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










Publication year range
1.
Commun Biol ; 4(1): 172, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558616

ABSTRACT

IL-36, which belongs to the IL-1 superfamily, is increasingly linked to neutrophilic inflammation. Here, we combined in vivo and in vitro approaches using primary mouse and human cells, as well as, acute and chronic mouse models of lung inflammation to provide mechanistic insight into the intercellular signaling pathways and mechanisms through which IL-36 promotes lung inflammation. IL-36 receptor deficient mice exposed to cigarette smoke or cigarette smoke and H1N1 influenza virus had attenuated lung inflammation compared with wild-type controls. We identified neutrophils as a source of IL-36 and show that IL-36 is a key upstream amplifier of lung inflammation by promoting activation of neutrophils, macrophages and fibroblasts through cooperation with GM-CSF and the viral mimic poly(I:C). Our data implicate IL-36, independent of other IL-1 family members, as a key upstream amplifier of neutrophilic lung inflammation, providing a rationale for targeting IL-36 to improve treatment of a variety of neutrophilic lung diseases.


Subject(s)
Interleukin-1/metabolism , Lung/metabolism , Neutrophil Activation , Neutrophils/metabolism , Orthomyxoviridae Infections/metabolism , Pneumonia, Viral/metabolism , Receptors, Interleukin-1/metabolism , Animals , Cells, Cultured , Cigarette Smoking , Disease Models, Animal , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-1/genetics , Lung/immunology , Lung/virology , Macrophage Activation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/virology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Receptors, Interleukin-1/genetics , Signal Transduction
2.
Immunology ; 143(3): 416-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24831554

ABSTRACT

Interleukin-4 (IL-4) and IL-13 are critical drivers of immune activation and inflammation in ulcerative colitis, asthma and other diseases. Because these cytokines may have redundant function, dual targeting holds promise for achieving greater efficacy. We have recently described a bifunctional therapeutic targeting IL-4 and IL-13 developed on a novel protein scaffold, generated by combining specific binding domains in an optimal configuration using appropriate linker regions. In the current study, the bifunctional IL-4/IL-13 antagonist was evaluated in the murine oxazolone-induced colitis model, which produces disease with features of ulcerative colitis. The bifunctional IL-4/IL-13 antagonist reduced body weight loss throughout the 7-day course of the model, and ameliorated the increased colon weight and decreased colon length that accompany disease. Colon tissue gene expression was modulated in accordance with the treatment effect. Concentrations of serum amyloid P were elevated in proportion to disease severity, making it an effective biomarker. Serum concentrations of the bifunctional IL-4/IL-13 antagonist were inversely proportional to disease severity, colon tissue expression of pro-inflammatory genes, and serum amyloid P concentration. Taken together, these results define a panel of biomarkers signifying engagement of the IL-4/IL-13 pathway, confirm the T helper type 2 nature of disease in this model, and demonstrate the effectiveness of dual cytokine blockade.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colitis, Ulcerative/metabolism , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Mice , Oxazolone/adverse effects , Recombinant Fusion Proteins/administration & dosage , Serum Amyloid A Protein/metabolism , Serum Amyloid P-Component/metabolism , Severity of Illness Index
3.
J Cell Biochem ; 115(9): 1539-48, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24633904

ABSTRACT

Fibrosis is a chronic disease characterized by an excessive deposition of scar tissue in the affected organs. A central mediator of this process is transforming growth factor-ß (TGF-ß), which stimulates the production of extracellular matrix proteins such as collagens. MicroRNAs (miRNAs) have been implicated in both fibrosis as well as in TGF-ß signaling, but the extent of their regulation has not been fully defined. A functional screen was conducted using a library of miRNA inhibitors to identify miRNAs that affect TGF-ß-induced type I collagen expression, a key event in the development of fibrosis. The inhibition of one miRNA in particular, miR-27b, caused a significant increase in type I collagen expression. We found that miR-27b directly targets Gremlin 1 by binding to its 3'-UTR, reducing its mRNA levels. TGF-ß signaling decreased miR-27b expression and caused a corresponding increase in Gremlin 1 levels, suggesting that TGF-ß regulates Gremlin 1 expression in part by modulating miR-27b expression. Reducing Gremlin 1 levels by either siRNA-mediated gene silencing or by using the miR-27b mimic inhibited the expression of several genes known to be involved in fibrosis, while increasing Gremlin 1 levels by the addition of either recombinant protein or the miR-27b inhibitor enhanced the expression of these genes. In summary, we have demonstrated that miR-27b targets Gremlin 1, and that this regulation likely represents an important control point in fibrotic pathways.


Subject(s)
Epithelial Cells/cytology , Fibroblasts/cytology , Intercellular Signaling Peptides and Proteins/genetics , Lung/cytology , MicroRNAs/genetics , 3' Untranslated Regions , Cell Line , Cell Proliferation , Collagen Type I/metabolism , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology
4.
Clin Sci (Lond) ; 125(10): 483-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23738811

ABSTRACT

Increased systemic and pulmonary levels of IL-6 (interleukin-6) are associated with the severity of exacerbations and decline of lung function in patients with COPD (chronic obstructive pulmonary disease). Whether IL-6 is directly involved or plays a bystander role in the pathophysiology of COPD remains unclear. Here we hypothesized that neutralizing circulating levels of IL-6 would modulate episodes of acute pulmonary inflammation following CS (cigarette smoke) exposure and virus-like challenges. For this purpose, we used a model where C57BL/6 mice were exposed to CS twice daily via a nose-only system, and concomitant periodic intranasal challenge with poly(I:C), a synthetic ligand for TLR3 (Toll-like receptor 3) that mimics the encounter with double stranded RNA that is carried by influenza-like viruses. This protocol recapitulates several aspects of acute pulmonary inflammation associated with COPD, including prominent airway neutrophilia, insensitivity to steroid treatment and increased levels of several inflammatory cytokines in BAL (bronchoalveolar lavage) samples. Although IL-6-deficient mice exposed to CS/poly(I:C) developed pulmonary inflammation similar to WT (wild-type) controls, WT mice exposed to CS/poly(I:C) and treated intraperitoneally with IL-6-neutralizing antibodies showed significantly lower blood counts of lymphocytes and monocytes, lower BAL levels of IL-6 and CXCL1 (CXC chemokine ligand 1)/KC (keratinocyte chemoattractant), as well as reduced numbers of BAL neutrophils, lymphocytes and macrophages. Our results thus indicate that the systemic neutralization of IL-6 significantly reduces CS/poly(I:C)-induced pulmonary inflammation, which may be a relevant approach to the treatment of episodes of acute pulmonary inflammation associated with COPD.


Subject(s)
Interleukin-6/antagonists & inhibitors , Pneumonia/prevention & control , Poly I-C/toxicity , Pulmonary Disease, Chronic Obstructive/prevention & control , Tobacco Smoke Pollution/adverse effects , Acute Disease , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cytokines/biosynthesis , Dexamethasone/therapeutic use , Drug Resistance/physiology , Female , Glucocorticoids/therapeutic use , Inflammation Mediators/metabolism , Interleukin-6/deficiency , Interleukin-6/physiology , Leukocyte Count , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pneumonia/etiology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology
5.
Am J Respir Cell Mol Biol ; 49(1): 37-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23449738

ABSTRACT

IL-4 and IL-13 comprise promising targets for therapeutic interventions in asthma and other Th2-associated diseases, but agents targeting either IL-4 or IL-13 alone have shown limited efficacy in human clinical studies. Because these cytokines may involve redundant function, dual targeting holds promise for achieving greater efficacy. We describe a bifunctional therapeutic targeting IL-4 and IL-13, developed by a combination of specific binding domains. IL-4-targeted and IL-13-targeted single chain variable fragments were joined in an optimal configuration, using appropriate linker regions on a novel protein scaffold. The bifunctional IL-4/IL-13 antagonist displayed high affinity for both cytokines. It was a potent and efficient neutralizer of both murine IL-4 and murine IL-13 bioactivity in cytokine-responsive Ba/F3 cells, and exhibited a half-life of approximately 4.7 days in mice. In a murine model of ovalbumin-induced ear swelling, the bifunctional molecule blocked both the IL-4/IL-13-dependent early-phase response and the IL-4-dependent late-phase response. In the ovalbumin-induced lung inflammation model, the bifunctional IL-4/IL-13 antagonist reduced the IL-4-dependent rise in serum IgE titers, and reduced IL-13-dependent airway hyperresponsiveness, lung inflammation, mucin gene expression, and serum chitinase responses. Taken together, these findings demonstrate the effective dual blockade of IL-4 and IL-13 with a single agent, which resulted in the modulation of a more extensive range of endpoints than could be achieved by targeting either cytokine alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunoglobulin E/immunology , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Pneumonia/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/immunology , Binding Sites , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , CHO Cells , Cricetinae , Cross-Linking Reagents/chemistry , Ear/physiopathology , Female , Half-Life , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Mice , Mice, Inbred C57BL , Molecular Conformation , Neutralization Tests , Ovalbumin/adverse effects , Ovalbumin/immunology , Pneumonia/immunology , Protein Binding , Protein Interaction Domains and Motifs , Single-Chain Antibodies/metabolism
6.
Pulm Pharmacol Ther ; 25(4): 286-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22613758

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a debilitating condition resulting from exposure to pollutants such as cigarette smoke. Pulmonary macrophages secrete a plethora of inflammatory mediators that are increased in the lungs of COPD patients, but whether this phenotype results directly from smoke exposure remains unknown. Using an in vitro model for alveolar macrophages (AM) derived from human peripheral blood monocytes with granulocyte-macrophage stimulating factor (GM-MØ), we analyzed the mechanistic connection between cigarette smoke exposure and histone deacetylase (HDAC) regulation, hypothesized to be a contributing factor in COPD pathophysiology. Here we show that acute smoke exposure inhibits HDAC enzymatic activity in GM-MØ. Analysis of mRNA and total cellular proteins for expression of class I (1, 2, 3 and 8), class II (4, 5, 6, 7, 9, 10), and class IV (11) HDAC revealed no effect of smoke exposure, whereas nuclear HDAC3 protein content was reduced. To better understand the physiological significance of reduced HDAC3 activity, we utilized siRNA to knockdown HDAC1, 2 and 3 individually. Interestingly, siRNA-mediated reduction of HDAC3 resulted in increased production of IL8 and IL1ß in response to LPS stimulation, while HDAC2 knockdown had no effect on either cytokine. Lower nuclear content of HDAC3 in the context of equivalent total HDAC protein levels following smoke exposure may reflect increased nuclear export of HDAC3, allowing increased nuclear factor kappa b (NF-κB ) driven cytokine expression that can contribute to inflammation.


Subject(s)
Cytokines/metabolism , Hazardous Substances/adverse effects , Histone Deacetylases/metabolism , Macrophages, Alveolar/metabolism , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/immunology , Tobacco Smoke Pollution/adverse effects , Down-Regulation/immunology , Enzyme Repression/immunology , Hazardous Substances/toxicity , Histone Deacetylase 2/metabolism , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Lung/physiopathology , Macrophages, Alveolar/drug effects , Time Factors
8.
Toxicol Pathol ; 40(2): 205-15, 2012.
Article in English | MEDLINE | ID: mdl-22301949

ABSTRACT

Cytokines are critical in allergic intercellular communication networks, and they contribute to disease pathology through the recruitment and activation of pro-inflammatory leukocytes and in chronic disease to pro-fibrotic/remodeling events. Th2 cytokines predominate primarily in mild to moderate allergic asthma, although clinical trials with inhibitors of IL-4 and IL-5 have not provided the robust efficacy observed in animal models of allergy. These results not only highlight the complexity of allergic disease, but they also point to the importance of other cytokine networks in driving pathology. The heterogeneous nature of the disease is emphasized by the fact that the Th2/Th1/Th17 cytokine balance can be influenced by the initiating allergic trigger. For example, the house dust mite allergen Der p 2 mimics the activity of MD-2 by presenting lipopolysaccharide to Toll-like receptor-4 for the activation of inflammatory genes including innate-type cytokines. Here we discuss the functions of the novel cytokine players, thymic stromal lymphopoetin (TSLP), IL-33, IL-25, and IL-9 and delineate nonredundant roles for IL-4 and IL-13 in allergic disease. Persistent efforts in the characterization of these and other cytokine networks will be essential for understanding the complex pathogenic mechanisms that underpin allergic disease and for guiding targeted therapeutic interventions.


Subject(s)
Cytokines/immunology , Hypersensitivity/immunology , Signal Transduction/immunology , Humans
9.
Am J Respir Cell Mol Biol ; 46(1): 71-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21836154

ABSTRACT

The expression of acidic mammalian chitinase (AMCase) is associated with Th2-driven respiratory disorders. To investigate the potentially pathological role of AMCase in allergic airway disease (AAD), we sensitized and challenged mice with ovalbumin or a combination of house dust mite (HDM) plus cockroach allergen. These mice were treated or not treated with small molecule inhibitors of AMCase, which significantly reduced allergen-induced chitinolytic activity in the airways, but exerted no apparent effect on pulmonary inflammation per se. Transgenic and AMCase-deficient mice were also submitted to protocols of allergen sensitization and challenge, yet we found little or no difference in the pattern of AAD between mutant mice and wild-type (WT) control mice. In a separate model, where mice were challenged only with intratracheal instillations of HDM without adjuvant, total bronchoalveolar lavage (BAL) cellularity, inflammatory infiltrates in lung tissues, and lung mechanics remained comparable between AMCase-deficient mice and WT control mice. However BAL neutrophil and lymphocyte counts were significantly increased in AMCase-deficient mice, whereas concentrations in BAL of IL-13 were significantly decreased compared with WT control mice. These results indicate that, although exposure to allergen stimulates the expression of AMCase and increased chitinolytic activity in murine airways, the overexpression or inhibition of AMCase exerts only a subtle impact on AAD. Conversely, the increased numbers of neutrophils and lymphocytes in BAL and the decreased concentrations of IL-13 in AMCase-deficient mice challenged intratracheally with HDM indicate that AMCase contributes to the Th1/Th2 balance in the lungs. This finding may be of particular relevance to patients with asthma and increased airway neutrophilia.


Subject(s)
Asthma/enzymology , Chitinases/antagonists & inhibitors , Hypersensitivity/enzymology , Allergens/immunology , Animals , Asthma/genetics , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Chitinases/deficiency , Chitinases/genetics , Chitinases/immunology , Female , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Interleukin-13/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neutrophils/immunology , Th1 Cells/immunology , Th2 Cells/immunology
10.
J Pharmacol Exp Ther ; 340(3): 656-65, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22160268

ABSTRACT

Asthma is a chronic inflammatory lung disease with considerable unmet medical needs for new and effective therapies. Cytosolic phospholipase A(2)α (cPLA(2)α) is the rate-limiting enzyme that is ultimately responsible for the production of eicosanoids implicated in the pathogenesis of asthma. We investigated a novel cPLA(2)α inhibitor, PF-5212372, to establish the potential of this drug as a treatment for asthma. PF-5212372 was a potent inhibitor of cPLA(2)α (7 nM) and was able to inhibit prostaglandin (PG)D(2) and cysteinyl leukotriene release from anti-IgE-stimulated human lung mast cells (0.29 and 0.45 nM, respectively). In a mixed human lung cell population, PF-5212372 was able to inhibit ionomycin-stimulated release of leukotriene B(4), thromboxane A(2), and PGD(2) (2.6, 2.6, and 4.0 nM, respectively) but was significantly less effective against PGE(2) release (>301 nM; p < 0.05). In an in vitro cell retention assay, PF-5212372 retained its potency up to 24 h after being washed off. In a sheep model of allergic inflammation, inhalation of PF-5212372 significantly inhibited late-phase bronchoconstriction (78% inhibition; p < 0.001) and airway hyper-responsiveness (94% inhibition; p < 0.001), and isolated sheep lung mast cell assays confirmed species translation via effective inhibition of PGD(2) release (0.78 nM). Finally, PF-5212372 was assessed for its ability to inhibit the contraction of human bronchi induced by AMP. PF5212372 significantly inhibited AMP-induced contraction of human bronchi (81% inhibition; p < 0.001); this finding, together with the ability of this drug to be effective in a wide range of preclinical asthma models, suggests that inhibition of cPLA(2)α with PF-5212372 may represent a new therapeutic option for the treatment of asthma.


Subject(s)
Asthma/drug therapy , Cytosol/enzymology , Enzyme Inhibitors/therapeutic use , Group IV Phospholipases A2/antagonists & inhibitors , Phenylpropionates/pharmacology , Sulfonamides/pharmacology , Animals , Antibodies, Anti-Idiotypic/pharmacology , Bronchoconstriction/drug effects , Calcium Ionophores/pharmacology , Cell Line , Humans , Mast Cells/physiology , Prostaglandin D2/metabolism , Sheep
11.
Inflamm Allergy Drug Targets ; 10(6): 486-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21864272

ABSTRACT

Atopic dermatitis (AD) is a pruritic chronic inflammatory disease of the skin that is triggered by an underlying complicated interplay between the genetics of the individual and stimulation by allergens. Patients with AD demonstrate compromised barrier function that leads to activation of keratinocytes and immune cells which favor a strong Th2 bias. As a result of this immunological bias such patients also suffer from secondary pathogenic infections. A wide array of cytokines and chemokines interact to yield symptoms characteristic of AD. In addition, the involvement of different immunological cell types compounds our difficulty in understanding its immunopathogenesis. The use of various mouse models and transgenics has allowed us to intricately examine the functioning of the various molecules identified to play a role in AD. Such mouse models have also aided in the testing and development of various therapeutics for AD. This review is focused on examining the various factors contributing to the pathogenesis and exacerbation of AD as well as current treatments for AD. There is scope for improving the therapy of AD patients and thereby allowing them a better quality of life.


Subject(s)
Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Administration, Topical , Animals , Chemokines/immunology , Chronic Disease , Cytokines/immunology , Dermatitis, Atopic/drug therapy , Dermatologic Agents/administration & dosage , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology
12.
J Med Chem ; 53(16): 6122-8, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20666458

ABSTRACT

Acidic mammalian chitinase (AMCase) is a member of the glycosyl hydrolase 18 family (EC 3.2.1.14) that has been implicated in the pathophysiology of allergic airway disease such as asthma. Small molecule inhibitors of AMCase were identified using a combination of high-throughput screening, fragment screening, and virtual screening techniques and characterized by enzyme inhibition and NMR and Biacore binding experiments. X-ray structures of the inhibitors in complex with AMCase revealed that the larger more potent HTS hits, e.g. 5-(4-(2-(4-bromophenoxy)ethyl)piperazine-1-yl)-1H-1,2,4-triazol-3-amine 1, spanned from the active site pocket to a hydrophobic pocket. Smaller fragments identified by FBS occupy both these pockets independently and suggest potential strategies for linking fragments. Compound 1 is a 200 nM AMCase inhibitor which reduced AMCase enzymatic activity in the bronchoalveolar lavage fluid in allergen-challenged mice after oral dosing.


Subject(s)
Chitinases/antagonists & inhibitors , Models, Molecular , Piperazines/chemical synthesis , Triazoles/chemical synthesis , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid , Catalytic Domain , Crystallography, X-Ray , Female , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Piperazines/chemistry , Piperazines/pharmacology , Protein Binding , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Structure-Activity Relationship , Surface Plasmon Resonance , Triazoles/chemistry , Triazoles/pharmacology
13.
Exp Lung Res ; 34(9): 599-629, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19005923

ABSTRACT

Pulmonary macrophages (MØs) are essential for clearance of inhaled particles, innate immunity, and lung tissue maintenance. However, the products of activated MØs have also been implicated in inflammation and tissue destruction, including in chronic obstructive pulmonary disease (COPD). Primary human alveolar macrophages (AMs) are available in limited numbers via bronchoalveolar lavage (BAL) or sputum induction, and BAL macrophages are not commonly available to all researchers. A readily available, plentiful, but representative surrogate for AMs would advance understanding of the contribution of macrophages to lung pathophysiology. Herein the authors describe a method for the in vitro derivation of AM-like cells using primary human peripheral blood monocytes differentiated in suspension with granulocyte-macrophage colony-stimulating factor (GM-CSF). The method produces a cell population with a consistent and stable phenotype. Flow cytometry reveals that GM-CSF-derived macrophages (GM-MØs) express lineage markers, immunoglobulin gamma (Fc gamma) receptors, adhesion molecules, antigen presentation coreceptors, and scavenger receptors akin to AMs. Functionally, cigarette smoke activates extracellular signal-related kinase (ERK) and p38 mitogen-activated protein (MAP) kinase, enhances interleukin 8 (IL8) production from GM-MØs and inhibits phagocytosis, phenotypes previously described for smokers' AMs. Global transcriptional profiling revealed significant overlap in regulated genes between smokers' AMs and GM-MØs treated with cigarette smoke preparations in vitro.


Subject(s)
Macrophages, Alveolar/metabolism , Smoke/adverse effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Inflammation/etiology , Interleukin-8/biosynthesis , Models, Biological , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Immunol ; 179(11): 7791-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025225

ABSTRACT

IL-17A and IL-17F are related homodimeric proteins of the IL-17 family produced by Th17 cells. In this study, we show that mouse Th17 cells also produce an IL-17F/A heterodimeric protein. Whereas naive CD4(+) T cells differentiating toward the Th17 cell lineage expressed IL-17F/A in higher amounts than IL-17A/A homodimer and in lower amounts than IL-17F/F homodimer, differentiated Th17 cells expressed IL-17F/A in higher amounts than either homodimer. In vitro, IL-17F/A was more potent than IL-17F/F and less potent than IL-17A/A in regulating CXCL1 expression. Neutralization of IL-17F/A with an IL-17A-specific Ab, and not with an IL-17F-specific Ab, reduced the majority of IL-17F/A-induced CXCL1 expression. To study these cytokines in vivo, we established a Th17 cell adoptive transfer model characterized by increased neutrophilia in the airways. An IL-17A-specific Ab completely prevented Th17 cell-induced neutrophilia and CXCL5 expression, whereas Abs specific for IL-17F or IL-22, a cytokine also produced by Th17 cells, had no effects. Direct administration of mouse IL-17A/A or IL-17F/A, and not IL-17F/F or IL-22, into the airways significantly increased neutrophil and chemokine expression. Taken together, our data elucidate the regulation of IL-17F/A heterodimer expression by Th17 cells and demonstrate an in vivo function for this cytokine in airway neutrophilia.


Subject(s)
Interleukin-17/biosynthesis , Lung/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer/methods , Animals , Dimerization , Disease Models, Animal , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic
15.
Ann Allergy Asthma Immunol ; 98(1): 83-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17225725

ABSTRACT

BACKGROUND: Several studies indicate that selectin-mediated leukocyte migration may depend on the types of initiating inflammatory stimuli or on the vascular beds involved in the inflammatory response. Thus, targeting selectin interactions to treat inflammation may have variable effects depending on the site and origin of the inflammatory response. OBJECTIVE: To address whether selectin-mediated leukocyte recruitment is stimulus or tissue dependent. METHODS: We examined pulmonary and cutaneous allergic inflammatory responses and silica-induced nonallergic lung inflammation and fibrosis in wild-type and P- and E-selectin-deficient (P/E-/-) double knockout mice. Allergen-sensitized wild-type and P/E-/- double knockout mice were challenged either intradermally or via the airways to induce allergic responses in the skin or lung, respectively. Other animals were subjected to intranasal silica administration to induce a nonallergic lung inflammatory/fibrotic response. RESULTS: The P/E-/- mice exhibited significantly reduced allergic inflammation in the skin and lung. Allergic late-phase ear swelling and allergic lung airway hyperresponsiveness were also significantly attenuated in the P/E-/- mice compared with identically treated wild-type animals. In contrast, pulmonary inflammation and fibrosis induced by intranasal administration of silica particles resulted in a more severe phenotype in the P/E-/- mice. CONCLUSIONS: Selectin interactions drive allergic inflammation in the lung and skin. Silica-induced pulmonary inflammation and fibrosis, however, was more pronounced in the absence of selectin interactions, suggesting that selectin-mediated leukocyte migration may depend on the types of initiating inflammatory stimuli.


Subject(s)
E-Selectin/metabolism , Endothelium, Vascular/metabolism , Hypersensitivity/immunology , Inflammation/metabolism , P-Selectin/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Cytokines/metabolism , E-Selectin/immunology , Endothelium, Vascular/immunology , Hypersensitivity/metabolism , Inflammation/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Ovalbumin/toxicity , P-Selectin/metabolism , Silicon Dioxide/toxicity , Skin/immunology
16.
Am J Respir Cell Mol Biol ; 35(3): 357-65, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16645179

ABSTRACT

Gob-5 is a member of the calcium-activated chloride channel family and has been associated with allergic response in mouse models of pulmonary inflammation. Gene expression of Gob-5 has been shown to be induced in allergic airways and has been strongly associated with mucin gene regulation and goblet cell hyperplasia. We investigated the physiologic role of Gob-5 in murine models of pulmonary inflammation using mice deficient in Gob-5. After sensitization and aerosol challenge with ovalbumin (OVA), Gob-5 knockout mice exhibit significantly increased bronchoalveolar lavage (BAL) inflammation as compared with wild-type controls. The augmented inflammation in BAL consisted predominantly of neutrophils. Examination of perivascular inflammation revealed that tissue inflammation was decreased in OVA-challenged Gob-5-/- mice. OVA-challenged Gob-5 knockout mice also had decreased goblet cell hyperplasia as well as decreased mucus production. These mice also had decreased airway hypersensitivity after cholinergic provocation with methacholine. Gob-5 knockout mice were also challenged via intranasal LPS, a TLR-4 agonist. Gob-5-/- mice responded with increased neutrophilic BAL inflammation and decreased perivascular tissue inflammation as compared with wild-type controls. There was little effect on goblet cell hyperplasia and mucus production after LPS challenge. These observations reinforce findings that associate Gob-5 with goblet cell hyperplasia and mucus production in the allergic immune response, but also implicate Gob-5 in the regulation of tissue inflammation in the innate immune response.


Subject(s)
Chloride Channels/physiology , Goblet Cells/pathology , Mucoproteins/physiology , Pneumonia/genetics , Pneumonia/pathology , Airway Resistance , Animals , Antigens/toxicity , Bronchoalveolar Lavage Fluid/cytology , Chemokines/metabolism , Chloride Channels/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Goblet Cells/drug effects , Goblet Cells/metabolism , Hyperplasia/genetics , Hyperplasia/pathology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Mucoproteins/genetics , Mucus/metabolism , Ovalbumin/toxicity , Pneumonia/chemically induced
17.
J Immunol ; 174(12): 7716-24, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15944273

ABSTRACT

IL-4 and IL-13 are potent cytokines that drive production of IgE, which is critical to the development of atopic disease. In this study, we directly compared IgE generation and IgE-dependent mast cell effector function in mouse strains lacking IL-4, IL-13, IL-4 + IL-13, or their common receptor component, IL-4Ralpha. Although serum IgE was undetectable under resting conditions in most animals deficient in one or both cytokines, peritoneal mast cells from mice lacking IL-4 or IL-13 had only partial reductions in surface IgE level. In contrast, peritoneal mast cells from IL-4/13(-/-) and IL-4Ralpha(-/-) animals were severely deficient in surface IgE, and showed no detectable degranulation following treatment with anti-IgE in vitro. Surprisingly, however, intradermal challenge with high concentrations of anti-IgE Ab induced an ear-swelling response in these strains, implying some capacity for IgE-mediated effector function in tissue mast cells. Furthermore, upon specific immunization with OVA, both IL-4/IL-13(-/-) and IL-4Ralpha(-/-) mice produced detectable levels of serum IgE and Ag-specific IgG1, and generated strong ear-swelling responses to intradermal administration of anti-IgE. These findings suggest that a mechanism for IgE production exists in vivo that is independent of IL-4 or IL-13.


Subject(s)
Immunoglobulin E/biosynthesis , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-4/deficiency , Interleukin-4/genetics , Mast Cells/immunology , Mast Cells/metabolism , Animals , Binding Sites, Antibody/genetics , Cell Count , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Separation , Dose-Response Relationship, Immunologic , Immunization, Secondary , Immunoglobulin E/blood , Immunoglobulin E/deficiency , Immunoglobulin E/physiology , Interleukin-13/metabolism , Interleukin-13/physiology , Interleukin-4/metabolism , Interleukin-4/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Passive Cutaneous Anaphylaxis , Peritoneal Cavity/cytology , Protein Binding/genetics , Protein Binding/immunology , Protein Subunits/deficiency , Protein Subunits/genetics , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/genetics , Up-Regulation/genetics , Up-Regulation/immunology
18.
Annu Rev Immunol ; 23: 749-86, 2005.
Article in English | MEDLINE | ID: mdl-15771585

ABSTRACT

This review focuses on recent progress in our understanding of how mast cells can contribute to the initiation, development, expression, and regulation of acquired immune responses, both those associated with IgE and those that are apparently expressed independently of this class of Ig. We emphasize findings derived from in vivo studies in mice, particularly those employing genetic approaches to influence mast cell numbers and/or to alter or delete components of pathways that can regulate mast cell development, signaling, or function. We advance the hypothesis that mast cells not only can function as proinflammatory effector cells and drivers of tissue remodeling in established acquired immune responses, but also may contribute to the initiation and regulation of such responses. That is, we propose that mast cells can also function as immunoregulatory cells. Finally, we show that the notion that mast cells have primarily two functional configurations, off (or resting) or on (or activated for extensive mediator release), markedly oversimplifies reality. Instead, we propose that mast cells are "tunable," by both genetic and environmental factors, such that, depending on the circumstances, the cell can be positioned phenotypically to express a wide spectrum of variation in the types, kinetics, and/or magnitude of its secretory functions.


Subject(s)
Mast Cells/immunology , Animals , Asthma/immunology , Basophils/immunology , Dermatitis, Contact/immunology , Humans , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , Mice , Mice, Mutant Strains , Models, Immunological , Receptors, IgE/metabolism , Signal Transduction , T-Lymphocytes/immunology
19.
J Allergy Clin Immunol ; 115(2): 309-15, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15696086

ABSTRACT

BACKGROUND: T(H)2-mediated allergic asthma is characterized by eosinophilia, mucus overproduction, and airway hyperresponsiveness (AHR). Although it is clear that T(H)2 cells and their cytokines play an important role in AHR, the roles of T(H)1 cells and neutrophils in AHR are controversial. OBJECTIVE: We sought to determine the roles of T(H)1 cells and neutrophils in AHR. METHODS: Ovalbumin-specific CD4(+) T cells were purified from DO11.10 mice, differentiated into T(H)1 cells, and injected into naive BALB/c, IL-4RalphaKO, or IL-8RKO mice. After ovalbumin antigen challenge, cytokine mRNA levels in lung samples, as well as inflammatory cell types and numbers in bronchoalveolar lavage fluid (BALF), were determined. AHR was assessed by measuring resistance in tracheostomized mice and enhanced pause in freely moving mice. RESULTS: T(H)1 cells induced AHR as robust as T(H)2 cells. They also induced lung inflammation dominated by neutrophils. Neither AHR nor inflammation were reduced when T(H)1 cells were transferred into IL-4RalphaKO mice. When IL-8RKO mice were used as recipients of T(H)1 cells, neutrophilia was greatly reduced, but the AHR was as strong as that seen in wild-type mice. On the other hand, dexamethasone treatment had no effect on neutrophilia but has significantly reduced AHR. Reduction in AHR was accompanied by a reduction in the numbers of lymphocytes and macrophages in BALF. CONCLUSIONS: T(H)1 cells can induce strong AHR independent of IL-4 and IL-13. The AHR is associated with the presence of lymphocytes and macrophages, but not neutrophils, in BALF. Our results point to a pathway whereby T(H)1 cells mediate AHR independent of neutrophilic inflammation.


Subject(s)
Bronchial Hyperreactivity/physiopathology , Bronchitis/pathology , Neutrophils/pathology , Th1 Cells/immunology , Adoptive Transfer , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Cytokines/genetics , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Interleukin-13/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucus/metabolism , Ovalbumin/pharmacology , RNA, Messenger/metabolism , Receptors, Interleukin-8A/deficiency , Severity of Illness Index , Th2 Cells/immunology
20.
J Immunol ; 171(1): 331-7, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12817015

ABSTRACT

Adenosine-induced bronchoconstriction is a well-recognized feature of atopic asthma. Adenosine acts through four different G protein-coupled receptors to produce a myriad of physiological effects. To examine the contribution of the A(3) adenosine receptor to adenosine-induced bronchoconstriction and to assess the contribution of mast cells to this process, we quantified airway responsiveness to aerosolized adenosine in wild-type, A(3) receptor-deficient, and mast cell-deficient mice. Compared with the robust airway responses elicited by adenosine in wild-type mice, both A(3)-deficient and mast cell-deficient mice exhibited a significantly attenuated response compared with their respective wild-type controls. Histological examination of the airways 4 h after adenosine exposure revealed extensive degranulation of airway mast cells as well as infiltration of neutrophils in wild-type mice, whereas these findings were much diminished in A(3)-deficient mice and were not different from those in PBS-treated controls. These data indicate that the airway responses to aerosolized adenosine in mice occur largely through A(3) receptor activation and that mast cells contribute significantly to these responses, but that activation of additional adenosine receptors on a cell type(s) other than mast cells also contributes to adenosine-induced airway responsiveness in mice. Finally, our findings indicate that adenosine exposure can result in A(3)-dependent airway inflammation, as reflected in neutrophil recruitment, as well as alterations in airway function.


Subject(s)
Adenosine/physiology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Mast Cells/physiology , Receptors, Purinergic P1/physiology , Adenosine/administration & dosage , Administration, Inhalation , Aerosols , Animals , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/genetics , Cell Degranulation/genetics , Down-Regulation/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Lung/pathology , Lung/physiology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Neutrophil Infiltration/genetics , Receptor, Adenosine A3 , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...