Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Exp Cell Res ; 398(2): 112416, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33307020

ABSTRACT

Idiopathic pulmonary fibrosis (IPF), a progressive disorder of unknown etiology, is characterized by pathological lung fibroblast activation and proliferation resulting in abnormal deposition of extracellular matrix proteins within the lung parenchyma. The pathophysiological roles of exosomal microRNAs in pulmonary fibrosis remain unclear; therefore, we aimed to identify and characterize fibrosis-responsive exosomal microRNAs. We used microRNA array analysis and profiled the expression of exosome-derived miRNA in sera of C57BL/6 mice exhibiting bleomycin-induced pulmonary fibrosis. The effect of microRNAs potentially involved in fibrosis was then evaluated in vivo and in vitro. The expression of exosomal microRNA-16 was increased by up to 8.0-fold on day 14 in bleomycin-treated mice, compared to vehicle-treated mice. MicroRNA-16 mimic administration on day 14 after bleomycin challenge ameliorated pulmonary fibrosis and suppressed lung and serum expression of secreted protein acidic and rich in cysteine (SPARC). Pretreatment of human lung fibroblasts with the microRNA-16 mimic decreased the expression of rapamycin-insensitive companion of mTOR (Rictor) and TGF-ß1-induced expression of SPARC. This is the first study reporting the anti-fibrotic properties of microRNA-16 and demonstrating that these effects occur via the mTORC2 pathway. These findings support that microRNA-16 may be a promising therapeutic target for IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , MicroRNAs/metabolism , Osteonectin/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Animals , Exosomes/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL
2.
Thorac Cancer ; 11(7): 1876-1884, 2020 07.
Article in English | MEDLINE | ID: mdl-32421226

ABSTRACT

BACKGROUND: Pleurodesis is the standard of care for non-small cell lung cancer (NSCLC) patients with symptomatic malignant pleural effusion (MPE). However, there is no standard management for MPE uncontrolled by pleurodesis. Most patients with unsuccessful MPE control are unable to receive effective chemotherapy. Vascular endothelial growth factor (VEGF) plays an important role in the pathogenesis of MPE. This multicenter, phase II study investigated the effects of bevacizumab plus chemotherapy in nonsquamous NSCLC patients with unsuccessful management of MPE. METHODS: Nonsquamous NSCLC patients with MPE following unsuccessful tube drainage or pleurodesis received bevacizumab (15 mg/kg) plus chemotherapy every three weeks. The primary endpoint was pleural effusion control rate (PECR), defined as the percentage of patients without reaccumulation of MPE at eight weeks. Secondary endpoints included pleural progression-free survival (PPFS), safety, and quality of life (QoL). RESULTS: A total of 20 patients (median age: 69 years; 14 males; 20 adenocarcinomas; six epidermal growth factor receptor mutations) were enrolled in nine centers. The PECR was 80% and the primary end point was met. The PPFS and the overall survival (OS) were 16.6 months and 19.6 months, respectively. Patients with high levels of VEGF in the MPE had shorter PPFS (P = 0.010) and OS (P = 0.002). Toxicities of grade ≥ 3 included neutropenia (50%), thrombocytopenia (10%), proteinuria (10%), and hypertension (2%). The cognitive QoL score improved after treatment. CONCLUSIONS: Bevacizumab plus chemotherapy is highly effective with acceptable toxicities in nonsquamous NSCLC patients with uncontrolled MPE, and should be considered as a standard therapy in this setting. KEY POINTS: SIGNIFICANT FINDINGS OF THE STUDY: Bevacizumab plus chemotherapy is highly effective with acceptable toxicities in nonsquamous NSCLC patients with uncontrolled MPE. WHAT THIS STUDY ADDS: Bevacizumab plus chemotherapy should be considered as a standard treatment option for patients with uncontrolled MPE. CLINICAL TRIAL REGISTRATION: UMIN000006868 was a phase II study of efficacy of bevacizumab plus chemotherapy for the management of malignant pleural effusion (MPE) in nonsquamous non-small cell lung cancer patients with MPE unsuccessfully controlled by tube drainage or pleurodesis (North East Japan Study Group Trial NEJ-013B) (http://umin.sc.jp/ctr/).


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pleural Effusion, Malignant/drug therapy , Pleurodesis/adverse effects , Adenocarcinoma of Lung/etiology , Adenocarcinoma of Lung/pathology , Aged , Bevacizumab/administration & dosage , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/pathology , Docetaxel/administration & dosage , Erlotinib Hydrochloride/administration & dosage , Female , Follow-Up Studies , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/pathology , Pemetrexed/administration & dosage , Pleural Effusion, Malignant/complications , Pleural Effusion, Malignant/pathology , Prognosis , Survival Rate
3.
J Nippon Med Sch ; 87(3): 118-128, 2020 Jul 13.
Article in English | MEDLINE | ID: mdl-31776321

ABSTRACT

BACKGROUND: Although aberrant proliferation and activation of lung fibroblasts are implicated in the initiation and progression of idiopathic pulmonary fibrosis (IPF), the underlying mechanisms are not well characterized. Numerous microRNAs (miRNAs) have been implicated in this process; however, miRNAs derived from exosomes and the relevance of such miRNAs to fibroblast-to-myofibroblast differentiation are not well understood. In this study, we attempted to identify exosome-derived miRNAs relevant to fibrosis development. METHODS: Using miRNA array analysis, we profiled exosome-derived miRNA expression in sera of C57BL/6 mice exhibiting bleomycin-induced pulmonary fibrosis. After validating a selected miRNA by quantitative reverse-transcription polymerase chain reaction, its effect on fibroblast-to-myofibroblast differentiation was investigated in human lung fibroblasts. Furthermore, we determined the role of the selected miRNA in an in vivo model of pulmonary fibrosis. RESULTS: MiRNA array analysis revealed that miR-22 expression was increased by up to 2 fold on day 7 after bleomycin treatment compared with that in vehicle-treated mice. In vitro, miR-22 transfection suppressed TGF-ß1-induced α-SMA expression. This was mediated via inhibition of the ERK1/2 pathway. Baseline α-SMA expression was increased upon miR-22 inhibitor transfection. Furthermore, miR-22 negatively regulated connective tissue growth factor expression in the presence of TGF-ß1. In vivo, administration of a miR-22 mimic on day 10 after bleomycin challenge ameliorated pulmonary fibrosis lesions accompanied by decreased α-SMA expression in the model mice. CONCLUSIONS: Exosomal miR-22 modulates fibroblast-to-myofibroblast differentiation. The present findings warrant further study, which could shed light on miR-22 as a novel therapeutic target in IPF.


Subject(s)
Cell Differentiation/genetics , Exosomes/genetics , Exosomes/physiology , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , MicroRNAs/physiology , Myofibroblasts/pathology , Actins/genetics , Actins/metabolism , Animals , Disease Models, Animal , Gene Expression/genetics , In Vitro Techniques , MAP Kinase Signaling System/genetics , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism
4.
Respir Res ; 19(1): 71, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29690905

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive disease with high mortality, and the pathogenesis of the disease is still incompletely understood. Although lymphocytes, especially CD4+CD25+FoxP3+ regulatory T cells (Tregs), have been implicated in the development of IPF, contradictory results have been reported regarding the contribution of Tregs to fibrosis both in animals and humans. The aim of this study was to investigate whether a specific T cell subset has therapeutic potential in inhibiting bleomycin (BLM)-induced murine pulmonary fibrosis. METHODS: C57BL/6 mice received BLM (100 mg/kg body weight) with osmotic pumps (day 0), and pulmonary fibrosis was induced. Then, splenocytes or Tregs were adoptively transferred via the tail vein. The lungs were removed and subjected to histological and biochemical examinations to study the effects of these cells on pulmonary fibrosis, and blood samples were collected by cardiac punctures to measure relevant cytokines by enzyme-linked immunosorbent assay. Tregs isolated from an interleukin (IL)-10 knock-out mice were used to assess the effect of this mediator. To determine the roles of the spleen in this model, spleen vessels were carefully cauterized and the spleen was removed either on day 0 or 14 after BLM challenge. RESULTS: Splenocytes significantly ameliorated BLM-induced pulmonary fibrosis when they were administered on day 14. This effect was abrogated by depleting Tregs with an anti-CD25 monoclonal antibody. Adoptive transfer of Tregs on day 14 after a BLM challenge significantly attenuated pulmonary fibrosis, and this was accompanied by decreased production of fibroblast growth factor (FGF) 9-positive cells bearing the morphology of alveolar epithelial cells. In addition, BLM-induced plasma IL-10 expression reverted to basal levels after adoptive transfer of Tregs. Moreover, BLM-induced fibrocyte chemoattractant chemokine (CC motif) ligand-2 production was significantly ameliorated by Treg adoptive transfer in lung homogenates, accompanied by reduced accumulation of bone-marrow derived fibrocytes. Genetic ablation of IL-10 abrogated the ameliorating effect of Tregs on pulmonary fibrosis. Finally, splenectomy on day 0 after a BLM challenge significantly ameliorated lung fibrosis, whereas splenectomy on day 14 had no effect. CONCLUSIONS: These findings warrant further investigations to develop a cell-based therapy using Tregs for treating IPF.


Subject(s)
Bleomycin/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/therapy , Spleen/transplantation , T-Lymphocytes, Regulatory/transplantation , Animals , Bleomycin/administration & dosage , Infusion Pumps, Implantable , Lymphocyte Transfusion/methods , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/metabolism , Spleen/cytology , T-Lymphocytes, Regulatory/metabolism
5.
Pulm Pharmacol Ther ; 44: 61-69, 2017 06.
Article in English | MEDLINE | ID: mdl-28315487

ABSTRACT

Pathogenesis of idiopathic pulmonary fibrosis (IPF) remains unclear. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that participates in the assembly and turnover of the extracellular matrix, whose expression is regulated by transforming growth factor (TGF)-ß1 through activation of mammalian target of rapamycin complex 2 (mTORC2). Exchange factor found in platelets, leukemic, and neuronal tissues (XPLN) is an endogenous inhibitor of mTORC2. However, whether XPLN modulates SPARC expression remains unknown. Herein, we investigated the regulatory mechanisms of XPLN in human lung fibroblasts. Effect of XPLN on mTORC2 activity was evaluated by silencing XPLN in human foetal lung fibroblasts (HFL-1 cells), using small interfering RNA. SPARC expression was quantified by quantitative real-time RT-PCR and western blotting. Fibroblasts were treated with TGF-ß1, histone deacetylase (HDAC) inhibitors, entinostat, or vorinostat, to assess their effects on XPLN expression. Moreover, the effect of mTORC1 inhibition on SPARC and XPLN was examined. XPLN depletion stimulated SPARC expression and Akt phosphorylation on Ser473. TGF-ß1 treatment down-regulated XPLN via Smad 2/3. XPLN mRNA expression was up-regulated upon treatment with HDAC inhibitors in a concentration-dependent manner, and TGF-ß1-induced SPARC expression was reversed by entinostat treatment. mTORC1 inhibition by rapamycin and Raptor depletion stimulated SPARC expression. In conclusion, this is the first study describing the involvement of XPLN in the regulation of SPARC. These findings may help uncover the regulatory mechanisms of the mTORC2-SPARC axis. The up-regulation of XPLN by HDAC inhibitors may be a novel therapeutic approach in patients with IPF.


Subject(s)
Fibroblasts/metabolism , Histone Deacetylase Inhibitors/pharmacology , Idiopathic Pulmonary Fibrosis/physiopathology , Rho Guanine Nucleotide Exchange Factors/drug effects , Benzamides/pharmacology , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Gene Silencing , Humans , Hydroxamic Acids/pharmacology , Lung/cytology , Lung/drug effects , Lung/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Osteonectin/genetics , Pyridines/pharmacology , RNA, Small Interfering/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/metabolism , Up-Regulation , Vorinostat
6.
Sarcoidosis Vasc Diffuse Lung Dis ; 33(3): 242-246, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27758989

ABSTRACT

BACKGROUND AND OBJECTIVE: There is growing evidence for anti-inflammatory activities of macrolides in chronic respiratory diseases, such as diffuse panbronchiolitis, cystic fibrosis, or chronic bronchitis. The long-term effect of macrolides in idiopathic pulmonary fibrosis (IPF) is unknown. This study was aimed to investigate the effect of macrolide therapy on the frequency of acute exacerbation (AE) and the mortality in IPF. METHODS: A total 52 IPF patients who were treated by combination of conventional agents with or without macrolides were retrospectively reviewed. The primary endpoint was the incidence of AE in IPF patients. We also observed survival rate after the treatment with or without macrolides. RESULTS: AE was observed in 4 of 29 cases (13.8%) treated with macrolides and 8 of 23 cases (34.8%) treated without macrolides, respectively during 36 months. AE free survival rate of macrolide group was significantly better than that of non-macrolide group (logrank p=0.027). Survival rate of IPF patients with macrolide therapy was significantly better than that of patients without macrolide therapy (p=0.047). CONCLUSION: Our results indicate the potential beneficial efficacy of macrolide therapy combined with oral corticosteroids, immunosuppressive or anti-fibrotic agents in IPF.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Macrolides/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Aged , Disease Progression , Drug Therapy, Combination , Female , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/mortality , Idiopathic Pulmonary Fibrosis/physiopathology , Immunosuppressive Agents/therapeutic use , Kaplan-Meier Estimate , Lung/physiopathology , Male , Middle Aged , Retrospective Studies , Time Factors , Treatment Outcome
7.
Pulm Pharmacol Ther ; 32: 29-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25843005

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with a high mortality rate. Signalling pathways activated by several tyrosine kinase receptors are known to be involved in lung fibrosis, and this knowledge has led to the development of the triple tyrosine kinase inhibitor nintedanib, an inhibitor of vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and fibroblast growth factor receptor (FGFR), for the treatment of IPF. Pulmonary surfactant protein D (SP-D), an important biomarker of IPF, reportedly attenuates bleomycin-induced pulmonary fibrosis in mice. In this study, we investigated whether nintedanib modulates SP-D expression in human lung epithelial (A549) cells using quantitative real-time reverse transcriptase polymerase chain reaction and western blotting. To investigate the mechanisms underlying the effects of nintedanib, we evaluated the phosphorylation of c-Jun N-terminal kinase (JNK) and its downstream target c-Jun. The effect of the JNK inhibitor SP600125 on c-Jun phosphorylation was also tested. Activation of activator protein-1 (AP-1) was examined using an enzyme-linked immunosorbent assay-based test, and cell proliferation assays were performed to estimate the effect of nintedanib on cell proliferation. Furthermore, we treated mice with nintedanib to examine its in vivo effect on SP-D levels in lungs. These experiments showed that nintedanib up-regulated SP-D messenger RNA expression in a dose-dependent manner at concentrations up to 5 µM, with significant SP-D induction observed at concentrations of 3 µM and 5 µM, in comparison with that observed in vehicle controls. Nintedanib stimulated a rapid increase in phosphorylated JNK in A549 cells within 30 min of treatment and stimulated c-Jun phosphorylation, which was inhibited by the JNK inhibitor SP600125. Additionally, nintedanib was found to activate AP-1. A549 cell proliferation was not affected by nintedanib at any of the tested concentrations. Moreover, blocking FGFR, PDGFR, and VEGFR function did not affect nintedanib-induced SP-D expression, suggesting that nintedanib mediates its effects through a mechanism that is distinct from its known role as a tyrosine kinase inhibitor. Nintedanib is also reported to inhibit Src kinase although pre-treatment of cells with a Src kinase inhibitor had no effect on nintedanib-induced SP-D expression. Increased expression of SFTPD mRNA and SP-D protein in the lungs of nintedanib-treated mice was also observed. In this work, we demonstrated that nintedanib up-regulated SP-D expression in A549 cells via the JNK-AP-1 pathway and did not affect cell proliferation. This is the first report describing SP-D induction by nintedanib.


Subject(s)
Epithelial Cells/drug effects , Indoles/pharmacology , Pulmonary Surfactant-Associated Protein D/genetics , Up-Regulation/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/physiopathology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/cytology , Lung/drug effects , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism
8.
Intern Med ; 51(21): 3085-8, 2012.
Article in English | MEDLINE | ID: mdl-23124156

ABSTRACT

We herein report a case of toxic shock syndrome (TSS) associated with the 2009 pandemic H1N1 (pH1N1) influenza virus and a community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) infection in a 16-year-old Vietnamese girl. Staphylococcal enterotoxin B (SEB) was detected in the patient's serum, and the level of anti-SEB antibodies was found to be elevated. A flow cytometric analysis showed evidence of activated SEB-reactive Vß3+ and Vß12+ T cells. These data suggest that the CA-MRSA-induced activation of SEB-reactive T cells may cause TSS in patients with pH1N1 virus infection. Moreover, this is the first report describing immunological confirmation of SEB contributing directly to TSS in a patient fulfilling the diagnostic criteria of TSS.


Subject(s)
Community-Acquired Infections/etiology , Enterotoxins/toxicity , Methicillin-Resistant Staphylococcus aureus , Shock, Septic/etiology , Staphylococcal Infections/etiology , Adolescent , Anti-Bacterial Agents/administration & dosage , Community-Acquired Infections/drug therapy , Community-Acquired Infections/microbiology , Female , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/complications , Pneumonia, Staphylococcal/drug therapy , Pneumonia, Staphylococcal/etiology , Pneumonia, Staphylococcal/microbiology , Shock, Septic/drug therapy , Shock, Septic/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
9.
J Nippon Med Sch ; 79(1): 46-59, 2012.
Article in English | MEDLINE | ID: mdl-22398790

ABSTRACT

Myofibroblasts play a critical role in tissue fibrosis. However, the intracellular signaling pathways in myofibroblast differentiation are poorly understood. Here, we studied the relationship between transforming growth factor-ß (TGF-ß)/Smad pathway activation and myofibroblast differentiation in both in vivo and in vitro experiments. In murine bleomycin-induced pulmonary fibrosis, nuclear localization of phosphorylated Smad2/3 (p-Smad2/3) was observed in pulmonary fibrotic lesions 7 days after bleomycin injection, whereas α-smooth muscle actin (ASMA)-positive myofibroblasts appeared in the lesions at 14 days, when the cytoplasmic localization of p-Smad2/3 was observed. We also compared the effects of TGF-ß1 on myofibroblast differentiation and on type I collagen expression in a murine lung fibroblast cell line (MLg2908). TGF-ß1 induced rapid expression of p-Smad2/3 in nuclei, after which ASMA organization in the cytoplasm of fibroblasts was observed. However, TGF-ß1 produced no effect on the quantity of ASMA, either in mRNA levels or protein levels, even after the phosphorylation of Smad2/3. In contrast, TGF-ß1 upregulated the expression of type I collagen mRNA. These findings suggest that in pulmonary fibrosis the molecular mechanism of myofibroblast differentiation is complex and that the difference between ASMA expression and type I collagen expression is mediated by the TGF-ß/Smad pathway.


Subject(s)
Cell Differentiation , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , 3T3 Cells , Actins/genetics , Actins/metabolism , Animals , Bleomycin , Cell Differentiation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Pulmonary Fibrosis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Smad Proteins/genetics , Time Factors , Transforming Growth Factor beta1/pharmacology
10.
Blood Purif ; 32(4): 310-6, 2011.
Article in English | MEDLINE | ID: mdl-21893977

ABSTRACT

BACKGROUND/AIM: Recent reports suggest that polymyxin B (PMX)-immobilized fiber may have beneficial effects in idiopathic pulmonary fibrosis (IPF) with acute exacerbation (AE). High mobility group box-1 (HMGB-1) is an important pro-inflammatory mediator that contributes to acute lung inflammation. This study was aimed to investigate whether PMX treatment affects serum HMGB-1 levels and oxygenation in IPF patients with AE. MATERIALS AND METHODS: Twenty IPF patients with AE were treated by PMX. PMX treatment was carried out once daily for 2 successive days. Serum HMGB-1 levels were measured before and after PMX treatment. We also monitored arterial oxygen tension (PaO(2))/inspiratory oxygen fraction (FiO(2)) (P/F) ratio. PMX fiber columns were analyzed to examine whether HMGB-1 was absorbed by PMX. RESULTS: PMX treatment significantly improved both the serum HMGB-1 level and P/F ratio. HMGB-1 was detected in washing medium from the PMX column. CONCLUSION: PMX treatment may reduce serum HMGB-1 and improve oxygenation in patients with IPF with AE.


Subject(s)
HMGB1 Protein/blood , Hemoperfusion , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/therapy , Polymyxin B/therapeutic use , Aged , Female , Hemoperfusion/methods , Humans , Male , Middle Aged , Oxygen/analysis , Polymyxin B/chemistry , Treatment Outcome
11.
Blood Purif ; 29(4): 321-6, 2010.
Article in English | MEDLINE | ID: mdl-20185904

ABSTRACT

BACKGROUND/AIMS: Polymyxin B-immobilized fiber (PMX) treatment has beneficial effects in patients with acute lung injury/acute respiratory distress syndrome or acute exacerbation of idiopathic pulmonary fibrosis. This study was aimed to clarify the mechanism of PMX treatment for acute exacerbation of interstitial pneumonia (IP). MATERIALS AND METHODS: Sixteen consecutive IP patients with acute exacerbation were included. The patients were treated with PMX once daily for 2 successive days at a flow rate of 80-100 ml/min for 6 h. Cells adsorbed by PMX were analyzed morphologically by electron microscopy. Surface markers of these cells were determined by flow cytometry. Serum matrix metalloproteinase (MMP)-9 was measured before and after PMX treatment. RESULTS: Cells adsorbed by PMX were neutrophils and highly expressed HLA-DR, CD14, CD62L and CD114. Serum MMP-9 levels were significantly decreased after PMX treatment. CONCLUSION: This pilot study demonstrated neutrophil adsorption by PMX and its possible clinical application for acute exacerbation of IP.


Subject(s)
Hemoperfusion/methods , Lung Diseases, Interstitial/therapy , Neutrophils/pathology , Polymyxin B/therapeutic use , Aged , Cell Separation , Cell Shape , Female , Flow Cytometry , Humans , Immunophenotyping , Lung Diseases, Interstitial/pathology , Male , Middle Aged , Observation/methods , Pilot Projects
12.
Respirology ; 15(1): 126-31, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19947989

ABSTRACT

BACKGROUND AND OBJECTIVE: The incidence of Pneumocystis jirovecii pneumonia (PCP) in patients with predisposing immunodeficiencies other than AIDS is growing. Knowing the different characteristics and outcomes of PCP according to HIV status would help physicians manage and treat patients with PCP. METHODS: The medical charts of all patients with a proven first episode of PCP, diagnosed between 1997 and 2007 were retrospectively reviewed, and clinical and laboratory data abstracted. RESULTS: Of the 35 patients with PCP, 18 were HIV-positive and 17 were HIV-negative with other immunosuppressive conditions. HIV-negative patients were significantly older than HIV-positive patients. The WCC (10 952 +/- 5669 vs 9750 +/- 3133/microL; P = 0.015), neutrophil counts (9631 +/- 5421 vs 5680 +/- 2628/microL; P = 0.01) and CD4+ lymphocyte counts (329 +/- 502 vs 47 +/- 50/microL; P < 0.001) were significantly higher in HIV-negative patients. Six of the 17 HIV-negative patients had a CD4+ lymphocyte count >300/microL. Serum IgG levels were lower in HIV-negative patients (943 +/- 379 vs 1635 +/- 657 mg/dL; P = 0.017). Mortality was higher in HIV-negative (52.9%) than in HIV-positive patients (0%). On univariate analysis, risk factors for mortality from PCP were the presence of underlying pulmonary disease (odds ratio 4.000, 95% CI: 1.501-10.658) and HIV-negative status (odds ratio 2.125, 95% CI: 1.283-3.518). CONCLUSIONS: The characteristics and outcomes of PCP differ significantly depending on HIV status. The existence of underlying pulmonary diseases may be associated with the prognosis of HIV-negative patients with PCP.


Subject(s)
AIDS-Related Opportunistic Infections/diagnosis , HIV Infections/complications , Immunocompromised Host , Pneumocystis carinii , Pneumonia, Pneumocystis/diagnosis , AIDS-Related Opportunistic Infections/immunology , AIDS-Related Opportunistic Infections/mortality , Adult , Aged , CD4 Lymphocyte Count , Diabetes Mellitus/immunology , Female , HIV Infections/immunology , Humans , Immunoglobulin G/blood , Lung Diseases/immunology , Male , Middle Aged , Neutrophils , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/mortality , Prognosis , Retrospective Studies , Risk Factors , Smoking/adverse effects , Treatment Outcome
13.
Nihon Kokyuki Gakkai Zasshi ; 47(2): 116-21, 2009 Feb.
Article in Japanese | MEDLINE | ID: mdl-19260534

ABSTRACT

An 82-year old man was admitted to our hospital for evaluation of progressive general malaise. He had previously been in good health. His chest roentgenogram showed reticular shadows and we suspected interstitial lung disease. On admission, his roentgenographic images showed deterioration compared with previous images. Acute lung injury was diagnosed by transbronchial lung biopsy, and steroid administration was started. He initially responded to treatment, but bilateral spontaneous pneumothorax occurred. Despite treatment, he died of respiratory failure. Amitani disease (idiopathic pulmonary upper lobe fibrosis) was suspected based on postmortem pathology, but his lung parenchyma was poor due to the presence of changes producing diffuse alveolar damage. We report and discuss this case because there are apparently no previous similar cases.


Subject(s)
Pulmonary Fibrosis/physiopathology , Aged, 80 and over , Humans , Male
14.
Exp Lung Res ; 34(6): 343-54, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18600500

ABSTRACT

Long-term, low-dose macrolide therapy has been proven to improve survival in patients with diffuse panbronchiolitis and cystic fibrosis, although the mechanisms by which it does so remain unknown. To elucidate the molecular mechanisms of the anti-inflammatory effects of macrolides, the authors examined the effects of erythromycin (EM-A) and new derivative EM703 on transforming growth factor (TGF)-beta /Smad signaling fibroblasts. EM-A and EM703 each inhibited fibroblast proliferation and the collagen production in human lung fibroblasts induced by TGF-beta. EM-A and EM703 inhibited the augmentation of Smad3 mRNA induced by TGF-beta. Smad7 mRNA was inhibited by TGF-beta, but augmented by coincubation with EM-A or EM703. EM-A and EM703 each inhibited p-Smad2/3 proteins induced by TGF-beta. Smad7 protein inhibited by TGF-beta restored beyond basal level by EM-A and EM703. These findings suggest that EM-A and EM703 inhibit TGF-beta signaling in human lung fibroblasts via inhibition of p-Smad2/3 through recovery of Smad7 level.


Subject(s)
Antifibrinolytic Agents/pharmacology , Erythromycin/analogs & derivatives , Fibroblasts/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Anti-Bacterial Agents/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen/metabolism , Drug Combinations , Erythromycin/pharmacology , Fibroblasts/metabolism , Gene Expression , Humans , Phosphorylation , RNA, Messenger/metabolism , Smad Proteins/genetics
15.
J Nippon Med Sch ; 75(2): 96-105, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18475030

ABSTRACT

Clinical studies have demonstrated that gefitinib, an epidermal growth factor receptor inhibitor, is an effective treatment for some patients with advanced non-small cell lung cancer and is generally well-tolerated. However, several reports have also suggested that gefitinib is associated with acute lung injury and subsequent fibrosis. One hypothesis is that gefitinib exacerbates lung injury induced by radiation therapy. It is important to confirm the safety of gefitinib in radiotherapy for patients with lung cancer. In this preclinical study we aimed to clarify the effect of gefitinib on thoracic radiotherapy. Six-week-old female C57BL/6 mice were immobilized in a plastic frame, and the thorax was irradiated once with a dose of 12 Gy on day 0. Gefitinib (20, 90 and 200 mg/kg/day) was administered on days 0 to 5 (acute phase) or days 14 to 19 (late phase) postirradiation. Thoracic irradiation induced lung injury and subsequent fibrosis 5 months later. Gefitinib, administered in the acute phase, had no effect on lung fibrosis or collagen levels induced by irradiation. A high dose of gefitinib (200 mg/kg/day) administered during the late phase significantly reduced fibrosis scores and collagen levels. These results suggest that gefitinib does not exacerbate radiation-induced lung injury and fibrosis in this strain of mice. Therefore, thoracic irradiation is unlikely to be a risk factor for lung injury associated with gefitinib treatment.


Subject(s)
Antineoplastic Agents/adverse effects , ErbB Receptors/antagonists & inhibitors , Lung/radiation effects , Quinazolines/adverse effects , Quinazolines/therapeutic use , Radiation Injuries, Experimental/pathology , Animals , Female , Gefitinib , Mice , Mice, Inbred C57BL
16.
Intern Med ; 47(7): 585-91, 2008.
Article in English | MEDLINE | ID: mdl-18379141

ABSTRACT

OBJECTIVE: Low-dose, long-term macrolide therapy has been shown to be effective for the treatment of diffuse panbronchiolitis (DPB) and similar disorders in terms of the presence of airway mucus hypersecretion such as bronchiectasis, chronic bronchitis and sinobronchial syndrome. However, there are some patients, especially advanced cases, whose volume of sputum does not decrease sufficiently with macrolide therapy. These patients suffer from copious expectoration. There is currently no effective treatment, and an effective therapy is therefore urgently required. The aim of this study was to clarify whether or not the inhalation of tiotropium improves the symptoms in these cases. METHODS: Tiotropium (18 microg/day) was administered to patients with DPB and similar disorders with airway mucus hypersecretion who did not respond to macrolide. The symptoms were evaluated by a visual analog scale (VAS) prior to and at 1 and 3 months after tiotropium administration. Radiological and pulmonary function tests were also performed to evaluate the effects of tiotropium. RESULTS: Thirteen patients (DPB 5, sinobronchial syndrome 5, bronchiectasis 3) were enrolled. The VAS scores were dramatically improved after the introduction of tiotropium. FEV(1) was significantly improved after 3 months of treatment with tiotropium. In contrast, the radiological findings remained unchanged. CONCLUSION: Tiotropium improved the symptoms of cough, sputum and breathlessness in the macrolide-resistant cases of DPB or similar disorders. These beneficial effects might be due to the suppression of airway secretion through the anticholinergic effect of tiotropium on the submucosal gland, however, the long-term efficiency of this treatment still needs to be further assessed.


Subject(s)
Bronchitis/drug therapy , Macrolides/therapeutic use , Mucus/metabolism , Scopolamine Derivatives/therapeutic use , Administration, Inhalation , Adult , Aged , Bronchitis/pathology , Chronic Disease , Drug Resistance, Microbial/physiology , Female , Humans , Macrolides/pharmacology , Male , Middle Aged , Mucus/drug effects , Pilot Projects , Prospective Studies , Scopolamine Derivatives/pharmacology , Tiotropium Bromide
17.
Intern Med ; 47(1): 15-20, 2008.
Article in English | MEDLINE | ID: mdl-18175999

ABSTRACT

BACKGROUND: Pneumocystis jiroveci pneumonia (PCP) is a potentially fatal complication in interstitial pneumonia patients receiving glucocorticoid therapy. Prophylaxis of PCP during glucocorticoid therapy is an important issue in the treatment of interstitial pneumonia. OBJECTIVE: We evaluated the prophylactic effect of sulfamethoxasole-trimethoprim (TMP-SMX) in interstitial pneumonia patients receiving glucocorticoids. METHODS: We retrospectively analyzed 74 interstitial pneumonia patients who received glucocorticoid therapy. RESULTS: Seven of the 74 patients developed PCP. At the time of diagnosis of PCP, the mean duration of glucocorticoid therapy was 71 days and the mean daily dose of prednisolone was 37 mg. Among the 7 patients, the circulating CD4+ lymphocyte count was 370 /microl on average and it was over 200 /microl in 3 cases. The PCP patients showed a significant reduction of the lymphocyte count at 4 weeks after initiation of steroid therapy. None of the patients who received prophylactic TMP-SMX therapy developed PCP even if the CD4+ lymphocyte count was less than 200 /microl. CONCLUSION: Interstitial pneumonia patients receiving glucocorticoid therapy can benefit from TMP-SMX prophylaxis against PCP. Development of PCP cannot be ruled out in patients with a CD4+ lymphocyte count of greater than 200 /microl.


Subject(s)
Anti-Infective Agents/therapeutic use , Lung Diseases, Interstitial/drug therapy , Pneumocystis carinii , Pneumonia, Pneumocystis/prevention & control , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Adult , Aged , Aged, 80 and over , Antibiotic Prophylaxis , CD4 Lymphocyte Count , Female , Glucocorticoids/adverse effects , Humans , Lung Diseases, Interstitial/diagnostic imaging , Male , Middle Aged , Pneumonia, Pneumocystis/diagnostic imaging , Pneumonia, Pneumocystis/microbiology , Prednisolone/adverse effects , Radiography , Retrospective Studies , Treatment Outcome
18.
Intern Med ; 46(20): 1739-43, 2007.
Article in English | MEDLINE | ID: mdl-17938531

ABSTRACT

A 64-year-old woman with rheumatoid arthritis and treated with bucillamine presented with a productive cough. No obvious infiltration was detected in chest radiography, but CT revealed patchy ground glass opacities in bilateral lung fields. Her serum KL-6 level was elevated and transbronchial lung biopsy showed interstitial pneumonia. Drug lymphocyte stimulation test (DLST) for bucillamine was negative for blood lymphocytes, but positive for bronchoalveolar lavage (BAL) lymphocytes. The pneumonitis improved after the cessation of bucillamine. We therefore made a diagnosis of bucillamine-induced interstitial pneumonia. DLST with BAL lymphocytes is thus suggested to be useful for such diagnoses.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Bronchoalveolar Lavage Fluid/cytology , Cysteine/analogs & derivatives , Lung Diseases, Interstitial/chemically induced , Lymphocyte Activation , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Cysteine/adverse effects , Female , Humans , Lung , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/diagnostic imaging , Middle Aged , Radiography
19.
Nihon Kokyuki Gakkai Zasshi ; 45(7): 551-5, 2007 Jul.
Article in Japanese | MEDLINE | ID: mdl-17682466

ABSTRACT

A 69-year-old man who had been followed for pneumoconiosis complained of dyspnea with effort. He was hospitalized because chest roentogenography showed pleural effusion. Further examination of this pleural effusion revealed an eosinophilic cell population and with a varied appearance. First, we suspected tuberculous pleuritis from the characteristics of the pleural effusion, but we could not demonstrate the existence of any acid-fast bacilli. During diagnostic studies, the patient's respiratory status gradually worsened, making it impossible to obtain essential findings. We initiated steroid administration as an antidote to progressive respiratory failure, and carried out bronchoscopy; As a result, we diagnosed secondary pulmonary cryptococcosis from bronchoalveolar lavarge and transbronchial lung biopsy. Pulmonary cryptococcosis with pleural effusion is rare, and this may be the first report of a case involving a type 1 allergy. We speculate that immunological dysfunction contributed to disease progression in this case.


Subject(s)
Cryptococcosis/etiology , Lung Diseases, Fungal/etiology , Pleural Effusion/complications , Pneumoconiosis/complications , Respiratory Hypersensitivity/complications , Disease Progression , Humans , Male , Middle Aged
20.
Intern Med ; 45(18): 1033-8, 2006.
Article in English | MEDLINE | ID: mdl-17043373

ABSTRACT

OBJECTIVE: This study was aimed to investigate the effect of polymyxin B-immobilized fiber column (PMX) hemoperfusion treatment on the acute exacerbation of idiopathic pulmonary fibrosis (IPF). PATIENTS AND METHODS: Six patients with a clinical diagnosis of idiopathic pulmonary fibrosis (IPF) who developed acute exacerbation were included in this study. Although five of six patients were treated with high-dose corticosteroid therapy, mechanical ventilation was necessary for all six patients due to severe respiratory failure. Blood endotoxin levels were undetectable in all patients. PMX treatment was performed on these six patients. RESULTS: In four of six patients, alveolar-arterial difference of oxygen (AaDO(2)), serum KL-6 and lactate dehydrogenase (LDH) were improved after PMX treatment. These four patients were successfully weaned from mechanical ventilation and survived more than 30 days after the initial PMX treatment. CONCLUSION: These data suggest a potential beneficial effect of PMX treatment on acute exacerbation of IPF.


Subject(s)
Hemoperfusion/methods , Polymyxin B/administration & dosage , Pulmonary Fibrosis/drug therapy , Acute Disease , Aged , Antigens, Neoplasm/blood , Female , Glucocorticoids/therapeutic use , Humans , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Mucin-1 , Mucins/blood , Oxygen/analysis , Pilot Projects , Pulmonary Fibrosis/therapy , Respiration, Artificial , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...