Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Exp Lung Res ; 47(9): 451-463, 2021 11.
Article in English | MEDLINE | ID: mdl-34739349

ABSTRACT

Purpose: IL-33 is known to induce corticosteroid-resistant eosinophilic inflammation and airway remodeling by activating type 2 innate lymphoid cells (ILC2s). Although the RAS signal pathway plays an important role in IL-33-induced ILC2s activation and airway remodeling, it is not known if RAS inhibitors are effective against refractory asthma. We examined the effects of the RAS inhibitor XRP44X in refractory asthma. Methods: RAS activity were examined by BAL fluid and T-cells isolated from spleen cells in Dermatophagoides pteronyssinus (Dp)-sensitized/challenged acute allergic airway inflammation model. A chronic allergic airway inflammation mouse model was generated by challenged with Dp. XRP44X and/or fluticasone were administrated nasally to different experimental groups. The effects of nasal simultaneous administration of XRP44X or fluticasone were assessed in mice administrated with IL-33 or Dp. Results: RAS activity in CD4+ T cells stimulated by Dp were suppressed by XRP44X. Although fluticasone and XRP44X only improved allergic airway inflammation in mice, XRP44X in combination with fluticasone produced further improvement in not only eosinophilic inflammation but also bronchial subepithelial thickness. XRP44X suppressed IL-5 and IL-13 production from ILC2s, although this effect was not suppressed by fluticasone. IL-33-induced airway inflammation resistant to fluticasone was ameliorated by XRP44X via regulating the accumulation of lung ILC2s. Conclusion: The RAS signal pathway plays a crucial role in allergen-induced airway remodeling associated with ILC2s. XRP44X may have therapeutic potential for refractory asthma.


Subject(s)
Hypersensitivity , Interleukin-33 , Airway Remodeling , Animals , Immunity, Innate , Lymphocytes , Mice
2.
Materials (Basel) ; 13(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066660

ABSTRACT

To develop a high-performance hydrogen gas sensor, we fabricated a composite film made of carbon nanotubes (CNTs) and palladium nanoparticles. Carbon nanotubes were spin-coated onto a glass substrate, and subsequently, palladium nanoparticles were sputtered onto this film. The response to hydrogen gas was measured during two seasons (summer and winter) using a vacuum chamber by introducing a hydrogen/argon gas mixture. There was a clear difference in the sensor response despite the temperature difference between summer and winter. In addition, since a clean chamber was used, fewer water molecules acted as a dopant, and the behavior of the CNT changed from p-type to n-type because of the dissociative adsorption of hydrogen. This phenomenon was confirmed as the Seebeck effect. Finally, the work functions of Pd, PdHx, and CNT were calculated by first-principle calculations. As predicted by previous studies, a decrease in work function due to hydrogen adsorption was confirmed; however, the electron transfer to CNT was not appropriate from the perspective of charge neutrality and was found to be localized at the Pd/CNT interface. It seems that the Seebeck effect causes the concentration of conductive carriers to change.

3.
J Med Invest ; 67(1.2): 102-112, 2020.
Article in English | MEDLINE | ID: mdl-32378592

ABSTRACT

Fibrocytes, which are bone marrow-derived collagen-producing cells, were reported to play a role in the pathogenesis of pulmonary fibrosis. However, their function in pulmonary fibrosis is unclear. We analyzed their function compared with that of monocytes and localization in fibrotic tissues in patients with idiopathic pulmonary fibrosis (IPF). We compared the gene expression profile of monocyte-derived fibrocytes with that of monocytes by microarray analysis. Proliferation and differentiation into myofibroblasts were examined by 3H-thymidine incorporation assay and Western blotting. We measured the level of growth factors in the culture supernatant of fibrocytes by ELISA. The localization of fibrocytes in lung tissues of patients with IPF was determined by immunofluorescence staining. Compared with monocytes, fibrocytes had higher expression of extracellular matrix- and growth factor-encoding genes, including PDGF-B, FGF-2 and VEGF-B. Although fibrocytes did not proliferate in response to PDGF, co-culture of fibrocytes stimulated the growth of lung fibroblasts through the production of PDGF-BB. In the lung of IPF patients, CD45+Collagen-I+FSP-1+ cells, which have a similar phenotype to fibrocytes, were detected and co-stained with anti-PDGF antibody. This study suggested that fibrocytes function in pulmonary fibrosis partly by producing PDGF in the lungs of IPF patients. J. Med. Invest. 67 : 102-112, February, 2020.


Subject(s)
Fibroblasts/physiology , Idiopathic Pulmonary Fibrosis/etiology , Monocytes/cytology , Becaplermin/analysis , Becaplermin/genetics , Becaplermin/physiology , Cell Differentiation , Cells, Cultured , Fibroblast Growth Factor 2/analysis , Fibroblasts/cytology , Humans , Lung/metabolism , Monocytes/metabolism , Myofibroblasts/cytology , Transcriptome
4.
Exp Lung Res ; 45(7): 188-199, 2019 09.
Article in English | MEDLINE | ID: mdl-31298961

ABSTRACT

Purpose/Aim of the Study: Wnt/ß-catenin signaling was reported to be activated in pulmonary fibrosis, and was focused on as a target for antifibrotic therapy. However, the mechanism how the inhibition of Wnt/ß-catenin signaling ameliorate pulmonary fibrosis has not been fully elucidated. The purpose of this study is to explore the target cells of Wnt/ß-catenin inhibition in pulmonary fibrosis and to examine the antifibrotic effect of the novel inhibitor PRI-724 specifically disrupting the interaction of ß-catenin and CBP. Materials and Methods: The effect of C-82, an active metabolite of PRI-724, on the expression of TGF-ß1 and α-smooth muscle actin (SMA) was examined on fibroblasts and macrophages. We also examined the effects of PRI-724 in mouse model of bleomycin-induced pulmonary fibrosis. Results: The activation and increased accumulation of ß-catenin in the canonical pathway were detected in lung fibroblasts as well as macrophages stimulated by Wnt3a using Western blotting. Treatment with C-82 reduced CBP protein and increased p300 protein binding to ß-catenin in the nucleus of lung fibroblasts. In addition, C-82 inhibited the expression of SMA in lung fibroblasts treated with TGF-ß, indicating the inhibition of myofibroblast differentiation. In the fibrotic lungs induced by bleomycin, ß-catenin was stained strongly in macrophages, but the staining of ß-catenin in alveolar epithelial cells and fibroblasts was weak. The administration of PRI-724 ameliorated pulmonary fibrosis induced by bleomycin in mice when administered with a late, but not an early, treatment schedule. Analysis of bronchoalveolar fluid (BALF) showed a decreased number of alveolar macrophages. In addition, the level of TGF-ß1 in BALF was decreased in mice treated with PRI-724. C-82 also inhibited the production of TGF-ß1 by alveolar macrophages. Conclusions: These results suggest that the ß-catenin/CBP inhibitor PRI-724 is a potent antifibrotic agent that acts by modulating the activity of macrophages in the lungs.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Pulmonary Fibrosis/drug therapy , Pyrimidinones/therapeutic use , beta Catenin/antagonists & inhibitors , Animals , Bleomycin , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pyrimidinones/pharmacology , Transforming Growth Factor beta1/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
5.
Cancer Lett ; 421: 17-27, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29448000

ABSTRACT

Cancer stem cells (CSCs) represent a minor population that have clonal tumor initiation and self-renewal capacity and are responsible for tumor initiation, metastasis, and therapeutic resistance. CSCs reside in niches, which are composed of diverse types of stromal cells and extracellular matrix components. These stromal cells regulate CSC-like properties by providing secreted factors or by physical contact. Fibrocytes are differentiated from bone marrow-derived CD14+ monocytes and have features of both macrophages and fibroblasts. Accumulating evidence has suggested that stromal fibrocytes might promote cancer progression. However, the role of fibrocytes in the CSC niches has not been revealed. We herein report that human fibrocytes enhanced the CSC-like properties of lung cancer cells through secreted factors, including osteopontin, CC-chemokine ligand 18, and plasminogen activator inhibitor-1. The PIK3K/AKT pathway was critical for fibrocytes to mediate the CSC-like functions of lung cancer cells. In human lung cancer specimens, the number of tumor-infiltrated fibrocytes was correlated with high expression of CSC-associated protein in cancer cells. These results suggest that fibrocytes may be a novel cell population that regulates the CSC-like properties of lung cancer cells in the CSC niches.


Subject(s)
Adenocarcinoma of Lung/pathology , Lung Neoplasms/pathology , Monocytes/pathology , Neoplastic Stem Cells/pathology , Tumor Microenvironment/physiology , A549 Cells , Adenocarcinoma of Lung/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Heterografts , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, SCID , Monocytes/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism
6.
PLoS One ; 13(12): e0209786, 2018.
Article in English | MEDLINE | ID: mdl-30596712

ABSTRACT

Platelet-derived growth factor (PDGF) has been implicated in the pathogenesis of pulmonary fibrosis. Nintedanib, a multi-kinase inhibitor that targets several tyrosine kinases, including PDGF receptor (PDGFR), was recently approved as an anti-fibrotic agent to reduce the deterioration of FVC in patients with idiopathic pulmonary fibrosis (IPF). However, the effects of PDGFR-α or -ß on pulmonary fibrosis remain unclear. In an attempt to clarify their effects, we herein used blocking antibodies specific for PDGFR-α (APA5) and -ß (APB5) in a bleomycin (BLM)-induced pulmonary fibrosis mouse model. The effects of these treatments on the growth of lung fibroblasts were examined using the 3H-thymidine incorporation assay in vitro. The anti-fibrotic effects of these antibodies were investigated with the Ashcroft score and collagen content of lungs treated with BLM. Their effects on inflammatory cells in the lungs were also analyzed using bronchoalveolar lavage fluid. We investigated damage to epithelial cells and the proliferation of fibroblasts in the lungs. APA5 and APB5 inhibited the phosphorylation of PDGFR-α and -ß as well as the proliferation of lung fibroblasts induced by PDGF-AA and BB. The administration of APB5, but not APA5 effectively inhibited BLM-induced pulmonary fibrosis in mice. Apoptosis and the proliferation of epithelial cells and fibroblasts were significantly decreased by the treatment with APB5, but not by APA5. The late treatment with APB5 also ameliorated fibrosis in lungs treated with BLM. These results suggest that PDGFR-α and -ß exert different effects on BLM-induced pulmonary fibrosis in mice. A specific approach using the blocking antibody for PDGFR-ß may be useful for the treatment of pulmonary fibrosis.


Subject(s)
Bleomycin/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Rats
7.
Respir Res ; 18(1): 172, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28915889

ABSTRACT

BACKGROUND: Nintedanib, a tyrosine kinase inhibitor that is specific for platelet-derived growth factor receptors (PDGFR), fibroblast growth factor receptors (FGFR), and vascular endothelial growth factor receptors (VEGFR), has recently been approved for idiopathic pulmonary fibrosis. Fibrocytes are bone marrow-derived progenitor cells that produce growth factors and contribute to fibrogenesis in the lungs. However, the effects of nintedanib on the functions of fibrocytes remain unclear. METHODS: Human monocytes were isolated from the peripheral blood of healthy volunteers. The expression of growth factors and their receptors in fibrocytes was analyzed using ELISA and Western blotting. The effects of nintedanib on the ability of fibrocytes to stimulate lung fibroblasts were examined in terms of their proliferation. The direct effects of nintedanib on the differentiation and migration of fibrocytes were also assessed. We investigated whether nintedanib affected the accumulation of fibrocytes in mouse lungs treated with bleomycin. RESULTS: Human fibrocytes produced PDGF, FGF2, and VEGF-A. Nintedanib and specific inhibitors for each growth factor receptor significantly inhibited the proliferation of lung fibroblasts stimulated by the supernatant of fibrocytes. Nintedanib inhibited the migration and differentiation of fibrocytes induced by growth factors in vitro. The number of fibrocytes in the bleomycin-induced lung fibrosis model was reduced by the administration of nintedanib, and this was associated with anti-fibrotic effects. CONCLUSIONS: These results support the role of fibrocytes as producers of and responders to growth factors, and suggest that the anti-fibrotic effects of nintedanib are at least partly mediated by suppression of fibrocyte function.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/metabolism , Indoles/therapeutic use , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
8.
Nat Commun ; 6: 8792, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26635184

ABSTRACT

Bevacizumab exerts anti-angiogenic effects in cancer patients by inhibiting vascular endothelial growth factor (VEGF). However, its use is still limited due to the development of resistance to the treatment. Such resistance can be regulated by various factors, although the underlying mechanisms remain incompletely understood. Here we show that bone marrow-derived fibrocyte-like cells, defined as alpha-1 type I collagen-positive and CXCR4-positive cells, contribute to the acquired resistance to bevacizumab. In mouse models of malignant pleural mesothelioma and lung cancer, fibrocyte-like cells mediate the resistance to bevacizumab as the main producer of fibroblast growth factor 2. In clinical specimens of lung cancer, the number of fibrocyte-like cells is significantly increased in bevacizumab-treated tumours, and correlates with the number of treatment cycles, as well as CD31-positive vessels. Our results identify fibrocyte-like cells as a promising cell biomarker and a potential therapeutic target to overcome resistance to anti-VEGF therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Bevacizumab/administration & dosage , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Lung Neoplasms/drug therapy , Animals , Cell Line, Tumor , Fibroblast Growth Factor 2/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/antagonists & inhibitors
9.
PLoS One ; 10(3): e0121615, 2015.
Article in English | MEDLINE | ID: mdl-25785861

ABSTRACT

Plasminogen activator inhibitor (PAI)-1 is the principal inhibitor of plasminogen activators, and is responsible for the degradation of fibrin and extracellular matrix. IMD-4690 is a newly synthesized inhibitor for PAI-1, whereas the effect on allergic airway inflammation and remodeling is still unclear. We examined the in vivo effects by using a chronic allergen exposure model of bronchial asthma in mice. The model was generated by an immune challenge for 8 weeks with house dust mite antigen, Dermatophagoides pteronyssinus (Dp). IMD-4690 was intraperitoneally administered during the challenge. Lung histopathology, hyperresponsiveness and the concentrations of mediators in lung homogenates were analyzed. The amount of active PAI-1 in the lungs was increased in mice treated with Dp. Administration with IMD-4690 reduced an active/total PAI-1 ratio. IMD-4690 also reduced the number of bronchial eosinophils in accordance with the decreased expressions of Th2 cytokines in the lung homogenates. Airway remodeling was inhibited by reducing subepithelial collagen deposition, smooth muscle hypertrophy, and angiogenesis. The effects of IMD-4690 were partly mediated by the regulation of TGF-ß, HGF and matrix metalloproteinase. These results suggest that PAI-1 plays crucial roles in airway inflammation and remodeling, and IMD-4690, a specific PAI-1 inhibitor, may have therapeutic potential for patients with refractory asthma due to airway remodeling.


Subject(s)
Acetates/pharmacology , Airway Remodeling/drug effects , Asthma/pathology , Asthma/physiopathology , Biphenyl Compounds/pharmacology , Bronchi/drug effects , Neovascularization, Pathologic/drug therapy , Plasminogen Activator Inhibitor 1/metabolism , Acetates/therapeutic use , Animals , Antigens, Dermatophagoides/adverse effects , Asthma/drug therapy , Asthma/immunology , Biphenyl Compounds/therapeutic use , Bronchi/blood supply , Bronchi/immunology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Chronic Disease , Cytokines/metabolism , Dermatophagoides pteronyssinus/immunology , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/immunology , Female , Immunoglobulin E/blood , Mice , Tissue Plasminogen Activator/metabolism
10.
Respir Res ; 15: 143, 2014 Nov 29.
Article in English | MEDLINE | ID: mdl-25472740

ABSTRACT

BACKGROUND: Surfactant protein D (SP-D) can regulate both innate and adaptive immunity. Recently, SP-D has been shown to contribute to the pathogenesis of airway allergic inflammation and bleomycin-induced pulmonary fibrosis. However, in allergic airways disease, the role of SP-D in airway remodeling remains unknown. The objective of this study was to determine the contribution of functional SP-D in regulating sub-epithelial fibrosis in a mouse chronic house dust mite model of allergic airways disease. METHODS: C57BL/6 wild-type (WT) and SP-D-/- mice (C57BL/6 background) were chronically challenged with house dust mite antigen (Dermatophagoides pteronyssinus, Dp). Studies with SP-D rescue and neutralization of TGF-ß were conducted. Lung histopathology and the concentrations of collagen, growth factors, and cytokines present in the airspace and lung tissue were determined. Cultured eosinophils were stimulated by Dp in presence or absence of SP-D. RESULTS: Dp-challenged SP-D-/- mice demonstrate increased sub-epithelial fibrosis, collagen production, eosinophil infiltration, TGF-ß1, and IL-13 production, when compared to Dp-challenged WT mice. By immunohistology, we detected an increase in TGF-ß1 and IL-13 positive eosinophils in SP-D-/- mice. Purified eosinophils stimulated with Dp produced TGF-ß1 and IL-13, which was prevented by co-incubation with SP-D. Additionally, treatment of Dp challenged SP-D-/- mice with exogenous SP-D was able to rescue the phenotypes observed in SP-D-/- mice and neutralization of TGF-ß1 reduced sub-epithelial fibrosis in Dp-challenged SP-D-/- mice. CONCLUSION: These data support a protective role for SP-D in the pathogenesis of sub-epithelial fibrosis in a mouse model of allergic inflammation through regulation of eosinophil-derived TGF-ß.


Subject(s)
Airway Remodeling , Asthma/metabolism , Epithelial Cells/metabolism , Lung/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/prevention & control , Pulmonary Surfactant-Associated Protein D/metabolism , Transforming Growth Factor beta1/metabolism , Airway Remodeling/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Antigens, Dermatophagoides , Arthropod Proteins , Asthma/immunology , Asthma/pathology , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Genotype , Interleukin-13/immunology , Interleukin-13/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Protein D/deficiency , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/pharmacology , Signal Transduction , Transforming Growth Factor beta1/antagonists & inhibitors
11.
Am J Respir Cell Mol Biol ; 51(6): 793-801, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24885373

ABSTRACT

Circulating fibrocytes have been reported to migrate into the injured lungs, and contribute to fibrogenesis via CXCL12-CXCR4 axis. In contrast, we report that imatinib mesylate prevented bleomycin (BLM)-induced pulmonary fibrosis in mice by inhibiting platelet-derived growth factor receptor (PDGFR), even when it was administered only in the early phase. The goal of this study was to test the hypothesis that platelet-derived growth factor (PDGF) might directly contribute to the migration of fibrocytes to the injured lungs. PDGFR expression in fibrocytes was examined by flow cytometry and RT-PCR. The migration of fibrocytes was evaluated by using a chemotaxis assay for human fibrocytes isolated from peripheral blood. The numbers of fibrocytes triple-stained for CD45, collagen-1, and CXCR4 were also examined in lung digests of BLM-treated mice. PDGFR mRNA levels in fibrocytes isolated from patients with idiopathic pulmonary fibrosis were investigated by real-time PCR. Fibrocytes expressed both PDGFR-α and -ß, and migrated in response to PDGFs. PDGFR inhibitors (imatinib, PDGFR-blocking antibodies) suppressed fibrocyte migration in vitro, and reduced the number of fibrocytes in the lungs of BLM-treated mice. PDGF-BB was a stronger chemoattractant than the other PDGFs in vitro, and anti-PDGFR-ß-blocking antibody decreased the numbers of fibrocytes in the lungs compared with anti-PDGFR-α antibody in vivo. Marked expression of PDGFR-ß was observed in fibrocytes from patients with idiopathic pulmonary fibrosis compared with healthy subjects. These results suggest that PDGF directly functions as a strong chemoattractant for fibrocytes. In particular, the PDGF-BB-PDGFR-ß biological axis might play a critical role in fibrocyte migration into the fibrotic lungs.


Subject(s)
Platelet-Derived Growth Factor/physiology , Pulmonary Fibrosis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Benzamides/administration & dosage , Case-Control Studies , Chemotaxis , Drug Evaluation, Preclinical , Female , Humans , Imatinib Mesylate , Injections, Intraperitoneal , Mice, Inbred C57BL , Piperazines/administration & dosage , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Pyrimidines/administration & dosage , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptors, CXCR4/metabolism , Signal Transduction
12.
Arthritis Rheumatol ; 66(3): 560-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24574215

ABSTRACT

OBJECTIVE: Thymidine phosphorylase (TP) in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) is induced by tumor necrosis factor α (TNFα) and other cytokines that have been reported to be major inflammation mediators in RA. We previously demonstrated that TP plays an important role in angiogenesis and tumor growth, invasion, and metastasis. The aim of this study was to investigate whether the role of TP in the pathogenesis of RA is similar to its role in tumors. METHODS: In FLS obtained from 2 patients with RA, the expression of TP, interferon-γ (IFNγ)-inducible protein 10 (CXCL10), and other cytokines was measured by quantitative real-time polymerase chain reaction, immunoblotting, and enzyme-linked immunosorbent assays. Microarray analysis was performed using FLS transfected with TYMP complementary DNA and treated with a TP inhibitor. RESULTS: The expression of TP in FLS was up-regulated by TNFα, interleukin-1ß (IL-1ß), IL-17, IFNγ, and lipopolysaccharide. Microarray analysis of FLS overexpressing TP identified CXCL10 as a thymidine phosphorylase-related gene. The expression of CXCL10 was induced by TNFα, and this induction was suppressed by TYMP small interfering RNA and TP inhibitor. Furthermore, the combination of TNFα and IFNγ synergistically augmented the expression of TP and CXCL10. TP-induced CXCL10 expression was suppressed by the antioxidant EUK-8. In the synovial tissue of patients with RA, TP levels were significantly correlated with CXCL10 expression. CONCLUSION: The combination of TNFα and IFNγ strongly induced the expression of thymidine phosphorylase in RA FLS. The induction of thymidine phosphorylase enhanced the expression of CXCL10, which may contribute to the Th1 phenotype and bone destruction observed in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokine CXCL10/metabolism , Interferon-gamma/metabolism , Synovial Membrane/metabolism , Thymidine Phosphorylase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokine CXCL10/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Interferon-gamma/genetics , Synovial Membrane/pathology , Thymidine Phosphorylase/genetics , Tumor Necrosis Factor-alpha/genetics
13.
J Med Invest ; 60(3-4): 175-83, 2013.
Article in English | MEDLINE | ID: mdl-24190033

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease characterized by the proliferation of fibroblasts and deposition of extracellular matrix. Since the prognosis of IPF is still poor, novel therapeutic modalities are strongly required. For this reason, to find molecular target for therapy of IPF is of much importance. The recent understanding of pathogenesis in IPF indicates the critical role of alveolar epithelial type II cells (AECII) and fibroblasts. Although the detailed mechanisms involved in IPF is still unclear, various profibrotic mediators which are produced by the injured AECII are thought to play a role in the progression of pulmonary fibrosis via stimulating fibroblasts. Among them, platelet-derived growth factor (PDGF) is one of critical growth factors by stimulating the proliferation and migration of fibroblasts. In this review, we discuss the role of PDGF in pulmonary fibrosis and the possibility as a therapeutic target for IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Platelet-Derived Growth Factor/antagonists & inhibitors , Alveolar Epithelial Cells/pathology , Animals , Benzamides/therapeutic use , Disease Models, Animal , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/physiopathology , Imatinib Mesylate , Piperazines/therapeutic use , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/physiology , Pyrimidines/therapeutic use , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/physiology , Signal Transduction
14.
Am J Respir Cell Mol Biol ; 49(4): 536-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23642017

ABSTRACT

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in various biological functions, including cell survival, proliferation, migration, and adhesion. FAK is an essential factor for transforming growth factor ß to induce myofibroblast differentiation. In the present study, we investigated whether the targeted inhibition of FAK by using a specific inhibitor, TAE226, has the potential to regulate pulmonary fibrosis. TAE226 showed inhibitory activity of autophosphorylation of FAK at tyrosine 397 in lung fibroblasts. The addition of TAE226 inhibited the proliferation of lung fibroblasts in response to various growth factors, including platelet-derived growth factor and insulin-like growth factor I, in vitro. TAE226 strongly suppressed the production of type I collagen by lung fibroblasts. Furthermore, treatment of fibroblasts with TAE226 reduced the expression of α-smooth muscle actin induced by transforming growth factor ß, indicating the inhibition of differentiation of fibroblasts to myofibroblasts. Administration of TAE226 ameliorated the pulmonary fibrosis induced by bleomycin in mice even when used late in the treatment. The number of proliferating mesenchymal cells was reduced in the lungs of TAE226-treated mice. These data suggest that FAK signal plays a significant role in the progression of pulmonary fibrosis and that it can become a promising target for therapeutic approaches to pulmonary fibrosis.


Subject(s)
Focal Adhesion Kinase 1/antagonists & inhibitors , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Actins/genetics , Actins/metabolism , Animals , Bleomycin , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , DNA/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tyrosine/genetics , Tyrosine/metabolism
15.
J Immunol ; 190(12): 6239-49, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23690472

ABSTRACT

Podoplanin (Aggrus), which is a type I transmembrane sialomucin-like glycoprotein, is highly expressed in malignant pleural mesothelioma (MPM). We previously reported the generation of a rat anti-human podoplanin Ab, NZ-1, which inhibited podoplanin-induced platelet aggregation and hematogenous metastasis. In this study, we examined the antitumor effector functions of NZ-1 and NZ-8, a novel rat-human chimeric Ab generated from NZ-1 including Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity against MPM in vitro and in vivo. Immunostaining with NZ-1 showed the expression of podoplanin in 73% (11 out of 15) of MPM cell lines and 92% (33 out of 36) of malignant mesothelioma tissues. NZ-1 could induce potent ADCC against podoplanin-positive MPM cells mediated by rat NK (CD161a(+)) cells, but not murine splenocytes or human mononuclear cells. Treatment with NZ-1 significantly reduced the growth of s.c. established tumors of MPM cells (ACC-MESO-4 or podoplanin-transfected MSTO-211H) in SCID mice, only when NZ-1 was administered with rat NK cells. In in vivo imaging, NZ-1 efficiently accumulated to xenograft of MPM, and its accumulation continued for 3 wk after systemic administration. Furthermore, NZ-8 preferentially recognized podoplanin expressing in MPM, but not in normal tissues. NZ-8 could induce higher ADCC mediated by human NK cells and complement-dependent cytotoxicity as compared with NZ-1. Treatment with NZ-8 and human NK cells significantly inhibited the growth of MPM cells in vivo. These results strongly suggest that targeting therapy to podoplanin with therapeutic Abs (i.e., NZ-8) derived from NZ-1 might be useful as a novel immunotherapy against MPM.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotherapy/methods , Membrane Glycoproteins/immunology , Mesothelioma/immunology , Pleural Neoplasms/immunology , Animals , Antibodies, Monoclonal/pharmacology , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Rats , Rats, Wistar , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Xenograft Model Antitumor Assays
16.
J Med Invest ; 60(1-2): 127-37, 2013.
Article in English | MEDLINE | ID: mdl-23614921

ABSTRACT

Circulating fibrocytes had been reported to migrate into the injured lungs, and contribute to fibrogenesis via chemokine-chemokine receptor systems including CXCL12-CXCR4 axis. Here we hypothesized that blockade of CXCR4 might inhibit the migration of fibrocytes to the injured lungs and the subsequent pulmonary fibrosis. To explore the antifibrotic effects of blockade of CXCR4, we used a specific antagonist for CXCR4, AMD3100, in bleomycin-induced pulmonary fibrosis model in mice. Administration of AMD3100 significantly improved the loss of body weight of mice treated with bleomycin, and inhibited the fibrotic lesion in subpleural areas of the lungs. The quantitative analysis demonstrated that treatment with AMD3100 reduced the collagen content and fibrotic score (Aschcroft score) in the lungs. Although AMD3100 did not affect cell classification in bronchoalveolar lavage fluid on day 7, the percentage of lymphocytes was reduced by AMD3100 on day 14. AMD3100 directly inhibited the migration of human fibrocytes in response to CXCL12 in vitro, and reduced the trafficking of fibrocytes into the lungs treated with bleocmycin in vivo. These results suggest that the blockade of CXCR4 might be useful strategy for therapy of patients with pulmonary fibrosis via inhibiting the migration of circulating fibrocytes.


Subject(s)
Bleomycin/toxicity , Heterocyclic Compounds/therapeutic use , Pulmonary Fibrosis/prevention & control , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Cell Movement/drug effects , Chemokine CXCL12/analysis , Chemotaxis/drug effects , Cyclams , Female , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced
17.
Am J Pathol ; 182(5): 1843-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23499372

ABSTRACT

Surfactant protein A (SP-A) is a large multimeric protein found in the lungs. In addition to its immunoregulatory function in infectious respiratory diseases, SP-A is also used as a marker of lung adenocarcinoma. Despite the finding that SP-A expression levels in cancer cells has a relationship with patient prognosis, the function of SP-A in lung cancer progression is unknown. We investigated the role of SP-A in lung cancer progression by introducing the SP-A gene into human lung adenocarcinoma cell lines. SP-A gene transduction suppressed the progression of tumor in subcutaneous xenograft or lung metastasis mouse models. Immunohistochemical analysis showed that the number of M1 antitumor tumor-associated macrophages (TAMs) was increased and the number of M2 tumor-promoting TAMs was not changed in the tumor tissue produced by SP-A-expressing cells. In addition, natural killer (NK) cells were also increased and activated in the SP-A-expressing tumor. Moreover, SP-A did not inhibit tumor progression in mice depleted of NK cells. Taking into account that SP-A did not directly activate NK cells, these results suggest that SP-A inhibited lung cancer progression by recruiting and activating NK cells via controlling the polarization of TAMs.


Subject(s)
Cell Polarity , Disease Progression , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophages/pathology , Pulmonary Surfactant-Associated Protein A/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Killer Cells, Natural/pathology , Macrophages/metabolism , Male , Mice , Mice, Nude , Neoplasm Metastasis , Subcutaneous Tissue/pathology , Xenograft Model Antitumor Assays
18.
J Med Invest ; 59(1-2): 174-85, 2012.
Article in English | MEDLINE | ID: mdl-22450006

ABSTRACT

BACKGROUND: Gefitinib, an inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase, has been reported to be associated with interstitial lung disorders, and their high incidence and mortality have become a matter of great concern, especially in Japan. In this study, we investigated the effect of gefitinib on different phases of radiation-induced lung disorders in an experimental model. METHODS: The thoraxes of Wistar rats were irradiated on day 1 with a single X-ray dose of 20 Gy, and gefitinib (50 mg/kg/day) was orally administered from day 1 to 14. The rat lungs were harvested on days 15 and 57 and the bronchoalveolar lavage (BAL) was performed. RESULTS: Gefitinib treatment increased the infiltration of inflammatory cells, which produced more pro-inflammatory cytokines (IL-6, IL-1ß), in the lungs of the irradiated rats on days 15 and 57, while gefitinib treatment reduced collagen content of the lungs in irradiated rats and decreased proliferation and EGFR expression in the lung fibroblasts from irradiated rats on day 57. CONCLUSIONS: In irradiated rats, gefitinib treatment augmented lung inflammation, including inflammatory cell infiltration and pro-inflammatory cytokine expression, while gefitinib treatment attenuated fibrotic lung remodeling due to the inhibition of lung fibroblast proliferation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Quinazolines/pharmacology , Radiation Pneumonitis/drug therapy , Animals , Disease Models, Animal , Gefitinib , Male , Protein Kinase Inhibitors/pharmacology , Radiation Pneumonitis/pathology , Rats , Rats, Wistar
19.
Am J Respir Crit Care Med ; 185(5): 525-36, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22198976

ABSTRACT

RATIONALE: Surfactant protein (SP)-D and SP-A have been implicated in immunomodulation in the lung. It has been reported that patients with idiopathic pulmonary fibrosis (IPF) often have elevated serum levels of SP-A and SP-D, although their role in the disease is not known. OBJECTIVES: The goal of this study was to test the hypothesis that SP-D plays an important role in lung fibrosis using a mouse model of fibrosis induced by bleomycin (BLM). METHODS: Triple transgenic inducible SP-D mice (iSP-D mice), in which rat SP-D is expressed in response to doxycycline (Dox) treatment, were administered BLM (100 U/kg) or saline subcutaneously using miniosmotic pumps. MEASUREMENTS AND MAIN RESULTS: BLM-treated iSP-D mice off Dox (SP-D off) had increased lung fibrosis compared with mice on Dox (SP-D on). SP-D deficiency also increased macrophage-dominant cell infiltration and the expression of profibrotic cytokines (transforming growth factor [TGF]-ß1, platelet-derived growth factor-AA). Alveolar macrophages isolated from BLM-treated iSP-D mice off Dox (SP-D off) secreted more TGF-ß1. Fibrocytes, which are bone marrow-derived mesenchymal progenitor cells, were increased to a greater extent in the lungs of the BLM-treated iSP-D mice off Dox (SP-D off). Fibrocytes isolated from BLM-treated iSP-D mice off Dox (SP-D off) expressed more of the profibrotic cytokine TGF-ß1 and more CXCR4, a chemokine receptor that is important in fibrocyte migration into the lungs. Exogenous SP-D administered intratracheally attenuated BLM-induced lung fibrosis in SP-D(-/-) mice. CONCLUSIONS: These data suggest that alveolar SP-D regulates numbers of macrophages and fibrocytes in the lungs, profibrotic cytokine expression, and fibrotic lung remodeling in response to BLM injury.


Subject(s)
Bleomycin/toxicity , Idiopathic Pulmonary Fibrosis/physiopathology , Pulmonary Surfactant-Associated Protein D/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/physiopathology , Airway Remodeling/physiology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/physiology , Disease Models, Animal , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pulmonary Surfactant-Associated Protein A/analysis , Pulmonary Surfactant-Associated Protein A/physiology , Pulmonary Surfactant-Associated Protein D/analysis , Rats
20.
J Med Invest ; 58(3-4): 188-96, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21921419

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive and lethal disease of the lung that is characterized by the proliferation of fibroblasts and increased deposition of the extracellular matrix. The CCN6/WISP-3 is a member of the CCN family of matricellular proteins, which consists of six members that are involved in many vital biological functions. However, the regulation of lung fibroblasts mediated by CCN6 protein has not been fully elucidated. Here, we demonstrated that CCN6 induced the proliferation of lung fibroblasts by binding to integrin ß1, leading to the phosphorylation of FAK(Y397). Furthermore, CCN6 showed a weak, but significant, ability to stimulate the expression of fibronectin. CCN6 was highly expressed in the lung tissues of mice treated with bleomycin. Our results suggest that CCN6 plays a role in the fibrogenesis of the lungs mainly by stimulating the growth of lung fibroblasts and is a potential target for the treatment of pulmonary fibrosis.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Integrin beta1/physiology , Pulmonary Fibrosis/etiology , Signal Transduction/physiology , Animals , Cell Proliferation , Extracellular Matrix Proteins/physiology , Female , Fibroblasts/physiology , Humans , Lung/cytology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...