Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Biochim Biophys Acta ; 1851(12): 1587-95, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26454245

RESUMEN

The scavenger receptor class B type 1 (SR-B1) is an important HDL receptor involved in cholesterol uptake and efflux, but its physiological role in human lipoprotein metabolism is not fully understood. Heterozygous carriers of the SR-B1(P297S) mutation are characterized by increased HDL cholesterol levels, impaired cholesterol efflux from macrophages and attenuated adrenal function. Here, the composition and function of lipoproteins were studied in SR-B1(P297S) heterozygotes.Lipoproteins from six SR-B1(P297S) carriers and six family controls were investigated. HDL and LDL/VLDL were isolated by ultracentrifugation and proteins were separated by two-dimensional gel electrophoresis and identified by mass spectrometry. HDL antioxidant properties, paraoxonase 1 activities, apoA-I methionine oxidations and HDL cholesterol efflux capacity were assessed.Multivariate modeling separated carriers from controls based on lipoprotein composition. Protein analyses showed a significant enrichment of apoE in LDL/VLDL and of apoL-1 in HDL from heterozygotes compared to controls. The relative distribution of plasma apoE was increased in LDL and in lipid-free form. There were no significant differences in paraoxonase 1 activities, HDL antioxidant properties or HDL cholesterol efflux capacity but heterozygotes showed a significant increase of oxidized methionines in apoA-I.The SR-B1(P297S) mutation affects both HDL and LDL/VLDL protein compositions. The increase of apoE in carriers suggests a compensatory mechanism for attenuated SR-B1 mediated cholesterol uptake by HDL. Increased methionine oxidation may affect HDL function by reducing apoA-I binding to its targets. The results illustrate the complexity of lipoprotein metabolism that has to be taken into account in future therapeutic strategies aiming at targeting SR-B1.


Asunto(s)
Heterocigoto , Lipoproteínas/sangre , Mutación Missense , Receptores Depuradores de Clase B/sangre , Receptores Depuradores de Clase B/genética , Sustitución de Aminoácidos , Antioxidantes/metabolismo , Arildialquilfosfatasa/metabolismo , Femenino , Humanos , Masculino
2.
N Engl J Med ; 364(2): 136-45, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21226579

RESUMEN

BACKGROUND: In mice, the scavenger receptor class B type I (SR-BI) is essential for the delivery of high-density lipoprotein (HDL) cholesterol to the liver and steroidogenic organs. Paradoxically, elevated HDL cholesterol levels are associated with increased atherosclerosis in SR-BI-knockout mice. It is unclear what role SR-BI plays in human metabolism. METHODS: We sequenced the gene encoding SR-BI in persons with elevated HDL cholesterol levels and identified a family with a new missense mutation (P297S). The functional effects of the P297S mutation on HDL binding, cellular cholesterol uptake and efflux, atherosclerosis, platelet function, and adrenal function were studied. RESULTS: Cholesterol uptake from HDL by primary murine hepatocytes that expressed mutant SR-BI was reduced to half of that of hepatocytes expressing wild-type SR-BI. Carriers of the P297S mutation had increased HDL cholesterol levels (70.4 mg per deciliter [1.8 mmol per liter], vs. 53.4 mg per deciliter [1.4 mmol per liter] in noncarriers; P<0.001) and a reduced capacity for efflux of cholesterol from macrophages, but the carotid artery intima-media thickness was similar in carriers and in family noncarriers. Platelets from carriers had increased unesterified cholesterol content and impaired function. In carriers, adrenal steroidogenesis was attenuated, as evidenced by decreased urinary excretion of sterol metabolites, a decreased response to corticotropin stimulation, and symptoms of diminished adrenal function. CONCLUSIONS: We identified a family with a functional mutation in SR-BI. The mutation carriers had increased HDL cholesterol levels and a reduction in cholesterol efflux from macrophages but no significant increase in atherosclerosis. Reduced SR-BI function was associated with altered platelet function and decreased adrenal steroidogenesis. (Funded by the European Community and others.).


Asunto(s)
Insuficiencia Suprarrenal/genética , Aterosclerosis/genética , HDL-Colesterol/sangre , Colesterol/metabolismo , Mutación Missense , Receptores Depuradores de Clase B/genética , Adolescente , Glándulas Suprarrenales/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Arterias Carótidas/anatomía & histología , Colesterol/sangre , Análisis Mutacional de ADN , Femenino , Heterocigoto , Homeostasis/genética , Humanos , Hidrocortisona/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Linaje , Activación Plaquetaria/genética , Triglicéridos/sangre , Adulto Joven
3.
J Gene Med ; 11(8): 697-707, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19431216

RESUMEN

BACKGROUND: Inherited apolipoprotein (Apo) A-I deficiency is an orphan disorder characterized by high-density lipoprotein (HDL)-cholesterol deficiency and premature atherosclerosis. Constitutive over-expression of ApoA-I might provide a means to treat this disease. The present study provides a comprehensive evaluation of adeno-associated virus (AAV)-mediated ApoA-I gene delivery to express human (h)ApoA-I and correct the low HDL-cholesterol phenotype associated with ApoA-I deficiency. METHODS: In an effort to maximize AAV-mediated gene expression, we performed head-to-head comparisons of recombinant AAVs with pseudotype capsids 1, 2, 6 and 8 administered by different routes with the use of five different liver-specific promoters in addition to cytomegalovirus as single-stranded or as self-complementary (sc) AAV vectors. RESULTS: Intravenous administration of 1 x 10(13) gc/kg scAAV8, in combination with the liver-specific promoter LP1, in female ApoA-I(-/-) mice resulted in hApoA-I expression levels of 634 +/- 69 mg/l, which persisted for the duration of the study (15 weeks). This treatment resulted in full recovery of HDL-cholesterol levels with correction of HDL particle size and apolipoprotein composition. In addition, we observed increased adrenal cholesterol content and a significant increase in bodyweight in treated mice. CONCLUSIONS: The present study demonstrates that systemic delivery of a scAAV8 vector provides a means for efficient liver expression of hApoA-I, thereby correcting the lipid abnormalities associated with murine ApoA-I deficiency. Importantly, the study demonstrates that AAV-based gene therapy can be used to express therapeutic proteins at a high level for a prolonged period of time and, as such, provides a basis for further development of this strategy to treat hApoA-I deficiency.


Asunto(s)
Apolipoproteína A-I/sangre , Apolipoproteína A-I/deficiencia , HDL-Colesterol/sangre , Dependovirus/genética , Terapia Genética , Animales , Apolipoproteína A-I/genética , Peso Corporal , Citomegalovirus/genética , Dependovirus/clasificación , Elementos de Facilitación Genéticos/genética , Vectores Genéticos/genética , Humanos , Inyecciones Intravenosas , Hígado/metabolismo , Ratones , Mutagénesis Insercional , Especificidad de Órganos , Fenotipo , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Serotipificación , Aumento de Peso
4.
Biochem Biophys Res Commun ; 379(2): 542-6, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19121291

RESUMEN

Variation in the apolipoprotein A5 (APOA5) gene has consistently been associated with increased plasma triglyceride (TG) levels in epidemiological studies. In vivo functionality of these variations, however, has thus far not been tested. Using adenoviral over-expression, we evaluated plasma expression levels and TG-lowering efficacies of wild-type human apoAV, two human apoAV variants associated with increased TG (S19W, G185C) and one variant (Q341H) that is predicted to have altered protein function. Injection of mice with adenovirus encoding wild-type or mutant apoAV resulted in an identical dose-dependent elevation of human apoAV levels in plasma. The increase in apoAV levels resulted in pronounced lowering of plasma TG levels at two viral dosages. Unexpectedly, the TG-lowering efficacy of all three apoAV variants was similar to wild-type apoAV. In addition, no effect on TG-hydrolysis-related plasma parameters (free fatty acids, glycerol and post-heparin lipoprotein lipase activity) was apparent upon expression of all apoAV variants. In conclusion, our data indicate that despite their association with hypertriglyceridemia and/or predicted protein dysfunction, the 19W, 185C and 341H apoAV variants are equally effective in reducing plasma TG levels in mice.


Asunto(s)
Apolipoproteínas A/metabolismo , Hipertrigliceridemia/metabolismo , Triglicéridos/sangre , Animales , Apolipoproteína A-V , Apolipoproteínas A/sangre , Apolipoproteínas A/genética , Humanos , Hipertrigliceridemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple
5.
Biochim Biophys Acta ; 1761(10): 1163-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16990047

RESUMEN

UNLABELLED: Intramuscular (IM) application of adeno-associated virus serotype 1 (AAV1) for the delivery of human lipoprotein lipase (hLPL) was previously shown efficacious in mice with chylomicronemia. The current study addresses whether AAV1-LPL(S447X) can reduce elevated triglyceride (TG) levels in mice with attenuated clearance of TG-rich remnant particles. METHODS: Female mice, expressing human apoE2 but deficient for endogenous apoE (apoE2KI) received IM injections of AAV1-LPL(S447X) (n=6; 8 x 10(12) gc/kg; 4-sites) or PBS (n=5). Following lipid monitoring, the mice were challenged with intravenous Intralipid injections, and sacrificed 3 months after treatment. RESULTS: In the mice that received LPL gene therapy, a marked increase of post-heparin hLPL protein levels (averaging 517+/-277 ng/mL vs. 4+/-3 ng/mL in apoE2KI-untreated) induced 20% reductions of fasting plasma TG levels (p<0.05). This was accompanied by two-fold increased TG clearance rates after Intralipid administration at 6 weeks after treatment (p<0.05). Post-mortem analyses revealed increased levels of TG (2-fold, p<0.005) and cholesterol (1.7-fold, p<0.001) in the treated muscles. CONCLUSIONS: IM application of AAV1-LPL(S447X) is effective in reducing TG levels in a mouse model for type III dyslipidemia. Thus, hypertriglyceridemia caused by attenuated uptake of TG-rich lipoproteins can be alleviated by increasing lipolytic function of the skeletal muscle tissue.


Asunto(s)
Apolipoproteína E2/genética , Terapia Genética , Hipertrigliceridemia/terapia , Lipoproteína Lipasa/genética , Animales , Dependovirus/genética , Femenino , Vectores Genéticos , Humanos , Inyecciones Intramusculares , Lípidos/sangre , Lipoproteína Lipasa/sangre , Ratones , Ratones Transgénicos , Músculos/metabolismo , Triglicéridos/sangre
6.
Hum Gene Ther ; 16(11): 1276-86, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16259561

RESUMEN

Lipoprotein lipase (LPL) deficiency causes hypertriglyceridemia and recurrent, potentially life-threatening pancreatitis. There currently is no adequate treatment for this disease. Previously, we showed that intramuscular administration of an adeno-associated virus serotype 1 (AAV1) vector encoding the human LPL(S447X) variant cDNA (AAV1-LPL(S447X)) normalized the dyslipidemia of LPL-/- mice for more than 1 year. In preparation for a clinical trial, we evaluated the safety and biodistribution of AAV1-LPL(S447X) in wild-type mice and fully characterized six LPL-deficient patients. Toxicological analysis in mice showed that intramuscular administration was well tolerated. Acute inflammatory response markers were transiently increased, and anti- AAV1 antibodies were generated. Histological analyses indicated a dose-dependent reversible spleen hyperplasia, and myositis at the injection sites. Biodistribution data showed short-term vector leakage from injection sites into the circulation, followed by liver-mediated clearance. Persistence of vector DNA was limited to the injected muscle and draining lymph nodes, and spread to reproductive organs was limited. Characterization of LPL-deficient patients showed that all patients presented with hypertriglyceridemia and recurrent pancreatitis. LPL catalytic activity was absent, but LPL protein levels were 20-100% of normal. Myoblasts derived from skeletal muscle biopsies of these patients were efficiently transduced by AAV1-LPL(S447X) and secreted active LPL. These data support the initiation of a clinical trial in LPL-deficient patients, for which regulatory approval has been granted.


Asunto(s)
Terapia Genética , Hiperlipoproteinemia Tipo I/terapia , Lipoproteína Lipasa/genética , Animales , Dependovirus/genética , Estudios de Factibilidad , Femenino , Terapia Genética/efectos adversos , Vectores Genéticos , Inyecciones Intramusculares , Lipoproteína Lipasa/administración & dosificación , Lipoproteína Lipasa/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Tisular
7.
Cell Metab ; 14(6): 811-8, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22152306

RESUMEN

Genome-wide association studies have identified GALNT2 as a candidate gene in lipid metabolism, but it is not known how the encoded enzyme ppGalNAc-T2, which contributes to the initiation of mucin-type O-linked glycosylation, mediates this effect. In two probands with elevated plasma high-density lipoprotein cholesterol and reduced triglycerides, we identified a mutation in GALNT2. It is shown that carriers have improved postprandial triglyceride clearance, which is likely attributable to attenuated glycosylation of apolipoprotein (apo) C-III, as observed in their plasma. This protein inhibits lipoprotein lipase (LPL), which hydrolyses plasma triglycerides. We show that an apoC-III-based peptide is a substrate for ppGalNAc-T2 while its glycosylation by the mutant enzyme is impaired. In addition, neuraminidase treatment of apoC-III which removes the sialic acids from its glycan chain decreases its potential to inhibit LPL. Combined, these data suggest that ppGalNAc-T2 can affect lipid metabolism through apoC-III glycosylation, thereby establishing GALNT2 as a lipid-modifying gene.


Asunto(s)
Apolipoproteína C-III/metabolismo , Heterocigoto , Lipasa/sangre , N-Acetilgalactosaminiltransferasas/genética , Péptidos/metabolismo , Periodo Posprandial/fisiología , Adulto , Anciano , Colesterol/sangre , Electroforesis en Gel Bidimensional , Femenino , Glicosilación , Humanos , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/metabolismo , Masculino , Persona de Mediana Edad , Modelos Biológicos , Mutación/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Polipéptido N-Acetilgalactosaminiltransferasa
8.
Circ Cardiovasc Genet ; 3(2): 169-78, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20124439

RESUMEN

BACKGROUND: Recent studies in mice have established that an endothelial cell protein, glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), is essential for the lipolytic processing of triglyceride-rich lipoproteins. METHODS AND RESULTS: We report the discovery of a homozygous missense mutation in GPIHBP1 in a young boy with severe chylomicronemia. The mutation, p.C65Y, replaces a conserved cysteine in the GPIHBP1 lymphocyte antigen 6 domain with a tyrosine and is predicted to perturb protein structure by interfering with the formation of a disulfide bond. Studies with transfected Chinese hamster ovary cells showed that GPIHBP1-C65Y reaches the cell surface but has lost the ability to bind lipoprotein lipase (LPL). When the GPIHBP1-C65Y homozygote was given an intravenous bolus of heparin, only trace amounts of LPL entered the plasma. We also observed very low levels of LPL in the postheparin plasma of a subject with chylomicronemia who was homozygous for a different GPIHBP1 mutation (p.Q115P). When the GPIHBP1-Q115P homozygote was given a 6-hour infusion of heparin, a significant amount of LPL appeared in the plasma, resulting in a fall in the plasma triglyceride levels from 1780 to 120 mg/dL. CONCLUSIONS: We identified a novel GPIHBP1 missense mutation (p.C65Y) associated with defective LPL binding in a young boy with severe chylomicronemia. We also show that homozygosity for the C65Y or Q115P mutations is associated with low levels of LPL in the postheparin plasma, demonstrating that GPIHBP1 is important for plasma triglyceride metabolism in humans.


Asunto(s)
Proteínas Portadoras/genética , Quilomicrones/sangre , Lipoproteína Lipasa/sangre , Sustitución de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/química , Preescolar , Cricetinae , Cricetulus , Exones , Heparina/farmacología , Homocigoto , Humanos , Lipoproteína Lipasa/metabolismo , Masculino , Mutación Missense , Receptores de Lipoproteína , Triglicéridos/sangre
9.
Atherosclerosis ; 194(1): 55-61, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17087965

RESUMEN

BACKGROUND: Overexpression of lipoprotein lipase (LPL) protects against atherosclerosis in genetically engineered mice. We tested whether a gene therapy vector that delivers human (h) LPL(S447X) cDNA to skeletal muscle could induce similar effects. METHODS: LDL receptor knockout (LDLr-/-) mice were injected intramuscular (i.m.) with adeno-associated virus serotype 1 (AAV1) LPL(S447X) or PBS. Four weeks later they were started on an atherogenic diet for 12 weeks. After termination, atherosclerosis was assessed and homogenates of muscle and liver tissue were analyzed. RESULTS: AAV1-treated mice showed hLPL concentrations of 768+/-293 ng/mL in post-heparin plasma associated with 48% reductions of fasting triglycerides (TG) levels (p<0.0001). In the absence of an effect on total cholesterol (TC) levels, no effects on atherosclerosis were found. An increase in lipid content of injected muscles was accompanied by a significant decrease of TG (-20%, p<0.0001) and free cholesterol (FC) content (-24%, p<0.0001) in liver homogenates. CONCLUSIONS: The data show that transgenic hLPL(S447X) on top of endogenous murine LPL reduces fasting TG levels in plasma but has no effect on atherosclerosis in LDLr-/- mice. While lipid accumulation in the injected muscle was anticipated, this coincided with an interesting decrease of both TG and FC in liver homogenates.


Asunto(s)
Aterosclerosis/terapia , Dependovirus/genética , Terapia Genética/métodos , Lipoproteína Lipasa/genética , Receptores de LDL/genética , Animales , Aterosclerosis/genética , Colesterol/sangre , Grasas de la Dieta/farmacología , Emulsiones Grasas Intravenosas/farmacología , Femenino , Vectores Genéticos/genética , Humanos , Inyecciones Intramusculares , Lipoproteína Lipasa/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/fisiología , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA