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










Publication year range
1.
Front Immunol ; 12: 740330, 2021.
Article in English | MEDLINE | ID: mdl-34603325

ABSTRACT

Rationale: The accumulation of macrophages in the airways and the pulmonary interstitium is a hallmark of cigarette smoke-associated inflammation. Notably, pulmonary macrophages are not a homogenous population but consist of several subpopulations. To date, the manner in which cigarette smoke exposure affects the relative composition and functional capacity of macrophage subpopulations has not been elucidated. Methods: Using a whole-body cigarette smoke exposure system, we investigated the impact of cigarette smoke on macrophage subpopulations in C57BL/6 mice using flow cytometry-based approaches. Moreover, we used bromodeoxyuridine labelling plus Il1a-/- and Il1r1-/- mice to assess the relative contribution of local proliferation and monocyte recruitment to macrophage accumulation. To assess the functional consequences of altered macrophage subpopulations, we used a model of concurrent bleomycin-induced lung injury and cigarette smoke exposure to examine tissue remodelling processes. Main Results: Cigarette smoke exposure altered the composition of pulmonary macrophages increasing CD11b+ subpopulations including monocyte-derived alveolar macrophages (Mo-AM) as well as interstitial macrophages (IM)1, -2 and -3. The increase in CD11b+ subpopulations was observed at multiple cigarette smoke exposure timepoints. Bromodeoxyuridine labelling and studies in Il1a-/- mice demonstrated that increased Mo-AM and IM3 turnover in the lungs of cigarette smoke-exposed mice was IL-1α dependent. Compositional changes in macrophage subpopulations were associated with impaired induction of fibrogenesis including decreased α-smooth muscle actin positive cells following intratracheal bleomycin treatment. Mechanistically, in vivo and ex vivo assays demonstrated predominant macrophage M1 polarisation and reduced matrix metallopeptidase 9 activity in cigarette smoke-exposed mice. Conclusion: Cigarette smoke exposure modified the composition of pulmonary macrophage by expanding CD11b+ subpopulations. These compositional changes were associated with attenuated fibrogenesis, as well as predominant M1 polarisation and decreased fibrotic activity. Overall, these data suggest that cigarette smoke exposure altered the composition of pulmonary macrophage subpopulations contributing to impaired tissue remodelling.


Subject(s)
Airway Remodeling/drug effects , Cigarette Smoking/adverse effects , Lung Injury/immunology , Lung/immunology , Macrophages/immunology , Animals , Bleomycin , CD11b Antigen/metabolism , Cells, Cultured , Disease Models, Animal , Female , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1alpha/metabolism , Lung Injury/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics
2.
Cells ; 8(2)2019 02 05.
Article in English | MEDLINE | ID: mdl-30764496

ABSTRACT

The accumulation of extracellular matrix in lung diseases involves numerous factors, including cytokines and chemokines that participate in cell activation in lung tissues and the circulation of fibrocytes that contribute to local fibrotic responses. The transient overexpression of the gp130 cytokine Oncostatin M can induce extracellular matrix (ECM) accumulation in mouse lungs, and here, we assess a role for IL-13 in this activity using gene deficient mice. The endotracheal administration of an adenovirus vector encoding Oncostatin M (AdOSM) caused increases in parenchymal lung collagen accumulation, neutrophil numbers, and CXCL1/KC chemokine elevation in bronchioalveolar lavage fluids. These effects were similar in IL-13-/- mice at day 7; however, the ECM matrix induced by Oncostatin M (OSM) was reduced at day 14 in the IL-13-/- mice. CD45+col1+ fibrocyte numbers were elevated at day 7 due to AdOSM whereas macrophages were not. Day 14 levels of CD45+col1+ fibrocytes were maintained in the wildtype mice treated with AdOSM but were reduced in IL-13-/- mice. The expression of the fibrocyte chemotactic factor CXCL12/SDF-1 was suppressed marginally by AdOSM in vivo and significantly in vitro in mouse lung fibroblast cell cultures. Thus, Oncostatin M can stimulate inflammation in an IL-13-independent manner in BALB/c lungs; however, the ECM remodeling and fibrocyte accumulation is reduced in IL-13 deficiency.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Lung/metabolism , Oncostatin M/metabolism , Animals , Bronchoalveolar Lavage Fluid , Chemokines/genetics , Chemokines/metabolism , Collagen/metabolism , Down-Regulation , Female , Inflammation/pathology , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Lab Invest ; 94(9): 1003-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24933422

ABSTRACT

Adverse health outcomes in pulmonary fibrosis are associated with extracellular matrix (ECM) accumulation. Although transforming growth factor-ß (TGF-ß) has been reported to be an important regulator of fibrosis pathogenesis, TGF-ß-independent pathways may also be involved. Here, we investigated responses of putative relatively fibrosis-resistant BALB/c mice to transient pulmonary overexpression of oncostatin M (OSM) using an adenovirus vector encoding OSM (AdOSM) and compared responses with the relatively fibrosis-prone C57Bl/6 strain. Interestingly, BALB/c mice showed similar ECM accumulation and collagen 1A1 and 3A1 mRNA elevation to C57Bl/6 mice 7 days after endotracheal administration of AdOSM. TGF-ß1 mRNA levels and pSMAD2 signal were not regulated in either strain in total lung extracts. In contrast to C57Bl/6 mice, BALB/c mice lacked eosinophil, Th2 cytokine, and pro-inflammatory cytokine elevation in the broncholveolar space. OSM overexpression induced STAT3 activation and SMAD1/5/8 signaling suppression in lung from both mice strains, which was associated with a downregulation of BMPR2 and BMP ligands, and increased expression of the BMP antagonist gremlin. Although we also observed STAT3 activation and SMAD1/5/8 signaling suppression in mouse lung fibroblast cultures in vitro upon OSM stimulation, immunohistochemistry analyses indicated that the AdOSM-induced pSMAD1/5/8 signal suppression was primarily localized to the airway epithelium. Other gp130 cytokines including IL-6, LIF, CT-1, but not IL-31, also induced STAT3 activation and SMAD1/5/8 signaling suppression in C10 mouse lung epithelial cells and BEAS 2B bronchial epithelial cells, and we found that pharmacological inhibition of STAT3 activation reversed OSM-induced SMAD1/5/8 signaling suppression in vitro. The results demonstrate that OSM induces ECM accumulation in fibrosis-resistant BALB/c mouse lung in the absence of Th2 inflammation or TGF-ß signaling, and highlight a dichotomy of STAT3 activation versus SMAD1 suppression in this process.


Subject(s)
Extracellular Matrix/metabolism , Lung/metabolism , Oncostatin M/metabolism , STAT3 Transcription Factor/metabolism , Smad1 Protein/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Bronchoalveolar Lavage Fluid , Female , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pulmonary Fibrosis , Reverse Transcriptase Polymerase Chain Reaction
4.
J Immunol ; 191(3): 1453-64, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23797667

ABSTRACT

Inducible BALT (iBALT) is associated with immune responses to respiratory infections as well as with local pathology derived from chronic inflammatory lung diseases. In this study, we assessed the role of oncostatin M (OSM) in B cell activation and iBALT formation in mouse lungs. We found that C57BL/6 mice responded to an endotracheally administered adenovirus vector expressing mouse OSM, with marked iBALT formation, increased cytokine (IL-4, IL-5, IL-6, IL-10, TNF-α, and IL-12), and chemokine (CXCL13, CCL20, CCL21, eotaxin-2, KC, and MCP-1) production as well as inflammatory cell accumulation in the airways. B cells, T cells, and dendritic cells were also recruited to the lung, where many displayed an activated phenotype. Mice treated with control adenovirus vector (Addl70) were not affected. Interestingly, IL-6 was required for inflammatory responses in the airways and for the expression of most cytokines and chemokines. However, iBALT formation and lymphocyte recruitment to the lung tissue occurred independently of IL-6 and STAT6 as assessed in gene-deficient mice. Collectively, these results support the ability of OSM to induce B cell activation and iBALT formation independently of IL-6 and highlight a role for IL-6 downstream of OSM in the induction of pulmonary inflammation.


Subject(s)
B-Lymphocytes/immunology , Interleukin-6/metabolism , Lymphoid Tissue/metabolism , Oncostatin M/metabolism , Pneumonia/metabolism , Animals , B-Lymphocytes/metabolism , Bronchoalveolar Lavage Fluid/cytology , Cell Line, Tumor , Cell Movement , Chemokines/biosynthesis , Cytokines/biosynthesis , HeLa Cells , Humans , Interleukin-6/deficiency , Interleukin-6/genetics , Lung/metabolism , Lymphocyte Activation , Lymphoid Tissue/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncostatin M/biosynthesis , Oncostatin M/genetics , Pneumonia/pathology , STAT6 Transcription Factor/metabolism , Transfection
5.
Chest ; 143(3): 856-858, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23460164

ABSTRACT

Airway inflammation is considered a central component of asthma and, therefore, international guidelines recommend antiinflammatory medications. We describe the clinical history of a 34-year-old woman with airway hyperresponsiveness and asthma who had a reduced ability to mount an inflammatory response due to two unrelated and rare genetic conditions: Fanconi anemia and incontinentia pigmenti. Absence of eosinophils in blood and sputum led to a successful reduction in the dose of corticosteroids without loss of asthma control demonstrating the clinical utility of monitoring treatment using biomarkers and the importance of recognizing the components of airway diseases that contribute to symptoms.


Subject(s)
Asthma/epidemiology , Fanconi Anemia/epidemiology , Incontinentia Pigmenti/epidemiology , Adult , Comorbidity , Eosinophils/metabolism , Female , Humans , Incontinentia Pigmenti/genetics , Sputum/cytology , Sputum/metabolism
6.
Respir Res ; 13: 81, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22992200

ABSTRACT

BACKGROUND: Evidence suggests that dendritic cells accumulate in the lungs of COPD patients and correlate with disease severity. We investigated the importance of IL-1R1 and its ligands IL-1α and ß to dendritic cell accumulation and maturation in response to cigarette smoke exposure. METHODS: Mice were exposed to cigarette smoke using a whole body smoke exposure system. IL-1R1-, TLR4-, and IL-1α-deficient mice, as well as anti-IL-1α and anti-IL-1ß blocking antibodies were used to study the importance of IL-1R1 and TLR4 to dendritic cell accumulation and activation. RESULTS: Acute and chronic cigarette smoke exposure led to increased frequency of lung dendritic cells. Accumulation and activation of dendritic cells was IL-1R1/IL-1α dependent, but TLR4- and IL-1ß-independent. Corroborating the cellular data, expression of CCL20, a potent dendritic cells chemoattractant, was IL-1R1/IL-1α-dependent. Studies using IL-1R1 bone marrow-chimeric mice revealed the importance of IL-1R1 signaling on lung structural cells for CCL20 expression. Consistent with the importance of dendritic cells in T cell activation, we observed decreased CD4+ and CD8+ T cell activation in cigarette smoke-exposed IL-1R1-deficient mice. CONCLUSION: Our findings convey the importance of IL-1R1/IL-1α to the recruitment and activation of dendritic cells in response to cigarette smoke exposure.


Subject(s)
Chemotaxis/drug effects , Dendritic Cells/drug effects , Interleukin-1alpha/metabolism , Lung/drug effects , Smoke/adverse effects , Smoking/adverse effects , Animals , Antibodies, Blocking , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL20/metabolism , Dendritic Cells/immunology , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/metabolism , Lung/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Signal Transduction , Time Factors , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Transplantation Chimera
7.
PLoS One ; 6(12): e28457, 2011.
Article in English | MEDLINE | ID: mdl-22163019

ABSTRACT

BACKGROUND: Cigarette smoking is the main risk factor for the development of chronic obstructive pulmonary disease (COPD), a major cause of morbidity and mortality worldwide. Despite this, the cellular and molecular mechanisms that contribute to COPD pathogenesis are still poorly understood. METHODOLOGY AND PRINCIPAL FINDINGS: The objective of this study was to assess IL-1 α and ß expression in COPD patients and to investigate their respective roles in perpetuating cigarette smoke-induced inflammation. Functional studies were pursued in smoke-exposed mice using gene-deficient animals, as well as blocking antibodies for IL-1α and ß. Here, we demonstrate an underappreciated role for IL-1α expression in COPD. While a strong correlation existed between IL-1α and ß levels in patients during stable disease and periods of exacerbation, neutrophilic inflammation was shown to be IL-1α-dependent, and IL-1ß- and caspase-1-independent in a murine model of cigarette smoke exposure. As IL-1α was predominantly expressed by hematopoietic cells in COPD patients and in mice exposed to cigarette smoke, studies pursued in bone marrow chimeric mice demonstrated that the crosstalk between IL-1α+ hematopoietic cells and the IL-1R1+ epithelial cells regulates smoke-induced inflammation. IL-1α/IL-1R1-dependent activation of the airway epithelium also led to exacerbated inflammatory responses in H1N1 influenza virus infected smoke-exposed mice, a previously reported model of COPD exacerbation. CONCLUSIONS AND SIGNIFICANCE: This study provides compelling evidence that IL-1α is central to the initiation of smoke-induced neutrophilic inflammation and suggests that IL-1α/IL-1R1 targeted therapies may be relevant for limiting inflammation and exacerbations in COPD.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin-1alpha/biosynthesis , Neutrophils/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Smoking , Animals , Biopsy , Caspase 1/metabolism , Humans , Inflammation , Interleukin-1beta/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Smoke , Sputum/metabolism
8.
Respir Res ; 12: 39, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21473774

ABSTRACT

BACKGROUND: While the presence of the chitinase-like molecule YKL40 has been reported in COPD and asthma, its relevance to inflammatory processes elicited by cigarette smoke and common environmental allergens, such as house dust mite (HDM), is not well understood. The objective of the current study was to assess expression and function of BRP-39, the murine equivalent of YKL40 in a murine model of cigarette smoke-induced inflammation and contrast expression and function to a model of HDM-induced allergic airway inflammation. METHODS: CD1, C57BL/6, and BALB/c mice were room air- or cigarette smoke-exposed for 4 days in a whole-body exposure system. In separate experiments, BALB/c mice were challenged with HDM extract once a day for 10 days. BRP-39 was assessed by ELISA and immunohistochemistry. IL-13, IL-1R1, IL-18, and BRP-39 knock out (KO) mice were utilized to assess the mechanism and relevance of BRP-39 in cigarette smoke- and HDM-induced airway inflammation. RESULTS: Cigarette smoke exposure elicited a robust induction of BRP-39 but not the catalytically active chitinase, AMCase, in lung epithelial cells and alveolar macrophages of all mouse strains tested. Both BRP-39 and AMCase were increased in lung tissue after HDM exposure. Examining smoke-exposed IL-1R1, IL-18, and IL-13 deficient mice, BRP-39 induction was found to be IL-1 and not IL-18 or IL-13 dependent, while induction of BRP-39 by HDM was independent of IL-1 and IL-13. Despite the importance of BRP-39 in cellular inflammation in HDM-induced airway inflammation, BRP-39 was found to be redundant for cigarette smoke-induced airway inflammation and the adjuvant properties of cigarette smoke. CONCLUSIONS: These data highlight the contrast between the importance of BRP-39 in HDM- and cigarette smoke-induced inflammation. While functionally important in HDM-induced inflammation, BRP-39 is a biomarker of cigarette smoke induced inflammation which is the byproduct of an IL-1 inflammatory pathway.


Subject(s)
Glycoproteins/metabolism , Lung/metabolism , Pneumonia/metabolism , Smoking/adverse effects , Animals , Antigens, Dermatophagoides , Chitinase-3-Like Protein 1 , Chitinases/metabolism , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Glycoproteins/deficiency , Glycoproteins/genetics , Immunohistochemistry , Inflammation Mediators/metabolism , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Pneumonia/etiology , Pneumonia/pathology , Pneumonia/physiopathology , Time Factors
9.
Am J Respir Cell Mol Biol ; 45(4): 753-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21317378

ABSTRACT

Although a similar prevalence of smoking is evident among patients with asthma and the general population, little is known about the impact of cigarette smoke on the immune inflammatory processes elicited by common environmental allergens. We investigated the impact of exposure to cigarette smoke on house dust mite (HDM)-induced allergic airway inflammation and its consequences for tissue remodeling and lung physiology in mice. BALB/c mice received intranasal HDMs daily, 5 days per week, for 3 weeks to establish chronic airway inflammation. Subsequently, mice were concurrently exposed to HDMs plus cigarette smoke, 5 days per week, for 2 weeks (HDMs + smoke). We observed significantly attenuated eosinophilia in the bronchoalveolar lavage of mice exposed to HDMs + smoke, compared with animals exposed only to HDMs. A similar activation of CD4 T cells and expression of IL-5, IL-13, and transforming growth factor-ß was observed between HDM-treated and HDM + smoke-treated animals. Consistent with an effect on eosinophil trafficking, HDMs + smoke exposure attenuated the HDM-induced expression of eotaxin-1 and vascular cell adhesion molecule-1, whereas the survival of eosinophils and the numbers of blood eosinophils were not affected. Exposure to cigarette smoke also reduced the activation of B cells and the concentrations of serum IgE. Although the production of mucus decreased, collagen deposition significantly increased in animals exposed to HDMs + smoke, compared with animals exposed only to HDMs. Although airway resistance was unaffected, tissue resistance was significantly decreased in mice exposed to HDMs + smoke. Our findings demonstrate that cigarette smoke affects eosinophil migration without affecting airway resistance or modifying Th2 cell adaptive immunity in a murine model of HDM-induced asthma.


Subject(s)
Airway Remodeling , Allergens , Asthma/immunology , Lung/immunology , Pulmonary Eosinophilia/prevention & control , Pyroglyphidae/immunology , Smoking/immunology , Airway Resistance , Animals , Asthma/pathology , Asthma/physiopathology , B-Lymphocytes/immunology , Bronchial Hyperreactivity/immunology , Chemokine CCL11/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Interleukin-13/metabolism , Interleukin-5/metabolism , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Pulmonary Eosinophilia/physiopathology , Smoking/pathology , Smoking/physiopathology , T-Lymphocytes/immunology , Time Factors , Transforming Growth Factor beta/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
10.
PLoS One ; 5(10): e13251, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20967263

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease is a progressive lung disease that is punctuated by periods of exacerbations (worsening of symptoms) that are attributable to viral infections. While rhinoviruses are most commonly isolated viruses during episodes of exacerbation, influenza viruses have the potential to become even more problematic with the increased likelihood of an epidemic. METHODOLOGY AND PRINCIPAL FINDINGS: This study examined the impact of current and potential pharmacological targets namely the systemic corticosteroid dexamethasone and the peroxisome proliferator-activated receptor-gamma agonist pioglitazone on the outcome of infection in smoke-exposed mice. C57BL/6 mice were exposed to room air or cigarette smoke for 4 days and subsequently inoculated with an H1N1 influenza A virus. Interventions were delivered daily during the course of infection. We show that smoke-exposed mice have an exacerbated inflammatory response following infection. While smoke exposure did not compromise viral clearance, precision cut lung slices from smoke-exposed mice showed greater expression of CC (MCP-1, -3), and CXC (KC, MIP-2, GCP-2) chemokines compared to controls when stimulated with a viral mimic or influenza A virus. While dexamethasone treatment partially attenuated the inflammatory response in the broncho-alveolar lavage of smoke-exposed, virally-infected animals, viral-induced neutrophilia was steroid insensitive. In contrast to controls, dexamethasone-treated smoke-exposed influenza-infected mice had a worsened health status. Pioglitazone treatment of virally-infected smoke-exposed mice proved more efficacious than the steroid intervention. Further mechanistic evaluation revealed that a deficiency in CCR2 did not improve the inflammatory outcome in smoke-exposed, virally-infected animals. CONCLUSIONS AND SIGNIFICANCE: This animal model of cigarette smoke and H1N1 influenza infection demonstrates that smoke-exposed animals are differentially primed to respond to viral insult. While providing a platform to test pharmacological interventions, this model demonstrates that treating viral exacerbations with alternative anti-inflammatory drugs, such as PPAR-gamma agonists should be further explored since they showed greater efficacy than systemic corticosteroids.


Subject(s)
Disease Models, Animal , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/complications , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking , Animals , Chemokines/metabolism , Humans , Influenza, Human/drug therapy , Influenza, Human/metabolism , Influenza, Human/virology , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Pioglitazone , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/virology , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
11.
Am J Respir Cell Mol Biol ; 42(4): 394-403, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19502389

ABSTRACT

The objective of this study was to characterize the impact of cigarette smoke exposure on lung immune and inflammatory processes. BALB/c and C57BL/6 mice were exposed to cigarette smoke for 4 days (acute) or at least 5 weeks (prolonged). Both mouse strains manifested an inflammatory response after acute smoke exposure, characterized by an influx of neutrophils and mononuclear cells. Multiplex analysis revealed a greater than twofold increase of the cytokines IL-1alpha, -5, -6, and -18, as well as the chemokines monocyte chemotactic protein-1 and -3, macrophage inflammatory protein-1alpha, -beta, and -gamma, -2, -3beta, macrophage defined chemokine, granulocyte chemotactic protein-2, and interferon-gamma-inducible protein-10. In BALB/c mice, neutrophilia persisted after prolonged exposure, whereas C57BL/6 showed evidence of attenuated neutrophilia both in the bronchoalveolar lavage and the lungs. In both mouse strains, cigarette smoke exposure was associated with an expansion of mature (CD11c(hi)/major histocompatibility complex class II(hi)) myeloid dendritic cells; we observed no changes in plasmacytoid dendritic cells. Lymphocytes in the lungs displayed an activated phenotype that persisted for CD4 T cells only after prolonged exposure. In BALB/c mice, T cells acquired T helper (Th) 1 and Th2 effector function after 5 weeks of smoke exposure, whereas, in C57BL/6 mice, neither Th1 nor Th2 cells were detected. In both mouse strains, cigarette smoke exposure led to an accumulation of FoxP3+ T regulatory cells in the lungs. Studies in RAG1 knockout mice suggest that these regulatory cells may participate in controlling smoke-induced inflammation. Acute and prolonged cigarette smoke exposure was associated with inflammation, activation of the adaptive immune system, and expansion of T regulatory cells in the lungs.


Subject(s)
Immunity, Innate/immunology , Pneumonia/immunology , Smoking/adverse effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Bronchoalveolar Lavage , CD11c Antigen/genetics , CD11c Antigen/immunology , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Immunity, Innate/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/pathology , Smoking/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Time Factors
12.
Am J Respir Cell Mol Biol ; 40(1): 38-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18635815

ABSTRACT

The impact of cigarette smoke on allergic asthma remains controversial both clinically and experimentally. The objective of this study was to investigate, in a murine model, how cigarette smoke affects immune inflammatory processes elicited by a surrogate allergen. In our experimental design, mice were concurrently exposed to cigarette smoke and ovalbumin (OVA), an innocuous antigen that, unless introduced in the context of an adjuvant, induces inhalation tolerance. We show that cigarette smoke exposure has adjuvant properties, allowing for allergic mucosal sensitization to OVA. Specifically, concurrent exposure to cigarette smoke and OVA for 2 weeks led to airway eosinophilia and goblet cell hyperplasia. In vivo OVA recall challenge 1 month after the last smoke exposure showed that concurrent exposure to OVA and cigarette smoke induced antigen-specific memory. Robust eosinophilia and OVA-specific IgG1 and IgE characterized the ensuing inflammatory response. Mechanistically, allergic sensitization was, in part, granulocyte macrophage colony-stimulating factor (GM-CSF) dependent, as a significant reduction in BAL eosinophilia was observed in mice treated with an anti-GM-CSF antibody. Of note, continuous smoke exposure attenuated the OVA recall response; decreased airway eosinophilia was observed in mice continuously exposed to cigarette smoke compared with mice that ceased the smoke exposure protocol. In conclusion, we demonstrate experimentally that while cigarette smoke acts as an adjuvant allowing for allergic sensitization, it also attenuates the ensuing eosinophilic inflammatory response.


Subject(s)
Adjuvants, Immunologic , Allergens/immunology , Asthma/immunology , Bronchial Hyperreactivity/immunology , Inflammation/immunology , Nicotiana , Smoke , Animals , Antigens, CD/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Immune Tolerance/physiology , Immunologic Memory , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , T-Lymphocytes/immunology
13.
Cell Immunol ; 253(1-2): 38-44, 2008.
Article in English | MEDLINE | ID: mdl-18533139

ABSTRACT

Although its direct effects cannot be discounted, tobacco's effects on the immune system have been proposed to play a key role in mediating its deleterious health impact. Studies in rats using high levels of smoke exposure have suggested that tobacco smoke exhausts cellular signal transduction cascades, making lymphocytes unresponsive to stimulation. In the present study, we show that purified B or T cells, and total lymphocytes from the lungs, lymph nodes and spleens of smoke-exposed mice fluxed calcium, proliferated, and secreted immunoglobulin or IFN-gamma similarly to control mice when stimulated with ligands including anti-IgM, and anti-CD3. Importantly, we recapitulated these findings in PBMCs from human smokers; cells from long-term smokers and never-smokers proliferated equivalently when stimulated ex vivo. Previous reports of lymphocyte unresponsiveness in rats are inconsistent with these findings, and may reflect a phenomenon observed only at levels of smoke exposure well above those seen in actual human smokers.


Subject(s)
B-Lymphocytes/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Subsets/drug effects , Nicotiana/adverse effects , Smoke/adverse effects , T-Lymphocytes/drug effects , Animals , B-Lymphocytes/immunology , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Lung/cytology , Lung/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Rats , Smoking/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Nicotiana/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...