Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 400
Filter
1.
BMC Genomics ; 25(1): 634, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918701

ABSTRACT

BACKGROUND: Previous studies have demonstrated the role of N6-methyladenosine (m6A) RNA methylation in various biological processes, our research is the first to elucidate its specific impact on LCAT mRNA stability and adipogenesis in poultry. RESULTS: The 6 100-day-old female chickens were categorized into high (n = 3) and low-fat chickens (n = 3) based on their abdominal fat ratios, and their abdominal fat tissues were processed for MeRIP-seq and RNA-seq. An integrated analysis of MeRIP-seq and RNA-seq omics data revealed 16 differentially expressed genes associated with to differential m6A modifications. Among them, ELOVL fatty acid elongase 2 (ELOVL2), pyruvate dehydrogenase kinase 4 (PDK4), fatty acid binding protein 9 (PMP2), fatty acid binding protein 1 (FABP1), lysosomal associated membrane protein 3 (LAMP3), lecithin-cholesterol acyltransferase (LCAT) and solute carrier family 2 member 1 (SLC2A1) have ever been reported to be associated with adipogenesis. Interestingly, LCAT was down-regulated and expressed along with decreased levels of mRNA methylation methylation in the low-fat group. Mechanistically, the highly expressed ALKBH5 gene regulates LCAT RNA demethylation and affects LCAT mRNA stability. In addition, LCAT inhibits preadipocyte proliferation and promotes preadipocyte differentiation, and plays a key role in adipogenesis. CONCLUSIONS: In conclusion, ALKBH5 mediates RNA stability of LCAT through demethylation and affects chicken adipogenesis. This study provides a theoretical basis for further understanding of RNA methylation regulation in chicken adipogenesis.


Subject(s)
Adenosine , Adipogenesis , AlkB Homolog 5, RNA Demethylase , Chickens , Phosphatidylcholine-Sterol O-Acyltransferase , RNA Stability , Animals , Adipogenesis/genetics , Chickens/genetics , Chickens/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Female , Adenosine/analogs & derivatives , Adenosine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Methylation
2.
Nutrients ; 15(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37447314

ABSTRACT

Dysregulation of lipid metabolism has been implicated in age-related macular degeneration (AMD), the leading cause of blindness among the elderly. Lecithin cholesterol acyltransferase (LCAT) is an important enzyme responsible for lipid metabolism, which could be regulated by DNA methylation during the development of various age-related diseases. This study aimed to assess the association between LCAT DNA methylation and the risk of AMD, and to examine whether plasma vitamin and carotenoid concentrations modified this association. A total of 126 cases of AMD and 174 controls were included in the present analysis. LCAT DNA methylation was detected by quantitative real-time methylation-1specific PCR (qMSP). Circulating vitamins and carotenoids were measured using reversed-phase high-performance liquid chromatography (RP-HPLC). DNA methylation of LCAT was significantly higher in patients with AMD than those in the control subjects. After multivariable adjustment, participants in the highest tertile of LCAT DNA methylation had a 5.37-fold higher risk (95% CI: 2.56, 11.28) of AMD compared with those in the lowest tertile. Each standard deviation (SD) increment of LCAT DNA methylation was associated with a 2.23-fold (95% CI: 1.58, 3.13) increased risk of AMD. There was a J-shaped association between LCAT DNA methylation and AMD risk (Pnon-linearity = 0.03). Higher concentrations of plasma retinol and ß-cryptoxanthin were significantly associated with decreased levels of LCAT DNA methylation, with the multivariate-adjusted ß coefficient being -0.05 (95% CI: -0.08, -0.01) and -0.25 (95% CI: -0.42, -0.08), respectively. In joint analyses of LCAT DNA methylation and plasma vitamin and carotenoid concentrations, the inverse association between increased LCAT DNA methylation and AMD risk was more pronounced among participants who had a lower concentration of plasma retinol and ß-cryptoxanthin. These findings highlight the importance of comprehensively assessing LCAT DNA methylation and increasing vitamin and carotenoid status for the prevention of AMD.


Subject(s)
Macular Degeneration , Vitamins , Humans , Aged , Carotenoids , Vitamin A , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , DNA Methylation , Beta-Cryptoxanthin , Macular Degeneration/prevention & control , Vitamin K
3.
Curr Opin Lipidol ; 34(2): 35-43, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36473023

ABSTRACT

PURPOSE OF REVIEW: Genetic lecithin:cholesterol acyltransferase (LCAT) deficiency is a rare, inherited, recessive disease, which manifests as two different syndromes: Familial LCAT deficiency (FLD) and Fish-eye disease (FED), characterized by low HDL-C and corneal opacity. FLD patients also develop anaemia and renal disease. There is currently no therapy for FLD, but novel therapeutics are at different stages of development. Here, we summarize the most recent advances and the opportunities for and barriers to the further development of such therapies. RECENT FINDINGS: Recent publications highlight the heterogeneous phenotype of FLD and the uncertainty over the natural history of disease and the factors contributing to disease progression. Therapies that restore LCAT function (protein and gene replacement therapies and LCAT activators) showed promising effects on markers of LCAT activity. Although they do not restore LCAT function, HDL mimetics may slow renal disease progression. SUMMARY: The further development of novel therapeutics requires the identification of efficacy endpoints, which include quantitative biomarkers of disease progression. Because of the heterogeneity of renal disease progression among FLD individuals, future treatments for FLD will have to be tailored based on the specific clinical characteristics of the patient. Extensive studies of the natural history and biomarkers of the disease will be required to achieve this goal.


Subject(s)
Kidney Diseases , Lecithin Cholesterol Acyltransferase Deficiency , Humans , Biomarkers , Disease Progression , Kidney , Lecithin Cholesterol Acyltransferase Deficiency/drug therapy , Phenotype , Phosphatidylcholine-Sterol O-Acyltransferase/genetics
4.
J Clin Lipidol ; 16(4): 423-433, 2022.
Article in English | MEDLINE | ID: mdl-35778257

ABSTRACT

The proband was a 53-year-old Japanese woman. Despite having no atherosclerotic vascular lesions on a physiological examination, markedly decreased levels of high-density lipoprotein (HDL) were always noted at her annual medical checkup. She also had corneal opacities but neither xanthoma nor tonsillar hypertrophy. A biochemical examination showed decreased levels of both apolipoprotein A-I (apoA-I) (<5 mg/dL) and lecithin-cholesterol acyltransferase (LCAT) activity. Her brother and son also had low concentrations of HDL-cholesterol, suggesting the presence of a genetic abnormality. Therefore, a sequence analysis of the genes for ABCA1, LCAT and apoA-I proteins was performed in the proband. The analysis of the APOA1 gene revealed a novel homozygous two-nucleotide deletion in exon 4 (c.614_615delTC), which causes a frameshift after residue 205 of the apoA-I protein (p.Leu205fs). Since no mutation has been found in the ABCA1 or LCAT gene, functional abnormalities of the carboxyl-terminal region of the apoA-I protein in lipid binding might have caused the low HDL-cholesterol levels and decreased LCAT activity, possibly associated with corneal opacities but not premature CAD, in the patient.


Subject(s)
Corneal Opacity , Lecithin Cholesterol Acyltransferase Deficiency , Apolipoprotein A-I/genetics , Cholesterol, HDL/genetics , Corneal Opacity/diagnosis , Corneal Opacity/genetics , Female , Frameshift Mutation , Humans , Lecithin Cholesterol Acyltransferase Deficiency/diagnosis , Lipoproteins, HDL/genetics , Male , Middle Aged , Phosphatidylcholine-Sterol O-Acyltransferase/genetics
5.
J Lipid Res ; 63(7): 100232, 2022 07.
Article in English | MEDLINE | ID: mdl-35598637

ABSTRACT

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


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/metabolism , Apolipoproteins B , Cholesterol/metabolism , Cholesterol Esters , Humans , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lipoproteins , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase 2
6.
Cell Metab ; 34(3): 441-457.e7, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235775

ABSTRACT

Hepatic osteodystrophy (HOD) is a metabolic bone disease that is often associated with chronic liver disease and is marked by bone loss. Here, we demonstrate that hepatic expression of the phosphatase PP2Acα is upregulated during HOD, leading to the downregulation of expression of the hepatokine lecithin-cholesterol acyltransferase (LCAT). Loss of LCAT function markedly exacerbates the bone loss phenotype of HOD in mice. In addition, we found that alterations in cholesterol levels are involved in the regulation of osteoblast and osteoclast activities. We also found that LCAT improves liver function and relieves liver fibrosis in the mouse HOD model by promoting reversal of cholesterol transport from the bone to the liver. In summary, defects in a liver-bone axis occur during HOD that can be targeted to ameliorate disease progression.


Subject(s)
Bone Diseases, Metabolic , Liver Cirrhosis , Animals , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Cholesterol/metabolism , Disease Models, Animal , Disease Progression , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism
7.
Biomed Pharmacother ; 147: 112677, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35121343

ABSTRACT

Lecithin: cholesterol acyltransferase (LCAT) is the only enzyme in plasma which is able to esterify cholesterol and boost cholesterol esterify with phospholipid-derived acyl chains. In order to better understand the progress of LCAT research, it is always inescapable that it is linked to high-density lipoprotein (HDL) metabolism and reverse cholesterol transport (RCT). Because LCAT plays a central role in HDL metabolism and RCT, many animal studies and clinical studies are currently aimed at improving plasma lipid metabolism by increasing LCAT activity in order to find better treatment options for familial LCAT deficiency (FLD), fish eye disease (FED), and cardiovascular disease. Recombinant human LCAT (rhLCAT) injections, cells and gene therapy, and small molecule activators have been carried out with promising results. Recently rhLCAT therapies have entered clinical phase II trials with good prospects. In this review, we discuss the diseases associated with LCAT and therapies that use LCAT as a target hoping to find out whether LCAT can be an effective therapeutic target for coronary heart disease and atherosclerosis. Also, probing the mechanism of action of LCAT may help better understand the heterogeneity of HDL and the action mechanism of dynamic lipoprotein particles.


Subject(s)
Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/pharmacology , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Coronary Artery Disease/drug therapy , Coronary Artery Disease/pathology , Genetic Therapy , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Lecithin Cholesterol Acyltransferase Deficiency/drug therapy , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Recombinant Proteins , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology
8.
J Lipid Res ; 63(3): 100169, 2022 03.
Article in English | MEDLINE | ID: mdl-35065092

ABSTRACT

Syndromes associated with LCAT deficiency, a rare autosomal recessive condition, include fish-eye disease (FED) and familial LCAT deficiency (FLD). FLD is more severe and characterized by early and progressive chronic kidney disease (CKD). No treatment is currently available for FLD, but novel therapeutics are under development. Furthermore, although biomarkers of LCAT deficiency have been identified, their suitability to monitor disease progression and therapeutic efficacy is unclear, as little data exist on the rate of progression of renal disease. Here, we systematically review observational studies of FLD, FED, and heterozygous subjects, which summarize available evidence on the natural history and biomarkers of LCAT deficiency, in order to guide the development of novel therapeutics. We identified 146 FLD and 53 FED patients from 219 publications, showing that both syndromes are characterized by early corneal opacity and markedly reduced HDL-C levels. Proteinuria/hematuria were the first signs of renal impairment in FLD, followed by rapid decline of renal function. Furthermore, LCAT activity toward endogenous substrates and the percentage of circulating esterified cholesterol (EC%) were the best discriminators between these two syndromes. In FLD, higher levels of total, non-HDL, and unesterified cholesterol were associated with severe CKD. We reveal a nonlinear association between LCAT activity and EC% levels, in which subnormal levels of LCAT activity were associated with normal EC%. This review provides the first step toward the identification of disease biomarkers to be used in clinical trials and suggests that restoring LCAT activity to subnormal levels may be sufficient to prevent renal disease progression.


Subject(s)
Lecithin Cholesterol Acyltransferase Deficiency , Humans , Biomarkers , Heterozygote , Lecithin Cholesterol Acyltransferase Deficiency/complications , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Mutation , Phosphatidylcholine-Sterol O-Acyltransferase/genetics
9.
Acta Clin Belg ; 77(6): 970-975, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34789074

ABSTRACT

BACKGROUND: Fish-eye disease (FED) is due to a partial deficiency in LCAT activity. Nevertheless, Familial lecithin-cholesterol acyltransferase deficiency (FLD), also called Norum disease, appears when the deficiency is complete. They are both rare genetic disorders inherited in an autosomal recessive manner. Clinical signs include decreased circulating HDL cholesterol and dense corneal opacity. Kidney injuries also affect patients suffering from FLD. The diagnosis of FLD is based on the presence of characteristic signs and symptoms and confirmed by genetic testing. CASE PRESENTATION: We present a case of a 63-year-old man showing an altered lipid profile with low HDL cholesterol, chronic kidney disease (CKD) and corneal disorders. He was referred to genetic counseling in order to discard genetic LCAT deficiency due to decreased visual acuity caused by corneal opacity. A massive DNA sequencing was conducted using a multigene panel associated with lipid metabolism disturbances. RESULTS AND GENETIC FINDINGS: Two likely pathogenic variants in LCAT were identified and later confirmed by Sanger sequencing. Both (c.491 G > A and c.496 G > A) were missense variants that originated an amino acid substitution (164Arginine for Histidine and 166Alanine for Threonine, respectively) modifying the protein sequence and its 3D structure. CONCLUSIONS: FLD and FED sharing common biochemical features, and the existence of other diseases with similar clinical profiles underline the need for a timely differential diagnosis aiming to address patients to preventive programs and future available therapies. This case, added to the reduced number of publications previously reported regarding FLD and FED, contributes to better understanding the genetic characteristics, clinical features, and diagnosis of these syndromes.


Subject(s)
Corneal Opacity , Lecithin Cholesterol Acyltransferase Deficiency , Humans , Male , Cholesterol, HDL , Corneal Opacity/etiology , Corneal Opacity/genetics , Histidine , Lecithin Cholesterol Acyltransferase Deficiency/complications , Lecithin Cholesterol Acyltransferase Deficiency/diagnosis , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithins , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase , Threonine
10.
Lipids Health Dis ; 20(1): 70, 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34256778

ABSTRACT

BACKGROUND: LCAT (lecithin-cholesterol acyltransferase) deficiency is characterized by two distinct phenotypes, familial LCAT deficiency (FLD) and Fish Eye disease (FED). This is the first systematic review evaluating the ethnic distribution of LCAT deficiency, with particular emphasis on Latin America and the discussion of three Mexican-Mestizo probands. METHODS: A systematic review was conducted following the PRISMA (Preferred Reporting Items for Systematic review and Meta-Analysis) Statement in Pubmed and SciELO. Articles which described subjects with LCAT deficiency syndromes and an assessment of the ethnic group to which the subject pertained, were included. RESULTS: The systematic review revealed 215 cases (154 FLD, 41 FED and 20 unclassified) pertaining to 33 ethnic/racial groups. There was no association between genetic alteration and ethnicity. The mean age of diagnosis was 42 ± 16.5 years, with fish eye disease identified later than familial LCAT deficiency (55 ± 13.8 vs. 41 ± 14.7 years respectively). The prevalence of premature coronary heart disease was significantly greater in FED vs. FLD. In Latin America, 48 cases of LCAT deficiency have been published from six countries (Argentina (1 unclassified), Brazil (38 FLD), Chile (1 FLD), Columbia (1 FLD), Ecuador (1 FLD) and Mexico (4 FLD, 1 FED and 1 unclassified). Of the Mexican probands, one showed a novel LCAT mutation. CONCLUSIONS: The systematic review shows that LCAT deficiency syndromes are clinically and genetically heterogeneous. No association was confirmed between ethnicity and LCAT mutation. There was a significantly greater risk of premature coronary artery disease in fish eye disease compared to familial LCAT deficiency. In FLD, the emphasis should be in preventing both cardiovascular disease and the progression of renal disease, while in FED, cardiovascular risk management should be the priority. The LCAT mutations discussed in this article are the only ones reported in the Mexican- Amerindian population.


Subject(s)
Ethnicity/genetics , Lecithin Cholesterol Acyltransferase Deficiency/ethnology , Ethnicity/statistics & numerical data , Genetic Predisposition to Disease/ethnology , Genetic Predisposition to Disease/genetics , Humans , Indians, North American/genetics , Indians, North American/statistics & numerical data , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Mexico , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Racial Groups/genetics , Racial Groups/statistics & numerical data
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(10): 159002, 2021 10.
Article in English | MEDLINE | ID: mdl-34197964

ABSTRACT

DHA (docosahexaenoic acid) is an essential fatty acid that is required for the normal development and function of the brain. Because of its inability to synthesize adequate amounts of DHA from the precursors, the brain has to acquire DHA from plasma through the blood brain barrier (BBB). Recent studies demonstrated the presence of a transporter at the BBB that specifically transports DHA into the brain in the form of lysophosphatidylcholine (LPC-DHA). However, the mechanism by which LPC-DHA is generated in the plasma is not known. Our previous studies showed that there are at least three different enzymes - lecithin cholesterol acyltransferase (LCAT), endothelial lipase (EL), and hepatic lipase (HL), which can generate LPC-DHA from sn-2 DHA phosphatidylcholine. Here we determined the relative contributions of these enzymes in the delivery of DHA to the brain by measuring the brain DHA levels in the mice deficient in each of these enzymes. The results show that the brain DHA levels of LCAT-deficient mice or EL-deficient mice were not significantly lower than those of their littermates. However, brain DHA was significantly decreased in HL deficient mice (13.5% of total fatty acids) compared to their littermates (17.1%) (p < 0.002), and further decreased to 8.3% of total fatty acids in mice deficient in both HL and EL. These results suggest that HL activity may be the major source for the generation of LPC-DHA in the plasma necessary for transport into the brain, and EL might contribute to this process in the absence of HL.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/metabolism , Lipase/deficiency , Lipase/metabolism , Lysophosphatidylcholines/metabolism , Animals , Blood-Brain Barrier/enzymology , Brain/pathology , Disease Models, Animal , Docosahexaenoic Acids/analysis , Humans , Lipase/genetics , Liver/enzymology , Lysophosphatidylcholines/analysis , Male , Mice , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism
12.
Sci Rep ; 11(1): 15357, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321507

ABSTRACT

Staphylococcus aureus virulence has been associated with the production of phenol-soluble modulins (PSMs). These PSMs have distinct virulence functions and are known to activate, attract and lyse neutrophils. These PSM-associated biological functions are inhibited by lipoproteins in vitro. We set out to address whether lipoproteins neutralize staphylococcal PSM-associated virulence in experimental animal models. Serum from both LCAT an ABCA1 knockout mice strains which are characterised by near absence of high-density lipoprotein (HDL) levels, was shown to fail to protect against PSM-induced neutrophil activation and lysis in vitro. Importantly, PSM-induced peritonitis in LCAT-/- mice resulted in increased lysis of resident peritoneal macrophages and enhanced neutrophil recruitment into the peritoneal cavity. Notably, LCAT-/- mice were more likely to succumb to staphylococcal bloodstream infections in a PSM-dependent manner. Plasma from homozygous carriers of ABCA1 variants characterized by very low HDL-cholesterol levels, was found to be less protective against PSM-mediated biological functions compared to healthy humans. Therefore, we conclude that lipoproteins present in blood can protect against staphylococcal PSMs, the key virulence factor of community-associated methicillin resistant S. aureus.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Lipoproteins, HDL/blood , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Staphylococcal Infections/genetics , Animals , Bacterial Toxins/genetics , Biofilms/drug effects , Disease Models, Animal , Humans , Lipoproteins, HDL/genetics , Mice , Mice, Knockout , Neutrophils/microbiology , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/genetics
13.
BMC Cancer ; 21(1): 599, 2021 May 25.
Article in English | MEDLINE | ID: mdl-34034705

ABSTRACT

BACKGROUND: The potential reversibility of aberrant DNA methylation indicates an opportunity for oncotherapy. This study aimed to integrate methylation-driven genes and pretreatment prognostic factors and then construct a new individual prognostic model in hepatocellular carcinoma (HCC) patients. METHODS: The gene methylation, gene expression dataset and clinical information of HCC patients were downloaded from The Cancer Genome Atlas (TCGA) database. Methylation-driven genes were screened with a Pearson's correlation coefficient less than - 0.3 and a P value less than 0.05. Univariable and multivariable Cox regression analyses were performed to construct a risk score model and identify independent prognostic factors from the clinical parameters of HCC patients. The least absolute shrinkage and selection operator (LASSO) technique was used to construct a nomogram that might act to predict an individual's OS, and then C-index, ROC curve and calibration plot were used to test the practicability. The correlation between clinical parameters and core methylation-driven genes of HCC patients was explored with Student's t-test. RESULTS: In this study, 44 methylation-driven genes were discovered, and three prognostic signatures (LCAT, RPS6KA6, and C5orf58) were screened to construct a prognostic risk model of HCC patients. Five clinical factors, including T stage, risk score, cancer status, surgical method and new tumor events, were identified from 13 clinical parameters as pretreatment-independent prognostic factors. To avoid overfitting, LASSO analysis was used to construct a nomogram that could be used to calculate the OS in HCC patients. The C-index was superior to that from previous studies (0.75 vs 0.717, 0.676). Furthermore, LCAT was found to be correlated with T stage and new tumor events, and RPS6KA6 was found to be correlated with T stage. CONCLUSION: We identified novel therapeutic targets and constructed an individual prognostic model that can be used to guide personalized treatment in HCC patients.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Nomograms , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , DNA Methylation , Datasets as Topic , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hepatectomy , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver/surgery , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Liver Neoplasms/surgery , Neoplasm Staging , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Preoperative Period , ROC Curve , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Sequence Analysis, RNA , Treatment Outcome
15.
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
17.
Metabolism ; 116: 154464, 2021 03.
Article in English | MEDLINE | ID: mdl-33309714

ABSTRACT

OBJECTIVE: CER-001 is an HDL mimetic that has been tested in different pathological conditions, but never with LCAT deficiency. This study was designed to investigate whether the absence of LCAT affects the catabolic fate of CER-001, and to evaluate the effects of CER-001 on kidney disease associated with LCAT deficiency. METHODS: Lcat-/- and wild-type mice received CER-001 (2.5, 5, 10 mg/kg) intravenously for 2 weeks. The plasma lipid/ lipoprotein profile and HDL subclasses were analyzed. In a second set of experiments, Lcat-/- mice were injected with LpX to induce renal disease and treated with CER-001 and then the plasma lipid profile, lipid accumulation in the kidney, albuminuria and glomerular podocyte markers were evaluated. RESULTS: In Lcat-/- mice a decrease in total cholesterol and triglycerides, and an increase in HDL-c was observed after CER-001 treatment. While in wild-type mice CER-001 entered the classical HDL remodeling pathway, in the absence of LCAT it disappeared from the plasma shortly after injection and ended up in the kidney. In a mouse model of renal disease in LCAT deficiency, treatment with CER-001 at 10 mg/kg for one month had beneficial effects not only on the lipid profile, but also on renal disease, by limiting albuminuria and podocyte dysfunction. CONCLUSIONS: Treatment with CER-001 ameliorates the dyslipidemia typically associated with LCAT deficiency and more importantly limits renal damage in a mouse model of renal disease in LCAT deficiency. The present results provide a rationale for using CER-001 in FLD patients.


Subject(s)
Apolipoprotein A-I/therapeutic use , Kidney Diseases/drug therapy , Lecithin Cholesterol Acyltransferase Deficiency/drug therapy , Lipid Metabolism/drug effects , Lipids/blood , Phospholipids/therapeutic use , Recombinant Proteins/therapeutic use , Animals , Apolipoprotein A-I/pharmacology , Cells, Cultured , Disease Models, Animal , Kidney Diseases/genetics , Kidney Diseases/pathology , Lecithin Cholesterol Acyltransferase Deficiency/genetics , Lecithin Cholesterol Acyltransferase Deficiency/metabolism , Lecithin Cholesterol Acyltransferase Deficiency/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phospholipids/pharmacology , Podocytes/drug effects , Podocytes/pathology , Podocytes/physiology , Recombinant Proteins/pharmacology
18.
Arterioscler Thromb Vasc Biol ; 41(1): 360-376, 2021 01.
Article in English | MEDLINE | ID: mdl-33086872

ABSTRACT

OBJECTIVE: Enhancement of LCAT (lecithin:cholesterol acyltransferase) activity has possibility to be beneficial for atherosclerosis. To evaluate this concept, we characterized our novel, orally administered, small-molecule LCAT activator DS-8190a, which was created from high-throughput screening and subsequent derivatization. We also focused on its mechanism of LCAT activation and the therapeutic activity with improvement of HDL (high-density lipoprotein) functionality. Approach and Results: DS-8190a activated human and cynomolgus monkey but not mouse LCAT enzymes in vitro. DS-8190a was orally administered to cynomolgus monkeys and dose dependently increased LCAT activity (2.0-fold in 3 mg/kg group on day 7), resulting in HDL cholesterol elevation without drastic changes of non-HDL cholesterol. Atheroprotective effects were then evaluated using Ldl-r KO×hLcat Tg mice fed a Western diet for 8 weeks. DS-8190a treatment achieved significant reduction of atherosclerotic lesion area (48.3% reduction in 10 mg/kg treatment group). Furthermore, we conducted reverse cholesterol transport study using Ldl-r KO×hLcat Tg mice intraperitoneally injected with J774A.1 cells loaded with [3H]-cholesterol and confirmed significant increases of [3H] count in plasma (1.4-fold) and feces (1.4-fold on day 2 and 1.5-fold on day3) in the DS-8190a-treated group. With regard to the molecular mechanism involved, direct binding of DS-8190a to human LCAT protein was confirmed by 2 different approaches: affinity purification by DS-8190a-immobilized beads and thermal shift assay. In addition, the candidate binding site of DS-8190a in human LCAT protein was identified by photoaffinity labeling. CONCLUSIONS: This study demonstrates the potential of DS-8190a as a novel therapeutic for atherosclerosis. In addition, this compound proves that a small-molecule direct LCAT activator can achieve HDL-C elevation in monkey and reduction of atherosclerotic lesion area with enhanced HDL function in rodent.


Subject(s)
Atherosclerosis/prevention & control , Enzyme Activators/pharmacology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Plaque, Atherosclerotic , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line , Cholesterol, HDL/blood , Disease Models, Animal , Enzyme Activation , Humans , Macaca fascicularis , Macrophages/drug effects , Macrophages/enzymology , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Species Specificity , Up-Regulation
19.
FEBS Lett ; 595(6): 773-788, 2021 03.
Article in English | MEDLINE | ID: mdl-33020907

ABSTRACT

Apolipoprotein (apo) A-I, the major structural protein of high-density lipoprotein (HDL), is present in human and mouse cerebrospinal fluid (CSF) despite its lack of expression in brain cells. To identify the origin of apoA-I in CSF, we generated intestine-specific and liver-specific Apoa1 knockout mice (Apoa1ΔInt and Apoa1Δliv mice, respectively). Lipoprotein profiles of Apoa1ΔInt and Apoa1ΔLiv mice resembled those of control littermates, whereas knockout of Apoa1 in both intestine and liver (Apoa1ΔIntΔLiv ) resulted in a 60-percent decrease in HDL-cholesterol levels, thus strongly mimicking the Apoa1-/- mice. Immunoassays revealed that mouse apoA-I was not present in the CSF of the Apoa1ΔIntΔLiv mice. Furthermore, apoA-I levels in CSF were highly correlated with plasma spherical HDL levels, which were regulated by ABCA1 and LCAT. Collectively, these results suggest that apoA-I protein in CSF originates in liver and small intestine and is taken up from the plasma.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoprotein A-I/blood , Apolipoprotein A-I/cerebrospinal fluid , Intestinal Mucosa/metabolism , Lipoproteins, HDL/metabolism , Liver/metabolism , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , ATP Binding Cassette Transporter 1/genetics , Animals , Lipoproteins, HDL/genetics , Mice , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics
20.
Plant J ; 105(1): 182-196, 2021 01.
Article in English | MEDLINE | ID: mdl-33107656

ABSTRACT

Production of hydroxy fatty acids (HFAs) in transgenic crops represents a promising strategy to meet our demands for specialized plant oils with industrial applications. The expression of Ricinus communis (castor) OLEATE 12-HYDROXYLASE (RcFAH12) in Arabidopsis has resulted in only limited accumulation of HFAs in seeds, which probably results from inefficient transfer of HFAs from their site of synthesis (phosphatidylcholine; PC) to triacylglycerol (TAG), especially at the sn-1/3 positions of TAG. Phospholipase As (PLAs) may be directly involved in the liberation of HFAs from PC, but the functions of their over-expression in HFA accumulation and distribution at TAG in transgenic plants have not been well studied. In this work, the functions of lecithin:cholesterol acyltransferase-like PLAs (LCAT-PLAs) in HFA biosynthesis were characterized. The LCAT-PLAs were shown to exhibit homology to LCAT and mammalian lysosomal PLA2 , and to contain a conserved and functional Ser/His/Asp catalytic triad. In vitro assays revealed that LCAT-PLAs from the HFA-accumulating plant species Physaria fendleri (PfLCAT-PLA) and castor (RcLCAT-PLA) could cleave acyl chains at both the sn-1 and sn-2 positions of PC, and displayed substrate selectivity towards sn-2-ricinoleoyl-PC over sn-2-oleoyl-PC. Furthermore, co-expression of RcFAH12 with PfLCAT-PLA or RcLCAT-PLA, but not Arabidopsis AtLCAT-PLA, resulted in increased occupation of HFA at the sn-1/3 positions of TAG as well as small but insignificant increases in HFA levels in Arabidopsis seeds compared with RcFAH12 expression alone. Therefore, PfLCAT-PLA and RcLCAT-PLA may contribute to HFA turnover on PC, and represent potential candidates for engineering the production of unusual fatty acids in crops.


Subject(s)
Brassicaceae/enzymology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phosphatidylcholines/metabolism , Plant Proteins/metabolism , Ricinus/enzymology , Arabidopsis/metabolism , Brassicaceae/genetics , Fatty Acids/metabolism , Lysophospholipids , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Plant Proteins/genetics , Plant Roots/metabolism , Plants, Genetically Modified , Protein Structure, Tertiary , Ricinus/genetics , Seeds/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...