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










Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 43(10): 1851-1866, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37589135

RESUMO

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.


Assuntos
Anexina A2 , Plasminogênio , Humanos , Plasminogênio/química , Plasminogênio/metabolismo , Lipoproteína(a)/metabolismo , Anexina A2/genética , Dextranos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas de Transporte , Apolipoproteínas A/metabolismo
2.
Am J Physiol Regul Integr Comp Physiol ; 319(3): R376-R386, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755464

RESUMO

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.


Assuntos
Lipase Lipoproteica/metabolismo , Lipoproteínas VLDL/sangue , Ovário/crescimento & desenvolvimento , Triglicerídeos/metabolismo , Animais , Apolipoproteínas E/metabolismo , Colesterol/sangue , Feminino , Lipoproteínas LDL/metabolismo , Ovário/metabolismo
3.
Epigenetics ; 15(9): 949-958, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32237968

RESUMO

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.


Assuntos
Metilação de DNA , Lipoproteína(a)/genética , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Idoso , Ilhas de CpG , Epigenoma , Feminino , Humanos , Lipoproteína(a)/sangue , Masculino , Pessoa de Meia-Idade
4.
PLoS One ; 15(2): e0228415, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32084149

RESUMO

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.


Assuntos
Antioxidantes/administração & dosagem , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Cisteína/administração & dosagem , Substâncias Protetoras/administração & dosagem , Ribose/administração & dosagem , Animais , Antioxidantes/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Cisteína/metabolismo , Feminino , Lipídeos/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Oxirredução , Substâncias Protetoras/metabolismo , Ribose/metabolismo
5.
J Lipid Res ; 61(3): 432-444, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31806727

RESUMO

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.


Assuntos
Apoproteína(a)/genética , Lipoproteína(a)/genética , Plasminogênio/genética , Polimorfismo de Nucleotídeo Único/genética , Adulto , Idoso , Alelos , Linhagem Celular Tumoral , Estudos de Coortes , Humanos , Lipoproteína(a)/sangue , Masculino , Pessoa de Meia-Idade , Mutação , Plasminogênio/deficiência
6.
Pathology ; 51(2): 155-164, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30595508

RESUMO

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.


Assuntos
Apolipoproteínas A/metabolismo , Doenças Cardiovasculares/metabolismo , Lipoproteína(a)/metabolismo , Receptores de Lipoproteínas/metabolismo , Receptores Depuradores/metabolismo , Animais , Apolipoproteínas A/genética , Estudo de Associação Genômica Ampla , Humanos , Rim/metabolismo , Lectinas/metabolismo , Lipoproteína(a)/genética , Fígado/metabolismo , Camundongos , Oxirredução , Fosfolipídeos/metabolismo , Plasminogênio/metabolismo , Receptores de Lipoproteínas/genética
7.
Biomed Res Int ; 2018: 4963942, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30596094

RESUMO

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).


Assuntos
Metabolismo dos Lipídeos/fisiologia , Lipoproteína(a)/metabolismo , Fígado/metabolismo , Estresse Oxidativo/fisiologia , Animais , Antioxidantes/metabolismo , Aterosclerose/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dislipidemias/metabolismo , Feminino , Células Hep G2 , Humanos , Hipercolesterolemia/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas/metabolismo , Proteômica/métodos
9.
Circ Res ; 120(7): 1091-1102, 2017 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-28003220

RESUMO

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).


Assuntos
Apolipoproteínas A/metabolismo , Endocitose , Endossomos/metabolismo , Receptores de Superfície Celular/metabolismo , Células Hep G2 , Humanos , Lisossomos/metabolismo , Transporte Proteico
10.
Am J Physiol Regul Integr Comp Physiol ; 310(5): R422-31, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26764051

RESUMO

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.


Assuntos
Androgênios/administração & dosagem , Anguilla/sangue , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Testosterona/análogos & derivados , Triglicerídeos/sangue , Androgênios/sangue , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Colesterol/sangue , Implantes de Medicamento , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Fígado/metabolismo , Masculino , RNA Mensageiro/metabolismo , Fatores Sexuais , Maturidade Sexual , Testosterona/administração & dosagem , Testosterona/sangue , Fatores de Tempo , Regulação para Cima
11.
PLoS One ; 11(1): e0148049, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26807857

RESUMO

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.


Assuntos
Antineoplásicos/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Transportadores de Cassetes de Ligação de ATP/sangue , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Antineoplásicos/uso terapêutico , Apolipoproteína A-I/sangue , Apolipoproteína A-I/metabolismo , Apolipoproteínas B/sangue , Apolipoproteínas B/metabolismo , Neoplasias da Mama/sangue , Neoplasias da Mama/metabolismo , HDL-Colesterol/sangue , Ciclofosfamida/efeitos adversos , Ciclofosfamida/uso terapêutico , Doxorrubicina/efeitos adversos , Doxorrubicina/uso terapêutico , Epirubicina/efeitos adversos , Epirubicina/uso terapêutico , Feminino , Humanos , Pessoa de Meia-Idade , PPAR gama/sangue , PPAR gama/metabolismo
12.
J Lipid Res ; 56(7): 1318-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25852127

RESUMO

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.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/genética , Antígenos CD36/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Lipoproteína(a)/farmacologia , Fosfolipídeos/metabolismo , Regulação para Cima/efeitos dos fármacos , Células Hep G2 , Humanos , Oxirredução/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos
13.
Am J Physiol Regul Integr Comp Physiol ; 308(11): R935-44, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810387

RESUMO

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.


Assuntos
Enguias/metabolismo , Metabolismo Energético , Oócitos/metabolismo , Oogênese , Ovário/metabolismo , Triglicerídeos/metabolismo , Adaptação Fisiológica , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Enguias/sangue , Enguias/genética , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Fígado/metabolismo , RNA Mensageiro/metabolismo , Triglicerídeos/sangue , Vitelogênese
14.
Atherosclerosis ; 237(2): 725-33, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25463112

RESUMO

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.


Assuntos
Antioxidantes/química , Cisteína/química , Glutationa/química , Lipoproteína(a)/genética , Lipoproteínas LDL/sangue , Ribose/química , Animais , Apolipoproteínas B/sangue , Doenças Cardiovasculares/metabolismo , Dinoprosta/análogos & derivados , Dinoprosta/química , Feminino , Humanos , Lipídeos/sangue , Lipoproteína(a)/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Oxigênio/química , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
15.
J Lipid Res ; 54(1): 55-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23087442

RESUMO

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.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Mutação de Sentido Incorreto/efeitos dos fármacos , Fenilbutiratos/farmacologia , Transportador 1 de Cassete de Ligação de ATP , HDL-Colesterol/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Células HEK293 , Humanos , Transporte Proteico/efeitos dos fármacos
16.
Clin Cardiol ; 35(8): 482-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22488248

RESUMO

BACKGROUND: This prospective study investigated the association between preprocedural biomarker levels and incident major adverse cardiac events (MACE) in complex patients undergoing percutaneous coronary intervention (PCI) with sirolimus-eluting stenting. HYPOTHESIS: Lipoprotein(a) (Lp[a]), interleukin-10 (IL-10), and high-sensitivity C-reactive protein (CRP) have long-term prognostic value in patients undergoing PCI. METHODS: Between April 2002 and February 2003, 161 patients were included in the study. Blood was drawn before the procedure, and biomarkers were measured. Patients were followed-up for MACE (death, nonfatal myocardial infarction, and repeat revascularization). Cox proportional hazard models were used to determine risk of MACE for tertiles of biomarkers. Both 1-year and long-term follow-up (median, 6 years; maximum, 8 years) were evaluated. RESULTS: Mean age was 59 years, and 68% were men. During long-term follow-up, 72 MACE occurred (overall crude cumulative incidence: 45% [95% confidence interval (CI): 37%-52%]). Lp(a) was associated with a higher 1-year risk of MACE, with an adjusted hazard ratio (HR) of 3.1 (95% CI: 1.1-8.6) for the highest vs the lowest tertile. This association weakened and lost significance with long-term follow-up. IL-10 showed a tendency toward an association with MACE. The 1-year HR was 2.1 (95% CI: 0.92-5.0). Long-term follow-up rendered a similar result. The association of CRP with MACE did not reach statistical significance at 1-year follow-up. However, CRP was associated with long-term risk of MACE, with an HR of 1.9 (95% CI: 1.0-3.5). CONCLUSIONS: In this prospective study, preprocedural Lp(a) level was associated with short-term prognosis after PCI. The preprocedural CRP level was associated with long-term prognosis after PCI.


Assuntos
Angioplastia Coronária com Balão , Proteína C-Reativa/metabolismo , Doença da Artéria Coronariana/terapia , Interleucina-10/sangue , Lipoproteína(a)/sangue , Biomarcadores/sangue , Intervalos de Confiança , Doença da Artéria Coronariana/sangue , Stents Farmacológicos , Feminino , Indicadores Básicos de Saúde , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Medição de Risco , Inquéritos e Questionários , Fatores de Tempo
17.
PLoS One ; 7(1): e30383, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22276189

RESUMO

BACKGROUND: Elevated low density lipoprotein (LDL) and lipoprotein(a) are independent risk factors for the development of atherosclerosis. Using a proteomic approach we aimed to determine early changes in arterial protein expression in transgenic mice containing both human LDL and lipoprotein(a) in circulation. METHODS AND RESULTS: Plasma lipid analyses showed the lipoprotein(a) transgenic mice had significantly higher lipid levels than wildtype, including a much increased LDL and high density lipoprotein (HDL) cholesterol. Analysis of aortae from lipoprotein(a) mice showed lipoprotein(a) accumulation but no lipid accumulation or foam cells, leaving the arteries essentially atherosclerosis free. Using two-dimensional gel electrophoresis and mass spectrometry, we identified 34 arterial proteins with significantly altered abundance (P<0.05) in lipoprotein(a) transgenic mice compared to wildtype including 17 that showed a ≥2 fold difference. Some proteins of interest showed a similarly altered abundance at the transcript level. These changes collectively indicated an initial metabolic response that included a down regulation in energy, redox and lipid metabolism proteins and changes in structural proteins at a stage when atherosclerosis had not yet developed. CONCLUSIONS: Our study shows that human LDL and lipoprotein(a) promote changes in the expression of a unique set of arterial proteins which may be early indicators of the metabolic disturbances preceding atherosclerosis.


Assuntos
Aorta/metabolismo , Aterosclerose/sangue , Aterosclerose/metabolismo , Lipoproteína(a)/sangue , Lipoproteínas LDL/sangue , Proteômica/métodos , Animais , Aterosclerose/genética , Biologia Computacional , Feminino , Humanos , Lipoproteína(a)/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Atherosclerosis ; 218(2): 404-10, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21763656

RESUMO

OBJECTIVE: To investigate the impact of the p.R1068H mutation on the structure and function of the ATP-binding cassette A1 (ABCA1) protein. METHODS: A homology model of the nucleotide binding domains of ABCA1 was constructed to identify the three-dimensional orientation of R1068. Cholesterol efflux assays were performed on fibroblasts obtained from members of a Tangier disease (TD) family carrying the p.R1068H mutation and in HEK293 cells transfected with a p.R1068H mutant cDNA vector. Confocal microscopy was used to investigate the localisation of the wildtype and mutant p.R1068H protein in HEK293 cells. RESULTS: Sequence alignments and modeling indicated residue R1068 to be located in an α-helix downstream of the Walker B motif in the first nucleotide binding domain (NBD-1), in a position to form ionic interactions with D1092 and E1093. Cholesterol efflux studies showed the efflux from TD fibroblasts and HEK293 cells expressing the mutant p.R1068H protein to be markedly reduced compared to wildtype. Localisation of the mutant p.R1068H protein in HEK293 cells showed intracellular retention of the protein indicating a defect in trafficking to the plasma membrane. CONCLUSION: Homology modeling of the ABCA1 protein showed that the p.R1068H mutation would likely disrupt the conformation of NBD-1. Functional studies of p.R1068H showed a lack of cholesterol efflux function due to defective trafficking to the plasma membrane, most likely caused by impaired oligomerisation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Mutação , Doença de Tangier/genética , Transportador 1 de Cassete de Ligação de ATP , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Biologia Computacional/métodos , Fibroblastos/metabolismo , Células HEK293 , Heterozigoto , Humanos , Microscopia Confocal/métodos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
19.
Atherosclerosis ; 218(1): 123-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21621776

RESUMO

OBJECTIVE: Cholesterol crystals have been shown to cause inflammation, and ultimately atherosclerotic lesions through the activation of the NLRP3 inflammasome. As cholesterol crystals have also been found in the walls of patients with abdominal aortic aneurysms (AAA), it is possible that the NLRP3 inflammasome is involved in AAA and genetic variability within this protein complex could alter disease risk. The primary objective of this study was to assess whether there is genetic evidence for a role of the NLRP3 inflammasome in AAA by testing for association of AAA with functional single nucleotide polymorphisms (SNPs) in the CARD8 and NLRP3 genes. METHODS: AAA patients (n=1151) and controls (n=727) were genotyped for CARD8 SNP rs2043211 and NLRP3 SNP rs35829419 using TaqMan SNP assays. IL1-ß, C-reactive protein (CRP), and lipoprotein (a) [Lp(a)] were measured in the plasma of a subset of study participants. The Kruskal-Wallis Rank test was conducted to test for differences in mean concentration of IL1-ß, CRP and Lp(a). Logistic regression was used to test for interaction between CARD8 and NLRP3. RESULTS: Significantly higher mean concentration of plasma IL1-ß was observed in study participants who were homozygous for the common C allele of NLRP3 rs35829419 (p=0.010). Interaction between rs2043211 and rs35829419 was observed in this dataset (χ(2)=6.22; p=0.044), which strengthened when adjusted for age, gender, smoking, diabetes, hypertension, and dyslipidemia (χ(2)=14.75; p=0.012); and separately for NOD2 genotype (χ(2)=14.06; p=0.015). CONCLUSION: Our finding suggests genetic variability within the NLRP3 inflammasome may be important in the pathophysiology of AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Idoso , Idoso de 80 Anos ou mais , Alelos , Aneurisma da Aorta Abdominal/genética , Proteína C-Reativa/metabolismo , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Transporte/genética , Colesterol/química , Feminino , Variação Genética , Genótipo , Humanos , Inflamação , Interleucina-1beta/metabolismo , Lipoproteína(a)/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas de Neoplasias/genética , Risco
20.
Biochem Biophys Res Commun ; 409(3): 400-5, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21575609

RESUMO

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.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Doença de Tangier/genética , Transportador 1 de Cassete de Ligação de ATP , Idoso , Colesterol/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Mutação , Linhagem , Doença de Tangier/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...