Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Clin Lipidol ; 16(5): 694-703, 2022.
Article in English | MEDLINE | ID: mdl-36002365

ABSTRACT

BACKGROUND: Loss of function variants of LIPG gene encoding endothelial lipase (EL) are associated with primary hyperalphalipoproteinemia (HALP), a lipid disorder characterized by elevated plasma levels of high density lipoprotein cholesterol (HDL-C). OBJECTIVE: Aim of the study was the phenotypic and genotypic characterization of a family with primary HALP. METHODS: HDL subclasses distribution was determined by polyacrylamide gradient gel electrophoresis. Serum content of preß-HDL was assessed by (2D)-electrophoresis. Cholesterol efflux capacity (CEC) of serum mediated by ABCA1, ABCG1 or SR-BI was assessed using cells expressing these proteins. Cholesterol loading capacity (CLC) of serum was assayed using cultured human macrophages. Next generation sequencing was used for DNA analysis. Plasma EL mass was determined by ELISA. RESULTS: Three family members had elevated plasma HDL-C, apoA-I and total phospholipids, as well as a reduced content of preß-HDL. These subjects were heterozygous carriers of a novel variant of LIPG gene [c.526 G>T, p.(Gly176Trp)] found to be deleterious in silico. Plasma EL mass in carriers was lower than in controls. CEC of sera mediated by ABCA1 and ABCG1 transporters was substantially reduced in the carriers. This effect was maintained after correction for serum HDL concentration. The sera of carriers were found to have a higher CLC in cultured human macrophages than control sera. CONCLUSION: The novel p.(Gly176Trp) variant of endothelial lipase is associated with changes in HDL composition and subclass distribution as well as with functional changes affecting cholesterol efflux capacity of serum which suggest a defect in the early steps of revere cholesterol transport.


Subject(s)
Cholesterol , High-Density Lipoproteins, Pre-beta , Humans , High-Density Lipoproteins, Pre-beta/metabolism , ATP Binding Cassette Transporter 1/genetics , Cholesterol, HDL , Lipase/genetics
2.
J Clin Lipidol ; 14(5): 730-739, 2020.
Article in English | MEDLINE | ID: mdl-32868248

ABSTRACT

BACKGROUND: Preß1-high-density lipoprotein (HDL) is a lipid-poor cholesterol acceptor that is converted to lipid-rich HDL by lecithin-cholesterol acyltransferase (LCAT). In patients receiving hemodialysis, preß1-HDL metabolism is hampered even if HDL cholesterol is normal. Hemodialysis may affect preß1-HDL metabolism by releasing lipases from the vascular wall due to heparin. OBJECTIVES: We investigated whether preß1-HDL metabolism is delayed in patients with chronic kidney disease (CKD) who are not receiving hemodialysis. METHODS: We examined 44 patients with Stage 3 or higher CKD and 22 healthy volunteers (Control group). The patients with CKD were divided into those without renal replacement therapy (CKD group, n = 22) and those undergoing continuous ambulatory peritoneal dialysis (CAPD group, n = 22). Plasma preß1-HDL concentrations were determined by immunoassay. During incubation at 37°C, we used 5,5-dithio-bis (2-nitrobenzoic acid) (DTNB) to inhibit LCAT activity and defined the conversion halftime of preß1-HDL (CHTpreß1) as the time required for the difference in preß1-HDL concentration in the presence and absence of 5,5-DTNB to reach half the baseline concentration. RESULTS: The absolute and relative preß1-HDL concentrations were higher, and CHTpreß1 was longer in the CKD and CAPD groups than in the Control group. Preß1-HDL concentration was significantly correlated with CHTpreß1 but not with LCAT activity in patients with CKD and CAPD. CONCLUSION: Preß1-HDL metabolism is delayed in patients with CKD who are not on hemodialysis. This preß1-HDL metabolic delay may progress as renal function declines.


Subject(s)
High-Density Lipoproteins, Pre-beta/metabolism , Renal Dialysis/methods , Renal Insufficiency, Chronic/metabolism , Renal Replacement Therapy/methods , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Prognosis , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/therapy
3.
Article in English | MEDLINE | ID: mdl-27418968

ABSTRACT

AIMS: Apolipoprotein A-1 (ApoA-1), based on epidemiology, is inversely associated with cardiovascular (CV) events. Human carriers of the ApoA-1 Milano variant have a reduced incidence of CV disease. Regression of atherosclerotic plaque burden was previously observed on intravascular ultrasound (IVUS) with ETC-216, a predecessor of MDCO-216. MDCO-216, a complex of dimeric ApoA-1 Milano and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine, is being developed to reduce atherosclerotic plaque burden and CV events. We investigated the efficacy and safety of a single infusion of MDCO-216 in healthy volunteers and in patients with coronary artery disease (CAD). METHODS AND RESULTS: Twenty-four healthy volunteers and 24 patients with documented CAD received a 2-h infusion of MDCO-216 in a randomized, placebo controlled, single ascending dose study. Five cohorts of healthy volunteers and four cohorts of CAD patients received ApoA-1 Milano doses ranging from 5 to 40 mg/kg. Subjects were followed for 30 days. Dose-dependent increases in ApoA-1, phospholipid, and pre-beta 1 HDL and decreases in ApoE were observed. Prominent and sustained increases in triglyceride, and decreases in HDL-C, endogenous ApoA-1 and ApoA-II occurred at doses >20 mg/kg and profound increases in ABCA1-mediated cholesterol efflux were observed. Other lipid and lipoprotein parameters were generally unchanged. MDCO-216 was well tolerated. CONCLUSIONS: MDCO-216-modulated lipid parameters profoundly increased ABCA1-mediated cholesterol efflux and was well tolerated. These single-dose data support further development of this agent for reducing atherosclerotic disease and subsequent CV events.


Subject(s)
Apolipoprotein A-I/pharmacology , Coronary Artery Disease/drug therapy , Phosphatidylcholines/pharmacology , ATP Binding Cassette Transporter 1/metabolism , Adult , Aged , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Cholesterol/metabolism , Coronary Artery Disease/metabolism , Drug Combinations , Female , Healthy Volunteers , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Male , Middle Aged , Phosphatidylcholines/administration & dosage , Phospholipids/metabolism , Triglycerides/metabolism
4.
PLoS One ; 10(7): e0131997, 2015.
Article in English | MEDLINE | ID: mdl-26207756

ABSTRACT

Apolipoprotein (apo) mimetic peptides replicate some aspects of HDL function. We have previously reported the effects of compound ATI-5261 on its ability to replicate many functions of native apo A-I in the process of HDL biogenesis. ATI-5261 induced muscle toxicity in wild type C57Bl/6 mice, increased CPK, ALT and AST and increase in triglyceride (Tg) levels. Aromatic phenylalanine residues on the non-polar face of ATI-5261, together with positively charged arginine residues at the lipid-water interface were responsible for these effects. This information was used to create a novel analog (CS-6253) that was non-toxic. We evaluated this peptide designed from the carboxyl terminus of apo E, in its ability to mimic apo A-I functionality. Our data shows that the lipidated particles generated by incubating cells overexpressing ABCA1 with lipid free CS-6253 enhances the rate of ABCA1 lipid efflux with high affinity interactions with native ABCA1 oligomeric forms and plasma membrane micro-domains. Interaction between ABCA1 and lipid free CS-6253 resulted in formation of nascent HDL-CS-6253 particles that are actively remodeled in plasma. Mature HDL-CS-6253 particles deliver cholesterol to liver cells via SR-BI in-vitro. CS-6253 significantly increases cholesterol efflux in murine macrophages and in human THP-1 macrophage-derived foam cells expressing ABCA1. Addition of CS-6253 to plasma dose-dependently displaced apo A-I from α-HDL particles and led to de novo formation of preß-1 HDL that stimulates ABCA1 dependent cholesterol efflux efficiently. When incubated with human plasma CS-6253 was also found to bind with HDL and LDL and promoted the transfer of cholesterol from HDL to LDL predominantly. Our data shows that CS-6253 mimics apo A-I in its ability to promote ABCA1-mediated formation of nascent HDL particles, and enhances formation of preß-1 HDL with increase in the cycling of apo A-I between the preß and α-HDL particles in-vitro. These mechanisms are potentially anti-atherogenic.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoproteins E/metabolism , Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Peptides/pharmacology , ATP Binding Cassette Transporter 1/agonists , ATP Binding Cassette Transporter 1/genetics , Animals , Apolipoprotein A-I/metabolism , Apolipoproteins E/chemistry , Biological Transport/drug effects , CD36 Antigens/metabolism , Cell Line , Cell Line, Tumor , Dose-Response Relationship, Drug , Foam Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Macrophages/metabolism , Male , Mice, Inbred C57BL , Peptides/metabolism , Phospholipids/metabolism , Rats , Time Factors
5.
J Nutr Biochem ; 26(2): 165-72, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25468613

ABSTRACT

High-density lipoproteins (HDLs) are atheroprotective because of their role in reverse cholesterol transport. The intestine is involved in this process because it synthesizes HDL, removes cholesterol from plasma and excretes it into the lumen. We investigated the role of selected dietary fatty acids on intestinal cholesterol uptake and HDL functionality. Caco-2 monolayers grown on Transwells were supplemented with either palmitic, palmitoleic, oleic, linoleic, docosahexaenoic, eicosapentaenoic, arachidonic or conjugated linoleic acids (CLAs): c9,t11-CLA; t9,t11-CLA; c10,t12-CLA. Cells synthesized HDL in the basolateral compartment for 24 h in the absence or presence of an antibody to SR-BI (aSR-BI), which inhibits its interaction with HDL. Free cholesterol (FC) accumulated to a greater extent in the presence than in the absence of aSR-BI, indicating net uptake of FC by SR-BI. Uptake's efficiency was significantly decreased when cells were treated with c9,t11-CLA relative to the other fatty acids. These differences were associated with lower HDL functionality, since neither SR-BI protein expression nor expression and alternative splicing of other genes involved lipid metabolism were affected. Only INSIG2 expression was decreased, with no increase of its target genes. Increasing pre-ß-HDL synthesis, by inducing ABCA1 and adding APOA1, resulted in reduced uptake of FC by SR-BI after c9,t11-CLA treatment, indicating reduced functionality of pre-ß-HDL. Conversely, treatment with c9,t11-CLA resulted in a greater uptake of FC and esterified cholesterol from mature HDL. Therefore, Caco-2 monolayers administered c9,t11-CLA produced a nonfunctional pre-ß-HDL but took up cholesterol more efficiently via SR-BI from mature HDL.


Subject(s)
Cholesterol, Dietary/metabolism , Cholesterol, HDL/metabolism , Enterocytes/metabolism , Enterohepatic Circulation , Intestinal Absorption , Linoleic Acids, Conjugated/metabolism , Lipoproteins, HDL/metabolism , Alternative Splicing , Biological Transport , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/genetics , CD36 Antigens/metabolism , Caco-2 Cells , Cell Polarity , Cholesterol Esters/metabolism , Cholesterol, HDL/blood , Enterocytes/cytology , Gene Expression Regulation , High-Density Lipoproteins, Pre-beta/genetics , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Lipoproteins, HDL/blood , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Stereoisomerism
6.
Biochem Biophys Res Commun ; 450(1): 124-8, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24866239

ABSTRACT

Apolipoprotein E3 (apoE3) is an anti-atherogenic apolipoprotein with the ability to exist in lipid-free and lipoprotein-associated states. During atherosclerosis, its function in promoting cholesterol efflux from macrophages via the ATP-binding cassette transporter A1 (ABCA1) takes a prominent role, leading to generation of nascent high density lipoprotein (nHDL) particles. The objective of this study is to understand the conformation adopted by apoE3 in macrophage-generated nHDL using a fluorescence spectroscopic approach involving pyrene. Pyrene-labeled recombinant human apoE3 displayed a robust ability to stimulate ABCA1-mediated cholesterol efflux from cholesterol-loaded J774 macrophages (which do not express apoE), comparable to that elicited by unlabeled apoE3. The nHDL recovered from the conditioned medium revealed the presence of apoE3 by immunoblot analysis. A heterogeneous population of nHDL bearing exogenously added apoE3 was generated with particle size varying from ∼12 to ∼19 nm in diameter, corresponding to molecular mass of ∼450 to ∼700 kDa. The lipid: apoE3 ratio varied from ∼60:1 to 10:1. A significant extent of pyrene excimer emission was noted in nHDL, indicative of spatial proximity between Cys112 on neighboring apoE3 molecules similar to that noted in reconstituted HDL. Cross-linking analysis using Cys-specific cross-linkers revealed the predominant presence of dimers. Taken together the data indicate a double belt arrangement of apoE molecules on nHDL. A similar organization of the C-terminal tail of apoE on nHDL was noted when pyrene-apoEA277C(201-299) was used as the cholesterol acceptor. These studies open up the possibility of using exogenously labeled apoE3 to generate nHDL for structural and conformational analysis.


Subject(s)
Apolipoprotein E3/chemistry , Apolipoprotein E3/metabolism , High-Density Lipoproteins, Pre-beta/chemistry , High-Density Lipoproteins, Pre-beta/metabolism , Macrophages/metabolism , Pyrenes/chemistry , Spectrometry, Fluorescence/methods , Animals , Cell Line , Humans , Mice , Microscopy, Fluorescence/methods , Protein Conformation , Pyrenes/metabolism , Staining and Labeling
7.
Eur J Clin Invest ; 44(3): 240-8, 2014.
Article in English | MEDLINE | ID: mdl-24325778

ABSTRACT

BACKGROUND: The importance of functional properties of high-density lipoproteins (HDL) for atheroprotection is increasingly recognized. We determined the impact of lipid-lowering therapy on 3 key HDL functionalities in Type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS: A placebo-controlled, randomized cross-over study (three 8-week treatment periods with simvastatin (40 mg daily), bezafibrate (400 mg daily), alone and in combination) was carried out in 14 men with T2DM. Cholesterol efflux was determined using human THP-1 monocyte-derived macrophages, HDL antioxidative capacity was measured as inhibition of low-density lipoprotein oxidation in vitro, and HDL anti-inflammatory capacity was assessed as suppression of thrombin-induced monocyte chemotactic protein 1 expression in human umbilical vein endothelial cells. Pre-ß-HDL was assayed using crossed immunoelectrophoresis. RESULTS: While cholesterol efflux increased in response to simvastatin, bezafibrate and combination treatment (+12 to +23%; anova, P = 0.001), HDL antioxidative capacity (P = 0.23) and HDL anti-inflammatory capacity (P = 0.15) did not change significantly. Averaged changes in cellular cholesterol efflux during active treatment were correlated positively with changes in HDL cholesterol, apoA-I and pre-ß-HDL (P < 0.05 to P < 0.001). There were no inter-relationships between changes in the three HDL functionalities during treatment (P > 0.10). Changes in HDL antioxidative capacity and anti-inflammatory capacity were also unrelated to changes in HDL cholesterol and apoA-I, while changes in HDL antioxidative capacity were related inversely to pre-ß-HDL (P < 0.05). CONCLUSION: Simvastatin and bezafibrate increase cholesterol efflux, parallel to HDL cholesterol and apoA-I responses. The antioxidative and anti-inflammatory properties of HDL are not to an important extent affected by these therapeutic interventions.


Subject(s)
Bezafibrate/therapeutic use , Cholesterol, HDL/metabolism , Diabetes Mellitus, Type 2/drug therapy , High-Density Lipoproteins, Pre-beta/metabolism , Hypolipidemic Agents/therapeutic use , Lipoproteins, LDL/metabolism , Simvastatin/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cholesterol/metabolism , Cross-Over Studies , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Drug Therapy, Combination , Humans , Male , Middle Aged
8.
Curr Opin Lipidol ; 23(4): 367-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22517613

ABSTRACT

PURPOSE OF REVIEW: A negative correlation between HDL cholesterol levels and risk of coronary artery disease has long been recognized. Emerging knowledge of the molecular speciation and functional properties of HDL provides an opportunity to study the atheroprotective effects of specific metabolic processes. The discovery of the quantum particle among the molecular species of HDL (prebeta-1 HDL) and its role in cholesterol efflux from the artery wall, offer a means of assessing the efficiency of efflux. This review presents observations on the structure and metabolism of this particle and its emerging role as a predictor of risk for atherosclerotic vascular disease. RECENT FINDINGS: Prebeta-1 HDL is now recognized as the primary acceptor of cholesterol effluxed by the dominant ATP-binding cassette A1 (ABCA1) transporter in arterial macrophages, a critical step in reverse cholesterol transport. Several studies have revealed an association between high levels of this particle and risk of globally defined coronary artery disease and carotid intima-media thickness. Recently, these findings have been confirmed and extended to include myocardial infarction. High levels of prebeta-1 HDL may serve as an index of functional impairment of cholesterol efflux or esterification, either of which would be expected to impede reverse cholesterol transport. SUMMARY: Recent studies underscore the critical role of prebeta-1 HDL in reverse cholesterol transport and its use as a marker of risk for structural coronary disease, myocardial infarction, and cerebral vascular disease.


Subject(s)
Coronary Disease/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Biological Transport , Cholesterol/metabolism , Humans
9.
J Lipid Res ; 53(5): 941-950, 2012 May.
Article in English | MEDLINE | ID: mdl-22389325

ABSTRACT

The lipidation of apoA-I in liver greatly influences HDL biogenesis and plasma HDL levels by stabilizing the secreted apoA-I. Niacin is the most effective lipid-regulating agent clinically available to raise HDL. This study was undertaken to identify regulatory mechanisms of niacin action in hepatic lipidation of apoA-I, a critical event involved in HDL biogenesis. In cultured human hepatocytes (HepG2), niacin increased: association of apoA-I with phospholipids and cholesterol by 46% and 23% respectively, formation of lipid-poor single apoA-I molecule-containing particles up to ~2.4-fold, and pre ß 1 and α migrating HDL particles. Niacin dose-dependently stimulated the cell efflux of phospholipid and cholesterol and increased transcription of ABCA1 gene and ABCA1 protein. Mutated DR4, a binding site for nuclear factor liver X receptor alpha (LXR α ) in the ABCA1 promoter, abolished niacin stimulatory effect. Further, knocking down LXR α or ABCA1 by RNA interference eliminated niacin-stimulated apoA-I lipidation. Niacin treatment did not change apoA-I gene expression. The present data indicate that niacin increases apoA-I lipidation by enhancing lipid efflux through a DR4-dependent transcription of ABCA1 gene in HepG2 cells. A stimulatory role of niacin in early hepatic formation of HDL particles suggests a new mechanism that contributes to niacin action to increase the stability of newly synthesized circulating HDL.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Apolipoprotein A-I/metabolism , Cholesterol, HDL/biosynthesis , Niacin/pharmacology , Repetitive Sequences, Nucleic Acid/genetics , Transcription, Genetic/drug effects , ATP Binding Cassette Transporter 1 , Biological Transport/drug effects , Culture Media/metabolism , Gene Expression Regulation/drug effects , Hep G2 Cells , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Movement/drug effects , Phospholipids/metabolism , Repetitive Sequences, Nucleic Acid/drug effects
10.
J Lipid Res ; 53(4): 767-75, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22338009

ABSTRACT

In familial hypercholesterolemia (FH), low HDL cholesterol (HDL-C) levels are associated with functional alterations of HDL particles that reduce their capacity to mediate the reverse cholesterol transport (RCT) pathway. The objective of this study was to evaluate the consequences of LDL apheresis on the efficacy of the RCT pathway in FH patients. LDL apheresis markedly reduced abnormal accelerated cholesteryl ester transfer protein (CETP)-mediated cholesteryl ester (CE) transfer from HDL to LDL, thus reducing their CE content. Equally, we observed a major decrease (-53%; P < 0.0001) in pre-ß1-HDL levels. The capacity of whole plasma to mediate free cholesterol efflux from human macrophages was reduced (-15%; P < 0.02) following LDL apheresis. Such reduction resulted from a marked decrease in the ABCA1-dependent efflux (-71%; P < 0.0001) in the scavenger receptor class B type I-dependent efflux (-21%; P < 0.0001) and in the ABCG1-dependent pathway (-15%; P < 0.04). However, HDL particles isolated from FH patients before and after LDL apheresis displayed a similar capacity to mediate cellular free cholesterol efflux or to deliver CE to hepatic cells. We demonstrate that rapid removal of circulating lipoprotein particles by LDL apheresis transitorily reduces RCT. However, LDL apheresis is without impact on the intrinsic ability of HDL particles to promote either cellular free cholesterol efflux from macrophages or to deliver CE to hepatic cells.


Subject(s)
Blood Component Removal/methods , Cholesterol, HDL/metabolism , Cholesterol, LDL/isolation & purification , Hyperlipoproteinemia Type II/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adult , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Biological Transport , CHO Cells , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Esters/metabolism , Cricetinae , Esterification , Female , High-Density Lipoproteins, Pre-beta/genetics , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/therapy , Lipid Metabolism , Macrophages/metabolism , Male , Mice , Middle Aged , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Young Adult
11.
J Cardiovasc Pharmacol Ther ; 17(3): 315-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22067613

ABSTRACT

High-density lipoprotein (HDL)-targeting therapies, including reconstituted HDL (rHDL), are attractive agents for treating dyslipidemia and atherosclerosis, as they may increase HDL levels and enhance therapeutic activities associated with HDL, including reverse cholesterol transport (RCT). Using CSL-111, a rHDL consisting of native human apolipoprotein AI (hApoAI) and phospholipids, we characterized the acute effects of rHDL administration in C57Bl/6 mice to (i) further our understanding of the mechanism of action of rHDL, and (ii) evaluate the usefulness of the mouse as a preclinical model for HDL-targeting therapies. After a single injection of CSL-111, there was a dose- and time-dependent increase of hApoAI, human pre-ß HDL, total cholesterol, and triglycerides in serum, consistent with the effects of CSL-111 in humans. However, unlike in humans, there was no measurable increase in cholesteryl esters. Evaluated ex vivo, the ATP binding cassette A1 (ABCA1)- and scavenger receptor type BI (SR-BI)-dependent cholesterol efflux capacity of serum from CSL-111-treated mice was increased compared with serum from vehicle-treated animals. Fractionation by size exclusion chromatography of lipoproteins in serum from treated mice revealed hApoAI in particles the size of endogenous HDL and slightly larger, cholesterol-enriched particles of all sizes, including sizes distinct from endogenous HDL or CSL-111 itself, and triglyceride-enriched particles the size of very-low-density lipoprotein (VLDL). These results suggest that in mouse blood CSL-111 is remodeled and generates enhanced cholesterol efflux capacity which increases mobilization of free cholesterol from peripheral tissues. Our findings complement the previous reports on CSL-111 in human participants and provide data with which to evaluate the potential utility of mouse models in mechanistic studies of HDL-targeting therapies.


Subject(s)
Cholesterol, HDL/pharmacology , Lipids/blood , Animals , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Cell Line , Cholesterol/metabolism , Cholesterol, HDL/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation , High-Density Lipoproteins, Pre-beta/metabolism , Injections, Intravenous , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphatidylcholines
12.
J Lipid Res ; 52(12): 2323-2331, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21971713

ABSTRACT

The composition of lipoproteins and the association of proteins with various particles are of much interest in the context of cardiovascular disease. Here, we describe a technique for the multidimensional analysis of lipoproteins and their associated apolipoproteins. Plasma is separated by size exclusion chromatography (SEC), and fractions are analyzed by reverse-phase arrays. SEC fractions are spotted on nitrocellulose slides and incubated with different antibodies against individual apolipoproteins or antibodies against various apolipoproteins. In this way, tens of analytes can be measured simultaneously in 100 µl of plasma from a single SEC separation. This methodology is particularly suited to simultaneous analysis of multiple proteins that may change their distribution to lipoproteins or alter their conformation, depending on factors that influence circulating lipoprotein size or composition. We observed changes in the distribution of exchangeable apolipoproteins following addition of recombinant apolipoproteins or interaction with exogenous compounds. While the cholesteryl ester transfer protein (CETP)-dependent formation of pre-ß-HDL was inhibited by the CETP inhibitors torcetrapib and anacetrapib, it was not reduced by the CETP modulator dalcetrapib. This finding was elucidated using this technique.


Subject(s)
Blood Chemical Analysis/methods , Chromatography, Gel/methods , Lipoproteins/blood , Lipoproteins/isolation & purification , Protein Array Analysis/methods , Antibody Specificity , Artifacts , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Ester Transfer Proteins/pharmacology , High-Density Lipoproteins, Pre-beta/blood , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Lipoproteins/immunology , Lipoproteins/metabolism , Quinolines/pharmacology , Reproducibility of Results
13.
Curr Opin Lipidol ; 22(5): 394-402, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21881503

ABSTRACT

PURPOSE OF REVIEW: Both quantity and quality of the circulating HDL particle matter for the optimal antiatherogenic potential of HDL. This review summarizes various mechanisms capable of inducing extracellular modifications of HDL and reducing the function of HDL subclasses as cholesterol acceptors. Special emphasis is laid on the proteolytic inactivation of lipid-poor preß-migrating HDL (preß-HDL). RECENT FINDINGS: HDL particles can undergo functional inactivation in vivo. During atherogenesis, different cell types in the arterial intima release enzymes into the intimal fluid, potentially capable of causing structural and chemical modifications of the various components present in the lipid core or in the polar surface of the HDL particles. Enzymatic oxidation, lipolysis and proteolysis, and nonenzymatic glycosylation are among the HDL modifications that adversely affect HDL functionality. Proteolysis of preß-HDL by various proteases present in the arterial intima has emerged as a potential mechanism that impairs the efficiency of HDL to promote cholesterol efflux from macrophage foam cells, the mast cell-derived neutral protease chymase being a prime example of such impairment. A paradigm of proteolytic inactivation of preß-HDL in vivo is emerging. SUMMARY: Several extracellular enzymes present in the arterial intima may compromise various cardioprotective functions of HDL. Observations on proteolysis of specific lipid-poor HDL subpopulations in vivo constitute the basis for future studies evaluating the actual impact of proteolytic microenvironments on the initiation and progression of atherosclerotic lesions.


Subject(s)
Atherosclerosis/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Lipoproteins, HDL/metabolism , Animals , Humans , Proteolysis
14.
Acta Biochim Pol ; 58(3): 275-85, 2011.
Article in English | MEDLINE | ID: mdl-21750785

ABSTRACT

The pre-ß HDL fraction constitutes a heterogeneous population of discoid nascent HDL particles. They transport from 1 to 25 % of total human plasma apo A-I. Pre-ß HDL particles are generated de novo by interaction between ABCA1 transporters and monomolecular lipid-free apo A-I. Most probably, the binding of apo A-I to ABCA1 initiates the generation of the phospholipid-apo A-I complex which induces free cholesterol efflux. The lipid-poor nascent pre-ß HDL particle associates with more lipids through exposure to the ABCG1 transporter and apo M. The maturation of pre-ß HDL into the spherical α-HDL containing apo A-I is mediated by LCAT, which esterifies free cholesterol and thereby forms a hydrophobic core of the lipoprotein particle. LCAT is also a key factor in promoting the formation of the HDL particle containing apo A-I and apo A-II by fusion of the spherical α-HDL containing apo A-I and the nascent discoid HDL containing apo A-II. The plasma remodelling of mature HDL particles by lipid transfer proteins and hepatic lipase causes the dissociation of lipid-free/lipid-poor apo A-I, which can either interact with ABCA1 transporters and be incorporated back into pre-existing HDL particles, or eventually be catabolized in the kidney. The formation of pre-ß HDL and the cycling of apo A-I between the pre-ß and α-HDL particles are thought to be crucial mechanisms of reverse cholesterol transport and the expression of ABCA1 in macrophages may play a main role in the protection against atherosclerosis.


Subject(s)
Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/metabolism , Apolipoproteins/metabolism , Apolipoproteins M , High-Density Lipoproteins, Pre-beta/chemistry , Humans , Lipase/metabolism , Lipocalins/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism
15.
Biophys J ; 101(2): 353-61, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21767487

ABSTRACT

Apolipoprotein A-I (ApoA-I) is the principle protein component of HDL, also known as "good cholesterol," which is an inverse marker for cardiovascular disease. The N-terminal 44 amino acids of ApoA-I (N44) are predicted to be responsible for stabilization of soluble ApoA-I, whereas the C-terminal 46 amino acids (C46) are predicted to initiate lipid binding and oligomerization. In this work, we apply what we believe to be a novel application of drop tensiometry to study the adsorption and desorption of N44 and C46 at a triolein/POPC/water (TO/POPC/W) interface. The amount of peptide that adsorbed to the surface was dependent on the surface concentration of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) and pressure (Π) before adsorption. At a TO/POPC/W interface, the exclusion pressure (Π(EX)) of C46 was 25.8 mN/m, and was 19.3 mN/m for N44. Once adsorbed, both peptides formed a homogeneous surface with POPC but were progressively ejected from the surface by compression. During a compression, C46 removed POPC from the surface whereas N44 did not. Repeated compressions caused C46 to deplete entirely the surface of phospholipid. If full-length ApoA-I could also remove phospholipid, this could provide a mechanism for the transfer of surface components of chylomicrons and very low density lipoprotein to high density lipoprotein with the assistance of phospholipid transfer protein.


Subject(s)
Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Phosphatidylcholines/chemistry , Phospholipids/isolation & purification , Triolein/chemistry , Water/chemistry , Adsorption , Models, Molecular , Peptides/metabolism , Structure-Activity Relationship , Temperature
16.
Atherosclerosis ; 219(1): 259-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21696738

ABSTRACT

OBJECTIVES: Increases of homocysteine (Hcy) by fenofibrate correlated inversely to changes in HDL-C and apoA-I in the FIELD study. This finding raised the question whether high Hcy may influence HDL function and counteract benefits of fenofibrate on cardiovascular outcomes. In a subset of the FIELD study we investigated whether fenofibrate therapy or high Hcy, separately or in concert, modulate: (1) ability of plasma or HDL to facilitate cholesterol efflux from THP-1 foam cells; (2) plasma potential to generate preß-HDL; (3) plasma phospholipid transfer protein (PLTP) activity, serum PON-1 mass and activity, HDL particle size and distribution. METHODS: We selected 33 subjects in the FIELD fenofibrate arm according to quartiles of Hcy at 5th year: 17 subjects were in the lowest (Low Hcy group) and 16 subjects were in the highest quartile (High Hcy group). In addition, 14 subjects allocated to placebo were matched by close-out Hcy levels to Low Hcy group. This design allowed us to examine the effects of both fenofibrate (comparison between placebo vs Low Hcy groups) and Hcy (comparison between close-out Low and High Hcy groups) on plasma and HDL ability to facilitate cellular cholesterol removal in the efflux assay in vitro using THP-1 foam cells. RESULTS: Hcy levels were 13.3±0.7 µmol/L (placebo), 13.2±2 µmol/L (Low Hcy) and 27.4±6.5 µmol/L (High Hcy). Cholesterol efflux values to HDL and plasma, percentage of plasma preß-HDL, PLTP activity, serum PON-1 mass and HDL particle size and distribution were similar in both fenofibrate groups and comparable to those of the placebo group. CONCLUSIONS: In the present study cohort fenofibrate and high Hcy levels did not modulate HDL and plasma functions in the first step of reverse cholesterol transport, cholesterol efflux from foam cells.


Subject(s)
Cholesterol, HDL/blood , Cholesterol/metabolism , Fenofibrate/therapeutic use , Foam Cells/metabolism , Homocysteine/blood , Aged , Aryldialkylphosphatase/blood , Cells, Cultured , Cholesterol/blood , Cohort Studies , Diabetes Mellitus, Type 2/drug therapy , Female , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Macrophages/metabolism , Male , Middle Aged , Particle Size , Phospholipid Transfer Proteins/blood
17.
J Lipid Res ; 51(12): 3443-54, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861162

ABSTRACT

The mechanism by which cholesteryl ester transfer protein (CETP) activity affects HDL metabolism was investigated using agents that selectively target CETP (dalcetrapib, torcetrapib, anacetrapib). In contrast with torcetrapib and anacetrapib, dalcetrapib requires cysteine 13 to decrease CETP activity, measured as transfer of cholesteryl ester (CE) from HDL to LDL, and does not affect transfer of CE from HDL3 to HDL2. Only dalcetrapib induced a conformational change in CETP, when added to human plasma in vitro, also observed in vivo and correlated with CETP activity. CETP-induced pre-ß-HDL formation in vitro in human plasma was unchanged by dalcetrapib ≤3 µM and increased at 10 µM. A dose-dependent inhibition of pre-ß-HDL formation by torcetrapib and anacetrapib (0.1 to 10 µM) suggested that dalcetrapib modulates CETP activity. In hamsters injected with [³H]cholesterol-labeled autologous macrophages, and given dalcetrapib (100 mg twice daily), torcetrapib [30 mg once daily (QD)], or anacetrapib (30 mg QD), only dalcetrapib significantly increased fecal elimination of both [³H]neutral sterols and [³H]bile acids, whereas all compounds increased plasma HDL-[³H]cholesterol. These data suggest that modulation of CETP activity by dalcetrapib does not inhibit CETP-induced pre-ß-HDL formation, which may be required to increase reverse cholesterol transport.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Amides , Animals , Bile Acids and Salts/metabolism , Binding Sites , Biological Transport/drug effects , Cholesterol/blood , Cholesterol Ester Transfer Proteins/blood , Cricetinae , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Esters , High-Density Lipoproteins, Pre-beta/blood , Humans , Oxazolidinones/pharmacology , Quinolines/pharmacology , Sulfhydryl Compounds/pharmacology
18.
Bioorg Med Chem Lett ; 20(1): 236-9, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19932961

ABSTRACT

Reverse cholesterol transport (RCT) is believed to be the primary mechanism by which HDL and its major protein apoA-I protect against atherosclerosis. Starting from the inactive 22-amino acid peptide representing the consensus sequence of the class A amphipathic helical repeats of apoA-I, we designed novel peptides able to mobilize cholesterol from macrophages in vitro, and to stimulate the formation of 'nascent HDL' particles, with potency comparable to the entire apoA-I protein.


Subject(s)
Apolipoprotein A-I/chemistry , Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Peptides/chemistry , Amino Acid Sequence , Animals , Apolipoprotein A-I/metabolism , Cell Line , Circular Dichroism , Macrophages/metabolism , Mice , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/toxicity , Protein Folding , Protein Structure, Secondary
19.
J Lipid Res ; 51(3): 514-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19767535

ABSTRACT

Apolipoprotein M (apoM) is a novel apolipoprotein that is reportedly necessary for pre beta HDL formation; however, its detailed function remains unknown. We investigated the biogenesis and properties of apoM and its effects on the initial steps of nascent pre beta HDL assembly by ABCA1 in HEK293 cells. Transiently transfected apoM was localized primarily in the endomembrane compartment. Pulse-chase analyses demonstrated that apoM is inefficiently secreted, relative to human serum albumin, and that approximately 50% remains membrane-associated after extraction with sodium carbonate, pH 11.5. To investigate the role of apoM in nascent pre beta HDL formation, ABCA1-expressing or control cells, transfected with empty vector, apoM, or C-terminal epitope-tagged apoM (apoM-C-FLAG), were incubated with (125)I-apoA-I for 24 h. Conditioned media were harvested and fractionated by fast-protein liquid chromatography (FPLC) to monitor HDL particle size. Pre beta HDL particles were formed effectively in the absence of apoM expression; however, increased apoM expression stimulated the formation of larger-sized nascent pre beta HDLs. Immunoprecipitation with anti-apoA-I antibody followed by apoM Western blot analysis revealed that little secreted apoM was physically associated with pre beta HDL. Our results suggest that apoM is an atypical secretory protein that is not necessary for ABCA1-dependent pre beta HDL formation but does stimulate the formation of larger-sized pre beta HDL. We propose that apoM may function catalytically at an intracellular site to transfer lipid onto pre beta HDL during or after their formation by ABCA1.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoproteins/metabolism , Gene Expression Regulation , High-Density Lipoproteins, Pre-beta/chemistry , High-Density Lipoproteins, Pre-beta/metabolism , Particle Size , ATP Binding Cassette Transporter 1 , Amino Acid Sequence , Animals , Apolipoproteins/chemistry , Apolipoproteins/genetics , Apolipoproteins/isolation & purification , Apolipoproteins M , Carbonates/chemistry , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Humans , Intracellular Space/metabolism , Lipocalins , Molecular Sequence Data
20.
Biochemistry ; 48(46): 11067-74, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19839639

ABSTRACT

The aim of this study was to correlate the lipid content and size of discoidal reconstituted HDL particles with their ability to promote cellular cholesterol efflux. Homogeneous discoidal rHDL particles containing apoA-I and POPC, with diameters of 7.8, 9.6, 10.8, 12.5, and 17.0 nm, were prepared by the cholate dialysis technique. Cholesterol efflux to rHDL was evaluated in pathway-specific cell models for ABCA1-, ABCG1-, and SR-BI-mediated efflux. ABCA1-mediated efflux was efficiently promoted by the 7.8 nm rHDL containing 82 POPC molecules per particle. This rHDL also promoted ABCG1, but not SR-BI, cholesterol efflux. All large and lipid-rich rHDLs, with a diameter of >or=9.6 nm and a phospholipid content of >/=202 molecules per particle, promoted both SR-BI- and ABCG1-mediated efflux. Our results indicated that the ABCA1-mediated cell cholesterol efflux can be efficiently driven not only by monomolecular lipid free/poor apoA-I but also by a small discoidal phospholipid-containing particle resembling plasma pre-beta1 HDL. This same particle also promotes ABCG1- but not SR-BI-mediated efflux. These results help to clarify the role of plasma pre-beta1 HDL in reverse cholesterol transport.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Alitretinoin , Animals , Apolipoprotein A-I/analysis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclopentanes/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , High-Density Lipoproteins, Pre-beta/chemical synthesis , High-Density Lipoproteins, Pre-beta/chemistry , Humans , Kinetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Particle Size , Phosphatidylcholines/analysis , Phosphatidylcholines/chemistry , Probucol/pharmacology , Rats , Scavenger Receptors, Class B/antagonists & inhibitors , Scavenger Receptors, Class B/metabolism , Tangier Disease/metabolism , Tangier Disease/pathology , Thiosemicarbazones/pharmacology , Transfection , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...