Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Respir Res ; 23(1): 180, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35804363

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a diffuse parenchymal lung disease characterized by exuberant deposition of extracellular matrix (ECM) proteins in the lung interstitium, which contributes to substantial morbidity and mortality in IPF patients. Matrix metalloproteinases (MMPs) are a large family of zinc-dependent endopeptidases, many of which have been implicated in the regulation of ECM degradation in lung fibrosis. However, the roles of MMP-2 and -9 (also termed gelatinases A and B) have not yet been explored in lung fibrosis in detail. METHODS: AdTGF-ß1 was applied via orotracheal routes to the lungs of WT, MMP-2 KO, MMP-9 KO and MMP-2/-9 dKO mice on day 0 to induce lung fibrosis. Using hydroxyproline assay, FlexiVent based lung function measurement, histopathology, western blot and ELISA techniques, we analyzed MMP-2 and MMP-9 levels in BAL fluid and lung, collagen contents in lung and lung function in mice on day 14 and 21 post-treatment. RESULT: IPF lung homogenates exhibited significantly increased levels of MMP-2 and MMP-9, relative to disease controls. Enzymatically active MMP-2 and MMP-9 was increased in lungs of mice exposed to adenoviral TGF-ß1, suggesting a role for these metalloproteinases in lung fibrogenesis. However, we found that neither MMP-2 or MMP-9 nor combined MMP-2/-9 deletion had any effect on experimental lung fibrosis in mice. CONCLUSION: Together, our data strongly suggest that both gelatinases MMP-2 and MMP-9 play only a subordinate role in experimental lung fibrosis in mice.


Subject(s)
Idiopathic Pulmonary Fibrosis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/metabolism , Mice
2.
J Immunol ; 208(5): 1259-1271, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35149532

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an irreversible, age-related diffuse parenchymal lung disease of poorly defined etiology. Many patients with IPF demonstrate distinctive lymphocytic interstitial infiltrations within remodeled lung tissue with uncertain pathogenetic relevance. Histopathological examination of explant lung tissue of patients with IPF revealed accentuated lymphoplasmacellular accumulations in close vicinity to, or even infiltrating, remodeled lung tissue. Similarly, we found significant accumulations of B cells interfused with T cells within remodeled lung tissue in two murine models of adenoviral TGF-ß1 or bleomycin (BLM)-induced lung fibrosis. Such B cell accumulations coincided with significantly increased lung collagen deposition, lung histopathology, and worsened lung function in wild-type (WT) mice. Surprisingly, B cell-deficient µMT knockout mice exhibited similar lung tissue remodeling and worsened lung function upon either AdTGF-ß1 or BLM as for WT mice. Comparative transcriptomic profiling of sorted B cells collected from lungs of AdTGF-ß1- and BLM-exposed WT mice identified a large set of commonly regulated genes, but with significant enrichment observed for Gene Ontology terms apparently not related to lung fibrogenesis. Collectively, although we observed B cell accumulations in lungs of IPF patients as well as two experimental models of lung fibrosis, comparative profiling of characteristic features of lung fibrosis between WT and B cell-deficient mice did not support a major involvement of B cells in lung fibrogenesis in mice.


Subject(s)
B-Lymphocytes/immunology , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Transforming Growth Factor beta1/metabolism , Animals , Bleomycin/toxicity , Collagen/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parenchymal Tissue/pathology , T-Lymphocytes/immunology
3.
Am J Physiol Lung Cell Mol Physiol ; 320(3): L377-L392, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33296268

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease (ILD) associated with high morbidity and mortality. Patients with ILD frequently develop an acute exacerbation of their disease, which may be triggered by viral and/or bacterial infections. Prostaglandin E2 (PGE2) is an eicosanoid released in a cyclooxygenase-2 (COX2)-dependent manner and is considered to contribute to regulation of lung fibrosis. However, its role in infection-induced exacerbation of lung fibrosis is poorly defined. We found significantly increased levels of PGE2 in lung tissue of patients with ILD. Increased levels of PGE2 were also found in lung tissue of mice with AdTGF-ß1-induced lung fibrosis and even more so in Streptococcus pneumoniae exacerbated lung fibrosis. Type II alveolar epithelial cells (AT II cells) and alveolar macrophages (AM) contributed to PGE2 release during exacerbating fibrosis. Application of parecoxib to inhibit PGE2 synthesis ameliorated lung fibrosis, whereas intratracheal application of PGE2 worsened lung fibrosis in mice. Both interventions had no effect on S. pneumoniae-exacerbated lung fibrosis. Together, we found that the COX2-PGE2 axis has dual roles in fibrosis that may offset each other: PGE2 helps resolve infection/attenuate inflammation in fibrosis exacerbation but accentuates TGF-ß/AT II cell-mediated fibrosis. These data support the efficacy of COX/PGE2 interventions in the setting of non-exacerbating lung fibrosis.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Pneumonia, Pneumococcal/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Streptococcus pneumoniae/metabolism , Alveolar Epithelial Cells/microbiology , Alveolar Epithelial Cells/pathology , Animals , Disease Models, Animal , Female , Isoxazoles/pharmacology , Mice , Pneumonia, Pneumococcal/pathology , Pulmonary Fibrosis/microbiology , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta/metabolism
4.
J Immunol ; 204(9): 2429-2438, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213566

ABSTRACT

Patients with idiopathic pulmonary fibrosis (IPF) can experience life-threatening episodes of acute worsening of their disease, termed acute exacerbation of IPF, which may be caused by bacterial and/or viral infections. The potential for regulatory T cells (Tregs) to limit disease progression in bacterially triggered fibrosis exacerbation has not been explored so far. In the current study, we show that the number of Tregs was significantly increased in mice with established AdTGF-ß1-induced lung fibrosis and further increased in mice with pneumococcal infection-induced lung fibrosis exacerbation. Diphtheria toxin-induced depletion of Tregs significantly worsened infection-induced fibrosis exacerbation as determined by increased lung collagen deposition, lung histology, and elevated pulmonary Th1/Th2 cytokine levels. Conversely, IL-2 complex-induced Treg expansion in wild-type mice with established lung fibrosis completely inhibited pneumococcal infection-induced fibrosis exacerbation as efficaciously as antibiotic treatment while preserving lung antibacterial immunity in mice. Collectively, these findings demonstrate the efficacy of Tregs as "silencers," suppressing infection-induced exacerbation of lung fibrosis in mice, and their expansion may offer a novel adjunctive treatment to limit acute exacerbations in patients with IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Disease Models, Animal , Disease Progression , Female , Idiopathic Pulmonary Fibrosis/microbiology , Interleukin-2/immunology , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Pneumococcal Infections/microbiology , Th1 Cells/immunology , Th17 Cells/immunology
5.
Eur Respir J ; 55(6)2020 06.
Article in English | MEDLINE | ID: mdl-32165401

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a complex disease of unknown aetiology, which makes drug development challenging. Single administration of bleomycin directly to the lungs of mice is a widely used experimental model for studying pulmonary fibrogenesis and evaluating the effect of therapeutic antifibrotic strategies. The model works by inducing an early inflammatory phase, which transitions into fibrosis after 5-7 days. This initial inflammation makes therapeutic timing crucial. To accurately assess antifibrotic efficacy, the intervention should inhibit fibrosis without impacting early inflammation.Studies published between 2008 and 2019 using the bleomycin model to investigate pulmonary fibrosis were retrieved from PubMed, and study characteristics were analysed. Intervention-based studies were classified as either preventative (starting <7 days after bleomycin installation) or therapeutic (>7 days). In addition, studies were cross-referenced with current major clinical trials to assess the availability of preclinical rationale.A total of 976 publications were evaluated. 726 investigated potential therapies, of which 443 (61.0%) were solely preventative, 166 (22.9%) were solely therapeutic and 105 (14.5%) were both. Of the 443 preventative studies, only 70 (15.8%) characterised inflammation during the model's early inflammatory phase. In the reported 145 IPF clinical trials investigating 93 compounds/combinations, only 25 (26.9%) interventions had any preclinical data on bleomycin available on PubMed.Since 2008, we observed a shift (from <5% to 37.4%) in the number of studies evaluating drugs in the therapeutic setting in the bleomycin model. While this shift is encouraging, further characterisation of early inflammation and appropriate preclinical therapeutic testing are still needed. This will facilitate fruitful drug development in IPF, and more therapeutic strategies for patients with this devastating disease.


Subject(s)
Bleomycin , Disease Models, Animal , Idiopathic Pulmonary Fibrosis , Animals , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Mice
6.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-32102985

ABSTRACT

Neutrophils are the most abundant inflammatory cells at the earliest stages of wound healing and play important roles in wound repair and fibrosis. Formyl peptide receptor 1 (FPR-1) is abundantly expressed on neutrophils and has been shown to regulate their function, yet the importance of FPR-1 in fibrosis remains ill defined. FPR-1-deficient (fpr1-/-) mice were protected from bleomycin-induced pulmonary fibrosis but developed renal and hepatic fibrosis normally. Mechanistically, we observed a failure to effectively recruit neutrophils to the lungs of fpr1-/- mice, whereas neutrophil recruitment was unaffected in the liver and kidney. Using an adoptive transfer model we demonstrated that the defect in neutrophil recruitment to the lung was intrinsic to the fpr1-/- neutrophils, as C57BL/6 neutrophils were recruited normally to the damaged lung in fpr1-/- mice. Finally, C57BL/6 mice in which neutrophils had been depleted were protected from pulmonary fibrosis. In conclusion, FPR-1 and FPR-1 ligands are required for effective neutrophil recruitment to the damaged lung. Failure to recruit neutrophils or depletion of neutrophils protects from pulmonary fibrosis.


Subject(s)
Neutrophil Infiltration/physiology , Pulmonary Fibrosis/physiopathology , Receptors, Formyl Peptide/physiology , Animals , Bleomycin/toxicity , Humans , Ligands , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism
7.
Int J Biochem Cell Biol ; 116: 105595, 2019 11.
Article in English | MEDLINE | ID: mdl-31473260

ABSTRACT

Fibroblasts are considered major contributors to the process of fibrogenesis and the progression of matrix deposition and tissue distortion in fibrotic diseases such as Pulmonary Fibrosis. Recent discovery of the fibrocyte, a circulating possible precursor cell to the tissue fibroblast in fibrosis, has raised issues regarding the characterization of fibrocytes with respect to their morphology, growth characteristics in vitro, their biological role in vivo and their potential utility as a biomarker and/ or treatment target in various human diseases. Characterization studies of the fibrocyte continue as does emerging conflicting data concerning the relationship to or with the lung fibroblast. The source of signals that direct the traffic of these cells, as well as their response to therapeutic intervention with newly available drugs, bring new insights to the understanding of this cell type. The identification of exosomes from fibrocytes that can affect resident fibroblast activities suggest mechanisms of their influence on pathogenesis. Moreover, interesting comparisons with other pathologies are emerging involving the influence of circulating mesenchymal precursor cells on tissue responses.


Subject(s)
Exosomes/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Pulmonary Fibrosis/genetics , Animals , Biomarkers/metabolism , Cell Differentiation , Cytokines/genetics , Cytokines/metabolism , Exosomes/chemistry , Exosomes/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/pathology , Fibroblasts/pathology , Gene Expression , Humans , Lung/metabolism , Lung/pathology , Mesenchymal Stem Cells/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
8.
Thorax ; 74(10): 947-957, 2019 10.
Article in English | MEDLINE | ID: mdl-31076499

ABSTRACT

RATIONALE: Dendritic cells (DC) accumulate in the lungs of patients with idiopathic lung fibrosis, but their pathogenetic relevance is poorly defined. OBJECTIVES: To assess the role of the FMS-like tyrosine kinase-3 ligand (Flt3L)-lung dendritic cell axis in lung fibrosis. MEASUREMENTS AND MAIN RESULTS: We demonstrate in a model of adenoviral gene transfer of active TGF-ß1 that established lung fibrosis was accompanied by elevated serum Flt3L levels and subsequent accumulation of CD11bpos DC in the lungs of mice. Patients with idiopathic pulmonary fibrosis also demonstrated increased levels of Flt3L protein in serum and lung tissue and accumulation of lung DC in explant subpleural lung tissue specimen. Mice lacking Flt3L showed significantly reduced lung DC along with worsened lung fibrosis and reduced lung function relative to wild-type (WT) mice, which could be inhibited by administration of recombinant Flt3L. Moreover, therapeutic Flt3L increased numbers of CD11bpos DC and improved lung fibrosis in WT mice exposed to AdTGF-ß1. In this line, RNA-sequencing analysis of CD11bpos DC revealed significantly enriched differentially expressed genes within extracellular matrix degrading enzyme and matrix metalloprotease gene clusters. In contrast, the CD103pos DC subset did not appear to be involved in pulmonary fibrogenesis. CONCLUSIONS: We show that Flt3L protein and numbers of lung DC are upregulated in mice and humans during pulmonary fibrogenesis, and increased mobilisation of lung CD11bpos DC limits the severity of lung fibrosis in mice. The current study helps to inform the development of DC-based immunotherapy as a novel intervention against lung fibrosis in humans.


Subject(s)
Collagen/metabolism , Dendritic Cells/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Dendritic Cells/pathology , Disease Models, Animal , Ligands , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Lab Invest ; 99(6): 830-852, 2019 06.
Article in English | MEDLINE | ID: mdl-30700849

ABSTRACT

In human idiopathic pulmonary fibrosis (IPF), collapse of distal airspaces occurs in areas of the lung not (yet) remodeled. Mice lungs overexpressing transforming growth factor-ß1 (TGF-ß1) recapitulate this abnormality: surfactant dysfunction results in alveolar collapse preceding fibrosis and loss of alveolar epithelial type II (AE2) cells' apical membrane surface area. Here we examined whether surfactant dysfunction-related alveolar collapse due to TGF-ß1 overexpression is linked to septal wall remodeling and AE2 cell abnormalities. Three and 6 days after gene transfer of TGF-ß1, mice received either intratracheal surfactant (Surf-groups: Curosurf®, 100 mg/kg bodyweight) or 0.9% NaCl (Saline-groups). On days 7 (D7) and 14 (D14), lung mechanics were assessed followed by design-based stereology at light and electron microscopic level to quantify structures. Compared with Saline, Surf showed significantly improved tissue elastance, increased numbers of open alveoli, as well as reduced alveolar size heterogeneity on D7. Deterioration in lung mechanics was highly correlated to the loss of open alveoli. On D14, lung mechanics, number of open alveoli, and alveolar size heterogeneity remained significantly improved in the Surf-group. Volumes of extracellular matrix and collagen fibrils in septal walls were significantly reduced, whereas the apical membrane surface area of AE2 cells was increased in Surf compared with Saline. In remodeled tissue with collapsed alveoli, three-dimensional reconstruction of AE2 cells based on scanning electron microscopy array tomography revealed that AE2 cells were trapped without contact to airspaces in the TGF-ß1 mouse model. Similar observations were made in human IPF. Based on correlation analyses, the number of open alveoli and of alveolar size heterogeneity were highly linked with the loss of apical membrane surface area of AE2 cells and deposition of collagen fibrils in septal walls on D14. In conclusion, surfactant replacement therapy stabilizes alveoli and prevents extracellular matrix deposition in septal walls in the TGF-ß1 model.


Subject(s)
Alveolar Epithelial Cells/drug effects , Pulmonary Fibrosis/prevention & control , Pulmonary Surfactants/therapeutic use , Airway Remodeling , Alveolar Epithelial Cells/ultrastructure , Animals , Disease Models, Animal , Female , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology , Pulmonary Surfactants/pharmacology , Respiratory Mechanics , Transforming Growth Factor beta1
10.
Thorax ; 74(5): 455-465, 2019 05.
Article in English | MEDLINE | ID: mdl-30808717

ABSTRACT

BACKGROUND: The role of mast cells accumulating in idiopathic pulmonary fibrosis (IPF) lungs is unknown. OBJECTIVES: We investigated the effect of fibrotic extracellular matrix (ECM) on mast cells in experimental and human pulmonary fibrosis. RESULTS: In IPF lungs, mast cell numbers were increased and correlated with disease severity (control vs 60%90% vs 60%90% vs FVC<60%, mean difference=-268.6, 95% CI of difference -441.0 to -96.17, p=0.0007). Plasma tryptase levels were increased in IPF and negatively correlated with FVC (control vs FVC<60%, mean difference=-17.12, 95% CI of difference -30.02 to -4.22, p=0.006: correlation curves R=-0.045, p=0.025). In a transforming growth factor (TGF)-ß1-induced pulmonary fibrosis model, chymase-positive and tryptase-positive mast cells accumulated in fibrotic lung. Lung tissue was decellularised and reseeded with bone marrow or peritoneum-derived mast cells; cells on fibrotic ECM released more TGF-ß1 compared with normal ECM (active TGF-ß1: bone marrow-derived mast cell (BMMC)-DL vs BMMC-TGF-ß1 p=0.0005, peritoneal mast cell (PMC)-DL vs PMC-TGF-ß1 p=0.0003, total TGF-ß1: BMMC-DL vs BMMC-TGF-ß1 p=0.013, PMC-DL vs PMC-TGF-ß1 p=0.001). Mechanical stretch of lungs caused mast cell degranulation; mast cell stabilisers inhibited degranulation (histamine: cont vs doxantrazole p=0.004, ß-hexosaminidase: cont vs doxantrazole, mean difference=1.007, 95% CI of difference 0.2700 to 1.744, p=0.007) and TGF-ß1 activation (pSmad2/Smad2: cont vs dox p=0.006). Cromoglycate attenuated pulmonary fibrosis in rats (collagen: phosphate-buffered saline (PBS) vs cromoglycate p=0.036, fibrotic area: PBS vs cromoglycate p=0.031). CONCLUSION: This study suggests that mast cells may contribute to the progression of pulmonary fibrosis.


Subject(s)
Cell Degranulation , Lung/pathology , Mast Cells/physiology , Pulmonary Fibrosis/metabolism , Stress, Mechanical , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Lung/metabolism , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
Eur Respir J ; 52(2)2018 08.
Article in English | MEDLINE | ID: mdl-29976656

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with an unknown cause. Two drugs, nintedanib and pirfenidone, have been shown to slow, but not stop, disease progression. Pulmonary hypertension (PH) is a frequent complication in IPF patients and is associated with poor prognosis. Macitentan is a dual endothelin receptor antagonist that is approved for pulmonary arterial hypertension treatment. We hypothesised that using macitentan to treat animals with pulmonary fibrosis induced by adenoviral vector encoding biologically active transforming growth factor-ß1 (AdTGF-ß1) would improve the PH caused by chronic lung disease and would limit the progression of fibrosis.Rats (Sprague Dawley) which received AdTGF-ß1 were treated by daily gavage of macitentan (100 mg·kg-1·day-1), pirfenidone (0.5% food admix) or a combination from day 14 to day 28. Pulmonary artery pressure (PAP) was measured before the rats were killed, and fibrosis was subsequently evaluated by morphometric measurements and hydroxyproline analysis.AdTGF-ß1 induced pulmonary fibrosis associated with significant PH. Macitentan reduced the increase in PAP and both macitentan and pirfenidone stopped fibrosis progression from day 14 to day 28. Macitentan protected endothelial cells from myofibroblast differentiation and apoptosis whereas pirfenidone only protected against fibroblast-to-myofibroblast differentiation. Both drugs induced apoptosis of differentiated myofibroblasts in vitro and in vivoOur results demonstrate that dual endothelin receptor antagonism was effective in both PH and lung fibrosis whereas pirfenidone only affected fibrosis.


Subject(s)
Hypertension, Pulmonary/drug therapy , Myofibroblasts/drug effects , Pulmonary Fibrosis/pathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Differentiation/drug effects , Disease Progression , Female , Humans , Hypertension, Pulmonary/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Male , Myofibroblasts/metabolism , Pulmonary Fibrosis/chemically induced , Pyridones/pharmacology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/pharmacology
12.
Elife ; 72018 07 03.
Article in English | MEDLINE | ID: mdl-29966587

ABSTRACT

Matrix stiffening with downstream activation of mechanosensitive pathways is strongly implicated in progressive fibrosis; however, pathologic changes in extracellular matrix (ECM) that initiate mechano-homeostasis dysregulation are not defined in human disease. By integrated multiscale biomechanical and biological analyses of idiopathic pulmonary fibrosis lung tissue, we identify that increased tissue stiffness is a function of dysregulated post-translational collagen cross-linking rather than any collagen concentration increase whilst at the nanometre-scale collagen fibrils are structurally and functionally abnormal with increased stiffness, reduced swelling ratio, and reduced diameter. In ex vivo and animal models of lung fibrosis, dual inhibition of lysyl oxidase-like (LOXL) 2 and LOXL3 was sufficient to normalise collagen fibrillogenesis, reduce tissue stiffness, and improve lung function in vivo. Thus, in human fibrosis, altered collagen architecture is a key determinant of abnormal ECM structure-function, and inhibition of pyridinoline cross-linking can maintain mechano-homeostasis to limit the self-sustaining effects of ECM on progressive fibrosis.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Collagen/chemistry , Enzyme Inhibitors/pharmacology , Extracellular Matrix/chemistry , Pulmonary Fibrosis/drug therapy , Reticulin/chemistry , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Amino Acids/chemistry , Animals , Biomechanical Phenomena , Case-Control Studies , Collagen/metabolism , Collagen/ultrastructure , Cross-Linking Reagents/chemistry , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Homeostasis/genetics , Humans , Lung/metabolism , Lung/pathology , Mechanotransduction, Cellular , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/antagonists & inhibitors , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein-Lysine 6-Oxidase , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Reticulin/metabolism , Reticulin/ultrastructure , Structure-Activity Relationship , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology
13.
Immunobiology ; 223(3): 331-341, 2018 03.
Article in English | MEDLINE | ID: mdl-29089144

ABSTRACT

Granulocyte macrophage-colony stimulating factor (GM-CSF) is a hematopoietic growth factor involved in differentiation, survival and activation of myeloid and non-myeloid cells with important implications for lung antibacterial immunity. Here we examined the effect of pulmonary adenoviral vector-mediated delivery of GM-CSF (AdGM-CSF) on anti-mycobacterial immunity in M. bovis BCG infected mice. Exposure of M. bovis BCG infected mice to AdGM-CSF either applied on 6h, or 6h and 7days post-infection substantially increased alveolar recruitment of iNOS and IL-12 expressing macrophages, and significantly increased accumulation of IFNγpos T cells and particularly regulatory T cells (Tregs). This was accompanied by significantly reduced mycobacterial loads in the lungs of mice. Importantly, diphtheria toxin-induced depletion of Tregs did not influence mycobacterial loads, but accentuated immunopathology in AdGM-CSF-exposed mice infected with M. bovis BCG. Together, the data demonstrate that AdGM-CSF therapy improves lung protective immunity against M. bovis BCG infection in mice independent of co-recruited Tregs, which however critically contribute to limit lung immunopathology in BCG-infected mice. These data may be relevant to the development of immunomodulatory strategies to limit immunopathology-based lung injury in tuberculosis in humans.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung/physiology , Macrophages/immunology , Mycobacterium bovis/physiology , T-Lymphocytes, Regulatory/immunology , Tuberculosis/immunology , Adenoviridae/genetics , Animals , Cell Movement , Cells, Cultured , Gene Transfer Techniques , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tuberculosis/veterinary
14.
Am J Respir Cell Mol Biol ; 58(4): 461-470, 2018 04.
Article in English | MEDLINE | ID: mdl-29115860

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by excessive deposition of extracellular matrix (ECM) in the lung parenchyma. The abnormal ECM deposition slowly overtakes normal lung tissue, disturbing gas exchange and leading to respiratory failure and death. ECM cross-linking and subsequent stiffening is thought to be a major contributor of disease progression and also promotes the activation of transforming growth factor (TGF)-ß1, one of the main profibrotic growth factors. Lysyl oxidase-like (LOXL) 1 belongs to the cross-linking enzyme family and has been shown to be up-regulated in active fibrotic regions of bleomycin-treated mice and patients with IPF. We demonstrate in this study that LOXL1-deficient mice are protected from experimental lung fibrosis induced by overexpression of TGF-ß1 using adenoviral (Ad) gene transfer (AdTGF-ß1). The lack of LOXL1 prevented accumulation of insoluble cross-linked collagen in the lungs, and therefore limited lung stiffness after AdTGF-ß1. In addition, we applied mechanical stretch to lung slices from LOXL1+/+ and LOXL1-/- mice treated with AdTGF-ß1. Lung stiffness (Young's modulus) of LOXL1-/- lung slices was significantly lower compared with LOXL1+/+ lung slices. Moreover, the release of activated TGF-ß1 after mechanical stretch was significantly lower in LOXL1-/- mice compared with LOXL1+/+ mice after AdTGF-ß1. These data support the concept that cross-linking enzyme inhibition represents an interesting therapeutic target for drug development in IPF.


Subject(s)
Adenoviridae/genetics , Amino Acid Oxidoreductases/deficiency , Collagen/metabolism , Gene Transfer Techniques , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/enzymology , Transforming Growth Factor beta1/genetics , Adenoviridae/metabolism , Amino Acid Oxidoreductases/genetics , Animals , Disease Models, Animal , Elastic Modulus , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Lung/physiopathology , Lung Compliance , Mechanotransduction, Cellular , Mice, Knockout , Pulmonary Stretch Receptors/metabolism , Transforming Growth Factor beta1/biosynthesis , Up-Regulation
15.
PLoS One ; 11(8): e0158047, 2016.
Article in English | MEDLINE | ID: mdl-27494713

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating, progressive disease with poor survival rates and limited treatment options. Upregulation of αvß6 integrins within the alveolar epithelial cells is a characteristic feature of IPF and correlates with poor patient survival. The pro-fibrotic cytokine TGFß1 can upregulate αvß6 integrin expression but the molecular mechanisms driving this effect have not previously been elucidated. We confirm that stimulation with exogenous TGFß1 increases expression of the integrin ß6 subunit gene (ITGB6) and αvß6 integrin cell surface expression in a time- and concentration-dependent manner. TGFß1-induced ITGB6 expression occurs via transcriptional activation of the ITGB6 gene, but does not result from effects on ITGB6 mRNA stability. Basal expression of ITGB6 in, and αvß6 integrins on, lung epithelial cells occurs via homeostatic αvß6-mediated TGFß1 activation in the absence of exogenous stimulation, and can be amplified by TGFß1 activation. Fundamentally, we show for the first time that TGFß1-induced ITGB6 expression occurs via canonical Smad signalling since dominant negative constructs directed against Smad3 and 4 inhibit ITGB6 transcriptional activity. Furthermore, disruption of a Smad binding site at -798 in the ITGB6 promoter abolishes TGFß1-induced ITGB6 transcriptional activity. Using chromatin immunoprecipitation we demonstrate that TGFß1 stimulation of lung epithelial cells results in direct binding of Smad3, and Smad4, to the ITGB6 gene promoter within this region. Finally, using an adenoviral TGFß1 over-expression model of pulmonary fibrosis we demonstrate that Smad3 is crucial for TGFß1-induced αvß6 integrin expression within the alveolar epithelium in vivo. Together, these data confirm that a homeostatic, autocrine loop of αvß6 integrin activated TGFß1-induced ITGB6 gene expression regulates epithelial basal αvß6 integrin expression, and demonstrates that this occurs via Smad-dependent transcriptional regulation at a single Smad binding site in the promoter of the ß6 subunit gene. Active TGFß1 amplifies this pathway both in vitro and in vivo, which may promote fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/pathology , Integrin beta Chains/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Binding Sites , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Integrin beta Chains/genetics , Integrins/genetics , Integrins/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Promoter Regions, Genetic , RNA Stability/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
J Pathol ; 240(2): 197-210, 2016 10.
Article in English | MEDLINE | ID: mdl-27425145

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibroblast and myofibroblast proliferation, and extensive deposition of extracellular matrix (ECM). Fibroblast growth factor-1 (FGF-1) belongs to the FGF family and has been shown to inhibit fibroblast collagen production and differentiation into myofibroblasts, and revert epithelial-mesenchymal transition by inhibiting TGF-ß1 signalling pathways. However, the precise role of FGF-1 in pulmonary fibrosis has not yet been elucidated. In this study, we explore the mechanisms underlying the anti-fibrogenic effect of FGF-1 in pulmonary fibrosis in vitro and in vivo by prolonged transient overexpression of FGF-1 (AdFGF-1) and TGF-ß1 (AdTGF-ß1) using adenoviral vectors. In vivo, FGF-1 overexpression markedly attenuated TGF-ß1-induced pulmonary fibrosis in rat lungs when given both concomitantly, or delayed, by enhancing proliferation and hyperplasia of alveolar epithelial cells (AECs). AdFGF-1 also attenuated the TGF-ß1 signalling pathway and induced FGFR1 expression in AECs. In vitro, AdFGF-1 prevented the increase in α-SMA and the decrease in E-cadherin induced by AdTGF-ß1 in normal human lung fibroblasts, primary human pulmonary AECs, and A549 cells. Concomitantly, AdTGF-ß1-induced Smad2 phosphorylation was significantly reduced by AdFGF-1 in both cell types. AdFGF-1 also attenuated the increase in TGFßR1 protein and mRNA levels in fibroblasts. In AECs, AdFGF-1 decreased TGFßR1 protein by favouring TGFßR1 degradation through the caveolin-1/proteasome pathway. Furthermore, FGFR1 expression was increased in AECs, whereas it was decreased in fibroblasts. In serum of IPF patients, FGF-1 levels were increased compared to controls. Interestingly, FGF-1 expression was restricted to areas of AEC hyperplasia, but not α-SMA-positive areas in IPF lung tissue. Our results demonstrate that FGF-1 may have preventative and therapeutic effects on TGF-ß1-driven pulmonary fibrosis via inhibiting myofibroblast differentiation, inducing AEC proliferation, regulating TGF-ß1 signalling by controlling TGFßR1 expression and degradation, and regulating FGFR1 expression. Thus, modulating FGF-1 signalling represents a potential therapy for the treatment of pulmonary fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Epithelial Cells/metabolism , Fibroblast Growth Factor 1/genetics , Fibroblasts/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/genetics , Animals , Cadherins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Epithelial Cells/pathology , Female , Fibroblast Growth Factor 1/metabolism , Fibroblasts/pathology , Humans , Phosphorylation , Proteasome Endopeptidase Complex , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Smad2 Protein/metabolism , Up-Regulation
17.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1260-71, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27106287

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) is involved in regulation of cellular proliferation, differentiation, and fibrogenesis, inducing myofibroblast migration and increasing extracellular matrix synthesis. Here, TGF-ß1 effects on pulmonary structure and function were analyzed. Adenovirus-mediated gene transfer of TGF-ß1 in mice lungs was performed and evaluated by design-based stereology, invasive pulmonary function testing, and detailed analyses of the surfactant system 1 and 2 wk after gene transfer. After 1 wk decreased static compliance was linked with a dramatic alveolar derecruitment without edema formation or increase in the volume of septal wall tissue or collagen fibrils. Abnormally high surface tension correlated with downregulation of surfactant proteins B and C. TTF-1 expression was reduced, and, using PLA (proximity ligand assay) technology, we found Smad3 and TTF-1 forming complexes in vivo, which are normally translocated into the nucleus of the alveolar epithelial type II cells (AE2C) but in the presence of TGF-ß1 remain in the cytoplasm. AE2C show altered morphology, resulting in loss of total apical surface area per lung and polarity. These changes of AE2C were progressive 2 wk after gene transfer and correlated with lung compliance. Although static lung compliance remained low, the volume of septal wall tissue and collagen fibrils increased 2 wk after gene transfer. In this animal model, the primary effect of TGF-ß1 signaling in the lung is downregulation of surfactant proteins, high surface tension, alveolar derecruitment, and mechanical stress, which precede fibrotic tissue remodeling and progressive loss of AE2C polarity. Initial TTF-1 dysfunction is potentially linked to downregulation of surfactant proteins.


Subject(s)
Lung Diseases, Interstitial/metabolism , Transforming Growth Factor beta1/genetics , Airway Remodeling , Alveolar Epithelial Cells/metabolism , Animals , Cell Polarity , DNA-Binding Proteins/metabolism , Down-Regulation , Fibrosis , Gene Expression , Lung/metabolism , Lung/pathology , Lung Diseases, Interstitial/physiopathology , Male , Mice, Inbred C57BL , Pulmonary Surfactants/metabolism , Signal Transduction , Smad3 Protein/metabolism , Transcription Factors , Transforming Growth Factor beta1/biosynthesis
20.
Am J Respir Crit Care Med ; 194(1): 84-96, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26771871

ABSTRACT

RATIONALE: Recent findings suggesting transforming growth factor (TGF)-ß1 activation by mechanical stimuli in vitro raised the question of whether this phenomenon was relevant in vivo in the context of pulmonary fibrosis. OBJECTIVES: To explore the effect of mechanical stress on TGF-ß1 activation and its signaling pathway in rat and human fibrotic lung tissue using a novel ex vivo model. METHODS: Rat lung fibrosis was induced using transient gene expression of active TGF-ß1. Lungs were harvested at Day 14 or 21 and submitted to various stimuli in a tissue bath equipped with a force transducer and servo-controlled arm. MEASUREMENTS AND MAIN RESULTS: Fibrotic lung strips responded to tensile force by releasing active TGF-ß1 from latent stores with subsequent increase in tissue phospho-Smad2/3. In contrast, measurable active TGF-ß1 and phospho-Smad2/3 were not induced by mechanical stress in nonfibrotic lungs. Protease inhibition did not affect the release of active TGF-ß1. A TGF-ß1 receptor inhibitor, Rho-associated protein kinase inhibitor, and αv integrin inhibitor all attenuated mechanical stretch-induced phospho-Smad2/3 in fibrotic lung strips. Furthermore, the induction of phospho-Smad2/3 was enhanced in whole fibrotic rat lungs undergoing ventilation pressure challenge compared with control lungs. Last, tissue slices from human lung with usual interstitial pneumonia submitted to mechanical force showed an increase in TGF-ß1 activation and induction of phospho-Smad2/3 in contrast with human nonfibrotic lungs. CONCLUSIONS: Mechanical tissue stretch contributes to the development of pulmonary fibrosis via mechanotransduced activation of TGF-ß1 in rodent and human pulmonary fibrosis.


Subject(s)
Lung/physiopathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/physiopathology , Stress, Mechanical , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Humans , Rats , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...