Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 206(4): 722-736, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33441441

ABSTRACT

Eosinophils mediate pathological manifestations during tropical pulmonary eosinophilia (TPE), a potentially fatal complication of lymphatic filariasis, by mechanisms that are incompletely understood. Using two-dimensional gel electrophoresis, mass spectrometry, flow cytometry, and pharmacological and functional studies, we identified acidic calcium-independent phospholipase A2 (aiPLA2) as the master regulator of TPE pathogenesis. FACS-sorted lung eosinophils from TPE mice exhibited aiPLA2-dependent activation characterized by heavy calcium influx, F-actin polymerization, increased degranulation, and heightened reactive oxygen species generation. Interestingly, aiPLA2 also promoted alternative activation in lung macrophages and regulated the release of inflammatory intermediates from them. Treatment of TPE mice with MJ33, a nontoxic pharmacological inhibitor of aiPLA2, lowered eosinophil counts in the bronchoalveolar lavage fluid, reduced eosinophil peroxidase and ß-hexosaminidase activity, increased airway width, improved lung endothelial barrier, and lowered the production of inflammatory lipid intermediates, which significantly improved the pathological condition of the lungs. Importantly, ex vivo reconstitution of arachidonic acid to eosinophils from MJ33-treated TPE mice increased eosinophil degranulation and inflammatory lipid intermediates underlining the pivotal role of aiPLA2 in arachidonic acid metabolism. Mechanistically, phosphorylation of JNK-1 regulated phospholipase activity of aiPLA2, whereas IgG cross-linking mediated pathological activation of eosinophils. Taken together, ours is the first study, to our knowledge, to report hitherto undocumented role of aiPLA2 in regulating TPE pathogenesis.


Subject(s)
Brugia malayi/immunology , Elephantiasis, Filarial/immunology , Eosinophils/immunology , Group VI Phospholipases A2/immunology , Macrophages/immunology , Pulmonary Eosinophilia/immunology , Animals , Disease Models, Animal , Elephantiasis, Filarial/pathology , Eosinophils/pathology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Pulmonary Eosinophilia/parasitology , Pulmonary Eosinophilia/pathology
2.
J Biol Chem ; 291(44): 23268-23281, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27650501

ABSTRACT

Macrophages are important in innate and adaptive immunity. Macrophage participation in inflammation or tissue repair is directed by various extracellular signals and mediated by multiple intracellular pathways. Activation of group VIA phospholipase A2 (iPLA2ß) causes accumulation of arachidonic acid, lysophospholipids, and eicosanoids that can promote inflammation and pathologic states. We examined the role of iPLA2ß in peritoneal macrophage immune function by comparing wild type (WT) and iPLA2ß-/- mouse macrophages. Compared with WT, iPLA2ß-/- macrophages exhibited reduced proinflammatory M1 markers when classically activated. In contrast, anti-inflammatory M2 markers were elevated under naïve conditions and induced to higher levels by alternative activation in iPLA2ß-/- macrophages compared with WT. Induction of eicosanoid (12-lipoxygenase (12-LO) and cyclooxygenase 2 (COX2))- and reactive oxygen species (NADPH oxidase 4 (NOX4))-generating enzymes by classical activation pathways was also blunted in iPLA2ß-/- macrophages compared with WT. The effects of inhibitors of iPLA2ß, COX2, or 12-LO to reduce M1 polarization were greater than those to enhance M2 polarization. Certain lipids (lysophosphatidylcholine, lysophosphatidic acid, and prostaglandin E2) recapitulated M1 phenotype in iPLA2ß-/- macrophages, but none tested promoted M2 phenotype. These findings suggest that (a) lipids generated by iPLA2ß and subsequently oxidized by cyclooxygenase and 12-LO favor macrophage inflammatory M1 polarization, and (b) the absence of iPLA2ß promotes macrophage M2 polarization. Reducing macrophage iPLA2ß activity and thereby attenuating macrophage M1 polarization might cause a shift from an inflammatory to a recovery/repair milieu.


Subject(s)
Cell Polarity , Group VI Phospholipases A2/immunology , Inflammation/enzymology , Macrophages/cytology , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Female , Group VI Phospholipases A2/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Macrophages/enzymology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/immunology
3.
J Trauma Acute Care Surg ; 79(2): 238-46, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26218692

ABSTRACT

BACKGROUND: The role of calcium-independent phospholipase A2 (iPLA2), a component of the three major PLA2 families, in acute/chronic inflammatory processes remains elusive. Previous investigations have documented iPLA2-mediated respiratory burst of neutrophils (PMNs); however, the causative isoform of iPLA2 is unidentified. We also demonstrated that the iPLA2γ-specific inhibitor attenuates trauma/hemorrhagic shock-induced lung injury. Therefore, iPLA2γ may be implicated in acute inflammation. In addition, arachidonic acid (AA), which is primarily produced by cytosolic PLA2 (cPLA2), is known to display PMN cytotoxicity, although the relationship between AA and the cytotoxic function is still being debated on. We therefore hypothesized that iPLA2γ regulates PMN cytotoxicity via AA-independent signaling pathways. The study aim was to distinguish the role of intracellular phospholipases A2, iPLA2, and cPLA2, in human PMN cytotoxicity and explore the possibility of the presence of signaling molecule(s) other than AA. METHODS: Isolated human PMNs were incubated with the PLA2 inhibitor selective for iPLA2ß, iPLA2γ, or cPLA2 and then activated with formyl-methionyl-leucyl-phenylalanine (fMLP) or phorbol 12-myristate 13-acetate (PMA). Superoxide production was assayed according to the superoxide dismutase-inhibitable cytochrome c reduction method, and the degree of elastase release was measured using a p-nitroanilide-conjugated elastase-specific substrate. In addition, chemotaxis toward platelet activating factor/fMLP was determined with a modified Boyden chamber system. RESULTS: The iPLA2γ-specific inhibitor reduced the fMLP/PMA-stimulated superoxide generation by 90% and 30%, respectively; in addition, the inhibitor completely blocked the fMLP/PMA-activated elastase release. However, the cPLA2-specific inhibitor did not abrogate these effects to any degree at all concentrations. Likewise, the inhibitor for iPLA2γ, but not iPLA2ß or cPLA2, completely inhibited the platelet activating factor/fMLP-induced chemotaxis. CONCLUSION: iPLA2 is involved in extracellular reactive oxygen species production, elastase release, and chemotaxis in response to well-defined stimuli. In addition, the ineffectiveness of the cPLA2 inhibitor suggests that AA may not be relevant to these cytotoxic functions.


Subject(s)
Cytotoxicity, Immunologic/immunology , Group VI Phospholipases A2/immunology , Neutrophils/immunology , Phospholipases A2/immunology , Cells, Cultured , Chemotaxis/immunology , Group VI Phospholipases A2/antagonists & inhibitors , Humans , Intracellular Space/immunology , Pancreatic Elastase/metabolism , Phospholipases A2, Cytosolic/antagonists & inhibitors , Phospholipases A2, Cytosolic/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Endocrinology ; 155(9): 3352-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25004092

ABSTRACT

Type 1 diabetes (T1D) results from autoimmune destruction of islet ß-cells, but the underlying mechanisms that contribute to this process are incompletely understood, especially the role of lipid signals generated by ß-cells. Proinflammatory cytokines induce ER stress in ß-cells and we previously found that the Ca(2+)-independent phospholipase A2ß (iPLA2ß) participates in ER stress-induced ß-cell apoptosis. In view of reports of elevated iPLA2ß in T1D, we examined if iPLA2ß participates in cytokine-mediated islet ß-cell apoptosis. We find that the proinflammatory cytokine combination IL-1ß+IFNγ, induces: a) ER stress, mSREBP-1, and iPLA2ß, b) lysophosphatidylcholine (LPC) generation, c) neutral sphingomyelinase-2 (NSMase2), d) ceramide accumulation, e) mitochondrial membrane decompensation, f) caspase-3 activation, and g) ß-cell apoptosis. The presence of a sterol regulatory element in the iPLA2ß gene raises the possibility that activation of SREBP-1 after proinflammatory cytokine exposure contributes to iPLA2ß induction. The IL-1ß+IFNγ-induced outcomes (b-g) are all inhibited by iPLA2ß inactivation, suggesting that iPLA2ß-derived lipid signals contribute to consequential islet ß-cell death. Consistent with this possibility, ER stress and ß-cell apoptosis induced by proinflammatory cytokines are exacerbated in islets from RIP-iPLA2ß-Tg mice and blunted in islets from iPLA2ß-KO mice. These observations suggest that iPLA2ß-mediated events participate in amplifying ß-cell apoptosis due to proinflammatory cytokines and also that iPLA2ß activation may have a reciprocal impact on ER stress development. They raise the possibility that iPLA2ß inhibition, leading to ameliorations in ER stress, apoptosis, and immune responses resulting from LPC-stimulated immune cell chemotaxis, may be beneficial in preserving ß-cell mass and delaying/preventing T1D evolution.


Subject(s)
Apoptosis , Cytokines/immunology , Diabetes Mellitus, Type 1/enzymology , Group VI Phospholipases A2/immunology , Interferon-gamma/immunology , Interleukin-1beta/immunology , Islets of Langerhans/cytology , Adult , Animals , Cytokines/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Endoplasmic Reticulum Stress , Female , Group VI Phospholipases A2/genetics , Humans , Interferon-gamma/genetics , Interleukin-1beta/genetics , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Male , Mice , Mice, Knockout , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology
5.
Am J Physiol Renal Physiol ; 302(7): F865-74, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22218592

ABSTRACT

Na(+)-K(+)-2Cl(-) cotransporter (NKCC2)-mediated NaCl reabsorption in the thick ascending limb (TAL) is stimulated by AVP via V2 receptor/PKA/cAMP signaling. This process is antagonized by locally produced eicosanoids such as 20-HETE or prostaglandin E(2), which are synthesized in a phospholipase A(2)-dependent reaction cascade. Using microarray-based gene expression analysis, we found evidence for an AVP-dependent downregulation of the calcium-independent isoform of PLA(2), iPLA(2)ß, in the outer medulla of rats. In the present study, we therefore examined the contribution of iPLA(2)ß to NKCC2 regulation. Immunoreactive iPLA(2)ß protein was detected in cultured mTAL cells as well as in the entire TAL of rodents and humans with the exception of the macula densa. Administration of the V2 receptor-selective agonist desmopressin (5 ng/h; 3 days) to AVP-deficient diabetes insipidus rats increased outer medullary phosphorylated NKCC2 (pNKCC2) levels more than twofold in association with a marked reduction in iPLA(2)ß abundance (-65%; P < 0.05), thus confirming microarray results. Inhibition of iPLA(2)ß in Sprague-Dawley rats with FKGK 11 (0.5 µM) or in mTAL cells with FKGK 11 (10 µM) or (S)-bromoenol lactone (5 µM) for 1 h markedly increased pNKCC2 levels without affecting total NKCC2 expression. Collectively, these data indicate that iPLA(2)ß acts as an inhibitory modulator of NKCC2 activity and suggest that downregulation of iPLA(2)ß may be a relevant step in AVP-mediated urine concentration.


Subject(s)
Group VI Phospholipases A2/metabolism , Loop of Henle/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Vasopressins/metabolism , Animals , Antibodies , Arachidonic Acids , Cells, Cultured , Deamino Arginine Vasopressin , Down-Regulation , Fluorocarbons , Gene Expression , Group VI Phospholipases A2/immunology , Guinea Pigs , Humans , Isoenzymes , Ketones , Kidney Medulla/metabolism , Male , Mice , Mice, Inbred C57BL , Naphthalenes , Organophosphonates , Phosphorylation , Pyrones , Rats , Rats, Brattleboro , Rats, Sprague-Dawley , Solute Carrier Family 12, Member 1
6.
J Exp Med ; 205(2): 347-59, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18208975

ABSTRACT

Monocyte chemoattractant protein-1 (MCP-1) directs migration of blood monocytes to inflamed tissues. Despite the central role of chemotaxis in immune responses, the regulation of chemotaxis by signal transduction pathways and their in vivo significance remain to be thoroughly deciphered. In this study, we examined the intracellular location and functions of two recently identified regulators of chemotaxis, Ca(2+)-independent phospholipase (iPLA(2)beta) and cytosolic phospholipase (cPLA(2)alpha), and substantiate their in vivo importance. These enzymes are cytoplasmic in unstimulated monocytes. Upon MCP-1 stimulation, iPLA(2)beta is recruited to the membrane-enriched pseudopod. In contrast, cPLA(2)alpha is recruited to the endoplasmic reticulum. Although iPLA(2)beta or cPLA(2)alpha antisense oligodeoxyribonucleotide (ODN)-treated monocytes display reduced speed, iPLA(2)beta also regulates directionality and actin polymerization. iPLA(2)beta or cPLA(2)alpha antisense ODN-treated adoptively transferred mouse monocytes display a profound defect in migration to the peritoneum in vivo. These converging observations reveal that iPLA(2)beta and cPLA(2)alpha regulate monocyte migration from different intracellular locations, with iPLA(2)beta acting as a critical regulator of the cellular compass, and identify them as potential targets for antiinflammatory strategies.


Subject(s)
Chemokine CCL2/immunology , Group IV Phospholipases A2/immunology , Group VI Phospholipases A2/immunology , Monocytes/immunology , Actins/metabolism , Adoptive Transfer , Animals , Arachidonic Acids/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/immunology , Female , Group IV Phospholipases A2/antagonists & inhibitors , Group VI Phospholipases A2/antagonists & inhibitors , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Naphthalenes/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Peritonitis/chemically induced , Peritonitis/immunology , Pyrones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...