Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(10): 1851-1866, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589135

RESUMEN

BACKGROUND: High levels of Lp(a) (lipoprotein(a)) are associated with multiple forms of cardiovascular disease. Lp(a) consists of an apoB100-containing particle attached to the plasminogen homologue apo(a). The pathways for Lp(a) clearance are not well understood. We previously discovered that the plasminogen receptor PlgRKT (plasminogen receptor with a C-terminal lysine) promoted Lp(a) uptake in liver cells. Here, we aimed to further define the role of PlgRKT and to investigate the role of 2 other plasminogen receptors, annexin A2 and S100A10 (S100 calcium-binding protein A10) in the endocytosis of Lp(a). METHODS: Human hepatocellular carcinoma (HepG2) cells and haploid human fibroblast-like (HAP1) cells were used for overexpression and knockout of plasminogen receptors. The uptake of Lp(a), LDL (low-density lipoprotein), apo(a), and endocytic cargos was visualized and quantified by confocal microscopy and Western blotting. RESULTS: The uptake of both Lp(a) and apo(a), but not LDL, was significantly increased in HepG2 and HAP1 cells overexpressing PlgRKT, annexin A2, or S100A10. Conversely, Lp(a) and apo(a), but not LDL, uptake was significantly reduced in HAP1 cells in which PlgRKT and S100A10 were knocked out. Surface binding studies in HepG2 cells showed that overexpression of PlgRKT, but not annexin A2 or S100A10, increased Lp(a) and apo(a) plasma membrane binding. Annexin A2 and S100A10, on the other hand, appeared to regulate macropinocytosis with both proteins significantly increasing the uptake of the macropinocytosis marker dextran when overexpressed in HepG2 and HAP1 cells and knockout of S100A10 significantly reducing dextran uptake. Bringing these observations together, we tested the effect of a PI3K (phosphoinositide-3-kinase) inhibitor, known to inhibit macropinocytosis, on Lp(a) uptake. Results showed a concentration-dependent reduction confirming that Lp(a) uptake was indeed mediated by macropinocytosis. CONCLUSIONS: These findings uncover a novel pathway for Lp(a) endocytosis involving multiple plasminogen receptors that enhance surface binding and stimulate macropinocytosis of Lp(a). Although the findings were produced in cell culture models that have limitations, they could have clinical relevance since drugs that inhibit macropinocytosis are in clinical use, that is, the PI3K inhibitors for cancer therapy and some antidepressant compounds.


Asunto(s)
Anexina A2 , Plasminógeno , Humanos , Plasminógeno/química , Plasminógeno/metabolismo , Lipoproteína(a)/metabolismo , Anexina A2/genética , Dextranos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Portadoras , Apolipoproteínas A/metabolismo
2.
J Lipid Res ; 61(3): 432-444, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31806727

RESUMEN

Plasma lipoprotein (a) [Lp(a)] levels are largely determined by variation in the LPA gene, which codes for apo(a). Genome-wide association studies (GWASs) have identified nonsynonymous variants in LPA that associate with low Lp(a) levels, although their effect on apo(a) function is unknown. We investigated two such variants, R990Q and R1771C, which were present in four null Lp(a) individuals, for structural and functional effects. Sequence alignments showed the R990 and R1771 residues to be highly conserved and homologous to each other and to residues associated with plasminogen deficiency. Structural modeling showed both residues to make several polar contacts with neighboring residues that would be ablated on substitution. Recombinant expression of the WT and R1771C apo(a) in liver and kidney cells showed an abundance of an immature form for both apo(a) proteins. A mature form of apo(a) was only seen with the WT protein. Imaging of the recombinant apo(a) proteins in conjunction with markers of the secretory pathway indicated a poor transit of R1771C into the Golgi. Furthermore, the R1771C mutant displayed a glycosylation pattern consistent with ER, but not Golgi, glycosylation. We conclude that R1771 and the equivalent R990 residue facilitate correct folding of the apo(a) kringle structure and mutations at these positions prevent the proper folding required for full maturation and secretion. To our knowledge, this is the first example of nonsynonymous variants in LPA being causative of a null Lp(a) phenotype.


Asunto(s)
Apoproteína(a)/genética , Lipoproteína(a)/genética , Plasminógeno/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Anciano , Alelos , Línea Celular Tumoral , Estudios de Cohortes , Humanos , Lipoproteína(a)/sangre , Masculino , Persona de Mediana Edad , Mutación , Plasminógeno/deficiencia
3.
Am J Physiol Regul Integr Comp Physiol ; 319(3): R376-R386, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32755464

RESUMEN

The present study was conducted to understand key biochemical, physiological, and molecular changes associated with ovarian growth and with lipid transfer and/or accumulation into the ovary during oogenesis in captive beluga sturgeon. Plasma levels of triacylglycerides, cholesterol, phospholipid, and sex steroid hormones were determined and all were found to increase notably throughout development from the perinucleolar to the tertiary yolk stage. Using fast protein liquid chromatography, we recognized three major lipoprotein peaks in chromatograms from all samples. These peaks were characterized as containing very low-density lipoprotein (Vldl), low-density lipoprotein/high-density lipoprotein (Ldl/Hdl), and plasma proteins. While Ldl/Hdl represented the most abundant lipoprotein fraction, the relative abundance of different lipoprotein classes did not change with the stage of oogenesis. Eluted lipoproteins were separated using sodium dodecyl-sulfate polyacrylamide gel electrophoresis and sequenced. The peptide sequence spectra for 66-kDa, 205-kDa, 29-kDa, and 70-kDa bands matched with albumin, vitellogenin (Vtg) AB2b, immunoglobulin light-chain precursor, and immunoglobulin heavy-chain, respectively. The large amount of albumin in the plasma protein peak and the confined presence of Vtg AB2b to within Ldl/Hdl reinforce the lipoprotein classification. Lastly, transcript levels of genes encoding ovarian lipoprotein lipase (lpl), apolipoprotein E (apoe), very low-density lipoprotein receptors (vldlr), and low-density lipoprotein receptor-related protein 8-like (lrp8) were estimated using quantitative RT-PCR. The high mRNA levels of lpl, apoe, and lipoprotein receptors vldlr and lrp8 in previtellogenic females suggest that sturgeon oocytes need to be prepared to accept and traffic Vtg and lipids internally, before the start of vitellogenesis.


Asunto(s)
Lipoproteína Lipasa/metabolismo , Lipoproteínas VLDL/sangre , Ovario/crecimiento & desarrollo , Triglicéridos/metabolismo , Animales , Apolipoproteínas E/metabolismo , Colesterol/sangre , Femenino , Lipoproteínas LDL/metabolismo , Ovario/metabolismo
4.
Circ Res ; 120(7): 1091-1102, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28003220

RESUMEN

RATIONALE: Lipoprotein(a) [Lp(a)] is a low-density lipoprotein-like lipoprotein and important cardiovascular risk factor whose cognate receptor and intracellular fate remains unknown. OBJECTIVE: Our study aimed to determine the intracellular trafficking pathway for Lp(a) and the receptor responsible for its uptake in liver cells. METHODS AND RESULTS: Human hepatoma cells were treated with Lp(a) purified from human plasma and Lp(a) uptake studied using Western blot analysis and intracellular localization of Lp(a) by confocal microscopy. Lp(a) was maximally internalized by 2 hours and was detected by an antiapo(a) antibody to be localized to Rab5-positive early endosomes, the trans-Golgi network, and subsequently Rab11-positive recycling endosomes. In human hepatoma cells, the apo(a) component from the internalized Lp(a) was resecreted back into the cellular media, whereas the low-density lipoprotein component was localized to the lysosomal compartment. Lp(a) internalization was reduced 0.35-fold in HAP1 and 0.33-fold in human hepatoma cells in which the plasminogen receptor (KT) was knocked out. Conversely, Lp(a) internalization was enhanced 2-fold in HAP1 and 1.6-fold in human hepatoma cells in which plasminogen receptor (KT) was overexpressed, showing for the first time the role of a specific plasminogen receptor in Lp(a) uptake. CONCLUSIONS: The novel findings that Lp(a) is internalized by the plasminogen receptor, plasminogen receptor (KT), and the apo(a) component is recycled may have important implications for the catabolism and function of Lp(a).


Asunto(s)
Apolipoproteínas A/metabolismo , Endocitosis , Endosomas/metabolismo , Receptores de Superficie Celular/metabolismo , Células Hep G2 , Humanos , Lisosomas/metabolismo , Transporte de Proteínas
5.
Am J Physiol Regul Integr Comp Physiol ; 310(5): R422-31, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26764051

RESUMEN

The importance of androgens (especially 11-ketotestosterone) during previtellogenesis in eels is well established. In wild pubertal migrants, circulating 11-ketotestosterone levels correlate with a number of morphological and molecular changes. Here, we test the prediction that this correlation represents a causal relationship by artificially raising the levels of circulating 11-ketotestosterone in prepubertal nonmigratory female and pubertal, migratory male short-finned eels (Anguilla australis) using sustained-release hormone implants. In females, increases in hepatosomatic index and transcript copy numbers of hepatic apolipoprotein B and microsomal triacylglyceride transfer protein indicated increased repackaging of endogenously sourced triacylglycerides. These changes in liver measures were reflected in increased concentrations of serum triacylglycerides. However, despite a small increase in gonadosomatic index, ovarian lipoprotein receptor transcript abundances were not affected by 11-ketotestosterone. Interestingly, no such changes in hepatic gene expression were detected in a dose-response experiment using males. We propose that the androgens are inducing the observed changes in previtellogenic females, although it remains unclear to what extent these effects are direct or indirect.


Asunto(s)
Andrógenos/administración & dosificación , Anguilla/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Testosterona/análogos & derivados , Triglicéridos/sangre , Andrógenos/sangre , Animales , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Colesterol/sangre , Implantes de Medicamentos , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Hígado/metabolismo , Masculino , ARN Mensajero/metabolismo , Factores Sexuales , Maduración Sexual , Testosterona/administración & dosificación , Testosterona/sangre , Factores de Tiempo , Regulación hacia Arriba
6.
J Lipid Res ; 56(7): 1318-28, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25852127

RESUMEN

Elevated levels of lipoprotein (a) [Lp(a)] are a well-established risk factor for developing CVD. While Lp(a) levels are thought to be independent of other plasma lipoproteins, some trials have reported a positive association between Lp(a) and HDL. Whether Lp(a) has a direct effect on HDL is not known. Here we investigated to determine whether Lp(a) had any effect on the ABCA1 pathway of HDL production in liver cells. Incubation of HepG2 cells with Lp(a) upregulated the PPARγ protein by 1.7-fold and the liver X receptor α protein by 3-fold. This was accompanied by a 1.8-fold increase in ABCA1 protein and a 1.5-fold increase in cholesterol efflux onto apoA1. We showed that Lp(a) was internalized by HepG2 cells, however, the ABCA1 response to Lp(a) was mediated by the selective uptake of oxidized phospholipids (oxPLs) from Lp(a) via the scavenger receptor-B1 and not by Lp(a) internalization per se. We conclude that there is a biological connection between Lp(a) and HDL through the ability of Lp(a)'s oxPLs to upregulate HDL biosynthesis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Antígenos CD36/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Lipoproteína(a)/farmacología , Fosfolípidos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Células Hep G2 , Humanos , Oxidación-Reducción/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
7.
Am J Physiol Regul Integr Comp Physiol ; 308(11): R935-44, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810387

RESUMEN

During certain stages in an animal's life cycle, energy requirements may exceed energy intake from the diet. The spawning migration of temperate eels is a textbook example of negative energy balance, forcing these fish to rely on stored fats (triacylglycerides) to provide their muscles with energy for swimming and their growing oocytes with the nutrients needed to develop and support healthy offspring. We predicted broad implications of this great need for endogenous triacylglycerides in terms of their packaging, transport, and ovarian uptake. To test this, serum lipid concentrations and transcript abundances of intestinal and hepatic triacylglyceride packagers and ovarian triacylglyceride modifiers and receivers were investigated throughout previtellogenesis (feeding phase) and into early vitellogenesis (fasting phase) in short-finned eels. A switch from exogenous to endogenous triacylglyceride packaging was seen as the liver upregulated transcript levels of apolipoprotein B and microsomal triacylglyceride transport protein and downregulated those of apolipoprotein E and lipoprotein lipase. In the intestine, the reverse response was observed. Furthermore, ovarian transcript abundances of triacylglyceride modifiers and receivers increased (apolipoprotein E, lipoprotein lipase, and vitellogenin receptor), indicative of increased triacylglyceride uptake during previtellogenesis. We propose that increased hepatic apolipoprotein B production is a conserved vertebrate response to prolonged periods of negative energy balance.


Asunto(s)
Anguilas/metabolismo , Metabolismo Energético , Oocitos/metabolismo , Oogénesis , Ovario/metabolismo , Triglicéridos/metabolismo , Adaptación Fisiológica , Animales , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Anguilas/sangre , Anguilas/genética , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Hígado/metabolismo , ARN Mensajero/metabolismo , Triglicéridos/sangre , Vitelogénesis
8.
J Lipid Res ; 54(1): 55-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23087442

RESUMEN

Mutations in the ATP-binding cassette transporter A1 (ABCA1) are a major cause of decreased HDL cholesterol (HDL-C), which infers an increased risk of cardiovascular disease (CVD). Many ABCA1 mutants show impaired localization to the plasma membrane. The aim of this study was to investigate whether the chemical chaperone, sodium 4-phenylbutyrate (4-PBA) could improve cellular localization and function of ABCA1 mutants. Nine different ABCA1 mutants (p.A594T, p.I659V, p.R1068H, p.T1512M, p.Y1767D, p.N1800H, p.R2004K, p.A2028V, p.Q2239N) expressed in HEK293 cells, displaying different degrees of mislocalization to the plasma membrane and discrete impacts on cholesterol efflux, were subject to treatment with 4-PBA. Treatment restored localization to the plasma membrane and increased cholesterol efflux function for the majority of mutants. Treatment with 4-PBA also increased ABCA1 protein expression in all transfected cell lines. In fibroblast cells obtained from low HDL-C subjects expressing two of the ABCA1 mutants (p.R1068H and p.N1800H), 4-PBA increased cholesterol efflux without any increase in ABCA1 expression. Our study is the first to investigate the effect of the chemical chaperone, 4-PBA on ABCA1 and shows that it is capable of restoring plasma membrane localization and enhancing the cholesterol efflux function of mutant ABCA1s both in vitro and ex vivo. These results suggest 4-PBA may warrant further investigation as a potential therapy for increasing cholesterol efflux and HDL-C levels.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Mutación Missense/efectos de los fármacos , Fenilbutiratos/farmacología , Transportador 1 de Casete de Unión a ATP , HDL-Colesterol/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HEK293 , Humanos , Transporte de Proteínas/efectos de los fármacos
9.
Biochem Biophys Res Commun ; 409(3): 400-5, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21575609

RESUMEN

The ATP binding cassette transporter (ABCA1) A1 is a key determinant of circulating high density lipoprotein cholesterol (HDL-C) levels. Mutations in ABCA1 are a major genetic contributor to low HDL-C levels within the general population. Following the finding of three different ABCA1 mutations, p.C978fsX988, p.T1512M and p.N1800H in a subject with hypoalphalipoproteinemia, we aimed to establish whether the p.C978fsX988 truncation exerted a dominant negative effect on the full-length ABCA1 alleles within family members as has been reported for other ABCA1 truncations. Characterisation of the p.C978fsX988 mutant in transfected HEK 293 cells showed it to be expressed as a GFP fusion protein but lacking in cholesterol efflux function. This was in keeping with results from cholesterol efflux assays in the fibroblasts of p.C978fsX988 carriers which also showed impaired efflux. Allele- specific quantification of p.C978fsX988 mRNA and analysis of ABCA1 protein levels in the fibroblasts of p.C978fsX988 heterozygotes showed negligible levels of mRNA and protein expression. There was no evidence of a dominant negative effect on wildtype or p.N1800H protein levels. We conclude that in the case of the p.C978fsX988 truncated mutant a lack of expression precludes it from having a dominant negative effect.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Enfermedad de Tangier/genética , Transportador 1 de Casete de Unión a ATP , Anciano , Colesterol/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Mutación , Linaje , Enfermedad de Tangier/metabolismo
10.
J Lipid Res ; 51(11): 3370-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20667818

RESUMEN

We present here a gel-based method for rapid purification of apolipoprotein A-I (apoA-I) from small volumes of human plasma. After isolation of high density lipoprotein from plasma, the apoA-I protein was separated by electrophoresis and the apoA-I band excised from the gel. The apoA-I was then eluted from the gel strip, concentrated, and delipidated ready for use. The structure and function of the gel-purified apoA-I protein was compared against apoA-I purified by the traditional size-exclusion chromatography method. The α-helical content of the gel-purified apoA-I as determined by circular dichroism was similar to chromatography-purified apoA-I. The functional activity of gel-purified apoA-I, as determined by cholesterol efflux assays in primary human fibroblasts and RAW264.7 macrophages, was also comparable with chromatography-purified apoA-I. This method is a valid alternative for apoA-I purification with some advantages over traditional chromatography purification including a much reduced plasma volume requirement, less time and cost, and a higher percentage protein recovery. The method is particularly suitable for applications requiring the purification of apoA-I from multiple human or animal samples of interest.


Asunto(s)
Apolipoproteína A-I/sangre , Apolipoproteína A-I/aislamiento & purificación , Fraccionamiento Químico/métodos , Geles , Humanos , Indicadores y Reactivos/química , Laboratorios
11.
PLoS One ; 15(2): e0228415, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32084149

RESUMEN

Ribose-cysteine is a synthetic compound designed to increase glutathione (GSH) synthesis. Low levels of GSH and the GSH-dependent enzyme, glutathione peroxidase (GPx), is associated with cardiovascular disease (CVD) in both mice and humans. Here we investigate the effect of ribose-cysteine on GSH, GPx, oxidised lipids and atherosclerosis development in apolipoprotein E-deficient (apoE-/-) mice. Female 12-week old apoE-/- mice (n = 15) were treated with 4-5 mg/day ribose-cysteine in drinking water for 8 weeks or left untreated. Blood and livers were assessed for GSH, GPx activity and 8-isoprostanes. Plasma alanine transferase (ALT) and lipid levels were measured. Aortae were quantified for atherosclerotic lesion area in the aortic sinus and brachiocephalic arch and 8-isoprostanes measured. Ribose-cysteine treatment significantly reduced ALT levels (p<0.0005) in the apoE-/- mice. Treatment promoted a significant increase in GSH concentrations in the liver (p<0.05) and significantly increased GPx activity in the liver and erythrocytes of apoE-/-mice (p<0.005). The level of 8-isoprostanes were significantly reduced in the livers and arteries of apoE-/- mice (p<0.05 and p<0.0005, respectively). Ribose-cysteine treatment showed a significant decrease in total and low density lipoprotein (LDL) cholesterol (p<0.05) with no effect on other plasma lipids with the LDL reduction likely through upregulation of scavenger receptor-B1 (SR-B1). Ribose-cysteine treatment significantly reduced atherosclerotic lesion area by >50% in both the aortic sinus and brachiocephalic branch (p<0.05). Ribose-cysteine promotes a significant GSH-based antioxidant effect in multiple tissues as well as an LDL-lowering response. These effects are accompanied by a marked reduction in atherosclerosis suggesting that ribose-cysteine might increase protection against CVD.


Asunto(s)
Antioxidantes/administración & dosificación , Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Cisteína/administración & dosificación , Sustancias Protectoras/administración & dosificación , Ribosa/administración & dosificación , Animales , Antioxidantes/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Cisteína/metabolismo , Femenino , Lípidos/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Oxidación-Reducción , Sustancias Protectoras/metabolismo , Ribosa/metabolismo
12.
Epigenetics ; 15(9): 949-958, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32237968

RESUMEN

Changes in whole blood DNA methylation levels at several CpG sites have been associated with circulating blood lipids, specifically high-density lipoprotein and triglycerides. This study performs a discovery and validation epigenome-wide association study (EWAS) for circulating lipoprotein(a) [Lp(a)], an independent risk factor for cardiovascular diseases. Whole-blood DNA methylation profiles were assessed in a cohort of 1020 elderly individuals using the Illumina EPIC array and independent validation in 359 elderly males using the Illumina 450 k array. Plasma Lp(a) was measured using an apolipoprotein(a)-size-independent ELISA. Epigenome-wide rank regression analysis identified and validated a single CpG site, cg17028067 located in intron 1 of the LPA gene, that was significantly associated with plasma Lp(a) levels after correction for multiple testing. Genotyping of the site identified a relatively uncommon SNP (rs76735376, MAF <0.02) at the CpG site that largely explained the observed methylation effect. Rs76735376 is an expression quantitative trait loci for the LPA gene and could affect expression by altering enhancer activity. This EWAS for plasma Lp(a) identified a single CpG site within LPA. This association is due to an uncommon, but highly effective genetic variant, which was not in significant linkage disequilibrium with other variants known to influence Lp(a) levels or apo(a) isoform size. This study highlights the utility of CpG site methylation to identify potentially important genetic associations that would not be readily apparent in a comparable size genetic association study.


Asunto(s)
Metilación de ADN , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Anciano , Islas de CpG , Epigenoma , Femenino , Humanos , Lipoproteína(a)/sangre , Masculino , Persona de Mediana Edad
13.
Arterioscler Thromb Vasc Biol ; 28(11): 2071-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18617650

RESUMEN

OBJECTIVE: Whereas the anti-inflammatory effects of high-density lipoprotein (HDL) on endothelial cells are well described, such effects on monocytes are less studied. METHODS AND RESULTS: Human monocytes were isolated from whole blood followed by assessment of CD11b activation/expression and cell adhesion under shear-flow. HDL caused a dose-dependent reduction in the activation of CD11b induced by PMA or receptor-dependent agonists. The constituent of HDL responsible for the antiinflammatory effects on CD11b activation was found to be apolipoprotein A-I (apoA-I). Cyclodextrin, but not cyclodextrin/cholesterol complex, also inhibited PMA-induced CD11b activation implicating cholesterol efflux as the main mechanism. This was further confirmed with the demonstration that cholesterol content of lipid rafts diminished after treatment with the cholesterol acceptors. Blocking ABCA1 with an anti-ABCA1 antibody abolished the effect of apoA-I. Furthermore, monocytes derived from a Tangier disease patient definitively confirmed the requirement of ABCA1 in apoA-I-mediated CD11b inhibition. The antiinflammatory effects of apoA-I were also observed in functional models including cell adhesion to an endothelial cell monolayer, monocytic spreading under shear flow, and transmigration. CONCLUSIONS: HDL and apoA-I exhibit an antiinflammatory effect on human monocytes by inhibiting activation of CD11b. ApoA-I acts through ABCA1, whereas HDL may act through several receptors.


Asunto(s)
Apolipoproteína A-I/metabolismo , Aterosclerosis/prevención & control , Inflamación/prevención & control , Lipoproteínas HDL/metabolismo , Monocitos/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Actinas/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Antígeno CD11b/metabolismo , Adhesión Celular , Forma de la Célula , Células Cultivadas , Colesterol/deficiencia , Colesterol/metabolismo , Ciclodextrinas/farmacología , Células Endoteliales/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Microdominios de Membrana/metabolismo , Monocitos/efectos de los fármacos , Monocitos/patología , Adhesividad Plaquetaria , Receptores Depuradores de Clase B , Estrés Mecánico , Enfermedad de Tangier/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factores de Tiempo
14.
Pathology ; 51(2): 155-164, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30595508

RESUMEN

Lipoprotein(a) [Lp(a)] is an apolipoprotein B (apoB)-containing plasma lipoprotein similar in structure to low-density lipoprotein (LDL). Lp(a) is more complex than LDL due to the presence of apolipoprotein(a) [apo(a)], a large glycoprotein sharing extensive homology with plasminogen, which confers some unique properties onto Lp(a) particles. ApoB and apo(a) are essential for the assembly and catabolism of Lp(a); however, other proteins associated with the particle may modify its metabolism. Lp(a) specifically carries a cargo of oxidised phospholipids (OxPL) bound to apo(a) which stimulates many proinflammatory pathways in cells of the arterial wall, a key property underlying its pathogenicity and association with cardiovascular disease (CVD). While the liver and kidney are the major tissues implicated in Lp(a) clearance, the pathways for Lp(a) uptake appear to be complex and are still under investigation. Biochemical studies have revealed an exceptional array of receptors that associate with Lp(a) either via its apoB, apo(a), or OxPL components. These receptors fall into five main categories, namely 'classical' lipoprotein receptors, toll-like and scavenger receptors, lectins, and plasminogen receptors. The roles of these receptors have largely been dissected by genetic manipulation in cells or mice, although their relative physiological importance for removal of Lp(a) from the circulation remains unclear. The LPA gene encoding apo(a) has an overwhelming effect on Lp(a) levels which precludes any clear associations between potential Lp(a) receptor genes and Lp(a) levels in population studies. Targeted approaches and the selection of unique Lp(a) phenotypes within populations has nevertheless allowed for some associations to be made. Few of the proposed Lp(a) receptors can specifically be manipulated with current drugs and, as such, it is not currently clear whether any of these receptors could provide relevant targets for therapeutic manipulation of Lp(a) levels. This review summarises the current status of knowledge about receptor-mediated pathways for Lp(a) catabolism.


Asunto(s)
Apolipoproteínas A/metabolismo , Enfermedades Cardiovasculares/metabolismo , Lipoproteína(a)/metabolismo , Receptores de Lipoproteína/metabolismo , Receptores Depuradores/metabolismo , Animales , Apolipoproteínas A/genética , Estudio de Asociación del Genoma Completo , Humanos , Riñón/metabolismo , Lectinas/metabolismo , Lipoproteína(a)/genética , Hígado/metabolismo , Ratones , Oxidación-Reducción , Fosfolípidos/metabolismo , Plasminógeno/metabolismo , Receptores de Lipoproteína/genética
15.
Biomed Res Int ; 2018: 4963942, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30596094

RESUMEN

BACKGROUND: Mouse models of hypercholesterolaemia have been used to identify arterial proteins involved in atherosclerosis. As the liver is extremely sensitive to dyslipidemia, one might expect major changes in the abundance of liver proteins in these models even before atherosclerosis develops. METHODS: Lipid levels were measured and a proteomic approach was used to quantify proteins in the livers of mice with an elevated low-density lipoprotein (LDL) and the presence of lipoprotein(a) [Lp(a)] but no atherosclerosis. RESULTS: The livers of Lp(a) mice showed an increased triglyceride but reduced phospholipid and oxidised lipid content. Two-dimensional gel electrophoresis and mass spectrometry analysis identified 24 liver proteins with significantly increased abundance in Lp(a) mice (P<0.05). A bioinformatic analysis of the 24 proteins showed the major effect was that of an enhanced antioxidant and lipid efflux response with significant increases in antioxidant (Park7, Gpx1, Prdx6, and Sod1) and lipid metabolism proteins (Fabp4, Acaa2, apoA4, and ApoA1). Interestingly, human liver cells treated with Lp(a) showed significant increases in Gpx1 and Prdx6 but not Sod1 or Park7. CONCLUSIONS: The presence of human LDL and Lp(a) in mice promotes an enhanced flux of lipids into the liver which elicits an antioxidant and lipid export response before the onset of atherosclerosis. The antioxidant response can be reproduced in human liver cells treated with Lp(a).


Asunto(s)
Metabolismo de los Lípidos/fisiología , Lipoproteína(a)/metabolismo , Hígado/metabolismo , Estrés Oxidativo/fisiología , Animales , Antioxidantes/metabolismo , Aterosclerosis/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Dislipidemias/metabolismo , Femenino , Células Hep G2 , Humanos , Hipercolesterolemia/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas/metabolismo , Proteómica/métodos
16.
Atherosclerosis ; 187(2): 393-400, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16225879

RESUMEN

Mutations in the ATP-binding cassette A1 (ABCA1) transporter cause the high-density lipoprotein (HDL) deficiency syndromes of Tangier disease and familial hypoalphalipoproteinemia (FHA). Between individuals carrying ABCA1 mutations, the expression of FHA can be highly variable. Using denaturing HPLC (dHPLC) and direct promoter sequencing we screened the ABCA1 gene of a family with Tangier disease and variable expression of FHA. A new mutation (R1068H) within the first ATP-binding domain was identified in homozygous form in the Tangier disease individual and was present in several family members. Haplotyping of both 1068H alleles in the proband showed homozygosity in the coding region, however, the maternal 1068H allele had three single nucleotide polymorphisms (SNPs) in the promoter previously reported to be associated with reduced ABCA1 expression and HDL levels. An analysis of HDL levels based on 1068H allele haplotype showed the paternal 1068H heterozygotes to have the expected low HDL levels (0.67+/-0.16mmol/L), while maternal 1068H heterozygotes showed normal HDL levels (1.18+/-0.14mmol/L). Haplotype analysis of the wildtype allele amongst heterozygotes showed no haplotype that was common to the paternal or maternal side. We propose that the paternal 1068H ABCA1 allele causes a negative effect on the function of the wildtype allele and is associated with low HDL levels. In contrast, the maternal 1068H allele has less effect and is associated with a relatively normal HDL level. We conclude that haplotypes of mutant ABCA1 alleles may contribute to the phenotypic variance shown between FHA individuals.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Regiones Promotoras Genéticas/genética , Enfermedad de Tangier/genética , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/química , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , HDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/genética , Salud de la Familia , Femenino , Pruebas Genéticas , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Estructura Terciaria de Proteína , Enfermedad de Tangier/sangre
17.
PLoS One ; 11(1): e0148049, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26807857

RESUMEN

Cardiovascular complications have emerged as a major concern for cancer patients. Many chemotherapy agents are cardiotoxic and some appear to also alter lipid profiles, although the mechanism for this is unknown. We studied plasma lipid levels in 12 breast cancer patients throughout their chemotherapy. Patients received either four cycles of doxorubicin and cyclophosphamide followed by weekly paclitaxel or three cycles of epirubicin, cyclophosphamide and 5'-fluorouracil followed by three cycles of docetaxel. Patients demonstrated a significant reduction (0.32 mmol/L) in high density lipoprotein cholesterol (HDL-C) and apolipoprotein A1 (apoA1) levels (0.18 g/L) and an elevation in apolipoprotein B (apoB) levels (0.15 g/L) after treatment. Investigation of the individual chemotherapy agents for their effect on genes involved in lipoprotein metabolism in liver cells showed that doxorubicin decreased ATP binding cassette transporter A1 (ABCA1) via a downregulation of the peroxisomal proliferator activated receptor γ (PPARγ) and liver X receptor α (LXRα) transcription factors. In contrast, ABCA1 levels were not affected by cyclophosphamide or paclitaxel. Likewise, apoA1 levels were reduced by doxorubicin and remained unaffected by cyclophosphamide and paclitaxel. Doxorubicin and paclitaxel both increased apoB protein levels and paclitaxel also decreased low density lipoprotein receptor (LDLR) protein levels. These findings correlate with the observed reduction in HDL-C and apoA1 and increase in apoB levels seen in these patients. The unfavourable lipid profiles produced by some chemotherapy agents may be detrimental in the longer term to cancer patients, especially those already at risk of cardiovascular disease (CVD). This knowledge may be useful in tailoring effective follow-up care plans for cancer survivors.


Asunto(s)
Antineoplásicos/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Transportadoras de Casetes de Unión a ATP/sangre , Transportadoras de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Antineoplásicos/uso terapéutico , Apolipoproteína A-I/sangre , Apolipoproteína A-I/metabolismo , Apolipoproteínas B/sangre , Apolipoproteínas B/metabolismo , Neoplasias de la Mama/sangre , Neoplasias de la Mama/metabolismo , HDL-Colesterol/sangre , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Epirrubicina/efectos adversos , Epirrubicina/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , PPAR gamma/sangre , PPAR gamma/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 23(3): 502-7, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12615683

RESUMEN

OBJECTIVE: We previously reported that human apolipoprotein B100 (apoB) amino acids 4330-4397 were important for the initial noncovalent binding to apolipoprotein(a) [apo(a)] that facilitates lipoprotein(a) [Lp(a)] assembly. In this study, we aimed to further define the apoB sequences within the 4330-4397 region that were important for the noncovalent binding to apo(a). METHODS AND RESULTS: Alignment of the human apoB4330-4397 sequence with mouse apoB, which also noncovalently binds apo(a), revealed stretches of similar sequence, including a lysine-rich sequence spanning apoB amino acids 4372-4392. Structural analysis of the apoB4372-4392 sequence using the WHEEL program predicted an amphipathic alpha-helix. Circular dichroism studies of a synthetic peptide spanning human apoB amino acids 4372-4392, both in the absence and presence of dimyristoylphosphatidylcholine, confirmed the alpha-helical nature of the sequence. We tested the ability of the apoB4372-4392 peptide to bind to apo(a) and found that the peptide bound to apo(a) with high affinity but not to Lp(a). The apoB4372-4392 peptide inhibited Lp(a) assembly in Lp(a) formation assays far more effectively than the lysine analogue, epsilon-amino-n-caproic acid (IC50=40 micromol/L versus 10 mmol/L, respectively). Incorporation of the apoB4372-4392 peptide onto dimyristoylphosphatidylcholine vesicles yielded an even more effective inhibitor (IC50=4 micromol/L). CONCLUSIONS: Our study shows that the apoB4372-4392 sequence mediates the initial noncovalent binding to apo(a) and has demonstrated that the apoB4372-4392 peptide is a novel and effective inhibitor of Lp(a) assembly.


Asunto(s)
Apolipoproteínas B/farmacología , Lipoproteína(a)/antagonistas & inhibidores , Lipoproteína(a)/biosíntesis , Fragmentos de Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Apolipoproteínas B/química , Apolipoproteínas B/metabolismo , Dicroismo Circular , Humanos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo
20.
Atherosclerosis ; 237(2): 725-33, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25463112

RESUMEN

OBJECTIVE: D-ribose-L-cysteine (ribose-cysteine) is a cysteine analogue designed to increase the synthesis of glutathione (GSH). GSH is a cofactor for glutathione peroxidase (GPx), the redox enzyme that catalyses the reduction of lipid peroxides. A low GPx activity and increased oxidised lipids are associated with the development of cardiovascular disease (CVD). Here we aimed to investigate the effect of ribose-cysteine supplementation on GSH, GPx, lipid oxidation products and plasma lipids in vivo. METHODS: Human lipoprotein(a) [Lp(a)] transgenic mice were treated with 4 mg/day ribose-cysteine (0.16 g/kg body weight) for 8 weeks. Livers and blood were harvested from treated and untreated controls (n = 9 per group) and GSH concentrations, GPx activity, thiobarbituric acid reactive substances (TBARS), 8-isoprostanes and plasma lipid concentrations were measured. RESULTS: Ribose-cysteine increased GSH concentrations in the liver and plasma (P < 0.05). GPx activity was increased in both liver (1.7 fold, P < 0.01) and erythrocytes (3.5 fold, P < 0.05). TBARS concentrations in the liver, plasma and aortae were significantly reduced with ribose-cysteine (P < 0.01, P < 0.0005 and P < 0.01, respectively) as were the concentrations of 8-isoprostanes in the liver and aortae (P < 0.0005, P < 0.01, respectively). Ribose-cysteine treated mice showed significant decreases in LDL, Lp(a) and apoB concentrations (P < 0.05, P < 0.01 and P < 0.05, respectively), an effect which was associated with upregulation of the LDL receptor (LDLR). CONCLUSIONS: As ribose-cysteine lowers LDL, Lp(a) and oxidised lipid concentrations, it might be an ideal intervention to increase protection against the development of atherosclerosis.


Asunto(s)
Antioxidantes/química , Cisteína/química , Glutatión/química , Lipoproteína(a)/genética , Lipoproteínas LDL/sangre , Ribosa/química , Animales , Apolipoproteínas B/sangre , Enfermedades Cardiovasculares/metabolismo , Dinoprost/análogos & derivados , Dinoprost/química , Femenino , Humanos , Lípidos/sangre , Lipoproteína(a)/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Oxígeno/química , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA