Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 6(5): e19938, 2011.
Article in English | MEDLINE | ID: mdl-21603616

ABSTRACT

The transmembrane protease ADAM17 regulates the release and density of various leukocyte cell surface proteins that modulate inflammation, including L-selectin, TNF-α, and IL-6R. At this time, its in vivo substrates and role in pulmonary inflammation have not been directly examined. Using conditional ADAM17 knock-out mice, we investigated leukocyte ADAM17 in acute lung inflammation. Alveolar TNF-α levels were significantly reduced (>95%) in ADAM17-null mice following LPS administration, as was the shedding of L-selectin, a neutrophil-expressed adhesion molecule. Alveolar IL-6R levels, however, were reduced by only ≈25% in ADAM17-null mice, indicating that ADAM17 is not its primary sheddase in our model. Neutrophil infiltration into the alveolar compartment is a key event in the pathophysiology of acute airway inflammation. Following LPS inhalation, alveolar neutrophil levels and lung inflammation in ADAM17-null mice were overall reduced when compared to control mice. Interestingly, however, neutrophil recruitment to the alveolar compartment occurred earlier in ADAM17-null mice after exposure to LPS. This decrease in alveolar neutrophil recruitment in ADAM17-null mice was accompanied by significantly diminished alveolar levels of the neutrophil-tropic chemokines CXCL1 and CXCL5. Altogether, our study suggests that leukocyte ADAM17 promotes inflammation in the lung, and thus this sheddase may be a potential target in the design of pharmacologic therapies for acute lung injury.


Subject(s)
ADAM Proteins/immunology , Leukocytes/immunology , Pneumonia/immunology , ADAM17 Protein , Acute Disease , Animals , Chemokine CXCL1 , Chemokine CXCL5 , L-Selectin , Leukocytes/enzymology , Lipopolysaccharides/pharmacology , Lung/chemistry , Lung/immunology , Mice , Mice, Knockout , Neutrophil Infiltration , Pneumonia/etiology , Tumor Necrosis Factor-alpha
2.
Exp Lung Res ; 35(8): 682-700, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19895322

ABSTRACT

Subsequent to the initial recruitment of neutrophils, monocytes are recruited to the lung after an injurious insult. Previously the authors have shown that inhibition of either p38 or c-Jun NH(2)-terminal kinase (JNK) decreased pulmonary neutrophil recruitment in mice exposed to lipopolysaccharide (LPS). As the signaling pathways regulating the influx of mononuclear cells to the lung are poorly understood, the authors undertook the present study to examine the roles of p38 and JNK. In a model of LPS-induced lung inflammation, systemic inhibition of JNK, but not p38, decreased the recruitment of mononuclear cells to the lung. Levels of CCL2 (monocyte chemoattractant protein 1 [MCP-1]) were decreased in the setting of JNK inhibition, with LPS-induced pulmonary mononuclear cell recruitment in CCL2-deficient mice similar to that found with JNK inhibition. The decrease in LPS-induced CCL2 levels in the lung seen with JNK inhibition, however, was independent of neutrophil recruitment, as systemic depletion of neutrophils had no effect on pulmonary CCL2 levels after LPS exposure. In sum, these results suggest that JNK, but not p38, regulates LPS-induced mononuclear cell recruitment to the lung, that this occurs through a CCL2-dependent pathway, and that LPS-induced pulmonary CCL2 expression is dependent on JNK but independent of pulmonary neutrophil recruitment.


Subject(s)
Chemokine CCL2/immunology , JNK Mitogen-Activated Protein Kinases/physiology , Lung/immunology , Neutrophil Infiltration/immunology , Animals , Chemokine CCL2/deficiency , Female , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , p38 Mitogen-Activated Protein Kinases
3.
Cell Immunol ; 258(1): 90-7, 2009.
Article in English | MEDLINE | ID: mdl-19393603

ABSTRACT

The acute inflammatory response involves neutrophils wherein recognition of bacterial products, such as lipopolysaccharide (LPS), activates intracellular signaling pathways. We have shown that the mitogen-activated protein kinase (MAPK) c-Jun NH(2) terminal kinase (JNK) is activated by LPS in neutrophils and plays a critical role in monocyte chemoattractant protein (MCP)-1 expression and actin assembly. As the Tec family kinases are expressed in neutrophils and regulate activation of the MAPKs in other cell systems, we hypothesized that the Tec kinases are an upstream component of the signaling pathway leading to LPS-induced MAPKs activation in neutrophils. Herein, we show that the Tec kinases are activated in LPS-stimulated human neutrophils and that inhibition of the Tec kinases, with leflunomide metabolite analog (LFM-A13), decreased LPS-induced JNK, but not p38, activity. Furthermore, LPS-induced actin polymerization as well as MCP-1, tumor necrosis factor-alpha, interleukin-6, and interleukin-1beta expression are dependent on Tec kinase activity.


Subject(s)
Actins/physiology , Chemokine CCL2/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Neutrophils/immunology , Protein-Tyrosine Kinases/metabolism , Amides/pharmacology , Cell Membrane/physiology , Chemokine CCL2/immunology , Cytokines/immunology , Cytokines/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/immunology , Neutrophils/drug effects , Nitriles/pharmacology , Phosphorylation/drug effects , Phosphorylation/immunology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Blood ; 109(3): 1248-56, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17018860

ABSTRACT

Production of tumor necrosis factor-alpha (TNFalpha) by the neutrophil (PMN) is a pivotal event in innate immunity, but the signals regulating TNFalpha induction in this primary cell are poorly understood. Herein, we use protein transduction to identify novel, opposing anti- and pro-cytokine-inducing roles for RhoA in the resting and lipopolysaccharide (LPS)-stimulated human PMN, respectively. In the resting cell, RhoA suppresses Cdc42 activation, IkappaBalpha degradation, nuclear factor-kappaB (NF-kappaB) activation, and induction of TNFalpha and NF-kappaB-dependent chemokines. Suppression of TNFalpha induction by RhoA is Rho kinase alpha (ROCKalpha) independent, but Cdc42 dependent, because TNFalpha induction by C3 transferase is attenuated by inhibition of Cdc42, and constitutively active Cdc42 suffices to activate NF-kappaB and induce TNFalpha. By contrast, we also place RhoA downstream of p38 mitogen-activated protein kinase and Cdc42 in a novel LPS-activated pathway in which p38, Cdc42, and ROCKalpha all promote TNFalpha protein expression. The p65 subunit of NF-kappaB coprecipitates with RhoA in a manner sensitive to the RhoA activation state. Our findings suggest a new, 2-faced role for RhoA as a checkpoint in innate immunity.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , Neutrophils/metabolism , rhoA GTP-Binding Protein/physiology , Down-Regulation , Humans , I-kappa B Proteins/metabolism , Immunity, Innate , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Neutrophils/immunology , Tumor Necrosis Factor-alpha/genetics , Up-Regulation , cdc42 GTP-Binding Protein/physiology
5.
J Immunol ; 177(10): 7233-41, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082641

ABSTRACT

Recruitment of neutrophils to the lung is a sentinel event in acute lung inflammation. Identifying mechanisms that regulate neutrophil recruitment to the lung may result in strategies to limit lung damage and improve clinical outcomes. Recently, the renin angiotensin system (RAS) has been shown to regulate neutrophil influx in acute inflammatory models of cardiac, neurologic, and gastrointestinal disease. As a role for the RAS in LPS-induced acute lung inflammation has not been described, we undertook this study to examine the possibility that the RAS regulates neutrophil recruitment to the lung after LPS exposure. Pretreatment of mice with the angiotensin-converting enzyme (ACE) inhibitor enalapril, but not the anti-hypertensive hydralazine, decreased pulmonary neutrophil recruitment after exposure to LPS. We hypothesize that inhibition of LPS-induced neutrophil accumulation to the lung with enalapril occurred through both an increase in bradykinin, and a decrease in angiotensin II (ATII), mediated signaling. Bradykinin receptor blockade reversed the inhibitory effect of enalapril on neutrophil recruitment. Similarly, pretreatment with bradykinin receptor agonists inhibited IL-8-induced neutrophil chemotaxis and LPS-induced neutrophil recruitment to the lung. Inhibition of ATII-mediated signaling, with the ATII receptor 1a inhibitor losartan, decreased LPS-induced pulmonary neutrophil recruitment, and this was suggested to occur through decreased PAI-1 levels. LPS-induced PAI-1 levels were diminished in animals pretreated with losartan and in those deficient for the ATII receptor 1a. Taken together, these results suggest that ACE regulates LPS-induced pulmonary neutrophil recruitment via modulation of both bradykinin- and ATII-mediated pathways, each regulating neutrophil recruitment by separate, but distinct, mechanisms.


Subject(s)
Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/physiology , Lipopolysaccharides/antagonists & inhibitors , Lung/enzymology , Lung/pathology , Signal Transduction/immunology , Angiotensin II/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Bradykinin/agonists , Cell Migration Inhibition , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines/antagonists & inhibitors , Chemokines/metabolism , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/metabolism , Chemotaxis, Leukocyte , Down-Regulation/immunology , Female , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Losartan/pharmacology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Bradykinin/agonists , Signal Transduction/drug effects
6.
J Immunol ; 177(1): 538-47, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785551

ABSTRACT

Bacterial pneumonia is a leading cause of mortality and is associated with extensive neutrophil accumulation. Major pathogens associated with this disease include nonflagellated Klebsiella pneumoniae (Kp) and flagellated Pseudomonas aeruginosa (Pa). TLRs are essential for innate immune defense. TIRAP (Toll/IL-1R domain-containing adaptor protein) is an adaptor in TLR1, TLR2, TLR4, and TLR6 signaling, whereas MyD88 is an adaptor for all TLRs. However, the importance of TIRAP in pulmonary defense against Kp or Pa has not been examined. To demonstrate the role of TIRAP, TIRAP-deficient and wild-type littermates were intratracheally inoculated with Kp or Pa. We found that TIRAP(-/-) mice had substantial mortality, higher bacterial burden in the lungs, and enhanced dissemination following Kp challenge. Furthermore, Kp-induced neutrophil sequestration, histopathology, and MIP-2, TNF-alpha, IL-6, and LIX (lipopolysaccharide-induced CXC chemokine) production were attenuated in the lungs of TIRAP(-/-) mice. In contrast, TIRAP is not required for Pa-induced mortality, pulmonary bacterial burden, bacterial dissemination, neutrophil accumulation, or histopathology, yet it is necessary for MIP-2, TNF-alpha, and IL-6 production, but not LIX production. However, both Kp- and Pa-induced neutrophil influxes are MyD88 dependent. To determine the mechanisms associated with Pa-induced neutrophil accumulation, we inoculated mice with a flagellin C mutant of Pa (PaDeltafliC) or purified flagellin, a TLR5 agonist. PaDeltafliC-induced neutrophil sequestration and LIX expression are dependent on TIRAP, whereas flagellin-induced neutrophil influx and LIX expression are independent of TIRAP. These novel findings illustrate a pathogen-specific role for TIRAP in pulmonary defense and suggest that TLR5 plays an essential role for Pa-induced neutrophil influx via LIX production.


Subject(s)
Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Membrane Glycoproteins/physiology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Receptors, Interleukin-1/physiology , Animals , Chemokines/biosynthesis , Cytokines/biosynthesis , Down-Regulation/immunology , Female , Flagellin/genetics , Intubation, Intratracheal , Klebsiella Infections/genetics , Klebsiella Infections/microbiology , Klebsiella Infections/mortality , Lung/immunology , Lung/microbiology , Lung/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality , Pseudomonas Infections/genetics , Pseudomonas Infections/microbiology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Survival Rate , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis
7.
J Immunol ; 175(6): 4049-59, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148154

ABSTRACT

The neutrophil is of undoubted importance in lung inflammation after exposure to LPS. We have shown recently that systemic inhibition of JNK decreased neutrophil recruitment to the lung after exposure to LPS, although the mechanisms underlying this inhibition are incompletely understood. As plasminogen activator inhibitor-1 (PAI-1) accentuates cell migration, with JNK activation recently shown to up-regulate PAI-1 expression, this suggested that systemic JNK inhibition may down-regulate LPS-induced pulmonary neutrophil recruitment through a decrease in PAI-1 expression. We show in this study that exposure of mice to aerosolized LPS increased PAI-1 expression in the lung and alveolar compartment, which was decreased by pretreatment with the JNK inhibitor SP600125. Exogenous, intratracheally administered PAI-1 prevented the inhibition of pulmonary neutrophil recruitment in the setting of systemic JNK inhibition, thereby suggesting a role for PAI-1 in the JNK-mediated pathway regulating LPS-induced neutrophil recruitment. In addition, PAI-1(-/-) mice had a decrease in neutrophil recruitment to the alveolar compartment after exposure to LPS, compared with wild-type controls, further suggesting a role for PAI-1 in LPS-induced lung inflammation. An increase in the intravascular level of KC is a likely mechanism for the inhibition of pulmonary neutrophil recruitment after LPS exposure in the setting of decreased PAI-1 expression, as systemic KC levels after exposure to LPS were increased in PAI-1-deficient mice and in mice pretreated with SP600125, with augmentation of intravascular KC levels inhibiting neutrophil recruitment to the lung after exposure to LPS.


Subject(s)
Inflammation/chemically induced , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Plasminogen Activator Inhibitor 1/physiology , Animals , Cell Line , Chemotaxis, Leukocyte , Female , Interleukin-8/blood , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/physiology , Plasminogen Activator Inhibitor 1/analysis , Plasminogen Activator Inhibitor 1/genetics , Protein Kinase Inhibitors/pharmacology , Up-Regulation
8.
Am J Respir Crit Care Med ; 171(9): 978-86, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15735060

ABSTRACT

The influx of neutrophils into the lung is a sentinel event in LPS-induced acute lung inflammation. Previous studies have shown that systemic inhibition of p38 decreases LPS-induced neutrophil influx into the alveolar space but has no effect on pulmonary parenchymal neutrophil accumulation or on microvascular leak, indicating other pathways are important in LPS-induced acute lung inflammation. This study examined the role of c-Jun N-terminal kinase in LPS-induced acute lung inflammation. Systemic inhibition of c-Jun N-terminal kinase, with the specific c-Jun N-terminal kinase inhibitor SP600125, decreased the LPS-induced accumulation of neutrophils into the lung parenchyma and alveolar space. In addition, increases in microvascular leak after LPS exposure were diminished by c-Jun N-terminal kinase inhibition. To determine mechanisms by which systemic c-Jun N-terminal kinase inhibition decreased pulmonary neutrophil influx, LPS and tumor necrosis factor alpha (TNF-alpha-)-induced neutrophil actin assembly and retention were examined. Neutrophil actin assembly was decreased after LPS and TNF-alpha stimulation with SP600125 pretreatment, as well as LPS-induced neutrophil retention. Finally, c-Jun N-terminal kinase inhibition decreased Cdc42 activation after LPS or TNF-alpha stimulation, thereby providing one mechanism by which c-Jun N-terminal kinase inhibition decreased actin assembly, and thereby pulmonary neutrophil accumulation.


Subject(s)
JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lung/immunology , Neutrophil Infiltration/physiology , Animals , Anthracenes/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability , Female , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli , Signal Transduction/physiology , cdc42 GTP-Binding Protein/physiology
9.
Am J Respir Crit Care Med ; 171(6): 606-15, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15591471

ABSTRACT

RATIONALE: A growing literature indicates that hydroxy-methylglutaryl coenzyme A reductase inhibitors (statins) modulate proinflammatory cellular signaling and functions. No studies to date, however, have addressed whether statins modulate pulmonary inflammation triggered by aerogenic stimuli or whether they affect host defense. OBJECTIVES: To test whether lovastatin modulates LPS-induced pulmonary inflammation and antibacterial host defense. METHODS: To address these questions, and to confirm any effect of statins as dependent on inhibition of hydroxy-methylglutaryl coenzyme A reductase, we treated C57Bl/6 mice with three oral doses of 10 mg/kg lovastatin (or vehicle) and three intraperitoneal doses of 10 mg/kg mevalonic acid (or saline), and then exposed them to the following: (1) aerosolized LPS, (2) intratracheal keratinocyte-derived chemokine (KC), or (3) intratracheal Klebsiella pneumoniae. MEASUREMENTS AND MAIN RESULTS: LPS- and KC-induced airspace neutrophils were reduced by lovastatin, an effect that was blocked by mevalonic acid cotreatment. Lovastatin was also associated with reduced parenchymal myeloperoxidase and microvascular permeability, and altered airspace and serum cytokines after LPS. Native pulmonary clearance of K. pneumoniae was inhibited by lovastatin and extrapulmonary dissemination was enhanced, both reversibly with mevalonic acid. Ex vivo studies of neutrophils isolated from lovastatin-treated mice confirmed inhibitory effects on Rac activation, actin polymerization, chemotaxis, and bacterial killing. CONCLUSION: Lovastatin attenuates pulmonary inflammation induced by aerosolized LPS and impairs host defense.


Subject(s)
Hydroxymethylglutaryl CoA Reductases/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Lung/immunology , Pneumonia/etiology , Pneumonia/immunology , Actins/metabolism , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Communication , Chemokines , Chemotaxis , Female , Humans , Klebsiella pneumoniae , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Pneumonia, Bacterial/immunology
10.
J Biol Chem ; 279(38): 39989-98, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15262974

ABSTRACT

Lipid rafts are cholesterol-rich membrane microdomains that are thought to act as coordinated signaling platforms by regulating dynamic, agonist-induced translocation of signaling proteins. They have been described to play a role in multiple prototypical cascades, among them the lipopolysaccharide pathway, and to host multiple signaling proteins, including kinases and low molecular weight G-proteins. Here we report lipopolysaccharide-induced activation of the Rho family GTPase Cdc42, and we show its activation in the human neutrophil to be mediated by a p38 mitogen-activated protein kinase-dependent mechanism. Subcellular fractionation reveals that lipopolysaccharide induces translocation of Cdc42 to lipid rafts, where it and p38 are both found to be activated. By contrast, lipopolysaccharide causes translocation of Rac from the polymorphonuclear leukocyte (PMN) rafts and does not induce its activation. With the use of methyl-beta-cyclodextrin, a cholesterol-depleting agent that reversibly disrupts rafts, we confirm an important regulatory role for rafts in the activation state of p38 and Cdc42 and in the Rho GTPase-dependent functions superoxide anion production and actin polymerization. Methyl-beta-cyclodextrin induces activation of p38 and Cdc42, but not Rac, in the nonstimulated PMN, yet inhibits subsequent lipopolysaccharide-induced activation of p38 and Cdc42. In parallel, methyl-beta-cyclodextrin primes the human PMN for subsequent superoxide release triggered by the formylated bacterial tripeptide formyl-Met-Leu-Phe, and induces actin polymerization in a subcellular distribution distinct from that induced by lipopolysaccharide. In sum, these findings provide evidence for an important regulatory role of cholesterol in both transmission of the lipopolysaccharide signal and the inflammatory phenotype of the human neutrophil.


Subject(s)
Membrane Microdomains/metabolism , Neutrophils/immunology , Neutrophils/metabolism , beta-Cyclodextrins , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Cholesterol/metabolism , Cyclodextrins/pharmacology , Humans , Lipopolysaccharides/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Mitogen-Activated Protein Kinases/metabolism , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
11.
J Biol Chem ; 279(12): 10883-91, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14699155

ABSTRACT

Polymorphonuclear leukocytes (neutrophils) respond to lipopolysaccharide (LPS) through the up-regulation of several pro-inflammatory mediators. We have recently shown that LPS-stimulated neutrophils express monocyte chemoattractant protein 1 (MCP-1), an AP-1-dependent gene, suggesting that LPS activates the c-Jun N-terminal kinase (JNK) pathway in neutrophils. Previously, we have shown the activation of p38 MAPK, but not JNK, in suspended neutrophils stimulated with LPS but have recently shown activation of JNK by TNF-alpha in an adherent neutrophil system. We show here that exposure to LPS activates JNK in non-suspended neutrophils and that LPS-induced MCP-1 expression, but not tumor necrosis factor-alpha (TNF-alpha) or interleukin-8 (IL-8), is dependent on JNK activation. In addition, LPS stimulation of non-suspended neutrophils activates Syk and phosphatidylinositol 3-kinase (PI3K). Inhibition of Syk with piceatannol or PI3K with wortmannin inhibited LPS-induced JNK activation and decreased MCP-1 expression after exposure to LPS, suggesting that both Syk and PI3K reside in a signaling pathway leading to LPS-induced JNK activation in neutrophils. This Syk- and PI3K-dependent pathway leading to JNK activation after LPS exposure in non-suspended neutrophils is specific for JNK, because inhibition of neither Syk nor PI3K decreased p38 activation after LPS stimulation. Furthermore we show that PI3K inhibition decreased LPS-induced Syk activation suggesting that PI3K resides upstream of Syk in this pathway. Finally, we show that Syk associates with Toll-like receptor 4 (TLR4) upon LPS stimulation further implicating Syk in the LPS-induced signaling pathway in neutrophils. Overall our data suggests that LPS induces JNK activation only in non-suspended neutrophils, which proceeds through Syk- and PI3K-dependent pathways, and that JNK activation is important for LPS-induced MCP-1 expression but not for TNF-alpha or IL-8 expression.


Subject(s)
Enzyme Precursors/metabolism , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Anthracenes/pharmacology , Base Sequence , Chemokine CCL2/metabolism , DNA Primers , Enzyme Activation , Enzyme Precursors/antagonists & inhibitors , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Neutrophils/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Syk Kinase
12.
Am J Physiol Lung Cell Mol Physiol ; 284(4): L663-70, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12495940

ABSTRACT

Neutrophils respond to infection by degranulation, release of reactive oxygen intermediates, and secretion of chemokines and cytokines; however, activation of neutrophil transcriptional machinery has been little appreciated. Recent findings suggest that gene expression may represent an additional neutrophil function after exposure to lipopolysaccharide (LPS). We performed microarray gene expression analysis of 4,608 mostly nonredundant genes on LPS-stimulated human neutrophils. Analysis of three donors indicated some variability but also a high degree of reproducibility in gene expression. Twenty-eight verifiable, distinct genes were induced by 4 h of LPS treatment, and 13 genes were repressed. Genes other than cytokines and chemokines are regulated; interestingly, genes involved in cell growth regulation and survival, transcriptional regulation, and interferon response are among those induced, whereas genes involved in cytoskeletal regulation are predominantly repressed. In addition, we identified monocyte chemoattractant protein-1 as a novel LPS-regulated chemokine in neutrophils. Included in these lists are five clones with no defined function. These data suggest molecular mechanisms by which neutrophils respond to infection and indicate that the transcriptional potential of neutrophils is greater than previously thought.


Subject(s)
Lipopolysaccharides/pharmacology , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis , Gene Expression Regulation/drug effects , Humans , Lung/immunology , Neutrophils/drug effects , Oligonucleotide Array Sequence Analysis/standards , Reproducibility of Results , Up-Regulation/genetics
13.
J Immunol ; 169(9): 5260-9, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391245

ABSTRACT

The p38 mitogen-activated protein kinase (MAPK) signaling pathway regulates a wide range of inflammatory responses in many different cells. Inhibition of p38 MAPK before exposing a cell to stress stimuli has profound anti-inflammatory effects, but little is known about the effects of p38 MAPK inhibition on ongoing inflammatory responses. LPS-induced activation of p38 MAPK in human neutrophils was inhibited by poststimulation exposure to a p38 MAPK inhibitor (M39). Release of TNF-alpha, macrophage-inflammatory protein (MIP)-2 (MIP-1beta), and IL-8 by LPS-stimulated neutrophils was also reduced by poststimulation p38 MAPK inhibition. In contrast, release of monocyte chemoattractant protein-1 was found to be p38 MAPK independent. Ongoing chemotaxis toward IL-8 was eliminated by p38 MAPK inhibition, although the rate of nondirectional movement was not reduced. A murine model of acute LPS-induced lung inflammation was used to study the effect of p38 MAPK inhibition in ongoing pulmonary inflammation. Initial pulmonary cell responses occur within 4 h of stimulation in this model, so M39 was administered 4 h or 12 h after exposure of the animals to aerosolized LPS to avoid inhibition of cytokine release. Quantities of TNF-alpha, MIP-2, KC, or monocyte chemoattractant protein-1 recovered from bronchial alveolar lavage or serum were not changed. Recruitment of neutrophils, but not other leukocytes, to the airspaces was significantly reduced. Together, these data demonstrate the selective reduction of LPS-induced neutrophil recruitment to the airspaces, independent of suppression of other inflammatory responses. These findings support the feasibility of p38 MAPK inhibition as a selective intervention to reduce neutrophilic inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Lung/enzymology , Lung/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neutrophil Infiltration/immunology , Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Animals , Cell Migration Inhibition , Chemotaxis, Leukocyte/drug effects , Cytokines/blood , Cytokines/metabolism , Drug Administration Schedule , Enzyme Activation/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Inflammation/enzymology , Inflammation/pathology , Inflammation/prevention & control , Intubation, Gastrointestinal , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...