Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 308(11): H1414-22, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25795713

ABSTRACT

Sudden cardiac arrest (SCA) is a leading cause of death in the United States. Despite return of spontaneous circulation, patients die due to post-SCA syndrome that includes myocardial dysfunction, brain injury, impaired metabolism, and inflammation. No medications improve SCA survival. Our prior work suggests that optimal Akt activation is critical for cooling protection and SCA recovery. Here, we investigate a small inhibitor of PTEN, an Akt-related phosphatase present in heart and brain, as a potential therapy in improving cardiac and neurological recovery after SCA. Anesthetized adult female wild-type C57BL/6 mice were randomized to pretreatment of VO-OHpic (VO) 30 min before SCA or vehicle control. Mice underwent 8 min of KCl-induced asystolic arrest followed by CPR. Resuscitated animals were hemodynamically monitored for 2 h and observed for 72 h. Outcomes included heart pressure-volume loops, energetics (phosphocreatine and ATP from (31)P NMR), protein phosphorylation of Akt, GSK3ß, pyruvate dehydrogenase (PDH) and phospholamban, circulating inflammatory cytokines, plasma lactate, and glucose as measures of systemic metabolic recovery. VO reduced deterioration of left ventricular maximum pressure, maximum rate of change in the left ventricular pressure, and Petco2 and improved 72 h neurological intact survival (50% vs. 10%; P < 0.05). It reduced plasma lactate, glucose, IL-1ß, and Pre-B cell colony enhancing factor, while increasing IL-10. VO increased phosphorylation of Akt and GSK3ß in both heart and brain, and cardiac phospholamban phosphorylation while reducing p-PDH. Moreover, VO improved cardiac bioenergetic recovery. We concluded that pharmacologic PTEN inhibition enhances Akt activation, improving metabolic, cardiovascular, and neurologic recovery with increased survival after SCA. PTEN inhibitors may be a novel pharmacologic strategy for treating SCA.


Subject(s)
Energy Metabolism , Enzyme Inhibitors/therapeutic use , Heart Arrest/drug therapy , Organometallic Compounds/therapeutic use , PTEN Phosphohydrolase/antagonists & inhibitors , Animals , Cytokines/blood , Female , Heart Arrest/metabolism , Hemodynamics , Mice , Mice, Inbred C57BL , Organometallic Compounds/pharmacology , Resuscitation/methods
2.
PLoS One ; 9(4): e95622, 2014.
Article in English | MEDLINE | ID: mdl-24752319

ABSTRACT

Recent work shows that cooling protection after mouse cardiac arrest and cardiomyocyte ischemia is mediated by Akt activation. The PI3K p85 subunit can either augment or inhibit Akt activation depending on its binding to p110 or PTEN respectively. To further clarify the role of PI3K p85 in cardioprotection, we studied novel TAT-p85 fusion proteins that selectively inhibit PI3K p85 binding. We hypothesized that TAT fused p85 lacking the PTEN binding site (TAT-ΔPTEN p85) would enhance Akt phosphorylation to afford cardioprotection. Conversely, TAT fused p85 lacking the p110 binding site (TAT-Δp110p85) would decrease Akt phosphorylation and abrogate cardioprotection. Microscopy and Western blot analysis demonstrated that TAT fusion protein was transduced into cardiomyocytes within 5 min and remained more than 2 h. Inhibition of PI3K/Akt by TAT-Δp110 p85 significantly increased cell death from 44.6±2.7% to 92.5±3.4% after simulated ischemia and reperfusion. By contrast, PTEN inhibition using TAT-ΔPTEN p85 decreased cell death to 11.9±5.3%, a similar level of cardioprotection seen with past cooling studies. Additional studies with the small molecule PTEN inhibitor VO-OHpic confirmed that PTEN inhibition was highly protective against cell death induced by ischemia and reperfusion. We conclude that blockade of p85-PTEN interaction and PTEN inhibition may be promising strategies for rescuing the heart from ischemia and reperfusion injury.


Subject(s)
Gene Products, tat/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Hydrogen Peroxide/pharmacology , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , PTEN Phosphohydrolase/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects
3.
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
4.
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.

5.
Respir Res ; 13: 4, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22257498

ABSTRACT

BACKGROUND: Proline-rich tyrosine kinase 2 (Pyk2) is essential in neutrophil degranulation and chemotaxis in vitro. However, its effect on the process of lung inflammation and edema formation during LPS induced acute lung injury (ALI) remains unknown. The goal of the present study was to determine the effect of inhibiting Pyk2 on LPS-induced acute lung inflammation and injury in vivo. METHODS: C57BL6 mice were given either 10 mg/kg LPS or saline intratracheally. Inhibition of Pyk2 was effected by intraperitoneal administration TAT-Pyk2-CT 1 h before challenge. Bronchoalveolar lavage analysis of cell counts, lung histology and protein concentration in BAL were analyzed at 18 h after LPS treatment. KC and MIP-2 concentrations in BAL were measured by a mouse cytokine multiplex kit. The static lung compliance was determined by pressure-volume curve using a computer-controlled small animal ventilator. The extravasated Evans blue concentration in lung homogenate was determined spectrophotometrically. RESULTS: Intratracheal instillation of LPS induced significant neutrophil infiltration into the lung interstitium and alveolar space, which was attenuated by pre-treatment with TAT-Pyk2-CT. TAT-Pyk2-CT pretreatment also attenuated 1) myeloperoxidase content in lung tissues, 2) vascular leakage as measured by Evans blue dye extravasation in the lungs and the increase in protein concentration in bronchoalveolar lavage, and 3) the decrease in lung compliance. In each paradigm, treatment with control protein TAT-GFP had no blocking effect. By contrast, production of neutrophil chemokines MIP-2 and keratinocyte-derived chemokine in the bronchoalveolar lavage was not reduced by TAT-Pyk2-CT. Western blot analysis confirmed that tyrosine phosphorylation of Pyk2 in LPS-challenged lungs was reduced to control levels by TAT-Pyk2-CT pretreatment. CONCLUSIONS: These results suggest that Pyk2 plays an important role in the development of acute lung injury in mice and that pharmacological inhibition of Pyk2 might provide a potential therapeutic strategy in the pretreatment for patients at imminent risk of developing acute lung injury.


Subject(s)
Acute Lung Injury/drug therapy , Focal Adhesion Kinase 2/antagonists & inhibitors , Lung/drug effects , Pneumonia/drug therapy , Recombinant Fusion Proteins/administration & dosage , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CXCL2/analysis , Chemokines/analysis , Disease Models, Animal , Female , Lipopolysaccharides/administration & dosage , Lung/enzymology , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Peroxidase/analysis , Pneumonia/chemically induced
6.
J Trauma ; 71(5): 1262-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22071928

ABSTRACT

BACKGROUND: Therapeutic hypothermia (TH) has demonstrated great potential for forestalling cardiovascular collapse and improving outcomes in the setting of severe hemorrhagic shock (HS). We used an established mouse model of severe HS to study the response of interrelated cardiac-signaling proteins p38, HspB1, and Akt to shock, resuscitation, and cardioprotective TH. METHODS: Adult female C57BL6/J mice were bled and maintained at a mean arterial pressure of 35 mm Hg. After 30 minutes, mice were randomized to 120 minutes of TH (33°C ± 0.5°C) or continued normothermia at 37°C. After 90 minutes, animals were resuscitated and monitored for 180 minutes. Cardiac p38, Akt, and HspB1 phosphorylation (p-p38, p-Akt, and p-HspB1), expression, and Akt/HspB1 interactions were measured at serial time points during HS and resuscitation. Markers of mitochondrial damage (plasma cytochrome c), inflammation (myeloperoxidase), and apoptosis (terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling) were analyzed. RESULTS: By 15 minutes HS, p-p38 and p-HspB1 significantly increased while p-Akt(T308) decreased (p < 0.05). TH attenuated phosphorylation of the p38α isoform during HS and increased phosphorylation of the p38γ isoform during both HS and early resuscitation (p < 0.05). TH increased Akt/HspB1 coimmunoprecipitation during early resuscitation and increased p-Akt and HspB1 expression during late resuscitation (p < 0.05). Finally, TH attenuated the myocardial myeloperoxidase and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining and plasma cytochrome c during late resuscitation. CONCLUSIONS: TH increases phosphorylation of p38γ during both HS and early resuscitation, but attenuates phosphorylation of p38α, increases Akt/HspB1 interaction, and modulates Akt phosphorylation during HS and resuscitation. Such TH-related signaling events are associated with reduced cardiac inflammation, apoptosis, and mitochondrial injury.


Subject(s)
Hypothermia, Induced , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/therapy , Analysis of Variance , Animals , Apoptosis , Cytochromes c/blood , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Female , Heat-Shock Proteins/metabolism , Immunoblotting , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Molecular Chaperones , Neoplasm Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Resuscitation/methods , Statistics, Nonparametric , p38 Mitogen-Activated Protein Kinases/metabolism
8.
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
9.
J Inflamm (Lond) ; 7: 26, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20507587

ABSTRACT

BACKGROUND: Pyk2 is a non-receptor cytoplasmic tyrosine kinase that belongs to the focal adhesion kinase family and has been implicated in neutrophil spreading and respiratory burst activity caused by TNF-alpha. However, the role of Pyk2 in neutrophil migration is incompletely defined. In this study, we tested the hypothesis that Pyk2 regulates the migration of neutrophil-like differentiated HL-60 cells subsequent to beta2-integrin mediated cell adhesion. METHODS: HL-60 cells were induced to differentiate into neutrophil-like cells (dHL60) by incubation in medium containing 1.25% DMSO for up to 4 days. Pyk2 expression and tyrosine phosphorylation was measured by Western blot analysis. Adhesion of dHL60 cells to plated fibrinogen was measured by residual myeloperoxidase activity. dHL60 cell migration was evaluated using a 96-well chemoTx chamber. RESULTS: Western blot analysis demonstrated that hematopoietic Pyk2 was predominantly expressed after HL60 cell differentiation. Pyk2 was tyrosine phosphorylated upon adhesion of dHL60 cells to plated fibrinogen in the presence of fMLP. By contrast, tyrosine phosphorylation of Pyk2 was insignificant in dHL60 cells treated in suspension with fMLP. Antibodies against CD18 blocked both phosphorylation of Pyk2 and adhesion of dHL60 cells to fibrinogen, demonstrating that phosphorylation of Pyk2 was beta2-integrin dependent. TAT-Pyk2-CT, a dominant negative fusion protein in which the TAT protein transduction domain was fused to the c-terminal Pyk2, attenuated fMLP-stimulated spreading, migration and phosphorylation of endogenous Pyk2 without blocking adhesion of dHL-60 cells to fibrinogen. Similarly, silencing of Pyk2 expression by siRNA in dHL60 cells also attenuated dHL60 cell migration caused by fMLP. Phospho-Pyk2 was evenly distributed around cell membrane circumferentially in unstimulated dHL-60 cells adherent to plated fibrinogen. In dHL60 cells treated with fMLP to cause cell spreading and polarization, Pyk2 was concentrated at the leading edge of pseudopods or at the trailing edge of uropods during migration of neutrophilic dHL-60 cells. CONCLUSIONS: We conclude that Pyk2 is activated by beta2-integrin adhesion. The activated concentration of Pyk2 and colocalization with F-actin in pseudopodia suggests that Pyk2 may regulate cell spreading and migration in dHL60 cells.

10.
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.

11.
Am J Respir Cell Mol Biol ; 42(4): 491-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19520918

ABSTRACT

The objective of this investigation was to determine the role of Pyk2, an intracellular nonreceptor protein tyrosine kinase for postadhesive inflammatory cell migration, on airway inflammation and hyperresponsiveness in immune-sensitized mice. Blockade of Pyk2 was effected by intraperitoneal administration of dominant-negative C-terminal Pyk2 fused to a TAT protein transduction domain (TAT-Pyk2-CT). Ovalbumin challenge elicited infiltration of both eosinophils and lymphocytes into airways, increased mucus-containing epithelial cells, and caused increased airway hyperresponsiveness to methacholine in immune-sensitized mice. Pretreatment with 10 mg/kg TAT-Pyk2-CT intraperitoneally blocked all of these effects and further decreased secretion of Th2 cytokine IL-4, IL-5, and IL-13 into the bronchoalveolar lavage fluid. Intranasal administration of IL-5 caused eosinophil migration into the airway lumen, which was attenuated by systemic pretreatment with TAT-Pyk2-CT. In each paradigm, treatment with control protein TAT-GFP had no blocking effect. We conclude that Pyk2, which is essential for inflammatory cell migration in vitro, regulates airway inflammation, Th2 cytokine secretion, and airway hyperresponsiveness in the ovalbumin-sensitized mice during antigen challenge in vivo.


Subject(s)
Asthma/metabolism , Cell Movement , Eosinophils/metabolism , Focal Adhesion Kinase 2/metabolism , Th2 Cells/metabolism , Animals , Antigens/adverse effects , Antigens/pharmacology , Asthma/chemically induced , Asthma/pathology , Cytokines/metabolism , Eosinophils/pathology , Female , Focal Adhesion Kinase 2/pharmacology , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Mice , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Th2 Cells/pathology
13.
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
17.
Am J Respir Cell Mol Biol ; 39(3): 263-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18367725

ABSTRACT

We examined the role of proline-rich tyrosine kinase (Pyk) 2 in the spreading and migration of human blood eosinophils after beta(2)-integrin ligation. Western blot analysis showed that Pyk2 was activated by phosphorylation at Y402 after eosinophil adhesion to BSA-coated plates after activation with IL-5, platelet-activating factor (PAF), formyl-met-leu-phe (fMLP), or Mn(2)(+). To determine the role of Pyk2 in regulating eosinophil migration, we used a transducable dominant-negative inhibitor of Pyk2, TAT-mediated protein transduction of dominant-negative C-terminal Pyk2 (TAT-Pyk2-CT), a fusion protein in which TAT peptide was fused to the C-terminal Pyk2. TAT-Pyk2-CT blocked tyrosine phosphorylation of Pyk2 caused by beta(2)-integrin adhesion, but did not block adhesion of eosinophils to plated BSA. TAT-Pyk2-CT also blocked subsequent spreading and migration of eosinophils caused by IL-5, PAF, or fMLP. Spreading eosinophils stained with FITC-conjugated phalloidin showed elongation and formation of multiple fillopodia and lamellipodia, whereas nonspreading eosinophils were smaller and round. Treatment of eosinophils with TAT-Pyk2-CT had no effect on the initial cell polarization, but blocked the formation of fillopodia and lamellipodia in adherent cells. Migration of eosinophils through Transwell plates caused by IL-5, PAF, or fMLP was blocked significantly after inhibition of Pyk2. These data indicate that Pyk2, although not involved in beta(2)-integrin adhesion, causes eosinophil spreading and regulates subsequent chemotactic migration after beta(2)-integrin ligation to endothelial counter ligands. We conclude that Pyk2 is activated by beta(2)-integrin adhesion and is a required signal for eosinophil spreading and subsequent chemotactic migration.


Subject(s)
CD18 Antigens/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Focal Adhesion Kinase 2/metabolism , Blotting, Western , Eosinophils/cytology , Humans , Phosphorylation , Tyrosine/metabolism
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...