Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 13(3): 423-436, 2020 05.
Article in English | MEDLINE | ID: mdl-31819170

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a highly prevalent respiratory disease characterized by airflow limitation and chronic inflammation. MiR-155 is described as an ancient regulator of the immune system. Our objective was to establish a role for miR-155 in cigarette smoke (CS)-induced inflammation and COPD. We demonstrate increased miR-155 expression by RT-qPCR in lung tissue of smokers without airflow limitation and patients with COPD compared to never smokers and in lung tissue and alveolar macrophages of CS-exposed mice compared to air-exposed mice. In addition, we exposed wild type and miR-155 deficient mice to CS and show an attenuated inflammatory profile in the latter. Alveolar macrophages were sorted by FACS from the different experimental groups and their gene expression profile was analyzed by RNA sequencing. This analysis revealed increased expression of miR-155 targets and an attenuation of the CS-induced increase in inflammation-related genes in miR-155 deficient mice. Moreover, intranasal instillation of a specific miR-155 inhibitor attenuated the CS-induced pulmonary inflammation in mice. Finally, elastase-induced emphysema and lung functional changes were significantly attenuated in miR-155 deficient mice. In conclusion, we highlight a role for miR-155 in CS-induced inflammation and the pathogenesis of COPD, implicating miR-155 as a new therapeutic target in COPD.


Subject(s)
Cigarette Smoking/adverse effects , Disease Susceptibility , MicroRNAs/genetics , Pneumonia/etiology , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Knockout , Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , RNA Interference
2.
Mucosal Immunol ; 12(2): 340-351, 2019 03.
Article in English | MEDLINE | ID: mdl-30542109

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is associated with abnormal inflammatory responses and airway wall remodeling, leading to reduced lung function. An association between the bone morphogenetic protein (BMP-6) locus and forced vital capacity has been found in a genome-wide association study. However, the role of BMP-6 in the pathogenesis of COPD remains unknown. The pulmonary expression of BMP-6 was analyzed in patients with COPD and in cigarette smoke (CS)-exposed mice. We evaluated lung function and histology in BMP-6 KO mice at baseline. We exposed BMP-6 KO mice to CS for 4 weeks and measured pulmonary inflammation and iron levels. Pulmonary mRNA levels of BMP-6 were decreased in smokers with and without COPD and in CS-exposed mice. Importantly, BMP-6 expression was lowest in severe COPD. Accordingly, protein levels of BMP-6 were decreased in patients with COPD. Lung function measurements demonstrated a decreased compliance and total lung capacity in BMP-6 KO mice, whereas lung histology was normal. Furthermore, BMP-6 KO mice displayed elevated iron levels and an aggravated CS-induced inflammatory response. These results suggest that BMP-6 is important for normal lung function and that downregulation of BMP-6-as observed in patients with COPD-contributes to pulmonary inflammation after CS exposure.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Iron/metabolism , Lung/metabolism , Pneumonia/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Aged , Animals , Bone Morphogenetic Protein 6/genetics , Cells, Cultured , Cigarette Smoking/adverse effects , Disease Models, Animal , Female , Humans , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pneumonia/etiology , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/immunology
3.
Mucosal Immunol ; 10(6): 1400-1411, 2017 11.
Article in English | MEDLINE | ID: mdl-28145442

ABSTRACT

The molecular mechanisms underlying the pathogenesis of chronic obstructive pulmonary disease (COPD) are still unclear, however signaling pathways associated with lung development, such as the transforming growth factor (TGF)-ß superfamily, could be implicated in COPD. Growth differentiation factor (GDF)-15, a member of the TGF-ß superfamily, is involved in inflammation, mucus secretion, and cachexia. We analyzed the pulmonary expression of GDF-15 in smokers and patients with COPD, in cigarette smoke (CS)-exposed cultures of primary human bronchial epithelial cells (pHBECs), and in CS-exposed mice. Next, we exposed GDF-15 KO and control mice to air or CS and evaluated pulmonary inflammation. GDF-15 levels were higher in sputum supernatant and lung tissue of patients with COPD and smokers without COPD compared with never smokers. Immunohistochemistry revealed GDF-15 staining in the airway epithelium. Increased expression and secretion of GDF-15 was confirmed in vitro in CS-exposed pHBECs compared with air-exposed pHBECs. Similarly, GDF-15 levels were increased in lungs of CS-exposed mice. Importantly, GDF-15 deficiency attenuated the CS-induced pulmonary inflammation. These results suggest that increased GDF-15-as observed in lungs of smokers and patients with COPD-contributes to CS-induced pulmonary inflammation.


Subject(s)
Cigarette Smoking/adverse effects , Epithelial Cells/immunology , Growth Differentiation Factor 15/metabolism , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Animals , Bronchi/pathology , Cells, Cultured , Cohort Studies , Female , Growth Differentiation Factor 15/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Up-Regulation
4.
Mol Cell Endocrinol ; 443: 23-31, 2017 03 05.
Article in English | MEDLINE | ID: mdl-27940297

ABSTRACT

INTRODUCTION: Steroid hormones are involved in lung development, pulmonary inflammation, and lung cancer. Estrogen signaling and exposure may play a role in pulmonary disorders, including COPD. In both genders, estrogens can be generated locally in the lungs and this contributes importantly to the tissue exposure to these steroids. OBJECTIVE: To characterize and assess differences in localization of estrogen receptors and enzymes involved in the local generation of estrogens in COPD. METHODS: Estrogen Receptor alpha (ERα/ESR1), Estrogen Receptor beta (ERß/ESR2) and G-protein-coupled estrogen receptor 1 (GPER) were explored by real-time (RT)-PCR analysis (mRNA expression), immunohistochemistry and western blotting in controls and COPD patients. mRNA expression of the enzymes involved in the local estrogen generation - i.e. aromatase (CYP19A1), 17beta-hydroxysteroid dehydrogenases (17ß-HSDs) 1, 2, 4, 5, 7 and 12, steroid sulfatase (STS) and sulfotransferase (SULT1E1) - were analyzed by RT-PCR. RESULTS: ERα, ERß and GPER were expressed in lung tissue, but no differences were observed between patients and controls. The main enzymes involved in local estrogen generation were also present in both normal and COPD lung tissue. In lungs of COPD patients compared with controls, we observed increased expression of the enzymes 17ß-HSD type 1 and aromatase (positive association), both involved in the local synthesis of active estrogens. CONCLUSION: All ER subtypes are present in the lung. The shift in local mRNA level of estrogen metabolic enzymes suggests that exposure to estrogens is involved in the pathogenesis of COPD.


Subject(s)
Estradiol/biosynthesis , Pulmonary Disease, Chronic Obstructive/enzymology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation , Humans , Lung/metabolism , Lung/pathology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism
5.
Eur Respir J ; 39(4): 830-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21920889

ABSTRACT

Pentraxin (PTX)3 is involved in antimicrobial defence, apoptotic cell clearance and extracellular matrix stability. As these processes are altered in chronic obstructive pulmonary disease (COPD), we aimed to investigate PTX3 expression in patients with this disease. PTX3 expression was quantified by immunohistochemical staining of lung tissue from never-smokers, smokers without COPD, and in patients with COPD of Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage I, II and III-IV. mRNA expression was examined in total lung tissue by quantitative RT-PCR. PTX3 concentration was measured in induced sputum and plasma by ELISA. PTX3 is mainly localised in the interstitium of the small airways and alveolar walls. There were no significant differences in pulmonary, sputum and plasma PTX3 expression between study groups. However, PTX3 expression in small airways correlated significantly with forced expiratory volume in 1 s (r = 0.35, p = 0.004). In the alveolar walls, PTX3 expression correlated significantly with carbon monoxide transfer coefficient (r = 0.28, p = 0.04). In sputum, PTX3 levels were highly correlated with the number of neutrophils. Finally, systemic levels of PTX3 tended to be lower in severe COPD compared with mild COPD. In COPD, airflow limitation and reduced transfer coefficient for carbon monoxide are associated with lower pulmonary interstitial expression of PTX3.


Subject(s)
C-Reactive Protein/genetics , Lung/physiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Serum Amyloid P-Component/genetics , Adult , Aged , Bronchioles/physiology , C-Reactive Protein/metabolism , Female , Humans , Male , Middle Aged , Mucous Membrane/metabolism , Pulmonary Alveoli/physiology , Pulmonary Artery/physiology , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/metabolism , Respiratory Mucosa/physiology , Serum Amyloid P-Component/metabolism , Smoking/metabolism , Smoking/physiopathology , Sputum/metabolism
6.
Eur Respir J ; 38(5): 1019-28, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21622588

ABSTRACT

Cigarette smoke (CS), the primary risk factor of chronic obstructive pulmonary disease (COPD), leads to pulmonary inflammation through interleukin-1 receptor (IL-1R)I signalling, as determined using COPD mouse models. It is unclear whether interleukin (IL)-1α or IL-1ß, activated by the Nlrp3/caspase-1 axis, is the predominant ligand for IL-1RI in CS-induced responses. We exposed wild-type mice (treated with anti-IL-1α or anti-IL-1ß antibodies), and IL-1RI knockout (KO), Nlrp3 KO and caspase-1 KO mice to CS for 3 days or 4 weeks and evaluated pulmonary inflammation. Additionally, we measured the levels of IL-1α and IL-1ß mRNA (in total lung tissue by RT-PCR) and protein (in induced sputum by ELISA) of never-smokers, smokers without COPD and patients with COPD. In CS-exposed mice, pulmonary inflammation was dependent on IL-1RI and could be significantly attenuated by neutralising IL-1α or IL-1ß. Interestingly, CS-induced inflammation occurred independently of IL-1ß activation by the Nlrp3/caspase-1 axis. In human subjects, IL-1α and IL-1ß were significantly increased in total lung tissue and induced sputum of patients with COPD, respectively, compared with never-smokers. These results suggest that not only IL-1ß but also IL-1α should be considered as an important mediator in CS-induced inflammation and COPD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking/adverse effects , Animals , Antibodies, Neutralizing/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Humans , Inflammation , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Lung/pathology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/pathology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Receptors, Interleukin-1/metabolism , Tobacco Smoke Pollution/adverse effects
8.
Eur Respir J ; 36(4): 781-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20351031

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are professional antigen-presenting cells with antiviral and tolerogenic capabilities. Viral infections and autoimmunity are proposed to be important mechanisms in the pathogenesis of chronic obstructive pulmonary disease (COPD). The study aimed to quantify blood dendritic cell antigen 2-positive pDCs in lungs of subjects with or without COPD by immunohistochemistry and flow cytometry, combined with the investigation of the influence of cigarette smoke extract (CSE) on the function of pDCs in vitro. pDCs were mainly located in lymphoid follicles, a finding compatible with their expression of lymphoid homing chemokine receptors CXCR3 and CXCR4. pDC accumulated in the lymphoid follicles and in lung digests of patients with mild to moderate COPD, compared with smokers without airflow limitation and patients with COPD Global Initiative for Chronic Obstructive Lung disease (GOLD) stage III-IV. Exposing maturing pDC of healthy subjects to CSE in vitro revealed an attenuation of the expression of co-stimulatory molecules and impaired interferon-α production. Maturing pDC from patients with COPD produced higher levels of tumour necrosis factor (TNF)-α and interleukin (IL)-8 compared to pDC from healthy subjects. CSE significantly impairs the antiviral function of pDCs. In COPD, a GOLD stage dependent accumulation of pDC in lymphoid follicles is present, combined with an enhanced production of TNF-α and IL-8 by maturing pDCs.


Subject(s)
Dendritic Cells/cytology , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Case-Control Studies , Dendritic Cells/pathology , Female , Flow Cytometry/methods , Humans , Immunohistochemistry/methods , Inflammation , Interleukin-8/metabolism , Lung/cytology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking/adverse effects , Tumor Necrosis Factor-alpha/metabolism
9.
Eur Respir J ; 34(1): 219-30, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19567605

ABSTRACT

Inflammation is a main pathogenetic factor in the development and progression of chronic obstructive pulmonary disease (COPD). Recently, it has become clear that not only the innate, but also the specific immune response plays a role. A striking finding, in particular in lungs of patients with severe COPD, often with a predominant emphysema phenotype, is the presence of B-cell follicles. As seen in other tissues, these follicles are the result of lymphoid neogenesis. The finding of oligoclonality in B-cell follicles in COPD suggests that they play a role in local antigen specific immune responses. To date, it is not known which antigens may be involved; microbial antigens, cigarette smoke-derived antigens and antigens from extracellular matrix breakdown products have been suggested. Consequently, the pathogenetic role of this follicular B-cell response is not yet clear. It might be protective against microbial colonisation and infection of the lower respiratory tract and, therefore, beneficial, or it could be of a more harmful (autoimmune) nature, directed against lung tissue components. It is necessary to determine the specific antigen(s) and to explore the exact role of the COPD related B-cell response in order to include modulation of this response and develop therapeutic options.


Subject(s)
Pulmonary Disease, Chronic Obstructive/physiopathology , Animals , B-Lymphocytes/metabolism , Dendritic Cells/cytology , Disease Models, Animal , Extracellular Matrix/metabolism , Humans , Immune System , Inflammation , Lymphocytes/metabolism , Mice , Mice, Transgenic , Models, Biological , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Emphysema/complications , Pulmonary Emphysema/diagnosis
10.
Eur Respir J ; 34(2): 405-16, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19164352

ABSTRACT

In chronic obstructive pulmonary disease (COPD), chronic inflammation is accompanied by peribronchial lymphoid aggregates. Lymphotoxin (LT)-alpha, crucial in secondary lymphoid organogenesis, may be involved in lymphoid neogenesis. We examined cigarette smoke (CS)-induced pulmonary lymphoid neogenesis and inflammation in vivo in LTalpha knockout (LTalpha(-/-)) and wild-type (WT) mice and studied the expression of lymphoid chemokines by lung fibroblasts in vitro. T-cell numbers (in bronchoalveolar lavage fluid (BALF) and lungs) and lymphoid aggregate numbers were significantly higher in air-exposed LTalpha(-/-) mice than in WT animals, and increased upon chronic CS exposure in both genotypes. In contrast, local immunoglobulin A responses upon chronic CS exposure were attenuated in LTalpha(-/-) mice. CXC chemokine ligand (CXCL) 13 and CC chemokine ligand (CCL) 19 mRNA in total lung and CXCL13 protein level in BALF increased upon CS exposure in WT, but not in LTalpha(-/-) mice. In vitro lymphotoxin-beta receptor (LTbetaR) stimulation induced CXCL13 and CCL19 mRNA in WT lung fibroblasts. Furthermore, in vitro exposure to CS extract upregulated CXCL13 mRNA expression in WT, but not in LTbetaR(-/-), lung fibroblasts. In this murine model of COPD, CS induces pulmonary expression of lymphoid chemokines CXCL13 and CCL19 in a LTalphabeta-LTbetaR-dependent fashion. However, LTalpha is not required for CS-induced pulmonary lymphocyte accumulation and neogenesis of lymphoid aggregates.


Subject(s)
Inflammation/chemically induced , Lymphotoxin-alpha/metabolism , Smoke , Smoking/adverse effects , Animals , Bronchoalveolar Lavage Fluid , Chemokine CCL19/biosynthesis , Chemokine CXCL13/biosynthesis , Fibroblasts/metabolism , Lung/cytology , Lung/drug effects , Lymphoid Tissue/drug effects , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Disease, Chronic Obstructive/physiopathology
11.
Thorax ; 64(1): 26-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18835960

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterised by an abnormal inflammatory reaction of the lungs involving activation of epithelial cells. Leptin is a pleiotropic cytokine important in the regulation of immune responses via its functional receptor Ob-Rb. This study was undertaken to test the hypothesis that severe COPD is associated with increased leptin expression in epithelial cells. METHODS: Immunohistochemistry for leptin was performed on peripheral lung specimens from 20 patients with COPD (GOLD stage 4), 14 asymptomatic ex-smokers and 13 never smokers. Leptin and Ob-Rb mRNA expression were determined by rtPCR in cultured primary bronchial epithelial cells and primary type II pneumocytes. NCI-H292 and A549 cell lines were used to study functional activation of leptin signalling. RESULTS: Leptin immunoreactivity in lung tissue was observed in bronchial epithelial cells, type II pneumocytes, macrophages (tissue/alveolar) and interstitial lymphocytic infiltrates. rtPCR analysis confirmed pulmonary leptin and Ob-Rb mRNA expression in primary bronchial epithelial cells and pneumocytes. Leptin-expressing bronchial epithelial cells and alveolar macrophages were markedly higher in patients with severe COPD and ex-smokers than in never smokers (p<0.02). Exposure of cultured primary bronchial epithelial cells to smoke resulted in increased expression of both leptin and Ob-Rb (p<0.05). Leptin induced phosphorylation of STAT3 in both NCI-H292 and A549 cells. CONCLUSIONS: Leptin expression is increased in bronchial epithelial cells and alveolar macrophages of ex-smokers with or without severe COPD compared with never smokers. A functional leptin signalling pathway is present in lung epithelial cells.


Subject(s)
Leptin/metabolism , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Leptin/metabolism , Smoking/metabolism , Blotting, Western , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , Epithelial Cells/metabolism , Female , Forced Expiratory Volume/physiology , Humans , Immunohistochemistry , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , RNA, Messenger/metabolism , Vital Capacity/physiology
12.
Clin Exp Allergy ; 37(10): 1467-79, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17883726

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterized by a chronic inflammatory response of the airways and lungs to noxious particles and gases, mostly cigarette smoke (CS). Pathological changes characteristic of COPD include airway wall thickening, peribronchial fibrosis, peribronchial lymphoid follicles and destruction of lung parenchyma (emphysema). The recruitment of inflammatory cells into the lung in response to CS is thought to play an important role in the development of COPD. OBJECTIVE: Our aim was to study the contribution of chemokine receptor 5 (CCR5) to the pathogenesis of COPD and specifically whether the development of airway remodelling is a direct result of airway inflammation or rather occurs through an independent mechanism. METHODS: In this study, C57BL/6 wild-type mice and CCR5-deficient mice were subjected to sub-acute (4 weeks) and chronic (24 weeks) CS exposure. RESULTS: Both sub-acute and chronic CS exposure significantly increased CCR5 mRNA expression and protein levels of CCR5 ligands [macrophage inflammatory protein-1alpha (MIP-1alpha), MIP-1beta and regulated upon activation, normal T expressed and secreted (RANTES)], and induced the recruitment of neutrophils, macrophages, dendritic cells, and lymphocytes to the bronchoalveolar lavage (BAL) of wild-type mice. Chronic CS exposure also increased the number and extent of peribronchial lymphoid follicles. In CCR5 knockout (KO) mice, these CS-induced increases in CCR5 ligands, inflammatory cells in BAL and peribronchial lymphoid follicles were all significantly attenuated compared with wild-type animals. Importantly, chronic CS exposure induced airspace enlargement in wild-type mice, while CCR5 KO mice were partially protected against the development of emphysema. However, CCR5 deficiency did not affect CS-induced airway wall remodelling, because chronic CS exposure induced a similar increase in airway wall thickness, smooth muscle mass and peribronchial deposition of collagen and fibronectin in both wild-type and CCR5 KO mice. CONCLUSION: Our data suggest that CCR5 contributes to pulmonary inflammation and to the development of emphysema in response to CS. CCR5 is, however, not implicated in CS-induced airway wall remodelling, suggesting that the mechanisms that lead to airway inflammation are distinct to those responsible for airway remodelling.


Subject(s)
Pulmonary Emphysema/immunology , Pulmonary Emphysema/pathology , Receptors, CCR5/deficiency , Smoking , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/analysis , Cytokines/metabolism , Mice , Mice, Knockout , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Emphysema/genetics , Receptors, CCR5/genetics
13.
Eur Respir J ; 28(1): 102-12, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16540505

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterised by a local pulmonary inflammatory response to respiratory pollutants and by systemic inflammation. Tumour necrosis factor (TNF)-alpha has been implicated in systemic effects of COPD and operates by binding the p55 (R1) and p75 (R2) TNF-alpha receptors. To investigate the contribution of each TNF-alpha receptor in the pathogenesis of COPD, the present study examined the effects of chronic air or cigarette smoke (CS) exposure in TNF-alpha R1 knockout (KO) mice, TNF-alpha R2 KO mice and wild type (WT) mice. CS was found to significantly increase the protein levels of soluble TNF-alpha R1 (by four-fold) and TNF-alpha R2 (by 10-fold) in the bronchoalveolar lavage of WT mice. After 3 months, CS induced a prominent pulmonary inflammatory cell influx in WT and TNF-alpha R1 KO mice. In TNF-alpha R2 KO mice, CS-induced pulmonary inflammation was clearly attenuated. After 6 months, no emphysema was observed in CS-exposed TNF-alpha R2 KO mice in contrast to WT and TNF-alpha R1 KO mice. CS-exposed WT and TNF-alpha R1 KO mice failed to gain weight, whereas the body mass of TNF-alpha R2 KO mice was not affected. These current findings suggest that both tumour necrosis factor-alpha receptors contribute to the pathogenesis of chronic obstructive pulmonary disease, but tumour necrosis factor-alpha receptor-2 is the most active receptor in the development of inflammation, emphysema and systemic weight loss in this murine model of chronic obstructive pulmonary disease.


Subject(s)
Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Receptors, Tumor Necrosis Factor, Type II/physiology , Animals , Apoptosis , Body Weight , Bronchoalveolar Lavage , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smoking , Time Factors , Tumor Necrosis Factor-alpha/metabolism
14.
Pulm Pharmacol Ther ; 19(3): 155-65, 2006.
Article in English | MEDLINE | ID: mdl-16084119

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by airflow limitation, that is not fully reversible, and that is associated with an abnormal inflammatory response of the airways and lungs to noxious particles and gases. The airflow limitation is caused by increased resistance of the small conducting airways and by decreased elastic recoil forces of the lung due to emphysematous destruction of the lung parenchyma. In vivo animal models can help to unravel the molecular and cellular mechanisms underlying the pathogenesis of COPD. Mice represent the most favored animal species with regard to the study of (both innate and adaptive) immune mechanisms, since they offer the opportunity to manipulate gene expression. Several experimental approaches are applied in order to mimic the different traits of COPD in these murine models. Firstly, the tracheal instillation of tissue-degrading enzymes induces emphysema-like lesions in the lung parenchyma, adding further proof to the protease-antiprotease imbalance hypothesis. Secondly, the inhalation of noxious stimuli, including tobacco smoke, sulfur dioxide, nitrogen dioxide, or oxidants such as ozone, may also lead to COPD-like lesions in mice, depending on concentration, duration of exposure and strainspecific genetic susceptibility. Thirdly, in transgenic mice, a specific gene is either overexpressed (non-specific or organ-specific) or selectively depleted (constitutively or conditionally). The study of these transgenic mice, either per se or in combination with the above mentioned experimental approaches (e.g. the inhalation of tobacco smoke), can offer valuable information on both the physiological function of the gene of interest as well as the pathophysiological mechanisms of diseases with complex traits such as COPD.


Subject(s)
Disease Models, Animal , Pulmonary Disease, Chronic Obstructive/physiopathology , Animals , Humans , Mice , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...