Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Transfusion ; 57(11): 2690-2700, 2017 11.
Article in English | MEDLINE | ID: mdl-28880373

ABSTRACT

BACKGROUND: Although transfusion is a lifesaving intervention, it may be associated with significant morbidity in injured patients. We hypothesize that stored red blood cells (RBCs) induce proinflammatory activation of human pulmonary microvascular endothelial cells (HMVECs) resulting in neutrophil (PMN) adhesion and predisposition to acute lung injury (ALI). STUDY DESIGN AND METHODS: Ten units of RBCs were collected; 50% (by weight) were leukoreduced (LR-RBCs) and the remainder was unmodified and stored in additive solution-5 (AS-5). An additional 10 units of RBCs were collected, leukoreduced, and stored in AS-3. HMVECs were incubated with [10%-40%]FINAL of the supernatants on Day (D)1 to D42 of storage, lipid extracts, and purified lipids. Endothelial surface expression of intercellular adhesion molecule-1 (ICAM-1), interleukin (IL)-8 release, and PMN adhesion to HMVECs were measured. HMVEC signaling via the BLT2 receptor was evaluated. Supernatants and lipids were also employed as the first event in a two-event model of ALI. RESULTS: The supernatants [10%-40%]FINAL from D21 LR-RBCs and D42 RBCs and LR-RBCs and the lipids from D42 stored in AS-5 induced increased ICAM-1 surface expression on endothelium, IL-8 release, and PMN adhesion. In addition, the supernatants [20%-40%]FINAL from D21 and D42 RBCs in AS-5 also increased endothelial surface expression of ICAM-1. D42 supernatants and lipids also caused coprecipitation of ß-arrestin-1 with BLT2, protein kinase C (PKC)ßI , and PKCδ and served as the first event in a two-event rodent model of ALI. CONCLUSION: Lipids that accumulate during RBC storage activate endothelium and predispose to ALI, which may explain some of the adverse events associated with the transfusion of critically injured patients.


Subject(s)
Blood Preservation/methods , Erythrocytes/cytology , Lipids/pharmacology , Lung/blood supply , Protein Kinase C/metabolism , Receptors, Leukotriene B4/metabolism , Acute Lung Injury/etiology , Culture Media, Conditioned/pharmacology , Endothelial Cells/metabolism , Enzyme Activation , Erythrocyte Transfusion/adverse effects , Humans , Microvessels/cytology , Pneumonia/etiology
2.
J Leukoc Biol ; 101(1): 261-273, 2017 01.
Article in English | MEDLINE | ID: mdl-27531930

ABSTRACT

Lysophosphatidylcholines (lysoPCs) are effective polymorphonuclear neutrophil (PMN) priming agents implicated in transfusion-related acute lung injury (TRALI). LysoPCs cause ligation of the G2A receptor, cytosolic Ca2+ flux, and activation of Hck. We hypothesize that lysoPCs induce Hck-dependent activation of protein kinase C (PKC), resulting in phosphorylation and membrane translocation of 47 kDa phagocyte oxidase protein (p47phox). PMNs, human or murine, were primed with lysoPCs and were smeared onto slides and examined by digital microscopy or separated into subcellular fractions or whole-cell lysates. Proteins were immunoprecipitated or separated by polyacrylamide gel electrophoresis and immunoblotted for proteins of interest. Wild-type (WT) and PKCγ knockout (KO) mice were used in a 2-event model of TRALI. LysoPCs induced Hck coprecipitation with PKCδ and PKCγ and the PKCδ:PKCγ complex also had a fluorescence resonance energy transfer (FRET)+ interaction with lipid rafts and Wiskott-Aldrich syndrome protein family verprolin-homologous protein 2 (WAVE2). PKCγ then coprecipitated with p47phox Immunoblotting, immunoprecipitation (IP), specific inhibitors, intracellular depletion of PKC isoforms, and PMNs from PKCγ KO mice demonstrated that Hck elicited activation/Tyr phosphorylation (Tyr311 and Tyr525) of PKCδ, which became Thr phosphorylated (Thr507). Activated PKCδ then caused activation of PKCγ, both by Tyr phosphorylation (Τyr514) and Ser phosphorylation, which induced phosphorylation and membrane translocation of p47phox In PKCγ KO PMNs, lysoPCs induced Hck translocation but did not evidence a FRET+ interaction between PKCδ and PKCγ nor prime PMNs. In WT mice, lysoPCs served as the second event in a 2-event in vivo model of TRALI but did not induce TRALI in PKCγ KO mice. We conclude that lysoPCs prime PMNs through Hck-dependent activation of PKCδ, which stimulates PKCγ, resulting in translocation of phosphorylated p47phox.


Subject(s)
Cell Membrane/metabolism , Lysophosphatidylcholines/pharmacology , NADPH Oxidases/metabolism , Neutrophils/metabolism , Protein Kinase C-delta/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Animals , Calcium/metabolism , Cell Membrane/drug effects , Enzyme Activation/drug effects , Humans , Lung Injury/pathology , Mice , Mice, Knockout , Neutrophils/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Recombinant Proteins/pharmacology
3.
Shock ; 44(2): 137-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25944790

ABSTRACT

UNLABELLED: Proinflammatory activation of vascular endothelium leading to increased surface expression of adhesion molecules and neutrophil (PMN) sequestration and subsequent activation is paramount in the development of acute lung injury and organ injury in injured patients. We hypothesize that α-enolase, which accumulates in injured patients, primes PMNs and causes proinflammatory activation of endothelial cells leading to PMN-mediated cytotoxicity. METHODS: Proteomic analyses of field plasma samples from injured versus healthy patients were used for protein identification. Human pulmonary microvascular endothelial cells (HMVECs) were incubated with α-enolase or thrombin, and intercellular adhesion molecule-1 surface expression was measured by flow cytometry. A two-event in vitro model of PMN cytotoxicity HMVECs activated with α-enolase, thrombin, or buffer was used as targets for lysophosphatidylcholine-primed or buffer-treated PMNs. The PMN priming activity of α-enolase was completed, and lysates from both PMNs and HMVECs were immunoblotted for protease-activated receptor 1 (PAR-1) and PAR-2 and coprecipitation of α-enolase with PAR-2 and plasminogen/plasmin. RESULTS: α-Enolase increased 10.8-fold in injured patients (P < 0.05). Thrombin and α-enolase significantly increased intercellular adhesion molecule-1 surface expression on HMVECs, which was inhibited by antiproteases, induced PMN adherence, and served as the first event in the two-event model of PMN cytotoxicity. α-Enolase coprecipitated with PAR-2 and plasminogen/plasmin on HMVECs and PMNs and induced PMN priming, which was inhibited by tranexamic acid, and enzymatic activity was not required. CONCLUSIONS: α-Enolase increases after injury and may activate pulmonary endothelial cells and prime PMNs through plasmin activity and PAR-2 activation. Such proinflammatory endothelial activation may predispose to PMN-mediated organ injury.


Subject(s)
Endothelial Cells/metabolism , Lung/blood supply , Microcirculation , Neutrophils/metabolism , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Receptor, PAR-2/metabolism , Acute Lung Injury/metabolism , Cell Membrane/metabolism , Endothelial Cells/cytology , Flow Cytometry , Humans , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Proteomics , Thrombin/metabolism , Tranexamic Acid/metabolism , Wounds, Nonpenetrating/blood
4.
Blood ; 123(22): 3488-95, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24747436

ABSTRACT

Transfusion-related acute lung injury (TRALI) remains a significant cause of transfusion-related mortality with red cell transfusion. We hypothesize that prestorage filtration may reduce proinflammatory activity in the red blood cell (RBC) supernatant and prevent TRALI. Filters were manufactured for both small volumes and RBC units. Plasma containing antibodies to human lymphocyte antigen (HLA)-A2 or human neutrophil antigen (HNA)-3a was filtered, and immunoglobulins and specific HNA-3a and HLA-2a neutrophil (PMN) priming activity were measured. Antibodies to OX27 were added to plasma, and filtration was evaluated in a 2-event animal model of TRALI. RBC units from 31 donors known to have antibodies against HLA antigens and from 16 antibody-negative controls were filtered. Furthermore, 4 RBC units were drawn and underwent standard leukoreduction. Immunoglobulins, HLA antibodies, PMN priming activity, and the ability to induce TRALI in an animal model were measured. Small-volume filtration of plasma removed >96% of IgG, antibodies to HLA-A2 and HNA-3a, and their respective priming activity, as well as mitigating antibody-mediated in vivo TRALI. In RBC units, experimental filtration removed antibodies to HLA antigens and inhibited the accumulation of lipid priming activity and lipid-mediated TRALI. We conclude that filtration removes proinflammatory activity and the ability to induce TRALI from RBCs and may represent a TRALI mitigation step.


Subject(s)
Acute Lung Injury/etiology , Antibodies/immunology , Blood Component Removal/methods , Blood Component Transfusion/adverse effects , Erythrocytes/immunology , Filtration/methods , Lipids/immunology , 2,3-Diphosphoglycerate/blood , Adenosine Triphosphate/blood , Animals , Blood Donors , Female , HLA Antigens/immunology , Humans , Hydrogen-Ion Concentration , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Plasma/immunology , Rats
5.
Transfusion ; 53(1): 147-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22563732

ABSTRACT

BACKGROUND: Lipids and other biologically active substances accumulate in platelet concentrates (PCs) during storage. Some of these substances have been suggested to modulate immune responses and to play a pathogenic role in the development of transfusion-related acute lung injury. This study compared the content and impact of some biological response modifiers in PCs treated with pathogen reduction (PR) technology and nontreated PCs. STUDY DESIGN AND METHODS: Apheresis PCs (n = 12) were split in two: one split was subjected to PR treatment (INTERCEPT, Cerus Corp.) and the other split was left untreated. Basic characterization and content of vascular endothelial growth factor (VEGF) and sCD154 were measured. Lipopolysaccharide (LPS)-induced secretion of interleukin-10 (IL-10) and tumor necrosis factor-α (TNF-α) was measured after incubation of heparinized whole blood with platelet (PLT) supernatants. The supernatants' neutrophil (PMN)-priming capacity, and thereby activation of the NADPH oxidase, was measured as the rate of superoxide anion production after formyl-Met-Leu-Phe activation. Lipids were extracted from the supernatants on Day 6 and tested for PMN-priming activity. RESULTS: Supernatants from PR-treated PCs demonstrated significantly higher mean PLT volume (MPV) and O(2) , lower pH, CO(2) , and HCO(3-) , and significantly less LPS-induced TNF-α secretion compared to untreated PCs. No differences in swirling, PLT count, potassium levels, glucose consumption, lactate production, IL-10, VEGF, sCD154, or PMN-priming activity were found between the groups over time. CONCLUSION: INTERCEPT PR treatment caused no substantial differences in PCs, except for minor changes in MPV and metabolic variables. Further studies are needed to explain the differences in the LPS-induced TNF-α secretion.


Subject(s)
Blood Component Removal , Blood Platelets/metabolism , Blood Platelets/drug effects , Humans , Hydrogen-Ion Concentration , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Photochemistry , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Pediatr Blood Cancer ; 58(3): 399-405, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21793188

ABSTRACT

BACKGROUND: Acute chest syndrome (ACS) in sickle cell disease is associated with elevation of secretory phospholipase A(2) (sPLA(2) ). We hypothesize that sPLA(2) cleaves membrane lipids from sickled red blood cells (RBCs) causing PMN-mediated endothelial cell injury (ECI) as the second event in a two-event model. METHODS: Whole blood was collected from children when in steady state or daily during admissions for vaso-occlusive pain (VOC) or ACS. The plasma and RBCs were separated, sPLA(2) levels were measured, and the RBCs were incubated with sPLA(2) . Plasma and lipids, extracted from the plasma or the supernatant of sPLA(2) -treated RBCs, were assayed for PMN priming activity and used as the second event in a model of PMN-mediated ECI. Phosphatidylserine (PS) surface expression on RBCs was quantified by flow cytometry. RESULTS: Increased sPLA(2) -IIa levels were associated with ACS. SPLA(2) -liberated lipids from VOC and the plasma, plasma lipids and sPLA(2) -liberated lipids from ACS primed PMNs and caused PMN-mediated ECI (P < 0.01). RBCs from VOC had increased in PS surface expression versus steady state. CONCLUSIONS: ACS plasma and lipids and sPLA(2) -released lipids from RBCs during VOC or ACS induce PMN-mediated ECI. VOC elicited increases in PS surface expression providing a membrane substrate for sPLA(2) lysis of sickle RBCs.


Subject(s)
Acute Chest Syndrome/physiopathology , Neutrophils/metabolism , Phospholipases A2, Secretory/blood , Adolescent , Child , Child, Preschool , Colorado , Endothelium, Vascular , Female , Humans , Infant , Lung/blood supply , Male
7.
Transfusion ; 51(12): 2549-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21615744

ABSTRACT

BACKGROUND: Lipids accumulate during the storage of red blood cells (RBCs), prime neutrophils (PMNs), and have been implicated in transfusion-related acute lung injury (TRALI). These lipids are composed of two classes: nonpolar lipids and lysophosphatidylcholines based on their retention time on separation by high-pressure liquid chromatography. Prestorage leukoreduction significantly decreases white blood cell and platelet contamination of RBCs; therefore, it is hypothesized that prestorage leukoreduction changes the classes of lipids that accumulate during storage, and these lipids prime PMNs and induce acute lung injury (ALI) as the second event in a two-event in vivo model. STUDY DESIGN AND METHODS: RBC units were divided: 50% was leukoreduced (LR-RBCs), stored, and sampled on Day 1 and at the end of storage, Day 42. Priming activity was evaluated on isolated PMNs, and the purified lipids from Day 1 or Day 42 were used as the second event in the in vivo model. RESULTS: The plasma and lipids from RBCs and LR-RBCs primed PMNs, and the LR-RBC activity decreased with longer storage. Unlike RBCs, nonpolar lipids comprised the PMN-priming activity from stored LR-RBCs. Mass spectroscopy identified these lipids as arachidonic acid and 5-, 12-, and 15-hydroxyeicsotetranoic acid. At concentrations from Day 42, but not Day 1, three of four of these lipids individually, and the mixture, primed PMNs. The mixture also caused ALI as the second event in a two-event model of TRALI. CONCLUSION: We conclude that the nonpolar lipids that accumulate during LR-RBC storage may represent the agents responsible for antibody-negative TRALI.


Subject(s)
Acute Lung Injury/etiology , Arachidonic Acid/analysis , Blood Preservation/adverse effects , Erythrocytes/chemistry , Hydroxyeicosatetraenoic Acids/analysis , Leukocytes, Mononuclear/chemistry , Arachidonic Acid/metabolism , Erythrocyte Transfusion/adverse effects , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Leukapheresis/methods , Leukocytes, Mononuclear/metabolism , Male , Mass Spectrometry , Plasma/chemistry , Plasma/metabolism , Time Factors
8.
Shock ; 35(3): 240-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20926984

ABSTRACT

Leukotrienes are proinflammatory lipid mediators, derived from arachidonic acid via 5-lipoxygenase (5-LO). Leukotriene B4 (LTB4) is an effective polymorphonuclear neutrophil (PMN) chemoattractant, as well as being a major product of PMN priming. Leukotriene B4 is rapidly metabolized into products that are thought to be inactive, and little is known about the effects of LTB4 on the pulmonary endothelium. We hypothesize that LTB4 and its metabolites are effective PMN priming agents and cause proinflammatory activation of pulmonary endothelial cells. Isolated PMNs were primed (5 min, 37°C) with serial concentrations 10 to 10 M of LTB4 and its metabolites: 6-trans-LTB4, 20-OH-LTB4, and 20-COOH-LTB4, and then activated with fMLP. Primary human pulmonary microvascular endothelial cells (HMVECs) were incubated with these lipids (6 h, 37°C, 5% CO2), and intercellular adhesion molecule 1 was measured by flow cytometry. Polymorphonuclear neutrophil adhesion was measured by myeloperoxidase assays, and to ensure that these reactions were specific to the LTB4 receptors, BLT1 and BLT2 were antagonized with CP105,696 (BLT1) or silenced with siRNA (BLT1 and BLT2). Leukotriene B4 and its metabolites primed PMNs over a wide range of concentrations, depending on the specific metabolite. In addition, at high concentrations these lipids also caused increases in the surface expression of intercellular adhesion molecule 1 on HMVECs and induced HMVEC-mediated adhesion of PMNs. Silencing of BLT2 abrogated HMVEC activation, and blockade of BLT1 inhibited the observed PMN priming activity. We conclude that LTB4 and its ω-oxidation and nonenzymatic metabolites prime PMNs over a range of concentrations and activate HMVECs. These data have expanded the repertoire of causative agents in acute lung injury and postinjury multiple organ failure.


Subject(s)
Endothelial Cells/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Oxidoreductases/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Leukotriene B4/genetics , Leukotriene B4/metabolism , Lung/cytology , Lung/metabolism , Neutrophils/immunology , RNA Interference , Receptors, Leukotriene B4/genetics , Receptors, Leukotriene B4/metabolism
9.
Biochem J ; 432(1): 35-45, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20799926

ABSTRACT

Lyso-PCs (lysophosphatidylcholines) are a mixture of lipids that accumulate during storage of cellular blood components, have been implicated in TRALI (transfusion-related acute lung injury) and directly affect the physiology of neutrophils [PMNs (polymorphonuclear leucocytes)]. Because the G2A receptor, expressed on PMNs, has been reported to recognize lyso-PCs, we hypothesize that lyso-PC activation of G2A causes the increases in cytosolic Ca²(+) via release of G(α) and G(ßγ) subunits, kinase activation, and the recruitment of clathrin, ß-arrestin-1 and GRK6 (G-protein receptor kinase 6) to G2A for signal transduction. PMNs were isolated by standard techniques, primed with lyso-PCs for 5-180 s, and lysed for Western blot analysis, immunoprecipitation or subcellular fractionation, or fixed and smeared on to slides for digital microscopy. The results demonstrated that lyso-PCs cause rapid activation of the G2A receptor through S-phosphorylation and internalization resulting in G(αi)₋1 and G(αq/)11 release leading to increases in cytosolic Ca²(+), which was inhibited by an antibody to G2A or intracellular neutralization of these subunits. Lyso-PCs also caused the release of the G(ßγ) subunit which demonstrated a physical interaction (FRET+) with activated Hck (haemopoietic cell kinase; Tyr4¹¹). Moreover, G2A recruited clathrin, ß-arrestin-1 and GRK6: clathrin is important for signal transduction, GRK6 for receptor de-sensitization, and ß-arrestin-1 both propagates and terminates signals. We conclude that lyso-PC activation of G2A caused release of G(αi)₋1, G(αq/)11 and G(ßγ), resulting in cytosolic Ca²(+) flux, Hck activation, and recruitment of clathrin, ß-arrestin-1 and GRK6.


Subject(s)
Calcium/metabolism , Cell Cycle Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Lysophosphatidylcholines/pharmacology , Neutrophils/drug effects , Protein Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Arrestins/metabolism , Blotting, Western , Cells, Cultured , Clathrin/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer , G-Protein-Coupled Receptor Kinases/metabolism , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Humans , Ion Transport/drug effects , Microscopy, Fluorescence/methods , Neutrophils/cytology , Neutrophils/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-hck/metabolism , Signal Transduction/drug effects , Time Factors , beta-Arrestin 1 , beta-Arrestins
10.
Am J Physiol Cell Physiol ; 298(3): C714-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19907017

ABSTRACT

Neutrophils (PMNs) are a vital part of host defense and are the principal leukocyte in innate immunity. Interleukin (IL)-18 is a proinflammatory cytokine with roles in both innate and adaptive immunity. We hypothesize that PMNs contain preformed IL-18, which is released in response to specific inflammatory stimuli. Isolated PMNs were stimulated with a battery of chemoattractants (5 min to 24 h), and IL-18 release was measured. PMNs were also separated into subcellular fractions and immunoblotted with antibodies against IL-18 or were fixed and probed with antibodies to IL-18 as well as to the contents of granules, intracellular organelles, and filamentous actin (F-actin), incubated with fluorescent secondary antibodies, and examined by digital microscopy. Quiescent PMNs contained IL-18 in the cytoplasm, associated with F-actin, as determined by positive fluorescence resonance energy transfer (FRET+). In turn, TNF-alpha stimulation disrupted the association of IL-18 with F-actin, induced a FRET+ interaction of IL-18 with lipid rafts, and elicited IL-18 release. Manipulation of F-actin status confirmed the relationship between IL-18 and F-actin in resting PMNs. Consequently, incubation with monomeric IL-18 binding protein inhibited TNF-alpha-mediated priming of the PMN oxidase. We conclude that human PMNs contain IL-18 associated with F-actin in the cytoplasm and TNF-alpha stimulation causes dissociation of IL-18 from F-actin, association with lipid rafts, and extracellular release. Extracellular IL-18 participates in TNF-alpha priming of the PMN oxidase as demonstrated by inhibition with the IL-18 binding protein.


Subject(s)
Cytosol/immunology , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Neutrophils/immunology , Tumor Necrosis Factor-alpha/metabolism , Actins/metabolism , Adaptive Immunity , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique, Indirect , Humans , Immunity, Innate , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Microdomains/immunology , Microscopy, Fluorescence , Phosphoproteins/metabolism , Time Factors
11.
Blood Rev ; 23(6): 245-55, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19699017

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the most common cause of serious morbidity and mortality due to hemotherapy. Although the pathogenesis has been related to the infusion of donor antibodies into the recipient, antibody negative TRALI has been reported. Changes in transfusion practices, especially the use of male-only plasma, have decreased the number of antibody-mediated cases and deaths; however, TRALI still occurs. The neutrophil appears to be the effector cell in TRALI and the pathophysiology is centered on neutrophil-mediated endothelial cell cytotoxicity resulting in capillary leak and ALI. This review will detail the pathophysiology of TRALI including recent pre-clinical data, provide insight into newer areas of research, and critically assess current practices to decrease it prevalence and to make transfusion safer.


Subject(s)
Blood Transfusion/mortality , Lung Injury/etiology , Transfusion Reaction , Animals , Disease Models, Animal , Female , Humans , Lung Injury/mortality , Male , Mice , Rats
12.
Am J Physiol Cell Physiol ; 297(4): C886-97, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19295175

ABSTRACT

Receptor signaling is integral for adhesion, emigration, phagocytosis, and reactive oxygen species production in polymorphonuclear neutrophils (PMNs). Priming is an important part of PMN emigration, but it can also lead to PMN-mediated organ injury in the host. Platelet-activating factor (PAF) primes PMNs through activation of a specific G protein-coupled receptor. We hypothesize that PAF priming of PMNs requires clathrin-mediated endocytosis (CME) of the PAF receptor (PAFr), and, therefore, amantadine, known to inhibit CME, significantly antagonizes PAF signaling. PMNs were isolated by standard techniques to >98% purity and tested for viability. Amantadine (1 mM) significantly inhibited the PAF-mediated changes in the cellular distribution of clathrin and the physical colocalization [fluorescence resonance energy transfer positive (FRET+)] of early endosome antigen-1 and Rab5a, known components of CME and similar to hypertonic saline, a known inhibitor of CME. Furthermore, amantadine had no effect on the PAF-induced cytosolic calcium flux; however, phosphorylation of p38 MAPK was significantly decreased. Amantadine inhibited PAF-mediated changes in PMN physiology, including priming of the NADPH oxidase and shape change with lesser inhibition of increases in CD11b surface expression and elastase release. Furthermore, rimantadine, an amantadine analog, was a more potent inhibitor of PAF priming of the N-formyl-methionyl-leucyl-phenylalanine-activated oxidase. PAF priming of PMNs requires clathrin-mediated endocytosis that is inhibited when PMNs are pretreated with either amantadine or rimantadine. Thus, amantadine and rimantadine have the potential to ameliorate PMN-mediated tissue damage in humans.


Subject(s)
Amantadine/pharmacology , Clathrin/metabolism , Endocytosis , Neutrophils/physiology , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Antigens, CD1/metabolism , Enzyme Activation , Humans , In Vitro Techniques , NADPH Oxidases/metabolism , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Rimantadine/pharmacology , Signal Transduction
13.
Blood ; 113(9): 2079-87, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19131548

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion death. We hypothesize that TRALI requires 2 events: (1) the clinical condition of the patient and (2) the infusion of antibodies against MHC class I antigens or the plasma from stored blood. A 2-event rat model was developed with saline (NS) or endotoxin (LPS) as the first event and the infusion of plasma from packed red blood cells (PRBCs) or antibodies (OX18 and OX27) against MHC class I antigens as the second event. ALI was determined by Evans blue dye leak from the plasma to the bronchoalveolar lavage fluid (BALF), protein and CINC-1 concentrations in the BALF, and the lung histology. NS-treated rats did not evidence ALI with any second events, and LPS did not cause ALI. LPS-treated animals demonstrated ALI in response to plasma from stored PRBCs, both prestorage leukoreduced and unmodified, and to OX18 and OX27, all in a concentration-dependent fashion. ALI was neutrophil (PMN) dependent, and OX18/OX27 localized to the PMN surface in vivo and primed the oxidase of rat PMNs. We conclude that TRALI is the result of 2 events with the second events consisting of the plasma from stored blood and antibodies that prime PMNs.


Subject(s)
Acute Lung Injury/etiology , Antibodies/adverse effects , Erythrocyte Transfusion/adverse effects , Erythrocytes/physiology , Histocompatibility Antigens Class I/immunology , Plasma/physiology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Blood Preservation/adverse effects , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Humans , Male , Neutrophil Activation/immunology , Plasma/immunology , Rats , Rats, Sprague-Dawley
14.
J Immunol ; 180(12): 8192-203, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523285

ABSTRACT

Neutrophils (polymorphonuclear leukocytes, PMNs) are vital to innate immunity and receive proinflammatory signals that activate G protein-coupled receptors (GPCRs). Because GPCRs transduce signals through clathrin-mediated endocytosis (CME), we hypothesized that platelet-activating factor (PAF), an effective chemoattractant that primes the PMN oxidase, would signal through CME, specifically via dynamin-2 activation and endosomal formation resulting in membrane translocation of cytosolic phagocyte oxidase (phox) proteins. PMNs were incubated with buffer or 2 muM PAF for 1-3 min, and in some cases activated with PMA, and O(2)(-) was measured, whole-cell lysates and subcellular fractions were prepared, or the PMNs were fixed onto slides for digital or electron microscopy. PAF caused activation of dynamin-2, resulting in endosomal formation that required PI3K and contained early endosomal Ag-1 (EEA-1) and Rab5a. The apoptosis signal-regulating kinase-1/MAPK kinase-3/p38 MAPK signalosome assembled on Rab5a and phosphorylated EEA-1 and Rab GDP dissociation inhibitor, with the latter causing Rab5a activation. Electron microscopy demonstrated that PAF caused two distinct sites for activation of p38 MAPK. EEA-1 provided a scaffold for recruitment of the p40(phox)-p67(phox) complex and PI3K-dependent Akt1 phosphorylation of these two phox proteins. PAF induced membrane translocation of p40(phox)-p67(phox) localizing to gp91(phox), which was PI3K-, but not p47(phox)-, dependent. In conclusion, PAF transduces signals through CME, and such GPCR signaling may allow for pharmacological manipulation of these cells to decrease PMN-mediated acute organ injury.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Platelet Activating Factor/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Membrane/enzymology , Dynamin II/metabolism , Endosomes/enzymology , Enzyme Activation/physiology , Fluorescence Resonance Energy Transfer , Humans , Ligands , MAP Kinase Signaling System/physiology , Neutrophils/enzymology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Vesicular Transport Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
15.
Clin Cancer Res ; 14(12): 3942-7, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559616

ABSTRACT

PURPOSE: Platelet concentrates are important for support of patients with malignancies requiring myelotoxic chemotherapy. During storage, 10% to 15% of platelets may become activated resulting in the release of alpha-granules, which contain growth factors. We hypothesize that, during storage, growth factors accumulate in the plasma, specifically platelet-derived growth factor, vascular endothelial growth factor (VEGF), transforming growth factor-beta, and fibroblast growth factor-2, which may adversely affect cancer patients. EXPERIMENTAL DESIGN: The concentrations of growth factors were measured by ELISA from the plasma of apheresis platelets serially throughout storage (days 1, 3, 5, and 7) and compared with concentrations in fresh plasma from healthy blood donors. Washing was evaluated as a method of growth factor removal, and an in vitro model of platelet transfusion in a patient receiving Bevacizumab (Avastin) using immunoprecipitation was employed to determine if Bevacizumab would be bound by the VEGF in apheresis platelets. RESULTS: VEGF, platelet-derived growth factor, and transforming growth factor-beta were increased on day 1 versus fresh plasma and throughout storage reaching a relative maximum at outdate (P < 0.01, day 5 or 7). Fibroblast growth factor-2 concentrations were significantly increased on day 7 alone versus day 1 or to fresh plasma (P < 0.01). Washing removed 41 +/- 11% to 56 +/- 2% of the growth factors. Bevacizumab effectively bound the VEGF from apheresis platelets, with significant amounts of VEGF remaining in the supernatant. CONCLUSIONS: Significant amounts of growth factors are present in apheresis platelets due to the isolation procedures, and these concentrations increase over storage, which may be partially removed by washing. In addition, apheresis platelet transfusion could affect cancer treatment by binding monoclonal antibodies directed against growth factors of tumor origin.


Subject(s)
Angiogenic Proteins/metabolism , Blood Preservation , Carcinogens/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Plateletpheresis , Angiogenesis Inhibitors/therapeutic use , Angiogenic Proteins/analysis , Angiogenic Proteins/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Blood Preservation/adverse effects , Carcinogens/analysis , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/blood , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/blood , Platelet-Derived Growth Factor/analysis , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/blood , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/immunology
16.
Blood ; 109(4): 1752-5, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17038531

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related mortality. Antibodies to HNA-3a are commonly implicated in TRALI. We hypothesized that HNA-3a antibodies prime neutrophils (PMNs) and cause PMN-mediated cytotoxicity through a two-event pathogenesis. Isolated HNA-3a+ or HNA-3a- PMNs were incubated with plasma containing HNA-3a antibodies implicated in TRALI, and their ability to prime the oxidase was measured. Human pulmonary microvascular endothelial cells (HMVECs) were activated with endotoxin or buffer, HNA-3a+ or HNA-3a- PMNs were added, and the coculture was incubated with plasma+/-antibodies to HNA-3a. PMN-mediated damage was measured by counting viable HMVECs/mm2. Plasma containing HNA-3a antibodies primed the fMLP-activated respiratory burst of HNA-3a+, but not HNA-3a-, PMNs and elicited PMN-mediated damage of LPS-activated HMVECs when HNA-3a+, but not HNA-3a-, PMNs were used. Thus, antibodies to HNA-3a primed PMNs and caused PMN-mediated HMVEC cytotoxicity in a two-event model identical to biologic response modifiers implicated in TRALI.


Subject(s)
Endothelium, Vascular/pathology , Isoantibodies/immunology , Isoantigens/immunology , Neutrophil Activation/immunology , Pulmonary Circulation , Respiratory Distress Syndrome/immunology , Transfusion Reaction , Blood Donors , Cell Survival , Coculture Techniques , Cytotoxicity, Immunologic , Endothelial Cells/cytology , Humans , Neutrophils/cytology , Respiratory Burst , Respiratory Distress Syndrome/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...