Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Clin Exp Allergy ; 47(9): 1214-1222, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28471517

ABSTRACT

BACKGROUND: The Duffy antigen receptor for chemokines (DARC) is an atypical receptor that regulates pro-inflammatory cytokines. However, the role of DARC in asthma pathophysiology is unknown. OBJECTIVE: To determine the role of DARC in allergic airways disease in mice, and the association between DARC single nucleotide polymorphisms (SNPs) and clinical outcomes in patients with asthma. METHODS: Mice with targeted disruption of the Darc gene (Darc∆E2 ) or WT mice were challenged over 3 weeks with house dust mite (HDM) antigen. Allergic airways disease was assessed 24 hours and 7 days following the final challenge. Additionally, associations between DARC SNPs and clinical outcomes were analysed in a cohort of poorly controlled asthmatics. RESULTS: Total airway inflammation following HDM did not differ between Darc∆E2 and WT mice. At 24 hours, Darc∆E2 mice had increased airway hyperresponsiveness; however, at 7 days airway hyperresponsiveness had completely resolved in Darc∆E2 but persisted in WT mice. In poorly controlled asthmatics, DARC SNPs were associated with worse asthma control at randomization and subsequent increased risk of healthcare utilization (odds ratio 3.13(1.37-7.27), P=.0062). CONCLUSIONS AND CLINICAL RELEVANCE: Our animal model and human patient data suggest a novel role for DARC in the temporal regulation in asthma pathophysiology and symptoms.


Subject(s)
Asthma , Chemokines , Duffy Blood-Group System , Receptors, Cell Surface , Animals , Female , Humans , Male , Mice , Antigens, Dermatophagoides/immunology , Asthma/diagnosis , Asthma/etiology , Asthma/metabolism , Chemokines/metabolism , Disease Models, Animal , Disease Susceptibility , Duffy Blood-Group System/genetics , Duffy Blood-Group System/metabolism , Gene Expression , Genetic Loci , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Patient Acceptance of Health Care , Patient Outcome Assessment , Phenotype , Polymorphism, Single Nucleotide , Prognosis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Respiratory Hypersensitivity/diagnosis , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Severity of Illness Index
2.
Mucosal Immunol ; 10(6): 1529-1541, 2017 11.
Article in English | MEDLINE | ID: mdl-28120850

ABSTRACT

Annual epidemics and unexpected pandemics of influenza are threats to human health. Lung immune and inflammatory responses, such as those induced by respiratory infection influenza virus, determine the outcome of pulmonary pathogenesis. Platelet-derived chemokine (C-X-C motif) ligand 4 (CXCL4) has an immunoregulatory role in inflammatory diseases. Here we show that CXCL4 is associated with pulmonary influenza infection and has a critical role in protecting mice from fatal H1N1 virus respiratory infection. CXCL4 knockout resulted in diminished viral clearance from the lung and decreased lung inflammation during early infection but more severe lung pathology relative to wild-type mice during late infection. Additionally, CXCL4 deficiency decreased leukocyte accumulation in the infected lung with markedly decreased neutrophil infiltration into the lung during early infection and extensive leukocyte, especially lymphocyte accumulation at the late infection stage. Loss of CXCL4 did not affect the activation of adaptive immune T and B lymphocytes during the late stage of lung infection. Further study revealed that CXCL4 deficiency inhibited neutrophil recruitment to the infected mouse lung. Thus the above results identify CXCL4 as a vital immunoregulatory chemokine essential for protecting mice against influenza A virus infection, especially as it affects the development of lung injury and neutrophil mobilization to the inflamed lung.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Lung/physiology , Neutrophils/immunology , Orthomyxoviridae Infections/immunology , Platelet Factor 4/metabolism , Animals , Cell Movement , Humans , Immunity, Innate , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Factor 4/genetics , Viral Load
3.
Am J Physiol Lung Cell Mol Physiol ; 281(4): L913-21, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557595

ABSTRACT

The long-term disposition of circulating neutrophils and the site of disappearance from circulation remain unclear. We investigated neutrophil localization in mice using (111)In-labeled murine peripheral blood neutrophils, mature bone marrow neutrophils, and peritoneal exudate neutrophils to track in vivo localization of these different cell populations. Infused peripheral neutrophils were found to localize equally between liver and marrow sites by 4 h (31.2 +/- 1.9 vs. 31.9 +/- 1.8%), whereas exudate neutrophils predominantly localized to liver (42.0 +/- 1.1%) and marrow-derived neutrophils to the marrow (65.9 +/- 6.6%) where they were found to localize predominantly in the hematopoietic cords. Stimulation of marrow neutrophils before infusion caused a shift in localization from marrow to liver, and subsequent induction of an inflammatory site after infusion and marrow sequestration led to remobilization of infused marrow neutrophils but not of peripheral neutrophils. These results indicate that the marrow participates in removing neutrophils from circulation, with evidence supporting both storage and perhaps disposal functions. Furthermore, models for circulating neutrophil homeostasis should consider that the site of retention is governed by the maturation and activation states of the cell.


Subject(s)
Chemotaxis, Leukocyte/immunology , Neutrophils/cytology , Neutrophils/immunology , Actins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Chemotaxis, Leukocyte/drug effects , Exudates and Transudates/cytology , Exudates and Transudates/immunology , Indium Radioisotopes , Kinetics , Lipopolysaccharides/pharmacology , Liver/cytology , Liver/immunology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Superoxides/metabolism
4.
J Virol ; 75(7): 3383-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238864

ABSTRACT

Human cytomegalovirus (HCMV) evades healthy immune responses during infection, and this evasion may allow HCMV to establish latency in the host. The human vasculature has been recognized as a site of HCMV infection and may also be a site of latent HCMV infection. As the interface between circulating cells and underlying parenchymal cells, the vascular endothelium provides signals for local reaction of inflammatory cells. We propose that HCMV down-regulates expression of the proinflammatory chemokine RANTES from the infected endothelium, which may result in reduced recruitment of mononuclear cells to the site of infection. Abortive HCMV infection of primary endothelial cells with the clinical isolate HCMV 4010, under conditions in which viral gene expression could not occur, induced high levels of RANTES expression. Replicative HCMV infection, however, induced cells in parallel cultures to express significantly lower levels of RANTES. Expression of the chemokines interleukin 8 and MCP-1 by endothelial cells was found to be unaffected by replicative HCMV infection and thus may not play an important role during early HCMV infection of the endothelium. HCMV may regulate RANTES expression from endothelial cells as a mechanism to evade the local immune response to infection.


Subject(s)
Chemokine CCL5/biosynthesis , Cytomegalovirus/physiology , Endothelium, Vascular/metabolism , Chemokine CCL5/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Humans , RNA, Messenger/analysis , RNA, Messenger/metabolism , Virus Replication
5.
J Biol Chem ; 276(3): 2189-99, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11053415

ABSTRACT

The intensity and duration of an inflammatory response depends on the balance of factors that favor perpetuation versus resolution. At sites of inflammation, neutrophils adherent to other cells or matrix components are exposed to tumor necrosis factor-alpha (TNFalpha). Although TNFalpha has been implicated in induction of pro-inflammatory responses, it may also inhibit the intensity of neutrophilic inflammation by promoting apoptosis. Since TNFalpha is not only an important activator of the stress-induced pathways leading to p38 MAPk and c-Jun N-terminal kinase (JNK) but also a potent effector of apoptosis, we investigated the effects of TNFalpha on the JNK pathway in adherent human neutrophils and the potential involvement of this pathway in neutrophil apoptosis. Stimulation with TNFalpha was found to result in beta2 integrin-mediated activation of the cytoplasmic tyrosine kinases Pyk2 and Syk, and activation of a three-part MAPk module composed of MEKK1, MKK7, and/or MKK4 and JNK1. JNK activation was attenuated by blocking antibodies to beta2 integrins, the tyrosine kinase inhibitors, genistein, and tyrphostin A9, a Pyk2-specific inhibitor, and piceatannol, a Syk-specific inhibitor. Exposure of adherent neutrophils to TNFalpha led to the rapid onset of apoptosis that was demonstrated by augmented annexin V binding and caspase-3 cleavage. TNFalpha-induced increases in annexin V binding to neutrophils were attenuated by blocking antibodies to beta2 integrins, and the caspase-3 cleavage was attenuated by tyrphostin A9. Hence, exposure of adherent neutrophils to TNFalpha leads to utilization of the JNK-signaling pathways that may contribute to diverse functional responses including induction of apoptosis and subsequent resolution of the inflammatory response.


Subject(s)
CD18 Antigens/physiology , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Cell Adhesion , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Neutrophils/cytology
6.
J Immunol ; 164(4): 2151-9, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657669

ABSTRACT

Early inflammatory events include cytokine release, activation, and rapid accumulation of neutrophils, with subsequent recruitment of mononuclear cells. The p38 mitogen-activated protein kinase (MAPK) intracellular signaling pathway plays a central role in regulating a wide range of inflammatory responses in many different cells. A murine model of mild LPS-induced lung inflammation was developed to investigate the role of the p38 MAPK pathway in the initiation of pulmonary inflammation. A novel p38 MAPK inhibitor, M39, was used to determine the functional consequences of p38 MAPK activation. In vitro exposure to M39 inhibited p38 MAPK activity in LPS-stimulated murine and human neutrophils and macrophages, blocked TNF-alpha and macrophage inflammatory protein-2 (MIP-2) release, and eliminated migration of murine neutrophils toward the chemokines MIP-2 and KC. In contrast, alveolar macrophages required a 1000-fold greater concentration of M39 to block release of TNF-alpha and MIP-2. Systemic inhibition of p38 MAPK resulted in significant decreases in the release of TNF-alpha and neutrophil accumulation in the airspaces following intratracheal administration of LPS. Recovery of MIP-2 and KC from the airspaces was not affected by inhibition of p38 MAPK, and accumulation of mononuclear cells was not significantly reduced. When KC was instilled as a proinflammatory stimulus, neutrophil accumulation was significantly decreased by p38 MAPK inhibition independent of TNF-alpha or LPS. Together, these results demonstrate a much greater dependence on the p38 MAPK cascade in the neutrophil when compared with other leukocytes, and suggest a means of selectively studying and potentially modulating early inflammation in the lung.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Lung/enzymology , Lung/pathology , Mitogen-Activated Protein Kinases , Aminopyridines/pharmacology , Animals , Bronchoalveolar Lavage Fluid/immunology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Chemotaxis, Leukocyte/immunology , Cytokines/metabolism , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Female , Humans , Imidazoles/pharmacology , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Intubation, Intratracheal , Leukocyte Count , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/pathology , p38 Mitogen-Activated Protein Kinases
7.
Clin Immunol ; 92(3): 300-10, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479535

ABSTRACT

Wound healing is a complex process that involves inflammation, apoptosis, growth, and tissue remodeling. The autoimmune-prone inbred mouse strain MRL/+ manifests accelerated and extensive healing to ear punch wounds, suggesting a link between immune defects and wound healing. Prior studies with lupus-prone mice have shown that hematopoietic cells of lupus-prone strains can transfer disease to otherwise non-autoimmune-prone recipients. In this study we performed reciprocal bone marrow transfers between MRL and the control strain B10.BR and found that radioresistant MRL/+ host cells, rather than hematopoietic cells, are required for the healing response. We have also made the novel observations that, compared to normal controls, MRL/+ hematopoietic cells overproduce TGF-beta1 and manifest impaired inflammatory responses to lipopolysaccharide challenge. These features suggest that the aberrant wound healing phenotype of MRL mice is independent of their propensity to develop autoimmunity.


Subject(s)
Mice, Inbred MRL lpr/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/physiology , Aging/genetics , Aging/physiology , Animals , Bone Marrow Transplantation , Bronchoalveolar Lavage Fluid/cytology , Genotype , Hematopoietic Stem Cells/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Neutrophils/cytology , Pneumonia/physiopathology , Transplantation Chimera , Wound Healing/genetics
10.
J Clin Invest ; 103(6): 851-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079106

ABSTRACT

Activation of leukocytes by proinflammatory stimuli selectively initiates intracellular signal transduction via sequential phosphorylation of kinases. Lipopolysaccharide (LPS) stimulation of human neutrophils is known to result in activation of p38 mitogen-activated protein kinase (MAPk); however, the upstream activator(s) of p38 MAPk is unknown, and consequences of p38 MAPk activation remain largely undefined. We investigated the MAPk kinase (MKK) that activates p38 MAPk in response to LPS, the p38 MAPk isoforms that are activated as part of this pathway, and the functional responses affected by p38 MAPk activation. Although MKK3, MKK4, and MKK6 all activated p38 MAPk in experimental models, only MKK3 was found to activate recombinant p38 MAPk in LPS-treated neutrophils. Of p38 MAPk isoforms studied, only p38alpha and p38delta were detected in neutrophils. LPS stimulation selectively activated p38alpha. Specific inhibitors of p38alpha MAPk blocked LPS-induced adhesion, nuclear factor-kappa B (NF-kappaB) activation, and synthesis of tumor necrosis factor-alpha (TNF-alpha). Inhibition of p38alpha MAPk resulted in a transient decrease in TNF-alpha mRNA accumulation but persistent loss of TNF-alpha synthesis. These findings support a pathway by which LPS stimulation of neutrophils results in activation of MKK3, which in turn activates p38alpha MAPk, ultimately regulating adhesion, NF-kappaB activation, enhanced gene expression of TNF-alpha, and regulation of TNF-alpha synthesis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Neutrophils/drug effects , Cell Adhesion , Down-Regulation , Enzyme Activation/drug effects , Humans , Isoenzymes/metabolism , Lipopolysaccharide Receptors/metabolism , MAP Kinase Kinase 3 , Models, Biological , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases
11.
J Virol ; 72(7): 5535-44, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9621010

ABSTRACT

In patients with impaired cell-mediated immune responses (e.g., lung transplant recipients and AIDS patients), cytomegalovirus (CMV) infection causes severe disease such as pneumonitis. However, although immunocompetency in the host can protect from CMV disease, the virus persists by evading the host immune defenses. A model of CMV infection of the endothelium has been developed in which inflammatory stimuli, such as the CC chemokine RANTES, bind to the endothelial cell surface, stimulating calcium flux during late times of CMV infection. At 96 h postinfection, CMV-infected cells express mRNA of the CMV-encoded CC chemokine receptor US28 but do not express mRNA of other CC chemokine receptors that bind RANTES (CCR1, CCR4, CCR5). Cloning and stable expression of the receptor CMV US28 in human kidney epithelial cells (293 cells) with and without the heterotrimeric G protein alpha16 indicated that CMV US28 couples to both Galphai and Galpha16 proteins to activate calcium flux in response to the chemokines RANTES and MCP-3. Furthermore, cells that coexpress US28 and Galpha16 responded to RANTES stimulation with activation of extracellular signal-regulated kinase, which could be attributed, in part, to specific Galpha16 coupling. Thus, through expression of the CC chemokine receptor US28, CMV may utilize resident G proteins of the infected cell to manipulate cellular responses stimulated by chemokines.


Subject(s)
Cytomegalovirus/physiology , Receptors, Chemokine/physiology , Signal Transduction , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Chemokine CCL5/metabolism , GTP-Binding Proteins/physiology , Humans , RNA, Messenger/analysis , Receptors, CCR2
12.
J Biol Chem ; 273(14): 8389-97, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525949

ABSTRACT

Human neutrophils undergo apoptosis spontaneously when cultured in vitro; however, the signal transduction pathways involved remain largely unknown. In some cell types, c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase (MAPK) have been implicated in the pathways leading to stress-induced apoptosis. In this study, we begin to define two pathways leading to apoptosis in the neutrophil induced either by stress stimuli (UV, hyperosmolarity, sphingosine) or by anti-Fas antibody or overnight culture in vitro (spontaneous apoptosis). Apoptosis induced by stress stimuli activated p38 MAPK, and apoptosis was inhibited by the specific p38 MAPK inhibitor, 6-(4-Fluorophenyl)-2.3-dihydro-5-(4-puridinyl)imidazo(2, 1-beta)thiazole dihydrochloride. Furthermore, differentiation of HL-60 cells toward the neutrophil phenotype resulted in a loss in c-Jun NH2-terminal kinase activation with concomitant acquisition of formylmethionylleucylphenylalanine-stimulatable and stress-inducible p38 MAPK activity as well as apoptosis blockade by the p38 MAPK inhibitor. In contrast, anti-Fas-induced or spontaneous apoptosis occurred independent of p38 MAPK activation and was not blocked by the inhibitor. Both pathways appear to utilize member(s) of the caspase family, since pretreatment with either Val-Ala-Asp-fluoromethyl ketone or Asp-Glu-Val-Asp-fluoromethyl ketone inhibited apoptosis induced by each of the stimuli. We propose the presence of at least two pathways leading to apoptosis in human neutrophils, a stress-activated pathway that is dependent on p38 MAPK activation and an anti-FAS/spontaneous pathway that is p38 MAPK-independent.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Neutrophils/pathology , Signal Transduction , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Neutrophils/metabolism , Stress, Mechanical , Thiazoles/pharmacology , p38 Mitogen-Activated Protein Kinases
13.
Am J Respir Cell Mol Biol ; 18(1): 100-10, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9448051

ABSTRACT

Neutrophils and platelets interact both physically and metabolically during inflammation and thrombosis, but the mechanisms responsible for their adhesion remain incompletely understood. Neutrophil-platelet adhesion was measured after specific stimulation of neutrophils, platelets, or both and quantified by flow cytometry. Specific stimulation of either the neutrophil or the platelet led to a marked increase in the percentage of neutrophils that bound platelets, although platelet stimulation led to a large increase and neutrophil stimulation to only a small increase in the number of platelets per neutrophil. Stimulation of both cells further increased the number of neutrophil-platelet adhesive events and led to large numbers of platelets binding to each neutrophil. Confirming previous observations, blocking antibodies to platelet P-selectin (CD62P) partially inhibited adhesion. However, blockade of the neutrophil beta2 integrin CD11b/CD18 also inhibited the percentage of neutrophils that bound platelets. Combining P-selectin and CD11b/18 blockade further inhibited the stimulated increase in the percentage of neutrophils binding platelets and the increased number of platelets per neutrophil. Both cell adhesion molecules were active even when only a single cell type was primarily activated, supporting physiologically important transcellular activation. These data suggest that: (1) neutrophil-platelet adhesion can be initiated by specific activation of either the neutrophil or the platelet and that specific activation of either cell type leads to distinct patterns of adhesion, and (2) neutrophil-platelet adhesion uses both platelet P-selectin and the neutrophil beta2 integrin CD11b/CD18 when the cells are primarily or secondarily activated.


Subject(s)
Blood Platelets/physiology , CD18 Antigens/physiology , Cell Adhesion , Neutrophils/physiology , P-Selectin/physiology , Antibodies/pharmacology , Blood Platelets/ultrastructure , CD18 Antigens/immunology , Humans , Kinetics , Microscopy, Electron , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/ultrastructure , Oligopeptides/pharmacology , P-Selectin/immunology , Platelet Aggregation Inhibitors/pharmacology , Thrombin/pharmacology
14.
Proc Natl Acad Sci U S A ; 94(7): 3052-7, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9096344

ABSTRACT

Chemoattractants and chemokines, such as interleukin 8 (IL-8), are defined by their ability to induce directed cell migration of responsive cells. The signal transduction pathway(s) leading to cell migration remain ill defined. We demonstrate that phosphatidylinositol-3-kinase (PI3K) activity, as determined by inhibition using wortmannin and LY294002, is required for IL-8-induced cell migration of human neutrophils. Recently we reported that IL-8 caused the activation of the Ras/Raf/extracellular signal-regulated kinase (ERK) pathway in human neutrophils and that this activation was dependent on PI3K activity. The regulation of cell migration by IL-8 is independent of ERK kinase and ERK activation since the ERK kinase inhibitor PD098059 had no effect on IL-8-induced cell migration of human neutrophils. Additionally, activation of p38-mitogen-activated protein kinase is insufficient and activation of c-Jun N-terminal kinase is unnecessary to induce cell migration of human neutrophils. Therefore, regulation of neutrophil migration appears to be largely independent of the activation of the mitogen-activated protein kinases. The data argue that PI3K activity plays a central role in multiple signal transduction pathways within the human neutrophil leading to distinct cell functions.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Interleukin-8/pharmacology , Mitogen-Activated Protein Kinases , Neutrophils/cytology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Androstadienes/pharmacology , Calcium/metabolism , Cells, Cultured , Chemotaxis, Leukocyte , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1 , Phosphatidylinositol 3-Kinases , Wortmannin , p38 Mitogen-Activated Protein Kinases
15.
J Clin Invest ; 99(5): 975-86, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9062356

ABSTRACT

Stimulation of human neutrophils with chemoattractants FMLP or platelet activating factor (PAF) results in different but overlapping functional responses. We questioned whether these differences might reflect patterns of intracellular signal transduction. Stimulation with either PAF or FMLP resulted in equivalent phosphorylation and activation of the mitogen-activated protein kinase (MAPk) homologue 38-kD murine MAP kinase homologous to HOG-1 (p38) MAPk. Neither FMLP nor PAF activated c-jun NH2-terminal MAPk (JNKs). Under identical conditions, FMLP but not PAF, resulted in significant p42/44 (ERK) MAPk activation. Both FMLP and PAF activated MAP kinase kinase-3 (MKK3), a known activator of p38 MAPk. Both MAP ERK kinase kinase-1 (MEKK1) and Raf are activated strongly by FMLP, but minimally by PAF. Pertussis toxin blocked FMLP-induced activation of the p42/44 (ERK) MAPk cascade, but not that of p38 MAPk. A specific p38 MAPk inhibitor (SK&F 86002) blocked superoxide anion production in response to FMLP and reduced adhesion and chemotaxis in response to PAF or FMLP. These results demonstrate distinct patterns of intracellular signaling for two chemoattractants and suggest that selective activation of intracellular signaling cascades may underlie different patterns of functional responses.


Subject(s)
MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Platelet Activating Factor/pharmacology , Saccharomyces cerevisiae Proteins , Signal Transduction , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Calcium-Calmodulin-Dependent Protein Kinases/isolation & purification , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Adhesion/drug effects , Chemotaxis/drug effects , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase Kinases , Pertussis Toxin , Phosphorylation , Precipitin Tests , Protein Kinases/immunology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/isolation & purification , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/isolation & purification , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-raf , Recombinant Proteins/pharmacology , Superoxides/metabolism , Thiazoles/pharmacology , Time Factors , Virulence Factors, Bordetella/pharmacology
16.
Am J Physiol ; 272(3 Pt 1): L471-8, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9124604

ABSTRACT

Diminished ability of neonatal neutrophils to orient and move in a chemotactic gradient has been linked to compromised pulmonary host defense. We investigated whether deficiency of neonatal neutrophil function in vitro was evident in acute pulmonary inflammation. Analysis of neutrophils in vitro showed impaired chemotaxis in 4-wk-old compared with adult rabbits. In vivo-directed migration of labeled neutrophils into the alveolar space of adult rabbits in response to C5f instillation was significantly less for neutrophils donated from 4-wk-old rabbits compared with those from adults. In contrast, there were no differences in the alveolar accumulation of 4-wk-old and adult labeled neutrophils in 4-wk-old rabbits in response to C5f instillation, although the response showed a shorter time course than seen in adult rabbits. Adult rabbits diverted 46% of the blood away from the right cranial lung lobe, whereas 4-wk-old rabbits showed no change in blood flow after C5f instillation. Megakaryocytes (a source of blood flow mediators) were 3.2-fold greater in adult compared with 4-wk-old lung. These data suggest that the lack of blood flow diversion from inflamed neonatal lung increases neutrophil migration into alveoli, allowing for preservation of an inflammatory response despite neutrophil deficiencies in chemotaxis.


Subject(s)
Aging , Animals, Newborn/physiology , Lung/physiopathology , Neutrophils/physiology , Pneumonia/physiopathology , Acute Disease , Animals , Chemotaxis, Leukocyte/drug effects , Complement C5a/administration & dosage , Inflammation/physiopathology , Megakaryocytes/pathology , Microcirculation , Pulmonary Circulation , Rabbits
17.
J Biol Chem ; 271(52): 33598-606, 1996 Dec 27.
Article in English | MEDLINE | ID: mdl-8969228

ABSTRACT

Mechanisms of neutrophil activation in response to chemoattractants remain incompletely understood. We have recently reported a Ras-mediated c-Raf pathway leading to the activation of mitogen-activated protein (MAP) kinase in human neutrophils stimulated with the chemoattractant formyl-Met-Leu-Phe (FMLP). However, concern that Raf activation may not fully account for the early FMLP-mediated human neutrophil responses prompted us to investigate the activation of MAP kinase/ERK kinase (MEK) by MEK kinase (MEKK). In cell lysates we identified protein species at 180, 160, 110, 72, and 54 kDa with a monoclonal antibody to MEKK. Activation of MEKK was determined on immunoprecipitates from FMLP-stimulated neutrophils by in vitro kinase assay, which utilized both MEK1 and MEK2 as substrates. It was rapid, detectable at 30 s and reaching a plateau at 5 min, and it was inhibited in a dose-dependent fashion by a specific phosphatidylinositol 3-kinase inhibitor, wortmannin. Partial inhibition by pertussis toxin was observed. We were unable to show inhibition of the MEKK response by GF 109203X, a protein kinase C-specific inhibitor. These data indicate that in neutrophils activation of MEKK in addition to Raf may underlie stimulation of MAP kinase and other MAP kinase homologues by FMLP.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , MAP Kinase Kinase Kinase 1 , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/enzymology , Protein Serine-Threonine Kinases/metabolism , Androstadienes/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Indoles/pharmacology , Maleimides/pharmacology , Neutrophils/drug effects , Pertussis Toxin , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Recombinant Proteins/metabolism , Virulence Factors, Bordetella/pharmacology , Wortmannin
18.
J Immunol ; 156(12): 4867-75, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8648136

ABSTRACT

Stimulation of human neutrophils by LPS is central to the pathogenesis of sepsis and the adult respiratory distress syndrome. The intracellular signaling pathway that results in cellular responses following LPS stimulation in neutrophils is unknown. We report that exposure of neutrophils to LPS results in the phosphorylation and activation of a p38 mitogen-activated protein (MAP) kinase, occurring in a concentration-dependent manner, with maximum response at 20 to 25 min. Partial purification of a p38 MAP kinase by ion exchange chromatography established it as distinct from the p42/p44 (extracellular signal-regulated kinases (ERK-1 and ERK-2) MAP kinases). Activation of the p38 MAP kinase by LPS in human neutrophils occurs via CD14, a proposed LPS receptor, and requires the presence of plasma containing the LPS-binding protein. This intracellular signaling pathway is independent of protein kinase C and does not involve Raf, MAP/ERK kinase kinase-1, MAP/ERK kinase-1, or MAP/ERK kinase-2 and does not result in the activation of the p42/p44 ERK MAP kinases or the c-jun N-terminal kinases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Neutrophils/enzymology , Adult , Amino Acid Sequence , Cell Adhesion , Cells, Cultured , Enzyme Activation , Humans , Lipopolysaccharide Receptors/physiology , Molecular Weight , Peptides/chemistry , Phosphorylation , Phosphotyrosine/physiology , Protein Kinase C/physiology , Signal Transduction
19.
J Biol Chem ; 271(5): 2832-8, 1996 Feb 02.
Article in English | MEDLINE | ID: mdl-8576262

ABSTRACT

Interleukin-8 (IL-8), the prototypic member of the CXC subfamily of chemokines, induces in neutrophils chemotaxis, the respiratory burst, granule release, and increased cell adhesion. The IL-8 receptor is a seven-transmembrane spanning receptor coupled to specific heterotrimeric G proteins including Gi and G16. IL-8 stimulation of its receptor on neutrophils activates Ras GTP loading and the mitogen-activated protein kinase (MAPK) pathway including Raf-1 and B-Raf. The properties of IL-8 stimulation of the MAPK pathway differ from those observed for chemoattractants such as C5a. Even though Ras GTP loading is similar for IL-8 and C5a, the maximal activation of Raf-1 and B-Raf is approximately 2-fold and 3-7-fold, respectively, less for IL-8 than that observed for C5a. Raf-1 activation is rapid but transient, returning to near basal levels by 10 min. B-Raf activation is slower in onset and does not return to basal levels for nearly 30 min. IL-8 activation of MAPK follows a time course suggesting an involvement of both Raf-1 and B-Raf. Surprisingly, wortmannin, at low concentrations, inhibits Raf-1, B-Raf, and MAPK activation in response to IL-8 and C5a demonstrating a role for phosphatidylinositol 3-kinase in the activation of Raf kinases in G protein-coupled receptor systems in human neutrophils. Furthermore, wortmannin inhibits IL-8 stimulated granule release and neutrophil adherence. These findings demonstrate the control of Raf kinases, the MAPK pathway and specific neutrophil functions by phosphatidylinositol 3-kinase enzymes.


Subject(s)
Interleukin-8/metabolism , Neutrophils/enzymology , Protein Kinases/metabolism , Androstadienes/pharmacology , Cell Adhesion , Complement C5a/metabolism , Enzyme Activation , Guanosine Triphosphate/metabolism , Humans , Kinetics , Neutrophils/cytology , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors , Signal Transduction , Wortmannin , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...