TY - JOUR AB - The human airway epithelium is a primary site of toxicant exposure and crucial in the pathogenesis of acute and chronic lung disease (CLD). In CLD, the airway epithelium is frequently altered and distorted, and its restoration is desirable. The mechanisms underlying human aberrant epithelial regeneration, however, are poorly understood. Importantly, our knowledge about airway epithelial injury and regeneration largely stems from mouse models, yet airways differ considerably between mice and humans. We hypothesized that treatment of differentiated primary human bronchial epithelial cells (phBECs, or HBEC) with polidocanol or naphthalene would allow for studying mechanisms of human airway epithelial injury and regeneration. Injury of differentiated phBECs with 0.04%, but not 0.1% PDOC, resulted in full restoration of a functional epithelium and epithelial barrier integrity as monitored by qRT-PCR analysis, immunofluorescence stainings, and transepithelial electrical resistance measurements. Regeneration was associated with a transient but not parallel increase of p21+ and KRT17+ cells. Providing proof-of-concept, DAPT, an inhibitor of Notch signaling, blunted the restoration of secretory cell types post 0.04% PDOC injury. Differentiation of phBECs in presence of cigarette smoke extract (CSE) or ethanol as first hit significantly impaired the regeneration capacity of phBECs. While naphthalene is known to specifically induce club cell depletion in mouse airways, it failed to do so in phBECs. In conclusion, using fully differentiated phBECs treated with PDOC, we successfully established and thoroughly characterized a human in vitro system that will facilitate studies of mechanisms involved in susceptibility to injury as well as human airway repair and regeneration. AU - Chakraborty, A. AU - Zöller, M. AU - Sardogan, A. AU - Klotz, M. AU - Mastalerz, M. AU - Marchi, H. AU - Meixner, R. AU - Hatz, R.A.* AU - Behr, J.* AU - Hilgendorff, A. AU - Nakayama, M. AU - Staab-Weijnitz, C.A. C1 - 74123 C2 - 57311 TI - Development of a polidocanol-based human in vitro model to explore airway epithelial repair. JO - Am. J. Respir. Cell Mol. Biol. PY - 2025 SN - 1044-1549 ER - TY - JOUR AB - The urgent need for effective treatments for acute and chronic lung diseases underscores the significance of developing innovative preclinical human research tools. The 2023 American Thoracic Society Workshop on Precision-Cut Lung Slices (PCLSs) brought together 35 experts to discuss and address the role of human tissue–derived PCLSs as a unique tool for target and drug discovery and validation in pulmonary medicine. With increasing interest and usage, together with advancements in methods and technology, there is a growing need for consensus on PCLS methodology and readouts. The current document recommends standard reporting criteria and emphasizes the requirement for careful collection and integration of clinical metadata. We further discuss current clinically relevant readouts that can be applied to PCLSs and highlight recent developments and future steps for implementing novel technologies for PCLS modeling and analysis. The collection and correlation of clinical metadata and multiomic analysis will further advance the integration of this preclinical platform into patient endotyping and the development of tailored therapies for patients with lung disease. AU - Lehmann, M. AU - Krishnan, R.* AU - Sucre, J.* AU - Kulkarni, H.S.* AU - Pineda, R.H.* AU - Anderson, C.* AU - Banovich, N.E.* AU - Behrsing, H.P.* AU - Dean, C.H.* AU - Haak, A.* AU - Gosens, R.* AU - Kaminski, N.* AU - Zagorska, A.* AU - Koziol-White, C.* AU - Metcalf, J.P.* AU - Kim, Y.H.* AU - Loebel, C.* AU - Neptune, E.R.* AU - Noël, A.* AU - Raghu, G.* AU - Sewald, K.* AU - Sharma, A.* AU - Suki, B.* AU - Sperling, A.* AU - Tatler, A.* AU - Turner, S.* AU - Rosas, I.O.* AU - Pam Van, R.Y.* AU - Wille, T.* AU - Randell, S.H.* AU - Pryhuber, G.* AU - Rojas, M.* AU - Bourke, J.* AU - Königshoff, M.* C1 - 73003 C2 - 56805 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 16-31 TI - Precision-cut lung slices: Emerging tools for preclinical and translational lung research an official american thoracic society workshop report. JO - Am. J. Respir. Cell Mol. Biol. VL - 72 IS - 1 PB - Amer Thoracic Soc PY - 2025 SN - 1044-1549 ER - TY - JOUR AB - Macrophage-mediated inflammation drives various lung diseases, including chronic obstructive pulmonary disease (COPD). COPD macrophages have dysfunctional mitochondrial metabolism and function which lead to a chronic inflammatory lung environment. However, the factors regulating this altered metabolism have not been elucidated. Adenine nucleotide translocase 1 (ANT1) is a mitochondrial ATP transporter critical to mitochondrial metabolism. We demonstrate that human alveolar macrophages from patients with moderate COPD (GOLD stage 2) have reduced ANT1 expression while macrophages from very severe COPD (GOLD stage 4) have elevated ANT1 compared to normal control subjects. Ant1-deficient mice were protected against cigarette smoke (CS)-induced emphysema with failure of recruited immune cells to migrate into alveoli. Ant1-null alveolar macrophages had reduced ATP production and mitochondrial respiration, upregulated fewer inflammatory pathways after CS and reduced migratory capacity. Conditional Ant1 knockout in Cx3cr1-positive monocytes and adoptive transfer of Ant1-deficient bone marrow into CS-treated mice phenocopied the migratory defect in the lung. Our data indicate that ANT1 is a critical regulator of lung macrophage inflammatory signaling and CS-triggered cell migration in the lung, suggesting that metabolic modulation may be a promising therapeutic avenue for COPD. AU - Sui, J.* AU - Johnson, A.R.* AU - Kapellos, T. AU - Shiva, S.* AU - Kliment, C.R.* C1 - 74824 C2 - 57617 TI - ANT1 deficiency impairs macrophage metabolism and migration, protecting against emphysema in COPD. JO - Am. J. Respir. Cell Mol. Biol. PY - 2025 SN - 1044-1549 ER - TY - JOUR AU - Staab-Weijnitz, C.A. C1 - 68938 C2 - 53780 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 155-156 TI - A NEAT discovery hints at altered golgi signaling in lung fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 70 IS - 3 PB - Amer Thoracic Soc PY - 2023 SN - 1044-1549 ER - TY - JOUR AB - ATP-binding cassette (ABC) subfamily A member 3 (ABCA3) is a lipid transporter expressed in alveolar type II cells and localized in the limiting membrane of lamellar bodies. It is crucial for pulmonary surfactant storage and homeostasis. Mutations in the ABCA3 gene are the most common genetic cause of respiratory distress syndrome in mature newborns and interstitial lung disease in children. Apart from lung transplantation, there is no cure available. To address the lack of causal therapeutic options for ABCA3 deficiency, a rapid and reliable approach is needed to investigate variant-specific molecular mechanisms and to identify pharmacological modulators for mono- or combination therapies. To this end, we developed a phenotypic cell-based assay to autonomously identify ABCA3 wild-type-like or mutant-like cells by using machine-learning algorithms aimed at identifying morphological differences in WT and mutant cells. The assay was subsequently used to identify new drug candidates for ABCA3 specific molecular correction by high-content screening of 1,280 food and drug administration-approved small molecules. Cyclosporin A (CsA) was identified as a potent corrector, specific for some, but not all ABCA3 variants. Results were validated by our previously established functional small format assays. Hence, CsA may be selected for orphan drug evaluation in controlled repurposing trials in patients. AU - Forstner, M.* AU - Lin, S. AU - Yang, X.* AU - Kinting, S.* AU - Rothenaigner, I. AU - Schorpp, K.K. AU - Li, Y.* AU - Hadian, K. AU - Griese, M.* C1 - 63883 C2 - 51781 SP - 382-390 TI - High-content screen identifies cyclosporin A as a novel ABCA3-specific molecular corrector. JO - Am. J. Respir. Cell Mol. Biol. VL - 66 IS - 4 PY - 2022 SN - 1044-1549 ER - TY - JOUR AB - Transient receptor potential ankyrin 1 (TRPA1) is a non-selective Ca2+ permeable cation channel, which was originally cloned from human lung fibroblasts (HLFs). TRPA1-mediated Ca2+ entry is evoked by exposure to several chemicals, including allyl isothiocyanate (AITC), and a protective effect of TRPA1 activation in the development of cardiac fibrosis has been proposed. Yet, the function of TRPA1 in transforming growth factor β1 (TGF-β1)-driven fibroblast to myofibroblast differentiation and the development of pulmonary fibrosis remains elusive. TRPA1 expression and function was analyzed in cultured primary HLFs, and mRNA levels were significantly reduced after adding TGF-β1. Expression of genes encoding fibrosis markers, e.g. alpha smooth muscle actin (ACTA2), plasminogen activator inhibitor 1 (SERPINE1), fibronectin (FN1) and type I collagen (COL1A1) was increased after siRNA-mediated down-regulation of TRPA1-mRNA in HLFs. Moreover, AITC-induced Ca2+ entry in HLFs was decreased after TGF-β1 treatment and by application of TRPA1 siRNAs, while AITC treatment alone did not reduce cell viability or enhanced apoptosis. Most interestingly, AITC-induced TRPA1 activation augmented ERK1/2 phosphorylation, which might inhibit TGF-β-receptor signaling. Our results suggest an inhibitory function of TRPA1 channels in TGF-β1-driven fibroblast to myofibroblast differentiation. Therefore, activation of TRPA1 channels might be protective during the development of pulmonary fibrosis in patients. AU - Geiger, F.* AU - Zeitlmayr, S.* AU - Staab-Weijnitz, C.A. AU - Rajan, S.* AU - Breit, A.* AU - Gudermann, T.* AU - Dietrich, A.* C1 - 66666 C2 - 52066 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 314-325 TI - An inhibitory function of TRPA1 channels in TGF-β1-driven fibroblast to myofibroblast differentiation. JO - Am. J. Respir. Cell Mol. Biol. VL - 68 IS - 3 PB - Amer Thoracic Soc PY - 2022 SN - 1044-1549 ER - TY - JOUR AB - Organ fibrosis is characterized by epithelial injury and aberrant tissue repair, where activated effector cells, mostly fibroblasts and myofibroblasts, excessively deposit collagen into the extracellular matrix. Fibrosis frequently results in organ failure and has been estimated to contribute to at least one third of all global deaths. Also lung fibrosis, in particular idiopathic pulmonary fibrosis (IPF), is a fatal disease with rising incidence worldwide. As current treatment options targeting fibrogenesis are insufficient, there is an urgent need for novel therapeutic strategies. During the last decade, several studies have proposed to target intra- and extracellular components of the collagen biosynthesis, maturation, and degradation machinery. This includes intra- and extracellular targets directly acting on collagen gene products, but also such that anabolize essential building blocks of collagen, in particular glycine and proline biosynthetic enzymes. Collagen, however, is a ubiquitous molecule in the body and fulfils essential functions as a macromolecular scaffold, growth factor reservoir, and receptor binding site in virtually every tissue. This review summarizes recent advances and future directions in this field. Evidence for the proposed therapeutic targets and where they currently stand in terms of clinical drug development for treatment of fibrotic disease is provided. The drug targets are furthermore discussed in light of (1) specificity for collagen biosynthesis, maturation and degradation, and (2) specificity for disease-associated collagen. As therapeutic success and safety of these drugs may largely depend on targeted delivery, different strategies for specific delivery to the main effector cells and to the extracellular matrix are discussed. AU - Staab-Weijnitz, C.A. C1 - 63722 C2 - 51655 SP - 363-381 TI - Fighting the fiber: Targeting collagen in lung fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 66 IS - 4 PY - 2022 SN - 1044-1549 ER - TY - JOUR AB - The National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health, along with the Longfonds BREATH consortium convened a working group to review the field of lung regeneration and suggest avenues for future research. The meeting took place on May 22, 2019 at the American Thoracic Society 2019 conference in Dallas, Texas, USA, and brought together investigators studying lung development, adult stem cell biology, induced pluripotent stem cells, biomaterials and respiratory disease. The purpose of the working group was 1) to examine the present status of basic science approaches to tackling lung disease and promoting lung regeneration in patients and 2) to determine priorities for future research in the field. AU - Hynds, R.E.* AU - Zacharias, W.J.* AU - Nikolić, M.Z.* AU - Königshoff, M. AU - Eickelberg, O. AU - Gosens, R.* AU - de Coppi, P.* AU - Janes, S.M.* AU - Morrisey, E.* AU - Clevers, H.* AU - Ryan, A.L.* AU - Stripp, B.R.* AU - Sun, X.* AU - Kim, C.F.* AU - Lin, Q.S.* C1 - 61483 C2 - 50286 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 22-29 TI - National heart, lung and blood institute and building respiratory epithelium and tissue for health (BREATH) consortium workshop report: Moving forward in lung regeneration. JO - Am. J. Respir. Cell Mol. Biol. VL - 65 IS - 1 PB - Amer Thoracic Soc PY - 2021 SN - 1044-1549 ER - TY - JOUR AB - Molecular patterns and pathways in idiopathic pulmonary fibrosis (IPF) have been extensively investigated but few studies have assimilated multi-omic platforms to provide an integrative understanding of molecular patterns that are relevant in IPF. Herein, we combine coding and non-coding transcriptome, DNA methylome, and proteome from IPF and healthy lung tissue to identify molecules and pathways associated with this disease. RNA sequencing, Illumina MethylationEPIC array, and liquid chromatography-mass spectrometry (LC-MS) proteomic data were collected on lung tissue from 24 IPF cases and 14 control subjects. Significant differential features were identified using linear models adjusting for age and sex, inflation and bias where appropriate. Data Integration Analysis for Biomarker discovery using a Latent component method for Omics studies (DIABLO) was used for integrative multi-omic analysis. We identified 4,643 differentially expressed transcripts aligning to 3,439 genes, 998 differentially abundant proteins, 2,500 differentially methylated regions (DMRs), and 1,269 differentially expressed lncRNAs that were significant after correcting for multiple tests (false discovery rate [FDR]<0.05). Unsupervised hierarchical clustering using 20 coding mRNA, protein, methylation, and lncRNA features with highest loadings on the top latent variable from the four datasets demonstrates perfect separation of IPF and control lungs. Our analysis confirmed previously validated molecules and pathways known to be dysregulated in disease, and implicated novel molecular features as potential drivers and modifiers of disease. For example, four proteins, 18 DMRs, and 10 lncRNAs were found to have strong correlations (|r|>0.8) with MMP7. Therefore, using a systems biology approach, we have identified novel molecular relationships in IPF. AU - Konigsberg, I.R.* AU - Borie, R.* AU - Walts, A.D.* AU - Cardwell, J.* AU - Rojas, M.* AU - Metzger, F. AU - Hauck, S.M. AU - Fingerlin, T.E.* AU - Yang, I.V.* AU - Schwartz, D.A.* C1 - 62221 C2 - 50730 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 430-441 TI - Molecular signatures of idiopathic pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 65 IS - 4 PB - Amer Thoracic Soc PY - 2021 SN - 1044-1549 ER - TY - JOUR AB - Current smoking contributes to worsened asthma prognosis and more severe symptoms and limits the beneficial effects of corticosteroids. As the nasal epithelium can reflect smoking-induced changes in the lower airways, it is a relevant source to investigate changes in gene expression and DNA methylation. This study explores gene expression and DNA methylation changes in current and ex-smokers with asthma. Matched gene expression and epigenome-wide DNA methylation samples collected from nasal brushings of 55 patients enrolled in a clinical trial investigation of current and ex-smoker patients with asthma were analyzed. Differential gene expression and DNA methylation analyses were conducted comparing current smokers with ex-smokers. Expression quantitative trait methylation (eQTM) analysis was completed to explore smoking-relevant genes by CpG sites that differ between current and ex-smokers. To investigate the relevance of the smoking-associated DNA methylation changes for the lower airways, significant CpG sites were explored in bronchial biopsies from patients who had stopped smoking. A total of 809 genes and 18,814 CpG sites were differentially associated with current smoking in the nose. The cis-eQTM analysis uncovered 171 CpG sites with a methylation status associated with smoking-related gene expression, including AHRR, ALDH3A1, CYP1A1, and CYP1B1. The methylation status of CpG sites altered by current smoking reversed with 1 year of smoking cessation. We confirm that current smoking alters epigenetic patterns and affects gene expression in the nasal epithelium of patients with asthma, which is partially reversible in bronchial biopsies after smoking cessation. We demonstrate the ability to discern molecular changes in the nasal epithelium, presenting this as a tool in future investigations into disease-relevant effects of tobacco smoke. AU - Reddy, K.D.* AU - Lan, A.* AU - Boudewijn, I.M.* AU - Rathnayake, S.N.H.* AU - Koppelman, G.H.* AU - Aliee, H. AU - Theis, F.J. AU - Oliver, B.G.* AU - van den Berge, M.* AU - Faiz, A.* C1 - 63260 C2 - 51413 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 366-377 TI - Current smoking alters gene expression and DNA methylation in the nasal epithelium of patients with asthma. JO - Am. J. Respir. Cell Mol. Biol. VL - 65 IS - 4 PB - Amer Thoracic Soc PY - 2021 SN - 1044-1549 ER - TY - JOUR AU - Wjst, M. C1 - 60778 C2 - 49650 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 271-272 TI - Scientific integrity is threatened by image duplications. JO - Am. J. Respir. Cell Mol. Biol. VL - 64 IS - 2 PB - Amer Thoracic Soc PY - 2021 SN - 1044-1549 ER - TY - JOUR AB - Chronic lung diseases (CLDs), such as chronic obstructive pulmonary disease, interstitial lung disease, and lung cancer, are among the leading causes of morbidity globally and impose major health and financial burdens on patients and society. Effective treatments are scarce, and relevant human model systems to effectively study CLD pathomechanisms and thus discover and validate potential new targets and therapies are needed. Precision-cut lung slices (PCLS) from healthy and diseased human tissue represent one promising tool that can closely recapitulate the complexity of the lung's native environment, and recently, improved methodologies and accessibility to human tissue have led to an increased use of PCLS in CLD research. Here, we discuss approaches that use human PCLS to advance our understanding of CLD development, as well as drug discovery and validation for CLDs. PCLS enable investigators to study complex interactions among different cell types and the extracellular matrix in the native three-dimensional architecture of the lung. PCLS further allow for high-resolution (live) imaging of cellular functions in several dimensions. Importantly, PCLS can be derived from diseased lung tissue upon lung surgery or transplantation, thus allowing the study of CLDs in living human tissue. Moreover, CLDs can be modeled in PCLS derived from normal lung tissue to mimic the onset and progression of CLDs, complementing studies in end-stage diseased tissue. Altogether, PCLS are emerging as a remarkable tool to further bridge the gap between target identification and translation into clinical studies, and thus open novel avenues for future precision medicine approaches. AU - Alsafadi, H.N. AU - Uhl, F.E.* AU - Pineda, R.H.* AU - Bailey, K.E.* AU - Rojas, M.* AU - Wagner, D.E. AU - Königshoff, M. C1 - 59403 C2 - 48796 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 681-691 TI - Applications and approaches for three-dimensional precision-cut lung slices disease modeling and drug discovery. JO - Am. J. Respir. Cell Mol. Biol. VL - 62 IS - 6 PB - Amer Thoracic Soc PY - 2020 SN - 1044-1549 ER - TY - JOUR AB - Lung disease accounts for every sixth death globally. Profiling the molecular state of all lung cell types in health and disease is currently revolutionizing the identification of disease mechanisms and will aid the design of novel diagnostic and personalized therapeutic regimens. Recent progress in high-throughput techniques for single-cell genomic and transcriptomic analyses has opened up new possibilities to study individual cells within a tissue, classify these into cell types, and characterize variations in their molecular profiles as a function of genetics, environment, cell-cell interactions, developmental processes, aging, or disease. Integration of these cell state definitions with spatial information allows the in-depth molecular description of cellular neighborhoods and tissue microenvironments, including the tissue resident structural and immune cells, the tissue matrix, and the microbiome. The Human Cell Atlas consortium aims to characterize all cells in the healthy human body and has prioritized lung tissue as one of the flagship projects. Here, we present the rationale, the approach, and the expected impact of a Human Lung Cell Atlas. AU - Schiller, H. B. AU - Montoro, D.T.* AU - Simon, L. AU - Rawlins, E.L.* AU - Meyer, K.B.* AU - Strunz, M. AU - Vieira Braga, F.* AU - Timens, W.* AU - Koppelman, G.H.* AU - Budinger, G.R.S.* AU - Burgess, J.K.* AU - van den Berge, M.* AU - Theis, F.J. AU - Regev, A.* AU - Kaminski, N.* AU - Rajagopal, J.* AU - Teichmann, S.A.* AU - Misharin, A.V.* AU - Nawijn, M.C.* C1 - 55908 C2 - 46697 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 31-41 TI - The Human Lung Cell Atlas: A high-resolution reference map of the human lung in health and disease. JO - Am. J. Respir. Cell Mol. Biol. VL - 61 IS - 1 PB - Amer Thoracic Soc PY - 2019 SN - 1044-1549 ER - TY - JOUR AB - Idiopathic pulmonary fibrosis (IPF) and lung cancer are progressive lung diseases with a poor prognosis. IPF is a risk factor for the development of lung cancer, and the incidence of lung cancer is increased in patients with IPF. The disease pathogenesis of IPF and lung cancer involves common genetic alterations, dysregulated pathways, and the emergence of hyperplastic and metaplastic epithelial cells. Here, we aimed to identify novel, common mediators that might contribute to epithelial cell reprogramming in IPF. Gene set enrichment analysis of publicly available non-small cell lung cancer and IPF datasets revealed a common pattern of misregulated genes linked to cell proliferation and transformation. The oncogene ECT2 (epithelial cell transforming sequence 2), a guanine nucleotide exchange factor for Rho GTPases, was highly enriched in both IPF and non-small cell lung cancer compared with nondiseased controls. Increased expression of ECT2 was verified by qPCR and Western blotting in bleomycin-induced lung fibrosis and human IPF tissue. Immunohistochemistry demonstrated strong expression of ECT2 staining in hyperplastic alveolar epithelial type II (ATII) cells in IPF, as well as its colocalization with proliferating cell nuclear antigen, a well-known proliferation marker. Increased ECT2 expression coincided with enhanced proliferation of primary mouse ATII cells as analyzed by flow cytometry. ECT2 knockdown in ATII cells resulted in decreased proliferation and collagen I expression in vitro. These data suggest that the oncogene ECT2 contributes to epithelial cell reprogramming in IPF, and further emphasize the hyperplastic, proliferative ATII cell as a potential target in patients with IPF and lung cancer. AU - Ulke, H.M. AU - Mutze, K. AU - Lehmann, M. AU - Wagner, D.E. AU - Heinzelmann, K. AU - Guenther, A.* AU - Eickelberg, O.* AU - Königshoff, M. C1 - 57618 C2 - 47867 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 713-726 TI - The oncogene ECT2 contributes to a hyperplastic, proliferative lung epithelial cell phenotype in idiopathic pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 61 IS - 6 PB - Amer Thoracic Soc PY - 2019 SN - 1044-1549 ER - TY - JOUR AB - Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive aging-associated disease of unknown etiology. A growing body of evidence indicates that aberrant activated alveolar epithelial cells induce the expansion and activation of the fibroblast population, leading to the destruction of the lung architecture. Some matrix metalloproteinases (MMPs) are upregulated in IPF, indicating that they may be important in the pathogenesis and/or progression of IPF. In the present study, we examined the expression of MMP28 in this disease and evaluated its functional effects in two alveolar epithelial cell lines and in human primary bronchial epithelial cells. We found that the enzyme is expressed in bronchial (apical and cytoplasmic localization) and alveolar (cytoplasmic and nuclear localization) epithelial cells in two different groups of patients with IPF. In vitro MMP28 epithelial silencing decreased the proliferation rate and delayed wound closing, whereas overexpression showed opposite effects, protecting from apoptosis and enhanced epithelial-mesenchymal transition. Our findings demonstrate that MMP28is upregulated in epithelial cells from IPF lungs, where it may play a role in increasing the proliferative and migratory phenotype in a catalysis-dependent manner. AU - Maldonado, M.* AU - Salgado-Aguayo, A.* AU - Herrera, I.* AU - Cabrera, S.* AU - Ortíz-Quintero, B.* AU - Staab-Weijnitz, C.A. AU - Eickelberg, O. AU - Ramírez, R.* AU - Manicone, A.M.* AU - Selman, M.* AU - Pardo, A.* C1 - 52806 C2 - 44316 CY - 25 Broadway, 18 Fl, New York, Ny 10004 Usa SP - 77-86 TI - Upregulation and nuclear location of MMP28 in alveolar epithelium of idiopathic pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 59 IS - 1 PB - Amer Thoracic Soc PY - 2018 SN - 1044-1549 ER - TY - JOUR AU - Costa, R. AU - Königshoff, M. C1 - 51128 C2 - 42781 CY - New York SP - 551-552 TI - Linking wnt signaling to mucosal inflammation. JO - Am. J. Respir. Cell Mol. Biol. VL - 56 IS - 5 PB - Amer Thoracic Soc PY - 2017 SN - 1044-1549 ER - TY - JOUR AB - RATIONALE: Idiopathic pulmonary fibrosis (IPF) is characterized by excessive deposition of extracellular matrix, in particular collagens. Two IPF therapeutics, nintedanib and pirfenidone, decelerate lung function decline, but their underlying mechanisms of action are poorly understood. In this study we sought to analyze their effects on collagen synthesis and maturation at important regulatory levels. METHODS: Primary human fibroblasts from IPF patients and healthy donors were treated with nintedanib (0.01-1.0µM) or pirfenidone (0.1-1.0mM) in absence or presence of TGF-β1. Effects on collagen, fibronectin, FKBP10, HSP47 expression and collagen I and III secretion were analyzed by qPCR and Western Blot. Appearance of collagen fibrils was monitored by scanning electron microscopy (SEM) and kinetics of collagen fibril assembly was assessed in a light scattering approach. RESULTS: In IPF fibroblasts, nintedanib reduced the expression of collagen I, V, fibronectin and FKBP10 and attenuated secretion of collagen I and III. Pirfenidone also downregulated collagen V, but otherwise showed fewer and less pronounced effects. By and large, effects were similar in donor fibroblasts. For both drugs, electron microscopy of IPF fibroblast cultures revealed fewer and thinner collagen fibrils compared with untreated controls. Finally, both drugs dose-dependently delayed fibril formation of purified collagen I. CONCLUSIONS: Both drugs act on important regulatory levels in collagen synthesis and processing. Nintedanib was more effective in downregulating profibrotic gene expression and collagen secretion. Importantly, both drugs inhibited collagen I fibril formation and caused reduction and an altered appearance of collagen fibril bundles, representing a completely novel mechanism of action for both drugs. AU - Knüppel, L. AU - Ishikawa, Y.* AU - Aichler, M. AU - Heinzelmann, K. AU - Hatz, R.* AU - Behr, J.* AU - Walch, A.K. AU - Bächinger, H.P.* AU - Eickelberg, O. AU - Staab-Weijnitz, C.A. C1 - 50620 C2 - 42460 CY - New York SP - 77-90 TI - A novel antifibrotic mechanism of nintedanib and pirfenidone: Inhibition of collagen fibril assembly. JO - Am. J. Respir. Cell Mol. Biol. VL - 57 IS - 1 PB - Amer Thoracic Soc PY - 2017 SN - 1044-1549 ER - TY - JOUR AB - Pulmonary fibrosis, particularly idiopathic pulmonary fibrosis, represents a chronic and progressive disease with high mortality and limited therapeutic options. Excessive deposition of extracellular matrix proteins results in fibrotic remodeling, alveolar destruction and irreversible loss of lung function. Both innate and adaptive immune mechanisms contribute to fibrogenesis at several cellular and non-cellular levels. Here, we summarize and discuss the role of immune cells (T cells, neutrophils, macrophages and fibrocytes) and soluble mediators (cytokines and chemokines) involved in pulmonary fibrosis, pointing towards novel immune-based therapeutic strategies in the field. AU - Kolahian, S.* AU - Fernandez, I.E. AU - Eickelberg, O. AU - Hartl, D.* C1 - 48537 C2 - 41133 CY - New York SP - 309-322 TI - Immune mechanisms in pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 55 IS - 3 PB - Amer Thoracic Soc PY - 2016 SN - 1044-1549 ER - TY - JOUR AB - Platelet-derived growth factors and their receptors (PDGF/PDGFRs) represent one of the most intensively studied families of growth factors in the last four decades. PDGF signaling plays an essential role in cell proliferation, differentiation, migration, and survival. In vivo studies documented an important role of PDGF signaling in normal development of several organs, such as the kidney, eye, or lung. Here, PDGF signaling is essential for the formation of intact mesenchymal cells during embryogenesis. Recently, this knowledge has been extended to a role of PDGF signaling in diseases in general, such as cancer and atherosclerosis, and more importantly in lung diseases, including PAH, lung cancer, and lung fibrosis. In this review, we provide an up-to-date overview of PDGF signaling, including tissue- and cell-type specific expression patterns and effects. We will highlight current therapeutic approaches modifying PDGF signaling in lung diseases and summarize clinical trials, in which PDGF signaling has been inhibited. In conclusion, while PDGF inhibition is currently used in multiple clinical trials, we suggest that more elaborate and specific approaches for spatio-temporal control of PDGF signaling are required for developing personalized approaches involving PDGF signaling in lung disease. AU - Noskovicova, N. AU - Petrek, M.* AU - Eickelberg, O. AU - Heinzelmann, K. C1 - 32520 C2 - 35103 CY - New York SP - 263-284 TI - Platelet-derived growth factor  signaling in the lung - from lung development and disease to clinical studies. JO - Am. J. Respir. Cell Mol. Biol. VL - 52 IS - 3 PB - Amer Thoracic Soc PY - 2015 SN - 1044-1549 ER - TY - JOUR AB - Chronic obstructive pulmonary disease (COPD) is characterized by an irreversible loss of lung function and is one of the most prevalent and severe diseases world-wide. A major feature of COPD is emphysema -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 cells (ATII). 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 siRNA-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 vs. 38.8±5.5 µm, p<0.01) and lung compliance (2.8±0.32 µl/cmH20 vs. 2.4±0.4 µl/cmH20, p<0.04) compared with controls. The knockdown of CARM1 in LA4 cells led to decreased SIRT1 expression (0.034±0.003 vs. 0.022±0.001, p<0.05), but increased expression of p16 (0.27±0.013 vs. 0.31±0.010, p<0.5), p21 (0.81±0.088 vs. 1.28± 0.063, p<0.01) and higher beta-galactosidase-positive senescent cells (50.57%±7.36 vs. 2.21%±0.34, p<0.001), compared with scrambled siRNA. We further demonstrated that CARM1 haploinsufficiency impairs trans-differentiation and wound healing (32.18%±0.9512 vs. 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. AU - Sarker, R.S. AU - John-Schuster, G. AU - Bohla, A. AU - Mutze, K. AU - Burgstaller, G. AU - Bedford, M.T.* AU - Königshoff, M. AU - Eickelberg, O. AU - Yildirim, A.Ö. C1 - 44476 C2 - 36950 SP - 769-781 TI - CARM1 regulates alveolar epithelial senescence and elastase-induced emphysema susceptibility. JO - Am. J. Respir. Cell Mol. Biol. VL - 53 IS - 6 PY - 2015 SN - 1044-1549 ER - TY - JOUR AB - Secreted phosphoprotein 1 (Spp1) is located within quantitative trait loci associated with lung function that was previously identified by contrasting C3H/HeJ and JF1/Msf mouse strains that have extremely divergent lung function. JF1/Msf mice with diminished lung function had reduced lung SPP1 transcript and protein during the peak stage of alveologenesis (postnatal day 14-28) as compared to C3H/HeJ mice. In addition to a previously identified genetic variant that altered runt related transcription factor 2 (RUNX2) binding in the Spp1 promoter, we identified another promoter variant in a putative RUNX2 binding site that increased the DNA protein binding. SPP1 induced dose dependent MLE-15 cell proliferation. Spp1((-/-)) mice have decreased specific total lung capacity/body weight, higher specific compliance, and increased mean airspace chord length (Lm) compared to Spp1((+/+)) mice. Microarray analysis revealed enriched gene ontogeny (GO) categories with numerous genes associated with lung development and/or respiratory disease. IGF1, HHIP, WNT5A, and NOTCH1 transcripts decreased in the lung of P14 Spp1((-/-)) mice as determined by qRT-PCR analysis. SPP1 promotes pneumocyte growth and mice lacking SPP1 have smaller, more compliant lungs with enlarged airspace (increased Lm). Microarray analysis suggests a dysregulation of key lung developmental transcripts in gene targeted Spp1((-/-)) mice particularly during the peak phase of alveologenesis. In addition to its known roles in lung disease, this study supports SPP1 as a determinant of lung development in mice. AU - Ganguly, K.* AU - Martin, T.M.* AU - Concel, V.J.* AU - Upadhyay, S.* AU - Bein, K.* AU - Brant, K.A.* AU - George, L.* AU - Mitra, A.* AU - Thimraj, T.A.* AU - Fabisiak, J.P.* AU - Vuga, L.J.* AU - Fattman, C.* AU - Kaminski, N.* AU - Schulz, H. AU - Leikauf, G.D.* C1 - 31538 C2 - 34537 CY - New York SP - 637-651 TI - Secreted phosphoprotein 1 is a determinant of lung function development in mice. JO - Am. J. Respir. Cell Mol. Biol. VL - 51 IS - 5 PB - Amer Thoracic Soc PY - 2014 SN - 1044-1549 ER - TY - JOUR AB - Neonatal chronic lung disease (nCLD), also known as bronchopulmonary dysplasia (BPD), is the most common complication of premature birth, affecting up to 30% of very low birth weight infants. Improved medical care has allowed for the survival of the most premature infants, and significantly changed the pathology of BPD from a disease marked by severe lung injury, to the "new" form characterized by alveolar hypoplasia and impaired vascular development. However, increased patient survival has led to a paucity of pathologic specimens available from infants with BPD. This, combined with the lack of a system to model alveolarization in vitro, has resulted in a great need for animal models that recapitulate key features of the disease. To this end, a number of animal models have been created, by exposing the immature lung to injuries induced by hyperoxia, mechanical stretch, and inflammation, and most recently by the genetic modification of mice. These animal studies have: (i) allowed insight into the mechanisms that determine alveolar growth; (ii) delineated factors central to the pathogenesis of nCLD; and (iii) informed the development of new therapies. In this review, we will summarize the key findings and limitations of the most common animal models of BPD, and discuss how knowledge obtained from these studies has informed clinical care. Future studies should aim to provide a more complete understanding of the pathways that both preserve and repair alveolar growth during injury, which might be translated into novel strategies to treat lung diseases in both infants and adults. AU - Hilgendorff, A. AU - Reiss, I.* AU - Ehrhardt, H.* AU - Eickelberg, O. AU - Alvira, C.M.* C1 - 28305 C2 - 33089 CY - New York SP - 233-245 TI - Chronic lung disease in the preterm infant : Lessons learned from animal models. JO - Am. J. Respir. Cell Mol. Biol. VL - 50 IS - 2 PB - Amer. Thoracic Soc. PY - 2014 SN - 1044-1549 ER - TY - JOUR AB - In inhalation therapy, drugs are deposited as aerosols onto the air-facing lung epithelium. The currently used in vitro cell assays for drug testing, however, typically dissolve drugs in the medium completely covering the cells which represents an unphysiological drug application scenario. While physiologically realistic in vitro cell culture models of the pulmonary air-blood barrier are available, reliable, easy-to-handle and efficient technologies for direct aerosol-to-cell delivery are lacking. Here, we introduce the ALICE-CLOUD technology, which utilizes principles of cloud motion for fast and quantitative delivery of aerosolized liquid drugs to pulmonary cells cultured under realistic air-liquid interface conditions. Aerosol-to-cell delivery proved to be highly efficient, reproducible and rapid when using aerosolized fluorescein as surrogate drug. As a proof-of-concept study for the ALICE-CLOUD we performed functional efficacy studies with the FDA-approved proteasome inhibitor Bortezomib, a novel candidate drug for inhalation therapy. Aerosolized Bortezomib had a pronounced anti-inflammatory effect on human epithelial lung cells (A549) as indicated by a significant reduction of (TNFα-induced) IL-8 promoter activation. Importantly, cell-based therapeutic efficacy of aerosolized Bortezomib at air-liquid interface conditions was similar to dissolved and non-aerosolized submerged conditions, but with faster uptake kinetics. Our data indicate that the ALICE-CLOUD is a reliable tool for aerosolized drug screening with cells cultured at air-liquid interface conditions, which combines ease-of-handling with rapid, efficient and dosimetrically accurate drug-to-cell delivery. This may pave the way for screening of inhalable drugs under physiologically more relevant and hence potentially more predictive conditions than the currently used submerged cell culture systems. AU - Lenz, A.-G. AU - Stöger, T. AU - Cei, D. AU - Schmidmeir, M. AU - Pfister, N. AU - Burgstaller, G. AU - Lentner, B. AU - Eickelberg, O. AU - Meiners, S. AU - Schmid, O. C1 - 31179 C2 - 34193 CY - New York SP - 526-535 TI - Efficient bioactive delivery of aerosolized drugs to human pulmonary epithelial cells cultured at air-liquid interface conditions. JO - Am. J. Respir. Cell Mol. Biol. VL - 51 IS - 4 PB - Amer Thoracic Soc PY - 2014 SN - 1044-1549 ER - TY - JOUR AB - The airway epithelium constitutes an essential immunological and cytoprotective barrier to inhaled insults, such as cigarette smoke, environmental particles, or viruses. While bronchial epithelial integrity is crucial for airway homeostasis, defective epithelial barrier function contributes to chronic obstructive pulmonary disease (COPD). Tight junctions at the apical side of epithelial cell-cell contacts determine epithelial permeability. Cigarette smoke exposure, the major risk factor for COPD, is suggested to impair tight junction integrity; however, detailed mechanisms thereof remain elusive. Here, we investigated whether cigarette smoke extract (CSE) and transforming growth factor (TGF)-β1 affected tight junction integrity. Exposure of human bronchial epithelial cells (16HBE14o-), as well as differentiated primary human bronchial epithelial cells (pHBECs), to CSE significantly disrupted tight junction integrity and barrier function. Specifically, CSE decreased transepithelial electrical resistance (TEER) and tight junction-associated protein levels. Zona occludens (ZO)-1 and -2 protein levels were significantly reduced and dislocated from the cell membrane, as observed by fractionation and immunofluorescence analysis. These findings were reproduced in isolated bronchi exposed to CSE ex vivo, as detected by qRT-PCR and immunohistochemistry. Importantly, combined treatment of 16HBE14o- cells or pHBECs with CSE and TGF-β1 restored ZO-1 and ZO-2 protein levels. TGF-β1 co-treatment restored not only membrane localization of ZO-1 and ZO-2 protein, but also prevented CSE-mediated TEER decrease. In conclusion, CSE led to the disruption of tight junctions of human bronchial epithelial cells, while TGF-β1 counteracted this CSE-induced effect. Thus, TGF-β1 may serve as a protective factor for bronchial epithelial cell homeostasis in diseases such as COPD. AU - Schamberger, A.C. AU - Mise, N. AU - Jia, J. AU - Genoyer, E. AU - Yildirim, A.Ö. AU - Meiners, S. AU - Eickelberg, O. C1 - 28865 C2 - 33551 CY - New York SP - 1040-1052 TI - Cigarette smoke-induced disruption of bronchial epithelial tight junctions is prevented by transforming growth factor-beta. JO - Am. J. Respir. Cell Mol. Biol. VL - 50 IS - 6 PB - Amer Thoracic Soc PY - 2014 SN - 1044-1549 ER - TY - JOUR AB - Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease of yet unknown etiology. It is characterised by alterations of the alveolar epithelium, myofibroblast activation, and increased extracellular matrix deposition. Recently, reactivation of WNT/β-catenin signaling has been linked with IPF. The cell-specific mechanisms and mediators of WNT/β-catenin signaling in the lung, however, remain elusive. Here, we applied an unbiased gene expression screen to identify epithelial cell-specific mediators of WNT/β-catenin signaling. We found the proinflammatory cytokine interleukin (IL) 1β as one of the most upregulated genes in primary murine alveolar epithelial type (AT) II cells after WNT3a treatment. Increased transcript and protein expression of IL-1β upon WNT3a treatment was further detected in primary ATII cells by qRT-PCR (log-fold change: 2.0 +/- 0.5) and ELISA (1.8 fold increase). We observed significant IL-1β and IL-6 upregulation in bronchoalveolar lavage fluid (BALF) in bleomycin induced lung fibrosis in vivo. Importantly, primary fibrotic ATII cells secreted enhanced IL-1β and IL-6 in vitro. Furthermore, orotracheal application of recombinant WNT protein in TOPGAL reporter animals led to WNT/β-catenin activation in epithelial cells along with a significant increase in IL-1β and IL-6 in vivo (2.7-fold and 6.0-fold, respectively). Finally, we found increased WNT3a protein in fibrotic alveolar epithelium accompanied by enhanced IL-1β and IL-6 level in BALF from IPF patients. Taken together, our findings revealed that the alveolar epithelium is a relevant source of proinflammatory cytokines induced by active WNT/β-catenin. Thus, WNT/interleukin signaling represents a novel link between developmental pathway reactivation and inflammation in the development of pulmonary fibrosis. AU - Aumiller, V. AU - Balsara, N.* AU - Wilhelm, J.* AU - Günther, A.* AU - Königshoff, M. C1 - 24883 C2 - 31712 SP - 96-104 TI - WNT/β-catenin signaling induces interleukin 1β expression by alveolar epithelial cells in pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 49 IS - 1 PB - Amer. Thoracic Soc. PY - 2013 SN - 1044-1549 ER - TY - JOUR AB - A genome-wide association study identified interferon-related development regulator-1 (IFRD1), a protein expressed by neutrophils, as a key modifier gene in cystic fibrosis (CF) lung disease. Here, we investigated the expression and regulation of IFRD1 in CF neutrophils. IFRD1 expression was quantified in peripheral blood and airway neutrophils from patients with CF, patients with non-CF lung disease, and healthy control subjects. The regulation of IFRD1 expression was analyzed using isolated neutrophils and ex vivo stimulation assays with CF airway fluids. IFRD1 single-nucleotide polymorphisms (SNPs) were analyzed in a CF cohort (n = 572) and correlated with longitudinal lung function and IFRD1 expression. Patients with CF expressed higher protein levels of IFRD1 in peripheral blood neutrophils compared with healthy or non-CF disease control subjects. Within patients with CF, IFRD1 protein expression levels in neutrophils were lower in airway fluids compared with peripheral blood. High IFRD1 expression was positively associated with the production of reactive oxygen species (ROS) in CF neutrophils. In vitro regulation studies showed that CF airway fluid and the CF-characteristic chemokines CXCL8 and CXCL2 down-regulated IFRD1 expression in neutrophils, an effect that was mediated through CXCR2. Genetic analyses showed that three IFRD1 SNPs were associated with longitudinal declines in lung function, and modulated IFRD1 expression. These studies demonstrate that IFRD1 expression is systemically up-regulated in human CF neutrophils, is linked to the production of ROS, and is modulated by chemokines in CF airway fluids, depending on the IFRD1 genotype. Understanding the regulation of IFRD1 may pave the way for novel therapeutic approaches to target neutrophilic inflammation in CF. AU - Hector, A.* AU - Kormann, M.* AU - Kammermeier, J.* AU - Burdi, S.* AU - Marcos, V.* AU - Rieber, N.* AU - Mays, L.* AU - Illig, T. AU - Klopp, N. AU - Falkenstein, F.* AU - Kappler, M.* AU - Riethmueller, J.* AU - Graepler-Mainka, U.* AU - Stern, M.* AU - Eickmeier, O.* AU - Serve, F.* AU - Zielen, S.* AU - Döring, G.* AU - Griese, M.* AU - Hartl, D.* C1 - 22462 C2 - 30871 SP - 71-77 TI - Expression and regulation of interferon-related development regulator-1 in cystic fibrosis neutrophils. JO - Am. J. Respir. Cell Mol. Biol. VL - 48 IS - 1 PB - American Thoracic Society PY - 2013 SN - 1044-1549 ER - TY - JOUR AB - Rationale: Macrophages are lung-resident cells that play key roles in fibrosis. Surprisingly, pathways that inhibit macrophage functions, especially in idiopathic pulmonary fibrosis (IPF), received little attention. The cell-surface molecule paired immunoglobulin-like receptor B (PIR-B) can suppress macrophage activation. Yet, its role in pulmonary fibrosis is unknown. Objective: To define the role of PIR-B in IPF. Methods: The expression of PIR-B was assessed (qPCR, flow cytometry) following bleomycin treatment. Differential cell counts, histopathology and profibrogenic-mediator expression (e.g. Collagen, α-SMA, Relm-α, MMP-12, TIMP-1) were determined (ELISA, qPCR, flow cytometry) in the lungs of wild type and Pirb-/- mice following bleomycin or IL-4 treatment. Bone marrow-derived wild-type and Pirb-/- macrophages were stimulated with IL-4 and assessed for Relm-α and MMP-12 expression. Measurements and Main Results: PIR-B was upregulated in lung myeloid cells following bleomycin administration. Bleomycin-treated Pirb-/- mice displayed increased lung histopathology, increased collagen expression and of the IL-4-associated profibrogenic markers Relm-α, MMP-12, TIMP-1 and osteopontin, which were localized to alveolar macrophages. Increased profibrogenic mediator expression in Pirb-/- mice was not due to increased IL-4/IL-13 levels, suggesting that PIR-B negatively regulates IL-4-induced macrophage activation. Indeed, IL-4 treated Pirb-/- mice displayed increased Relm-α expression and Relm-α+ macrophage levels. IL-4-activated Pirb-/- macrophages displayed increased Relm-α and MMP-12 induction. Finally, LILRB3/ILT-5, the human PIR-B ortholog was expressed and upregulated in lung biopsies from IPF patients. Conclusions: Our results establish a key role for PIR-B in IPF, likely via regulation of macrophage activation. Therefore, PIR-B/LILRB3 may be a possible target for suppressing macrophage profibrogenic activity in IPF. AU - Karo-Atar, D.* AU - Moshkovits, I.* AU - Eickelberg, O. AU - Königshoff, M. AU - Munitz, A.* C1 - 11743 C2 - 30801 SP - 456-464 TI - Paired immunoglobulin-like receptor-B inhibits pulmonary fibrosis by suppressing profibrogenic properties of alveolar macrophages. JO - Am. J. Respir. Cell Mol. Biol. VL - 48 IS - 4 PB - American Thoracic Society PY - 2013 SN - 1044-1549 ER - TY - JOUR AB - In this study, we explored the regulation and the role of up-regulated microRNAs in idiopathic pulmonary fibrosis (IPF), a progressive interstitial lung disease of unknown origin. We analyzed the expression of microRNAs in IPF lungs and identified 43 significantly up-regulated microRNAs. Twenty-four of the 43 increased microRNAs were localized to the chromosome 14q32 microRNA cluster. We validated the increased expression of miR-154, miR-134, miR-299-5p, miR-410, miR-382, miR-409-3p, miR-487b, miR-31, and miR-127 by quantitative RT-PCR and determined that they were similarly expressed in embryonic lungs. We did not find evidence for differential methylation in this region, but analysis of transcription factor binding sites identified multiple SMAD3-binding elements in the 14q32 microRNA cluster. TGF-β1 stimulation of normal human lung fibroblasts (NHLF) caused up-regulation of microRNAs on chr14q32 that were also increased in IPF lungs. Chromatin immunoprecipitation confirmed binding of SMAD3 to the putative promoter of miR-154. Mir-154 was increased in IPF fibroblasts, and transfection of NHLF with miR-154 caused significant increases in cell proliferation and migration. The increase in proliferation induced by TGF-β was not observed when NHLF or IPF fibroblasts were transfected with a mir-154 inhibitor. Transfection with miR-154 caused activation of the WNT pathway in NHLF. ICG-001 and XAV939, inhibitors of the WNT/β-catenin pathway, reduced the proliferative effect of miR-154. The potential role of miR-154, one of multiple chr14q32 microRNA cluster members up-regulated in IPF and a regulator of fibroblast migration and proliferation, should be further explored in IPF. AU - Milosevic, J.* AU - Pandit, K.* AU - Magister, M.* AU - Rabinovich, E.* AU - Ellwanger, D.C. AU - Yu, G.* AU - Vuga, L.J.* AU - Weksler, B.* AU - Benos, P.V.* AU - Gibson, K.F.* AU - McMillan, M.* AU - Kahn, M.* AU - Kaminski, N.* C1 - 11314 C2 - 30607 SP - 879-887 TI - Profibrotic role of miR-154 in pulmonary fibrosis. JO - Am. J. Respir. Cell Mol. Biol. VL - 47 IS - 6 PB - American Thoracic Society PY - 2012 SN - 1044-1549 ER - TY - JOUR AB - High concentrations of particulate matter (PM(10)) were measured in classrooms. This study addresses the hazard of indoor particles in comparison to the better-studied outdoor particles. Samples were taken from six schools during teaching hours. Genome-wide gene expression in human BEAS-2B lung epithelial cells was analyzed and verified by quantitative PCR. Polycyclic aromatic hydrocarbons, endotoxin, and cat allergen (Fel d 1) were analyzed by standard methods. Enhancement of allergic reactivity by PM(10) was confirmed in human primary basophils. Acceleration of human blood coagulation was determined with supernatants of PM(10)-exposed human peripheral blood monocytes. Indoor PM(10) induced serine protease inhibitor B2 (involved in blood coagulation) and inflammatory genes (such as CXCL6, CXCL1, IL6, IL8; all P < 0.001). Outdoor PM(10) induced xenobiotic metabolizing enzymes (cytochrome P450 [CYP] 1A1, CYP1B1, TIPARP; all P < 0.001). The induction of inflammatory genes by indoor PM(10) was explained by endotoxin (indoor 128.5 ± 42.2 EU/mg versus outdoor 13.4 ± 21.5 EU/mg; P < 0.001), the induction of CYP by outdoor polycyclic aromatic hydrocarbons (indoor 8.3 ± 4.9 ng/mg versus outdoor 16.7 ± 15.2 ng/mg; P < 0.01). The induction of serine protease inhibitor B2 was confirmed by a more rapid human blood coagulation (P < 0.05). Indoor PM(10) only affected allergic reactivity from human primary basophils from cat-allergic individuals. This was explained by varying Fel d 1 concentrations in indoor PM(10) (P < 0.001). Indoor PM(10), compared with outdoor PM(10), was six times higher and, on an equal weight basis, induced more inflammatory and allergenic reactions, and accelerated blood coagulation. Outdoor PM(10) had significantly lower effects, but induced detoxifying enzymes. Therefore, preliminary interventions for the reduction of classroom PM(10) seem reasonable, perhaps through intensified ventilation. AU - Oeder, S. AU - Jörres, R.A.* AU - Weichenmeier, I. AU - Pusch, G. AU - Schober, W. AU - Pfab, F. AU - Behrendt, H.* AU - Schierl, R.* AU - Kronseder, A.* AU - Nowak, D.* AU - Dietrich, S.* AU - Fernández-Caldas, E.* AU - Lintelmann, J. AU - Zimmermann, R. AU - Lang, R.* AU - Mages, J.* AU - Fromme, H.* AU - Buters, J.T.M. C1 - 11286 C2 - 30585 SP - 575-582 TI - Airborne indoor particles from schools are more toxic than outdoor particles. JO - Am. J. Respir. Cell Mol. Biol. VL - 47 IS - 5 PB - American Thoracic Society PY - 2012 SN - 1044-1549 ER - TY - JOUR AB - The WNT family of signaling proteins is essential to organ development in general and lung morphogenesis in particular. Originally identified as a developmentally active signaling pathway, the WNT pathway has recently been linked to the pathogenesis of important lung diseases, in particular lung cancer and pulmonary fibrosis. This review summarizes our current understanding about WNT signaling in lung development and disease, and is structured into three chapters. The first chapter presents an introduction to WNT signaling, outlining WNT proteins, their receptors and signaling intermediates, as well as the regulation of this complex pathway. The second chapter focuses on the role of WNT signaling in the normal embryonic and adult lung, and highlights recent findings of altered WNT signaling in lung diseases, such as lung cancer, pulmonary fibrosis, or pulmonary arterial hypertension. In the last chapter, we will discuss novel data and ideas about the biological effects of WNT signaling on the cellular level, highlighting pleiotropic effects induced by WNT ligands on distinct cell types, and how these cellular effects may be relevant to the pathogenesis of the aforementioned diseases. AU - Königshoff, M. AU - Eickelberg, O. C1 - 252 C2 - 26815 SP - 21-31 TI - WNT signaling in lung disease: A failure or a regeneration signal? JO - Am. J. Respir. Cell Mol. Biol. VL - 42 IS - 1 PB - American Thoracic Society PY - 2010 SN - 1044-1549 ER - TY - JOUR AB - The role of macrophages in the clearance of particles with diameters less than 100 nm (ultrafine or nanoparticles) is not well established, although these particles deposit highly efficiently in peripheral lungs, where particle phagocytosis by macrophages is the primary clearance mechanism. To investigate the uptake of nanoparticles by lung phagocytes, we analyzed the distribution of titanium dioxide particles of 20 nm count median diameter in macrophages obtained by bronchoalveolar lavage at 1 hour and 24 hours after a 1-hour aerosol inhalation. Differential cell counts revealing greater than 96% macrophages and less than 1% neutrophils and lymphocytes excluded inflammatory cell responses. Employing energy-filtering transmission electron microscopy (EFTEM) for elemental microanalysis, we examined 1,594 macrophage profiles in the 1-hour group (n = 6) and 1,609 in the 24-hour group (n = 6). We found 4 particles in 3 macrophage profiles at 1 hour and 47 particles in 27 macrophage profiles at 24 hours. Model-based data analysis revealed an uptake of 0.06 to 0.12% ultrafine titanium-dioxide particles by lung-surface macrophages within 24 hours. Mean (SD) particle diameters were 31 (8) nm at 1 hour and 34 (10) nm at 24 hours. Particles were localized adjacent (within 13-83 nm) to the membrane in vesicles with mean (SD) diameters of 592 (375) nm at 1 hour and 414 (309) nm at 24 hours, containing other material like surfactant. Additional screening of macrophage profiles by conventional TEM revealed no evidence for agglomerated nanoparticles. These results give evidence for a sporadic and rather unspecific uptake of TiO(2)-nanoparticles by lung-surface macrophages within 24 hours after their deposition, and hence for an insufficient role of the key clearance mechanism in peripheral lungs. AU - Geiser, M.* AU - Casaulta, M.* AU - Kupferschmid, B.* AU - Schulz, S. AU - Semmler-Behnke, M. AU - Kreyling, W.G. C1 - 1536 C2 - 25436 SP - 371-376 TI - The role of macrophages in the clearance of inhaled ultrafine titanium dioxide particles. JO - Am. J. Respir. Cell Mol. Biol. VL - 38 IS - 3 PB - Amer. Thoracic Soc. PY - 2008 SN - 1044-1549 ER - TY - JOUR AB - Human pleural macrophages (PLM) have been studied in effusions, but little is known about normal human PLM. We therefore analyzed resting human PLM recovered by lavage before lobe resection from patients with a central bronchial tumor, not involving the pleura, and from patients with pulmonary chondroma, intrapulmonary hemorrhage, and pneumothorax. Analysis of surface antigens, phagocytosis capacity, and cytokine production was done in comparison to the regular CD14(++) blood monocytes and the recently described blood monocyte subset CD14(+)CD16(+) monocytes. When defining fluorescence intensity for the various markers on CD14(++) monocytes as 100%, the PLM gave the following pattern: CD14, 45%; CD32, 200%; CD64, 72%; CD11b, 128%; CD33, 74%; CD54, 299%; and HLA-DR, 1,906%. When CD16 on the CD14(+)CD16(+) monocytes was set as 100%, the level of CD16 expression on PLM was 7.7%. Taken together, when compared to blood monocytes, PLM appear to represent a cell-type intermediate of regular CD14(++) monocytes and the CD14(+)CD16(+) subset. In functional studies, we demonstrate that PLM can perform efficient Fc-receptor-mediated phagocytosis of antibody-coated sheep red blood cells. Compared with blood monocytes, the capacity of PLM to produce tumor necrosis factor is similar, but a striking finding in PLM was the constitutive interleukin-10 messenger RNA expression that could not be substantially increased by lipopolysaccharide stimulation. This first characterization of normal, noneffusion human PLM can form the basis for a better interpretation of findings in malignant and inflammatory exudates. AU - Frankenberger, M. AU - Passlick, B.* AU - Hofer, T.P.* AU - Siebeck, M.* AU - Maier, K.L.* AU - Ziegler-Heitbrock, L.* C1 - 24074 C2 - 31419 SP - 419-426 TI - Immunologic characterization of normal human pleural macrophages. JO - Am. J. Respir. Cell Mol. Biol. VL - 23 IS - 3 PB - Amer. Thoracic Soc. PY - 2000 SN - 1044-1549 ER - TY - JOUR AB - Intracellular dissolution of inhaled particles is an important pathway of clearance of potentially toxic materials. To study this process, monolayers of human and canine alveolar macrophages (AM) were maintained alive and functional in vitro for more than 2 wk. Complete phagocytosis of moderately soluble, monodisperse 57Co3O4 test particles of four different sizes was obtained by optimizing the cell density of the monolayer and the particle-to-cell ratio. The fraction of the initial particle mass that was soluble increased over time when the particles were ingested by AM but remained constant when in culture medium alone. Smaller particle sizes had a faster characteristic intracellular dissolution rate constant than did larger particles. The dissolution rates differed between AM obtained from two human volunteers as compared to those obtained from six mongrel dogs. These in vitro dissolution rates were very similar to in vivo translocation rates previously obtained from human and canine lung clearance studies after inhalation of the same or similar monodisperse, homogeneous 57Co3O4 test particles. We believe an important clearance mechanism for inhaled aerosol particles deposited in the lungs can be simulated in vitro in a cell culture system. AU - Kreyling, W.G. AU - Godleski, J.J. AU - Kariya, S.T. AU - Rose, R.M. AU - Brain, J.D. C1 - 33821 C2 - 36406 SP - 413-422 TI - In vitro dissolution of uniform cobalt oxide particles by human and canine alveolar macrophages. JO - Am. J. Respir. Cell Mol. Biol. VL - 2 IS - 5 PY - 1990 SN - 1044-1549 ER -