Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Atherosclerosis ; 222(2): 367-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22494626

ABSTRACT

OBJECTIVE: Increased secreted phospholipase A(2) (sPLA(2)) activity has been documented in several inflammatory disorders. Among sPLA(2)s, the human group X (hGX)-sPLA(2) has the highest catalytic activity towards phosphatidylcholine (PC), the major phospholipid of cell membranes and blood lipoproteins. hGX-sPLA(2) has been detected in human atherosclerotic lesions, indicating that sPLA(2)s are an important link between lipids and inflammation, both involved in atherosclerosis. The presence of dendritic cells (DC), the most potent antigen presenting cells, in atherosclerotic lesions has raised the question about their role in disease progression. METHODS AND RESULTS: In this study, we show that hGX-sPLA(2)-treated LDL induces human monocyte-derived DC maturation, resulting in a characteristic mature DC phenotype and enhanced DC ability to activate IFNγ secretion from T cells. hGX-sPLA(2) phospholipolysis of LDL produces high levels of lipid mediators, such as lysophosphatidylcholine (LPC) and free fatty acids (FFAs), which also modulate DC maturation. The major molecular species of LPC containing a palmitic or stearic acid esterified in the sn-1 position induce DC maturation, whereas the FFAs can positively or negatively modulate DC maturation depending on their nature. hGX-sPLA(2) added alone can also activate DC in vitro through the hydrolysis of the DC membrane phospholipids leading, however, to a different cytokine profile secretion pattern than the one observed with hGX-sPLA(2)-phospholipolysed LDL. CONCLUSION: hGX-sPLA(2) secreted in inflamed tissues can contribute to local DC maturation, resulting in pro-Th1 cells, through the production of various lipid mediators from hydrolysis of either LDL and/or cell plasma membrane.


Subject(s)
Atherosclerosis/enzymology , Dendritic Cells/enzymology , Group X Phospholipases A2/metabolism , Lipoproteins, LDL/metabolism , Atherosclerosis/immunology , Coculture Techniques , Dendritic Cells/immunology , Fatty Acids, Nonesterified/metabolism , Humans , Hydrolysis , Interferon-gamma/metabolism , Lysophosphatidylcholines/metabolism , Phenotype , Phosphatidylcholines/metabolism , Th1 Cells/immunology
2.
Atherosclerosis ; 218(1): 117-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21616491

ABSTRACT

OBJECTIVE: We set out to investigate the relative contribution of genetic and environmental effect on two inflammatory CVD biomarkers; lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) and anti-phosphorylcholine IgM (anti-PC). Their relationships and possible co-regulation with other established CVD biomarkers are also examined. METHODS: Lp-PLA(2) activity (N=1600) and anti-PC (N=2036) levels were measured in elderly Swedish twins. Correlation analyses and heritability estimation were conducted by structural equation modeling. RESULTS: We attribute 0.37 of the variance of Lp-PLA(2) and 0.40 of anti-PC variance to genetic variance. In addition, a bivariate heritability of 0.33, 0.35 and 0.36 could be detected for levels of Lp-PLA(2) together with ApoB, total cholesterol and LDL, respectively. Anti-PC was only weakly related to other biomarkers of CVD, which may suggest a more independent role of anti-PC as a biomarker. CONCLUSIONS: In this large sample, Lp-PLA(2) activity has lower heritability and higher environmental regulation than previously reported. Anti-PC levels are partly influenced by dominance genetics and appear to be regulated independently of more established CVD biomarkers.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Cardiovascular Diseases/blood , Immunoglobulin M/blood , Aged , Biomarkers/metabolism , Diseases in Twins , Environment , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Inflammation , Male , Middle Aged , Models, Genetic , Phosphorylcholine/chemistry , Sweden
3.
FASEB J ; 24(9): 3284-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20430794

ABSTRACT

Secreted phospholipases A2 (sPLA2s) are present in atherosclerotic plaques and are now considered novel attractive therapeutic targets and potential biomarkers as they contribute to the development of atherosclerosis through lipoprotein-dependent and independent mechanisms. We have previously shown that hGX-sPLA2-phospholipolyzed LDL (LDL-X) induces proinflammatory responses in human umbilical endothelial cells (HUVECs); here we explore the molecular mechanisms involved. Global transcriptional gene expression profiling of the response of endothelial cells exposed to either LDL or LDL-X revealed that LDL-X activates multiple distinct cellular pathways including the unfolded protein response (UPR). Mechanistic insight showed that LDL-X activates UPR through calcium depletion of intracellular stores, which in turn disturbs cytoskeleton organization. Treatment of HUVECs and aortic endothelial cells (HAECs) with LDL-X led to activation of all 3 proximal initiators of UPR: eIF-2alpha, IRE1alpha, and ATF6. In parallel, we observed a sustained phosphorylation of the p38 pathway resulting in the phosphorylation of AP-1 downstream targets. This was accompanied by significant production of the proinflammatory cytokines IL-6 and IL-8. Our study demonstrates that phospholipolyzed LDL uses a range of molecular pathways including UPR to initiate endothelial cell perturbation and thus provides an LDL oxidation-independent mechanism for the initiation of vascular inflammation in atherosclerosis.


Subject(s)
Endoplasmic Reticulum/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipoproteins, LDL/pharmacology , Activating Transcription Factor 6/metabolism , Blotting, Western , Cells, Cultured , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Fluorescent Antibody Technique , Gene Silencing , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipoproteins, LDL/metabolism , Oligonucleotide Array Sequence Analysis , Phospholipases A2, Secretory , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Umbilical Veins/cytology , Unfolded Protein Response
4.
Biochimie ; 92(6): 594-600, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20153800

ABSTRACT

Phospholipases A2 (PLA2) are a family of enzymes that catalyze the hydrolysis of the sn-2 ester bond of glycerophospholipids liberating lysophospholipids and free fatty acids; important second messengers involved in atherogenesis. Plasma PAF-acetylhydrolase (PAF-AH) or Lp-PLA2 is a Ca(2+)-independent PLA2 which is produced by monocyte-derived macrophages and by activated platelets, and circulates in plasma associated with lipoproteins. PAF-AH catalyzes the removal of the acetyl/short acyl group at the sn-2 position of PAF and oxidized phospholipids produced during inflammation and oxidative stress. In humans, PAF-AH is mainly associated with small dense LDL and to a lesser extent with HDL and with lipoprotein(a). PAF-AH is N-glycosylated prior to secretion which diminishes its association with HDL raising the question of its distribution between the proatherogenic LDL vs the antiatherogenic HDL. Hypercholesterolemic patients have higher plasma PAF-AH activity which is reduced upon hypolipidemic therapy. PAF-AH specific inhibitor darapladib stabilizes human and swine plaques, therefore challenging the antiatherogenic potential of PAF-AH shown in small animal models. Among secreted PLA2s (sPLA2), the group X sPLA2 (PLA2GX), due to its very high activity towards phosphatidylcholine the main phospholipid of LDL, became an attractive target in atherosclerosis. We showed that PLA2GX is present in human atherosclerotic lesions and that the PLA2GX-phospholipolysed LDL triggers human macrophage-foam cell formation. In contrast to other sPLA2s, including group IB, IIA and V, PLA2GX can efficiently hydrolyze PAF present in lipoproteins or vesicles indicating that PLA2GX may be a novel player in PAF regulation upon inflammatory processes. By a genetic approach we uncovered a relatively rare polymorphism (Arg38Cys) which produces a catalytically inactive PLA2GX; although no association was observed with cardiovascular risk factors in the AtheroGene study, this result should be replicated in cohorts of other inflammatory diseases. We anticipate that mores studies will be necessary to sort out the exact role of extracellular PLA2 family members in atherosclerosis initiation and progression.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Atherosclerosis/enzymology , Animals , Atherosclerosis/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Lipoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...