Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
BMB Rep ; 53(12): 664-669, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33298249

ABSTRACT

Breast cancer is one of the most frequently diagnosed cancers. Although biomarkers are continuously being discovered, few specific markers, rather than classification markers, representing the aggressiveness and invasiveness of breast cancer are known. In this study, we used samples from canine mammary tumors in a comparative approach. We subjected 36 fractions of both canine normal and mammary tumor plasmas to highperformance quantitative proteomics analysis. Among the identified proteins, LCAT was selectively expressed in mixed tumor samples. With further MRM and Western blot validation, we discovered that the LCAT protein is an indicator of aggressive mammary tumors, an advanced stage of cancer, possibly highly metastatic. Interestingly, we also found that LCAT is overexpressed in high-grade and lymph-node-positive breast cancer in silico data. We also demonstrated that LCAT is highly expressed in the sera of advanced-stage human breast cancers within the same classification. In conclusion, we identified a possible common plasma protein biomarker, LCAT, that is highly expressed in aggressive human breast cancer and canine mammary tumor. [BMB Reports 2020; 53(12): 664-669].


Subject(s)
Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Animals , Biomarkers, Tumor/genetics , Blood Proteins/analysis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Databases, Genetic , Dogs , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Neoplasm Invasiveness/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Plasma/chemistry , Proteomics/methods
3.
Exp Biol Med (Maywood) ; 241(17): 1933-1942, 2016 11.
Article in English | MEDLINE | ID: mdl-27354333

ABSTRACT

In sickle cell disease (SCD) cholesterol metabolism appears dysfunctional as evidenced by abnormal plasma cholesterol content in a subpopulation of SCD patients. Specific activity of the high density lipoprotein (HDL)-bound lecithin cholesterol acyltransferase (LCAT) enzyme, which catalyzes esterification of cholesterol, and generates lysoPC (LPC) was significantly lower in sickle plasma compared to normal. Inhibitory amounts of LPC were present in sickle plasma, and the red blood cell (RBC) lysophosphatidylcholine acyltransferase (LPCAT), essential for the removal of LPC, displayed a broad range of activity. The functionality of sickle HDL appeared to be altered as evidenced by a decreased HDL-Apolipoprotein A-I exchange in sickle plasma as compared to control. Increased levels of oxidized proteins including ApoA-I were detected in sickle plasma. In vitro incubation of sickle plasma with washed erythrocytes affected the ApoA-I-exchange supporting the view that the RBC blood compartment can affect cholesterol metabolism in plasma. HDL functionality appeared to decrease during acute vaso-occlusive episodes in sickle patients and was associated with an increase of secretory PLA2, a marker for increased inflammation. Simvastatin treatment to improve the anti-inflammatory function of HDL did not ameliorate HDL-ApoA-I exchange in sickle patients. Thus, the cumulative effect of an inflammatory and highly oxidative environment in sickle blood contributes to a decrease in cholesterol esterification and HDL function, related to hypocholesterolemia in SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Apolipoprotein A-I/physiology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Adult , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/physiopathology , Biomarkers/blood , Case-Control Studies , Erythrocytes/chemistry , Fetal Hemoglobin/analysis , Humans , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
4.
J Lipid Res ; 57(5): 882-93, 2016 05.
Article in English | MEDLINE | ID: mdl-27015744

ABSTRACT

Lipoproteins play a key role in the development of CVD, but the dynamics of lipoprotein metabolism are difficult to address experimentally. This article describes a novel two-step combined in vitro and in silico approach that enables the estimation of key reactions in lipoprotein metabolism using just one blood sample. Lipoproteins were isolated by ultracentrifugation from fasting plasma stored at 4°C. Plasma incubated at 37°C is no longer in a steady state, and changes in composition may be determined. From these changes, we estimated rates for reactions like LCAT (56.3 µM/h), ß-LCAT (15.62 µM/h), and cholesteryl ester (CE) transfer protein-mediated flux of CE from HDL to IDL/VLDL (21.5 µM/h) based on data from 15 healthy individuals. In a second step, we estimated LDL's HL activity (3.19 pools/day) and, for the very first time, selective CE efflux from LDL (8.39 µM/h) by relying on the previously derived reaction rates. The estimated metabolic rates were then confirmed in an independent group (n = 10). Although measurement uncertainties do not permit us to estimate parameters in individuals, the novel approach we describe here offers the unique possibility to investigate lipoprotein dynamics in various diseases like atherosclerosis or diabetes.


Subject(s)
Lipoproteins, LDL/blood , Adult , Algorithms , Cholesterol Ester Transfer Proteins/physiology , Computer Simulation , Esterification , Female , Humans , Hydrolysis , Male , Middle Aged , Models, Biological , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Triglycerides/physiology , Young Adult
5.
J Neurochem ; 130(1): 97-108, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24620755

ABSTRACT

Cholesterol is mostly removed from the CNS by its conversion to cerebrosterol (24(S)-hydroxycholesterol, 24(S)OH-C), which is transported to the circulation for bile formation in liver. A neurotoxic role of this oxysterol was previously demonstrated in cell culture. Here, we provide evidence that the enzyme lecithin-cholesterol acyltransferase, long known to esterify cholesterol, also produces monoesters of 24(S)OH-C. Proteoliposomes containing apolipoprotein A-I or apolipoprotein E were used to stimulate the enzyme activity and entrap the formed esters. Proteoliposomes with apolipoprotein A-I were found to be more active than those with apolipoprotein E in stimulating the production of oxysteryl esters. Cholesterol and 24(S)OH-C were found to compete for enzyme activity. High levels of haptoglobin, as those circulating during the acute inflammatory phase, inhibited 24(S)OH-C esterification. When highly neurotoxic 24(S)OH-C was treated with enzyme and proteoliposomes before incubation with differentiated SH-SY5Y cells, the neuron survival improved. The esters of 24(S)OH-C, embedded into proteoliposomes by the enzyme and isolated from unesterified 24(S)OH-C by gel filtration chromatography, did not enter the neurons in culture. These results suggest that the enzyme, in the presence of the apolipoproteins, converts 24(S)OH-C into esters restricted to the extracellular environment, thus preventing or limiting oxysterol-induced neurotoxic injuries to neurons in culture. 24-hydroxycholesterol (24(S)OH-C) is neurotoxic. The enzyme lecithin-cholesterol acyltransferase (LCAT) synthesizes monoesters of 24(S)OH-C in reaction mixtures with proteoliposomes containing phospholipids and apolipoprotein A-I or apolipoprotein E. The esters, also produced by incubation of cerebrospinal fluid only with tritiated 24(S)OH-C, are embedded into lipoproteins that do not enter neurons in culture. The enzyme activity limits the toxicity of 24-hydroxycholesterol in neuron culture.


Subject(s)
Hydroxycholesterols/antagonists & inhibitors , Hydroxycholesterols/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Cell Line, Tumor , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Esterification/drug effects , Esterification/physiology , Humans , Hydroxycholesterols/toxicity , Male , Middle Aged , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism
6.
Panminerva Med ; 54(2): 83-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22525563

ABSTRACT

High-density lipoprotein (HDL) is protective against atherosclerosis development. Other than its central role in reverse cholesterol transport, HDL exhibits several other mechanisms by which it is protective. These include antioxidative, anti-inflammatory and antiapoptopic activities and the normalisation of vascular function. In light of the current view that oxidative modification of low-density lipoprotein (LDL) is essential for the initiation and progression of atherosclerosis, the antioxidative properties of HDL may be an important protective mechanism. HDL can retard the oxidation of LDL and limit its atherogenicity. Several proteins are present on HDL and the evidence that some of them metabolise lipid peroxidation products of phospholipids, cholesteryl esters and triglycerides associated with LDL and vascular cell membranes are discussed in this review.


Subject(s)
Antioxidants/pharmacology , Lipoproteins, HDL/pharmacology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Animals , Apolipoprotein A-I/physiology , Aryldialkylphosphatase/physiology , Atherosclerosis/metabolism , Humans , Lipoproteins, LDL/metabolism , Oxidation-Reduction , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
7.
J Pharmacol Exp Ther ; 340(3): 716-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22171091

ABSTRACT

The acute-phase protein haptoglobin (Hpt) binds apolipoprotein A-I (ApoA-I) and impairs its action on lecithin-cholesterol acyltransferase, an enzyme that plays a key role in reverse cholesterol transport. We have previously shown that an ApoA-I mimetic peptide, P2a, displaces Hpt from ApoA-I, restoring the enzyme activity in vitro. The aim of this study was to evaluate whether P2a displaces Hpt from ApoA-I in vivo and whether this event leads to anti-inflammatory activity. Mice received subplantar injections of carrageenan. Paw volume was measured before the injection and 2, 4, 6, 24, 48, 72, and 96 h thereafter. At the same time points, concentrations of HDL cholesterol (C) and cholesterol esters (CEs) were measured by high-performance liquid chromatography, and Hpt and ApoA-I plasma levels were evaluated by enzyme-linked immunosorbent assay. Western blotting analysis for nitric-oxide synthase and cyclooxygenase (COX) isoforms was also performed on paw homogenates. CEs significantly decreased in carrageenan-treated mice during edema development and negatively correlated with the Hpt/ApoA-I ratio. P2a administration significantly restored the CE/C ratio. In addition, P2a displayed an anti-inflammatory effect on the late phase of edema with a significant reduction in COX2 expression coupled to an inhibition of prostaglandin E(2) synthesis, implying that, in the presence of P2a, CE/C ratio rescue and edema inhibition were strictly related. In conclusion, the P2a effect is due to its binding to Hpt with consequent displacement of ApoA-I that exerts anti-inflammatory activity. Therefore, it is feasible to design drugs that, by enhancing the physiological endogenous protective role of ApoA-I, may be useful in inflammation-based diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apolipoprotein A-I/pharmacology , Cholesterol Esters/metabolism , Peptides/pharmacology , Amino Acid Sequence , Animals , Apolipoprotein A-I/blood , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Edema/metabolism , Esterification , Haptoglobins/metabolism , Male , Mice , Molecular Sequence Data , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
8.
J Clin Endocrinol Metab ; 97(2): E248-56, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090275

ABSTRACT

BACKGROUND: Epidemiologically, high-density lipoprotein (HDL) cholesterol levels associate inversely with risk of ischemic cardiovascular disease. Whether this is a causal relation is unclear. METHODS: We studied 10,281 participants in the Copenhagen City Heart Study (CCHS) and 50,523 participants in the Copenhagen General Population Study (CGPS), of which 991 and 1,693 participants, respectively, had developed myocardial infarction (MI) by August 2010. Participants in the CCHS were genotyped for all six variants identified by resequencing lecithin-cholesterol acyltransferase in 380 individuals. One variant, S208T (rs4986970, allele frequency 4%), associated with HDL cholesterol levels in both the CCHS and the CGPS was used to study causality of HDL cholesterol using instrumental variable analysis. RESULTS: Epidemiologically, in the CCHS, a 13% (0.21 mmol/liter) decrease in plasma HDL cholesterol levels was associated with an 18% increase in risk of MI. S208T associated with a 13% (0.21 mmol/liter) decrease in HDL cholesterol levels but not with increased risk of MI or other ischemic end points. The causal odds ratio for MI for a 50% reduction in plasma HDL cholesterol due to S208T genotype in both studies combined was 0.49 (0.11-2.16), whereas the hazard ratio for MI for a 50% reduction in plasma HDL cholesterol in the CCHS was 2.11 (1.70-2.62) (P(comparison) = 0.03). CONCLUSION: Low plasma HDL cholesterol levels robustly associated with increased risk of MI but genetically decreased HDL cholesterol did not. This may suggest that low HDL cholesterol levels per se do not cause MI.


Subject(s)
Cholesterol, HDL/blood , Mendelian Randomization Analysis , Myocardial Ischemia/blood , Myocardial Ischemia/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/blood , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Cholesterol, HDL/analysis , Cholesterol, HDL/genetics , Cohort Studies , Cross-Sectional Studies , Denmark , Female , Humans , Male , Middle Aged , Myocardial Ischemia/epidemiology , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Polymorphism, Single Nucleotide/physiology , Registries , Young Adult
9.
Atherosclerosis ; 208(1): 155-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19679306

ABSTRACT

Cholesterol is esterified in mammals by two enzymes: LCAT (lecithin cholesterol acyltransferase) in plasma and ACAT(1) and ACAT(2) (acyl-CoA cholesterol acyltransferases) in the tissues. We hypothesized that the sterol structure may have significant effects on the outcome of esterification by these enzymes. To test this hypothesis, we analyzed sterol esters in plasma and tissues in patients having non-cholesterol sterols (sitosterolemia and Smith-Lemli-Opitz syndrome). The esterification of a given sterol was defined as the sterol ester percentage of total sterols. The esterification of cholesterol in plasma by LCAT was 67% and in tissues by ACAT was 64%. Esterification of nine sterols (cholesterol, cholestanol, campesterol, stigmasterol, sitosterol, campestanol, sitostanol, 7-dehydrocholesterol and 8-dehydrocholesterol) was examined. The relative esterification (cholesterol being 1.0) of these sterols by the plasma LCAT was 1.00, 0.95, 0.89, 0.40, 0.85, 0.82 and 0.80, 0.69 and 0.82, respectively. The esterification by the tissue ACAT was 1.00, 1.29, 0.75, 0.49, 0.45, 1.21 and 0.74, respectively. The predominant fatty acid of the sterol esters was linoleic acid for LCAT and oleic acid for ACAT. We compared the esterification of two sterols differing by only one functional group (a chemical group attached to sterol nucleus) and were able to quantify the effects of individual functional groups on sterol esterification. The saturation of the A ring of cholesterol increased ester formation by ACAT by 29% and decreased the esterification by LCAT by 5.9%. Esterification by ACAT and LCAT was reduced, respectively, by 25 and 11% by the presence of an additional methyl group on the side chain of cholesterol at the C-24 position. This data supports our hypothesis that the structure of the sterol substrate has a significant effect on its esterification by ACAT or LCAT.


Subject(s)
Lipid Metabolism Disorders/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Sitosterols/metabolism , Smith-Lemli-Opitz Syndrome/metabolism , Sterol O-Acyltransferase/physiology , Sterols/metabolism , Adolescent , Adult , Child , Child, Preschool , Esterification , Female , Humans , Infant , Infant, Newborn , Male , Young Adult , Sterol O-Acyltransferase 2
10.
MMW Fortschr Med ; 152 Suppl 2: 47-55, 2010 Jul 01.
Article in German | MEDLINE | ID: mdl-21591319

ABSTRACT

Large epidemiological studies have consistently shown that plasma levels of high-density lipoprotein (HDL) correlate inversely with cardiovascular risk. The apparent cardioprotective role of HDL has primarily been attributed to its participation in reverse cholesterol transport (RCT) but there is also substantial evidence that supports the concept of HDL and apoA-I preventing oxidative damage, inhibiting systemic inflammation, promoting vascular integrity and preventing thrombosis. Besides conventional therapy to increase HDL like physical exercise, weight loss and dietary changes new strategies to intervene at various steps of its metabolism have been proposed and are in development. One of the most promising approaches is inhibiting cholesteryl ester transfer protein (CETP)which plays a central role in RCT by transferring cholesteryl esters from HDL to apoB containing lipoproteins in exchange for triglycerides. The failure of the CETP inhibitor torcetrapib, however, to cause any benefit on cardiovascular outcomes despite significantly increased HDL levels in several clinical trials casted doubts upon the concept of CETP inhibition. Meanwhile, off target toxicity could be shown for torcetrapib and a new generation of CETP inhibitors stands ready to be tested in large clinical trials. This article describes the formation and remodeling of HDL, how HDL is thought to be beneficial for the vasculature and what options we have today to increase HDL levels with a special focus on CETP inhibition.


Subject(s)
Cardiovascular Diseases/physiopathology , Cholesterol Ester Transfer Proteins/physiology , Lipoproteins, HDL/blood , Anticholesteremic Agents/pharmacology , Apolipoprotein A-I/blood , Cardiovascular Diseases/prevention & control , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol, HDL/blood , Diet, Fat-Restricted , Exercise/physiology , Humans , Lipase/physiology , Liver/physiopathology , Oxidative Stress/physiology , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Phospholipid Transfer Proteins/physiology , Quinolines/pharmacology , Triglycerides/blood
11.
Circulation ; 120(7): 628-35, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19687369

ABSTRACT

BACKGROUND: Mutations in the LCAT gene cause lecithin:cholesterol acyltransferase (LCAT) deficiency, a very rare metabolic disorder with 2 hypoalphalipoproteinemia syndromes: classic familial LCAT deficiency (Online Mendelian Inheritance in Man No. 245900), characterized by complete lack of enzyme activity, and fish-eye disease (Online Mendelian Inheritance in Man No. 136120), with a partially defective enzyme. Theoretically, hypoalphalipoproteinemia cases with LCAT deficiency should be at increased cardiovascular risk because of high-density lipoprotein deficiency and defective reverse cholesterol transport. METHODS AND RESULTS: The extent of preclinical atherosclerosis was assessed in 40 carriers of LCAT gene mutations from 13 Italian families and 80 healthy controls by measuring carotid intima-media thickness (IMT). The average and maximum IMT values in the carriers were 0.07 and 0.21 mm smaller than in controls (P=0.0003 and P=0.0027), respectively. Moreover, the inheritance of a mutated LCAT genotype had a remarkable gene-dose-dependent effect in reducing carotid IMT (P=0.0003 for average IMT; P=0.001 for maximum IMT). Finally, no significant difference in carotid IMT was found between carriers of LCAT gene mutations that cause total or partial LCAT deficiency (ie, familial LCAT deficiency or fish-eye disease). CONCLUSIONS: Genetically determined low LCAT activity in Italian families is not associated with enhanced preclinical atherosclerosis despite low high-density lipoprotein cholesterol levels. This finding challenges the notion that LCAT is required for effective atheroprotection and suggests that elevating LCAT expression or activity is not a promising therapeutic strategy to reduce cardiovascular risk.


Subject(s)
Atherosclerosis/prevention & control , Atherosclerosis/physiopathology , Mutation/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Adult , Alleles , Atherosclerosis/epidemiology , Carotid Arteries/diagnostic imaging , Case-Control Studies , Cholesterol, HDL/blood , Diagnosis, Differential , Female , Humans , Italy , Lecithin Cholesterol Acyltransferase Deficiency/diagnosis , Lecithin Cholesterol Acyltransferase Deficiency/ethnology , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Male , Middle Aged , Risk Factors , Tunica Intima/diagnostic imaging , Tunica Media/diagnostic imaging , Ultrasonography
12.
BMB Rep ; 42(7): 393-400, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19643035

ABSTRACT

High-density lipoprotein (HDL) is a proven biomarker for the monitoring of changes in antioxidant and anti-inflammation capability of body fluids. The beneficial virtues of HDL are highly dependent on its lipids and protein compositions, and their ratios. In normal state, the HDL particle is enriched with lipids and several HDL-associated enzymes, which are responsible for its antioxidant activity. Lower HDL-cholesterol levels (40 mg/dL) have been recognized as an independent risk factor for coronary artery disease, as well as being a known component of metabolic syndrome. Functional and structural changes of HDL have been recognized as factors pivotal to the evaluation of HDL-quality. In this review, I have elected to focus on the functional and structural correlations of HDL and the roles of HDL-associated apolipoproteins and enzymes. Recent clinical applications of HDL have also been reviewed, particularly the therapeutic targeting of HDL metabolism and reconstituted HDL; these techniques represent promising emerging strategies for the treatment of cardiovascular disease, for drug or gene therapy.


Subject(s)
Lipoproteins, HDL/chemistry , Lipoproteins, HDL/physiology , Lipoproteins, HDL/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Apolipoproteins/metabolism , Apolipoproteins/physiology , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/physiology , Atherosclerosis/prevention & control , Biomedical Technology/methods , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Ester Transfer Proteins/physiology , Drug Delivery Systems/methods , Humans , Lipoproteins, HDL/metabolism , Models, Biological , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
14.
Atherosclerosis ; 206(2): 321-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19362310

ABSTRACT

The beneficial therapeutic effects of raising HDL cholesterol are proving difficult to confirm in humans. The evaluation of antiatherogenic functions of HDL is an important area of research which includes the role of HDL in reverse cholesterol transport (RCT), especially macrophage-specific RCT, and its antioxidant and antiinflammatory roles. The antioxidant and antiinflammatory functions of HDL can be assessed using cell-free and cell-based assays. Also, a new approach was developed to measure RCT from labeled-cholesterol macrophages to liver and feces of mice. Studies in genetically engineered animals indicate that these major HDL antiatherogenic functions are better predictors of atherosclerosis susceptibility than HDL cholesterol or total RCT. Thus, functional testing of the antiatherogenic functions of HDL in experimental animal models may facilitate the development of new strategies for the prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cholesterol, HDL/metabolism , Macrophages/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/physiology , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Antioxidants/metabolism , Antioxidants/therapeutic use , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/therapeutic use , Apolipoprotein A-II/metabolism , Aryldialkylphosphatase/physiology , CD36 Antigens/metabolism , Cholesterol Ester Transfer Proteins/physiology , Humans , Lipase/physiology , Lipoproteins/metabolism , Lipoproteins/physiology , Liver X Receptors , Mice , Mice, Transgenic , Orphan Nuclear Receptors/agonists , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Phospholipid Transfer Proteins/physiology
15.
Gene Ther ; 16(6): 757-65, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19242527

ABSTRACT

Plasma levels of high-density lipoprotein (HDL) cholesterol and its major apolipoprotein (apo), apo A-I, are inversely correlated with the incidence of ischemic cardiovascular diseases. Reverse cholesterol transport is likely the main mechanism underlying the atheroprotective effects of HDL. Here, we investigated whether increased HDL cholesterol following hepatocyte-directed adenoviral rabbit apo A-I (AdrA-I) or rabbit lecithin-cholesterol acyltransferase (LCAT) (AdrLCAT) transfer may induce cholesterol unloading in complex atherosclerotic lesions in heterozygous low-density lipoprotein receptor-deficient rabbits fed a 0.15% cholesterol diet for 420 days before and for 120 days after transfer. HDL cholesterol levels increased 2.0-fold (P<0.001) and 1.9-fold (P<0.001) in the 120 days after transfer with AdrA-I and AdrLCAT, respectively, compared to levels just before transfer whereas non-HDL cholesterol remained unchanged. Increased HDL cholesterol following AdrA-I and AdrLCAT transfer resulted in a 31% (P<0.05) reduction of the intima/media ratio in comparison with the control progression group. Compared to the baseline group killed after 420 days of cholesterol diet, AdrA-I and AdrLCAT transfer reduced the percentage of Oil Red O area 1.6-fold (P<0.001) and 1.4-fold (P<0.001), respectively. In conclusion, increased HDL cholesterol after AdrA-I and AdrLCAT transfer inhibits progression of atherosclerosis and induces cholesterol unloading in complex lesions in rabbits.


Subject(s)
Apolipoprotein A-I/genetics , Atherosclerosis/metabolism , Cholesterol, HDL/metabolism , Gene Transfer Techniques , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Adenoviridae/genetics , Animals , Aorta/metabolism , Apolipoprotein A-I/metabolism , Atherosclerosis/pathology , Azo Compounds , Biological Transport/physiology , Cholesterol, HDL/blood , Coloring Agents , Dietary Fats/administration & dosage , Disease Models, Animal , Disease Progression , Genetic Therapy/methods , Genetic Vectors , Lecithin Cholesterol Acyltransferase Deficiency , Liver/metabolism , Particle Size , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Rabbits , Time Factors , Tunica Intima/pathology
16.
Free Radic Biol Med ; 43(11): 1484-93, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17964419

ABSTRACT

Lecithin:cholesterol acyltransferase (LCAT) is a key enzyme involved in lipoprotein metabolism. It mediates the transesterification of free cholesterol to cholesteryl ester in an apoprotein A-I-dependent process. We have isolated purified LCAT from human plasma using anion-exchange chromatography and characterized the extracted LCAT in terms of its molecular weight, molar absorption coefficient, and enzymatic activity. The participation of LCAT in the oxidation of very low density lipoproteins (VLDL) and low-density lipoproteins (LDL) was examined by supplementing lipoproteins with exogenous LCAT over a range of protein concentrations. LCAT-depleted lipoproteins were also prepared and their oxidation kinetics examined. Our results provide evidence for a dual role for LCAT in lipoprotein oxidation, whereby it acts in a dose-responsive manner as a potent pro-oxidant during VLDL oxidation, but as an antioxidant during LDL oxidation. We believe this novel pro-oxidant effect may be attributable to the LCAT-mediated formation of oxidized cholesteryl ester in VLDL, whereas the antioxidant effect is similar to that of chain-breaking antioxidants. Thus, we have demonstrated that the high-density lipoprotein-associated enzyme LCAT may have a significant role to play in lipoprotein modification and hence atherogenesis.


Subject(s)
Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Humans , Lipoproteins, HDL/metabolism , Oxidation-Reduction , Phenylmethylsulfonyl Fluoride/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Phosphatidylcholine-Sterol O-Acyltransferase/isolation & purification
18.
J Lipid Res ; 48(3): 592-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17183024

ABSTRACT

To better understand the role of LCAT in HDL metabolism, we compared HDL subpopulations in subjects with homozygous (n = 11) and heterozygous (n = 11) LCAT deficiency with controls (n = 22). Distribution and concentrations of apolipoprotein A-I (apoA-I)-, apoA-II-, apoA-IV-, apoC-I-, apoC-III-, and apoE-containing HDL subpopulations were assessed. Compared with controls, homozygotes and heterozygotes had lower LCAT masses (-77% and -13%), and LCAT activities (-99% and -39%), respectively. In homozygotes, the majority of apoA-I was found in small, disc-shaped, poorly lipidated prebeta-1 and alpha-4 HDL particles, and some apoA-I was found in larger, lipid-poor, discoidal HDL particles with alpha-mobility. No apoC-I-containing HDL was noted, and all apoA-II and apoC-III was detected in lipid-poor, prebeta-mobility particles. ApoE-containing particles were more disperse than normal. ApoA-IV-containing particles were normal. Heterozygotes had profiles similar to controls, except that apoC-III was found only in small HDL with prebeta-mobility. Our data are consistent with the concepts that LCAT activity: 1) is essential for developing large, spherical, apoA-I-containing HDL and for the formation of normal-sized apoC-I and apoC-III HDL; and 2) has little affect on the conversion of prebeta-1 into alpha-4 HDL, only slight effects on apoE HDL, and no effect on apoA-IV HDL particles.


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency/blood , Lipoproteins, HDL/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Analysis of Variance , Apolipoprotein A-I/blood , Apolipoprotein A-II/blood , Apolipoprotein C-I/blood , Apolipoprotein C-III/blood , Apolipoproteins A/blood , Apolipoproteins E/blood , Female , Heterozygote , Homozygote , Humans , Lipoproteins, HDL/blood , Male , Models, Biological , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
19.
J Mol Med (Berl) ; 84(4): 276-94, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16501936

ABSTRACT

The concentration, composition, shape, and size of plasma high-density lipoprotein (HDL) are determined by numerous proteins that influence its biogenesis, remodeling, and catabolism. The discoveries of the HDL receptor (scavenger receptor class B type I, SR-BI) and the ABCA1 (ATP-binding cassette transporter A1) lipid transporter provided two missing links that were necessary to understand the biogenesis and some of the functions of HDL. Existing data indicate that functional interactions between apoA-I and ABCA1 are necessary for the initial lipidation of apoA-I. Through a series of intermediate steps, lipidated apoA-I proceeds to form discoidal HDL particles that can be converted to spherical particles by the action of lecithin:cholesterol acyltransferase (LCAT). Discoidal and spherical HDL can interact functionally with SR-BI and these interactions lead to selective lipid uptake and net efflux of cholesterol and thus remodel HDL. Defective apoA-I/ABCA1 interactions prevent lipidation of apoA-I that is necessary for the formation of HDL particles. In the same way, specific mutations in apoA-I or LCAT prevent the conversion of discoidal to spherical HDL particles. The interactions of lipid-bound apoA-I with SR-BI are affected in vitro by specific mutations in apoA-I or SR-BI. Furthermore, deficiency of SR-BI affects the lipid and apolipoprotein composition of HDL and is associated with increased susceptibility to atherosclerosis. Here we review the current status of the pathway of HDL biogenesis and mutations in apoA-I, ABCA1, and SR-BI that disrupt different steps of the pathway and may lead to dyslipidemia and atherosclerosis in mouse models. The phenotypes generated in experimental mouse models for apoA-I, ABCA1, LCAT, SR-BI, and other proteins of the HDL pathway may facilitate early diagnosis of similar phenotypes in the human population and provide guidance for proper treatment.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Apolipoprotein A-I/physiology , CD36 Antigens/physiology , Cholesterol, HDL/biosynthesis , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/metabolism , Atherosclerosis/prevention & control , Biological Transport , Dyslipidemias/genetics , Humans , Lipid Metabolism/physiology , Lipoproteins/metabolism , Models, Biological , Models, Molecular , Mutation/physiology , Protein Binding , Signal Transduction
20.
Atherosclerosis ; 187(1): 101-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16216249

ABSTRACT

The esterification of free cholesterol (FC) in plasma, catalyzed by the enzyme lecithin:cholesterol acyltransferase (LCAT; EC 2.3.1.43), is a key process in lipoprotein metabolism. The resulting cholesteryl esters (CE) represent the main core lipids of low (LDL) and high density lipoproteins (HDL). Primary (familial) LCAT-deficiency (FLD) is a rare autosomal recessive genetic disease caused by the complete or near absence of LCAT activity. In fish-eye disease (FED), residual LCAT activity is still detectable. Here, we describe a 32-year-old patient with corneal opacity, very low LCAT activity, reduced amounts of CE (low HDL-cholesterol level), and elevated triglyceride (TG) values. The lipoprotein pattern was abnormal with regard to lipoprotein composition and concentration, but distinct lipoprotein classes were still present. Despite of typical features of glomerular proteinuria, creatinine clearance was normal. DNA sequencing and restiction fragment analyses revealed two separate mutations in the patient's LCAT gene: a previously described G to A transition in exon 4 converting Arg140 to His, inherited from his mother, and a novel G to C transversion in exon 2 converting Gly71 to Arg, inherited from his father, indicating that M.P. was a compound heterozygote. Determination of enzyme activities of recombinant LCAT proteins obtained upon transfection of COS-7 cells with plasmids containing G71R-LCAT or wild-type LCAT cDNA revealed very low alpha- and absence of beta-LCAT activity for the G71R mutant. The identification of the novel G71R LCAT mutation supports the proposed molecular model for the enzyme implying that the "lid" domain at residues 50-74 is involved in enzyme:substrate interaction. Our data are in line with the hypothesis that a key event in the etiology of FLD is the loss of distinct lipoprotein fractions.


Subject(s)
Heterozygote , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/physiology , Adult , Animals , COS Cells , Chlorocebus aethiops , Cholesterol/metabolism , DNA, Complementary/metabolism , Female , Gene Expression Regulation , Humans , Kidney/metabolism , Lipoproteins/chemistry , Male , Phenotype , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...