Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nature ; 588(7836): 151-156, 2020 12.
Article in English | MEDLINE | ID: mdl-33149305

ABSTRACT

Lymphotoxin ß-receptor (LTßR) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures1,2, which are associated with severe chronic inflammatory diseases that span several organ systems3-6. How LTßR signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LTßR blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LTßR ligands in adaptive and innate immune cells, enhanced non-canonical NF-κB signalling, and enriched LTßR target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LTßR signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LTßR signalling dampened epithelial non-canonical activation of NF-κB, reduced TGFß signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/ß-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LTßR signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures1 and inhibition of apoptosis with tissue-regenerative strategies.


Subject(s)
Lung/drug effects , Lung/physiology , Lymphotoxin beta Receptor/antagonists & inhibitors , Regeneration/drug effects , Signal Transduction/drug effects , Wnt Proteins/agonists , Adaptive Immunity , Aging/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Apoptosis/drug effects , Emphysema/metabolism , Female , Humans , Immunity, Innate , Lung/metabolism , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Smoke/adverse effects , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
3.
J Immunol ; 200(5): 1570-1579, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29352004

ABSTRACT

Epidemiological and experimental studies have shown that exposure to the gastric bacterium Helicobacter pylori, especially in early life, prevents the development of asthma. Recent mouse studies have shown that this protective effect does not require live bacteria and that treatment with an extract of H. pylori in neonates prevents the development of airway inflammation and goblet cell metaplasia. In the current study, the effect of administration of an extract of H. pylori was assessed in a therapeutic study design with application of the extract just prior to allergen challenge. C57BL/6 mice were sensitized and challenged with OVA or house dust mite. Treatment with H. pylori extract just prior to the challenge significantly reduced airway inflammation, as assessed in bronchoalveolar lavage fluid and lung tissue, and reduced airway remodeling, as assessed by goblet cell quantification. These effects were apparent in the OVA model and in the house dust mite model. Injection of H. pylori extract reduced the processing of allergen by dendritic cells in the lungs and mediastinal lymph node. Bone marrow-derived dendritic cells exposed to H. pylori extract were affected with regard to their ability to process Ag. These data show that application of H. pylori extract after sensitization effectively inhibits allergic airway disease.


Subject(s)
Allergens/immunology , Asthma/immunology , Helicobacter pylori/immunology , Hypersensitivity/immunology , Respiratory Hypersensitivity/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Dendritic Cells/immunology , Female , Goblet Cells/immunology , Inflammation/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pyroglyphidae/immunology
4.
Int Arch Allergy Immunol ; 180(1): 1-9, 2019.
Article in English | MEDLINE | ID: mdl-31242493

ABSTRACT

BACKGROUND: An inverse relation between Helicobacter pylori infection and asthma has been shown in epidemiological studies. Infection with H. pylori, or application of an extract of it before or after sensitization, inhibits allergic airway disease in mice. OBJECTIVES: The aim of this study was to investigate the effect of an extract of H. pylori on allergic airway disease induced by repeated allergen exposure in mice that were sensitized and challenged prior to extract application. METHOD: C57BL/6 mice were intranasally (i.n.) sensitized and challenged with house dust mite (HDM). After a minimum of 4 weeks, mice received the H. pylori extract intraperitoneally and were rechallenged i.n. with HDM. Allergen-specific antibodies were measured by ELISA. Cells present in the bronchoalveolar lavage fluid and dendritic cell (DC) subsets in the lung tissue were analyzed by flow cytometry. Tissue inflammation and goblet cell hyperplasia were assessed by histology. Cells of the mediastinal lymph node (mLN) were isolated and in vitro restimulated with HDM or H. pylori extract. RESULTS: Treatment with H. pylori extract before rechallenge reduced allergen-specific IgE, the DC numbers in the tissue, and goblet cell hyperplasia. Cells isolated from mLN of mice treated with the extract produced significantly more IL-10 and IL-17 after in vitro restimulation with HDM. mLN cells of H. pylori-treated mice that were re-exposed to the H. pylori extract produced significantly more interferon gamma. CONCLUSIONS: An extract of H. pylori is effective in reducing mucus production and various features of inflammation in HDM rechallenged mice.


Subject(s)
Allergens/immunology , Antigens, Bacterial/immunology , Goblet Cells/pathology , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Animals , Biomarkers , Biopsy , Cytokines/metabolism , Environmental Exposure , Female , Helicobacter Infections/microbiology , Hyperplasia , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Immunophenotyping , Mice , Pyroglyphidae/immunology
5.
Pulm Pharmacol Ther ; 45: 210-217, 2017 08.
Article in English | MEDLINE | ID: mdl-28687462

ABSTRACT

One of the major goals of asthma therapy is to maintain asthma control and prevent acute exacerbations. Long-acting bronchodilators are regularly used for the treatment of asthma patients and in clinical studies the anti-cholinergic tiotropium has recently been shown to reduce exacerbations in patients with asthma. So far it is unclear how tiotropium exerts this effect. For this purpose, we designed an allergen-driven rechallenge model of allergic airway inflammation in mice, to assess the effectiveness of tiotropium and the long-acting ß-2 adrenoceptor agonist olodaterol on allergen-induced exacerbations of airway disease. Female C57BL/6J mice were sensitized intranasally (i.n.) with 1 µg of house dust mite (HDM) extract followed by a challenge regime (5 consecutive days 10 µg HDM extract i.n.) after one week. Mice were exposed to a secondary challenge five weeks after sensitization and were treated i.n. with different concentrations of tiotropium or olodaterol (1, 10 and 100 µg/kg) or a combination thereof (10 µg/kg each) prior to and during the secondary challenge period. Three days after the last challenge, bronchoalveolar lavage (BAL) fluid and lung tissue were collected for flow cytometry and histologic analysis, respectively. Secondary challenge with HDM extract strongly induced allergic airway disease reflected by inflammatory cell infiltration and goblet cell metaplasia. Treatment with tiotropium, but not with olodaterol reduced tissue inflammation and goblet cell metaplasia in a dose-dependent manner. The combination of tiotropium and olodaterol was more effective in significantly reducing tissue inflammation compared to tiotropium treatment alone, and also led to a decrease in BAL cell counts. These data suggest that in a model of relapsing allergic airway disease tiotropium directly prevents exacerbations by reducing inflammation and mucus production in the airways. In addition, the combination of tiotropium and olodaterol exerts synergistic effects.


Subject(s)
Asthma/drug therapy , Benzoxazines/pharmacology , Bronchodilator Agents/pharmacology , Tiotropium Bromide/pharmacology , Allergens/immunology , Animals , Asthma/immunology , Benzoxazines/administration & dosage , Bronchoalveolar Lavage Fluid , Bronchodilator Agents/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Flow Cytometry , Goblet Cells , Inflammation/drug therapy , Inflammation/immunology , Mice , Mice, Inbred C57BL , Pyroglyphidae/immunology , Tiotropium Bromide/administration & dosage
6.
Am J Respir Cell Mol Biol ; 53(6): 769-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25906418

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by an irreversible loss of lung function and is one of the most prevalent and severe diseases worldwide. A major feature of COPD is emphysema, which is the progressive loss of alveolar tissue. Coactivator-associated arginine methyltransferase-1 (CARM1) regulates histone methylation and the transcription of genes involved in senescence, proliferation, and differentiation. Complete loss of CARM1 leads to disrupted differentiation and maturation of alveolar epithelial type II (ATII) cells. We thus hypothesized that CARM1 regulates the development and progression of emphysema. To address this, we investigated the contribution of CARM1 to alveolar rarefication using the mouse model of elastase-induced emphysema in vivo and small interfering (si)RNA-mediated knockdown in ATII-like LA4 cells in vitro. We demonstrate that emphysema progression in vivo is associated with a time-dependent down-regulation of CARM1. Importantly, elastase-treated CARM1 haploinsufficient mice show significantly increased airspace enlargement (52.5 ± 9.6 µm versus 38.8 ± 5.5 µm; P < 0.01) and lung compliance (2.8 ± 0.32 µl/cm H2O versus 2.4 ± 0.4 µl/cm H2O; P < 0.04) compared with controls. The knockdown of CARM1 in LA4 cells led to decreased sirtuin 1 expression (0.034 ± 0.003 versus 0.022 ± 0.001; P < 0.05) but increased expression of p16 (0.27 ± 0.013 versus 0.31 ± 0.010; P < 0.5) and p21 (0.81 ± 0.088 versus 1.28 ± 0.063; P < 0.01) and higher ß-galactosidase-positive senescent cells (50.57 ± 7.36% versus 2.21 ± 0.34%; P < 0.001) compared with scrambled siRNA. We further demonstrated that CARM1 haploinsufficiency impairs transdifferentiation and wound healing (32.18 ± 0.9512% versus 8.769 ± 1.967%; P < 0.001) of alveolar epithelial cells. Overall, these results reveal a novel function of CARM1 in regulating emphysema development and premature lung aging via alveolar senescence as well as impaired regeneration, repair, and differentiation of ATII cells.


Subject(s)
Alveolar Epithelial Cells/enzymology , Protein-Arginine N-Methyltransferases/physiology , Pulmonary Emphysema/enzymology , Animals , Cell Differentiation , Cell Line , Cellular Senescence , Female , Genetic Predisposition to Disease , Haploinsufficiency , Mice, Inbred C57BL , Pancreatic Elastase , Pulmonary Emphysema/chemically induced
7.
Am J Physiol Lung Cell Mol Physiol ; 307(9): L692-706, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25128521

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by a progressive decline in lung function, caused by exposure to exogenous particles, mainly cigarette smoke (CS). COPD is initiated and perpetuated by an abnormal CS-induced inflammatory response of the lungs, involving both innate and adaptive immunity. Specifically, B cells organized in iBALT structures and macrophages accumulate in the lungs and contribute to CS-induced emphysema, but the mechanisms thereof remain unclear. Here, we demonstrate that B cell-deficient mice are significantly protected against CS-induced emphysema. Chronic CS exposure led to an increased size and number of iBALT structures, and increased lung compliance and mean linear chord length in wild-type (WT) but not in B cell-deficient mice. The increased accumulation of lung resident macrophages around iBALT and in emphysematous alveolar areas in CS-exposed WT mice coincided with upregulated MMP12 expression. In vitro coculture experiments using B cells and macrophages demonstrated that B cell-derived IL-10 drives macrophage activation and MMP12 upregulation, which could be inhibited by an anti-IL-10 antibody. In summary, B cell function in iBALT formation seems necessary for macrophage activation and tissue destruction in CS-induced emphysema and possibly provides a new target for therapeutic intervention in COPD.


Subject(s)
B-Lymphocytes/immunology , Macrophage Activation , Macrophages/immunology , Matrix Metalloproteinase 12/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Emphysema/immunology , Tobacco Smoke Pollution/adverse effects , Animals , Antibodies/pharmacology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Movement , Coculture Techniques , Disease Models, Animal , Gene Expression Regulation , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Matrix Metalloproteinase 12/genetics , Mice , Mice, Knockout , Pancreatic Elastase , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Respiratory Function Tests
8.
Sci Rep ; 8(1): 7547, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765129

ABSTRACT

Prenatal exposure to tobacco smoke is a significant risk-factor for airway disease development. Furthermore, the high prevalence of pregnant smoking women requires the establishment of strategies for offspring lung protection. Therefore, we here aimed to understand the molecular mechanism of how prenatal smoke exposure affects fetal lung development. We used a mouse model recapitulating clinical findings of prenatally exposed children, where pregnant mice were exposed to smoke until c-section or spontaneous delivery, and offspring weight development and lung function was monitored. Additionally, we investigated pulmonary transcriptome changes in fetal lungs (GD18.5) by mRNA/miRNA arrays, network analyses and qPCR. The results demonstrated that prenatally exposed mice showed intrauterine and postnatal growth retardation, and impaired lung function. 1340 genes and 133 miRNAs were found to be significantly dysregulated by in utero smoke exposure, and we identified Insulin-like growth factor 1 (Igf1) as a top hierarchical node in a network analysis. Moreover, Igf1 mRNA was increased in female murine offspring and in prenatally exposed children. These findings suggest that prenatal smoking is associated with a dysregulation of several genes, including Igf1 in a sex-specific manner. Thus, our results could represent a novel link between smoke exposure, abberant lung development and impaired lung function.


Subject(s)
Gene Expression Profiling/methods , Insulin-Like Growth Factor I/genetics , Lung/embryology , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/genetics , Tobacco Smoke Pollution/adverse effects , Adolescent , Animals , Child , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Lung/cytology , Lung/physiopathology , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Sex Characteristics
9.
J Exp Med ; 214(1): 143-163, 2017 01.
Article in English | MEDLINE | ID: mdl-27979969

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide. One main pathological feature of COPD is the loss of functional alveolar tissue without adequate repair (emphysema), yet the underlying mechanisms are poorly defined. Reduced WNT-ß-catenin signaling is linked to impaired lung repair in COPD; however, the factors responsible for attenuating this pathway remain to be elucidated. Here, we identify a canonical to noncanonical WNT signaling shift contributing to COPD pathogenesis. We demonstrate enhanced expression of noncanonical WNT-5A in two experimental models of COPD and increased posttranslationally modified WNT-5A in human COPD tissue specimens. WNT-5A was increased in primary lung fibroblasts from COPD patients and induced by COPD-related stimuli, such as TGF-ß, cigarette smoke (CS), and cellular senescence. Functionally, mature WNT-5A attenuated canonical WNT-driven alveolar epithelial cell wound healing and transdifferentiation in vitro. Lung-specific WNT-5A overexpression exacerbated airspace enlargement in elastase-induced emphysema in vivo. Accordingly, inhibition of WNT-5A in vivo attenuated lung tissue destruction, improved lung function, and restored expression of ß-catenin-driven target genes and alveolar epithelial cell markers in the elastase, as well as in CS-induced models of COPD. We thus identify a novel essential mechanism involved in impaired mesenchymal-epithelial cross talk in COPD pathogenesis, which is amenable to therapy.


Subject(s)
Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Wnt Signaling Pathway/physiology , Wnt-5a Protein/physiology , Animals , Cells, Cultured , Emphysema/etiology , Female , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , beta Catenin/physiology
10.
Oncotarget ; 7(21): 30068-83, 2016 May 24.
Article in English | MEDLINE | ID: mdl-26284585

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is related to an abnormal chronic inflammatory response of the lung to mainly cigarette smoke (CS) and the disease risk is increased in aged individuals. The source of this chronic inflammation is due to the repeated and progressive activation of immune cells. We hypothesize that in a chronic CS-induced mouse model, the predisposition to COPD pathogenesis in aged mice is characterized by an elevated immune response compared to young animals. We measured several characteristics of COPD in young and old mice (2 and 12 months of age) exposed to CS for 3 months. CS-exposed aged mice exhibited increased lung compliance (0.061 ± 0.008 vs. 0.055 ± 0.006 ml/cm H2O, p < 0.01), emphysema development (35.36 ± 0.71 vs. 25.31 ± 0.005 µm; p < 0.01) and airway remodeling (2.15 ± 0.37 vs. 1.09 ± 0.64 µm3/µm2; p < 0.01) compared to control animals, which was not seen in CS-exposed young mice. Quantification of lung tissue inflammation revealed a significantly greater volume of inducible bronchus-associated lymphoid tissue structures in aged mice after CS exposure (5.94 ± 2.89 vs. 2.37 ± 1.69 µm3/µm2; p < 0.01). Our results indicate that age-induced lung inflammation is further elevated after CS exposure in old mice, potentially via an age-induced change in immune cell susceptibility to CS thereby accelerating the pathophysiological hallmarks of COPD.


Subject(s)
Disease Susceptibility/immunology , Inflammation/immunology , Lymphocyte Activation , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/adverse effects , Age Factors , Airway Remodeling , Animals , B-Lymphocytes/immunology , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Disease Models, Animal , Disease Susceptibility/chemically induced , Female , Humans , Inflammation/chemically induced , Lung/cytology , Lung/pathology , Lung Compliance/immunology , Mice , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive/etiology , Smoke/adverse effects , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology , Nicotiana/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL