Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 295(30): 10092-10111, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32482892

ABSTRACT

Aortic dissection is a life-threatening aortopathy involving separation of the aortic wall, whose underlying mechanisms are still incompletely understood. Epidemiological evidence suggests that unsaturated fatty acids improve cardiovascular health. Here, using quantitative RT-PCR, histological analyses, magnetic cell sorting and flow cytometry assays, and MS-based lipidomics, we show that the activity of a lipid-metabolizing enzyme, secreted phospholipase A2 group V (sPLA2-V), protects against aortic dissection by endogenously mobilizing vasoprotective lipids. Global and endothelial cell-specific sPLA2-V-deficient mice frequently developed aortic dissection shortly after infusion of angiotensin II (AT-II). We observed that in the AT-II-treated aorta, endothelial sPLA2-V mobilized oleic and linoleic acids, which attenuated endoplasmic reticulum stress, increased the expression of lysyl oxidase, and thereby stabilized the extracellular matrix in the aorta. Of note, dietary supplementation with oleic or linoleic acid reversed the increased susceptibility of sPLA2-V-deficient mice to aortic dissection. These findings reveal an unexplored functional link between sPLA2-driven phospholipid metabolism and aortic stability, possibly contributing to the development of improved diagnostic and/or therapeutic strategies for preventing aortic dissection.


Subject(s)
Aorta/metabolism , Aortic Dissection/metabolism , Endoplasmic Reticulum Stress , Group V Phospholipases A2/metabolism , Phospholipids/metabolism , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Aorta/pathology , Disease Models, Animal , Group V Phospholipases A2/genetics , Linoleic Acid/genetics , Linoleic Acid/metabolism , Mice , Mice, Knockout , Oleic Acid/genetics , Oleic Acid/metabolism , Phospholipids/genetics
2.
Microvasc Res ; 129: 103954, 2020 05.
Article in English | MEDLINE | ID: mdl-31730773

ABSTRACT

Group V secretory phospholipase A2 (gVPLA2) is a potent inflammatory mediator in mammalian tissues that hydrolyzes phospholipids and initiates eicosanoid biosynthesis. Previous work has demonstrated that multiple inflammatory stimuli induce its expression and secretion in several cell types, including the lung endothelium. However, little is known about the mechanism(s) by which gVPLA2 inflammatory signaling is subsequently downregulated. Therefore, in this study we characterized potential clearance mechanisms for gVPLA2 in lung endothelial cells (EC). We observed that exogenous gVPLA2 is taken up rapidly by nutrient-starved human pulmonary artery EC (HPAEC) in vitro, and its cellular expression subsequently is reduced over several hours. In parallel experiments performed in pulmonary vascular EC isolated from mice genetically deficient in gVPLA2, the degradation of exogenously applied gVPLA2 occurs in a qualitatively similar fashion. This degradation is significantly attenuated in EC treated with ammonium chloride or chloroquine, which are lysosomal inhibitors that block autophagic flux. In contrast, the proteasomal inhibitor MG132 fails to prevent the clearance of gVPLA2. Both immunofluorescence microscopy and proximity ligation assay demonstrate the co-localization of LC3 and gVPLA2 during this process, indicating the association of gVPLA2 with autophagosomes. Nutrient starvation, a known inducer of autophagy, is sufficient to stimulate gVPLA2 degradation. These results suggest that a lysosome-mediated autophagy pathway contributes to gVPLA2 clearance from lung EC. These novel observations advance our understanding of the mechanism by which this key inflammatory enzyme is downregulated in the lung vasculature.


Subject(s)
Autophagy , Endothelial Cells/enzymology , Group V Phospholipases A2/metabolism , Lysosomes/enzymology , Pulmonary Artery/enzymology , Animals , Cells, Cultured , Enzyme Stability , Group V Phospholipases A2/deficiency , Group V Phospholipases A2/genetics , Humans , Mice, Knockout , Proteolysis , Time Factors
3.
J Recept Signal Transduct Res ; 39(1): 60-66, 2019 Feb.
Article in English | MEDLINE | ID: mdl-31084404

ABSTRACT

sPLA2 is released under inflammatory conditions from neutrophils, basophils and T-cells. They cleave the cellular phospholipids leading to the release of arachidonic acid and there by provide intermediates for biosynthesis of inflammatory mediators. The focus of this study is on the interaction of hesperidin, a natural flavonoid with Group IB, IIA, and V and X isozymes of sPLA2. Affinity of hesperidin towards PLA2 isozymes was analyzed through enzymatic studies and molecular modeling. The experiments showed that hesperidin competitively inhibited PLA2 with IC50 of 5.1 µM. Molecular modeling studies revealed the association of hesperidin with the docking scores -6.90, -9.53, -5.63 and -8.29 kcal for isozymes Group IB, IIA, V and X of PLA2 respectively. Their binding energy values were calculated as -20.25, -21.63, -21.66 and -33.43 kcal for the Group IB, IIA, V and X respectively. Structural model for Group V was made by homology modeling since no structural coordinates were available. Molecular dynamics studies were carried out to evaluate the structural stability of protein ligand complex. The analyses showed that hesperidin blocked the entry of the substrate to the active site of PLA2 and it was indifferent to the differences of the isozymes. Hence, hesperidin might serve as lead for designing highly specific anti-inflammatory drugs directed to the PLA2 isozyme specific to various diseases, with IC50 value of therapeutic significance.


Subject(s)
Group II Phospholipases A2/metabolism , Group V Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Hesperidin/pharmacology , Amino Acid Sequence , Calcium , Catalytic Domain , Computer Simulation , Group II Phospholipases A2/antagonists & inhibitors , Group V Phospholipases A2/antagonists & inhibitors , Group X Phospholipases A2/antagonists & inhibitors , Humans , Isoenzymes , Ligands , Molecular Docking Simulation , Protein Conformation , Sequence Homology
4.
Bioorg Chem ; 92: 103218, 2019 11.
Article in English | MEDLINE | ID: mdl-31536956

ABSTRACT

To enhance the cytotoxicity of benzimidazole and/or benzoxazole core, the benzimidazole/benzoxazole azo-pyrimidine were synthesized through diazo-coupling of 3-aminophenybenzimidazole (6a) or 3-aminophenylbenzoxazole (6b) with diethyl malonate. The new azo-molanates 6a&b mixed with urea in sodium ethoxide to afford the benzimidazolo/benzoxazolopyrimidine 7a&b. The structure elucidation of new synthesized targets was proved using spectroscopic techniques NMR, IR and elemental analysis. The cytoxicity screening had been carried out against five cancer cell lines: prostate cancer (PC-3), lung cancer (A-549), breast cancer (MCF-7), pancreas cancer (PaCa-2) and colon cancer (HT-29). Furthermore, the antioxidant activity, phospholipase A2-V and cyclooxygenases inhibitory activities of the target compounds 7a&b were evaluated and the new compounds showed potent activity (cytotoxicity IC50 range from 4.3 to 9.2 µm, antioxidant activity from 40% to 80%, COXs or LOX inhibitory activity from 1.92 µM to 8.21 µM). The docking of 7a&b was made to confirm the mechanism of action.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Phospholipase A2 Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Biphenyl Compounds/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Group V Phospholipases A2/antagonists & inhibitors , Group V Phospholipases A2/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Phospholipase A2 Inhibitors/chemical synthesis , Phospholipase A2 Inhibitors/chemistry , Picrates/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship
5.
J Immunol ; 194(7): 3327-39, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25725101

ABSTRACT

Studies on the heterogeneity and plasticity of macrophage populations led to the identification of two major polarization states: classically activated macrophages or M1, induced by IFN-γ plus LPS, and alternatively activated macrophages, induced by IL-4. We studied the expression of multiple phospholipase A2 enzymes in human macrophages and the effect that polarization of the cells has on their levels. At least 11 phospholipase A2 genes were found at significant levels in human macrophages, as detected by quantitative PCR. None of these exhibited marked changes after treating the cells with IFN-γ plus LPS. However, macrophage treatment with IL-4 led to strong upregulation of the secreted group V phospholipase A2 (sPLA2-V), both at the mRNA and protein levels. In parallel with increasing sPLA2-V expression levels, IL-4-treated macrophages exhibited increased phagocytosis of yeast-derived zymosan and bacteria, and we show that both events are causally related, because cells deficient in sPLA2-V exhibited decreased phagocytosis, and cells overexpressing the enzyme manifested higher rates of phagocytosis. Mass spectrometry analyses of lipid changes in the IL-4-treated macrophages suggest that ethanolamine lysophospholipid (LPE) is an sPLA2-V-derived product that may be involved in regulating phagocytosis. Cellular levels of LPE are selectively maintained by sPLA2-V. By supplementing sPLA2-V-deficient cells with LPE, phagocytosis of zymosan or bacteria was fully restored in IL-4-treated cells. Collectively, our results show that sPLA2-V is required for efficient phagocytosis by IL-4-treated human macrophages and provide evidence that sPLA2-V-derived LPE is involved in the process.


Subject(s)
Group V Phospholipases A2/genetics , Interleukin-4/metabolism , Macrophages/immunology , Macrophages/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Phosphatidylethanolamines/metabolism , Gene Expression Regulation/drug effects , Group V Phospholipases A2/deficiency , Group V Phospholipases A2/metabolism , Healthy Volunteers , Humans , Hydrolysis , Interleukin-4/pharmacology , Isoenzymes , Lipid Metabolism , Macrophages/drug effects , Male , Phagocytosis/drug effects , Phosphatidylethanolamines/pharmacology
6.
Article in English | MEDLINE | ID: mdl-26711221

ABSTRACT

Mitogenesis of Vascular Smooth Muscle Cells (VSMC) plays an important role in atherogenesis. Until recently, the effect of lipid subfractions has not been clarified. Secretory phospholipases A2 (sPLA2's) hydrolyse glycerophospholipids and release pro-inflammatory lyso-lipids, oxidized and non-oxidized fatty acids and isoprostanes. They localize in the vascular wall. We hypothesized that structurally similar sPLA2's may exert different impact on VSMC. The influence of sPLA2's, IIA, V, X, HDL, LDL, and hydrolysis products was tested on mitogenesis of VSMC, i.e., the early effect on the cell membrane phospholipids, and on PGE2 and LTB4 release, i.e., late effect of Cyclooxygenase and 5-lipooxygenase activity in VSMC. Mitogenesis was significantly enhanced by HDL and LDL, and by products of sPLA2 hydrolysis. Hydrolysis of HDL or LDL enhanced mitogenic activity in order V>X>IIA. The release of PGE2 was enhanced by group X sPLA2 and by HDL hydrolyzed by groups V and X. LDL and its hydrolysis products enhanced the release of PGE2 in order X>V>IIA. The release of LTB4 was markedly increased by LDL and HDL, and by hydrolytic products of group V and X, but not group IIA sPLA2. Our study demonstrates a diverse interaction of pro-inflammatory sPLA2's with HDL and LDL affecting both mitogenesis and eicosanoid release from VSMC, therefore potentially enhancing their pro-atherogenic activity.


Subject(s)
Eicosanoids/metabolism , Lipoproteins/metabolism , Myocytes, Smooth Muscle/metabolism , Phospholipases A2, Secretory/metabolism , Arachidonate 5-Lipoxygenase/metabolism , Cells, Cultured , Dinoprostone/metabolism , Group II Phospholipases A2/metabolism , Group V Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Humans , Hydrolysis , Leukotriene B4/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Mitosis , Muscle, Smooth, Vascular/cytology , Prostaglandin-Endoperoxide Synthases/metabolism , Time Factors
7.
J Cell Biochem ; 116(9): 1898-907, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25716287

ABSTRACT

We examined the effect of the cellular sphingolipid level on the release of arachidonic acid (AA) and the activity of secretory phospholipase A2 (sPLA2 ) using two Chinese hamster ovary (CHO)-K1 cell mutants, LY-B and LY-A cells, deficient in sphingolipid synthesis. In LY-B cells, deficiency of sphingolipids enhanced the release of AA induced by bee venom sPLA2-III or human sPLA2-V. These alterations were reversed by replenishment of exogenous sphingomyelin (SM). In LY-A cells, deficiency of SM increased the release of AA induced by sPLA2. In CHO-K1 cells, decrease and increase of SM level in the plasma membrane by pharmacological methods increased and inhibited the release of AA, respectively. SM inhibited the activity of sPLA2 in vitro. Niemann-Pick disease type C (NPC) is a lysosomal storage disorder caused by mutation of either the NPC1 or NPC2 gene, and is characterized by accumulation of cholesterol and sphingolipids including SM in late endosomes/lysosomes. Increased levels of AA and sPLA2 activity are involved in various neurodegenerative diseases. In CHO cells lacking NPC1 (A101 cells), SM level was lower in the plasma membrane, while it was higher in late endosomes/lysosomes. The release of AA induced by sPLA2 was increased in A101 cells than that in parental cells (JP17 cells), which was attenuated by adding exogenous SM. In addition, sPLA2 -III-induced cytotoxicity in A101 cells was much higher than that in JP17 cells. These results suggest that SM in the plasma membrane plays important roles in regulating sPLA2 activity and the enzyme-induced cytotoxicity in A101 cells.


Subject(s)
Arachidonic Acid/biosynthesis , Cell Membrane/metabolism , Niemann-Pick Disease, Type C/enzymology , Phospholipases A2, Secretory/metabolism , Sphingomyelins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Group III Phospholipases A2/metabolism , Group III Phospholipases A2/pharmacology , Group V Phospholipases A2/metabolism , Group V Phospholipases A2/pharmacology , Humans , Membrane Glycoproteins/deficiency , Models, Biological , Phospholipases A2, Secretory/pharmacology , Sphingomyelins/deficiency
8.
J Immunol ; 190(12): 5927-38, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23650617

ABSTRACT

We reported that Pla2g5-null mice lacking group V secretory phospholipase A2 (gV-sPLA2) showed reduced eosinophilic pulmonary inflammation and Th2 cytokine generation when challenged with an extract from house dust mite Dermatophagoides farinae, compared with wild-type (WT) controls. Adoptive transfer studies suggested that gV-sPLA2 in dendritic cells was necessary for sensitization of Pla2g5-null mice, but was not sufficient to induce the effector phase of pulmonary inflammation. In this study, we demonstrate that gV-sPLA2 is inducibly expressed in mouse and human macrophages (M) activated by IL-4 and is required for the acquisition of M effector functions that facilitate the effector phase of pulmonary inflammation. We demonstrate that gV-sPLA2 expression in M is sufficient for the development of pulmonary inflammation, even when inflammation is induced by intrapulmonary administration of IL-4. The concentrations of CCL22/CCL17 and effector T cell recruitment are severely impaired in Pla2g5-null mice. Intratracheal transfers of enriched CD68(+) cells isolated from the lungs of D. farinae-challenged WT donor mice induce eosinophilia, chemokine production, and recruitment of T cells into the lungs of Pla2g5-null recipients previously sensitized by WT D. farinae-loaded dendritic cells. Our studies identified a unique function of gV-sPLA2 in activation of M and in their capacity to recruit T cells to amplify the effector phase of pulmonary inflammation.


Subject(s)
Group V Phospholipases A2/immunology , Hypersensitivity/immunology , Macrophage Activation/immunology , Pneumonia/immunology , Animals , Fluorescent Antibody Technique , Group V Phospholipases A2/metabolism , Humans , Hypersensitivity/metabolism , Immunohistochemistry , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Am J Hum Genet ; 89(6): 782-91, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22137173

ABSTRACT

Flecked-retina syndromes, including fundus flavimaculatus, fundus albipunctatus, and benign fleck retina, comprise a group of disorders with widespread or limited distribution of yellow-white retinal lesions of various sizes and configurations. Three siblings who have benign fleck retina and were born to consanguineous parents are the basis of this report. A combination of homozygosity mapping and exome sequencing helped to identify a homozygous missense mutation, c.133G>T (p.Gly45Cys), in PLA2G5, a gene encoding a secreted phospholipase (group V phospholipase A(2)). A screen of a further four unrelated individuals with benign fleck retina detected biallelic variants in the same gene in three patients. In contrast, no loss of function or common (minor-allele frequency>0.05%) nonsynonymous PLA2G5 variants have been previously reported (EVS, dbSNP, 1000 Genomes Project) or were detected in an internal database of 224 exomes (from subjects with adult onset neurodegenerative disease and without a diagnosis of ophthalmic disease). All seven affected individuals had fundoscopic features compatible with those previously described in benign fleck retina and no visual or electrophysiological deficits. No medical history of major illness was reported. Levels of low-density lipoprotein were mildly elevated in two patients. Optical coherence tomography and fundus autofluorescence findings suggest that group V phospholipase A(2) plays a role in the phagocytosis of photoreceptor outer-segment discs by the retinal pigment epithelium. Surprisingly, immunohistochemical staining of human retinal tissue revealed localization of the protein predominantly in the inner and outer plexiform layers.


Subject(s)
Eye Abnormalities/genetics , Group V Phospholipases A2/genetics , Homozygote , Mutation, Missense , Retina/abnormalities , Adult , Aged, 80 and over , Alternative Splicing , Amino Acid Sequence , Base Sequence , Child , Consanguinity , Female , Genetic Association Studies , Group V Phospholipases A2/metabolism , Humans , Male , Molecular Sequence Data , Pedigree , Polymorphism, Single Nucleotide , Protein Transport , Retina/metabolism
10.
Blood ; 119(12): 2914-21, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22167755

ABSTRACT

The endothelial protein C receptor (EPCR) plays an important role in cardiovascular disease by binding protein C/activated protein C (APC). EPCR structure contains a hydrophobic groove filled with an unknown phospholipid needed to perform its function. It has not been established whether lipid exchange takes place in EPCR as a regulatory mechanism of its activity. Our objective was to identify this phospholipid and to explore the possibility of lipid exchange as a regulatory mechanism of EPCR activity driven by the endothelially expressed secretory group V phospholipase A(2) (sPLA(2)-V). We identified phosphatidylcholine (PCh) as the major phospholipid bound to human soluble EPCR (sEPCR). PCh in EPCR could be exchanged for lysophosphatidylcholine (lysoPCh) and platelet activating factor (PAF). Remarkably, lysoPCh and PAF impaired the protein C binding ability of sEPCR. Inhibition of sPLA(2)-V, responsible for lysoPCh and PAF generation, improved APC binding to endothelial cells. EPCR-dependent protein C activation and APC antiapoptotic effect were thus significantly enhanced. In contrast, endothelial cell supplementation with sPLA(2)-V inhibited both APC generation and its antiapoptotic effects. We conclude that APC generation and function can be modulated by changes in phospholipid occupancy of its endothelial cell receptor.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Group V Phospholipases A2/metabolism , Lysophosphatidylcholines/chemistry , Platelet Activating Factor/chemistry , Protein C/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Animals , Chromatography, Thin Layer , Endothelial Cells/metabolism , Endothelial Protein C Receptor , Enzyme Activation/physiology , Flow Cytometry , Humans , Immunohistochemistry , Lysophosphatidylcholines/metabolism , Mass Spectrometry , Mice , Platelet Activating Factor/metabolism , Protein Structure, Tertiary , Spectrometry, Fluorescence
11.
Biochim Biophys Acta ; 1821(2): 257-67, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22041135

ABSTRACT

Local acidic areas characterize diffuse intimal thickening (DIT) and advanced atherosclerotic lesions. The role of acidity in the modification and extra- and intracellular accumulation of triglyceride-rich VLDL and IDL particles has not been studied before. Here, we examined the effects of acidic pH on the activity of recombinant human group V secreted phospholipase A(2) (sPLA(2)-V) toward small VLDL (sVLDL), IDL, and LDL, on the binding of these apoB-100-containing lipoproteins to human aortic proteoglycans, and on their uptake by human monocyte-derived macrophages. At acidic pH, the ability of sPLA(2)-V to lipolyze the apoB-100-containing lipoproteins was moderately, but significantly, increased while binding of the lipoproteins to proteoglycans increased >60-fold and sPLA(2)-V-modification further doubled the binding. Moreover, acidic pH more than doubled macrophage uptake of soluble complexes of sPLA(2)-V-LDL with aortic proteoglycans. Proteoglycan-affinity chromatography at pH 7.5 and 5.5 revealed that sVLDL, IDL, and LDL consisted of populations with different proteoglycan-binding affinities, and, surprisingly, the sVLDL fractions with the highest proteoglycan-affinity contained only low amounts of apolipoproteins E and C-III. Our results suggest that in atherosclerotic lesions with acidic extracellular pH, sPLA(2)-V is able to lipolyze sVLDL, IDL, and LDL, and increase their binding to proteoglycans. This is likely to provoke extracellular accumulation of lipids derived from these atherogenic lipoprotein particles and to increase the progression of the atherosclerotic lesions.


Subject(s)
Acids/metabolism , Aorta/metabolism , Apolipoprotein B-100/metabolism , Group V Phospholipases A2/metabolism , Lipolysis , Lipoproteins/metabolism , Proteoglycans/metabolism , Aorta/pathology , Apolipoprotein C-III/metabolism , Apolipoproteins E/metabolism , Chromatography, Affinity , Fatty Acids, Nonesterified/metabolism , Humans , Hydrogen-Ion Concentration , Macrophages/metabolism , Protein Binding , Substrate Specificity , Tritium
12.
Am J Pathol ; 181(3): 1088-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22813854

ABSTRACT

Abdominal aortic aneurysms (AAAs) and heart failure are complex life-threatening diseases whose etiology is not completely understood. In this study, we investigated whether deficiency of group V secretory phospholipase A(2) (GV sPLA(2)) protects from experimental AAA. The impact of GV sPLA(2) deficiency on angiotensin (Ang) II-induced cardiac fibrosis was also investigated. Apolipoprotein E (apoE)(-/-) mice and apoE(-/-) mice lacking GV sPLA(2) (GV DKO) were infused with 1000 ng/kg per minute Ang II for up to 28 days. Increases in systolic blood pressure, plasma aldosterone level, and urinary and heart prostanoids were similar in apoE(-/-) and GV DKO mice after Ang II infusion. The incidence of aortic rupture in Ang II-infused GV DKO mice (10%) was significantly reduced compared with apoE(-/-) mice (29.4%). Although the incidence of AAA in GV DKO mice (81.3%) and apoE(-/-) mice (100%) was similar, the mean percentage increase in maximal luminal diameter of abdominal aortas was significantly smaller in GV DKO mice (68.5% ± 7.7%) compared with apoE(-/-) mice (92.6% ± 8.3%). Deficiency of GV sPLA(2) resulted in increased Ang II-induced cardiac fibrosis that was most pronounced in perivascular regions. Perivascular collagen, visualized by picrosirius red staining, was associated with increased TUNEL staining and increased immunopositivity for macrophages and myofibroblasts and nicotinamide adenine dinucleotide phosphate oxidase (NOX)-2 and NOX-4, respectively. Our findings indicate that GV sPLA(2) modulates pathological responses to Ang II, with different outcomes for AAA and cardiac fibrosis.


Subject(s)
Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Disease Progression , Group V Phospholipases A2/metabolism , Myocardium/pathology , Angiotensin II/administration & dosage , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Rupture/enzymology , Aortic Rupture/pathology , Apolipoproteins E/metabolism , Apoptosis/drug effects , Collagen/metabolism , Fibrosis , Group V Phospholipases A2/deficiency , Immunohistochemistry , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/enzymology , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Prostaglandins/metabolism
13.
J Biol Chem ; 286(41): 35650-35662, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21849511

ABSTRACT

Group V-secreted phospholipase A(2) (GV sPLA(2)) hydrolyzes bacterial phospholipids and initiates eicosanoid biosynthesis. Here, we elucidate the role of GV sPLA(2) in the pathophysiology of Escherichia coli pneumonia. Inflammatory cells and bronchial epithelial cells both express GV sPLA(2) after pulmonary E. coli infection. GV(-/-) mice accumulate fewer polymorphonuclear leukocytes in alveoli, have higher levels of E. coli in bronchoalveolar lavage fluid and lung, and develop respiratory acidosis, more severe hypothermia, and higher IL-6, IL-10, and TNF-α levels than GV(+/+) mice after pulmonary E. coli infection. Eicosanoid levels in bronchoalveolar lavage are similar in GV(+/+) and GV(-/-) mice after lung E. coli infection. In contrast, GV(+/+) mice have higher levels of prostaglandin D(2) (PGD(2)), PGF(2α), and 15-keto-PGE(2) in lung and express higher levels of ICAM-1 and PECAM-1 on pulmonary endothelial cells than GV(-/-) mice after lung infection with E. coli. Selective deletion of GV sPLA(2) in non-myeloid cells impairs leukocyte accumulation after pulmonary E. coli infection, and lack of GV sPLA(2) in either bone marrow-derived myeloid cells or non-myeloid cells attenuates E. coli clearance from the alveolar space and the lung parenchyma. These observations show that GV sPLA(2) in bone marrow-derived myeloid cells as well as non-myeloid cells, which are likely bronchial epithelial cells, participate in the regulation of the innate immune response to pulmonary infection with E. coli.


Subject(s)
Bone Marrow Cells/immunology , Bronchi/immunology , Epithelial Cells/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Group V Phospholipases A2/immunology , Immunity, Innate , Myeloid Cells/immunology , Pneumonia, Bacterial/immunology , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Bronchi/enzymology , Bronchi/pathology , Bronchoalveolar Lavage , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/enzymology , Epithelial Cells/pathology , Escherichia coli/metabolism , Escherichia coli Infections/enzymology , Escherichia coli Infections/genetics , Escherichia coli Infections/pathology , Group V Phospholipases A2/genetics , Group V Phospholipases A2/metabolism , Hydrolysis , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Knockout , Myeloid Cells/enzymology , Myeloid Cells/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pneumonia, Bacterial/enzymology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Prostaglandin D2/genetics , Prostaglandin D2/immunology , Prostaglandin D2/metabolism , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology
14.
Cell Mol Biol Lett ; 17(3): 459-78, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22706677

ABSTRACT

Secretory phospholipases A(2) (sPLA(2)s) are a diverse family of low molecular mass enzymes (13-18 kDa) that hydrolyze the sn-2 fatty acid ester bond of glycerophospholipids to produce free fatty acids and lysophospholipids. We have previously shown that group X sPLA(2) (sPLA(2)-X) had a strong hydrolyzing activity toward phosphatidylcholine in low-density lipoprotein (LDL) linked to the formation of lipid droplets in the cytoplasm of macrophages. Here, we show that group V sPLA(2) (sPLA(2)-V) can also cause the lipolysis of LDL, but its action differs remarkably from that of sPLA(2)-X in several respects. Although sPLA(2)-V released almost the same amount of fatty acids from LDL, it released more linoleic acid and less arachidonic acid than sPLA(2)-X. In addition, the requirement of Ca(2+) for the lipolysis of LDL was about 10-fold higher for sPLA(2)-V than sPLA(2)-X. In fact, the release of fatty acids from human serum was hardly detectable upon incubation with sPLA(2)-V in the presence of sodium citrate, which contrasted with the potent response to sPLA(2)-X. Moreover, sPLA(2)-X, but not sPLA(2)-V, was found to specifically interact with LDL among the serum proteins, as assessed by gel-filtration chromatography as well as sandwich enzyme-immunosorbent assay using anti-sPLA(2)-X and anti-apoB antibodies. Surface plasmon resonance studies have revealed that sPLA2-X can bind to LDL with high-affinity (K(d) = 3.1 nM) in the presence of Ca(2+). Selective interaction of sPLA(2)-X with LDL might be involved in the efficient hydrolysis of cell surface or intracellular phospholipids during foam cell formation.


Subject(s)
Arachidonic Acid , Group V Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Linoleic Acid , Lipoproteins, HDL , Lipoproteins, LDL , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Calcium/chemistry , Citrates/chemistry , Group V Phospholipases A2/chemistry , Group X Phospholipases A2/chemistry , Humans , Hydrolysis , Linoleic Acid/chemistry , Linoleic Acid/metabolism , Lipolysis , Lipoproteins , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Binding , Serum/chemistry , Serum/metabolism , Sodium Citrate , Surface Plasmon Resonance
15.
Am J Respir Cell Mol Biol ; 44(3): 361-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20448053

ABSTRACT

We examined the functional role of 14-kD secretory group V phospholipase A(2) (gVPLA(2)) on the barrier function of pulmonary endothelial cells (ECs) after LPS activation in vitro. Expression of gVPLA(2) was elicited by 20 ng/ml LPS as demonstrated by increased (1) mRNA, (2) protein content, and (3) cell surface expression of gVPLA(2) within 4 hours. The effect of LPS on EC barrier function was measured by transendothelial monolayer electrical resistance (TER). LPS increased permeability across EC monolayers at 2-3 hours, and was sustained for 10 hours or more. Blockade of gVPLA(2) with mouse monoclonal 3G1 (MCL-3G1) monoclonal antibody directed against gVPLA(2) inhibited EC barrier dysfunction elicited by LPS in a time- and concentration-dependent manner; control IgG had no effect on TER. Like LPS, exogenous gVPLA(2) caused increased EC permeability in a time- and concentration-dependent manner; neither gIIaPLA(2), a close homolog of gVPLA(2), nor W31A, an inactive mutant of gVPLA(2), caused a decrease in EC TER. Immunofluorescence analysis revealed comparable F-actin stress fiber and intercellular gap formation for ECs treated with either gVPLA(2) or LPS. Treatment with gVPLA(2) disrupted vascular endothelial-cadherin junctional complexes on ECs. Coincubation of ECs with MCL-3G1 substantially attenuated the structural changes caused by gVPLA(2) or LPS. We demonstrate that (1) gVPLA(2) is constitutively expressed in ECs and is up-regulated after LPS activation, (2) endogenously secreted gVPLA(2) from ECs after LPS increases EC permeability through F-actin and junctional complex rearrangement, and (3) inhibition of endogenous gVPLA(2) from ECs is sufficient to block disruption of the EC barrier function after LPS in vitro.


Subject(s)
Endothelial Cells/cytology , Group V Phospholipases A2/metabolism , Lipopolysaccharides/metabolism , Lung/enzymology , Actins/metabolism , Antibodies, Monoclonal/chemistry , Cells, Cultured , Dextrans/chemistry , Humans , In Vitro Techniques , Microcirculation , Microscopy, Fluorescence/methods , Mutation , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Time Factors
16.
J Biol Chem ; 285(14): 10993-1002, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20139082

ABSTRACT

Some isoforms of secretory phospholipase A(2) (sPLA(2)) distinguish between healthy and damaged or apoptotic cells. This distinction reflects differences in membrane physical properties. Because various sPLA(2) isoforms respond differently to properties of artificial membranes such as surface charge, they should also behave differently as these properties evolve during a dynamic physiological process such as apoptosis. To test this idea, S49 lymphoma cell death was induced by glucocorticoid (6-48 h) or calcium ionophore. Rates of membrane hydrolysis catalyzed by various concentrations of snake venom and human groups IIa, V, and X sPLA(2) were compared after each treatment condition. The data were analyzed using a model that evaluates the adsorption of enzyme to the membrane surface and subsequent binding of substrate to the active site. Results were compared temporally to changes in membrane biophysics and composition. Under control conditions, membrane hydrolysis was confined to the few unhealthy cells present in each sample. Increased hydrolysis during apoptosis and necrosis appeared to reflect substrate access to adsorbed enzyme for the snake venom and group X isoforms corresponding to weakened lipid-lipid interactions in the membrane. In contrast, apoptosis promoted initial adsorption of human groups V and IIa concurrent with phosphatidylserine exposure on the membrane surface. However, this observation was inadequate to explain the behavior of the groups V and IIa enzymes toward necrotic cells where hydrolysis was reduced or absent. Thus, a combination of changes in cell membrane properties during apoptosis and necrosis capacitates the cell for hydrolysis differently by each isoform.


Subject(s)
Apoptosis , Cell Membrane/pathology , Group II Phospholipases A2/metabolism , Group V Phospholipases A2/metabolism , Group X Phospholipases A2/metabolism , Lymphoma/pathology , Phospholipases A2, Secretory/metabolism , Anti-Inflammatory Agents/pharmacology , Calcium/metabolism , Cell Membrane/enzymology , Cell Membrane Permeability , Dexamethasone/pharmacology , Flow Cytometry , Humans , Hydrolysis , Ionophores/pharmacology , Kinetics , Lymphoma/enzymology , Membrane Fluidity , Necrosis , Snake Venoms/enzymology
17.
J Immunol ; 182(8): 4891-8, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342668

ABSTRACT

Phospholipase A(2) (PLA(2)) hydrolyzes the sn-2 position of cell membrane phospholipids to release fatty acids and lysophospholipids. We have previously reported that group V secretory PLA(2) (sPLA(2)) translocates from the Golgi and recycling endosomes of mouse peritoneal macrophages to newly formed phagosomes and regulates the phagocytosis of zymosan, suggesting a role in innate immunity. Here we report that in macrophages lacking group V sPLA(2), phagosome maturation was reduced 50-60% at early time points while the binding of zymosan was unimpaired. The ability of group V sPLA(2) to regulate phagocytosis extended to phagocytosis of IgG- and complement-opsonized sheep RBC. Moreover, macrophages lacking group V sPLA(2) had delays in phagocytosis, phagosome maturation, and killing of Candida albicans. Cytokine production and eicosanoid generation were not impaired by the lack of group V sPLA(2). Furthermore, in a model of systemic candidiasis, mice lacking group V sPLA(2) had an increased fungal burden in the kidney, liver, and spleen at day 7 postinfection and increased mortality. Thus, group V sPLA(2) regulates phagocytosis through major phagocytic receptors and contributes to the innate immune response against C. albicans by regulating phagocytosis and killing through a mechanism that is likely dependent on phagolysosome fusion.


Subject(s)
Candida albicans/immunology , Group V Phospholipases A2/metabolism , Immunity, Innate/immunology , Phagosomes/enzymology , Phagosomes/immunology , Animals , Candidiasis/genetics , Candidiasis/immunology , Candidiasis/metabolism , Candidiasis/pathology , Group V Phospholipases A2/deficiency , Group V Phospholipases A2/genetics , Lectins, C-Type , Macrophages/enzymology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Phagocytosis , Survival Rate , Tumor Necrosis Factor-alpha/biosynthesis , Zymosan/metabolism
18.
J Lipid Res ; 51(7): 1801-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20124257

ABSTRACT

LDL particles that enter the arterial intima become exposed to proteolytic and lipolytic modifications. The extracellular hydrolases potentially involved in LDL modification include proteolytic enzymes, such as chymase, cathepsin S, and plasmin, and phospholipolytic enzymes, such as secretory phospholipases A2 (sPLA2-IIa and sPLA2-V) and secretory acid sphingomyelinase (sSMase). Here, LDL was first proteolyzed and then subjected to lipolysis, after which the effects of combined proteolysis and lipolysis on LDL fusion and on binding to human aortic proteoglycans (PG) were studied. Chymase and cathepsin S led to more extensive proteolysis and release of peptide fragments from LDL than did plasmin. sPLA2-IIa was not able to hydrolyze unmodified LDL, and even preproteolysis of LDL particles failed to enhance lipolysis by this enzyme. However, preproteolysis with chymase and cathepsin S accelerated lipolysis by sPLA2-V and sSMase, which resulted in enhanced fusion and proteoglycan binding of the preproteolyzed LDL particles. Taken together, the results revealed that proteolysis sensitizes the LDL particles to hydrolysis by sPLA2-V and sSMase. By promoting fusion and binding of LDL to human aortic proteoglycans, the combination of proteolysis and phospholipolysis of LDL particles potentially enhances extracellular accumulation of LDL-derived lipids during atherogenesis.


Subject(s)
Group V Phospholipases A2/metabolism , Lipoproteins, LDL/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Aorta/metabolism , Cathepsins/metabolism , Chymases/metabolism , Fibrinolysin/metabolism , Humans , Hydrolysis , Lipolysis , Particle Size , Proteoglycans/metabolism
19.
J Cell Physiol ; 224(1): 127-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20232296

ABSTRACT

Secreted phospholipases A(2) (sPLA(2)s) are well known for their contribution in the biosynthesis of inflammatory eicosanoids. These enzymes also participate in the inflammatory process by regulating chemokine production and protein expression of adhesion molecules. The majority of sPLA(2) isoforms are up-regulated by proinflammatory stimuli such as bacterial lipopolysaccharide (LPS), which predominantly increases the expression of group V sPLA(2) (sPLA(2)-V). Furthermore, it has recently been shown that sPLA(2)-V is a critical messenger in the regulation of cell migration during allergic airway responsiveness. Herein, we investigated the effect of sPLA(2)-V on LPS-mediated leukocyte recruitment and its capacity to modulate adhesion molecule expression. We conducted our study in the murine air pouch model, using sPLA(2)-V null mice (sPLA(2)-V(-/-)) and control wild-type (WT) littermates. We observed that LPS (1 microg/ml)-mediated leukocyte emigration in sPLA(2)-V(-/-) was attenuated by 52% and 86% upon 6 and 12 h of treatment respectively, as compared to WT mice. In WT mice, treatment with the cell-permeable sPLA(2) inhibitor (12-epi-scalaradial; SLD) reduced LPS-mediated leukocyte recruitment by 67%, but had no additional inhibitory effect in sPLA(2)-V(-/-) mice. Protein analyses from the air pouch skin were carried out upon LPS-challenge, and the expression of intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 were both significantly reduced in sPLA(2)-V(-/-) mice as compared to control WT mice. Together, our data demonstrate the role of sPLA(2)-V in LPS-induced ICAM-1 and VCAM-1 protein overexpression and leukocyte recruitment, supporting the contribution of sPLA(2)-V in the development of inflammatory innate immune responses.


Subject(s)
Chemotaxis, Leukocyte , Group V Phospholipases A2/metabolism , Inflammation Mediators/metabolism , Inflammation/enzymology , Leukocytes/enzymology , Neutrophil Infiltration , Animals , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Group V Phospholipases A2/antagonists & inhibitors , Group V Phospholipases A2/deficiency , Group V Phospholipases A2/genetics , Immunity, Innate , Inflammation/chemically induced , Inflammation/immunology , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/drug effects , Leukocytes/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism
20.
Cytokine ; 50(1): 50-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20138782

ABSTRACT

OBJECTIVE: Previous studies have established that hydrolysis of LDL by Group V secretory phospholipase A(2) (GV sPLA(2)) generates a modified particle capable of inducing macrophage foam cell formation. The aim of the present study was to determine whether GV sPLA(2)-hydrolyzed LDL (GV-LDL) produces pro-atherogenic effects in macrophages independent of cholesterol accumulation. METHODS AND RESULTS: J-774 cells incubated with GV-LDL produced more TNF-alpha and IL-6 compared to cells incubated with control-LDL. Indirect immunofluorescence showed that GV-LDL but not control-LDL induced nuclear translocation of NFkappaB. Inhibitors of NFkappaB activation, effectively blocked cytokine production induced by GV-LDL. Control-LDL and GV-LDL were separated from albumin present in reaction mixtures by ultracentrifugation. The albumin fraction derived from GV-LDL contained 80% of the FFA generated and was more potent than the re-isolated GV-LDL in inducing pro-inflammatory cytokine secretion. Linoleic acid (18:2) and oleic acid (18:1) were the most abundant FFAs generated, whereas newly formed lyso-PCs contained 14:0 (myristic), 16:1 (palmitic), and 18:2 fatty acyl groups. Experiments with synthetic FFA showed that 18:1 induced J-774 cells to secrete TNF-alpha and IL-6. CONCLUSIONS: These results indicate that in addition to promoting atherosclerotic lipid accumulation in macrophages, GV sPLA(2) hydrolysis of LDL leads to activation of NFkappaB, a key regulator of inflammation.


Subject(s)
Cholesterol, LDL/metabolism , Group V Phospholipases A2/metabolism , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Macrophage Activation , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , Albumins/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Fatty Acids/metabolism , Humans , Hydrolysis , Lipolysis , Mice , NF-kappa B/antagonists & inhibitors , Protein Binding , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL