Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lipids Health Dis ; 12: 70, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23663650

RESUMEN

OBJECTIVES: Variants of the secreted glycoprotein, proprotein convertase subtilisin/kexin 9 (PCSK9), associate with both hypo- and hyper-cholesterolemic phenotypes. Herein, we carried out full exonic sequencing of PCSK9 documenting the frequency of single and multiple PCSK9 variations and their effects on serum lipoprotein and PCSK9 levels in Caucasian Canadians. METHODS: The 12 exons of PCSK9 were sequenced in 207 unrelated Caucasian Canadians. Minor allele frequencies of PCSK9 variants were compared amongst LDL cholesterol (LDLC) quintiles. Serum PCSK9 levels were measured by ELISA and lipoproteins by enzymatic methods. Comparisons were made with a Caucasian family cohort (n=51) and first generation African Canadians (n=31). RESULTS: In Caucasians, but not African Canadians, the c.61_63insCTG (denoted L10Ins) and A53V PCSK9 variations were linked and their frequency was significantly higher among Caucasian Canadians with LDLC levels in the <25th percentile. In both the unrelated and family Caucasian cohorts those carrying the L10A53V PCSK9 variant had significantly lower LDLC without reduction in plasma PCSK9. The I474V PCSK9 variant associated with significantly lower serum PCSK9 and LDLC. A novel PCSK9 variant was identified; E206K. We found that the frequency of multiple PCSK9 variations was higher in first generation African Canadians. CONCLUSIONS: We showed that the L10A53V and I474V PCSK9 variants were significantly associated with lower LDLC levels in Caucasian Canadians but differed in their effect on serum PCSK9 concentrations, illuminating differences in their mechanism of inaction and indicating that that PCSK9 measurement alone may not always be a good indicator of PCSK9 function.Full exonic sequencing of PCSK9 pointed to factors that may contribute to L10Ins PCSK9 variant loss of function in Canadians of Caucasian but not those of African descent. These included; (1) its tight linkage with the A53V variant in Caucasians and/or (2) for both the L10 and I474V, the combined (and negating) effect of multiple, differing phenotypic PCSK9 variants within individuals of African ancestry for which combinations of PCSK9 variations and their overall frequency was higher. No population studies, to our knowledge, have addressed or accessed the effect of multiple PCSK9 variants on cholesterol profiles. Our results indicate that this should be considered.


Asunto(s)
Colesterol/sangre , Estudios de Asociación Genética , Hipercolesterolemia/genética , Proproteína Convertasas/genética , Receptores de LDL/genética , Serina Endopeptidasas/genética , Población Negra/genética , Canadá , Colesterol/genética , Exones , Femenino , Frecuencia de los Genes , Heterocigoto , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Proproteína Convertasa 9 , Receptores de LDL/metabolismo , Población Blanca/genética
2.
Clin Chem ; 57(10): 1415-23, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21813713

RESUMEN

BACKGROUND: PCSK9 (proprotein convertase subtilisin/kexin type 9) is a polymorphic gene whose protein product regulates plasma LDL cholesterol (LDLC) concentrations by shuttling liver LDL receptors (LDLRs) for degradation. PCSK9 variants that cause a gain or loss of PCSK9 function are associated with hyper- or hypocholesterolemia, which increases or reduces the risk of cardiovascular disease, respectively. We studied the clinical and molecular characteristics of a novel PCSK9 loss-of-function sequence variant in a white French-Canadian family. METHODS: In vivo plasma and ex vivo secreted PCSK9 concentrations were measured with a commercial ELISA. We sequenced the PCSK9 exons for 15 members of a family, the proband of which exhibited very low plasma PCSK9 and LDLC concentrations. We then conducted a structure/function analysis of the novel PCSK9 variant in cell culture to identify its phenotypic basis. RESULTS: We identified a PCSK9 sequence variant in the French-Canadian family that produced the PCSK9 Q152H substitution. Family members carrying this variant had mean decreases in circulating PCSK9 and LDLC concentrations of 79% and 48%, respectively, compared with unrelated noncarriers (n=210). In cell culture, the proPCSK9-Q152H variant did not undergo efficient autocatalytic cleavage and was not secreted. Cells transiently transfected with PCSK9-Q152H cDNA had LDLR concentrations that were significantly higher than those of cells overproducing wild-type PCSK9 (PCSK9-WT). Cotransfection of PCSK9-Q152H and PCSK9-WT cDNAs produced a 78% decrease in the secreted PCSK9-WT protein compared with control cells. CONCLUSIONS: Collectively, our results demonstrate that the PCSK9-Q152H variant markedly lowers plasma PCSK9 and LDLC concentrations in heterozygous carriers via decreased autocatalytic processing and secretion, and hence, inactivity on the LDLR.


Asunto(s)
LDL-Colesterol/sangre , Serina Endopeptidasas/genética , Adulto , Anciano de 80 o más Años , Células Cultivadas , Femenino , Variación Genética , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Proproteína Convertasa 9 , Proproteína Convertasas , Serina Endopeptidasas/sangre , Serina Endopeptidasas/metabolismo , Población Blanca , Adulto Joven
3.
J Lipid Res ; 51(1): 140-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19571328

RESUMEN

The proprotein convertase subtilisin kexin-9 (PCSK9) circulates in plasma as mature and furin-cleaved forms. A polyclonal antibody against human PCSK9 was used to develop an ELISA that measures total plasma PCSK9 rather than only the mature form. A cross-sectional study evaluated plasma levels in normal (n = 254) and hypercholesterolemic (n = 200) subjects treated or untreated with statins or statin plus ezetimibe. In controls, mean plasma PCSK9 (89.5 +/- 31.9 ng/ml) correlated positively with age, total cholesterol, LDL-cholesterol (LDL-C), triglycerides, and fasting glucose. Sequencing PCSK9 from individuals at the extremes of the normal PCSK9 distribution identified a new loss-of-function R434W variant associated with lower levels of circulating PCSK9 and LDL-C. In hypercholesterolemic subjects, PCSK9 levels were higher than in controls (99.3 +/- 31.7 ng/ml, P < 0.04) and increased in proportion to the statin dose, combined or not with ezetimibe. In treated patients (n = 139), those with familial hypercholesterolemia (FH; due to LDL receptor gene mutations) had higher PCSK9 values than non-FH (147.01 +/- 42.5 vs. 127.2 +/- 40.8 ng/ml, P < 0.005), but LDL-C reduction correlated positively with achieved plasma PCSK9 levels to a similar extent in both subsets (r = 0.316, P < 0.02 in FH and r = 0.275, P < 0.009 in non-FH). The detection of circulating PCSK9 in both FH and non-FH subjects means that this assay could be used to monitor response to therapy in a wide range of patients.


Asunto(s)
Monitoreo de Drogas/métodos , Ensayo de Inmunoadsorción Enzimática , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipoproteinemia Tipo II/sangre , Serina Endopeptidasas/sangre , Anticuerpos/inmunología , Azetidinas/uso terapéutico , Glucemia/análisis , Línea Celular , Colesterol/sangre , LDL-Colesterol/sangre , Estudios Transversales , Ezetimiba , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Proproteína Convertasa 9 , Proproteína Convertasas , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/inmunología , Triglicéridos/sangre
4.
J Cell Biochem ; 106(4): 608-17, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19130493

RESUMEN

Apolipoprotein E (apoE), a key regulator of lipid metabolism, is highly produced by adipose tissue and adipocytes. However, there is little information about its role on adipocyte functions. Because apoE-deficiency in adipocytes was shown to impair adipocyte differentiation, we investigated the consequences of apoE high expression on differentiation and proliferation of a human adipocytic cell line (SW872). SW872 cells were transfected with human apoE to induce a fivefold increase in apoE production and secretion. Adipocyte differentiation and proliferation were assayed by measuring lipid content, adipogenic gene expression, cell number, cell resistance to serum deprivation, and cell division kinetics. Cultured apoE-transfected cells accumulated less triglycerides and less cholesterol than control cells. This decrease in lipid accumulation was associated with a strong downregulation of peroxisome proliferator-activated receptors gamma1 and gamma2 and stearoyl-CoA desaturase 1. The decrease in lipid accumulation was not dependent on the presence of lipids, lipoproteins, or PPAR-gamma agonists in the culture medium, nor was it observed with exogenously added apoE. Moreover, we observed that apoE-transfected cells were more resistant to death induced by serum deprivation, and that these cells underwent more cell divisions than control cells. These results bring new evidence of apoE-involvement in metabolic disorders at the adipocyte level.


Asunto(s)
Adipocitos/citología , Apolipoproteínas E/fisiología , Proliferación Celular , Metabolismo de los Lípidos , Acilcoenzima A/genética , Apolipoproteínas E/genética , Diferenciación Celular , Línea Celular , Regulación hacia Abajo/genética , Humanos , Receptores Activados del Proliferador del Peroxisoma/genética
5.
DNA Cell Biol ; 26(9): 665-71, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17683244

RESUMEN

Niemann-pick type C (NPC) disease is characterized by endosomal and lysosomal accumulation of lipids, impaired tubulovesicular trafficking, and neurodegeneration leading to premature death. Current treatment options are limited to mainly symptomatic treatments. Thus, new and efficient drug targets are needed, and therefore we performed a Gene Set Enrichment Analysis (GSEA) on NPC and healthy fibroblasts to identify globally affected pathways in NPC that could serve as targets for later drug discovery programs. Cell lines were characterized by analyzing cellular concentrations of cholesterol, its precursors and metabolites, as well as cellular plant sterol levels. Gene expression analyses were performed with Sentrix Human-8 Expression BeadChips, analyzing 23,000 transcripts. Pathway analysis of the expression data was performed using the GSEA method. Twenty-seven upregulated and 33 downregulated pathways emerged as globally affected in the GSEA analysis. These pathways included, for example, mitochondrial pathway, caspase cascade, as well as prostaglandin and leukotriene metabolism. Based on the present results and earlier published data, anti-inflammatory and antiapoptotic treatment could be beneficial in NPC.


Asunto(s)
Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Enfermedad de Niemann-Pick Tipo C/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Transducción de Señal , Biomarcadores/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Fibroblastos/citología , Humanos , Metabolismo de los Lípidos , Enfermedad de Niemann-Pick Tipo C/metabolismo
6.
DNA Cell Biol ; 26(11): 765-72, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17867930

RESUMEN

Sterol regulatory element-binding proteins (SREBPs) are transcription factors governing transcription of genes related to cholesterol and fatty acid metabolism. To become active, SREBPs must undergo a proteolytic cleavage to allow an active NH(2)-terminal segment to translocate into the nucleus. SKI-1/S1P is the first protease in the proteolytic activation cascade of SREBPs. SREBP inhibition may be useful, for example, in the treatment of liver steatosis caused by homocysteine-induced lipid synthesis. Accordingly, we overexpressed inhibitory prodomains (proSKI) of SKI-1/S1P in HepG2 cells to block SREBP activation to evaluate the potential of SKI-1/S1P in controlling cellular cholesterol synthesis. SKI-1/S1P inhibition resulted in reduced cholesterol synthesis and mRNA levels of the rate-limiting enzymes, HMG-CoA reductase and squalene epoxidase, in the cholesterol synthetic pathway. The inhibitory effect was maintained in the presence of homocysteine-induced endoplasmic reticulum stress. A gene set enrichment analysis was performed to elucidate other metabolic effects caused by SKI-1/S1P inhibition. SKI-1/S1P inhibition was observed to affect a number of other metabolic pathways, including glycolysis and citric acid cycle. These results demonstrate that inhibition of SREBPs decreases cholesterol synthesis in HepG2 cells both in the absence and presence of homocysteine. SKI-1/S1P inhibition may cause widespread changes in other key metabolic pathways.


Asunto(s)
Colesterol/metabolismo , Homocisteína/metabolismo , Proproteína Convertasas/antagonistas & inhibidores , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Línea Celular , Glucólisis , Humanos , Redes y Vías Metabólicas , Análisis de Secuencia por Matrices de Oligonucleótidos , Proproteína Convertasas/farmacología , ARN Mensajero/metabolismo , Serina Endopeptidasas
7.
Free Radic Biol Med ; 41(5): 786-96, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16895799

RESUMEN

Oxidized low-density lipoproteins (Ox-LDL) are key elements in atherogenesis. Apolipoprotein AI (apoAI) is an active component of the antiatherogenic high-density lipoproteins (HDL). In contrast, plasma apolipoprotein B (apoB), the main component of LDL, is highly correlated with coronary risk. Our results, obtained in HepG2 cells, show that Ox-LDL, unlike native LDL, leads to opposite effects on apoB and apoAI, namely a decrease in apoAI and an increase in apoB secretion as evaluated by [(3)H]leucine incorporation and specific immunoprecipitation. Parallel pulse-chase studies show that Ox-LDL impaired apoB degradation, whereas apoAI degradation was increased and mRNA levels were decreased. We also found that enhanced lipid biosynthesis of both triglycerides and cholesterol esters was involved in the Ox-LDL-induced increase in apoB secretion. Our data suggest that the increase in apoB and decrease in apoAI secretion may in part contribute to the known atherogenicity of Ox-LDL through an elevated LDL/HDL ratio, a strong predictor of coronary risk in patients.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteínas B/química , Lipoproteínas LDL/química , Oxígeno/metabolismo , Aterosclerosis , Línea Celular , Ésteres del Colesterol/química , Radicales Libres , Humanos , Leucina/química , Lípidos/química , Oxígeno/química , ARN Mensajero/metabolismo , Factores de Tiempo , Triglicéridos/química
8.
Atherosclerosis ; 178(2): 257-64, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15694932

RESUMEN

ApoC-I plays an important role in controlling plasma lipid metabolism, however little is known about factors regulating the hepatic synthesis and secretion of this apolipoprotein. In the present study, we have carried out experiments with human hepatoma (HepG2) cells, in order to determine the effect of different tissue culture conditions on cellular lipid levels and on the production of apoC-I (and apoE) at the protein and mRNA level. Cells incubated for 48 h with 10% human serum had significantly higher cellular triglyceride (22%, P<0.05) and cholesterol levels (19%, P<0.01), higher medium apoC-I and apoE levels (2.6- and 2.9-fold, respectively), but similar levels of apoC-I and apoE mRNA, compared to cells incubated with 10% human lipoprotein-deficient serum (LPDS). Serum containing only HDL, or containing HDL with LDL, also increased cellular lipids and increased secreted apoC-I and apoE levels without altering apoC-I and apoE mRNA levels. Incubation of cells with Intralipid triglyceride (625 microM), increased cellular triglyceride (2.8-fold, P<0.001), decreased cellular cholesterol (32%, P<0.01), decreased cellular and medium apoC-I (24 and 26%, P<0.01) and had no effect on apoC-I mRNA levels. Additional experiments in which cells were loaded with cholesterol (incubation with 10 microg/ml cholesterol plus 1 microg/ml 25-hydroxycholesterol) or depleted of cholesterol (statin treatment) confirmed that secretion of apoC-I by HepG2 cells was dependent on cellular cholesterol levels and independent of changes in apoC-I mRNA levels. These results demonstrate that cellular cholesterol rather than triglyceride levels play a role in controlling apoC-I production by HepG2 cells and that this regulation occurs at a post-transcriptional level.


Asunto(s)
Apolipoproteínas C/biosíntesis , Apolipoproteínas C/metabolismo , Arteriosclerosis/fisiopatología , Colesterol/farmacología , Apolipoproteína C-I , Carcinoma Hepatocelular/patología , Humanos , Líquido Intracelular/química , Neoplasias Hepáticas/patología , Procesamiento Postranscripcional del ARN , ARN Mensajero/biosíntesis , Células Tumorales Cultivadas
9.
Arterioscler Thromb Vasc Biol ; 24(8): 1454-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15178557

RESUMEN

OBJECTIVE: Neural apoptosis-regulated convertase (NARC)-1 is the newest member of the proprotein convertase family implicated in the cleavage of a variety of protein precursors. The NARC-1 gene, PCSK9, has been identified recently as the third locus implicated in autosomal dominant hypercholesterolemia (ADH). The 2 other known genes implicated in ADH encode the low-density lipoprotein receptor and apolipoprotein B. As an approach toward the elucidation of the physiological role(s) of NARC-1, we studied its transcriptional regulation. METHODS AND RESULTS: Using quantitative RT-PCR, we assessed NARC-1 regulation under conditions known to regulate genes involved in cholesterol metabolism in HepG2 cells and in human primary hepatocytes. We found that NARC-1 expression was strongly induced by statins in a dose-dependent manner and that this induction was efficiently reversed by mevalonate. NARC-1 mRNA level was increased by cholesterol depletion but insensitive to liver X receptor activation. Human, mouse, and rat PCSK9 promoters contain 2 typical conserved motifs for cholesterol regulation: a sterol regulatory element (SRE) and an Sp1 site. CONCLUSIONS: PCSK9 regulation is typical of that of the genes implicated in lipoprotein metabolism. In vivo, PCSK9 is probably a target of SRE-binding protein (SREBP)-2.


Asunto(s)
Colesterol/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/análogos & derivados , Ácido Mevalónico/análogos & derivados , Serina Endopeptidasas/genética , Alitretinoína , Animales , Atorvastatina , Secuencia de Bases , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Colesterol/farmacología , Secuencia de Consenso , Proteínas de Unión al ADN/fisiología , Hepatocitos/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Homeostasis , Humanos , Hidroxicolesteroles/farmacología , Hidroximetilglutaril-CoA Reductasas/fisiología , Receptores X del Hígado , Lovastatina/farmacología , Ácido Mevalónico/farmacología , Ratones , Receptores Nucleares Huérfanos , Regiones Promotoras Genéticas/genética , Proproteína Convertasa 9 , Proproteína Convertasas , Piridinas/farmacología , Pirroles/farmacología , Quinolinas/farmacología , ARN Mensajero/biosíntesis , Ratas , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Serina Endopeptidasas/biosíntesis , Simvastatina/farmacología , Factor de Transcripción Sp1/fisiología , Especificidad de la Especie , Proteína 2 de Unión a Elementos Reguladores de Esteroles , Factores de Transcripción/fisiología , Tretinoina/farmacología
10.
Atherosclerosis ; 169(1): 63-70, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12860251

RESUMEN

An Hpa I restriction site located 317 bp upstream of the transcription initiation site of the apoC-I gene has been shown to increase apoC-I gene transcription in vitro. The aim of the present study was to determine whether this genetic polymorphism was associated in vivo with increased plasma levels of apoC-I. In a cohort of French-Canadians (n=391) recruited for a family study, we found strong linkage disequilibrium between the genes for apoC-I and apoE (as reported before for European-Americans), such that the apoC-I Hpa I-negative (H1) allele was strongly associated with apoE epsilon 3, whereas the apoC-I Hpa I-positive (H2) allele was strongly associated with apoE epsilon 2 and epsilon 4. ApoC-I and apoE were measured by ELISA in total plasma and in very low-density lipoproteins (VLDL) separated by ultracentrifugation (d<1.006 g/ml), and then by difference for the non-VLDL fraction (d>1.006 g/ml), in a subset of families selected for their diverse apoE genotypes. Subjects were divided into normolipidemic (NL, n=89, TG<2.3 mmol/l, LDL-C<3.8 mmol/l) and hyperlipidemic groups (HL, n=88, TG>2.3 mmol/l and/or LDL-C>3.8 mmol/l). In NL subjects, apoC-I levels were not significantly associated with apoC-I genotype (H1/H1, H1/H2 or H2/H2). They were, however, related to apoE genotype, such that apoE3/2 subjects tended to have higher and apoE4/3 subjects tended to have lower concentrations of total plasma and non-VLDL apoC-I and apoE. Total plasma, VLDL and non-VLDL apoC-I and E levels were also higher in HL subjects with an apoE2/2 or apoE3/2 genotype. These results suggest that plasma levels of apoC-I are more strongly influenced by apoE genotype than by the Hpa I apoC-I promoter polymorphism, which probably reflects an effect of different apoE isoforms on plasma lipoprotein and plasma apoC-I metabolism, rather than a direct effect of apoE alleles on apoC-I transcription.


Asunto(s)
Apolipoproteínas C/sangre , Apolipoproteínas E/genética , Desequilibrio de Ligamiento , Polimorfismo Genético , Regiones Promotoras Genéticas/genética , Adulto , Alelos , Apolipoproteína C-I , Apolipoproteínas C/análisis , Apolipoproteínas C/genética , Canadá , Femenino , Francia/etnología , Genotipo , Humanos , Hiperlipidemias/genética , Lípidos/sangre , Lipoproteínas VLDL/química , Masculino , Persona de Mediana Edad
11.
Toxicol Sci ; 125(2): 586-94, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22106037

RESUMEN

LY2541546 is a humanized monoclonal antibody (IgG(4)) that has been optimized for neutralizing activity against sclerostin. In 5-week and 6-month nonclinical safety studies in rats, LY2541546 caused dose-dependent reversible decreases in platelet counts accompanied by accelerated platelet production, increased megakaryocytes, and altered megakaryocyte morphology. These treatment-related effects resulted in altered primary hemostasis as manifested by prolonged bleeding after phlebotomy or incidental toenail break. In some cases, the defects in hemostasis were sufficient to result in death of the affected rats. There was no evidence in rats of general bone marrow suppression or processes (e.g., disseminated intravascular coagulopathy) that may result in thrombocytopenia. Cynomolgus monkeys given LY2541546 for 5 weeks or 9 months had no changes in platelet count or megakaryocytes. In vitro cross-reactivity studies in rats, cynomolgus monkeys, and humans revealed LY2541546-bound rat but not cynomolgus monkey or human platelets and megakaryocytes. These data taken together demonstrated that the platelet and megakaryocyte effects in rats had a species-specific pathogenesis which likely involved LY2541546 binding of a rat-specific antigen on the surface of platelets and megakaryocytes resulting in the increased clearance of platelets and megakaryocyte hyperplasia. The species-specific nature of these reversible toxicological findings combined with the ease of clinical monitoring using standard hematology enabled the safe initiation of clinical studies in human volunteers.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Plaquetas/efectos de los fármacos , Proteínas Morfogenéticas Óseas/inmunología , Megacariocitos/efectos de los fármacos , Trombocitopenia/inducido químicamente , Animales , Especificidad de Anticuerpos , Plaquetas/patología , Reacciones Cruzadas , Relación Dosis-Respuesta a Droga , Femenino , Hemostasis/efectos de los fármacos , Humanos , Hiperostosis/inducido químicamente , Macaca fascicularis , Masculino , Megacariocitos/patología , Recuento de Plaquetas , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Trombocitopenia/sangre , Trombocitopenia/patología
13.
Atherosclerosis ; 207(2): 452-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19515369

RESUMEN

Familial LCAT deficiency (FLD) is a disease characterized by a defect in the enzyme lecithin:cholesterol acyltransferase (LCAT) resulting in low HDL-C, premature corneal opacities, anemia as well as proteinuria and renal failure. We have identified the first French Canadian kindred with familial LCAT deficiency. Two brothers, presenting classical signs of FLD, were shown to be homozygous for a novel LCAT mutation. This c.102delG mutation occurs at the codon for His35 and causes a frameshift that stops transcription at codon 61 abolishing LCAT enzymatic activity both in vivo and in vitro. It has a dramatic effect on the lipoprotein profile, with an important reduction of HDL-C in both heterozygotes (22%) and homozygotes (88%) and a significant decrease in LDL-C in heterozygotes (35%) as well as homozygotes (58%). Furthermore, the lipoprotein profile differs markedly between the two affected brothers who had different APOE genotypes. We propose that APOE could be an important modifier gene explaining heterogeneity in lipoprotein profiles observed among FLD patients. Our results suggest that a LCAT-/- genotype associated with an APOE epsilon2 allele could be a novel mechanism leading to dysbetalipoproteinemia.


Asunto(s)
Apolipoproteína E2/genética , Mutación del Sistema de Lectura , Hiperlipoproteinemia Tipo III/genética , Deficiencia de la Lecitina Colesterol Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Eliminación de Secuencia , Adulto , Anciano , Apolipoproteína A-I/sangre , Apolipoproteína E2/sangre , Apolipoproteínas B/sangre , Biomarcadores/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Cromatografía en Gel , Análisis Mutacional de ADN , Electroforesis en Gel de Agar , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Hiperlipoproteinemia Tipo III/sangre , Hiperlipoproteinemia Tipo III/enzimología , Deficiencia de la Lecitina Colesterol Aciltransferasa/sangre , Deficiencia de la Lecitina Colesterol Aciltransferasa/enzimología , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Quebec , Factores de Riesgo , Triglicéridos/sangre , Adulto Joven
14.
FEBS Lett ; 583(12): 2042-8, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19450584

RESUMEN

Oxidative stress in adipose tissue constitutes a pathological process involved in obesity-linked metabolic disorders. Apolipoprotein E (apoE), which exhibits antioxidant properties in plasma and brain, is highly produced by adipose tissue and adipocytes. In this study, we investigated the role of apoE in the human adipocyte response to oxidative stress. We first demonstrated that apoE secretion by adipocytes was stimulated by oxidative stress. We also observed that apoE overexpression protected adipocytes from hydrogen peroxide-induced damages, by mitigating intracellular oxidation and exerting extracellular antioxidant properties. Our findings clearly show a novel antioxidant role for apoE in adipose tissue.


Asunto(s)
Adipocitos/metabolismo , Apolipoproteínas E/metabolismo , Adipocitos/efectos de los fármacos , Adiponectina/genética , Antioxidantes/metabolismo , Apolipoproteínas E/genética , Línea Celular Tumoral , Células Cultivadas , Expresión Génica , Humanos , Peróxido de Hidrógeno/toxicidad , Leptina/genética , Estrés Oxidativo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
15.
Atherosclerosis ; 197(1): 118-24, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17399720

RESUMEN

In the French Canadian population six mutations appear to be responsible for about 85% of FH cases. Two of these mutations are large deletions. The most prevalent deletion is a >15 kb deletion of the promoter and first exon; the second, a 5 kb deletion that removes exons 2 and 3. The high frequency of these deletions in the French Canadian population has been attributed to a founder effect. Other mutations are present in the population but at a much lower prevalence. We recently identified two new large deletions in FH patients of French Canadian descent. Carriers of the new deletions were identified because of an unusual pattern of band migration on Southern blots. We have identified and sequenced the deletions' boundaries. The first deletion covers 3813 bp and removes exons 7 and 8. The second deletion covers 5994 bp and removes exons 3-6. These deletions have not been previously reported. They would have been missed if a PCR-based method had been used instead of Southern blot analysis.


Asunto(s)
Análisis Mutacional de ADN , Eliminación de Gen , Hiperlipoproteinemia Tipo II/etnología , Hiperlipoproteinemia Tipo II/genética , Adulto , Southern Blotting , Exones/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ontario/epidemiología , Reacción en Cadena de la Polimerasa , Prevalencia , Mapeo Restrictivo
16.
J Nutr ; 134(11): 2935-41, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15514255

RESUMEN

Little is known about the regulation of apolipoprotein (apo) C-I production by human adipocytes. The aim of the present study, therefore, was to investigate the effect of different tissue culture conditions on the synthesis and secretion of apoC-I and apoE in human SW872 liposarcoma cells. After 3-4 d in culture (0.5 x 10(6) cells/well, DMEM/F-12 medium with 10% fetal calf serum), cells reached confluence and became growth arrested. The molar ratio of apoE:apoC-I in the cell was 8.9 +/- 0.6 and in the medium was 6.6 +/- 0.5. After 17 d in culture, SW872 cells contained significantly more cholesterol (100%) and triglyceride (3-fold) and secreted more apoC-I [4 vs. 17 d: 0.11 +/- 0.01 vs. 0.23 +/- 0.01 pmol/(10(6) cells . 24 h), P < 0.001] and apoE [0.7 +/- 0.1 vs. 3.1 +/- 0.3 pmol/(10(6) cells . 24 h), P < 0.001]. Cellular apoC-I increased 7-fold and apoE increased 16-fold. Cell maturation was associated with significantly higher levels of apoE mRNA but not apoC-I mRNA. Increases in cell lipids, apoC-I, and apoE were not dependent on the presence of extracellular lipids because similar changes occurred in cells incubated with lipoprotein-deficient serum or in cells incubated without serum. Treatment (7 d) of cells during maturation with insulin (10 or 1000 nmol/L) significantly reduced the secretion of apoC-I and apoE. These results demonstrate that in maturing SW872 cells, cholesterol and triglyceride accumulation in the presence or absence of extracellular lipids, is associated with increased apoC-I and apoE production. Furthermore, apoC-I and apoE production are differentially regulated at the transcriptional level, and long-term treatment with insulin has an inhibitory rather than stimulatory effect on apoC-I and apoE production.


Asunto(s)
Adipocitos/metabolismo , Apolipoproteínas C/biosíntesis , Apolipoproteínas C/metabolismo , Apolipoproteínas E/biosíntesis , Apolipoproteínas E/metabolismo , Adipocitos/química , Adipocitos/efectos de los fármacos , Apolipoproteínas C/genética , Apolipoproteínas E/genética , División Celular , Colesterol/metabolismo , Medios de Cultivo , Humanos , Insulina/farmacología , Liposarcoma , PPAR alfa/genética , PPAR gamma/genética , ARN Mensajero/análisis , Triglicéridos/metabolismo , Células Tumorales Cultivadas
17.
J Cardiovasc Pharmacol ; 42(2): 251-7, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12883330

RESUMEN

Apolipoprotein (apo) E and C-I are plasma apolipoproteins that have been implicated in the etiology of atherosclerosis and obesity, respectively. Both proteins are synthesized and secreted by macrophages, though pharmacological regulation of their production is poorly understood. The authors compared the effect of 2 HMG-CoA reductase inhibitors, atorvastatin and cerivastatin, on the synthesis and secretion of apoE and apoC-I by THP-1 macrophages. Atorvastatin reduced medium apoE and cellular apoE mRNA of PMA-activated THP-1 cells in a dose-dependent manner (-24% and -22%, respectively, at 1-micromol/L, P < 0.01). ApoC-I in the medium was also reduced by atorvastatin in a dose-dependent manner, though to a lesser extent (-15% at 1-micromol/L, P < 0.05). Cerivastatin similarly reduced medium apoE (-20% at 1-micromol/L, P < 0.05) and cellular apoE mRNA (-31% at 1-micromol/L, P < 0.05), and significantly lowered cellular apoC-I mRNA (-15%, P < 0.05), but not apoC-I in the medium. In experiments with THP-1 macrophages loaded with cholesterol (ie, 24-hour incubation with acetyl-LDL), atorvastatin and cerivastatin (1-micromol/L) significantly (P < 0.05) reduced both medium apoE (-30% and -25%, respectively) and cellular apoE mRNA (-25% and -17%, respectively). A lower and less consistent effect was observed on medium apoC-I (-6% and -18%, respectively) and cellular apoC-I mRNA (-13% and -19%, respectively). These data demonstrate that statins have the capacity to reduce the synthesis and secretion of both apoE and apoC-I in THP-1 macrophages loaded or unloaded with cholesterol.


Asunto(s)
Apolipoproteínas C/biosíntesis , Apolipoproteínas E/biosíntesis , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macrófagos/efectos de los fármacos , Pirroles/farmacología , Apolipoproteína C-I , Apolipoproteínas C/metabolismo , Apolipoproteínas E/metabolismo , Atorvastatina , Células Cultivadas , Macrófagos/metabolismo
18.
J Cell Biochem ; 90(4): 766-76, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14587032

RESUMEN

As an inflammatory cell, the macrophage produces various oxidizing agents, such as free radical species. These can modify LDL as a secondary effect and doing so may favor atherogenic processes. Any molecule able to counteract these reactions would be of much benefit, especially if secreted by the macrophage itself at the lesion site. Such is the case for apolipoprotein E (apoE), which has been shown to exert antioxidant properties in some studies, mostly in relation to Alzheimer's disease. In this study, we assessed the antioxidant potential of the various isoforms of apoE (E2, E3, and E4) using a metal-induced LDL oxidation system with exogenous recombinant apoE and an in vitro model of macrophage-mediated LDL oxidation. We found that all three isoforms had an antioxidant capacity. However, whereas apoE2 was the most protective isoform in the cell-free system, the opposite was observed in apoE-transfected J774 macrophages. In the latter model, cellular cholesterol efflux was found to be more important with apoE2, possibly explaining the larger quantity of oxidative indices observed in the medium. It is proposed that the antioxidant property of apoE results from a balance between direct apoE antioxidant capacities, such as the ability to trap free radicals, and potentially pro-oxidative indirect events associated with cholesterol efflux from cells. Our observations add to the therapeutic potential of apoE. However, they also suggest the need for more experiments in order to achieve careful selection of the apoE isoform to be targeted, especially in the perspective of apoE transgene use.


Asunto(s)
Apolipoproteínas E/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Animales , Apolipoproteínas E/genética , Línea Celular , Colesterol/metabolismo , Cobre/metabolismo , Radicales Libres/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Oxidación-Reducción , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Tiempo
19.
J Lipid Res ; 43(10): 1680-7, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12364552

RESUMEN

ApoC-I has several different lipid-regulating functions including, inhibition of receptor-mediated uptake of plasma triglyceride-rich lipoproteins, inhibition of cholesteryl ester transfer activity, and mediation of tissue fatty acid uptake. Since little is known about the rate of production and catabolism of plasma apoC-I in humans, the present study was undertaken to determine the plasma kinetics of VLDL and HDL apoC-I using a primed constant (12 h) intravenous infusion of deuterium-labeled leucine. Data were obtained for 14 subjects: normolipidemics (NL, n = 4), hypertriglyceridemics (HTG, n = 4) and combined hyperlipidemics (CHL, n = 6). Plasma VLDL triglyceride (TG) levels were 0.59 +/- 0.03, 4.32 +/- 0.77 (P < 0.01 vs. NL), and 2.20 +/- 0.39 mmol/l (P < 0.01 vs. NL), and plasma LDL cholesterol (LDL-C) levels were 2.34 +/- 0.22, 2.48 +/- 0.26, and 5.35 +/- 0.48 mmol/l (P < 0.01 vs. NL), respectively. HTG and CHL had significantly (P < 0.05) increased levels of total plasma apoC-I (12.5 +/- 1.2 and 12.4 +/- 1.3 mg/dl, respectively) versus NL (7.9 +/- 0.6 mg/dl), due to significantly (P < 0.01) elevated levels of VLDL apoC-I (5.8 +/- 0.8 and 4.5 +/- 0.8 vs. 0.3 +/- 0.1 mg/dl). HTG and CHL also had increased rates of VLDL apoC-I transport (i.e., production) versus NL: 2.29 +/- 0.34 and 3.04 +/- 0.53 versus 0.24 +/- 0.11 mg/kg.day (P < 0.01), with no significant change in VLDL apoC-I residence times (RT): 1.16 +/- 0.12 versus 0.69 +/- 0.06 versus 0.74 +/- 0.17. Although HDL apoC-I concentrations were not significantly lower in HTG and CHL versus NL, HDL apoC-I rates of transport were inversely related to plasma and VLDL-TG levels (r = -0.63 and -0.62, respectively, P < 0.05). Our results demonstrate that increased levels of plasma and VLDL apoC-I in hypertriglyceridemic subjects (with or without elevated LDL-C levels) are associated with increased levels of plasma VLDL apoC-I production.


Asunto(s)
Apolipoproteínas C/sangre , Hipertrigliceridemia/sangre , Lipoproteínas HDL/sangre , Lipoproteínas VLDL/sangre , Adulto , Apolipoproteínas C/clasificación , Interpretación Estadística de Datos , Deuterio , Electroforesis en Gel de Poliacrilamida/métodos , Femenino , Humanos , Cinética , Masculino , Programas Informáticos
20.
J Biol Chem ; 279(39): 41197-207, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15269218

RESUMEN

ABCA1 is a cholesterol transporter that is widely expressed throughout the body. Outside the central nervous system (CNS), ABCA1 functions in the biogenesis of high-density lipoprotein (HDL), where it mediates the efflux of cholesterol and phospholipids to apolipoprotein (apo) A-I. Deficiency of ABCA1 results in lack of circulating HDL and greatly reduced levels of apoA-I. ABCA1 is also expressed in cells within the CNS, but its roles in brain lipid metabolism are not yet fully understood. In the brain, glia synthesize the apolipoproteins involved in CNS lipid metabolism. Here we demonstrate that glial ABCA1 is required for cholesterol efflux to apoA-I and plays a key role in facilitating cholesterol efflux to apoE, which is the major apolipoprotein in the brain. In both astrocytes and microglia, ABCA1 deficiency reduces lipid efflux to exogenous apoE. The impaired ability to efflux lipids in ABCA1-/- glia results in lipid accumulation in both astrocytes and microglia under normal culture conditions. Additionally, apoE secretion is compromised in ABCA1-/- astrocytes and microglia. In vivo, deficiency of ABCA1 results in a 65% decrease in apoE levels in whole brain, and a 75-80% decrease in apoE levels in hippocampus and striatum. Additionally, the effect of ABCA1 on apoE is selective, as apoJ levels are unchanged in brains of ABCA1-/- mice. Taken together, these results show that glial ABCA1 is a key influence on apoE metabolism in the CNS.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/fisiología , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Transportador 1 de Casete de Unión a ATP , Animales , Apolipoproteínas/metabolismo , Astrocitos/metabolismo , Transporte Biológico , Western Blotting , Sistema Nervioso Central/metabolismo , Cerebelo/metabolismo , Colesterol/metabolismo , Metabolismo de los Lípidos , Lipoproteínas HDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Microglía/metabolismo , Microscopía Fluorescente , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA