Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Am J Physiol Cell Physiol ; 326(6): C1637-C1647, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38646782

ABSTRACT

Bleomycin (BLM)-induced lung injury in mice is a valuable model for investigating the molecular mechanisms that drive inflammation and fibrosis and for evaluating potential therapeutic approaches to treat the disease. Given high variability in the BLM model, it is critical to accurately phenotype the animals in the course of an experiment. In the present study, we aimed to demonstrate the utility of microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation for rapid phenotyping of BLM mice. µCT was performed in freely breathing C57BL/6J mice under isoflurane anesthesia on days 7 and 21 after BLM administration. Terminal invasive lung function measurement and histological assessment of the left lung collagen content were conducted as well. µCT image analysis demonstrated gradual and time-dependent development of lung injury as evident by alterations in the lung density, air-to-tissue volume ratio, and lung aeration in mice treated with BLM. The right and left lung were unequally affected. µCT-derived parameters such as lung density, air-to-tissue volume ratio, and nonaerated lung volume correlated well with the invasive lung function measurement and left lung collagen content. Our study demonstrates the utility of AI-CNN-powered µCT image analysis for rapid and accurate phenotyping of BLM mice in the course of disease development and progression.NEW & NOTEWORTHY Microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation is a rapid and powerful tool for noninvasive phenotyping of bleomycin mice over the course of the disease. This, in turn, allows earlier and more reliable identification of therapeutic effects of new drug candidates, ultimately leading to the reduction of unnecessary procedures in animals in pharmacological research.


Subject(s)
Bleomycin , Lung Injury , Lung , Mice, Inbred C57BL , Neural Networks, Computer , Phenotype , Animals , Bleomycin/toxicity , Lung Injury/chemically induced , Lung Injury/diagnostic imaging , Lung Injury/pathology , Lung Injury/metabolism , Lung/diagnostic imaging , Lung/drug effects , Lung/pathology , Lung/metabolism , Mice , X-Ray Microtomography/methods , Disease Models, Animal , Artificial Intelligence , Male , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/diagnostic imaging , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Collagen/metabolism
2.
Am J Respir Crit Care Med ; 207(3): 283-299, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36047984

ABSTRACT

Rationale: Although type II alveolar epithelial cells (AEC2s) are chronically injured in idiopathic pulmonary fibrosis (IPF), they contribute to epithelial regeneration in IPF. Objectives: We hypothesized that Notch signaling may contribute to AEC2 proliferation, dedifferentiation characterized by loss of surfactant processing machinery, and lung fibrosis in IPF. Methods: We applied microarray analysis, kinome profiling, flow cytometry, immunofluorescence analysis, western blotting, quantitative PCR, and proliferation and surface activity analysis to study epithelial differentiation, proliferation, and matrix deposition in vitro (AEC2 lines, primary murine/human AEC2s), ex vivo (human IPF-derived precision-cut lung slices), and in vivo (bleomycin and pepstatin application, Notch1 [Notch receptor 1] intracellular domain overexpression). Measurements and Main Results: We document here extensive SP-B and -C (surfactant protein-B and -C) processing defects in IPF AEC2s, due to loss of Napsin A, resulting in increased intra-alveolar surface tension and alveolar collapse and induction of endoplasmic reticulum stress in AEC2s. In vivo pharmacological inhibition of Napsin A results in the development of AEC2 injury and overt lung fibrosis. We also demonstrate that Notch1 signaling is already activated early in IPF and determines AEC2 fate by inhibiting differentiation (reduced lamellar body compartment, reduced capacity to process hydrophobic SP) and by causing increased epithelial proliferation and development of lung fibrosis, putatively via altered JAK (Janus kinase)/Stat (signal transducer and activator of transcription) signaling in AEC2s. Conversely, inhibition of Notch signaling in IPF-derived precision-cut lung slices improved the surfactant processing capacity of AEC2s and reversed fibrosis. Conclusions: Notch1 is a central regulator of AEC2 fate in IPF. It induces alveolar epithelial proliferation and loss of Napsin A and of surfactant proprotein processing, and it contributes to fibroproliferation.


Subject(s)
Idiopathic Pulmonary Fibrosis , Pulmonary Surfactants , Humans , Mice , Animals , Surface-Active Agents , Lung , Alveolar Epithelial Cells , Bleomycin , Receptor, Notch1
3.
FASEB J ; 33(11): 12392-12408, 2019 11.
Article in English | MEDLINE | ID: mdl-31431059

ABSTRACT

Insufficient autophagy has been reported in idiopathic pulmonary fibrosis (IPF) lungs. Specific roles of autophagy-related proteins in lung fibrosis development remain largely unknown. Here, we investigated the role of autophagy marker protein microtubule-associated protein 1 light chain 3ß (LC3B) in the development of lung fibrosis. LC3B-/- mice upon aging show smaller lamellar body profiles, increased cellularity, alveolar epithelial cell type II (AECII) apoptosis, surfactant alterations, and lysosomal and endoplasmic reticulum stress. Autophagosomal soluble N-ethylmaleimide-sensitive factor attachment protein receptor syntaxin 17 is increased in the AECII of aged LC3B-/- mice and patients with IPF. Proteasomal activity, however, remained unaltered in LC3B-/- mice. In vitro knockdown of LC3B sensitized mouse lung epithelial cells to bleomycin-induced apoptosis, but its overexpression was protective. In vivo, LC3B-/- mice displayed increased susceptibility to bleomycin-induced lung injury and fibrosis. We identified cathepsin A as a novel LC3B binding partner and its overexpression in vitro drives MLE12 cells to apoptosis. Additionally, cathepsin A is increased in the AECII of aged LC3B-/- mice and in the lungs of patients with IPF. Our study reveals that LC3B mediated autophagy plays essential roles in AECII by modulating the functions of proteins like cathepsin A and protects alveolar epithelial cells from apoptosis and subsequent lung injury and fibrosis.-Kesireddy, V. S., Chillappagari, S., Ahuja, S., Knudsen, L., Henneke, I., Graumann, J., Meiners, S., Ochs, M., Ruppert, C., Korfei, M., Seeger, W., Mahavadi, P. Susceptibility of microtubule-associated protein 1 light chain 3ß (MAP1LC3B/LC3B) knockout mice to lung injury and fibrosis.


Subject(s)
Alveolar Epithelial Cells , Apoptosis/genetics , Genetic Predisposition to Disease , Microtubule-Associated Proteins/deficiency , Pulmonary Fibrosis , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Cathepsin A/genetics , Cathepsin A/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism
4.
Am J Respir Crit Care Med ; 199(11): 1407-1420, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30557519

ABSTRACT

Rationale: Pulmonary arterial hypertension (PAH) is characterized by vascular remodeling and excessive proliferation of pulmonary artery smooth muscle cells (PASMCs). Fucoidan, a polysaccharidic ligand of the adhesion molecule P-selectin, exhibits antiproliferative properties. The effects of the fucoidan/P-selectin axis on vascular remodeling and pulmonary hypertension (PH) after hypoxia remain unexplored. Objectives: We aimed to evaluate the therapeutic potential of targeting the fucoidan/P-selectin axis in PH. Methods: Mice with PH induced by chronic hypoxia (35 d) were given either fucoidan (from Fucus vesiculosus) or anti-P-selectin antibody (Rb40.34) during Days 21-35. Right ventricular (RV) function was determined by echocardiography. Vascular morphometry was assessed by immunohistochemistry. Human and experimental PH lungs and PASMCs were used for assessment of P-selectin expression and function. Measurements and Main Results: Fucoidan attenuated chronic hypoxia-induced PH in mice, reducing pulmonary vascular remodeling and restoring RV function. In vitro, fucoidan inhibited hypoxia and growth factor-stimulated PASMC proliferation and migration. Chronic hypoxia caused an upregulation of P-selectin in the medial layer of the small pulmonary arteries. P-selectin was persistently upregulated in PASMCs of human and hypoxia-induced experimental PH. HIF-1α (hypoxia-inducible factor 1α) directly bound to the P-selectin promoter and transcriptionally activated P-selectin in hypoxia. P-selectin blockage resulted in a marked reduction of PASMC proliferation in vitro. Blockage of P-selectin by administration of anti-P-selectin Rb40.34 antibody and P-selectin-deficient mice improved vascular remodeling and restored RV function. Conclusions: Fucoidan is a potent natural adjuvant that represents a promising therapeutic approach for PH. Our data indicate a previously unrecognized role of P-selectin in the proliferative response of PASMCs associated with PH.


Subject(s)
Anticoagulants/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypoxia/complications , Myocytes, Smooth Muscle/drug effects , P-Selectin/blood , Polysaccharides/therapeutic use , Animals , Biomarkers/blood , Disease Models, Animal , Fucus/chemistry , Humans , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Mice
6.
Am J Respir Crit Care Med ; 196(2): 186-199, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28005404

ABSTRACT

RATIONALE: Acute respiratory distress syndrome is characterized by alveolar epithelial cell injury, edema formation, and intraalveolar contact phase activation. OBJECTIVES: To explore whether C1 esterase inhibitor (C1INH), an endogenous inhibitor of the contact phase, may protect from lung injury in vivo and to decipher the possible underlying mechanisms mediating protection. METHODS: The ability of C1INH to control the inflammatory processes was studied in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Here, we demonstrate that application of C1INH alleviates bleomycin-induced lung injury via direct interaction with extracellular histones. In vitro, C1INH was found to bind all histone types. Interaction with histones was independent of its protease inhibitory activity, as demonstrated by the use of reactive-center-cleaved C1INH, but dependent on its glycosylation status. C1INH sialylated-N- and -O-glycans were not only essential for its interaction with histones but also to protect against histone-induced cell death. In vivo, histone-C1INH complexes were detected in bronchoalveolar lavage fluid from patients with acute respiratory distress syndrome and multiple models of lung injury. Furthermore, reactive-center-cleaved C1INH attenuated pulmonary damage evoked by intravenous histone instillation. CONCLUSIONS: Collectively, C1INH administration provides a new therapeutic option for disorders associated with histone release.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Histones/metabolism , Lung Injury/prevention & control , Respiratory Distress Syndrome/physiopathology , Animals , Bronchoalveolar Lavage Fluid , Complement C1 Inhibitor Protein/metabolism , Disease Models, Animal , Humans , Lung/metabolism , Lung/physiopathology , Lung Injury/physiopathology , Mice , Mice, Inbred C57BL
8.
Am J Physiol Lung Cell Mol Physiol ; 310(6): L519-31, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26719147

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disorder, and some patients with HPS develop pulmonary fibrosis, known as HPS-associated interstitial pneumonia (HPSIP). We have previously reported that HPSIP is associated with severe surfactant accumulation, lysosomal stress, and alveolar epithelial cell type II (AECII) apoptosis. Here, we hypothesized that defective autophagy might result in excessive lysosomal stress in HPSIP. Key autophagy proteins, including LC3B lipidation and p62, were increased in HPS1/2 mice lungs. Electron microscopy demonstrated a preferable binding of LC3B to the interior of lamellar bodies in the AECII of HPS1/2 mice, whereas in wild-type mice it was present on the limiting membrane in addition to the interior of the lamellar bodies. Similar observations were noted in human HPS1 lung sections. In vitro knockdown of HPS1 revealed increased LC3B lipidation and p62 accumulation, associated with an increase in proapoptotic caspases. Overexpression of LC3B decreased the HPS1 knockdown-induced p62 accumulation, whereas rapamycin treatment did not show the same effect. We conclude that loss of HPS1 protein results in impaired autophagy that is restored by exogenous LC3B and that defective autophagy might therefore play a critical role in the development and progression of HPSIP.


Subject(s)
Alveolar Epithelial Cells/physiology , Autophagy , Hermanski-Pudlak Syndrome/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Female , Hermanski-Pudlak Syndrome/pathology , Humans , Lung/metabolism , Lung/pathology , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics
9.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L63-75, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25957292

ABSTRACT

Chronic injury of alveolar epithelial type II cells (AE2 cells) represents a key event in the development of lung fibrosis in animal models and in humans, such as idiopathic pulmonary fibrosis (IPF). Intratracheal delivery of amiodarone to mice results in a profound injury and macroautophagy-dependent apoptosis of AE2 cells. Increased autophagy manifested in AE2 cells by disturbances of the intracellular surfactant. Hence, we hypothesized that ultrastructural alterations of the intracellular surfactant pool are signs of epithelial stress correlating with the severity of fibrotic remodeling. With the use of design-based stereology, the amiodarone model of pulmonary fibrosis in mice was characterized at the light and ultrastructural level during progression. Mean volume of AE2 cells, volume of lamellar bodies per AE2 cell, and mean size of lamellar bodies were correlated to structural parameters reflecting severity of fibrosis like collagen content. Within 2 wk amiodarone leads to an increase in septal wall thickness and a decrease in alveolar numbers due to irreversible alveolar collapse associated with alveolar surfactant dysfunction. Progressive hypertrophy of AE2 cells and increase in mean individual size and total volume of lamellar bodies per AE2 cell were observed. A high positive correlation of these AE2 cell-related ultrastructural changes and the deposition of collagen fibrils within septal walls were established. Qualitatively, similar alterations could be found in IPF samples with mild to moderate fibrosis. We conclude that ultrastructural alterations of AE2 cells including the surfactant system are tightly correlated with the progression of fibrotic remodeling.


Subject(s)
Airway Remodeling/physiology , Idiopathic Pulmonary Fibrosis/pathology , Pulmonary Alveoli/pathology , Pulmonary Surfactants/metabolism , Respiratory Mucosa/pathology , Amiodarone/toxicity , Animals , Apoptosis/drug effects , Cell Size/drug effects , Disease Models, Animal , Epithelial Cells/pathology , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Respiratory Mucosa/cytology , Vasodilator Agents/toxicity
10.
Am J Physiol Lung Cell Mol Physiol ; 308(10): L1014-24, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25820524

ABSTRACT

Fibroblast growth factors (Fgfs) mediate organ repair. Lung epithelial cell overexpression of Fgf10 postbleomycin injury is both protective and therapeutic, characterized by increased survival and attenuated fibrosis. Exogenous administration of FGF7 (palifermin) also showed prophylactic survival benefits in mice. The role of endogenous Fgfr2b ligands on bleomycin-induced lung fibrosis is still elusive. This study reports the expression of endogenous Fgfr2b ligands, receptors, and signaling targets in wild-type mice following bleomycin lung injury. In addition, the impact of attenuating endogenous Fgfr2b-ligands following bleomycin-induced fibrosis was tested by using a doxycycline (dox)-based inducible, soluble, dominant-negative form of the Fgfr2b receptor. Double-transgenic (DTG) Rosa26(rtTA/+);tet(O)solFgfr2b mice were validated for the expression and activity of soluble Fgfr2b (failure to regenerate maxillary incisors, attenuated recombinant FGF7 signal in the lung). As previously reported, no defects in lung morphometry were detected in DTG (+dox) mice exposed from postnatal days (PN) 1 through PN105. Female single-transgenic (STG) and DTG mice were subjected to various levels of bleomycin injury (1.0, 2.0, and 3.0 U/kg). Fgfr2b ligands were attenuated either throughout injury (days 0-11; days 0-28) or during later stages (days 6-28 and 14-28). No significant changes in survival, weight, lung function, confluent areas of fibrosis, or hydroxyproline deposition were detected in DTG mice. These results indicate that endogenous Fgfr2b ligands do not significantly protect against bleomycin injury, nor do they expedite the resolution of bleomycin-induced lung injury in mice.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Fibroblast Growth Factor 7/pharmacology , Pulmonary Fibrosis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Female , Mice , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptor, Fibroblast Growth Factor, Type 2/agonists , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/genetics
11.
Thorax ; 70(11): 1022-32, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26359372

ABSTRACT

BACKGROUND: Activation and differentiation of fibroblasts into contractile protein-expressing myofibroblasts and their acquired apoptosis-resistant phenotype are critical factors towards the development of idiopathic pulmonary fibrosis (IPF), a fatal disease characterised by distorted pulmonary structure and excessive extracellular matrix (ECM) deposition. The molecular mechanisms underlying these processes in IPF remain incompletely understood. We investigated the possible implication of aberrant overexpression and activity of histone deacetylases (HDACs) in IPF. METHODS: We analysed lung tissues from patients with sporadic IPF (n=26) and non-diseased control lungs (n=16) for expression of class I and II HDACs. Primary IPF fibroblasts were treated with HDAC inhibitors (HDACi) LBH589 or valproic acid (VPA). RESULTS: Compared to control lungs, protein levels of class I (HDAC1, HDAC2, HDAC3, HDAC8) and class II HDACs (HDAC4, HDAC 5, HDAC 7, HDAC 9) were significantly elevated in IPF lungs. Using immunohistochemistry, strong induction of nearly all HDAC enzymes was observed in myofibroblasts of fibroblast foci and in abnormal bronchiolar basal cells at sites of aberrant re-epithelialisation in IPF lungs, but not in controls. Treatment of primary IPF fibroblasts with the pan-HDACi LBH589 resulted in significantly reduced expression of genes associated with ECM synthesis, proliferation and cell survival, as well as in suppression of HDAC7, and was paralleled by induction of endoplasmic reticulum stress and apoptosis. The profibrotic and apoptosis-resistant phenotype of IPF fibroblasts was also partly attenuated by the class I HDACi VPA. CONCLUSIONS: Aberrant overexpression of HDACs in basal cells of IPF lungs may contribute to the bronchiolisation process in this disease. Similarly, generation and apoptosis resistance of IPF fibroblasts are mediated by enhanced activity of HDAC enzymes. Therefore, pan-HDAC inhibition by LBH589 may present a novel therapeutic option for patients with IPF.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/genetics , Idiopathic Pulmonary Fibrosis/genetics , RNA/genetics , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Histone Deacetylase 2/biosynthesis , Histone Deacetylase 2/genetics , Histone Deacetylases/biosynthesis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Male , Middle Aged , Real-Time Polymerase Chain Reaction
12.
Respir Res ; 16: 83, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138239

ABSTRACT

BACKGROUND: Recent clinical studies show that tyrosine kinase inhibitors slow the rate of lung function decline and decrease the number of acute exacerbations in patients with Idiopathic Pulmonary Fibrosis (IPF). However, in the murine bleomycin model of fibrosis, not all tyrosine kinase signaling is detrimental. Exogenous ligands Fibroblast Growth Factor (FGF) 7 and 10 improve murine lung repair and increase survival after injury via tyrosine kinase FGF receptor 2b-signaling. Therefore, the level and location of FGF/FGFR expression as well as the exogenous effect of the most highly expressed FGFR2b ligand, FGF1, was analyzed on human lung fibroblasts. METHODS: FGF ligand and receptor expression was evaluated in donor and IPF whole lung homogenates using western blotting and qPCR. Immunohistochemistry for FGF1 and FGFR1/2/3/4 were performed on human lung tissue. Lastly, the effects of FGF1, a potent, multi-FGFR ligand, were studied on primary cultures of IPF and non-IPF donor fibroblasts. Western blots for pro-fibrotic markers, proliferation, FACS for apoptosis, transwell assays and MetaMorph analyses on cell cultures were performed. RESULTS: Whole lung homogenate analyses revealed decreased FGFR b-isoform expression, and an increase in FGFR c-isoform expression. Of the FGFR2b-ligands, FGF1 was the most significantly increased in IPF patients; downstream targets of FGF-signaling, p-ERK1/2 and p-AKT were also increased. Immunohistochemistry revealed FGF1 co-localization within basal cell sheets, myofibroblast foci, and Surfactant protein-C positive alveolar epithelial type-II cells as well as co-localization with FGFR1, FGFR2, FGFR3, FGFR4 and myofibroblasts expressing the migratory marker Fascin. Both alone and in the presence of heparin, FGF1 led to increased MAPK-signaling in primary lung fibroblasts. While smooth muscle actin was unchanged, heparin + FGF1 decreased collagen production in IPF fibroblasts. In addition, FGF1 + heparin increased apoptosis and cell migration. The FGFR inhibitor (PD173074) attenuated these effects. CONCLUSIONS: Strong expression of FGF1/FGFRs in pathogenic regions of IPF suggest that aberrant FGF1-FGFR signaling is increased in IPF patients and may contribute to the pathogenesis of lung fibrosis by supporting fibroblast migration and increased MAPK-signaling.


Subject(s)
Fibroblast Growth Factor 1/biosynthesis , Idiopathic Pulmonary Fibrosis/metabolism , Receptors, Fibroblast Growth Factor/biosynthesis , Cell Movement/physiology , Cells, Cultured , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology
13.
Cell Mol Life Sci ; 70(19): 3695-708, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23619613

ABSTRACT

Posttranslational modification of the neural cell adhesion molecule (NCAM) by polysialic acid (polySia) is well studied in the nervous system and described as a dynamic modulator of plastic processes like precursor cell migration, axon fasciculation, and synaptic plasticity. Here, we describe a novel function of polysialylated NCAM (polySia-NCAM) in innate immunity of the lung. In mature lung tissue of healthy donors, polySia was exclusively attached to the transmembrane isoform NCAM-140 and located to intracellular compartments of epithelial cells. In patients with chronic obstructive pulmonary disease, however, increased polySia levels and processing of the NCAM carrier were observed. Processing of polysialylated NCAM was reproduced in a mouse model by bleomycin administration leading to an activation of the inflammasome and secretion of interleukin (IL)-1ß. As shown in a cell culture model, polySia-NCAM-140 was kept in the late trans-Golgi apparatus of lung epithelial cells and stimulation by IL-1ß or lipopolysaccharide induced metalloprotease-mediated ectodomain shedding, resulting in the secretion of soluble polySia-NCAM. Interestingly, polySia chains of secreted NCAM neutralized the cytotoxic activity of extracellular histones as well as DNA/histone-network-containing "neutrophil extracellular traps", which are formed during invasion of microorganisms. Thus, shedding of polySia-NCAM by lung epithelial cells may provide a host-protective mechanism to reduce tissue damage during inflammatory processes.


Subject(s)
Immunity, Innate/immunology , Lung/immunology , Neural Cell Adhesion Molecules/immunology , Sialic Acids/immunology , Adult , Animals , Cell Line, Tumor , Epithelial Cells/immunology , Female , Histones/immunology , Humans , Inflammasomes/immunology , Interleukin-1beta/immunology , Lipopolysaccharides/immunology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Models, Molecular , Neutrophils/immunology , Protein Isoforms , Protein Processing, Post-Translational , Pulmonary Disease, Chronic Obstructive/immunology , Up-Regulation/immunology , trans-Golgi Network/immunology
14.
Am J Respir Crit Care Med ; 183(12): 1703-14, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21471103

ABSTRACT

RATIONALE: Activation of the coagulation cascade has been demonstrated in pulmonary fibrosis. In addition to its procoagulant function, various coagulation proteases exhibit cellular effects that may also contribute to fibrotic processes in the lung. OBJECTIVE: To investigate the importance of protease-activated receptor (PAR)-2 and its activators, coagulation factor VIIa (FVIIa)/tissue factor (TF), in the development of idiopathic pulmonary fibrosis (IPF). METHODS: Expression and localization of PAR-2 and its activators were examined in IPF lung tissue. The ability of PAR-2 to mediate various cellular processes was studied in vitro. MEASUREMENTS AND MAIN RESULTS: Expression of PAR-2 was strongly elevated in IPF lungs and was attributable to alveolar type II cells and fibroblasts/myofibroblasts. Transforming growth factor-ß(1), a key profibrotic cytokine, considerably enhanced PAR-2 expression in human lung fibroblasts. FVIIa stimulated proliferation of human lung fibroblasts and extracellular matrix production in a PAR-2-dependent manner, but did not initiate differentiation of fibroblasts into myofibroblasts. PAR-2/FVIIa-driven mitogenic activities were mediated via the p44/42 mitogen-activated protein kinase pathway and were independent of factor Xa and thrombin production. Proproliferative properties of FVIIa were markedly potentiated in the presence of TF and abrogated by TF antisense oligonucleotides. Hyperplastic alveolar type II cells overlying fibroblastic foci were found to be the source of FVII in IPF lungs. Moreover, TF colocalized with PAR-2 on fibroblasts/myofibroblasts in IPF lungs. CONCLUSIONS: The PAR-2/TF/FVIIa axis may contribute to the development of pulmonary fibrosis; thus, interference with this pathway confers novel therapeutic potential for the treatment of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/etiology , Receptor, PAR-2/physiology , Cell Differentiation/physiology , Factor VIIa/physiology , Factor Xa/physiology , Female , Fibroblasts/pathology , Fibronectins/biosynthesis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , In Vitro Techniques , Lung/pathology , Male , Middle Aged , Mitosis , Myofibroblasts/pathology , Osteopontin/biosynthesis , Pulmonary Alveoli/pathology , Receptor, PAR-2/analysis , Thrombin/biosynthesis , Thromboplastin/physiology , Transforming Growth Factor beta/pharmacology
15.
J Proteome Res ; 10(5): 2185-205, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21319792

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease for which no effective therapy exists to date. To identify the molecular mechanisms underlying IPF, we performed comparative proteome analysis of lung tissue from patients with sporadic IPF (n = 14) and human donor lungs (controls, n = 10) using two-dimensional gel electrophoresis and MALDI-TOF-MS. Eighty-nine differentially expressed proteins were identified, from which 51 were up-regulated and 38 down-regulated in IPF. Increased expression of markers for the unfolded protein response (UPR), heat-shock proteins, and DNA damage stress markers indicated a chronic cell stress-response in IPF lungs. By means of immunohistochemistry, induction of UPR markers was encountered in type-II alveolar epithelial cells of IPF but not of control lungs. In contrast, up-regulation of heat-shock protein 27 (Hsp27) was exclusively observed in proliferating bronchiolar basal cells and associated with aberrant re-epithelialization at the bronchiolo-alveolar junctions. Among the down-regulated proteins in IPF were antioxidants, members of the annexin family, and structural epithelial proteins. In summary, our results indicate that IPF is characterized by epithelial cell injury, apoptosis, and aberrant epithelial proliferation.


Subject(s)
Gene Expression Regulation/genetics , Genetic Markers/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Proteins/metabolism , Proteomics/methods , Transplants , DNA Damage/genetics , Electrophoresis, Gel, Two-Dimensional , Heat-Shock Proteins/genetics , Humans , Idiopathic Pulmonary Fibrosis/pathology , Immunohistochemistry , Lung/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Unfolded Protein Response/genetics
16.
Blood ; 113(22): 5588-98, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19182206

ABSTRACT

Cell surface-associated proteolysis plays a crucial role in the migration of mononuclear phagocytes to sites of inflammation. The glycolytic enzyme enolase-1 (ENO-1) binds plasminogen at the cell surface, enhancing local plasmin production. This study addressed the role played by ENO-1 in lipopolysaccharide (LPS)-driven chemokine-directed monocyte migration and matrix invasion in vitro, as well as recruitment of monocytes to the alveolar compartment in vivo. LPS rapidly up-regulated ENO-1 cell-surface expression on human blood monocytes and U937 cells due to protein translocation from cytosolic pools, which increased plasmin generation, enhanced monocyte migration through epithelial monolayers, and promoted matrix degradation. These effects were abrogated by antibodies directed against the plasminogen binding site of ENO-1. Overexpression of ENO-1 in U937 cells increased their migratory and matrix-penetrating capacity, which was suppressed by overexpression of a truncated ENO-1 variant lacking the plasminogen binding site (ENO-1DeltaPLG). In vivo, intratracheal LPS application in mice promoted alveolar recruitment of monocytic cells that overexpressed ENO-1, but not of cells overexpressing ENO-1DeltaPLG. Consistent with these data, pneumonia-patients exhibited increased ENO-1 cell-surface expression on blood monocytes and intense ENO-1 staining of mononuclear cells in the alveolar space. These data suggest an important mechanism of inflammatory cell invasion mediated by increased cell-surface expression of ENO-1.


Subject(s)
Biomarkers, Tumor/physiology , Chemotaxis, Leukocyte/drug effects , DNA-Binding Proteins/physiology , Monocytes/drug effects , Phosphopyruvate Hydratase/physiology , Plasminogen/pharmacology , Pneumonia/immunology , Tumor Suppressor Proteins/physiology , Acute Disease , Animals , Antibodies/pharmacology , Antigens, Surface/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Chemotaxis, Leukocyte/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Monocytes/metabolism , Phosphopyruvate Hydratase/antagonists & inhibitors , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Pneumonia/pathology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , U937 Cells
17.
Am J Respir Crit Care Med ; 182(2): 207-19, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20378731

ABSTRACT

RATIONALE: The molecular mechanisms underlying Hermansky-Pudlak syndrome-associated interstitial pneumonia (HPSIP) are poorly understood but, as in idiopathic pulmonary fibrosis, may be linked to chronic alveolar epithelial type II cell (AECII) injury. OBJECTIVES: We studied the development of fibrosis and the role of AECII injury in various murine models of HPS. METHODS: HPS1, HPS2, and HPS6 monomutant mice, and HPS1/2 and HPS1/6 double-mutant and genetic background mice, were killed at 3 and 9 months of age. Quantitative morphometry was undertaken in lung sections stained with hemalaun-eosin. The extent of lung fibrosis was assessed by trichrome staining and hydroxyproline measurement. Surfactant lipids were analyzed by electrospray ionization mass spectrometry. Surfactant proteins, apoptosis, and lysosomal and endoplasmic reticulum stress markers were studied by Western blotting and immunohistochemistry. Cell proliferation was measured by water-soluble tetrazolium salt-1 and bromodeoxyuridine assays. MEASUREMENTS AND MAIN RESULTS: Spontaneous and slowly progressive HPSIP was observed in HPS1/2 double mutants, but not in other HPS mutants, with subpleural onset at 3 months and full-blown fibrosis at 9 months. In these mice, extensive surfactant abnormalities were encountered in AECII and were paralleled by early lysosomal stress (cathepsin D induction), late endoplasmic reticulum stress (activating transcription factor-4 [ATF4], C/EBP homologous protein [CHOP] induction), and marked apoptosis. These findings were fully corroborated in human HPSIP. In addition, cathepsin D overexpression resulted in apoptosis of MLE-12 cells and increased proliferation of NIH 3T3 fibroblasts incubated with conditioned medium of the transfected cells. CONCLUSIONS: Extensively impaired surfactant trafficking and secretion underlie lysosomal and endoplasmic reticulum stress with apoptosis of AECII in HPSIP, thereby causing the development of HPSIP.


Subject(s)
Apoptosis , Hermanski-Pudlak Syndrome/pathology , Lung Diseases, Interstitial/pathology , Lung/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Bronchoalveolar Lavage Fluid , Cathepsin D/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Epithelial Cells/pathology , Fibrosis , Intercellular Signaling Peptides and Proteins , Lung/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Mice , Peptides/metabolism , Phospholipids/metabolism , Pulmonary Alveoli/cytology , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C , Transcription Factor 4 , Transcription Factor CHOP/metabolism , Up-Regulation
18.
Am J Respir Crit Care Med ; 181(6): 611-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20056905

ABSTRACT

RATIONALE: Lung cancer is the most common malignancy in humans. Urokinase (uPA) plays a crucial role in carcinogenesis by facilitating tumor cell invasion and metastasis. OBJECTIVES: We investigated the effect of the highly specific urokinase inhibitor CJ-463 (benzylsulfonyl-D-Ser-Ser-4-amidinobenzylamide) on tumor growth, metastasis formation, and tumor vascularization in the murine Lewis lung carcinoma (LLC) and a human small lung cancer model. METHODS: A quantity of 3 x 10(6) LLC cells were subcutaneously injected into the right flank of C57Bl6/N mice, uPA knock out, and uPA receptor knockout mice. Seven days later mice were randomized to receive intraperitoneally either saline (control group), CJ-463 (10 and 100 mg/kg, twice a day), or its ineffective stereoisomer (10 mg/kg, twice a day). Tumor volume was measured every second day and metastasis formation was monitored by volumetric-computed tomography. Twelve days after onset of treatment mice were killed and tumors were prepared for histologic examination. MEASUREMENTS AND MAIN RESULTS: Treatment with CJ-463 resulted in a significant inhibition of primary tumor growth, with the highest efficacy seen in the 100 mg/kg group. In addition, histological analysis of the lung revealed a significant reduction in lung micrometastasis in the 100 mg/kg group. Similarly, a reduced seeding of tumor cells into the lung after intravenous injection of LLC cells was observed in inhibitor-treated mice. In these mice, treatment with CJ-463 appeared not to significantly alter the relative extent of tumor vascularization. In vitro, proliferation of LLC cells remained unchanged upon inhibitor treatment. CJ-463 was found to similarly reduce tumor growth in uPA receptor knockout mice, but was ineffective in uPA knockout mice. CONCLUSIONS: Our results suggest that synthetic low-molecular-weight uPA-inhibitors offer as novel agents for treatment of lung cancer.


Subject(s)
Carcinoma, Lewis Lung/enzymology , Small Cell Lung Carcinoma/enzymology , Urokinase-Type Plasminogen Activator/drug effects , Animals , Benzamidines , Blotting, Western/methods , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/secondary , Cell Culture Techniques/methods , Cell Proliferation/drug effects , Cone-Beam Computed Tomography/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors , Humans , Lung/diagnostic imaging , Lung/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/secondary , Sodium Chloride/administration & dosage , Treatment Outcome , Tumor Burden/drug effects
19.
Crit Care Med ; 38(7): 1584-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20400897

ABSTRACT

OBJECTIVE: Inhalative application of substantial amounts of pulmonary surfactant to the acutely inflamed lung represents a desirable therapeutic approach but was impossible under clinical conditions because of the technical limitations of currently available devices. We developed a new dry powder aerosolizer for administration of a recombinant surfactant protein-C-based surfactant, determined aerosol characteristics, and evaluated its use in animal models of acute lung injury. DESIGN: Laboratory experiment. SETTING: University laboratory. SUBJECTS: Rabbits and mice. INTERVENTIONS: The efficacy of an aerosol application of recombinant surfactant protein-C surfactant was assessed in three animal models of acute lung injury: in rabbits with acute lung injury caused by repetitive lavage with prolonged and injurious ventilation; in rabbits at day 4 after inhalative application of bleomycin; and in bleomycin-challenged, spontaneously breathing mice. MEASUREMENTS AND MAIN RESULTS: Analysis of aerosolizer characteristics revealed favorable properties making inhalative surfactant treatment in acute lung injury/acute respiratory distress syndrome possible. The generated aerosol had a mass median aerodynamic diameter of 1.6 microm, with 85% of all particles being smaller than 5 microm. The average mass of surfactant being aerosolized was approximately 800 mg/min, thus allowing delivery of large amounts of surfactant. Biochemical and biophysical surfactant properties remained unaltered after aerosolization. In both rabbit models aerosolization of approximately 500 mg recombinant surfactant protein-C surfactant resulted in a far-reaching restoration of gas exchange and compliance, with Pao2/Fio2 values approaching control values. In bleomycin-challenged, spontaneously breathing mice, surfactant aerosolization resulted in a restoration of compliance. CONCLUSIONS: The described dry powder aerosolizer may be applicable to surfactant therapy of acute lung injury/acute respiratory distress syndrome. This conclusion is based on four main factors. High doses comparable to those used for intratracheal instillation in humans can be generated within a relatively short time period, the device can be connected to the inspiratory limb of the ventilator circuit, the aerosolized surfactant material is biophysically fully active, and therapeutic efficacy was proven in three different animal models of acute lung injury/acute respiratory distress syndrome.


Subject(s)
Acute Lung Injury/drug therapy , Pulmonary Surfactant-Associated Protein C/therapeutic use , Acute Lung Injury/physiopathology , Administration, Inhalation , Aerosols , Animals , Bronchoalveolar Lavage Fluid/chemistry , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Particle Size , Powders , Pulmonary Surfactant-Associated Protein C/administration & dosage , Pulmonary Surfactant-Associated Protein C/chemistry , Rabbits , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Species Specificity
20.
Am J Respir Crit Care Med ; 179(6): 474-83, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19136374

ABSTRACT

RATIONALE: Acute lung injury (ALI) remains an important cause of mortality in intensive care units. Inflammation is controlled by cytokines and eicosanoids derived from the n-6 fatty acid (FA) arachidonic acid (AA). The n-3 FA eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and mediators derived from EPA and DHA possess reduced inflammatory potency. OBJECTIVES: To determine whether the ability of fat-1 mice to endogenously convert n-6 to n-3 FA, and thus generate an increased ratio of n-3 to n-6 FA, impacts experimental ALI. METHODS: We investigated ALI induced by intratracheal instillation of endotoxin in fat-1 and wild-type (WT) mice, assessing leukocyte numbers, protein concentration, and prostaglandin and cytokine levels in bronchoalveolar lavage fluid, as well as free FA in plasma, and lung ventilator compliance. Body temperature and motor activity of mice--markers of sickness behavior--were also recorded. MEASUREMENTS AND MAIN RESULTS: In ALI, fat-1 mice exhibited significantly reduced leukocyte invasion, protein leakage, and macrophage inflammatory protein-2 and thromboxane B(2) levels in lavage fluid compared with WT mice. Free AA levels were increased in the plasma of WT mice in response to endotoxin, whereas EPA and DHA were increased in the fat-1 group. Ventilator compliance was significantly improved in fat-1 mice. Body temperature and motor activity were decreased in ALI. fat-1 Mice recovered body temperature and motor activity faster. CONCLUSIONS: fat-1 Mice exhibited reduced features of ALI and sickness behavior. Increasing the availability of n-3 FA may thus be beneficial in critically ill patients with ALI.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Eicosapentaenoic Acid/metabolism , Animals , Arachidonic Acid/metabolism , Behavior, Animal , Body Temperature , Bronchoalveolar Lavage Fluid/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chemokine CXCL2/analysis , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Fat Emulsions, Intravenous/pharmacology , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fish Oils/pharmacology , Leukocyte Count , Leukocytes/pathology , Lung/metabolism , Lung/pathology , Mice , Mice, Transgenic , Motor Activity , NF-kappa B/metabolism , Respiration, Artificial , Thromboxane B2/analysis , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL