Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Eur J Prev Cardiol ; 31(10): 1303-1311, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-38593219

RESUMO

AIMS: Cohort studies have demonstrated associations between calcific aortic valve disease (CAVD) and Lp(a). As Lp(a) is almost entirely genetically determined, in this study, we aim to determine whether Lp(a), when predicted from genetic data, is associated with CAVD and major adverse cardiovascular events (MACEs). METHODS AND RESULTS: Patients undergoing coronary angiography between January 2012 and May 2013 were invited to participate in the study. Of 752 analysable participants, 446 had their Lp(a) measured and 703 had a calculable LPA genetic risk score (GRS). The primary outcomes were the presence of CAVD at baseline and MACE over a 7-year follow-up. The GRS explained 45% of variation in Lp(a). After adjustment for cardiac risk factors and coronary artery disease (CAD), the odds of CAVD increased with increasing Lp(a) [odds ratio (OR) 1.039 per 10-unit increase, 95% confidence interval (CI) 1.022-1.057, P < 0.001] and GRS (OR 1.054 per 10-unit increase, 95% CI 1.024-1.086; P < 0.001). Lipoprotein(a) and the GRS as continuous variables were not associated with subsequent MACEs. A dichotomized GRS (>54) was associated with MACE, but this relationship became non-significant when CAD classification was added into the model (OR 1.333, 95% CI 0.927-1.912; P = 0.12). CONCLUSION: An LPA GRS can explain 45% of variation in Lp(a) levels, and both Lp(a) and the GRS are associated with CAVD. An elevated GRS is associated with future cardiac events in a secondary risk setting, but, if the CAD status is known, it does not provide additional prognostic information.


Lipoprotein (a) [Lp(a)] is a type of cholesterol that is determined almost entirely by genetics. It is associated with heart disease and also stiffening of the heart valves. Recent advancements have made it possible to predict Lp(a) levels by analysing a person's DNA. This study examines the association between genetically predicted Lp(a) and adverse outcomes.Genetically predicted Lp(a) accounts for 45% of the variability in the actual Lp(a) level.Both actual and genetically predicted Lp(a) are associated with heart valve disease and adverse heart outcomes. If the degree of narrowing of the arteries in the heart is already known, genetically predicted Lp(a) does not help further predict risk.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Calcinose , Estratificação de Risco Genético , Lipoproteína(a) , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valva Aórtica/diagnóstico por imagem , Valva Aórtica/patologia , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/diagnóstico por imagem , Estenose da Valva Aórtica/epidemiologia , Calcinose/genética , Calcinose/diagnóstico por imagem , Angiografia Coronária , Lipoproteína(a)/sangue , Lipoproteína(a)/genética , Prognóstico , Medição de Risco , Fatores de Tempo
2.
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
3.
Biomolecules ; 12(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35204658

RESUMO

Endocytosis is the process by which molecules are actively transported into cells. It can take on a variety of forms depending on the cellular machinery involved ranging from specific receptor-mediated endocytosis to the less selective and actin-driven macropinocytosis. The plasma lipoproteins, which deliver lipids and other cargo to cells, have been intensely studied with respect to their endocytic uptake. One of the first molecules to be visualised undergoing endocytosis via a receptor-mediated, clathrin-dependent pathway was low-density lipoprotein (LDL). The LDL molecule has subsequently been shown to be internalised through multiple endocytic pathways. Dissecting the pathways of lipoprotein endocytosis has been crucial to understanding the regulation of plasma lipid levels and how lipids enter cells in the arterial wall to promote atherosclerosis. It has also aided understanding of the dysregulation that occurs in plasma lipid levels when molecules involved in uptake are defective, as is the case in familial hypercholesterolemia (FH). The aim of this review is to outline the many endocytic pathways utilised for lipoprotein uptake. It explores the various experimental approaches that have been applied to visualise lipoprotein endocytosis with an emphasis on LDL and its more complex counterpart, lipoprotein(a) [Lp(a)]. Finally, we look at new developments in lipoprotein visualisation that hold promise for scrutinising endocytic pathways to finer detail in the future.


Assuntos
Hiperlipoproteinemia Tipo II , Lipoproteínas LDL , Clatrina/metabolismo , Endocitose/fisiologia , Humanos , Lipoproteínas/metabolismo , Lipoproteínas LDL/metabolismo
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.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Free Radic Biol Med ; 51(7): 1445-53, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21798344

RESUMO

Members of the peroxidase-cyclooxygenase superfamily catalyze biochemical reactions essential to a broad spectrum of biological processes, including host defense, thyroid hormone biosynthesis, and modification of extracellular matrix, as well as contributing to the pathogenesis of chronic inflammatory diseases. We recently identified a novel member of this family, vascular peroxidase-1 (VPO1), that is highly expressed in the human cardiovascular system. Its biosynthesis and enzymatic properties are largely unknown. Here, we report that VPO1 was rapidly and efficiently secreted into the extracellular space when the gene was stably expressed in human embryonic kidney (HEK) cells. Secreted VPO1 is a monomer with complex N-linked oligosaccharides and exhibits peroxidase activity. Biosynthesis of endogenous VPO1 by cultured human umbilical vein endothelial cells (HUVECs) shares features exhibited by heterologous expression of recombinant VPO1 (rVPO1) in HEK cells. The proinflammatory agents lipopolysaccharide and tumor necrosis factor-α induce expression of VPO1 mRNA and protein in HUVECs. Furthermore, murine and bovine sera and human plasma contain enzymatically active VPO1. rVPO1 exhibits spectral and enzymatic properties characteristic of the peroxidase-cyclooxygenase family, except with regard to its heat stability. rVPO1 catalyzes tyrosyl radical formation and promotes dityrosine cross-linking. Taken together, these data demonstrate that VPO1 is a glycosylated heme peroxidase that is actively secreted into circulating plasma by vascular endothelial cells and shares several features with other members of the peroxidase-cyclooxygenase family, including the catalysis of dityrosine formation.


Assuntos
Células Endoteliais da Veia Umbilical Humana/enzimologia , Peroxidases , Tirosina/análogos & derivados , Tirosina/sangue , Animais , Biocatálise , Bovinos , Estabilidade Enzimática , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Oxirredução , Peroxidases/sangue , Peroxidases/química , Peroxidases/genética , Peroxidases/isolamento & purificação , Plasmídeos , Conformação Proteica , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
13.
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
14.
FEBS J ; 278(12): 2034-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21481187

RESUMO

The peptide antibiotic bacitracin is widely used as an inhibitor of protein disulfide isomerase (PDI) to demonstrate the role of the protein-folding catalyst in a variety of molecular pathways. Commercial bacitracin is a mixture of at least 22 structurally related peptides. The inhibitory activity of individual bacitracin analogs on PDI is unknown. For the present study, we purified the major bacitracin analogs, A, B, H, and F, and tested their ability to inhibit the reductive activity of PDI by use of an insulin aggregation assay. All analogs inhibited PDI, but the activity (IC(50) ) ranged from 20 µm for bacitracin F to 1050 µm for bacitracin B. The mechanism of PDI inhibition by bacitracin is unknown. Here, we show, by MALDI-TOF/TOF MS, a direct interaction of bacitracin with PDI, involving disulfide bond formation between an open thiol form of the bacitracin thiazoline ring and cysteines in the substrate-binding domain of PDI.


Assuntos
Bacitracina/farmacologia , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Animais , Bacitracina/análogos & derivados , Bacitracina/química , Domínio Catalítico , Bovinos , Cisteína/química , Dissulfetos/química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Estrutura Molecular , Isomerases de Dissulfetos de Proteínas/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
15.
Br J Pharmacol ; 164(2b): 507-20, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21501142

RESUMO

BACKGROUND: Celastrol is one of several bioactive compounds extracted from the medicinal plant Tripterygium wilfordii. Celastrol is used to treat inflammatory conditions, and shows benefits in models of neurodegenerative disease, cancer and arthritis, although its mechanism of action is incompletely understood. EXPERIMENTAL APPROACH: Celastrol was tested on human NADPH oxidases (NOXs) using a panel of experiments: production of reactive oxygen species and oxygen consumption by NOX enzymes, xanthine oxidase activity, cell toxicity, phagocyte oxidase subunit translocation, and binding to cytosolic subunits of NOX enzymes. The effect of celastrol was compared with diphenyleneiodonium, an established inhibitor of flavoproteins. KEY RESULTS: Low concentrations of celastrol completely inhibited NOX1, NOX2, NOX4 and NOX5 within minutes with concentration-response curves exhibiting higher Hill coefficients and lower IC50 values for NOX1 and NOX2 compared with NOX4 and NOX5, suggesting differences in their mode of action. In a cell-free system, celastrol had an IC50 of 1.24 and 8.4 µM for NOX2 and NOX5, respectively. Cytotoxicity, oxidant scavenging, and inhibition of p47(phox) translocation could not account for NOX inhibition. Celastrol bound to a recombinant p47(phox) and disrupted the binding of the proline rich region of p22(phox) to the tandem SH3 domain of p47(phox) and NOXO1, the cytosolic subunits of NOX2 and NOX1, respectively. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that celastrol is a potent inhibitor of NOX enzymes in general with increased potency against NOX1 and NOX2. Furthermore, inhibition of NOX1 and NOX2 was mediated via a novel mode of action, namely inhibition of a functional association between cytosolic subunits and the membrane flavocytochrome.


Assuntos
NADPH Oxidases/antagonistas & inibidores , Triterpenos/farmacologia , Animais , Células CHO , Linhagem Celular , Linhagem Celular Transformada , Cricetinae , Citosol/efeitos dos fármacos , Citosol/metabolismo , Células HEK293 , Humanos , Peróxido de Hidrogênio/metabolismo , NADPH Oxidases/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Oniocompostos/farmacologia , Oxirredutases/metabolismo , Oxigênio/metabolismo , Triterpenos Pentacíclicos , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Domínios de Homologia de src/efeitos dos fármacos
16.
Clin Chem ; 53(4): 679-85, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17303687

RESUMO

BACKGROUND: Increased lipoprotein(a) [Lp(a)] concentrations are predictive for coronary artery disease (CAD). The risk conferred by Lp(a) for other types of vascular disease compared with CAD has not been investigated within a single population. This study aimed to investigate Lp(a) risk association for 4 different types of vascular disease (including CAD) within a predominantly white population. METHODS: We used an Lp(a) ELISA that measures Lp(a) independently of apolipoprotein(a) size to measure plasma Lp(a) in patients [384 CAD, 262 peripheral vascular disease, 184 ischemic stroke (stroke), 425 abdominal aortic aneurysm] and 230 disease-free controls. We then conducted association studies with logistic regression, integrating the potential confounding effects of age, sex, diabetes, plasma lipids, and a history of previous hypertension, hypercholesterolemia, and smoking. RESULTS: Multivariate analyses with Lp(a) concentrations of >45 nmol/L (the 75th percentile value for controls) as the clinical cutoff showed increased Lp(a) concentrations to be a risk factor for all disease groups, with adjusted odds ratios ranging from 1.96 [95% confidence interval (CI) 1.24-3.08] for CAD to 2.33 (95% CI 1.39-3.89) for PVD. The risk conferred by Lp(a) appeared to be independent of other confounders, including exposure to statin/fibrate therapies. Similar odds ratios and CIs between disease groups indicated that increased Lp(a) conferred a similar risk for all groups studied. CONCLUSIONS: Lp(a) constitutes a stable risk factor of similar magnitude for 4 major forms of vascular disease. This association was not altered by exposure to standard lipid-lowering therapy.


Assuntos
Aneurisma da Aorta Abdominal/epidemiologia , Doença da Artéria Coronariana/epidemiologia , Lipoproteína(a)/sangue , Doenças Vasculares Periféricas/epidemiologia , Acidente Vascular Cerebral/epidemiologia , Idoso , Aneurisma da Aorta Abdominal/etnologia , Doença da Artéria Coronariana/etnologia , Feminino , Humanos , Masculino , Doenças Vasculares Periféricas/etnologia , Plasma , Valores de Referência , Risco , População Branca
17.
J Biol Chem ; 281(41): 30336-46, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16895900

RESUMO

The heterodimeric flavocytochrome b558, comprised of the two integral membrane proteins p22phox and gp91phox, mediates the transfer of electrons from NADPH to molecular oxygen in the phagocyte NADPH oxidase to generate the superoxide precursor of microbicidal oxidants. This study uses deletion mutagenesis to identify regions of p22phox required for maturation of gp91phox and for NADPH oxidase activity. N-terminal, C-terminal, or internal deletions of human p22phox were generated and expressed in Chinese hamster ovary cells with transgenes for gp91phox and two other NADPH oxidase subunits, p47phox, and p67phox. The results demonstrate that p22phox-dependent maturation of gp91phox carbohydrate, cell surface expression of gp91phox, and the enzymatic function of flavocytochrome b558 are closely correlated. Whereas the 5 N-terminal and 25 C-terminal amino acids are dispensable for these functions, the N-terminal 11 amino acids of p22phox are required, as is a hydrophilic region between amino acids 65 and 90. Upon deletion of 54 residues at the C terminus of p22phox (amino acids 142-195), maturation and cell surface expression of gp91phox was still preserved, although NADPH oxidase activity was absent, as expected, due to removal of a proline-rich domain between amino acids 151-160 that is required for recruitment of p47phox. Antibody binding studies indicate that the extreme N terminus of p22phox is inaccessible in the absence of cell permeabilization, supporting a model in which both the N- and C-terminal domains of p22phox extend into the cytoplasm, anchored by two membrane-embedded regions.


Assuntos
Grupo dos Citocromos b/genética , Deleção de Genes , Glicoproteínas de Membrana/genética , Mutagênese , NADPH Oxidases/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Membrana Celular/metabolismo , Humanos , Dados de Sequência Molecular , NADPH Oxidase 2 , NADPH Oxidases/genética , Homologia de Sequência de Aminoácidos
18.
J Vasc Res ; 42(3): 237-46, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15860923

RESUMO

The aortae from male apolipoprotein E knockout (apoE-/-) mice were examined by serial section immunohistochemistry and electron microscopy to determine the continuity of the internal elastic lamina (IEL) and its association with developing intimal lesions. While in this model, defects in the elastic laminae have previously been described beneath advanced xanthomatous lesions, this study demonstrates that disruption of the IEL may be a primary factor in the localization and pathogenesis of intimal lesions in the apoE-/- mouse. IEL defects were found beneath early lesions in animals as young as 8 weeks of age. Small defects without associated intimal alteration were also observed. The elastic tissue defects beneath early intimal lesions were usually transversely orientated with abrupt "break" edges. Regions consistent with direct enzymatic digestion of the IEL were relatively rare and only observed beneath advanced plaques, particularly in the brachiocephalic artery. The presence of IEL defects around branch sites of old C57BL/6J control mice along with their matching localization and morphology in apoE-/- appears consistent with biomechanical fatigue rather than direct enzymatic degradation. In conclusion, disruption of the IEL appears to be a prominent early, if not initial, feature of the apoE-/- model of atherosclerosis and may act as the nidus upon which intimal lesions develop.


Assuntos
Doenças da Aorta/etiologia , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Tecido Elástico/patologia , Animais , Aorta/patologia , Apolipoproteínas E/genética , Arteriosclerose/etiologia , Arteriosclerose/patologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Microscopia Eletrônica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA