Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Exp Cell Res ; 398(2): 112416, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33307020

RESUMO

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.


Assuntos
Fibrose Pulmonar Idiopática/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , MicroRNAs/metabolismo , Osteonectina/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Animais , Exossomos/metabolismo , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Respir Res ; 19(1): 71, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29690905

RESUMO

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.


Assuntos
Bleomicina/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/terapia , Baço/transplante , Linfócitos T Reguladores/transplante , Animais , Bleomicina/administração & dosagem , Bombas de Infusão Implantáveis , Transfusão de Linfócitos/métodos , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibrose Pulmonar/metabolismo , Baço/citologia , Linfócitos T Reguladores/metabolismo
3.
Pulm Pharmacol Ther ; 44: 61-69, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28315487

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Fibrose Pulmonar Idiopática/fisiopatologia , Fatores de Troca de Nucleotídeo Guanina Rho/efeitos dos fármacos , Benzamidas/farmacologia , Células Cultivadas , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Inativação Gênica , Humanos , Ácidos Hidroxâmicos/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Osteonectina/genética , Piridinas/farmacologia , RNA Interferente Pequeno/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Fator de Crescimento Transformador beta1/administração & dosagem , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima , Vorinostat
4.
Pulm Pharmacol Ther ; 32: 29-36, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25843005

RESUMO

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.


Assuntos
Células Epiteliais/efeitos dos fármacos , Indóis/farmacologia , Proteína D Associada a Surfactante Pulmonar/genética , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/fisiopatologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/metabolismo
5.
Blood Purif ; 32(4): 310-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21893977

RESUMO

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.


Assuntos
Proteína HMGB1/sangue , Hemoperfusão , Fibrose Pulmonar Idiopática/sangue , Fibrose Pulmonar Idiopática/terapia , Polimixina B/uso terapêutico , Idoso , Feminino , Hemoperfusão/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Oxigênio/análise , Polimixina B/química , Resultado do Tratamento
6.
Respirology ; 15(1): 126-31, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19947989

RESUMO

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.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/diagnóstico , Infecções por HIV/complicações , Hospedeiro Imunocomprometido , Pneumocystis carinii , Pneumonia por Pneumocystis/diagnóstico , Infecções Oportunistas Relacionadas com a AIDS/imunologia , Infecções Oportunistas Relacionadas com a AIDS/mortalidade , Adulto , Idoso , Contagem de Linfócito CD4 , Diabetes Mellitus/imunologia , Feminino , Infecções por HIV/imunologia , Humanos , Imunoglobulina G/sangue , Pneumopatias/imunologia , Masculino , Pessoa de Meia-Idade , Neutrófilos , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/mortalidade , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Fumar/efeitos adversos , Resultado do Tratamento
7.
Blood Purif ; 29(4): 321-6, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20185904

RESUMO

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.


Assuntos
Hemoperfusão/métodos , Doenças Pulmonares Intersticiais/terapia , Neutrófilos/patologia , Polimixina B/uso terapêutico , Idoso , Separação Celular , Forma Celular , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Doenças Pulmonares Intersticiais/patologia , Masculino , Pessoa de Meia-Idade , Observação/métodos , Projetos Piloto
8.
J Nippon Med Sch ; 87(3): 118-128, 2020 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-31776321

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Exossomos/genética , Exossomos/fisiologia , Fibroblastos/patologia , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , MicroRNAs/fisiologia , Miofibroblastos/patologia , Actinas/genética , Actinas/metabolismo , Animais , Modelos Animais de Doenças , Expressão Gênica/genética , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo
9.
Thorac Cancer ; 11(7): 1876-1884, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32421226

RESUMO

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/).


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Derrame Pleural Maligno/tratamento farmacológico , Pleurodese/efeitos adversos , Adenocarcinoma de Pulmão/etiologia , Adenocarcinoma de Pulmão/patologia , Idoso , Bevacizumab/administração & dosagem , Carboplatina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Docetaxel/administração & dosagem , Cloridrato de Erlotinib/administração & dosagem , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/etiologia , Recidiva Local de Neoplasia/patologia , Pemetrexede/administração & dosagem , Derrame Pleural Maligno/complicações , Derrame Pleural Maligno/patologia , Prognóstico , Taxa de Sobrevida
10.
Nihon Kokyuki Gakkai Zasshi ; 47(2): 116-21, 2009 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-19260534

RESUMO

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.


Assuntos
Fibrose Pulmonar/fisiopatologia , Idoso de 80 Anos ou mais , Humanos , Masculino
11.
Exp Lung Res ; 34(6): 343-54, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18600500

RESUMO

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.


Assuntos
Antifibrinolíticos/farmacologia , Eritromicina/análogos & derivados , Fibroblastos/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Antibacterianos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colágeno/metabolismo , Combinação de Medicamentos , Eritromicina/farmacologia , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Fosforilação , RNA Mensageiro/metabolismo , Proteínas Smad/genética
12.
J Nippon Med Sch ; 75(2): 96-105, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18475030

RESUMO

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.


Assuntos
Antineoplásicos/efeitos adversos , Receptores ErbB/antagonistas & inibidores , Pulmão/efeitos da radiação , Quinazolinas/efeitos adversos , Quinazolinas/uso terapêutico , Lesões Experimentais por Radiação/patologia , Animais , Feminino , Gefitinibe , Camundongos , Camundongos Endogâmicos C57BL
13.
Nihon Kokyuki Gakkai Zasshi ; 45(7): 551-5, 2007 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-17682466

RESUMO

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.


Assuntos
Criptococose/etiologia , Pneumopatias Fúngicas/etiologia , Derrame Pleural/complicações , Pneumoconiose/complicações , Hipersensibilidade Respiratória/complicações , Progressão da Doença , Humanos , Masculino , Pessoa de Meia-Idade
14.
Respir Res ; 7: 16, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16438734

RESUMO

BACKGROUND: Fourteen-membered ring macrolides have been effective in reducing chronic airway inflammation and also preventing lung injury and fibrosis in bleomycin-challenged mice via anti-inflammatory effects. EM703 is a new derivative of erythromycin (EM) without the bactericidal effects. We investigated the anti-inflammatory and antifibrotic effects of EM703 in an experimental model of bleomycin-induced lung injury and subsequent fibrosis in mice. METHODS: Seven-week-old male ICR mice were used. All experiments used eight mice/group, unless otherwise noted in the figure legends. Bleomycin was administered intravenously to the mice on day 0. EM703 was orally administered daily to mice. All groups were examined for cell populations in the bronchoalveolar lavage (BAL) fluid and for induction of messenger RNA (mRNA) of Smad3 and Smad4 in the lung tissues by reverse transcriptase (RT)-polymerase chainreaction (PCR) on day 7. Fibroblastic foci were assessed histologically, and the hydroxyproline content was chemically determined in the lung tissues on day 28. We performed assay of proliferation and soluble collagen production, and examined the induction of mRNA of Smad3 and Smad4 by RT-PCR in murine lung fibroblast cell line MLg2908. We also examined Smad3, Smad4 and phosphorylated Smad2/3 (p-Smad2/3) protein assay by western blotting in MLg2908. RESULTS: Bleomycin-induced lung fibrosis, and the infiltration of macrophages and neutrophils into the airspace were inhibited by EM703. The expression of Smad3 and Smad4 mRNA was clearly attenuated by bleomycin, but was recovered by EM703. EM703 also inhibited fibroblast proliferation and the collagen production in lung fibroblasts induced by Transforming growth factor-beta (TGF-beta). The expression of Smad3 and Smad4 mRNA in murine lung fibroblasts disappeared due to TGF-beta, but was recovered by EM703. EM703 inhibited the expression of p-Smad2/3 and Smad4 protein in murine lung fibroblasts induced by TGF-beta. CONCLUSION: These findings suggest that EM703 improves bleomycin-induced pulmonary fibrosis in mice by actions of anti-inflammation and regulation of TGF-beta signaling in lung fibroblasts.


Assuntos
Anti-Inflamatórios/farmacologia , Eritromicina/análogos & derivados , Pulmão/efeitos dos fármacos , Fibrose Pulmonar/prevenção & controle , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Sequência de Bases , Bleomicina , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Eritromicina/farmacologia , Eritromicina/uso terapêutico , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica , Hidroxiprolina/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/farmacologia
15.
Nihon Kokyuki Gakkai Zasshi ; 44(12): 938-43, 2006 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-17233391

RESUMO

A 68-year-old woman had been given a diagnosis of interstitial pneumonia (NSIP pattern) and followed up at our hospital for 3 years. She was admitted to our hospital because of dyspnea, lower limb edema and myalgia. On admission, serum CPK and CRP levels were elevated and an electromyogram suggested inflammatory myopathy. We diagnosed polymyositis (PM) with progressive interstitial pneumonia (IP). Although methylprednisolone pulse therapy and immunosuppressive agents were administered, pulmonary lesions became aggravated. The patient died due to respiratory failure as a result of the progress of IP. The autopsy lung revealed diffuse alveolar damage (DAD) at the both acute and fibrotic phases, suggesting that DAD could coincide with PM. We report here a rare case of polymyositis with diffuse alveolar damage (DAD).


Assuntos
Doenças Pulmonares Intersticiais/complicações , Polimiosite/etiologia , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/etiologia , Idoso , Eletromiografia , Feminino , Humanos , Doenças Pulmonares Intersticiais/patologia , Polimiosite/diagnóstico , Fibrose Pulmonar/patologia
16.
Sarcoidosis Vasc Diffuse Lung Dis ; 33(3): 242-246, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27758989

RESUMO

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.


Assuntos
Antibacterianos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Fibrose Pulmonar Idiopática/tratamento farmacológico , Pulmão/efeitos dos fármacos , Macrolídeos/uso terapêutico , Corticosteroides/uso terapêutico , Idoso , Progressão da Doença , Quimioterapia Combinada , Feminino , Humanos , Fibrose Pulmonar Idiopática/diagnóstico , Fibrose Pulmonar Idiopática/mortalidade , Fibrose Pulmonar Idiopática/fisiopatologia , Imunossupressores/uso terapêutico , Estimativa de Kaplan-Meier , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento
17.
Nihon Kokyuki Gakkai Zasshi ; 43(12): 725-30, 2005 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-16457333

RESUMO

We evaluated the clinical features of pneumocystis jiroveci pneumonia (PCP) as a complication of glucocorticoid therapy for interstitial pneumonia We analyzed 74 interstitial pneumonia patients receiving glucocorticoid therapy, of whom 7 patients developed PCP. At the time of PCP diagnosis, the average duration of the glucocorticoid therapy was 71 days and the average daily dose of predonisolone was 37 mg. Circulating CD4+ lymphocyte counts were 370/microl on the average and more than 200/microl in three cases. PCP cases showed less circulating lymphocyte counts four weeks after the initiation of the therapy. Any cases receiving sulfamethoxazole-trimethoprim (TMP-SMX) did not develop PCP. In conclusion, interstitial pneumonia patients, who are treated with glucocorticoid, are benefit from TMP-SMX as PCP prophylaxis, but CD4 + lymphocyte counts greater than 200/microl is no reason to denying PCP.


Assuntos
Glucocorticoides/efeitos adversos , Doenças Pulmonares Intersticiais/complicações , Pneumonia por Pneumocystis/etiologia , Prednisolona/efeitos adversos , Idoso , Anti-Infecciosos/administração & dosagem , Esquema de Medicação , Feminino , Glucocorticoides/administração & dosagem , Humanos , Doenças Pulmonares Intersticiais/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Infecções Oportunistas/imunologia , Infecções Oportunistas/prevenção & controle , Pneumocystis carinii/isolamento & purificação , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/prevenção & controle , Prednisolona/administração & dosagem , Estudos Retrospectivos , Combinação Trimetoprima e Sulfametoxazol/administração & dosagem
18.
Chest ; 123(6): 2007-11, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12796182

RESUMO

STUDY OBJECTIVE: Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease of unknown etiology. The incidence of IPF increases with age. Aging contributes to lifestyle-related diseases, including diabetes mellitus (DM); therefore, it is possible that lifestyle-related diseases may affect either the initiation or progression of IPF. DESIGN: Case-control study. SETTING: Nippon Medical School and Omiya City Clinic. PATIENTS AND PARTICIPANTS: Sixty-five consecutive patients with IPF who were admitted to Nippon Medical School Hospital from 1995 to 2000, and 184 control subjects selected from 15,798 subjects who were admitted to Omiya City Clinic for routine medical examination between September 1999 and August 2000. MEASUREMENTS: Age, sex, smoking history, and results of physical examinations, blood examinations, and lung function testing were extracted from medical records and were compared with the diagnostic criteria for lifestyle-related diseases. RESULTS: The adjusted odds ratios for cigarette smoking were 5.40 (95% confidence interval [CI], 2.30 to 12.66) and 4.06 (95% CI, 1.80 to 9.15) for diabetes. There were no differences in clinical characteristics of patients with IPF that could be related to the presence of DM. CONCLUSION: DM may be a risk factor for IPF.


Assuntos
Complicações do Diabetes , Fibrose Pulmonar/etiologia , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Razão de Chances , Fatores de Risco , Fumar/efeitos adversos
19.
Chest ; 122(6): 2137-45, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12475858

RESUMO

BACKGROUND AND OBJECTIVE: Although the pathogenesis of interstitial pneumonia and pulmonary fibrosis are not well understood, it has been reported that inflammatory cells, especially neutrophils, and the injurious substances produced by them play important roles in the progression of interstitial pneumonia and subsequent fibrosis. Erythromycin and other 14-membered ring macrolides (14-MRMLs) have been reported to improve the survival of patients with diffuse panbronchiolitis by antineutrophil and several other anti-inflammatory mechanisms. The present study was undertaken to investigate the effects of 14-MRMLs on an experimental model of bleomycin-induced acute lung injury and subsequent fibrosis in mice. METHODS: Bleomycin was administered IV to ICR mice. At 28 days after bleomycin injection, fibrotic foci were histologically observed in left lung tissues, and hydroxyproline content in right lung tissues was chemically analyzed. The inhibitory effects of 14-MRMLs were assessed by overall comparison between control (normal saline solution [NS] alone), untreated (bleomycin alone), and treated (bleomycin plus 14-MRMLs) groups. For evaluation of early-phase inflammation, cell populations in BAL fluid and induction of messenger RNA (mRNA) of adhesion molecules (E-selectin, P-selectin, intercellular adhesion molecule 1 [ICAM-1], and vascular cell adhesion molecule 1 [VCAM-1]) in lung tissues were examined at 0 to 13 days after bleomycin treatment. These parameters were also compared with those for the control (NS alone), 14-MRML untreated (bleomycin alone), and 14-MRML pretreated (bleomycin plus 14-MRML pretreated) groups. RESULTS: Bleomycin-induced pulmonary fibrosis was inhibited by erythromycin and other 14-MRMLs on day 28 after bleomycin injection in ICR mice, especially those pretreated with 14-MRMLs. Hydroxyproline content in lung tissues was also decreased in the 14-MRML-pretreated groups. The number of neutrophils in BAL fluid significantly increased, with two peaks at 1 day and 9 days (from 6 to 11 days) after bleomycin administration. 14-MRMLs significantly inhibited both peaks of neutrophil infiltration into the airspace. Changes in mRNA expression of adhesion molecules (E-selectin, P-selectin, ICAM-1, VCAM-1) were associated with leukocyte migration into the airspace. 14-MRMLs clearly inhibited the induction of VCAM-1 mRNA, and tended to attenuate that of ICAM-1 mRNA, but inhibited the induction of neither E-selectin mRNA nor P-selectin mRNA. CONCLUSION: These findings indicate that attenuation of inflammatory cell migration into the airspace by 14-MRMLs, especially of neutrophils and macrophages, resulted in inhibition of lung injury and subsequent fibrosis. 14-MRMLs clearly attenuated the expression of VCAM-1 mRNA during the early phase of bleomycin-induced lung injury, and this might be one mechanism of inhibition of neutrophil and macrophage migration into the airspace by 14-MRMLs. This may be one mechanism of the anti-inflammatory and antifibrotic effects of 14-MRMLs. These findings suggest that prophylactic administration of 14-MRMLs may be clinically efficacious in preventing acute exacerbation of interstitial pneumonia and acute lung injury.


Assuntos
Antibacterianos/farmacologia , Movimento Celular/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Doenças Pulmonares Intersticiais/prevenção & controle , Pulmão/patologia , RNA Mensageiro/biossíntese , Molécula 1 de Adesão de Célula Vascular/genética , Animais , Bleomicina , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Eritromicina/farmacologia , Hidroxiprolina/análise , Inflamação/patologia , Molécula 1 de Adesão Intercelular/genética , Doenças Pulmonares Intersticiais/induzido quimicamente , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Selectina-P/genética
20.
Sarcoidosis Vasc Diffuse Lung Dis ; 20(2): 152-4, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12870727

RESUMO

Dendriform pulmonary ossification is an extremely rare disease, which is usually found and analysed postmortem. Pathogenesis of pulmonary ossification is therefore still unknown. We describe two males in the same family (a 29 year-old patients and his 58 year-old father) with pulmonary ossification. The young male was symptomatic pneumothorax but his father had been asymptomatic since youth. Familial clustering of pulmonary ossifications strongly suggests a genetic predisposition for the pathogenesis. This is the first report of two cases of dendriform pulmonary ossification in the same family.


Assuntos
Predisposição Genética para Doença , Pneumopatias/genética , Ossificação Heterotópica/genética , Adulto , Humanos , Pneumopatias/diagnóstico por imagem , Pneumopatias/patologia , Masculino , Pessoa de Meia-Idade , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/patologia , Pneumotórax/etiologia , Radiografia Torácica , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa