Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Metabolism ; 116: 154464, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309714

RESUMEN

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.


Asunto(s)
Apolipoproteína A-I/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Deficiencia de la Lecitina Colesterol Aciltransferasa/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/sangre , Fosfolípidos/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Animales , Apolipoproteína A-I/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Enfermedades Renales/genética , Enfermedades Renales/patología , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfolípidos/farmacología , Podocitos/efectos de los fármacos , Podocitos/patología , Podocitos/fisiología , Proteínas Recombinantes/farmacología
2.
Curr Opin Lipidol ; 31(4): 232-237, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32618730

RESUMEN

PURPOSE OF REVIEW: Genetic LCAT deficiency is a rare metabolic disorder characterized by low-plasma HDL cholesterol levels. Clinical manifestations of the disease include corneal opacification, anemia, and renal disease, which represents the major cause of morbidity and mortality in carriers. RECENT FINDINGS: Biochemical and clinical manifestations of the disease are very heterogeneous among carriers. The collection of large series of affected individuals is needed to answer various open questions on this rare disorder of lipid metabolism, such as the cause of renal damage in patients with complete LCAT deficiency and the cardiovascular risk in carriers of different LCAT gene mutations. SUMMARY: Familial LCAT deficiency is a rare disease, with serious clinical manifestations, which can occur in the first decades of life, and presently with no cure. The timely diagnosis in carriers, together with the identification of disease biomarkers able to predict the evolution of clinical manifestations, would be of great help in the identification of carriers to address to future available therapies.


Asunto(s)
Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Humanos , Mutación , Factores de Riesgo
3.
Biomolecules ; 9(12)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779197

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) is an enzyme secreted by the liver and circulates with high-density lipoprotein (HDL) in the blood. The enzyme esterifies plasma cholesterol and increases the capacity of HDL to carry and potentially remove cholesterol from tissues. Cholesterol accumulates within the extracellular connective tissue matrix of the cornea stroma in individuals with genetic deficiency of LCAT. LCAT can be activated by apolipoproteins (Apo) including ApoD and ApoA1. ApoA1 also mediates cellular synthesis of HDL. This study examined the expression of LCAT by epithelial cells, keratocytes, and endothelial cells, the cell types that comprise from anterior to posterior the three layers of the cornea. LCAT and ApoD were immunolocalized to all three cell types within the cornea, while ApoA1 was immunolocalized to keratocytes and endothelium but not epithelium. In situ hybridization was used to detect LCAT, ApoD, and ApoA1 mRNA to learn what cell types within the cornea synthesize these proteins. No corneal cells showed mRNA for ApoA1. Keratocytes and endothelium both showed ApoD mRNA, but epithelium did not. Epithelium and endothelium both showed LCAT mRNA, but despite the presence of LCAT protein in keratocytes, keratocytes did not show LCAT mRNA. RNA sequencing analysis of serum-cultured dedifferentiated keratocytes (commonly referred to as corneal stromal fibroblasts) revealed the presence of both LCAT and ApoD (but not ApoA1) mRNA, which was accompanied by their respective proteins detected by immunolabeling of the cultured keratocytes and Western blot analysis of keratocyte lysates. The results indicate that keratocytes in vivo show both ApoA1 and LCAT proteins, but do not synthesize these proteins. Rather, keratocytes in vivo must take up ApoA1 and LCAT from the corneal interstitial tissue fluid.


Asunto(s)
Apolipoproteína A-I/metabolismo , Apolipoproteínas D/metabolismo , Colesterol/metabolismo , Córnea/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Anciano , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Apolipoproteínas D/sangre , Apolipoproteínas D/genética , Córnea/enzimología , Córnea/patología , Córnea/ultraestructura , Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/metabolismo , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Queratinocitos/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteínas HDL/sangre , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfolípidos/metabolismo , RNA-Seq , Enfermedad de Tangier/genética , Enfermedad de Tangier/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1351-1360, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30742993

RESUMEN

High density lipoprotein (HDL) has attracted the attention of biomedical community due to its well-documented role in atheroprotection. HDL has also been recently implicated in the regulation of islets of Langerhans secretory function and in the etiology of peripheral insulin sensitivity. Indeed, data from numerous studies strongly indicate that the functions of pancreatic ß-cells, skeletal muscles and adipose tissue could benefit from improved HDL functionality. To better understand how changes in HDL structure may affect diet-induced obesity and type 2 diabetes we aimed at investigating the impact of Apoa1 or Lcat deficiency, two key proteins of peripheral HDL metabolic pathway, on these pathological conditions in mouse models. We report that universal deletion of apoa1 or lcat expression in mice fed western-type diet results in increased sensitivity to body-weight gain compared to control C57BL/6 group. These changes in mouse genome correlate with discrete effects on white adipose tissue (WAT) metabolic activation and plasma glucose homeostasis. Apoa1-deficiency results in reduced WAT mitochondrial non-shivering thermogenesis. Lcat-deficiency causes a concerted reduction in both WAT oxidative phosphorylation and non-shivering thermogenesis, rendering lcat-/- mice the most sensitive to weight gain out of the three strains tested, followed by apoa1-/- mice. Nevertheless, only apoa1-/- mice show disturbed plasma glucose homeostasis due to dysfunctional glucose-stimulated insulin secretion in pancreatic ß-islets and insulin resistant skeletal muscles. Our analyses show that both apoa1-/- and lcat-/- mice fed high-fat diet have no measurable Apoa1 levels in their plasma, suggesting no direct involvement of Apoa1 in the observed phenotypic differences among groups.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Apolipoproteína A-I/genética , Glucosa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Obesidad/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Tejido Adiposo Blanco/patología , Animales , Apolipoproteína A-I/deficiencia , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Homeostasis/genética , Insulina/metabolismo , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Deficiencia de la Lecitina Colesterol Aciltransferasa/etiología , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas HDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Fosforilación Oxidativa , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Transducción de Señal , Termogénesis/genética , Aumento de Peso/genética
5.
J Pharmacol Exp Ther ; 368(3): 423-434, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30563940

RESUMEN

Familial LCAT deficiency (FLD) is due to mutations in lecithin:cholesterol acyltransferase (LCAT), a plasma enzyme that esterifies cholesterol on lipoproteins. FLD is associated with markedly reduced levels of plasma high-density lipoprotein and cholesteryl ester and the formation of a nephrotoxic lipoprotein called LpX. We used a mouse model in which the LCAT gene is deleted and a truncated version of the SREBP1a gene is expressed in the liver under the control of a protein-rich/carbohydrate-low (PRCL) diet-regulated PEPCK promoter. This mouse was found to form abundant amounts of LpX in the plasma and was used to determine whether treatment with recombinant human LCAT (rhLCAT) could prevent LpX formation and renal injury. After 9 days on the PRCL diet, plasma total and free cholesterol, as well as phospholipids, increased 6.1 ± 0.6-, 9.6 ± 0.9-, and 6.7 ± 0.7-fold, respectively, and liver cholesterol and triglyceride concentrations increased 1.7 ± 0.4- and 2.8 ±0.9-fold, respectively, compared with chow-fed animals. Transmission electron microscopy revealed robust accumulation of lipid droplets in hepatocytes and the appearance of multilamellar LpX particles in liver sinusoids and bile canaliculi. In the kidney, LpX was found in glomerular endothelial cells, podocytes, the glomerular basement membrane, and the mesangium. The urine albumin/creatinine ratio increased 30-fold on the PRCL diet compared with chow-fed controls. Treatment of these mice with intravenous rhLCAT restored the normal lipoprotein profile, eliminated LpX in plasma and kidneys, and markedly decreased proteinuria. The combined results suggest that rhLCAT infusion could be an effective therapy for the prevention of renal disease in patients with FLD.


Asunto(s)
Modelos Animales de Enfermedad , Riñón/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/tratamiento farmacológico , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteína X/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/administración & dosificación , Animales , Dieta Baja en Carbohidratos/efectos adversos , Proteínas en la Dieta/efectos adversos , Femenino , Riñón/efectos de los fármacos , Riñón/patología , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteína X/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
6.
J Clin Lipidol ; 12(5): 1157-1163, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30055975

RESUMEN

BACKGROUND: The significance of Lp8, that is, abnormal lipoprotein(s) detected in fraction 8 by combined high-performance liquid chromatography/gel filtration column in patients with familial lecithin:cholesterol acyltransferase (LCAT) syndrome, in relation to the severity of LCAT deficiency has not been analyzed. OBJECTIVE: We have studied Lp8 in a patient with primary biliary cirrhosis. METHODS: Plasma lipoproteins were analyzed using high-performance liquid chromatography/gel filtration column in the course of treatment of a 47-year-old female patient with primary biliary cirrhosis. RESULTS: Electrophoretic lipoprotein analyses showed massive accumulation of abnormal ß- and preß-lipoproteins with a minor lipoprotein fraction at a position near the cathode corresponding to Lp-X, on day A (status: hypercholesterolemia, LCAT activity undetectable). Chromatographic lipoprotein subfraction analysis revealed free cholesterol- and phospholipid-rich lipoproteins in fractions 1-6, corresponding to chylomicrons and very low-density lipoprotein, and phospholipid- and triglyceride-rich lipoproteins with increased free cholesterol, that is, Lp8, in fractions 7-9 (corresponding to low-density lipoprotein). On day B, after additional treatment for 7 months (status: almost normolipidemia, decreased LCAT activity), although the abnormal lipoprotein and the lipoproteins in fractions 1-6, were drastically decreased, the presence of Lp8 persisted. CONCLUSIONS: Lp8 likely is a minor abnormal lipoprotein fraction in patients with mildly decreased secondary LCAT activity, as well as with severely reduced primary LCAT activity.


Asunto(s)
Deficiencia de la Lecitina Colesterol Aciltransferasa/complicaciones , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteínas/metabolismo , Cirrosis Hepática Biliar/complicaciones , Dislipidemias/complicaciones , Femenino , Humanos , Cirrosis Hepática Biliar/metabolismo , Persona de Mediana Edad
7.
J Pharmacol Exp Ther ; 362(2): 306-318, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28576974

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) catalyzes plasma cholesteryl ester formation and is defective in familial lecithin:cholesterol acyltransferase deficiency (FLD), an autosomal recessive disorder characterized by low high-density lipoprotein, anemia, and renal disease. This study aimed to investigate the mechanism by which compound A [3-(5-(ethylthio)-1,3,4-thiadiazol-2-ylthio)pyrazine-2-carbonitrile], a small heterocyclic amine, activates LCAT. The effect of compound A on LCAT was tested in human plasma and with recombinant LCAT. Mass spectrometry and nuclear magnetic resonance were used to determine compound A adduct formation with LCAT. Molecular modeling was performed to gain insight into the effects of compound A on LCAT structure and activity. Compound A increased LCAT activity in a subset (three of nine) of LCAT mutations to levels comparable to FLD heterozygotes. The site-directed mutation LCAT-Cys31Gly prevented activation by compound A. Substitution of Cys31 with charged residues (Glu, Arg, and Lys) decreased LCAT activity, whereas bulky hydrophobic groups (Trp, Leu, Phe, and Met) increased activity up to 3-fold (P < 0.005). Mass spectrometry of a tryptic digestion of LCAT incubated with compound A revealed a +103.017 m/z adduct on Cys31, consistent with the addition of a single hydrophobic cyanopyrazine ring. Molecular modeling identified potential interactions of compound A near Cys31 and structural changes correlating with enhanced activity. Functional groups important for LCAT activation by compound A were identified by testing compound A derivatives. Finally, sulfhydryl-reactive ß-lactams were developed as a new class of LCAT activators. In conclusion, compound A activates LCAT, including some FLD mutations, by forming a hydrophobic adduct with Cys31, thus providing a mechanistic rationale for the design of future LCAT activators.


Asunto(s)
Cisteína/fisiología , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Compuestos de Sulfhidrilo/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Células HEK293 , Humanos , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Modelos Moleculares , Fosfatidilcolina-Esterol O-Aciltransferasa/química , Compuestos de Sulfhidrilo/química
8.
J Lipid Res ; 58(5): 994-1001, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28351888

RESUMEN

The aim of this study was to evaluate the vasoprotective effects of HDL isolated from carriers of LCAT deficiency, which are characterized by a selective depletion of LpA-I:A-II particles and predominance of preß migrating HDL. HDLs were isolated from LCAT-deficient carriers and tested in vitro for their capacity to promote NO production and to inhibit vascular cell adhesion molecule-1 (VCAM-1) expression in cultured endothelial cells. HDLs from carriers were more effective than control HDLs in promoting eNOS activation with a gene-dose-dependent effect (PTrend = 0.048). As a consequence, NO production induced by HDL from carriers was significantly higher than that promoted by control HDL (1.63 ± 0.24-fold vs. 1.34 ± 0.07-fold, P = 0.031). HDLs from carriers were also more effective than control HDLs in inhibiting the expression of VCAM-1 (homozygotes, 65.0 ± 8.6%; heterozygotes, 53.1 ± 7.2%; controls, 44.4 ± 4.1%; PTrend = 0.0003). The increased efficiency of carrier HDL was likely due to the depletion in LpA-I:A-II particles. The in vitro findings might explain why carriers of LCAT deficiency showed flow-mediated vasodilation and plasma-soluble cell adhesion molecule concentrations comparable to controls, despite low HDL-cholesterol levels. These results indicate that selective depletion of apoA-II-containing HDL, as observed in carriers of LCAT deficiency, leads to an increased capacity of HDL to stimulate endothelial NO production, suggesting that changes in HDL apolipoprotein composition may be the target of therapeutic interventions designed to improve HDL functionality.


Asunto(s)
Apolipoproteína A-II/deficiencia , Apolipoproteína A-I/deficiencia , Células Endoteliales/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas HDL/metabolismo , Adulto , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
Biochim Biophys Acta ; 1861(7): 594-605, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27090939

RESUMEN

Non-alcoholic steatohepatitis (NASH), is the form of non-alcoholic fatty liver disease posing risk to progress into serious long term complications. Human and pre-clinical models implicate cellular cholesterol dysregulation playing important role in its development. Mouse model studies suggest synergism between dietary cholesterol and fat in contributing to NASH but the mechanisms remain poorly understood. Our laboratory previously reported the primary importance of hepatic endoplasmic reticulum cholesterol (ER-Chol) in regulating hepatic ER stress by comparing the responses of wild type, Ldlr-/-xLcat+/+ and Ldlr-/-xLcat-/- mice, to a 2% high cholesterol diet (HCD). Here we further investigated the roles of ER-Chol and ER stress in HFHS diet-induced NASH using the same strains. With HFHS diet feeding, both WT and Ldlr-/-xLcat+/+ accumulate ER-Chol in association with ER stress and inflammasome activation but the Ldlr-/-xLcat-/- mice are protected. By contrast, all three strains accumulate cholesterol crystal, in correlation with ER-Chol, albeit less so in Ldlr-/-xLcat-/- mice. By comparison, HCD feeding per se (i) is sufficient to promote steatosis and activate inflammasomes, and (ii) results in dramatic accumulation of cholesterol crystal which is linked to inflammasome activation in Ldlr-/-xLcat-/- mice, independent of ER-Chol. Our data suggest that both dietary fat and cholesterol each independently promote steatosis, cholesterol crystal accumulation and inflammasome activation through distinct but complementary pathways. In vitro studies using palmitate-induced hepatic steatosis in HepG2 cells confirm the key roles by cellular cholesterol in the induction of steatosis and inflammasome activations. These novel findings provide opportunities for exploring a cellular cholesterol-focused strategy for treatment of NASH.


Asunto(s)
Colesterol en la Dieta/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Receptores de LDL/genética , Animales , Colesterol en la Dieta/efectos adversos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Femenino , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Metabolismo de los Lípidos/genética , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Oxidación-Reducción , Ácido Palmítico/farmacología , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Receptores de LDL/deficiencia , Transducción de Señal
10.
PLoS One ; 11(2): e0150083, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26919698

RESUMEN

Human familial lecithin:cholesterol acyltransferase (LCAT) deficiency (FLD) is characterized by low HDL, accumulation of an abnormal cholesterol-rich multilamellar particle called lipoprotein-X (LpX) in plasma, and renal disease. The aim of our study was to determine if LpX is nephrotoxic and to gain insight into the pathogenesis of FLD renal disease. We administered a synthetic LpX, nearly identical to endogenous LpX in its physical, chemical and biologic characteristics, to wild-type and Lcat-/- mice. Our in vitro and in vivo studies demonstrated an apoA-I and LCAT-dependent pathway for LpX conversion to HDL-like particles, which likely mediates normal plasma clearance of LpX. Plasma clearance of exogenous LpX was markedly delayed in Lcat-/- mice, which have low HDL, but only minimal amounts of endogenous LpX and do not spontaneously develop renal disease. Chronically administered exogenous LpX deposited in all renal glomerular cellular and matrical compartments of Lcat-/- mice, and induced proteinuria and nephrotoxic gene changes, as well as all of the hallmarks of FLD renal disease as assessed by histological, TEM, and SEM analyses. Extensive in vivo EM studies revealed LpX uptake by macropinocytosis into mouse glomerular endothelial cells, podocytes, and mesangial cells and delivery to lysosomes where it was degraded. Endocytosed LpX appeared to be degraded by both human podocyte and mesangial cell lysosomal PLA2 and induced podocyte secretion of pro-inflammatory IL-6 in vitro and renal Cxl10 expression in Lcat-/- mice. In conclusion, LpX is a nephrotoxic particle that in the absence of Lcat induces all of the histological and functional hallmarks of FLD and hence may serve as a biomarker for monitoring recombinant LCAT therapy. In addition, our studies suggest that LpX-induced loss of endothelial barrier function and release of cytokines by renal glomerular cells likely plays a role in the initiation and progression of FLD nephrosis.


Asunto(s)
Glomérulos Renales/efectos de los fármacos , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteína X/toxicidad , Proteinuria/etiología , Animales , Apolipoproteína A-I/metabolismo , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura , Células Endoteliales/metabolismo , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Membrana Basal Glomerular/efectos de los fármacos , Membrana Basal Glomerular/patología , Mesangio Glomerular/citología , Mesangio Glomerular/metabolismo , Mesangio Glomerular/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-6/metabolismo , Glomérulos Renales/patología , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteína X/metabolismo , Lipoproteína X/farmacocinética , Lipoproteínas HDL/metabolismo , Lisosomas/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Fosfolipasas A2/metabolismo , Pinocitosis , Podocitos/metabolismo , Podocitos/patología , Proteinuria/inducido químicamente , Proteinuria/genética , Proteinuria/patología
11.
J Biol Chem ; 290(51): 30514-29, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26494623

RESUMEN

Our laboratory previously reported that lecithin:cholesterol acyltransferase (LCAT) and LDL receptor double knock-out mice (Ldlr(-/-)xLcat(-/-) or DKO) spontaneously develop functioning ectopic brown adipose tissue (BAT) in skeletal muscle, putatively contributing to protection from the diet-induced obesity phenotype. Here we further investigated their developmental origin and the mechanistic role of LCAT deficiency. Gene profiling of skeletal muscle in DKO newborns and adults revealed a classical lineage. Primary quiescent satellite cells (SC) from chow-fed DKO mice, not in Ldlr(-/-)xLcat(+/+) single-knock-out (SKO) or C57BL/6 wild type, were found to (i) express exclusively classical BAT-selective genes, (ii) be primed to express key functional BAT genes, and (iii) exhibit markedly increased ex vivo adipogenic differentiation into brown adipocytes. This gene priming effect was abrogated upon feeding the mice a 2% high cholesterol diet in association with accumulation of excess intracellular cholesterol. Ex vivo cholesterol loading of chow-fed DKO SC recapitulated the effect, indicating that cellular cholesterol is a key regulator of SC-to-BAT differentiation. Comparing adipogenicity of Ldlr(+/+)xLcat(-/-) (LCAT-KO) SC with DKO SC identified a role for LCAT deficiency in priming SC to express BAT genes. Additionally, we found that reduced cellular cholesterol is important for adipogenic differentiation, evidenced by increased induction of adipogenesis in cholesterol-depleted SC from both LCAT-KO and SKO mice. Taken together, we conclude that ectopic BAT in DKO mice is classical in origin, and its development begins in utero. We further showed complementary roles of LCAT deficiency and cellular cholesterol reduction in the SC-to-BAT adipogenesis.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Diferenciación Celular , Colesterol/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Adipocitos Marrones/patología , Animales , Colesterol/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Ratones , Ratones Noqueados , Células Satélite del Músculo Esquelético/patología
12.
Vascul Pharmacol ; 74: 114-121, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26254103

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) deficiency is associated with hypoalphalipoproteinemia, generally a predisposing factor for premature coronary heart disease. The evidence of accelerated atherosclerosis in LCAT-deficient subjects is however controversial. In this study, the effect of LCAT deficiency on vascular tone and endothelial function was investigated in LCAT knockout mice, which reproduce the human lipoprotein phenotype. Aortas from wild-type (Lcat(wt)) and LCAT knockout (Lcat(KO)) mice exposed to noradrenaline showed reduced contractility in Lcat(KO) mice (P<0.005), whereas acetylcholine exposure showed a lower NO-dependent relaxation in Lcat(KO) mice (P<0.05). Quantitative PCR and Western blotting analyses suggested an adequate eNOS expression in Lcat(KO) mouse aortas. Real-time PCR analysis indicated increased expression of ß2-adrenergic receptors vs wild-type mice. Aorta stimulation with noradrenaline in the presence of propranolol, to abolish the ß-mediated relaxation, showed the same contractile response in the two mouse lines. Furthermore, propranolol pretreatment of mouse aortas exposed to L-NAME prevented the difference in responses between Lcat(wt) and Lcat(KO) mice. The results indicate that LCAT deficiency leads to increased ß2-adrenergic relaxation and to a consequently decreased NO-mediated vasodilation that can be reversed to guarantee a correct vascular tone. The present study suggests that LCAT deficiency is not associated with an impaired vascular reactivity.


Asunto(s)
Adrenérgicos/farmacología , Deficiencia de la Lecitina Colesterol Aciltransferasa/tratamiento farmacológico , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lecitinas/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Vasodilatación/efectos de los fármacos
13.
Clin Biochem ; 48(7-8): 472-5, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25172171

RESUMEN

Lecithin cholesterol acyl transferase (LCAT) is a plasma enzyme which esterifies cholesterol, and plays a key role in the metabolism of high-density lipoprotein cholesterol (HDL-C). Genetic disorders of LCAT are associated with lipoprotein abnormalities including low levels of HDL-C and presence of lipoprotein X, and clinical features mainly corneal opacities, changes in erythrocyte morphology and renal failure. Recombinant LCAT is being developed for the treatment of patients with LCAT deficiency.


Asunto(s)
HDL-Colesterol/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/diagnóstico , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Humanos , Deficiencia de la Lecitina Colesterol Aciltransferasa/enzimología , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteína X/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/genética
14.
J Lipid Res ; 55(8): 1721-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24950691

RESUMEN

A key step in plasma HDL maturation from discoidal to spherical particles is the esterification of cholesterol to cholesteryl ester, which is catalyzed by LCAT. HDL-like lipoproteins in cerebrospinal fluid (CSF) are also spherical, whereas nascent lipoprotein particles secreted from astrocytes are discoidal, suggesting that LCAT may play a similar role in the CNS. In plasma, apoA-I is the main LCAT activator, while in the CNS, it is believed to be apoE. apoE is directly involved in the pathological progression of Alzheimer's disease, including facilitating ß-amyloid (Aß) clearance from the brain, a function that requires its lipidation by ABCA1. However, whether apoE particle maturation by LCAT is also required for Aß clearance is unknown. Here we characterized the impact of LCAT deficiency on CNS lipoprotein metabolism and amyloid pathology. Deletion of LCAT from APP/PS1 mice resulted in a pronounced decrease of apoA-I in plasma that was paralleled by decreased apoA-I levels in CSF and brain tissue, whereas apoE levels were unaffected. Furthermore, LCAT deficiency did not increase Aß or amyloid in APP/PS1 LCAT(-/-) mice. Finally, LCAT expression and plasma activity were unaffected by age or the onset of Alzheimer's-like pathology in APP/PS1 mice. Taken together, these results suggest that apoE-containing discoidal HDLs do not require LCAT-dependent maturation to mediate efficient Aß clearance.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteína A-I/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Animales , Apolipoproteína A-I/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Ratones , Ratones Noqueados , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo
15.
J Nutr Biochem ; 24(3): 567-77, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22819565

RESUMEN

Lecithin/cholesterol acyltransferase (LCAT) is responsible for the esterification of the free cholesterol of plasma lipoproteins. Here, we investigated the involvement of LCAT in mechanisms associated with diet-induced hepatic triglyceride accumulation in mice. LCAT-deficient (LCAT(-/-)) and control C57BL/6 mice were placed on a Western-type diet (17.3% protein, 48.5% carbohydrate, 21.2% fat, 0.2% cholesterol, 4.5kcal/g) for 24weeks, then histopathological and biochemical analyses were performed. We report that, in our experimental setup, male LCAT(-/-) mice are characterized by increased diet-induced hepatic triglyceride deposition and impaired hepatic histology and architecture. Mechanistic analyses indicated that LCAT deficiency was associated with enhanced intestinal absorption of dietary triglycerides (3.6±0.5mg/dl per minute for LCAT(-/-) vs. 2.0±0.7mg/dl per minute for C57BL/6 mice; P<.05), accelerated clearance of postprandial triglycerides and a reduced rate of hepatic very low density lipoprotein triglyceride secretion (9.8±1.1mg/dl per minute for LCAT(-/-) vs. 12.5±1.3mg/dl per minute for C57BL/6 mice, P<.05). No statistical difference in the average daily food consumption between mouse strains was observed. Adenovirus-mediated gene transfer of LCAT in LCAT(-/-) mice that were fed a Western-type diet for 12weeks resulted in a significant reduction in hepatic triglyceride content (121.2±5.9mg/g for control infected mice vs. 95.1±5.8mg/g for mice infected with Ad-LCAT, P<.05) and a great improvement of hepatic histology and architecture. Our data extend the current knowledge on the functions of LCAT, indicating that LCAT activity is an important modulator of processes associated with diet-induced hepatic lipid deposition.


Asunto(s)
Lipoproteínas VLDL/sangre , Hígado/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Triglicéridos/sangre , Adenoviridae/genética , Animales , Peso Corporal , Dieta , Técnicas de Transferencia de Gen , Vectores Genéticos , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Aceite de Oliva , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Aceites de Plantas/administración & dosificación , Periodo Posprandial , Triglicéridos/metabolismo
16.
Osteoarthritis Cartilage ; 21(2): 322-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23151457

RESUMEN

OBJECTIVE: Recent data suggest that obesity and related metabolic aberrations are associated with osteoarthritis (OA) development, a phenomenon that is attributed at least in part to the consumption of lipid-rich diets. To date, the molecular mechanisms that govern the lipid-OA connection remain largely unknown. Given the important role of high-density lipoprotein (HDL) in plasma and tissue lipid metabolism, the main purpose of the present study was to investigate the role of HDL metabolism in the pathobiology of OA. METHODS: We used apolipoprotein A-I (apoA-I)(-/-) mice that lack classical apoA-I containing HDL, LCAT(-/-) mice that have only immature HDL and relatively reduced HDL-cholesterol levels and control C57BL/6 mice. Mice were placed on chow or western-type (WTD) and monitored for 24 weeks. Knee joints were removed and articular cartilage was isolated for further analyses. RESULTS: The LCAT(-/-) mice were significantly more sensitive to the development of diet-induced obesity compared to the C57BL/6 and apoA-I(-/-) mice. Morphological, biochemical and molecular analyses revealed that the LCAT(-/-) obese mice developed OA, while the C57BL/6 mice that were fed WTD did not. Notably, apoA-I(-/-) mice that received WTD also developed OA although their body-weight gain was similar to their wild-type counterparts. Interestingly, bone marrow from LCAT(-/-) and apoA-I(-/-) mice contained significantly increased number of adipocytes, compared to the other groups. CONCLUSIONS: Our findings suggest that perturbations in HDL metabolism predispose to OA following chronic insult with WTD and raise the challenging possibility that HDL has a causative relation to OA in patients with metabolic syndrome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Lipoproteínas HDL/metabolismo , Redes y Vías Metabólicas/fisiología , Obesidad/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/fisiopatología , Animales , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Peso Corporal/fisiología , Causalidad , Modelos Animales de Enfermedad , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Deficiencia de la Lecitina Colesterol Aciltransferasa/fisiopatología , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/fisiopatología , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Factores de Tiempo , Mundo Occidental
17.
J Clin Endocrinol Metab ; 97(10): 3399-407, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23043194

RESUMEN

Patients with extremely low high-density lipoprotein-cholesterol (HDL-C) pose distinct challenges to clinical diagnosis and management. Confirmation of HDL-C levels below 20 mg/dl in the absence of severe hypertriglyceridemia should be followed by evaluation for secondary causes, such as androgen use, malignancy, and primary monogenic disorders, namely, apolipoprotein A-I mutations, Tangier disease, and lecithin-cholesterol acyltransferase deficiency. Global cardiovascular risk assessment is a critical component of comprehensive evaluation, although the association between extremely low HDL-C levels and atherosclerosis remains unclear. Therapeutic interventions address reversible causes of low HDL-C, multiorgan abnormalities that may accompany primary disorders and cardiovascular risk modification when appropriate. Uncommon encounters with patients exhibiting extremely low HDL-C provide an opportunity to directly observe the role of HDL metabolism in atherosclerosis and beyond the vascular system.


Asunto(s)
HDL-Colesterol/deficiencia , Enfermedad de Tangier/diagnóstico , Apolipoproteína A-I/deficiencia , HDL-Colesterol/sangre , Diagnóstico Diferencial , Humanos , Deficiencia de la Lecitina Colesterol Aciltransferasa/diagnóstico , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Masculino , Paraproteinemias/diagnóstico , Paraproteinemias/metabolismo , Enfermedad de Tangier/metabolismo , Adulto Joven
18.
J Biol Chem ; 287(24): 20755-68, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22500017

RESUMEN

We recently reported that lecithin:cholesterol acyltransferase (LCAT) knock-out mice, particularly in the LDL receptor knock-out background, are hypersensitive to insulin and resistant to high fat diet-induced insulin resistance (IR) and obesity. We demonstrated that chow-fed Ldlr-/-xLcat+/+ mice have elevated hepatic endoplasmic reticulum (ER) stress, which promotes IR, compared with wild-type controls, and this effect is normalized in Ldlr-/-xLcat-/- mice. In the present study, we tested the hypothesis that hepatic ER cholesterol metabolism differentially regulates ER stress using these models. We observed that the Ldlr-/-xLcat+/+ mice accumulate excess hepatic total and ER cholesterol primarily attributed to increased reuptake of biliary cholesterol as we observed reduced biliary cholesterol in conjunction with decreased hepatic Abcg5/g8 mRNA, increased Npc1l1 mRNA, and decreased Hmgr mRNA and nuclear SREBP2 protein. Intestinal NPC1L1 protein was induced. Expression of these genes was reversed in the Ldlr-/-xLcat-/- mice, accounting for the normalization of total and ER cholesterol and ER stress. Upon feeding a 2% high cholesterol diet (HCD), Ldlr-/-xLcat-/- mice accumulated a similar amount of total hepatic cholesterol compared with the Ldlr-/-xLcat+/+ mice, but the hepatic ER cholesterol levels remained low in conjunction with being protected from HCD-induced ER stress and IR. Hepatic ER stress correlates strongly with hepatic ER free cholesterol but poorly with hepatic tissue free cholesterol. The unexpectedly low ER cholesterol seen in HCD-fed Ldlr-/-xLcat-/- mice was attributable to a coordinated marked up-regulation of ACAT2 and suppressed SREBP2 processing. Thus, factors influencing the accumulation of ER cholesterol may be important for the development of hepatic insulin resistance.


Asunto(s)
Colesterol/metabolismo , Estrés del Retículo Endoplásmico , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Hígado/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa , Receptores de LDL/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/genética , Animales , Colesterol/genética , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/fisiología , Regulación de la Expresión Génica/genética , Resistencia a la Insulina/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/patología , Lipoproteínas/biosíntesis , Lipoproteínas/genética , Hígado/patología , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de LDL/genética , Esterol O-Aciltransferasa/genética , Esterol O-Aciltransferasa/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Esterol O-Aciltransferasa 2
19.
Biochim Biophys Acta ; 1821(4): 654-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22326749

RESUMEN

Lecithin cholesterol acyltransferase (LCAT) is the key enzyme in mediating the esterification of cholesterol on circulating lipoproteins. It has long been suggested that LCAT plays a crucial role in reverse cholesterol transport, a process depicting the removal of cellular cholesterol through efflux to high density lipoproteins (HDL) and its delivery to the liver for eventual excretion from the body. Although loss-of-function LCAT mutations invariably result in profound HDL deficiency, the role of LCAT in atherogenesis continues to be clouded with controversy. Increasing number of large scale, population-based studies failed to detect an elevated cardiac risk with reduced blood levels of LCAT, suggesting that reduced LCAT activity may not be a risk factor nor a therapeutic target. More recent studies in human LCAT gene mutation carriers tend to suggest that atherogenicity in LCAT deficiency may be dependent on the nature of the mutations, providing plausible explanations for the otherwise contradictory findings. Genetic models of LCAT excess or deficiency yielded mixed findings. Despite its known profound effects on HDL and triglyceride metabolism, the role of LCAT in metabolic disorders, including obesity and diabetes, has not received much attention. Recent studies in LCAT deficient mouse models suggest that absence of LCAT may protect against insulin resistance, diabetes and obesity. Coordinated modulation of a number of anti-obesity and insulin sensitizing pathways has been implicated. Further studies to explore the role of LCAT in the modulation of cardiometabolic disorders and the underlying mechanisms are warranted.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Lipoproteínas HDL/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Animales , Aterosclerosis/enzimología , Aterosclerosis/genética , Colesterol/sangre , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Humanos , Resistencia a la Insulina/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/enzimología , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteínas HDL/sangre , Ratones , Obesidad/enzimología , Obesidad/genética , Obesidad/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/genética
20.
J Clin Lipidol ; 5(6): 493-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22108153

RESUMEN

BACKGROUND: A case of homozygous familial lecithin:cholesterol acyltransferase (LCAT) deficiency with a novel homozygous LCAT missense mutation (replacement of methionine by arginine at position 293 in the amino acid sequence of the LCAT protein) is reported. METHODS AND RESULTS: The probable diagnosis was suggested by findings of marked high density lipoprotein (HDL) deficiency, corneal opacification, anemia, and renal insufficiency. The diagnosis was confirmed by two dimensional gel electrophoresis of HDL, the measurement of free and esterified cholesterol, and sequencing of the LCAT gene. CONCLUSIONS: In our view the most important aspects of therapy to prevent the kidney disease that these patients develop is careful control of blood pressure and lifestyle measures to optimize non HDL lipoproteins. In the future replacement therapy by gene transfer or other methods may become available.


Asunto(s)
Homocigoto , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Mutación Missense , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Secuencia de Aminoácidos , Apolipoproteína A-I/sangre , Colesterol/sangre , Opacidad de la Córnea/genética , Opacidad de la Córnea/metabolismo , Electroforesis en Gel Bidimensional , Humanos , Immunoblotting , Deficiencia de la Lecitina Colesterol Aciltransferasa/sangre , Deficiencia de la Lecitina Colesterol Aciltransferasa/metabolismo , Lipoproteínas HDL/sangre , Masculino , Persona de Mediana Edad , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proteinuria/genética , Proteinuria/metabolismo , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...