Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Biochem Biophys Res Commun ; 622: 108-114, 2022 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-35843089

RESUMEN

Apolipoprotein A-I (apoA-I), the main protein component of High-Density Lipoprotein (HDL), is modified in plasma and the arterial wall by various enzymes. Myeloperoxidase (MPO), a leukocyte-derived peroxidase, is highly expressed during inflammation and associates with HDL reducing its functionality and contributing to atherosclerosis. In the present study we sought to explore further the effect of MPO on HDL structure and functionality in vivo using adenovirus-mediated gene transfer of human MPO combined with human apoA-I forms containing substitutions at MPO-sensitive sites or wild type apoA-I. We found that overexpression of MPO in mice significantly increased plasma apoA-I and HDL levels without affecting the expression of genes involved in HDL biogenesis or catabolism in the liver. Overexpression of MPO in the liver reduced the expression of pro-inflammatory genes and increased or did not affect the expression of anti-inflammatory genes suggesting that MPO had no toxic effects in this organ. In the plasma of mice overexpressing MPO, no significant alterations in HDL size or electrophoretic mobility was observed with the exception of mice expressing apoA-I (M148A) which showed enriched pre-ß relative to α HDL particles, suggesting that the apoA-I (M148A) mutation may interfere with HDL remodelling. Overexpression of MPO was associated with reduced anti-oxidant capacity of HDL particles in all mice. Interestingly, HDL particles bearing apoA-I (Y192A) showed enhanced ABCA1-dependent cholesterol efflux from macrophages which was not affected by MPO and these mice had reduced levels of LDL-c. These findings provide new insights on the role of specific amino acid residues of apoA-I in HDL structure and function following modification by MPO. This knowledge may facilitate the development of novel therapies based on improved HDL forms for patients with chronic diseases that are characterized by dysfunctional HDL.


Asunto(s)
Infecciones por Adenoviridae , Apolipoproteína A-I , Transportador 1 de Casete de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Humanos , Lipoproteínas HDL , Ratones , Peroxidasa/genética , Peroxidasa/metabolismo
2.
Biochem J ; 475(10): 1839-1859, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29743204

RESUMEN

The ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) initiates the production of amyloid-ß peptide (Aß), which is central to the pathogenesis of Alzheimer's disease (AD). Changes in brain cholesterol homeostasis have been suggested to affect Aß metabolism. Cholesterol homeostasis is maintained in the brain by apolipoprotein E (apoE). The apoE4 isoform constitutes the major risk factor for AD. Here, we investigated the effect of apoE forms on Aß generation and on BACE1 levels. We also examined the potential involvement in these processes of cholesterol transporters ABCG1 and ABCG4 or the lipoprotein receptor SR-BI, which are implicated in cholesterol efflux to apoE. It was found that reconstituted lipoprotein-associated apoE isoforms promoted the increase of Aß production and oligomerization and of BACE1 levels in human neuroblastoma SK-N-SH cells, with an apoE4 ≥ apoE3 > apoE2 potency rank order. Progressive carboxyl-terminal apoE4 deletions between residues 230-299 decreased the protein's ability to increase BACE1, while further truncations up to residue 166 prevented apoE4 from increasing BACE1 and Aß levels in SK-N-SH and primary mouse neuronal cells. ABCG1, but not ABCG4 or SR-BI, moderately increased Aß production and BACE1 levels in SK-N-SH cells. All apoE forms affected Aß production/oligomerization and BACE1 levels in a pattern that did not follow that of their capacity to promote ABCG1, ABCG4 or SR-BI-mediated cholesterol efflux. Overall, our data indicate that apoE-containing lipoprotein particles can have a direct effect on BACE1 levels and Aß secretion and possibly contribute to AD pathogenetic processes, independently of their capacity to promote cholesterol efflux.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Colesterol/metabolismo , Neuroblastoma/metabolismo , Neuronas/metabolismo , Animales , Apolipoproteína E4/genética , Transporte Biológico , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Neuronas/citología , Isoformas de Proteínas , Eliminación de Secuencia
3.
Lipids ; 52(12): 991-998, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29094255

RESUMEN

Lipid core nanoparticles (LDE) resembling LDL behave similarly to native LDL when injected in animals or subjects. In contact with plasma, LDE acquires apolipoproteins (apo) E, A-I and C and bind to LDL receptors. LDE can be used to explore LDL metabolism or as a vehicle of drugs directed against tumoral or atherosclerotic sites. The aim was to investigate in knockout (KO) and transgenic mice the plasma clearance and tissue uptake of LDE labeled with 3H-cholesteryl ether. LDE clearance was lower in LDLR KO and apoE KO mice than in wild type (WT) mice (p < 0.05). However, infusion of human apoE3 into the apoE KO mice increased LDE clearance. LDE clearance was higher in apoA-I KO than in WT. In apoA-I transgenic mice, LDE clearance was lower than in apoA-I KO and than in apoA-I KO infusion with human HDL. Infusion of human HDL into the apoA-I KO mice resulted in higher LDE clearance than in the apoA-I transgenic mice (p < 0.05). In apoA-I KO and apoA-I KO infused human HDL, the liver uptake was greater than in WT animals and apoA-I transgenic animals (p < 0.05). LDE clearance was lower in apoE/A-I KO than in WT. Infusion of human HDL increased LDE clearance in those double KO mice. No difference among the groups in LDE uptake by the tissues occurred. In conclusion, results support LDLR and apoE as the key players for LDE clearance, apoA-I also influences those processes.


Asunto(s)
Apolipoproteínas E/genética , Lípidos/sangre , Receptores de LDL/genética , Animales , Línea Celular , Humanos , Lipoproteínas HDL/administración & dosificación , Ratones , Ratones Noqueados , Ratones Transgénicos
4.
Biochemistry ; 54(46): 6931-41, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26506427

RESUMEN

Macrophage ABCA1 effluxes lipid and has anti-inflammatory activity. The syntrophins, which are cytoplasmic PDZ protein scaffolding factors, can bind ABCA1 and modulate its activity. However, many of the data assessing the function of the ABCA1-syntrophin interaction are based on overexpression in nonmacrophage cells. To assess endogenous complex function in macrophages, we derived immortalized macrophages from Abca1(+/+) and Abca1(-/-) mice and show their phenotype recapitulates primary macrophages. Abca1(+/+) lines express the CD11B and F4/80 macrophage markers and markedly upregulate cholesterol efflux in response to LXR nuclear hormone agonists. In contrast, immortalized Abca1(-/-) macrophages show no efflux to apoA-I. In response to LPS, Abca1(-/-) macrophages display pro-inflammatory changes, including an increased level of expression of cell surface CD14, and 11-26-fold higher levels of IL-6 and IL-12 mRNA. Given recapitulation of phenotype, we show with these lines that the ABCA1-syntrophin protein complex is upregulated by LXR agonists and can bind apoA-I. Moreover, in immortalized macrophages, combined α1/ß2-syntrophin loss modulated ABCA1 cell surface levels and induced pro-inflammatory gene expression. However, loss of all three syntrophin isoforms known to bind ABCA1 did not impair lipid efflux in immortalized or primary macrophages. Thus, the ABCA1-syntrophin protein complex is not essential for ABCA1 macrophage lipid efflux but does directly interact with apoA-I and can modulate the pool of cell surface ABCA1 stabilized by apoA-I.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Apolipoproteína A-I/metabolismo , Proteínas Asociadas a la Distrofina/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/agonistas , Transportador 1 de Casete de Unión a ATP/deficiencia , Transportador 1 de Casete de Unión a ATP/genética , Animales , Transporte Biológico Activo , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Proteínas Asociadas a la Distrofina/deficiencia , Proteínas Asociadas a la Distrofina/genética , Hidrocarburos Fluorados/farmacología , Metabolismo de los Lípidos , Receptores X del Hígado , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sulfonamidas/farmacología , Regulación hacia Arriba
5.
Atherosclerosis ; 243(1): 77-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26363436

RESUMEN

OBJECTIVE: Mutations in human apolipoprotein A-I (apoA-I) are associated with low high-density lipoprotein (HDL) cholesterol levels and pathological conditions such as premature atherosclerosis and amyloidosis. In this study we functionally characterized two natural human apoA-I mutations, L141RPisa and L159RFIN, in vivo. METHODS: We generated transgenic mice expressing either wild-type (WT) or the two mutant forms of human apoA-I on a mouse apoA-I(-/-) background and analyzed for abnormalities in their lipid and lipoprotein profiles. HDL structure and functionality, as well as atherosclerosis development following a 14-week high-fat diet were assessed in these mice. RESULTS: The expression of either apoA-I mutant was associated with markedly reduced serum apoA-I (<10% of WT apoA-I), total and HDL-cholesterol levels (∼20% and ∼7% of WT apoA-I, respectively) and the formation of few small size HDL particles with preß2 and α3, α4 electrophoretic mobility. HDL particles containing either of the two apoA-I mutants exhibited attenuated anti-oxidative properties as indicated by their inability to prevent low-density lipoprotein oxidation, and by decreased activities of paraoxonase-1 and platelet-activating factor acetylhydrolase. However, the apoA-I(L141R)Pisa or apoA-I(L159R)FIN-containing HDL particles demonstrated increased capacity to promote ATP-Binding Cassette Transporter A1-mediated cholesterol efflux from macrophages. Expression of apoA-I(L141R)Pisa or apoA-I(L159R)FIN mutations in mice was associated with increased diet-induced atherosclerosis compared to either WT apoA-I transgenic or apoA-I(-/-) mice. CONCLUSIONS: These findings suggest that natural apoA-I mutations L141RPisa and L159RFIN affect the biogenesis and the functionality of HDL in vivo and predispose to diet-induced atherosclerosis in the absence of any other genetic defect.


Asunto(s)
Apolipoproteína A-I/genética , Aterosclerosis/sangre , Lipoproteínas HDL/sangre , Lipoproteínas LDL/genética , Mutación , Transportador 1 de Casete de Unión a ATP/metabolismo , Alimentación Animal , Animales , Antioxidantes/química , Arildialquilfosfatasa/metabolismo , Electroforesis en Gel Bidimensional , Humanos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Activación Plaquetaria/metabolismo
6.
J Lipid Res ; 55(7): 1310-23, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24776540

RESUMEN

The K146N/R147W substitutions in apoE3 were described in patients with a dominant form of type III hyperlipoproteinemia. The effects of these mutations on the in vivo functions of apoE were studied by adenovirus-mediated gene transfer in different mouse models. Expression of the apoE3[K146N/R147W] mutant in apoE-deficient (apoE(-/-)) or apoA-I-deficient (apoA-I(-/-))×apoE(-/-) mice exacerbated the hypercholesterolemia and increased plasma apoE and triglyceride levels. In apoE(-/-) mice, the apoE3[K146N/R147W] mutant displaced apoA-I from the VLDL/LDL/HDL region and caused the accumulation of discoidal apoE-containing HDL. The WT apoE3 cleared the cholesterol of apoE(-/-) mice without induction of hypertriglyceridemia and promoted formation of spherical HDL. A unique property of the truncated apoE3[K146N/R147W]202 mutant, compared with similarly truncated apoE forms, is that it did not correct the hypercholesterolemia. The contribution of LPL and LCAT in the induction of the dyslipidemia was studied. Treatment of apoE(-/-) mice with apoE3[K146N/R147W] and LPL corrected the hypertriglyceridemia, but did not prevent the formation of discoidal HDL. Treatment with LCAT corrected hypertriglyceridemia and generated spherical HDL. The combined data indicate that the K146N/R147W substitutions convert the full-length and the truncated apoE3[K146N/R147W] mutant into a dominant negative ligand that prevents receptor-mediated remnant clearance, exacerbates the dyslipidemia, and inhibits the biogenesis of HDL.


Asunto(s)
Apolipoproteína E3/metabolismo , Lipoproteínas HDL/biosíntesis , Mutación Missense , Sustitución de Aminoácidos , Animales , Apolipoproteína E3/genética , Femenino , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Hipertrigliceridemia/patología , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Lipoproteínas HDL/genética , Masculino , Ratones , Ratones Noqueados , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo
7.
J Lipid Res ; 54(12): 3293-302, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24123812

RESUMEN

We studied the significance of four hydrophobic residues within the 225-230 region of apoA-I on its structure and functions and their contribution to the biogenesis of HDL. Adenovirus-mediated gene transfer of an apoA-I[F225A/V227A/F229A/L230A] mutant in apoA-I⁻/⁻ mice decreased plasma cholesterol, HDL cholesterol, and apoA-I levels. When expressed in apoA-I⁻/⁻ × apoE⁻/⁻ mice, approximately 40% of the mutant apoA-I as well as mouse apoA-IV and apoB-48 appeared in the VLDL/IDL/LDL. In both mouse models, the apoA-I mutant generated small spherical particles of pre-ß- and α4-HDL mobility. Coexpression of the apoA-I mutant and LCAT increased and shifted the-HDL cholesterol peak toward lower densities, created normal αHDL subpopulations, and generated spherical-HDL particles. Biophysical analyses suggested that the apoA-I[225-230] mutations led to a more compact folding that may limit the conformational flexibility of the protein. The mutations also reduced the ability of apoA-I to promote ABCA1-mediated cholesterol efflux and to activate LCAT to 31% and 66%, respectively, of the WT control. Overall, the apoA-I[225-230] mutations inhibited the biogenesis of-HDL and led to the accumulation of immature pre-ß- and α4-HDL particles, a phenotype that could be corrected by administration of LCAT.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Lipoproteínas HDL/biosíntesis , Adenoviridae/genética , Animales , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Fenómenos Químicos , Células HEK293 , Humanos , Ratones , Mutación
8.
J Lipid Res ; 54(12): 3281-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23990662

RESUMEN

We investigated the significance of hydrophobic and charged residues 218-226 on the structure and functions of apoA-I and their contribution to the biogenesis of HDL. Adenovirus-mediated gene transfer of apoA-I[L218A/L219A/V221A/L222A] in apoA-I⁻/⁻ mice decreased plasma cholesterol and apoA-I levels to 15% of wild-type (WT) control mice and generated pre-ß- and α4-HDL particles. In apoA-I⁻/⁻ × apoE⁻/⁻ mice, the same mutant formed few discoidal and pre-ß-HDL particles that could not be converted to mature α-HDL particles by excess LCAT. Expression of the apoA-I[E223A/K226A] mutant in apoA-I⁻/⁻ mice caused lesser but discrete alterations in the HDL phenotype. The apoA-I[218-222] and apoA-I[E223A/K226A] mutants had 20% and normal capacity, respectively, to promote ABCA1-mediated cholesterol efflux. Both mutants had ∼65% of normal capacity to activate LCAT in vitro. Biophysical analyses suggested that both mutants affected in a distinct manner the structural integrity and plasticity of apoA-I that is necessary for normal functions. We conclude that the alteration of the hydrophobic 218-222 residues of apoA-I disrupts apoA-I/ABCA1 interactions and promotes the generation of defective pre-ß particles that fail to mature into α-HDL subpopulations, thus resulting in low plasma apoA-I and HDL. Alterations of the charged 223, 226 residues caused milder but discrete changes in HDL phenotype.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Lipoproteínas HDL/biosíntesis , Adenoviridae/genética , Animales , Apolipoproteína A-I/sangre , Apolipoproteína A-I/genética , Línea Celular , Humanos , Lipoproteínas HDL/sangre , Ratones , Mutación , Estructura Secundaria de Proteína , Desplegamiento Proteico , Temperatura , Transgenes/genética
9.
J Lipid Res ; 54(1): 107-15, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23132909

RESUMEN

The objective of this study was to establish the role of apoA-IV, ABCA1, and LCAT in the biogenesis of apoA-IV-containing HDL (HDL-A-IV) using different mouse models. Adenovirus-mediated gene transfer of apoA-IV in apoA-I(-/-) mice did not change plasma lipid levels. ApoA-IV floated in the HDL2/HDL3 region, promoted the formation of spherical HDL particles as determined by electron microscopy, and generated mostly α- and a few pre-ß-like HDL subpopulations. Gene transfer of apoA-IV in apoA-I(-/-) × apoE(-/-) mice increased plasma cholesterol and triglyceride levels, and 80% of the protein was distributed in the VLDL/IDL/LDL region. This treatment likewise generated α- and pre-ß-like HDL subpopulations. Spherical and α-migrating HDL particles were not detectable following gene transfer of apoA-IV in ABCA1(-/-) or LCAT(-/-) mice. Coexpression of apoA-IV and LCAT in apoA-I(-/-) mice restored the formation of HDL-A-IV. Lipid-free apoA-IV and reconstituted HDL-A-IV promoted ABCA1 and scavenger receptor BI (SR-BI)-mediated cholesterol efflux, respectively, as efficiently as apoA-I and apoE. Our findings are consistent with a novel function of apoA-IV in the biogenesis of discrete HDL-A-IV particles with the participation of ABCA1 and LCAT, and may explain previously reported anti-inflammatory and atheroprotective properties of apoA-IV.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Apolipoproteínas A/metabolismo , Lipoproteínas HDL/biosíntesis , Lipoproteínas HDL/química , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Transportador 1 de Casete de Unión a ATP , Animales , Células HEK293 , Humanos , Ratones
10.
Biochem Pharmacol ; 84(11): 1451-8, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22985620

RESUMEN

Physiological levels of wild-type (wt) apolipoprotein E (apoE) in plasma mediate the clearance of cholesterol-rich atherogenic lipoprotein remnants while higher than normal plasma apoE concentrations fail to do so and trigger hypertriglyceridemia. This property of wt apoE reduces significantly its therapeutic value as a lead biological for the treatment of dyslipidemia. Recently, we reported the generation of a recombinant apoE variant, apoE4 [L261A, W264A, F265A, L268A, V269A] (apoE4mut1) with improved biological functions. Specifically, in apoE-deficient (apoE(-/-)) mice this variant can normalize high plasma cholesterol levels without triggering hypertriglyceridemia, even at supraphysiological levels of expression. In the present study we performed pharmacodynamic and pharmacokinetic analysis of apoE4mut1 in experimental mice. Using adenovirus-mediated gene transfer in LDL receptor deficient (LDLr(-/-)) mice, we show that the cholesterol lowering potential of apoE4mut1 is dependent on the expression of a functional classical LDLr. Bolus infusion of apoE4mut1-containing proteoliposomes in apoE(-/-) mice fed western-type diet for 6 weeks indicated that exogenously synthesized apoE4mut1 maintains intact its ability to normalize the high cholesterol levels of these mice with a maximum pharmacological effect obtained at 10h post-treatment. Interestingly, plasma cholesterol levels remained significantly reduced up to 24h following intravenous administration of apoE4mut1 proteoliposomes. Measurements of plasma apoE levels indicated that apoE4mut1 in the form of proteoliposomes used in the study has a half-life of 15.8h. Our data suggest that purified apoE4mut1 may be an attractive new candidate for the acute correction of hypercholesterolemia in subjects expressing functional LDL receptor.


Asunto(s)
Apolipoproteína E4/farmacología , Animales , Apolipoproteína E4/farmacocinética , Apolipoproteína E4/uso terapéutico , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Modelos Animales de Enfermedad , Hipercolesterolemia/tratamiento farmacológico , Ratones , Ratones Noqueados , Microscopía Electrónica , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de LDL/genética , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
11.
PLoS One ; 6(11): e27037, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22069485

RESUMEN

BACKGROUND: Apolipoprotein E (apoE) is a major protein of the lipoprotein transport system that plays important roles in lipid homeostasis and protection from atherosclerosis. ApoE is characterized by structural plasticity and thermodynamic instability and can undergo significant structural rearrangements as part of its biological function. Mutations in the 136-150 region of the N-terminal domain of apoE, reduce its low density lipoprotein (LDL) receptor binding capacity and have been linked with lipoprotein disorders, such as type III hyperlipoproteinemia (HLP) in humans. However, the LDL-receptor binding defects for these apoE variants do not correlate well with the severity of dyslipidemia, indicating that these variants may carry additional properties that contribute to their pathogenic potential. METHODOLOGY/PRINCIPAL FINDINGS: In this study we examined whether three type III HLP predisposing apoE3 variants, namely R136S, R145C and K146E affect the biophysical properties of the protein. Circular dichroism (CD) spectroscopy revealed that these mutations do not significantly alter the secondary structure of the protein. Thermal and chemical unfolding analysis revealed small thermodynamic alterations in each variant compared to wild-type apoE3, as well as effects in the reversibility of the unfolding transition. All variants were able to remodel multillamelar 1,2-Dimyristoyl-sn-glycero-3-phosphocholine (DMPC) vesicles, but R136S and R145C had reduced kinetics. Dynamic light scattering analysis indicated that the variant R136S exists in a higher-order oligomerization state in solution. Finally, 1-anilinonaphthalene-8-sulfonic acid (ANS) binding suggested that the variant R145C exposes a larger amount of hydrophobic surface to the solvent. CONCLUSIONS/SIGNIFICANCE: Overall, our findings suggest that single amino acid changes in the functionally important region 136-150 of apoE3 can affect the molecule's stability and conformation in solution and may underlie functional consequences. However, the magnitude and the non-concerted nature of these changes, make it unlikely that they constitute a distinct unifying mechanism leading to type III HLP pathogenesis.


Asunto(s)
Apolipoproteína E3/química , Apolipoproteína E3/genética , Astrocitoma/genética , Biofisica , Variación Genética , Hiperlipoproteinemia Tipo III/genética , Naftalenosulfonatos de Anilina/farmacología , Apolipoproteína E3/metabolismo , Astrocitoma/metabolismo , Astrocitoma/patología , Dicroismo Circular , Colorantes Fluorescentes/farmacología , Humanos , Hiperlipoproteinemia Tipo III/patología , Cinética , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Receptores de LDL/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
12.
J Biol Chem ; 286(42): 36331-9, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21873422

RESUMEN

Septic shock results from bacterial infection and is associated with multi-organ failure, high mortality, and cardiac dysfunction. Sepsis causes both myocardial inflammation and energy depletion. We hypothesized that reduced cardiac energy production is a primary cause of ventricular dysfunction in sepsis. The JNK pathway is activated in sepsis and has also been implicated in impaired fatty acid oxidation in several tissues. Therefore, we tested whether JNK activation inhibits cardiac fatty acid oxidation and whether blocking JNK would restore fatty acid oxidation during LPS treatment. LPS treatment of C57BL/6 mice and adenovirus-mediated activation of the JNK pathway in cardiomyocytes inhibited peroxisome proliferator-activated receptor α expression and fatty acid oxidation. Surprisingly, none of the adaptive responses that have been described in other types of heart failure, such as increased glucose utilization, reduced αMHC:ßMHC ratio or induction of certain microRNAs, occurred in LPS-treated mice. Treatment of C57BL/6 mice with a general JNK inhibitor (SP600125) increased fatty acid oxidation in mice and a cardiomyocyte-derived cell line. JNK inhibition also prevented LPS-mediated reduction in fatty acid oxidation and cardiac dysfunction. Inflammation was not alleviated in LPS-treated mice that received the JNK inhibitor. We conclude that activation of JNK signaling reduces fatty acid oxidation and prevents the peroxisome proliferator-activated receptor α down-regulation that occurs with LPS.


Asunto(s)
Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Cardiopatías/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/toxicidad , Proteínas Musculares/biosíntesis , Miocardio/metabolismo , PPAR alfa/biosíntesis , Animales , Antracenos/farmacología , Línea Celular , Ácidos Grasos/genética , Cardiopatías/inducido químicamente , Cardiopatías/genética , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Ratones Noqueados , Proteínas Musculares/antagonistas & inhibidores , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción/efectos de los fármacos , PPAR alfa/genética
13.
Ann Med ; 43(4): 302-11, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21604997

RESUMEN

INTRODUCTION. We have studied the functions of truncated apoE4 forms in vitro and in vivo in order to identify the domains of apoE4 required for the biogenesis of apoE-containing high-density lipoprotein (HDL). RESULTS. We have found that apoE4-185, -202, -229, or -259 could promote ATP-binding cassette transporter A1 (ABCA1)-dependent cholesterol efflux in vitro, although less efficiently than Full-length apoE4, and had diminished capacity to activate lecithin cholesterol acyltransferase (LCAT). Formation of HDL in vivo was assessed by various methods following gene transfer in apolipoprotein A-I(-/-) × apoE(-/-) mice. Fast protein liquid chromatography of plasma showed that the truncated apoE forms, except apoE4-185, generated an apoE-containing HDL peak. Two-dimensional gel electrophoresis of plasma and electron microscopy showed that truncated apoE forms generated distinct HDL subpopulations and formed discoidal HDL particles which could be converted to spherical by co-administration of truncated apoE4-202 and LCAT. CONCLUSION. Overall, the in-vivo and in-vitro data are consistent and indicate that apoE4-185 is the shortest truncated form that supports formation of discoidal apoE4-containing HDL particles.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Apolipoproteína A-I/fisiología , Apolipoproteína E4/química , Apolipoproteína E4/fisiología , Apolipoproteínas E/biosíntesis , Lipoproteínas HDL/biosíntesis , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenoviridae/genética , Animales , Apolipoproteína A-I/metabolismo , Apolipoproteína E4/biosíntesis , Apolipoproteína E4/metabolismo , Apolipoproteínas E/metabolismo , Femenino , Humanos , Lipoproteínas/biosíntesis , Lipoproteínas/metabolismo , Lipoproteínas HDL/metabolismo , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Noqueados/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , ARN Mensajero , Transducción Genética
14.
J Lipid Res ; 52(7): 1363-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21504968

RESUMEN

In this study, we investigated the role of positively and negatively charged amino acids within the 89-99 region of apolipoprotein A-I (apoA-I), which are highly conserved in mammals, on plasma lipid homeostasis and the biogenesis of HDL. We previously showed that deletion of the 89-99 region of apoA-I increased plasma cholesterol and phospholipids, but it did not affect plasma triglycerides. Functional studies using adenovirus-mediated gene transfer of two apoA-I mutants in apoA-I-deficient mice showed that apoA-I[D89A/E91A/E92A] increased plasma cholesterol and caused severe hypertriglyceridemia. HDL levels were reduced, and approximately 40% of the apoA-I was distributed in VLDL/IDL. The HDL consisted of mostly spherical and a few discoidal particles and contained preß1 and α4-HDL subpopulations. The lipid, lipoprotein, and HDL profiles generated by the apoA-I[K94A/K96A] mutant were similar to those of wild-type (WT) apoA-I. Coexpression of apoA-I[D89A/E91A/E92A] and human lipoprotein lipase abolished hypertriglyceridemia, restored in part the α1,2,3,4 HDL subpopulations, and redistributed apoA-I in the HDL2/HDL3 regions, but it did not prevent the formation of discoidal HDL particles. Physicochemical studies showed that the apoA-I[D89A/E91A/E92A] mutant had reduced α-helical content and effective enthalpy of thermal denaturation, increased exposure of hydrophobic surfaces, and increased affinity for triglyceride-rich emulsions. We conclude that residues D89, E91, and E92 of apoA-I are important for plasma cholesterol and triglyceride homeostasis as well as for the maturation of HDL.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Colesterol/metabolismo , Homeostasis , Lipoproteínas HDL/biosíntesis , Triglicéridos/metabolismo , Naftalenosulfonatos de Anilina/metabolismo , Animales , Apolipoproteína A-I/genética , Colesterol/sangre , Secuencia Conservada , Dimiristoilfosfatidilcolina/metabolismo , Emulsiones , Regulación de la Expresión Génica , Homeostasis/genética , Humanos , Cinética , Lipoproteínas HDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Temperatura , Triglicéridos/sangre
15.
J Lipid Res ; 52(1): 45-56, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20861163

RESUMEN

We have used adenovirus-mediated gene transfer in apolipoprotein (apo)E(-/-) mice to elucidate the molecular etiology of a dominant form of type III hyperlipoproteinemia (HLP) caused by the R142C substitution in apoE4. It was found that low doses of adenovirus expressing apoE4 cleared cholesterol, whereas comparable doses of apoE4[R142C] greatly increased plasma cholesterol, triglyceride, and apoE levels, caused accumulation of apoE in VLDL/IDL/LDL region, and promoted the formation of discoidal HDL. Co-expression of apoE4[R142C] with lecithin cholesterol acyltransferase (LCAT) or lipoprotein lipase (LPL) in apoE(-/-) mice partially corrected the apoE4[R142C]-induced dyslipidemia. High doses of C-terminally truncated apoE4[R142C]-202 partially cleared cholesterol in apoE(-/-) mice and promoted formation of discoidal HDL. The findings establish that apoE4[R142C] causes accumulation of apoE in VLDL/IDL/LDL region and affects in vivo the activity of LCAT and LPL, the maturation of HDL, and the clearance of triglyceride-rich lipoproteins. The prevention of apoE4[R142C]-induced dyslipidemia by deletion of the 203-299 residues suggests that, in the full-length protein, the R142C substitution may have altered the conformation of apoE bound to VLDL/IDL/LDL in ways that prevent triglyceride hydrolysis, cholesterol esterification, and receptor-mediated clearance in vivo.


Asunto(s)
Apolipoproteína E4/genética , Hiperlipoproteinemia Tipo III/genética , Animales , Células Cultivadas , Humanos , Hiperlipoproteinemia Tipo III/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mutación , Fenotipo , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Triglicéridos/sangre , Triglicéridos/metabolismo
16.
J Biol Chem ; 285(40): 30719-30, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20660599

RESUMEN

Apolipoprotein M (apoM) plays an important role in the biogenesis and the metabolism of anti-atherogenic HDL particles in plasma and is expressed primarily in the liver and the kidney. We investigated the role of hormone nuclear receptors in apoM gene regulation in hepatic cells. Overexpression via adenovirus-mediated gene transfer and siRNA-mediated gene silencing established that hepatocyte nuclear factor 4 (HNF-4) is an important regulator of apoM gene transcription in hepatic cells. apoM promoter deletion analysis combined with DNA affinity precipitation and chromatin immunoprecipitation assays revealed that HNF-4 binds to a hormone-response element (HRE) in the proximal apoM promoter (nucleotides -33 to -21). Mutagenesis of this HRE decreased basal hepatic apoM promoter activity to 10% of control and abolished the HNF4-mediated transactivation of the apoM promoter. In addition to HNF-4, homodimers of retinoid X receptor and heterodimers of retinoid X receptor with receptors for retinoic acid, thyroid hormone, fibrates (peroxisome proliferator-activated receptor), and oxysterols (liver X receptor) were shown to bind with different affinities to the proximal HRE in vitro and in vivo. Ligands of these receptors strongly induced human apoM gene transcription and apoM promoter activity in HepG2 cells, whereas mutations in the proximal HRE abolished this induction. These findings provide novel insights into the role of apoM in the regulation of HDL by steroid hormones and into the development of novel HDL-based therapies for diseases such as diabetes, obesity, metabolic syndrome, and coronary artery disease that affect a large proportion of the population in Western countries.


Asunto(s)
Apolipoproteínas/biosíntesis , Regulación de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/metabolismo , Elementos de Respuesta , Apolipoproteínas/genética , Apolipoproteínas M , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hormonas/metabolismo , Hormonas/farmacología , Humanos , Lipocalinas , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Esteroides/metabolismo , Esteroides/farmacología
17.
J Neurochem ; 115(4): 873-84, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20412390

RESUMEN

Apolipoprotein E (apoE) plays a crucial role in lipid transport in circulation and the brain. The apoE4 isoform is a major risk factor for Alzheimer's disease (AD). ApoE4 is more susceptible to proteolysis than other apoE isoforms and apoE4 fragments have been found in brains of AD patients. These apoE4 fragments have been hypothesized to be involved in the pathogenesis of AD, although the mechanism is not clear. In this study we examined the effect of lipid-free apoE4 on amyloid precursor protein processing and 40-amino-acid Aß variant and 42-amino-acid Aß variant levels in human neuroblastoma SK-N-SH cells. We discovered that a specific apoE4 fragment, apoE4[Δ(166-299)], can promote the cellular uptake of extracellular 40-amino-acid Aß variant and 42-amino-acid Aß variant either generated after amyloid precursor protein transfection or added exogenously. A longer length fragment, apoE4[Δ(186-299)], or full-length apoE4 failed to elicit this effect. ApoE4[Δ(166-299)] effected a 20% reduction of cellular sphingomyelin levels, as well as changes in cellular membrane micro-fluidity. Following uptake, approximately 50% of 42-amino-acid Aß variant remained within the cell for at least 24 h, and led to increased formation of reactive oxygen species. Overall, our findings suggest a direct link between two early events in the pathogenesis of AD, apoE4 proteolysis and intraneuronal presence of amyloid beta peptide.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/fisiología , Líquido Intracelular/metabolismo , Fragmentos de Péptidos/fisiología , Línea Celular , Línea Celular Tumoral , Humanos , Fragmentos de Péptidos/metabolismo , Isoformas de Proteínas/fisiología
18.
J Lipid Res ; 51(6): 1513-23, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20124555

RESUMEN

We previously observed that treatment of mice with a dominant negative form of cJun (dn-cJun) increased the expression of genes involved in lipid metabolism and modulated the expression of nine microRNAs (miR). To investigate the potential effect of these miRs on the expression of the genes of lipid metabolism, we performed studies in cultured HepG2 cells. Transfection of HepG2 cells with sense or antisense miR-370 or miR-122 upregulated and downregulated, respectively, the transcription factor sterol-regulatory element binding protein 1c (SREBP-1c) and the enzymes diacylglycerol acyltransferase-2 (DGAT2), fatty acid synthase (FAS), and acyl-CoA carboxylase 1 (ACC1) that regulate fatty acid and triglyceride biosynthesis. The other seven miRs identified by the miR array screening did not affect the expression of lipogenic genes. miR-370 upregulated the expression of miR-122. Furthermore, the effect of miR-370 on the expression of the lipogenic genes was abolished by antisense miR-122. miR-370 targets the 3' untranslated region (UTR) of Cpt1alpha, and it downregulated the expression of the carnitine palmitoyl transferase 1alpha (Cpt1alpha) gene as well as the rate of beta oxidation. Our data suggest that miR-370 acting via miR-122 may have a causative role in the accumulation of hepatic triglycerides by modulating initially the expression of SREBP-1c, DGAT2, and Cpt1alpha and, subsequently, the expression of other genes that affect lipid metabolism.


Asunto(s)
Carnitina O-Palmitoiltransferasa/genética , Regulación de la Expresión Génica/genética , Metabolismo de los Lípidos/genética , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Acetiltransferasas/genética , Adenoviridae/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Biología Computacional , Diacilglicerol O-Acetiltransferasa/genética , Regulación hacia Abajo/genética , Espacio Extracelular/metabolismo , Ácidos Grasos/metabolismo , Genes jun/genética , Humanos , Lipogénesis/genética , Hígado/metabolismo , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Oxidación-Reducción , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Triglicéridos/metabolismo , Regulación hacia Arriba/genética , Receptor fas/genética
19.
Ann Med ; 40 Suppl 1: 14-28, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18246469

RESUMEN

Using adenovirus-mediated gene transfer in apolipoprotein A-I (apoA-I)-deficient mice, we have established that apoA-I mutations inhibit discrete steps in a pathway that leads to the biogenesis and remodeling of high-density lipoprotein (HDL). To this point, five discrete categories of apoA-I mutants have been characterized that may affect the interactions of apoA-I with ATP-binding cassette superfamily A, member 1 (ABCA1) or lecithin:cholesterol acyl transferase (LCAT) or may influence the plasma phospholipid transfer protein activity or may cause various forms of dyslipidemia. Biogenesis of HDL is not a unique property of apoA-I. Using adenovirus-mediated gene transfer of apoE in apoA-I- or ABCA1-deficient mice, we have established that apolipoprotein E (apoE) also participates in a novel pathway of biogenesis of apoE-containing HDL particles. This process requires the functions of the ABCA1 lipid transporter and LCAT, and it is promoted by substitution of hydrophobic residues in the 261 to 269 region of apoE by Ala. The apoE-containing HDL particles formed in the circulation may have atheroprotective properties. ApoE-containing HDL may also have important biological functions in the brain that confer protection from Alzheimer's disease.


Asunto(s)
Apolipoproteína A-I/metabolismo , Apolipoproteínas E/metabolismo , Lipoproteínas HDL/biosíntesis , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenoviridae/genética , Animales , Apolipoproteína A-I/genética , Técnicas de Transferencia de Gen , Ratones , Ratones Noqueados , Modelos Animales , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo
20.
Biochemistry ; 46(41): 11473-83, 2007 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17887732

RESUMEN

The lipid transporter ATP binding cassette transporter A1 (ABCA1) promotes the efflux of cellular phospholipids and cholesterol to lipid-free apolipoprotein A-I and thus initiates the biogenesis of high-density lipoprotein (HDL). The expression of the ABCA1 gene is controlled, coordinately with other genes of HDL metabolism, by liver X receptor/retinoid X receptor (LXR/RXR) heterodimers and their ligands oxysterols and retinoids. In the present study, we show that the oxysterol/retinoid-induced transcription of the ABCA1 gene is modulated by the ubiquitous transcription factor Sp1 that binds to the proximal ABCA1 promoter, adjacently to the LXR/RXR responsive element. The response of the ABCA1 gene to oxysterols/retinoids as well as the ligand-inducible recruitment of Sp1 and RXRalpha/LXRalpha heterodimers to the ABCA1 promoter was blocked by mithramycin A, a well-known Sp1 inhibitor. Using SL2 cells which lack endogenous Sp1, we showed that activation of the ABCA1 promoter by LXRalpha/RXRalpha heterodimers and their ligands requires Sp1. Functional interactions between these factors were demonstrated using the GAL4 transactivation system. Using both in vitro and in vivo assays, we show that physical interactions between Sp1 and LXRalpha require the N-terminal region of LXRalpha, which includes the AF1 and DNA binding domains and two different domains of Sp1: the transactivation domain B and the DNA binding domain. Overall, the present study revealed a novel mechanism of regulation of the human ABCA1 transporter which involves synergistic interactions between oxysterol/retinoid-inducible hormone nuclear receptors and the transcription factor Sp1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores X Retinoide/química , Receptores X Retinoide/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Biomarcadores de Tumor/química , Biomarcadores de Tumor/metabolismo , Biotinilación , Cartilla de ADN , Regulación de la Expresión Génica , Humanos , Receptores X del Hígado , Receptores Nucleares Huérfanos , Plásmidos , Regiones Promotoras Genéticas , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA