Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 304(10): L689-700, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525785

ABSTRACT

Ventilation at high tidal volume may cause lung inflammation and barrier dysfunction that culminates in ventilator-induced lung injury (VILI). However, the mechanisms by which mechanical stimulation triggers the inflammatory response have not been fully elucidated. This study tested the hypothesis that onset of VILI is triggered by activation of secretory group V phospholipase A(2) (gVPLA2) in pulmonary vascular endothelium exposed to excessive mechanical stretch. High-magnitude cyclic stretch (18% CS) increased expression and surface exposure of gVPLA2 in human pulmonary endothelial cells (EC). CS-induced gVPLA2 activation was required for activation of ICAM-1 expression and polymorphonuclear neutrophil (PMN) adhesion to CS-preconditioned EC. By contrast, physiological CS (5% CS) had no effect on gVPLA2 activation or EC-PMN adhesion. CS-induced ICAM-1 expression and EC-PMN adhesion were attenuated by the gVPLA2-blocking antibody (MCL-3G1), general inhibitor of soluble PLA2, LY311727, or siRNA-induced EC gVPLA2 knockdown. In vivo, ventilator-induced lung leukocyte recruitment, cell and protein accumulation in the alveolar space, and total lung myeloperoxidase activity were strongly suppressed in gVPLA2 mouse knockout model or upon administration of MCL-3G1. These results demonstrate a novel role for gVPLA2 as the downstream effector of pathological mechanical stretch leading to an inflammatory response associated with VILI.


Subject(s)
Acute Lung Injury/enzymology , Phospholipases A2/biosynthesis , Pneumonia/enzymology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Induction , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/enzymology , Leukocytes/metabolism , Leukocytes/pathology , Lung/enzymology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/metabolism , Pneumonia/pathology , Stress, Mechanical , Tidal Volume/physiology , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
2.
Pulm Circ ; 2(2): 182-92, 2012.
Article in English | MEDLINE | ID: mdl-22837859

ABSTRACT

Acute lung injury (ALI) is characterized by inflammatory disruption of the alveolar-vascular barrier, resulting in severe respiratory compromise. Inhibition of the intercellular messenger protein, Group V phospholipase A(2) (gVPLA(2)), blocks vascular permeability caused by LPS both in vivo and in vitro. In this investigation we studied the mechanism by which recombinant gVPLA(2) increases permeability of cultured human pulmonary endothelial cells (EC). Exogenous gVPLA(2) (500 nM), a highly hydrolytic enzyme, caused a significant increase in EC permeability that began within minutes and persisted for >10 hours. However, the major hydrolysis products of gVPLA(2) (Lyso-PC, Lyso-PG, LPA, arachidonic acid) did not cause EC structural rearrangement or loss of barrier function at concentrations <10 µM. Higher concentrations (≥ 30 µM) of these membrane hydrolysis products caused some increased permeability but were associated with EC toxicity (measured by propidium iodide incorporation) that did not occur with barrier disruption by gVPLA(2) (500 nM). Pharmacologic inhibition of multiple intracellular signaling pathways induced by gVPLA(2) activity (ERK, p38, PI3K, cytosolic gIVPLA(2)) also did not prevent EC barrier disruption by gVPLA(2). Finally, pretreatment with heparinase to prevent internalization of gVPLA(2) did not inhibit EC barrier disruption by gVPLA(2). Our data thus indicate that gVPLA(2) increases pulmonary EC permeability directly through action as a membrane hydrolytic agent. Disruption of EC barrier function does not depend upon membrane hydrolysis products, gVPLA(2) internalization, or upregulation of downstream intracellular signaling.

3.
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
4.
J Inflamm (Lond) ; 7: 14, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20298597

ABSTRACT

BACKGROUND: Cytosolic gIVaPLA2 is a critical enzyme in the generation of arachidonate metabolites and in induction of beta2-integrin adhesion in granulocytes. We hypothesized that gIVaPLA2 activation also is an essential downstream step for post adhesive migration of PMN in vitro. METHODS: Migration of PMNs caused by IL-8/CXCL8 was assessed using a transwell migration chamber. PMNs were pretreated with two structurally unrelated inhibitors of gIVaPLA2, arachidonyl trifluoromethylketone (TFMK) or pyrrophenone, prior to IL-8/CXCL8 exposure. The fraction of migrated PMNs present in the lower chamber was measured as total myeloperoxidase content. GIVaPLA2 enzyme activity was analyzed using [14C-PAPC] as specific substrate F-actin polymerization and cell structure were examined after rhodamine-phalloidin staining. RESULTS: IL-8/CXCL8-induced migration of PMNs was elicited in concentration- and time-dependent manner. Time-related phosphorylation and translocation of cytosolic gIVaPLA2 to the nucleus was observed for PMNs stimulated with IL-8/CXCL8 in concentration sufficient to cause upstream phosphorylation of MAPKs (ERK-1/2 and p38) and Akt/PKB. Inhibition of gIVaPLA2 corresponded to the magnitude of blockade of PMN migration. Neither AA nor LTB4 secretion was elicited following IL-8/CXCL8 activation. In unstimulated PMNs, F-actin was located diffusely in the cytosol; however, a clear polarized morphology with F-actin-rich ruffles around the edges of the cell was observed after activation with IL-8/CXCL8. Inhibition of gIVaPLA2 blocked change in cell shape and migration caused by IL-8/CXCL8 but did not cause F-actin polymerization or translocation of cytosolic F-actin to inner leaflet of the PMN membrane. CONCLUSION: We demonstrate that IL-8/CXCL8 causes a) phosphorylation and translocation of cytosolic gIVaPLA2 to the nucleus, b) change in cell shape, c) polymerization of F-actin, and d) chemoattractant/migration of PMN in vitro. Inhibition of gIVaPLA2 blocks the deformability and subsequent migration of PMNs caused by IL-8/CXCL8. Our data suggest that activation of gIVaPLA2 is an essential step in PMN migration in vitro.

5.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L879-87, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19286925

ABSTRACT

We investigated the regulatory role of 14-kDa secretory group V phospholipase A(2) (gVPLA(2)) in the development of acute lung injury (ALI) and neutrophilic inflammation (NI) caused by intratracheal administration of LPS. Experiments were conducted in gVPLA(2) knockout (pla2g5(-/-)) mice, which lack the gene, and gVPLA(2) wild-type littermate control (pla2g5(+/+)) mice. Indices of pulmonary injury were evaluated 24 h after intratracheal administration of LPS. Expression of gVPLA(2) in microsections of airways and mRNA content in lung homogenates were increased substantially in pla2g5(+/+) mice after LPS-administered compared with saline-treated pla2g5(+/+) mice. By contrast, expression of gVPLA(2) was neither localized in LPS- nor saline-treated pla2g5(-/-) mice. LPS also caused 1) reduced transthoracic static compliance, 2) lung edema, 3) neutrophilic infiltration, and 4) increased neutrophil myeloperoxidase activity in pla2g5(+/+) mice. These events were attenuated in pla2g5(-/-) mice exposed to LPS or in pla2g5(+/+) mice receiving MCL-3G1, a neutralizing MAb directed against gVPLA(2), before LPS administration. Our data demonstrate that gVPLA(2) is an inducible protein in pla2g5(+/+) mice but not in pla2g5(-/-) mice within 24 h after LPS treatment. Specific inhibition of gVPLA(2) with MCL-3G1 or gene-targeted mice lacking gVPLA(2) blocks ALI and attenuates NI caused by LPS.


Subject(s)
Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Group V Phospholipases A2/genetics , Group V Phospholipases A2/metabolism , Neutrophils/immunology , Acute Lung Injury/chemically induced , Animals , Antibodies, Monoclonal/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Extravascular Lung Water/metabolism , Group V Phospholipases A2/immunology , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/metabolism , Lung/pathology , Lung Compliance/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Peroxidase/metabolism , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/metabolism , RNA, Messenger/metabolism , Second Messenger Systems/immunology
6.
J Leukoc Biol ; 83(2): 344-51, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17971499

ABSTRACT

We examined the effect of glucocorticoid stimulation in blocking beta 2-integrin adhesion of polymorphonuclear leukocytes (PMNs) isolated from human subjects. Surface expression of CD11b and ERK-1/2-mediated gIVaPLA2 phosphorylation, which are required for beta 2-integrin adhesion, were not affected by treatment with < or =10(-6) M fluticasone propionate (FP) for PMNs activated by either 10(-7) M LTB4 or 30 ng/ml TNF-alpha and caused no significant blockade of beta 2-integrin adhesion in vitro. Baseline expression of annexin-1 (ANXA1) synthesis was increased only after 10(-6) M FP for PMNs; by contrast, comparable increase in ANXA1 expression was demonstrated in human eosinophils from the same subjects with 10(-8) M FP. Viability of PMNs was verified by propidium iodide and by the persistence of beta 2-integrin adhesion in treated groups. Exogenous administration of ANXA1 mimetic peptide fragment blocked significantly and comparably the beta 2-integrin adhesion in PMNs activated by LTB4 and TNF-alpha and in eosinophils activated by IL-5. Translocation of gIVaPLA2 from the cytosol to the nucleus also was refractory for activated PMNs treated with > or =10(-7) M FP; by contrast, complete blockade of nuclear translocation of cytosolic gIVaPLA2 was effected by 10(-9) M FP in eosinophils. Our data indicate that the cell surface ANXA1 synthesis is capable of blocking beta 2-integrin adhesion in both PMNs and eosinophils. However, in contrast to eosinophils, FP does not cause either substantial ANXA1 synthesis or nuclear transport of cytosolic gIVaPLA2 in PMNs and thus does not block beta2-integrin adhesion, a necessary step for granulocyte cell migration in vivo.


Subject(s)
Androstadienes/pharmacology , Annexin A1/physiology , CD18 Antigens/physiology , Cell Adhesion/drug effects , Neutrophils/drug effects , Active Transport, Cell Nucleus/drug effects , Adult , Annexin A1/antagonists & inhibitors , Annexin A1/pharmacology , Cell Nucleus/enzymology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Cytosol/enzymology , Eosinophils/cytology , Eosinophils/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Fluticasone , Group IV Phospholipases A2 , Humans , Hypersensitivity, Immediate/blood , Interleukin-5/pharmacology , Leukotriene B4/pharmacology , Middle Aged , Neutrophils/cytology , Peptides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
7.
Proc Am Thorac Soc ; 4(8): 659-66, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18073398

ABSTRACT

Therapies currently used to reduce exacerbations of chronic obstructive pulmonary disease (COPD) are compounds used almost entirely for asthma therapy. A notable exception is tiotropium, a long-acting parasympatholytic agent. This compound and its precursor, iprotropium, are only occasionally used for asthma therapy. Likewise, leukotriene-modifying drugs are used occasionally for the treatment of COPD. In neither circumstance is there agency-approved indication for these particular cross-over therapies, but the use of long-acting beta(2)-adrenergic compounds and high-solubility inhaled steroids is a mainstay for therapy in both asthma and COPD. Similarly, theophylline, although less often used for either process, is therapeutically applicable to both asthma and COPD. Although overlap syndromes point to the occurrence of a common pathway in some cases, the inflammatory process for asthma and chronic obstructive pulmonary disease (COPD) differs substantially in most cases. Hence, the application of therapies designed to relax airway smooth muscle and ameliorate asthmatic inflammation lacks a therapeutic rationale for a disease characterized by predominant neutrophilic inflammation occurring in the small airways and alveoli. By definition, COPD is poorly reversible airflow obstruction; hence, the use of drugs designed to relax airway smooth muscle is somewhat counterintuitive and does not address the pathophysiological process of the disease.


Subject(s)
Pulmonary Disease, Chronic Obstructive/metabolism , Annexin A1/metabolism , Anti-Inflammatory Agents/pharmacology , Group IV Phospholipases A2/metabolism , Humans , Inflammation/metabolism , Integrins/physiology , Neutrophils/metabolism , Phospholipases A2/metabolism , Phosphorylation , Transduction, Genetic , ras Proteins/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/metabolism
8.
J Allergy Clin Immunol ; 120(6): 1316-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17904626

ABSTRACT

BACKGROUND: Cysteinyl leukotriene (cysLT) antagonism attenuates migration of eosinophils into airways during immune challenge in human subjects and animal models. The intracellular signaling mechanism by which this occurs has not been elucidated. OBJECTIVE: We sought to determine the relative efficacy and mechanism by which 5-lipoxygenase (5-LO) inhibition and cysLT(1) receptor (cysLT(1)R) antagonism block beta(2)-integrin adhesion in isolated human eosinophils in vitro. METHODS: Human blood eosinophils were isolated by means of immunomagnetic separation. Upregulation of CD11b expression, active conformation of CD11b, and focal clustering of beta(2)-integrin caused by IL-5, eotaxin-1 or leukotriene (LT) B(4) was assessed by means of flow cytometry and confocal microscopy. The effect and mechanism of cysLT(1)R or 5-LO blockade on these components of beta(2)-integrin adhesion were determined. RESULTS: Montelukast, a cysLT(1)R antagonist, and AA861, a 5-LO enzyme inhibitor, blocked (1) avidity of beta(2)-integrin, (2) beta(2)-integrin-mediated adhesion to intercellular adhesion molecule 1, and (3) focal clustering of CD11b elicited by LTB(4). However, adhesion caused by either IL-5 or eotaxin-1 was not attenuated for eosinophils pretreated with either montelukast or AA861. CONCLUSION: Our data demonstrate that (1) LTB(4) causes autocrine upregulation of adhesion through secretion of cysLTs, and (2) blockade of cysLT(1)R blocks the avidity and focal clustering of CD11b/CD18 for eosinophils activated by LTB(4) but not by IL-5 or eotaxin-1. CLINICAL IMPLICATIONS: Unlike cysLT-induced adhesion, adhesion caused by IL-5 or eotaxin-1 is not regulated through the cysLT(1)R, suggesting that cysLTs have specific but limited potential to upregulate eosinophil adhesion.


Subject(s)
CD18 Antigens/metabolism , Cysteine/antagonists & inhibitors , Eosinophils/immunology , Eosinophils/metabolism , Inflammation Mediators/antagonists & inhibitors , Leukotriene Antagonists/pharmacology , Acetates/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Benzoquinones/pharmacology , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , CD18 Antigens/physiology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cells, Cultured , Chemokine CCL11/physiology , Cyclopropanes , Cysteine/physiology , Eosinophils/drug effects , Eosinophils/enzymology , Humans , Inflammation Mediators/physiology , Interleukin-5/physiology , Leukotriene B4/physiology , Leukotrienes/physiology , Lipoxygenase Inhibitors/pharmacology , Quinolines/pharmacology , Sulfides
9.
J Immunol ; 179(7): 4800-7, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878379

ABSTRACT

We investigated the role of group V phospholipase A2 (gVPLA2) in OVA-induced inflammatory cell migration and airway hyperresponsiveness (AHR) in C57BL/6 mice. Repeated allergen challenge induced biosynthesis of gVPLA2 in airways. By aerosol, gVPLA2 caused dose-related increase in airway resistance in saline-treated mice; in allergic mice, gVPLA2 caused persistent airway narrowing. Neither group IIa phospholipase A2, a close homolog of gVPLA2, nor W31A, an inactive gVPLA2 mutant with reduced activity, caused airway narrowing in immune-sensitized mice. Pretreatment with MCL-3G1, a blocking Ab against gVPLA2, before OVA challenge blocked fully gVPLA2-induced cell migration and airway narrowing as marked by reduction of migrating leukocytes in bronchoalveolar lavage fluid and decreased airway resistance. We also assessed whether nonspecific AHR caused by methacholine challenge was elicited by gVPLA2 secreted from resident airway cells of immune-sensitized mice. MCL-3G1 also blocked methacholine-induced airway bronchoconstriction in allergic mice. Blockade of bronchoconstriction by MCL-3G1 was replicated in allergic pla2g5-/- mice, which lack the gene encoding gVPLA2. Bronchoconstriction caused by gVPLA2 in pla2g4-/- mice was comparable to that in pla2g4+/+ mice. Our data demonstrate that gVPLA2 is a critical messenger enzyme in the development of AHR and regulation of cell migration during immunosensitization by a pathway that is independent of group IVa phospholipase A2.


Subject(s)
Cell Movement/immunology , Ovalbumin/immunology , Phospholipases A/deficiency , Phospholipases A/metabolism , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/pathology , Animals , Antibodies/immunology , Antigens/immunology , Gene Expression Regulation, Enzymologic , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Methacholine Chloride/pharmacology , Mice , Mice, Knockout , Phospholipases A/genetics , Phospholipases A2 , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/immunology , Up-Regulation
10.
Am J Respir Cell Mol Biol ; 36(5): 585-93, 2007 May.
Article in English | MEDLINE | ID: mdl-17218614

ABSTRACT

We examined the mechanism by which lysophosphatidylcholine (LPC) regulates beta2-integrin-mediated adhesion of eosiniophils. Eosinophils were isolated from blood of mildly atopic volunteers by negative immunomagnetic selection. beta2-integrin-dependent adhesion of eosinophils to plated bovine serum albumin (BSA) was measured by residual eosinophil peroxidase activity. LPC caused maximal adhesion of eosinophils to plated BSA at 4 microM. Lysophosphatidylinositol, which has a similar molecular shape, mimicked the effect of LPC on eosinophil adhesion, while neither lysophosphatidylserine nor lysophosphatidylethanolamine had any effect. Phosphatidylethanolamine, a lipid that has a molecular orientation that is the inverse of LPC, blocked eosinophil adhesion caused by LPC. Unlike platelet-activating factor, a G-protein-coupled receptor agonist, LPC did not cause Ca2+-store depletion, but caused increased Ca2+ influx upon addition of Ca2+ to extracellular medium. This influx was not inhibited by U73122, a phospholipase C inhibitor, demonstrating independence from the G protein-activated phospholipase C pathway. Ca2+ influx was inhibited by either preincubation of phosphotidylethanolamine or La3+, a broad spectrum blocker of cation channels. LPC induced up-regulation of the active conformation of CD11b, which was blocked by preincubation with phosphatidylethanolamine. These data suggest that LPC causes a non-store-operated Ca2+ influx into eosinophils, which subsequently activates CD11b/CD18 to promote eosinophil adhesion.


Subject(s)
Calcium Channels/metabolism , Eosinophils/cytology , Eosinophils/drug effects , Lysophosphatidylcholines/pharmacology , CD11b Antigen/metabolism , Cell Adhesion/drug effects , Egtazic Acid/pharmacology , GTP-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Macrophage-1 Antigen/metabolism , Phosphatidylethanolamines/pharmacology , Protein Conformation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serum Albumin, Bovine/metabolism , Signal Transduction/drug effects , Solubility/drug effects , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
11.
J Immunol ; 177(1): 574-82, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785555

ABSTRACT

We examined the mechanism by which secretory group V phospholipase A(2) (gVPLA(2)) secreted from stimulated epithelial cells activates eosinophil adhesion to ICAM-1 surrogate protein and secretion of leukotriene (LT)C(4). Exogenous human group V PLA(2) (hVPLA(2)) caused an increase in surface CD11b expression and focal clustering of this integrin, which corresponded to increased beta(2) integrin-mediated adhesion. Human IIaPLA(2), a close homolog of hVPLA(2), or W31A, an inactive mutant of hVPLA(2), did not affect these responses. Exogenous lysophosphatidylcholine but not arachidonic acid mimicked the beta(2) integrin-mediated adhesion caused by hVPLA(2) activation. Inhibition of hVPLA(2) with MCL-3G1, a mAb against gVPLA(2), or with LY311727, a global secretory phospholipase A(2) (PLA(2)) inhibitor, attenuated the activity of hVPLA(2); trifluoromethylketone, an inhibitor of cytosolic group IVA PLA(2) (gIVA-PLA(2)), had no inhibitory effect on hVPLA(2)-mediated adhesion. Activation of beta(2) integrin-dependent adhesion by hVPLA(2) did not cause ERK1/2 activation and was independent of gIVA-PLA(2) phosphorylation. In other studies, eosinophils cocultured with epithelial cells were stimulated with FMLP/cytochalasin B (FMLP/B) and/or endothelin-1 (ET-1) before LTC(4) assay. FMLP/B alone caused release of LTC(4) from eosinophils, which was augmented by coculture with epithelial cells activated with ET-1. Addition of MCL-3G1 to cocultured cells caused approximately 50% inhibition of LTC(4) secretion elicited by ET-1, which was blocked further by trifluoromethylketone. Our data indicate that hVPLA(2) causes focal clustering of CD11b and beta(2) integrin adhesion by a novel mechanism that is independent of arachidonic acid synthesis and gIVA-PLA(2) activation. We also demonstrate that gVPLA(2), endogenously secreted from activated epithelial cells, promotes secretion of LTC(4) in cocultured eosinophils.


Subject(s)
CD18 Antigens/physiology , Eosinophils/immunology , Eosinophils/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Leukotriene C4/biosynthesis , Phospholipases A/metabolism , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/physiology , Cell Adhesion/immunology , Cell Line , Cytosol/enzymology , Group V Phospholipases A2 , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoenzymes/physiology , Lysophosphatidylcholines , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phospholipases A/physiology , Phospholipases A2 , Phosphorylation , Protein Transport
12.
Nat Protoc ; 1(6): 2613-20, 2006.
Article in English | MEDLINE | ID: mdl-17406516

ABSTRACT

Eosinophils are a minority constituent in human peripheral blood. The study of eosinophils has been limited by difficulty in achieving sufficient cell number and purity. We describe a modified protocol for immunomagnetic cell separation for efficient isolation of human peripheral blood eosinophils. We employ a mixture of monoclonal antibodies (mAbs) directed against cell-surface antigens on human hematopoietic cells combined with secondary labeling with a colloidal suspension of magnetic dextran-iron particles for negative selection of eosinophils. Unwanted labeled cells are retained in the magnetized column, permitting high recovery (70%) and purity (>98%) of eosinophils while retaining cell viability. Eosinophils remain quiescent after isolation, and stimulation caused by cytokines upregulates (i) cell-cell or cell-extracellular matrix protein adhesion, (ii) secretion of bioactive mediators and (iii) cell-surface adhesion molecules. This method for purified isolation is accomplished in < or = 4 h and preserves eosinophils in a quiescent, viable state.


Subject(s)
Eosinophils , Immunomagnetic Separation/methods , Eosinophils/immunology , Humans
13.
Am J Respir Cell Mol Biol ; 33(1): 65-70, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15802551

ABSTRACT

We examined the role of phosphoinositide 3-kinase (PI3K) in integrin-mediated eosinophil adhesion. Deltap85, a dominant-negative form of the class IA PI3K adaptor subunit, was fused to an HIV-TAT protein transduction domain (TAT-Deltap85). Recombinant TAT-Deltap85 inhibited interleukin (IL)-5-stimulated phosphorylation of protein kinase B, a downstream target of PI3K. beta(2)-Integrin-dependent adhesion caused by IL-5 to the plated intracellular adhesion molecule-1 surrogate, bovine serum albumin, was inhibited by TAT-Deltap85 in a concentration-dependent manner. Similarly, two PI3K inhibitors, wortmannin and LY294002, blocked eosinophil adhesion to plated bovine serum albumin. By contrast, beta(1)-integrin-mediated eosinophil adhesion to vascular cell adhesion moelcule-1 was not blocked by TAT-Deltap85, wortmannin, or LY294002. Rottlerin, a protein kinase C (PKC)-delta inhibitor, also blocked beta(2)-integrin adhesion of eosinophils caused by IL-5, whereas beta(1) adhesion to vascular cell adhesion molecule-1 was not affected. IL-5 caused translocation of PKCdelta from the cytosol to cell membrane; inhibition of PI3K by wortmannin blocked translocation of PKCdelta. Western blot analysis demonstrated that extracellular signal-regulated kinase phosphorylation, a critical intermediary in adhesion elicited by IL-5, was blocked by inhibition of either PI3K or PKC-delta. These data suggest that extracellular signal-regulated kinase-mediated adhesion of beta(2)-integrin caused by IL-5 is mediated in human eosinophils by a class IA PI3K through activation of a PKCdelta pathway.


Subject(s)
CD18 Antigens/metabolism , Cell Adhesion , Eosinophils/metabolism , Interleukin-5/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Androstadienes/metabolism , Androstadienes/pharmacology , Blotting, Western , Cell Membrane/metabolism , Cell Separation , Chromones/pharmacology , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Products, tat/genetics , Genes, Dominant , HIV Long Terminal Repeat , Humans , Hypersensitivity, Immediate/immunology , Immunoblotting , Immunoprecipitation , Morpholines/pharmacology , Phosphorylation , Protein Binding , Protein Isoforms , Protein Kinase C/metabolism , Protein Kinase C-delta , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Serum Albumin, Bovine/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism , Wortmannin
14.
J Allergy Clin Immunol ; 115(3): 493-500, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15753895

ABSTRACT

BACKGROUND: Glucocorticoids attenuate the population of eosinophils and T lymphocytes in asthmatic airways. The decrease in airway eosinophilia is caused both by accelerated cell death and by induction of blockade of integrin adhesion. In this study, we examined the hypothesis that annexin 1 surface expression, which is upregulated by the glucocorticoid receptor, prevents integrin adhesion essential to cell migration by blocking intracellular translocation of cytosolic group IV phospholipase A2 (cPLA2). OBJECTIVE: To examine the relationship of the glucocorticoid on annexin 1 expression and the effect of blockade of annexin 1 activity on adhesion of human eosinophils in vitro. To determine the relationship between annexin 1surface expression and nuclear membrane translocation of cPLA2. METHODS: Eosinophils isolated from human peripheral blood were pretreated with fluticasone propionate (FP), and beta2-integrin adhesion was measured after stimulation with IL-5 or eotaxin. Effects of FP on cPLA2 expression, phosphorylation, and translocation were determined. The role of annexin 1 was examined by using annexin 1 blocking antibody and/or mimetic peptides. RESULTS: Fluticasone propionate decreased stimulated eosinophil adhesion and caused 4-fold increase in annexin 1 expression on the plasma membrane. Inhibition of adhesion by FP was blocked with annexin 1 blocking antibody. Annexin 1 N-terminal mimetic peptide also blocked beta2-integrin adhesion. Translocation of cPLA2 to the nuclear membrane was significantly blocked by incubation with FP. Blockade was reversed with annexin 1 blocking antibody. CONCLUSION: Blockade of beta2-integrin adhesion by glucocorticoid is regulated by annexin 1, which blocks cPLA2 translocation to nuclear membrane.


Subject(s)
Annexins/drug effects , Cell Adhesion/drug effects , Eosinophils/drug effects , Glucocorticoids/pharmacology , Integrin beta Chains/metabolism , Intercellular Adhesion Molecule-1/drug effects , Phospholipases A/metabolism , Androstadienes/pharmacology , Annexins/biosynthesis , Anti-Inflammatory Agents/pharmacology , Biomarkers , Blotting, Western , Cell Adhesion/immunology , Eosinophils/immunology , Eosinophils/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Fluticasone , Group IV Phospholipases A2 , Humans , Integrin beta Chains/drug effects , Integrin beta Chains/immunology , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Microscopy, Fluorescence , Nuclear Envelope/drug effects , Nuclear Envelope/immunology , Nuclear Envelope/metabolism , Phospholipases A2 , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/immunology
15.
Pulm Pharmacol Ther ; 17(2): 73-9, 2004.
Article in English | MEDLINE | ID: mdl-15123228

ABSTRACT

We investigated the effect and mechanism(s) of PDE-4 treatment with concurrent beta2-adrenoceptor stimulation on human eosinophil adhesion mediated by beta2-integrin in vitro. Eosinophils were pretreated with either rolipram, a PDE-4 inhibitor, alone or combined with salmeterol, a beta2-adrenoceptor agonist, before activation with either eotaxin or IL-5. Beta2-integrin mediated adhesion was assessed as adherence to BSA, an established surrogate for ICAM-1. Rolipram caused progressive blockade (77.7 +/- 6.2%) of adhesion elicited by eotaxin. Maximal blockade of IL-5-activated adhesion by rolipram was substantially less (29.9 +/- 5.2%). Salmeterol + rolipram synergistically enhanced the blockade of eotaxin-activated adhesion. Eotaxin also caused approximately 50% increase in surface CD11b expression, which was blocked additively by rolipram + salmeterol. By contrast, CD11b upregulation caused by IL-5 was not blocked by rolipram + salmeterol. Rolipram also attenuated cPLA2 phosphorylation caused by eotaxin but did not block IL-5-induced phosphorylation. We conclude that rolipram blocks expression of CD11b and inhibits cPLA2 phosphorylation in human eosinophils, thus blocking eotaxin-induced adhesion of beta2-integrin. IL-5-induced adhesion likely utilizes a different upstream mechanism in regulation of integrin adhesion.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adrenergic beta-2 Receptor Agonists , Albuterol/analogs & derivatives , CD18 Antigens/physiology , Chemokines, CC/pharmacology , Eosinophils/drug effects , Interleukin-5/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adrenergic beta-Agonists/pharmacology , Albuterol/pharmacology , CD11b Antigen/biosynthesis , CD18 Antigens/biosynthesis , Cell Adhesion/drug effects , Chemokine CCL11 , Chemokines, CC/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Drug Synergism , Enzyme Activation , Eosinophils/metabolism , Eosinophils/physiology , Group IV Phospholipases A2 , Humans , In Vitro Techniques , Interleukin-5/physiology , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Phosphorylation , Rolipram/pharmacology , Salmeterol Xinafoate , Up-Regulation
16.
J Exp Med ; 198(10): 1573-82, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14623911

ABSTRACT

Phosphoinositide 3-kinase (PI3K) is thought to contribute to the pathogenesis of asthma by effecting the recruitment, activation, and apoptosis of inflammatory cells. We examined the role of class IA PI3K in antigen-induced airway inflammation and hyperresponsiveness by i.p. administration into mice of Deltap85 protein, a dominant negative form of the class IA PI3K regulatory subunit, p85alpha, which was fused to HIV-TAT (TAT-Deltap85). Intraperitoneal administration of TAT-Deltap85 caused time-dependent transduction into blood leukocytes, and inhibited activated phosphorylation of protein kinase B (PKB), a downstream target of PI3K, in lung tissues in mice receiving intranasal FMLP. Antigen challenge elicited pulmonary infiltration of lymphocytes, eosinophils and neutrophils, increase in mucus-containing epithelial cells, and airway hyperresponsiveness to methacholine. Except for modest airway neutrophilia, these effects all were blocked by treatment with 3-10 mg/kg of TAT-Deltap85. There was also significant reduction in IL-5 and IL-4 secretion into the BAL. Intranasal administration of IL-5 caused eosinophil migration into the airway lumen, which was attenuated by systemic pretreatment with TAT-Deltap85. We conclude that PI3K has a regulatory role in Th2-cell cytokine secretion, airway inflammation, and airway hyperresponsiveness in mice.


Subject(s)
Hypersensitivity/prevention & control , Inflammation/prevention & control , Phosphatidylinositol 3-Kinases/genetics , Animals , Hypersensitivity/enzymology , Hypersensitivity/genetics , Inflammation/enzymology , Inflammation/genetics , Interleukin-5/metabolism , Lung/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Respiratory Tract Diseases/enzymology , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/prevention & control
17.
J Immunol ; 171(8): 4379-84, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530363

ABSTRACT

We have reported previously that HIV-TAT-dominant negative (dn) Ras inhibits eosinophil adhesion to ICAM-1 after activation by IL-5 and eotaxin. In this study, we evaluated the role of Ras in Ag-induced airway inflammation and hyperresponsiveness by i.p. administration into mice of dnRas, which was fused to an HIV-TAT protein transduction domain (TAT-dnRas). Uptake of TAT-dnRas (t(1/2) = 12 h) was demonstrated in leukocytes after i.p. administration. OVA-sensitization significantly increased eosinophil and lymphocyte numbers in bronchoalveolar lavage fluid 24 h after final challenge. Treatment of animals with 3-10 mg/kg TAT-dnRas blocked the migration of eosinophils from 464 +/- 91 x 10(3)/ml to 288 +/- 79 x 10(3)/ml with 3 mg/kg of TAT-dnRas (p < 0.05), and further decreased to 116 +/- 63 x 10(3)/ml after 10 mg/kg TAT-dnRas (p < 0.01). Histological examination demonstrated that inflammatory cell infiltration (largely eosinophils and mononuclear cells) and mucin production around the airways caused by OVA were blocked by TAT-dnRas. OVA challenge also caused airway hyperresponsiveness to methacholine, which was dose dependently blocked by treatment with TAT-dnRas. TAT-dnRas also blocked Ag-induced IL-4 and IL-5, but not IFN-gamma, production in lung tissue. Intranasal administration of IL-5 caused eosinophil migration into the airway lumen, which was attenuated by pretreatment with TAT-dnRas. By contrast, TAT-green fluorescent protein or dnRas lacking the TAT protein transduction domain did not block airway inflammation, cytokine production, or airway hyperresponsiveness. We conclude that Ras mediates Th2 cytokine production, airway inflammation, and airway hyperresponsiveness in immune-sensitized mice.


Subject(s)
Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/prevention & control , Gene Products, tat/administration & dosage , HIV/immunology , Inflammation Mediators/administration & dosage , Lung/pathology , Recombinant Fusion Proteins/administration & dosage , ras Proteins/administration & dosage , Administration, Intranasal , Animals , Antigens/administration & dosage , Antigens/immunology , Bronchial Hyperreactivity/pathology , Cell Migration Inhibition , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Eosinophils/immunology , Eosinophils/pathology , Gene Products, tat/genetics , HIV/genetics , Humans , Injections, Intraperitoneal , Interleukin-5/administration & dosage , Kinetics , Lung/immunology , Lung/metabolism , Lymphocytes/immunology , Lymphocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mucus/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Recombinant Fusion Proteins/genetics , Th2 Cells/immunology , Th2 Cells/metabolism , Transduction, Genetic , ras Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus
18.
J Immunol ; 171(8): 4399-405, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530366

ABSTRACT

Activation of group IV cytosolic phospholipase A(2) (gIV-PLA(2)) is the essential first step in the synthesis of inflammatory eicosanoids and in integrin-mediated adhesion of leukocytes. Prior investigations have demonstrated that phosphorylation of gIV-PLA(2) results from activation of at least two isoforms of mitogen-activated protein kinase (MAPK). We investigated the potential role of phosphoinositide 3-kinase (PI3K) in the activation of gIV-PLA(2) and the hydrolysis of membrane phosphatidylcholine in fMLP-stimulated human blood eosinophils. Transduction into eosinophils of Deltap85, a dominant negative form of class IA PI3K adaptor subunit, fused to an HIV-TAT protein transduction domain (TAT-Deltap85) concentration dependently inhibited fMLP-stimulated phosphorylation of protein kinase B, a downstream target of PI3K. FMLP caused increased arachidonic acid (AA) release and secretion of leukotriene C(4) (LTC(4)). TAT-Deltap85 and LY294002, a PI3K inhibitor, blocked the phosphorylation of gIV-PLA(2) at Ser(505) caused by fMLP, thus inhibiting gIV-PLA(2) hydrolysis and production of AA and LTC(4) in eosinophils. FMLP also caused extracellular signal-related kinases 1 and 2 and p38 MAPK phosphorylation in eosinophils; however, neither phosphorylation of extracellular signal-related kinases 1 and 2 nor p38 was inhibited by TAT-Deltap85 or LY294002. Inhibition of 1) p70 S6 kinase by rapamycin, 2) protein kinase B by Akt inhibitor, or 3) protein kinase C by Ro-31-8220, the potential downstream targets of PI3K for activation of gIV-PLA(2), had no effect on AA release or LTC(4) secretion caused by fMLP. We find that PI3K is required for gIV-PLA(2) activation and hydrolytic production of AA in activated eosinophils. Our data suggest that this essential PI3K independently activates gIV-PLA(2) through a pathway that does not involve MAPK.


Subject(s)
Eosinophils/enzymology , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/physiology , Phospholipases A/metabolism , Arachidonic Acid/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Enzyme Activation/genetics , Eosinophils/metabolism , Eosinophils/physiology , Gene Products, tat/genetics , Gene Products, tat/metabolism , Group IV Phospholipases A2 , Humans , Isoenzymes/biosynthesis , Leukotriene C4/biosynthesis , MAP Kinase Signaling System/genetics , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phospholipases A2 , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Transduction, Genetic , Tritium
19.
J Allergy Clin Immunol ; 112(2): 404-10, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897749

ABSTRACT

BACKGROUND: Prior investigations have demonstrated that beta(2)-adrenoceptor stimulation is ineffective in inhibiting synthesis of eicosanoids in human eosinophils. This effect has been postulated to relate to density or structural differences in the beta(2)-adrenoceptor or its coupled G-protein. However, recent reports indicate that cAMP-specific PDE4 activity in eosinophils is 10-fold that of other inflammatory cells. We postulated that selective blockade of PDE4 in eosinophils would unmask the inhibitory effect of beta(2)-adrenoceptor stimulation and that this inhibition would result from decreased phosphor-ylation of cytosolic group IV-PLA(2) (gIV-PLA(2)). OBJECTIVE: To determine (a) whether PDE4 inhibition alone with rolipram blocked secretions of arachidonic acid (AA) and leukotriene C(4) (LTC(4)) caused by activation of eosinophils with formyl-met-leu-phe plus cytochalasin B (FMLP/B), (b) to determine if PDE4 inhibition plus beta(2)-adrenoceptor agonist act additively to augment endogenous cAMP concentration, and (c) to determine the mechanism by which additive inhibition of AA and LTC(4) synthesis is regulated by cAMP. METHODS: Human eosinophils were pretreated with buffer, salmeterol or rolipram (singly or combination) before FMLP/B activation. Release of AA and LTC(4), intracellular cAMP concentration, and phosphorylation and activation of gIV-PLA(2) were determined. RESULTS: Rolipram unmasked the inhibitory effect of beta(2)-adrenoceptor stimulation with salmeterol and significantly attenuated the stimulated release of AA and subsequent LTC(4). Inhibition corresponded to increased cAMP production caused by rolipram alone or rolipram plus salmeterol and blocked proportionately the phosphorylation and activation of gIV-PLA(2) in FMLP/B-activated eosinophils. CONCLUSIONS: Inhibition of PDE4 by rolipram unmasks beta(2)-adrenergic blockade of LTC(4) synthesis caused by FMLP/B.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adrenergic beta-Agonists/pharmacology , Albuterol/analogs & derivatives , Albuterol/pharmacology , Eosinophils/metabolism , Leukotriene C4/antagonists & inhibitors , Phospholipases A/metabolism , Adult , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/pharmacology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cytosol/enzymology , Drug Synergism , Female , Group IV Phospholipases A2 , Humans , Intracellular Membranes/metabolism , Leukotriene C4/biosynthesis , Male , Osmolar Concentration , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A2 , Phosphorylation/drug effects , Rolipram/pharmacology , Salmeterol Xinafoate
20.
J Biol Chem ; 278(40): 38813-20, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12796497

ABSTRACT

We previously reported that exogenously added human group V phospholipase A2 (hVPLA2) could elicit leukotriene B4 biosynthesis in human neutrophils through the activation of group IVA phospholipase A2 (cPLA2) (Kim, Y. J., Kim, K. P., Han, S. K., Munoz, N. M., Zhu, X., Sano, H., Leff, A. R., and Cho, W. (2002) J. Biol. Chem. 277, 36479-36488). In this study, we determined the functional significance and mechanism of the exogenous hVPLA2-induced arachidonic acid (AA) release and leukotriene C4 (LTC4) synthesis in isolated human peripheral blood eosinophils. As low a concentration as 10 nm exogenous hVPLA2 was able to elicit the significant release of AA and LTC4 from unstimulated eosinophils, which depended on its ability to act on phosphatidylcholine membranes. hVPLA2 also augmented the release of AA and LTC4 from eosinophils activated with formyl-Met-Leu-Phe + cytochalasin B. A cellular fluorescent PLA2 assay showed that hVPLA2 had a lipolytic action first on the outer plasma membrane and then on the perinuclear region. hVPLA2 also caused the translocation of 5-lipoxygenase from the cytosol to the nuclear membrane and a 2-fold increase in 5-lipoxygenase activity. However, hVPLA2 induced neither the increase in intracellular calcium concentration nor cPLA2 phosphorylation; consequently, cPLA2 activity was not affected by hVPLA2. Pharmacological inhibition of cPLA2 and the hVPLA2-induced activation of eosinophils derived from the cPLA2-deficient mouse corroborated that hVPLA2 mediates the release of AA and leukotriene in a cPLA2-independent manner. As such, this study represents a unique example in which a secretory phospholipase induces the eicosanoid formation in inflammatory cells, completely independent of cPLA2 activation.


Subject(s)
Cysteine/chemistry , Eosinophils/metabolism , Leukotrienes/biosynthesis , Phospholipases A/metabolism , Phospholipases A/physiology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acid/metabolism , Blotting, Western , Bone Marrow Cells , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cytochalasin B/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Group IV Phospholipases A2 , Group V Phospholipases A2 , Humans , Hydrolysis , Immunohistochemistry , Leukotriene C4/metabolism , Mice , Microscopy, Fluorescence , Neutrophils/metabolism , Phosphatidylcholines/metabolism , Phospholipases A2 , Phosphorylation , Protein Isoforms , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...