Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Atheroscler Thromb ; 28(7): 679-691, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33867422

ABSTRACT

Lecithin cholesterol acyltransferase (LCAT) is a lipid-modification enzyme that catalyzes the transfer of the acyl chain from the second position of lecithin to the hydroxyl group of cholesterol (FC) on plasma lipoproteins to form cholesteryl acylester and lysolecithin. Familial LCAT deficiency is an intractable autosomal recessive disorder caused by inherited dysfunction of the LCAT enzyme. The disease appears in two different phenotypes depending on the position of the gene mutation: familial LCAT deficiency (FLD, OMIM 245900) that lacks esterification activity on both HDL and ApoB-containing lipoproteins, and fish-eye disease (FED, OMIM 136120) that lacks activity only on HDL. Impaired metabolism of cholesterol and phospholipids due to LCAT dysfunction results in abnormal concentrations, composition and morphology of plasma lipoproteins and further causes ectopic lipid accumulation and/or abnormal lipid composition in certain tissues/cells, and serious dysfunction and complications in certain organs. Marked reduction of plasma HDL-cholesterol (HDL-C) and corneal opacity are common clinical manifestations of FLD and FED. FLD is also accompanied by anemia, proteinuria and progressive renal failure that eventually requires hemodialysis. Replacement therapy with the LCAT enzyme should prevent progression of serious complications, particularly renal dysfunction and corneal opacity. A clinical research project aiming at gene/cell therapy is currently underway.


Subject(s)
Enzyme Replacement Therapy/methods , Lecithin Cholesterol Acyltransferase Deficiency , Lipoproteins , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Corneal Opacity/etiology , Corneal Opacity/prevention & control , Humans , Japan/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/blood , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/physiopathology , Lecithin Cholesterol Acyltransferase Deficiency/therapy , Lipoproteins/blood , Lipoproteins/metabolism , Mutation , Phosphatidylcholine-Sterol O-Acyltransferase/pharmacology , Phospholipids/blood , Phospholipids/metabolism , Renal Insufficiency/etiology , Renal Insufficiency/prevention & control
2.
Lipids Health Dis ; 18(1): 132, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31164121

ABSTRACT

BACKGROUND: Lecithin-cholesterol acyltransferase (LCAT) is a plasma enzyme that esterifies cholesterol in high- and low-density lipoproteins (HDL and LDL). Mutations in LCAT gene causes familial LCAT deficiency, which is characterized by very low plasma HDL-cholesterol levels (Hypoalphalipoproteinemia), corneal opacity and anemia, among other lipid-related traits. Our aim is to evaluate clinical/biochemical features of a Chilean family with a proband showing clinical signs of familial LCAT deficiency, as well as to identify and assess the functional effects of LCAT mutations. METHODS: An adult female proband with hypoalphalipoproteinemia, corneal opacity and mild anemia, as well as her first-degree relatives, were recruited for clinical, biochemical, genetic, in-silico and in-vitro LCAT analysis. Sequencing of exons and intron-exon boundaries was performed to identify mutations. Site-directed mutagenesis was carried out to generate plasmids containing cDNA with wild type or mutant sequences. Such expression vectors were transfected to HEK-239 T cells to asses the effect of LCAT variants in expression, synthesis, secretion and enzyme activity. In-silico prediction analysis and molecular modeling was also used to evaluate the effect of LCAT variants. RESULTS: LCAT sequencing identified rare p.V333 M and p.M404 V missense mutations in compound heterozygous state in the proband, as well the common synonymous p.L363 L variant. LCAT protein was detected in proband's plasma, but with undetectable enzyme activity compared to control relatives. HEK-293 T transfected cells with vector expression plasmids containing either p.M404 V or p.V333 M cDNA showed detectable LCAT protein expression both in supernatants and lysates from cultured cells, but with much lower enzyme activity compared to cells transfected with the wild-type sequence. Bioinformatic analyses also supported a causal role of such rare variations in LCAT lack of function. Additionally, the proband carried the minor allele of the synonymous p.L363 L variant. However, this variant is unlikely to affect the clinical phenotype of the proband given its relatively high frequency in the Chilean population (4%) and its small putative effect on plasma HDL-cholesterol levels. CONCLUSION: Genetic, biochemical, in vitro and in silico analyses indicate that the rare mutations p.M404 V and p.V333 M in LCAT gene lead to suppression of LCAT enzyme activity and cause clinical features of familial LCAT deficiency.


Subject(s)
Hypoalphalipoproteinemias/genetics , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lipids/blood , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Adult , Aged , Chile/epidemiology , Cholesterol/blood , Cholesterol, HDL/blood , Corneal Opacity/genetics , Corneal Opacity/pathology , Exons/genetics , Female , HEK293 Cells , Humans , Hypoalphalipoproteinemias/blood , Hypoalphalipoproteinemias/epidemiology , Hypoalphalipoproteinemias/pathology , Lecithin Cholesterol Acyltransferase Deficiency/blood , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Lipoproteins, HDL/blood , Molecular Dynamics Simulation , Mutation, Missense/genetics , Pedigree , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Structure-Activity Relationship
3.
Nutr Metab Cardiovasc Dis ; 29(1): 4-8, 2019 01.
Article in English | MEDLINE | ID: mdl-30503707

ABSTRACT

AIMS: To review the formation, catabolism, and the possible atherogenic properties of Lp-X. DATA SYNTHESIS: The conversion of cholesterol to bile acids is regulated by several mechanisms including cholesterol 7 alpha hydroxylase, fibroblast growth factor 19, and farnesoid X receptors. During cholestasis these mechanisms are altered and there is an accumulation of bile acids and cholesterol in plasma. The hypercholesterolemia observed in cholestasis is due to the presence of an anomalous lipoprotein called lipoprotein-X (Lp-X). Lp-X is a lipoprotein rich in phospholipid and free cholesterol present in plasma of patients with cholestasis and, with some variations, in patients with lecithin-cholesterol-acyl-transferase deficiency (LCAT), and after lipid infusion. Lp-X is formed from a bile lipoprotein moving to the blood vessels where it incorporates small quantities of triglycerides, apo-C and esterified cholesterol and becomes a "mature" Lp-X. The activity of the phosphatidilcholine canalicular transporter Mdr2 P-glycoprotein (homologous to the human ABCB4) is essential for LpX appearance, since its suppression abolishes Lp-X formation. However, the concentration of Lp-X in plasma is determined also by the degree of the cholestasis, the residual liver function, and the LCAT deficiency. The Lp-X catabolism seems to be mediated by the reticuloendothelial system and possibly the kidney. CONCLUSIONS: Lp-X might be considered a defense mechanism against the toxic effect of free cholesterol in cholestasis. The frequency of cardiovascular events in patients affected by primary biliary cholangitis, in whom the Lp-X is present in high concentration, are not increased. Further studies could now clarify the remaining open questions on the role of Lp-X in the dyslipidemia of cholestasis.


Subject(s)
Cholestasis/blood , Hypercholesterolemia/blood , Lipoprotein-X/blood , Liver/metabolism , Animals , Biological Transport , Cholestasis/epidemiology , Cholestasis/history , History, 20th Century , History, 21st Century , Humans , Hypercholesterolemia/epidemiology , Hypercholesterolemia/history , Lecithin Cholesterol Acyltransferase Deficiency/blood , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lipoprotein-X/history , Prognosis , Risk Factors
4.
Hum Mutat ; 32(11): 1290-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21901787

ABSTRACT

Lecithin:cholesterol acyltransferase (LCAT) is crucial to the maturation of high-density lipoprotein (HDL). Homozygosity for LCAT mutations underlies rare disorders characterized by HDL-cholesterol (HDL-c) deficiency while heterozygotes have half normal HDL-c levels. We studied the prevalence of LCAT mutations in referred patients with low HDL-c to better understand the molecular basis of low HDL-c in our patients. LCAT was sequenced in 98 patients referred for HDL-c <5th percentile and in four patients referred for low HDL-c and corneal opacities. LCAT mutations were highly prevalent: in 28 of the 98 participants (29%), heterozygosity for nonsynonymous mutations was identified while 18 patients carried the same mutation (p.T147I). The four patients with corneal opacity were compound heterozygotes. All previously identified mutations are documented to cause loss of catalytic activity. Nine novel mutations-c.402G>T (p.E134D), c.403T>A (p.Y135N), c.964C>T (p.R322C), c.296G>C (p.W99S), c.736G>T (p.V246F), c.802C>T (p.R268C), c.945G>A (p.W315X), c.1012C>T (p.L338F), and c.1039C>T (p.R347C)--were shown to be functional through in vitro characterization. The effect of several mutations on the core protein structure was studied by a three-dimensional (3D) model. Unlike previous reports, functional mutations in LCAT were found in 29% of patients with low HDL-c, thus constituting a common cause of low HDL-c in referred patients in The Netherlands.


Subject(s)
Cholesterol, HDL/genetics , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Mutation , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Adult , Aged , Animals , COS Cells , Child, Preschool , Chlorocebus aethiops , Cholesterol, HDL/blood , Corneal Opacity/genetics , Female , Genetic Variation , Heterozygote , Humans , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Male , Middle Aged , Netherlands/epidemiology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Prevalence
5.
G Ital Nefrol ; 28(4): 369-82, 2011.
Article in Italian | MEDLINE | ID: mdl-21809306

ABSTRACT

A genetic mendelian autosomal recessive condition of deficiency of lecithin- cholesterol acyltransferase (LCAT) can produce two different diseases: one highly interesting nephrologic picture of complete enzymatic deficiency (lecithin:cholesterol acyltransferase deficiency; OMIM ID #245900; FLD), characterized by the association of dyslipidemia, corneal opacities, anemia and progressive nephropathy; and a partial form (fish eye disease; OMIM ID #136120; FED) with dyslipidemia and progressive corneal opacities only. The diagnosis of FLD falls first of all under the competence of nephrologists, because end-stage renal disease appears to be its most severe outcome. The diagnostic suspicion is based on clinical signs (corneal opacities, more severe anemia than expected for the degree of chronic renal failure, progressive proteinuric nephropathy) combined with histology obtained by kidney biopsy (glomerulopathy evolving toward sclerosis with distinctive lipid deposition). However, the final diagnosis, starting with a finding of extremely low levels of HDL-cholesterol, requires collaboration with lipidology Centers that can perform sophisticated investigations unavailable in common laboratories. To be heterozygous for a mutation of the LCAT gene is one of the monogenic conditions underlying primary hypoalphalipoproteinemia (OMIM ID #604091). This disease, which is characterized by levels of HDL-cholesterol below the 5th percentile of those of the examined population (<28 mg/dL for Italians), has heritability estimates between 40% and 60% and is considered to be a predisposing condition for coronary artery disease. Nevertheless, some monogenic forms, and especially those associated with LCAT deficiency, seem to break the rule, confirming once more the value of a proper diagnosis before drawing prognostic conclusions from a laboratory marker. As in many other rare illnesses, trying to discover all the existing cases will contribute to allow studies broad enough to pave the way for further therapies, in this case also fostering the production by industries of the lacking enzyme by genetic engineering. Epidemiological studies, although done on selected populations such as hypoalphalipoproteinemia patients on dialysis and with the effective genetic tools of today, have been disappointing in elucidating the disease. Spreading the clinical knowledge of the disease and its diagnostic course among nephrologists seems to be the best choice, and this is the aim of our work.


Subject(s)
Cholesterol, HDL/blood , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Lecithin Cholesterol Acyltransferase Deficiency/diagnosis , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Anemia/etiology , Biomarkers/blood , Biopsy , Cholesterol, HDL/metabolism , Corneal Opacity/etiology , Coronary Artery Disease/prevention & control , Disease Progression , Dyslipidemias/etiology , Genetic Engineering , Heterozygote , Humans , Italy/epidemiology , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Diseases/therapy , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Lecithin Cholesterol Acyltransferase Deficiency/complications , Lecithin Cholesterol Acyltransferase Deficiency/enzymology , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/therapy , Mutation , Proteinuria/etiology , Risk Factors , Treatment Outcome
6.
J Clin Invest ; 94(6): 2330-40, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7989589

ABSTRACT

The composition of lipoproteins in the plasma of patients with LCAT deficiency (LCAT-D) is grossly altered due to the lack of cholesteryl esters which form the core of normal lipoproteins. When plasma from LCAT-D patients and their relatives was examined we found that nine heterozygotes had plasma Lp(a) levels of 2-13 mg/dl whereas none of 11 affected homozygous individuals from different families contained detectable amounts of Lp(a) in their plasma. Therefore, the binding of apo(a) to LDL density particles was studied in vitro using LDL density fractions prepared from patients, and recombinant apo(a) [r-apo(a)], which was expressed and secreted by transfected COS-7 cells. The LDL from heterozygotes were chemically indistinguishable from normal LDL and homogeneous with regard to morphology, whereas the crude LDL floating fraction from homozygotes consisted of a heterogeneous mixture of large vesicles, and small spheres resembling normal LDL. The LDL density fraction from the LCAT-D patient lacked almost completely cholesteryl esters. Incubation of LCAT-D plasma with active LCAT caused a substantial augmentation of the original subfraction which morphologically resembled normal LDL. Using r-apo(a) and normal LDL or LDL of heterozygous individuals, apoB:r-apo(a) complexes were formed when incubated at 37 degrees C in vitro for 20 h. In contrast, the total LDL floating fraction from a homozygous LCAT-D patient failed to form apoB:r-apo(a) complexes. After treatment with active LCAT, a significant apoB:r-apo(a) association was observed with LCAT-D LDL-density particles. Our data emphasize the importance of the integrity of LDL structure and composition for the formation of Lp(a). In addition, we demonstrate that the absence of LCAT activity has a fundamental impact on the regulation of plasma Lp(a) levels.


Subject(s)
Apolipoproteins A/biosynthesis , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lipoproteins, LDL/biosynthesis , Apolipoproteins A/blood , Apolipoproteins A/metabolism , Apolipoproteins B/metabolism , Austria/epidemiology , Female , Homozygote , Humans , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/ultrastructure , Male , Pedigree , Protein Binding
7.
Intern Med ; 33(11): 677-82, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7849380

ABSTRACT

We present findings in the ninth known Japanese family with lecithin:cholesterol acyltransferase (LCAT) deficiency. A 54-year-old man (proband) and his 58-year-old brother presented with corneal opacity. Both subjects showed a marked decrease in serum high density lipoprotein (HDL)-cholesterol and in the cholesteryl ester ratio. Although apo A-I and A-II were low, apo E tended to be high. Serum LCAT activity and mass were not detectable. Urinary examination showed microhematuria or proteinuria. Renal function was normal and no anemia was demonstrated, but blood smears showed poikilocytosis with target cells. The serum LCAT activity of the proband's three sons, obligate heterozygotes of LCAT deficiency, was about one-half the normal level, and HDL-cholesterol and apo A-I levels were low normal.


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency/genetics , Adult , Cholesterol, HDL/blood , Corneal Opacity/genetics , Female , Heterozygote , Homozygote , Humans , Japan/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Lipoproteins/blood , Male , Middle Aged , Pedigree , Phosphatidylcholine-Sterol O-Acyltransferase/blood
8.
Arteriosclerosis ; 9(1 Suppl): I164-8, 1989.
Article in English | MEDLINE | ID: mdl-2463828

ABSTRACT

Some 50 years ago, Müller described hypercholesterolemia, xanthomas, and coronary heart disease as symptoms of a genetic disorder. In the 1930s, other important discoveries concerning inborn errors of metabolism were made in Norway. Følling described phenylketonuria, and Refsum examined his first patients with heredopathia atactica polyneuritiformis (phytanic acid storage disease). Several other inborn errors of metabolism have been discovered in Norway: familial lecithin-cholesterol acyltransferase deficiency, methylmalonic acidemia, beta-methylcrotonyl-coenzyme A carboxylase deficiency, pyroglutamic aciduria, and N-acetyl aspartic aciduria. Metabolic and biochemical studies in these patients have revealed new and important metabolic pathways. Studies on patients with inborn errors not first described in Norway have also given important information on key enzymes in metabolic pathways. Thus, studies on patients with cerebrotendinous xanthomatosis and those with Zellweger's syndrome have revealed the normal metabolic route for conversion of cholesterol to bile acids. The discoveries and the clinical and biochemical research in former days were mostly good examples of serendipity combined with excellent clinical alertness. In more recent years, several of the discoveries have resulted from systematic biochemical screening of urine, plasma, or other body fluids from patients with unusual clinical syndromes.


Subject(s)
Metabolism, Inborn Errors/epidemiology , Cholesterol/metabolism , Humans , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Norway , Phenylketonurias/epidemiology , Refsum Disease/epidemiology
9.
Atherosclerosis ; 43(2-3): 369-79, 1982 Jun.
Article in English | MEDLINE | ID: mdl-7115467

ABSTRACT

Plasma unesterified cholesterol is converted to cholesteryl ester by the enzyme lecithin-cholesterol acyltransferase (LCAT). Plasma levels of LCAT were measured by a sensitive double antibody radioimmunoassay in a sample from an adult employee population, ages 20-59 years, in the Pacific Northwest. After adjusting for differences in relative body mass, women had significantly higher LCAT levels (5.90 +/- 1.06, n = 154) than men (5.49 +/- 0.89, n = 83). For ages 20-59 years, LCAT levels showed a slight association with age: r = 0.13 for men and 0.29 for women. LCAT was positively correlated with relative body mass, total cholesterol, and LDL cholesterol. Men who smoked cigarettes had significantly lower LCAT mass than men who did not smoke cigarettes. No statistical differences in mean LCAT values were found between drinkers and nondrinkers. The 5th percentile LCAT value was 4.3 micrograms/ml for both men and women not using hormones. The 95th percentile value was 7.3 micrograms/ml for men and 7.8 micrograms/ml for women regardless of hormone use. Subjects phenotypically LCAT-deficient by clinical criteria and by the absence or near absence of LCAT activity had levels of LCAT mass well below the reference values: 0.73 +/- 0.70, range 0.10 micrograms/ml to 2.65 micrograms/ml, n = 20. Parents or children of LCAT-deficient subjects, i.e., obligate heterozygotes for familial LCAT deficiency, had reduced levels: 3.59 +/- 0.69, range 2.59-4.61 micrograms/ml, n = 19.


Subject(s)
Hypolipoproteinemias/epidemiology , Lecithin Cholesterol Acyltransferase Deficiency/epidemiology , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Population , Adult , Aged , Aging , Alcohol Drinking , Body Weight , Cholesterol/blood , Contraceptives, Oral/pharmacology , Estrogens/pharmacology , Female , Health Surveys , Humans , Japan , Lecithin Cholesterol Acyltransferase Deficiency/blood , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Male , Middle Aged , Norway , Reference Values , Smoking , Triglycerides/blood , Washington
SELECTION OF CITATIONS
SEARCH DETAIL