Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
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.
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
3.
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
4.
J Leukoc Biol ; 73(4): 511-24, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12660226

ABSTRACT

A mixture of lysophosphatidylcholines (lyso-PCs) are generated during blood storage and are etiologic in models of acute lung injury. We hypothesize that lyso-PCs stimulate polymorphonuclear neutrophils (PMNs) through Ca(2)(+)-dependent signaling. The lyso-PC mix (0.45-14.5 micro M) and the individual lyso-PCs primed formyl-Met-Leu-Phe (fMLP) activation of the oxidase (1.8- to 15.7-fold and 1.7- to 14.8-fold; P<0.05). Labeled lyso-PCs demonstrated a membrane association with PMNs and caused rapid increases in cytosolic Ca(2)(+). Receptor desensitization studies implicated a common receptor or a family of receptors for the observed lyso-PC-mediated changes in PMN priming, and cytosolic Ca(2)(+) functions were pertussis toxin-sensitive. Lyso-PCs caused rapid serine phosphorylation of a 68-kD protein but did not activate mitogen-activated protein kinases or cause changes in tyrosine phosphorylation. With respect to alterations in PMN function, lyso-PCs caused PMN adherence, increased expression of CD11b and the fMLP receptor, reduced chemotaxis, provoked changes in morphology, elicited degranulation, and augmented fMLP-induced azurophilic degranulation (P<0.05). Cytosolic Ca(2)(+) chelation inhibited lyso-PC-mediated priming of the oxidase, CD11b surface expression, changes in PMN morphology, and serine phosphorylation of the 68-kD protein. In conclusion, lyso-PCs affect multiple PMN functions in a Ca(2)(+)-dependent manner that involves the activation of a pertussis toxin-sensitive G-protein.


Subject(s)
Calcium/metabolism , Lysophosphatidylcholines/pharmacology , NADPH Oxidases/metabolism , Neutrophils/physiology , Receptors, G-Protein-Coupled , CD11 Antigens/metabolism , Calcium Signaling , Cell Adhesion/drug effects , Chemotaxis/drug effects , Cytosol , Enzyme Activation , Humans , Intercellular Signaling Peptides and Proteins , Lactoferrin/metabolism , Lysophosphatidylcholines/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , N-Formylmethionine Leucyl-Phenylalanine , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Peroxidase/metabolism , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Formyl Peptide , Receptors, Immunologic/metabolism , Receptors, Peptide/metabolism , Serine/metabolism , Tyrosine/metabolism
5.
Am J Physiol Cell Physiol ; 283(6): C1592-603, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12388071

ABSTRACT

Lysophosphatidylcholines (lyso-PCs), generated during blood storage, are etiologic in a two-insult, sepsis-based model of transfusion-related acute lung injury (TRALI). Individually, endotoxin (LPS) and lyso-PCs prime but do not activate neutrophils (PMNs). We hypothesized that priming of PMNs alters their reactivity such that a second priming agent causes PMN activation and endothelial cell damage. PMNs were primed or not with LPS and then treated with lyso-PCs, and oxidase activation and elastase release were measured. For coculture experiments, activation of human pulmonary microvascular endothelial cells (HMVECs) was assessed by ICAM-1 expression and chemokine release. HMVECs were stimulated or not with LPS, PMNs were added, cells were incubated with lyso-PCs, and the number of viable HMVECs was counted. Lyso-PCs activated LPS-primed PMNs. HMVEC activation resulted in increased ICAM-1 and release of ENA-78, GRO alpha, and IL-8. PMN-mediated HMVEC damage was dependent on LPS activation of HMVECs, chemokine release, PMN adhesion, and lyso-PC activation of the oxidase. In conclusion, sequential exposure of PMNs to priming agents activates the microbicidal arsenal, and PMN-mediated HMVEC damage was the result of two insults: HMVEC activation and PMN oxidase assembly.


Subject(s)
Chemokines, CXC , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Interleukin-8/analogs & derivatives , Neutrophils/physiology , Pulmonary Circulation , Cell Adhesion , Cell Count , Cells, Cultured , Chemokine CXCL1 , Chemokine CXCL5 , Chemokines/metabolism , Chemotactic Factors/metabolism , Coculture Techniques , Enzyme Activation , Humans , Intercellular Adhesion Molecule-1/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Lysophosphatidylcholines/pharmacology , Microcirculation , Neutrophil Activation , Neutrophils/drug effects , Oxidoreductases/metabolism , Pancreatic Elastase/metabolism
6.
Am J Physiol Cell Physiol ; 283(4): C1102-13, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12225974

ABSTRACT

Secretory phospholipase A(2) (sPLA(2)) produces lipids that stimulate polymorphonuclear neutrophils (PMNs). With the discovery of sPLA(2) receptors (sPLA(2)-R), we hypothesize that sPLA(2) stimulates PMNs through a receptor. Scatchard analysis was used to determine the presence of a sPLA(2) ligand. Lysates were probed with an antibody to the M-type sPLA(2)-R, and the immunoreactivity was localized. PMNs were treated with active and inactive (+EGTA) sPLA(2) (1-100 units of enzyme activity/ml, types IA, IB, and IIA), and elastase release and PMN adhesion were measured. PMNs incubated with inactive, FITC-linked sPLA(2)-IB, but not sPLA(2)-IA, demonstrated the presence of a sPLA(2)-R with saturation at 2.77 fM and a K(d) of 167 pM. sPLA(2)-R immunoreactivity was present at 185 kDa and localized to the membrane. Inactive sPLA(2)-IB activated p38 MAPK, and p38 MAPK inhibition attenuated elastase release. Active sPLA(2)-IA caused elastase release, but inactive type IA did not. sPLA(2)-IB stimulated elastase release independent of activity; inactive sPLA(2)-IIA partially stimulated PMNs. sPLA(2)-IB and sPLA(2)-IIA caused PMN adhesion. We conclude that PMNs contain a membrane M-type sPLA(2)-R that activates p38 MAPK.


Subject(s)
Leukocyte Elastase/metabolism , Neutrophils/metabolism , Receptors, Cell Surface/physiology , Binding, Competitive/drug effects , Calcium/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Cell Separation , Chelating Agents/metabolism , Chelating Agents/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Fluorescent Dyes , Group IB Phospholipases A2 , Group II Phospholipases A2 , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/drug effects , Phospholipases A/antagonists & inhibitors , Phospholipases A/pharmacology , Receptors, Phospholipase A2 , p38 Mitogen-Activated Protein Kinases
7.
J Leukoc Biol ; 72(2): 401-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12149432

ABSTRACT

Patients with sepsis and acute lung injury have increased interleukin (IL)-18 levels systemically. We hypothesize that IL-18 stimulates neutrophils (PMNs) at physiologic concentrations. IL-18 primed the oxidase at 15 min (10-100 ng/ml), 30 min (0.1-100 ng/ml), and 60 min (100 ng/ml; P<0.05) and caused translocation of p47(phox) to the membrane similar to lipopolysaccharides. CD11b surface expression was increased by IL-18 in a time- and concentration-dependent manner. IL-18 caused up-regulation of the formyl-Met-Leu-Phe receptor, changes in PMN size, and elastase release. Investigation of signaling demonstrated IL-18-mediated activation of p38 mitogen-activated protein (MAP) kinase in a concentration (0.1-100 ng/ml)-, time (5-15 min)-, and Ca2+-dependent manner. IL-18 directly increased cytosolic Ca2+ concentration. IL-18 activation of PMNs was blocked by inhibition of p38 MAP kinase activity (SB203580) or by inhibition of p38 MAP kinase activation by chelation of cytosolic Ca2+. We conclude that IL-18, at physiologic concentrations, is an effective PMN priming agent that requires p38 MAP kinase activity.


Subject(s)
Egtazic Acid/analogs & derivatives , Interleukin-18/physiology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/physiology , Neutrophils/drug effects , Adult , Calcium , Calcium Signaling/drug effects , Cell Size , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Interleukin-18/pharmacology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophage-1 Antigen/biosynthesis , Macrophage-1 Antigen/genetics , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/ultrastructure , Phosphoproteins/metabolism , Protein Transport/drug effects , Pyridines/pharmacology , Receptors, Formyl Peptide , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Peptide/biosynthesis , Receptors, Peptide/genetics , Respiratory Burst/drug effects , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...