB patients with normal outcomes on functional assays. In all six patients, pathogenic LDLR mutations were found, including three previously undescribed mutations, suggesting that failure of the functional assays explains the normal results found in some phenotypic FH/FDB patients and illustrating the need for DNA based screening techniques for routine genetic diagnosis in FH/FDB.


Subject(s)
Apolipoproteins B/genetics , Genetic Testing , Hyperlipoproteinemia Type II/genetics , Mutation , Receptors, LDL/genetics , Apolipoprotein B-100 , Apolipoproteins B/blood , DNA Mutational Analysis , HeLa Cells , Humans , Hyperlipoproteinemia Type II/blood , Receptors, LDL/blood
14.
Biochim Biophys Acta ; 1358(1): 103-12, 1997 Aug 21.
Article in English | MEDLINE | ID: mdl-9296527

ABSTRACT

Previous studies have indicated that in HepG2 cells HDL3-signalling involves glycosylphosphatidylinositol (GPI) anchored proteins. HDL3-binding to HepG2 cells was found to be enhanced by cellular preincubation with PI-PLC inhibitors and sensitive to a cellular preincubation with exogenous PI-PLC, suggesting that HDL3 binds directly on GPI-anchored proteins to initiate signaling. Moreover HDL3-binding was found to be partly inhibited by antibodies against the HDL-binding protein (AbHBP). HDL3, when binding to HepG2 cells, promoted the release in the culture medium of a 110 kDa protein that binds AbHBP, while a cellular preincubation with antibodies against the inositol-phosphoglycan (IPG) moiety of GPI-anchor (AbIPG), used to block lipolytic cleavage of the GPI-anchor, inhibits HDL3-induced release of the 110 kDa protein in the culture medium. In [3H]-PC prelabeled HepG2 cells, AbHBP were found to stimulate PC-hydrolysis and DAG generation within 5 min as did HDL3 stimulation. Cellular preincubation with AbIPG was found to inhibit only the HDL3-signal and not the AbHBP-signal, while a prior cellular pretreatment with PI-PLC from Bacillus cereus was found to inhibit the HDL3-and AbHBP-signal. Moreover cellular preincubation with AbHBP for 1 h at 37 degrees C was found to inhibit HDL3-signalling pathways. Our results suggest that in HepG2 cells a 110 kDa protein, which could be HBP, can be anchored to the membrane via GPI, and can function in HDL3-signalling pathways as binding sites.


Subject(s)
Carrier Proteins , Glycosylphosphatidylinositols/metabolism , Lipoproteins, HDL/metabolism , Membrane Proteins/metabolism , RNA-Binding Proteins , Receptors, Lipoprotein/metabolism , Antibodies/pharmacology , Binding Sites , Humans , Membrane Proteins/immunology , Phosphatidylcholines/metabolism , Phospholipases/antagonists & inhibitors , Receptors, Lipoprotein/immunology , Signal Transduction , Tumor Cells, Cultured
15.
Biochem J ; 328 ( Pt 2): 415-23, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9371696

ABSTRACT

To elucidate further the binding of high-density-lipoprotein subfraction 3 (HDL3) to cells, the involvement of glycosylphosphatidylinositol-anchored proteins (GPI-proteins) was studied. Treatment of cultured cells, such as fibroblasts or SK-MES-1 cells, with a phosphatidylinositol-specific phospholipase C (PI-PLC) significantly decreases specific HDL3 binding. Moreover, PI-PLC treatment of cultured cells or cellular plasma membrane fractions results in releasing proteins. These proteins have a soluble form and can also bind HDL3, as revealed by ligand blotting experiments with HDL3. In order to obtain enriched GPI-proteins, we used a detergent-free purification method to prepare a caveolar membrane fraction. In the caveolar fraction, we obtained, by ligand blotting experiments, the enrichment of two HDL3-binding proteins with molecular masses of 120 and 80 kDa. These proteins were also revealed in a plasma membrane preparation with two other proteins, with molecular masses of 150 and 104 kDa, and were sensitive to PI-PLC treatment. Electron microscopy also showed the binding of Au-labelled HDL3 inside the caveolar membrane invaginations. In SK-MES-1 cells, HDL3 are internalized into a particular structure, resulting in the accumulation and concentration of such specific membrane domains. To sum up, a demonstration has been made of the implication of GPI-proteins as well as caveolae in the binding of HDL3 to cells.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Endocytosis , Glycosylphosphatidylinositols/metabolism , Lipoproteins, HDL/metabolism , Biological Transport , Cells, Cultured , Fibroblasts/metabolism , Humans , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Protein Binding/drug effects , Skin/cytology , Skin/metabolism , Type C Phospholipases/pharmacology
16.
Atherosclerosis ; 120(1-2): 209-19, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8645362

ABSTRACT

The goal of this study was to compare the structural and biological characteristics of apolipoprotein (apo) B-100-containing particle subfractions isolated from poorly controlled diabetic patients with insulin-dependent diabetes (IDDM), and healthy controls matched for sex, age and body mass index (BMI). Different apo B-containing particles were isolated by sequential immunochromatography and were free of apo A-I, apo A-II, apo A-IV and apo(a). Particles lipoprotein (Lp) B/C-III contained apo B and apo C-III. They were free of apo E. Particles Lp B/E contained apo B and apo E. They were free of apo C-III. Particles Lp B were devoided of apo C-III and apo E. All these particles could contain other known apolipoproteins not cited here, as for example apo C-II and/or apo C-I. The plasma levels of cholesterol, triglycerides, phospholipids, apo A-I, B-100, C-III, E, total Lp B/C-III, total Lp B/E were not different between patients and controls. The physico-chemical properties of Lp B/C-III and Lp B/E were similar in both groups. Only Lp B from patients exhibited some changes, an increase in the size and a decrease in the cholesterol and cholesteryl ester levels. The conformational properties of the lipoproteins were studied through their immunoreactivity against four different anti-apo B-100 monoclonal antibodies (MAb) for which sequential epitopes have been located on the protein, and one MAb for which the epitope is conformationally expressed. Again, minor changes were observed between patients and controls, and only a slight decrease in the immunoreactivity of the epitope encompassing amino-acid residues 405 to 539 of Lp B and of the conformationally expressed epitope of Lp B/C-III were found in patients. Nevertheless, whatever these conformational and/or physico-chemical modifications may be, they were not sufficient to induce functional alterations in the binding of the particles from the patients to the LDL-receptor of HeLa cells. This study shows that IDDM is not associated with any significant abnormalities in the apo-containing lipoprotein particles. The excessive occurrence of coronary heart disease (CHD) and other atherosclerotic vascular disease in patients with IDDM must have other causes.


Subject(s)
Apolipoproteins B/analysis , Diabetes Mellitus, Type 1/blood , Lipoproteins/chemistry , Adult , Aged , Apolipoprotein B-100 , Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Blood Glucose/analysis , Coronary Disease/etiology , Coronary Disease/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Female , HeLa Cells/metabolism , Humans , Insulin/therapeutic use , Insulin Resistance , Lipids/blood , Male , Middle Aged , Receptors, LDL/metabolism
17.
J Lipid Res ; 36(5): 919-30, 1995 May.
Article in English | MEDLINE | ID: mdl-7658164

ABSTRACT

We studied the effect of in vitro moderate oxidation on low density lipoprotein (LDL) conformation and metabolism. LDL was modified with either copper ions or phospholipase A2 plus lipoxygenase and, in both cases, mild oxidative conditions were used. The resulting conformational changes were investigated by studying immunological and biological properties of oxidized LDL. The immunoreactivity of apolipoprotein (apo) B-100 was examined using seven monoclonal antibodies. The biological implications of conformational changes were provided by cell-lipoprotein interaction studies using human fibroblasts and mouse peritoneal macrophages. Enzymatically treated LDL presented a relatively less oxidative degree of modification because it generated lower levels of TBARS, and displayed a lower electronegativity and more comparable cellular interactions with those of native LDL. Nevertheless, dramatic immunological changes were measured on both forms of LDL, i.e., a significant increase in the immunoreactivity of an epitope located in the B/E receptor binding domain, but also at epitopes far from this site and located in the N-terminal part of the apoB-100 molecule. The immunoreactivity of the C-terminal region was in contrast, decreased. Yet, as compared with enzymatically oxidized LDL, much more dramatic structural changes with chemically modified LDL were observed, resulting in such a particular conformation of lipoprotein that its interaction with the macrophagic scavenger receptor was favored, but its recognition by the B/E receptor of fibroblast was abolished. In contrast, despite a lower interaction between enzymatically modified LDL and the B/E receptor, the metabolism of this lipoprotein was quite comparable with that of native LDL and its degradation with cultured macrophages was poor. The use of in vitro models is common for study of the relationship between oxidized LDL and atherogenesis in humans. The choice of the more appropriate way to modify lipoproteins is of interest and is discussed.


Subject(s)
Apolipoproteins B/immunology , Copper/pharmacology , Lipoproteins, LDL/physiology , Lipoxygenase/metabolism , Phospholipases A/metabolism , Antibodies, Monoclonal , Apolipoprotein B-100 , Fibroblasts/metabolism , Humans , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/drug effects , Macrophages/metabolism , Oxidation-Reduction , Phospholipases A2 , Reference Values , Structure-Activity Relationship
18.
Cell Mol Biol (Noisy-le-grand) ; 40(4): 461-81, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8061563

ABSTRACT

Lipoprotein receptors are membrane proteins which play a central role in lipid metabolism. Although cells are capable of synthetizing de novo cholesterol from acetate, cholesterol is mostly of food origin or synthetized by the liver. The liver is the only organ which can catabolize the cholesterol and clear it from the circulation into biliary acids. Cholesterol, triglycerides and phospholipids are carried in the blood and in the interstitial fluid in association with specific proteins called apolipoproteins (apo), and form the lipoproteins. Although lipoproteins can be separated by their physico-chemical properties (i.e. density), they are the result of continuous exchanges of lipids and apolipoproteins. Lipoproteins are secreted by the intestine and the liver. Enterocytes and hepatocytes associate, in their endoplasmic reticulum, apolipoproteins and lipids from dietary intake and/or endogenous synthesis to form chylomicrons (intestine) or Very Low Density Lipoproteins (VLDL, in the liver). Lipolysis by the lipases of the triglycerides leads to fatty acids which are delivered to cells by a non-receptor pathway. On the contrary, the delivery of cholesterol to cells is dependent of receptors which recognize the lipoproteins by their protein moiety. Peripheric cells use cholesterol from the Low Density Lipoproteins (LDL, final product of VLDL intravascular catabolism) by the LDL receptor pathway. By this receptor, hepatocytes can also perform the clearance of LDL from the organism. The LDL receptor, or B/E receptor, can recognize lipoproteins by both apo B or apo E. However, other receptors might exist to explain the normal catabolism of apo E- containing lipoproteins in patients genetically deficient in LDL receptor. One of the most characterized candidate protein for chylomicrons receptor is the LRP (LDL receptor Related Protein) which shares a strong homology with some domains of the LDL receptor, and which is shown to be the alpha 2-macroglobulin receptor previously described. Due to the delay in clearance by the liver, LDL can undergo oxidation. Oxidized LDL are not recognized by LDL receptor but rather "scavenger" receptors in macrophages and vascular endothelial smooth muscle cells. This metabolism leads to the formation of atherosclerotic plaques. High Density Lipoproteins (HDL) are implicated in the removal of excess cholesterol from peripheral cells and the transport to the liver. Specific HDL binding sites to several mammalian cells have been shown by numerous investigators and one candidate protein has been cloned. Analysis of HDL-induced signal transduction has been a very active field of research.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cholesterol/metabolism , Receptors, LDL/metabolism , Animals , Arteriosclerosis/metabolism , Humans , Receptors, LDL/chemistry
19.
Biochim Biophys Acta ; 1127(2): 124-30, 1992 Jul 29.
Article in English | MEDLINE | ID: mdl-1322706

ABSTRACT

Lipoprotein Lp(a) was isolated by immunoaffinity chromatography using anti apolipoprotein B and anti apolipoprotein (a) immunosorbents. Besides apolipoproteins (a) and B, this fraction was shown to contain apolipoproteins C and E. Therefore, it was decided to further purify this crude Lp(a) into particles containing apolipoprotein E and particles free of apo E, using chromatography with an anti apolipoprotein E immunosorbent. Lp(a), free of apolipoprotein E was cholesterol ester rich and triacylglycerol poor and was found mainly in the LDL size range. In contrast, Lp(a) containing apolipoprotein E was triacylglycerol rich and was distributed mainly in the VLDL and IDL size range. Binding of these two fractions, one containing apo E and one free of it, to the apo B/E receptor of HeLa cells was studied. Both fractions bound to the receptor but the one containing apo E had a better affinity than the one free of apo E. Further studies are needed to identify the clinical importance of these two different entities.


Subject(s)
Apolipoproteins E/isolation & purification , Apolipoproteins/isolation & purification , Lipoproteins/isolation & purification , Receptors, Lipoprotein , Apolipoproteins/chemistry , Apolipoproteins/metabolism , Apoprotein(a) , Chromatography, Affinity , HeLa Cells/metabolism , Humans , Lipids/blood , Lipoprotein(a) , Lipoproteins/chemistry , Lipoproteins/metabolism , Particle Size , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL