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

Country/Region as subject
Affiliation country
Publication year range
1.
J Cell Physiol ; 237(1): 566-579, 2022 01.
Article in English | MEDLINE | ID: mdl-34231213

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is defined as a specific form of chronic, progressive fibrosing interstitial pneumonia. It is unknown why fibrosis in IPF distributes in the peripheral or named sub-pleural area. Migration of pleural mesothelial cells (PMC) should contribute to sub-pleural fibrosis. Calpain is known to be involved in cell migration, but the role of calpain in PMC migration has not been investigated. In this study, we found that PMCs migrated into lung parenchyma in patients with IPF. Then using Wt1tm1(EGFP/Cre)Wtp /J knock-in mice, we observed PMC migration into lung parenchyma in bleomycin-induced pleural fibrosis models, and calpain inhibitor attenuated pulmonary fibrosis with prevention of PMC migration. In vitro studies revealed that bleomycin and transforming growth factor-ß1 increased calpain activity in PMCs, and activated calpain-mediated focal adhesion (FA) turnover as well as cell migration, cell proliferation, and collagen-I synthesis. Furthermore, we determined that calpain cleaved FA kinase in both C-terminal and N-terminal regions, which mediated FA turnover. Lastly, the data revealed that activated calpain was also involved in phosphorylation of cofilin-1, and p-cofilin-1 induced PMC migration. Taken together, this study provides evidence that calpain mediates PMC migration into lung parenchyma to promote sub-pleural fibrosis in IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Actin Depolymerizing Factors/metabolism , Animals , Bleomycin/pharmacology , Calpain/metabolism , Cell Movement , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/pathology , Mice , Transforming Growth Factor beta1/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L990-L1004, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33787325

ABSTRACT

The distribution of fibrosis in idiopathic pulmonary fibrosis (IPF) is subpleural with basal predominance. Alveolar epithelial cell was considered as the key cell in the initial phase of IPF. However, the idea of activation and damage of alveolar epithelial cells is very difficult to explain why fibrosis distributes in the subpleural area. In this study, human pleural mesothelial cell (PMC) line and primary rat PMC was used as in vitro model. Intraperitoneal injection of bleomycin was used for making a pulmonary fibrosis model. The integrity of cultured monolayer PMCs was determined by transepithelial electric resistance (TEER). Pleural permeability was estimated by measuring paracellular transport of fluorescein isothiocyanate (FITC)-conjugated dextran. Changes in lung tissue of patients with IPF were analyzed by Masson's and immunofluorescence staining. We found bleomycin induced PMCs damage and increased PMCs permeability; increased PMCs permeability aggravated bleomycin-induced subpleural inflammation and pulmonary fibrosis. Moreover, bleomycin was found to activate VEGF/Src signaling which increased PMCs permeability. In vivo, inhibition of VEGF/Src signaling prevented bleomycin-induced subpleural pulmonary fibrosis. At last, activation of VEGF/Src signaling was confirmed in subpleural area in patients with IPF. Taken together, our findings indicate that VEGF/Src signaling mediated pleural barrier damage and increased permeability which contributes to subpleural pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/pathology , Permeability/drug effects , Pleura/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Bleomycin/pharmacology , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelium/drug effects , Epithelium/pathology , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Lung/metabolism , Pleura/drug effects , Rats , Signal Transduction/drug effects
3.
Exp Cell Res ; 396(1): 112295, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32971116

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal fibrosing interstitial lung disease with limited therapeutic options and a median survival of 3 years after diagnosis. Dysregulated epithelial regeneration is key event involved in initiating and sustaining IPF. The type II alveolar epithelial cells (AECIIs) play a crucial role for epithelial regeneration and stabilisation of alveoli. Loss of cell apical-basal polarity contributes to fibrosis. AECII has apical-basal polarity, but it is poorly understood whether AECII apical-basal polarity loss is involved in fibrosis. Bleomycin is a traditional inducer of pulmonary fibrosis. Here firstly we observed that bleomycin induced apical-basal polarity loss in cultured AECIIs. Next, cell polarity proteins lethal (2) giant larvae 1 (Lgl1), PAR-3A, aPKC and PAR-6B were investigated. We found bleomycin induced increases of Lgl1 protein and decreases of PAR-3A protein, and bleomycin-induced PAR-3A depression was mediated by increased-Lgl1. Then Lgl1 siRNA was transfected into AECIIs. Lgl1 siRNA prevented apical-basal polarity loss in bleomycin-treated AECIIs. At last, Lgl1-conditional knockout mice were applied in making animal models. Bleomycin induced pulmonary fibrosis, but this was attenuated in Lgl1-conditional knockout mice. Together, these data indicated that bleomycin mediated AECII apical-basal polarity loss which contributed to experimental pulmonary fibrosis. Inhibition of Lgl1 should be a potential therapeutic strategy for the disease.


Subject(s)
Alveolar Epithelial Cells/drug effects , Bleomycin/pharmacology , Cell Polarity/drug effects , Glycoproteins/genetics , Pulmonary Fibrosis/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Polarity/genetics , Disease Models, Animal , Gene Expression Regulation , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice, Knockout , Primary Cell Culture , Protein Kinase C/genetics , Protein Kinase C/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction
4.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1201-1210, 2018 09.
Article in English | MEDLINE | ID: mdl-29842893

ABSTRACT

Pleural fibrosis is barely reversible and the underlying mechanisms are poorly understood. Pleural mesothelial cells (PMCs) which have apical-basal polarity play a key role in pleural fibrosis. Loss of cell polarity is involved in the development of fibrotic diseases. Partition defective protein (PAR) complex is a key regulator of cell polarity. However, changes of PMC polarity and PAR complex in pleural fibrosis are still unknown. In this study, we observed that PMC polarity was lost in fibrotic pleura. Next we found increased Lethal (2) giant larvae (Lgl) bound with aPKC and PAR-6B competing against PAR-3A in PAR complex, which led to cell polarity loss. Then we demonstrated that Lgl1 siRNA prevented cell polarity loss in PMCs, and Lgl1 conditional knockout (ER-Cre+/-Lgl1flox/flox) attenuated pleural fibrosis in a mouse model. Our data indicated that Lgl1 regulates cell polarity of PMCs, inhibition of Lgl1 and maintenance of cell polarity in PMCs could be a potential therapeutic treatment approach for pleural fibrosis.


Subject(s)
Epithelial Cells/cytology , Glycoproteins/genetics , Glycoproteins/metabolism , Pleura/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Polarity , Disease Models, Animal , Epithelial Cells/metabolism , Female , Fibrosis , Gene Knockout Techniques , Humans , Male , Mice , Pleura/metabolism , Protein Kinase C/metabolism , Rats
5.
Pulm Pharmacol Ther ; 48: 46-52, 2018 02.
Article in English | MEDLINE | ID: mdl-29107090

ABSTRACT

Pleural fibrosis is associated with various inflammatory processes such as tuberculous pleurisy and bacterial empyema. There is currently no ideal therapeutic to attenuate pleural fibrosis. Some pro-fibrogenic mediators induce fibrosis through inflammatory processes, suggesting that blockage of these mediators might prevent pleural fibrosis. The MeT-5A human pleural mesothelial cell line (PMC) was used in this study as an in vitro model of fibrosis; and intra-pleural injection of bleomycin with carbon particles was used as an in vivo mouse model of pleural fibrosis. Calpain knockout mice, calpain inhibitor (calpeptin), and angiotensin (Ang) II type 1 receptor (AT1R) antagonist (losartan) were evaluated in prevention of experimental pleural fibrosis. We found that bleomycin and carbon particles induced calpain activation in cultured PMCs. This in vitro response was associated with increased collagen-I synthesis, and was blocked by calpain inhibitor or AT1R antagonist. Calpain genetic or treatment with calpeptin or losartan prevented pleural fibrosis in a mouse model induced by bleomycin and carbon particles. Our findings indicate that Ang II signaling and calpain activation induce collagen-I synthesis and contribute to fibrotic alterations in pleural fibrosis. Inhibition of Ang II and calpain might therefore be a novel strategy in treatment of pleural fibrosis.


Subject(s)
Calpain/genetics , Dipeptides/pharmacology , Losartan/pharmacology , Pleural Diseases/drug therapy , Angiotensin II/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Bleomycin/toxicity , Calpain/antagonists & inhibitors , Carbon/toxicity , Cell Line , Collagen Type I/metabolism , Disease Models, Animal , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pleural Diseases/physiopathology
6.
Mol Ther ; 25(3): 728-738, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28131417

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease that typically leads to respiratory failure and death within 3-5 years of diagnosis. Sub-pleural pulmonary fibrosis is a pathological hallmark of IPF. Bleomycin treatment of mice is a an established pulmonary fibrosis model. We recently showed that bleomycin-induced epithelial-mesenchymal transition (EMT) contributes to pleural mesothelial cell (PMC) migration and sub-pleural pulmonary fibrosis. MicroRNA (miRNA) expression has recently been implicated in the pathogenesis of IPF. However, changes in miRNA expression in PMCs and sub-pleural fibrosis have not been reported. Using cultured PMCs and a pulmonary fibrosis animal model, we found that miR-18a-5p was reduced in PMCs treated with bleomycin and that downregulation of miR-18a-5p contributed to EMT of PMCs. Furthermore, we determined that miR-18a-5p binds to the 3' UTR region of transforming growth factor ß receptor II (TGF-ßRII) mRNA, and this is associated with reduced TGF-ßRII expression and suppression of TGF-ß-Smad2/3 signaling. Overexpression of miR-18a-5p prevented bleomycin-induced EMT of PMC and inhibited bleomycin-induced sub-pleural fibrosis in mice. Taken together, our data indicate that downregulated miR-18a-5p mediates sub-pleural pulmonary fibrosis through upregulation of its target, TGF-ßRII, and that overexpression of miR-18a-5p might therefore provide a novel approach to the treatment of IPF.


Subject(s)
Gene Expression Regulation , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptors, Transforming Growth Factor beta/genetics , Animals , Bleomycin/pharmacology , Cats , Cell Movement/genetics , Cluster Analysis , Disease Models, Animal , Epithelial-Mesenchymal Transition , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Mice , Pleura/metabolism , Pleura/pathology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
7.
Biochim Biophys Acta ; 1852(9): 1796-804, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26071646

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease of unknown cause that typically leads to respiratory failure and death within 3-5years of diagnosis. TGF-ß1 is considered a major profibrotic factor. However, TGF-ß1 is necessary but not sufficient to the pathogenesis of fibrotic lesion of the lungs. Recent observations have revealed that calpain, a calcium dependent protease, plays a pivotal role in tissue remodeling and fibrosis. However, the mechanism of calpain mediating pulmonary fibrosis is not understood. Calpain conditional knockout (ER-Cre(+/-)capns1(flox/flox)) mice and primary human lung fibroblasts (HLFs) were used here to investigate the relationship between calpain and TGF-ß1. Calpain knockout mice were protected from fibrotic effects of bleomycin. Bleomycin induced increases in TGF-ß1 via calpain activation in HLFs. Moreover, TGF-ß1 also activated calpain. This crosstalk between calpain activation and TGF-ß1 triggered the downstream signaling pathway including TGF-ß1 Smad2/3 and non-Smad (Akt) pathways, as well as collagen-I synthesis. Taken together, our data indicate that the crosstalk between calpain activation and TGF-ß1 augments collagen-I synthesis in HLFs and in pulmonary fibrosis. Intervention in the crosstalk between calpain activation and TGF-ß1 is a novel potential strategy to prevent pulmonary fibrosis.

8.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L145-53, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27261452

ABSTRACT

Pleural fibrosis is defined as an excessive deposition of extracellular matrix (ECM) components that results in destruction of the normal pleural tissue architecture. It can result from diverse inflammatory conditions, especially tuberculous pleurisy. Pleural mesothelial cells (PMCs) play a pivotal role in pleural fibrosis. Calpain is a family of calcium-dependent endopeptidases, which plays an important role in ECM remodeling. However, the role of calpain in pleural fibrosis remains unknown. In the present study, we found that tuberculous pleural effusion (TPE) induced calpain activation in PMCs and that inhibition of calpain prevented TPE-induced collagen-I synthesis and cell proliferation of PMCs. Moreover, our data revealed that the levels of angiotensin (ANG)-converting enzyme (ACE) were significantly higher in pleural fluid of patients with TPE than those with malignant pleural effusion, and ACE-ANG II in TPE resulted in activation of calpain and subsequent triggering of the phosphatidylinositol 3-kinase (PI3K)/Akt/NF-κB signaling pathway in PMCs. Finally, calpain activation in PMCs and collagen depositions were confirmed in pleural biopsy specimens from patients with tuberculous pleurisy. Together, these studies demonstrated that calpain is activated by renin-angiotensin system in pleural fibrosis and mediates TPE-induced collagen-I synthesis and proliferation of PMCs via the PI3K/Akt/NF-κB signaling pathway. Calpain in PMCs might be a novel target for intervention in tuberculous pleural fibrosis.


Subject(s)
Calpain/metabolism , Tuberculosis, Pleural/enzymology , Adolescent , Adult , Aged , Angiotensin II/physiology , Cell Proliferation , Cells, Cultured , Collagen Type I/biosynthesis , Enzyme Activation , Epithelium/pathology , Female , Fibrosis , Humans , Male , Middle Aged , Pleura/microbiology , Pleura/pathology , Pleural Effusion, Malignant/enzymology , Renin-Angiotensin System , Signal Transduction , Tuberculosis, Pleural/pathology , Young Adult
9.
Toxicol Appl Pharmacol ; 283(2): 75-82, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25595642

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease characterized by the development of subpleural foci of myofibroblasts that contribute to the exuberant fibrosis. Recent studies revealed that pleural mesothelial cells (PMCs) undergo epithelial-mesenchymal transition (EMT) and play a pivotal role in IPF. In animal model, bleomycin induces pulmonary fibrosis exhibiting subpleural fibrosis similar to what is seen in human IPF. It is not known yet whether bleomycin induces EMT in PMCs. In the present study, PMCs were cultured and treated with bleomycin. The protein levels of collagen-I, mesenchymal phenotypic markers (vimentin and α-smooth muscle actin), and epithelial phenotypic markers (cytokeratin-8 and E-cadherin) were measured by Western blot. PMC migration was evaluated using wound-healing assay of culture PMCs in vitro, and in vivo by monitoring the localization of PMC marker, calretinin, in the lung sections of bleomycin-induced lung fibrosis. The results showed that bleomycin induced increases in collagen-I synthesis in PMC. Bleomycin induced significant increases in mesenchymal phenotypic markers and decreases in epithelial phenotypic markers in PMC, and promoted PMC migration in vitro and in vivo. Moreover, TGF-ß1-Smad2/3 signaling pathway involved in the EMT of PMC was demonstrated. Taken together, our results indicate that bleomycin induces characteristic changes of EMT in PMC and the latter contributes to subpleural fibrosis.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Epithelial-Mesenchymal Transition/drug effects , Respiratory Mucosa/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Epithelium/drug effects , Epithelium/pathology , Humans , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Respiratory Mucosa/pathology
10.
J Immunol ; 185(10): 6348-54, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20952674

ABSTRACT

IL-17-producing CD4(+) T (Th17) cells have been found to be increased in some human cancers; however, the possible implication of Th17 cells in regulating antitumor responses in malignant pleural effusion (MPE) remains to be elucidated. In the current study, distribution and phenotypic features of Th17 cells in both MPE and peripheral blood from patients with lung cancer were determined by flow cytometry or double immunofluorescence staining. The impacts of cytokines on Th17 cell generation and differentiation were explored. The chemoattractant activity of chemokines CCL20 and CCL22 for Th17 cells in vitro was also observed. It was found that the increased Th17 cells could be found in MPE compared with blood. The in vitro experiments showed that IL-1ß, IL-6, IL-23, or their various combinations could promote Th17 cell generation and differentiation from naive CD4(+) T cells. MPE was chemotactic for Th17 cells, and this activity was partly blocked by anti-CCL20 and/or CCL22 Abs. Our data also showed that the accumulation of Th17 cells in MPE predicted improved patient survival. It could be concluded that the overrepresentation of Th17 cells in MPE might be due to Th17 cell differentiation and expansion stimulated by pleural proinflammatory cytokines and to recruitment of Th17 cells from peripheral blood induced by pleural chemokines CCL20 and CCL22. Furthermore, the accumulation of Th17 cells in MPE predicted improved patient survival. These data provide the basis for developing immune-boosting strategies based on ridding the cancer patient of this cell population.


Subject(s)
Cell Differentiation/immunology , Chemotaxis, Leukocyte/immunology , Lung Neoplasms/immunology , Pleural Effusion, Malignant/immunology , Th17 Cells/cytology , Adult , Aged , Aged, 80 and over , Cell Separation , Chemokines/analysis , Chemokines/immunology , Chemokines/metabolism , Cytokines/analysis , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Middle Aged , Pleural Effusion, Malignant/cytology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
11.
Respir Res ; 12: 77, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663645

ABSTRACT

BACKGROUND: Both regulatory T cells (Tregs) and T helper IL-17-producing cells (Th17 cells) have been found to be involved in human malignancies, however, the possible implication of Tregs in regulating generation and differentiation of Th17 cells in malignant pleural effusion remains to be elucidated. METHODS: The numbers of both CD39(+)Tregs and Th17 cells in malignant pleural effusion and peripheral blood from patients with lung cancer were determined by flow cytometry. The regulation and mechanism of Tregs on generation and differentiation of Th17 cells were explored. RESULTS: Both CD39(+)Tregs and Th17 cells were increased in malignant pleural effusion when compared with blood, and the numbers of CD39(+)Tregs were correlated negatively with those of Th17 cells. It was also noted that high levels of IL-1ß, IL-6, and TGF-ß1 could be observed in malignant pleural effusion when compared the corresponding serum, and that pleural CD39(+)Tregs could express latency-associated peptide on their surface. When naïve CD4(+) T cells were cocultured with CD39(+)Tregs, Th17 cell numbers decreased as CD39(+)Treg numbers increased, addition of the anti-latency-associated peptide mAb to the coculture reverted the inhibitory effect exerted by CD39(+)Tregs. CONCLUSIONS: Therefore, the above results indicate that CD39(+)Tregs inhibit generation and differentiation of Th17 cells via a latency-associated peptide-dependent mechanism.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Cell Differentiation , Cell Proliferation , Lung Neoplasms/immunology , Peptides/metabolism , Pleural Effusion, Malignant/immunology , Protein Precursors/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Adult , Aged , Cells, Cultured , Coculture Techniques , Flow Cytometry , Humans , Immunophenotyping , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung Neoplasms/complications , Middle Aged , Signal Transduction , Transforming Growth Factor beta1/metabolism
12.
Respirology ; 16(3): 473-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21299686

ABSTRACT

BACKGROUND AND OBJECTIVE: The diagnosis of tuberculous pleurisy by analysis of pleural fluid using standard diagnostic tools is difficult. Recently, T-cell interferon-γ release assays (IGRA) have been introduced for the diagnosis of tuberculous pleurisy. The aim of the present meta-analysis was to establish the overall diagnostic accuracy of IGRA on both pleural fluid and peripheral blood, for diagnosing tuberculous pleurisy. METHODS: A systematic review was performed of English language publications. Sensitivity, specificity and other measures of the accuracy of IGRA for the diagnosis tuberculous pleurisy using both pleural fluid and blood were pooled using a random-effects model or a fixed-effects model. Receiver operating characteristic curves were used to summarize overall test performance. RESULTS: Seven out of eight studies met the inclusion criteria. The summary estimates of sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, positive predictive value, negative predictive value and diagnostic odds ratio were, for pleural fluid: 0.75, 0.82, 3.49, 0.24, 0.85, 0.70 and 19.04, respectively; and for blood: 0.80, 0.72, 2.86, 0.28, 0.78, 0.74 and 11.06, respectively. CONCLUSIONS: As almost 20% of non-tuberculosis patients would be erroneously treated for tuberculosis and 25% of patients with tuberculous pleurisy would be missed, pleural fluid IGRA are not useful for the clinical diagnosis of tuberculous pleurisy.


Subject(s)
Interferon-gamma/immunology , T-Lymphocytes/immunology , Tuberculosis, Pleural/diagnosis , Humans , Interferon-gamma/blood , Pleural Effusion/blood , Pleural Effusion/diagnosis , ROC Curve , Sensitivity and Specificity , Tuberculosis, Pleural/blood
13.
Zhonghua Jie He He Hu Xi Za Zhi ; 33(1): 33-6, 2010 Jan.
Article in Zh | MEDLINE | ID: mdl-20368022

ABSTRACT

OBJECTIVE: to investigate the levels of peripheral CD(8)(+)CD(25)(+)Foxp(3)(+) regulatory T cells (CD(8)(+)CD(25)(+)Treg) in stable chronic obstructive pulmonary disease (COPD) patients, and the effect of muscarinic cholinergic receptor antagonist, tiotropium bromide, on the expression of CD(8)(+)CD(25)(+)Treg. METHODS: from Oct. 2007 to Mar. 2008, 23 patients with stable moderate to severe COPD received tiotropium bromide inhalation (18 microg daily) for 3 months. Before and after the use of tiotropium bromide, lung function was performed and peripheral vein blood samples were collected from the patient. Lymphocytes were isolated by three-color labeled monoclonal antibodies flow cytometry to detect the quantity and percentage of CD(8)(+)T cell, CD(8)(+)CD(25)(+) T cell, CD(8)(+)CD(25)(+)Treg, CD(4)(+)T cell, CD(4)(+)CD(25)(+) T cell and CD(4)(+)CD(25)(+)Treg, respectively. Paired t test was used for comparison between data before and after treatment. RESULTS: in patients with stable COPD, after tiotropium bromide treatment, the percentage of CD(4)(+) T cells was increased from (28 +/- 10)% to (36 +/- 6)%, and the difference was significant (t = 3.20, P < 0.01). CD(4)(+)CD(25)(+) was decreased from (10 +/- 7)% to (4 +/- 3)% (t = 3.78, P < 0.01), and CD(8)(+)CD(25)(+)Treg was increased from (8 +/- 8)% to (21 +/- 21)% (t = 2.72, P < 0.05). At baseline, CD(8)(+) cells, CD(8)(+)CD(25)(+) cells and CD(4)(+)CD(25)(+)Treg were detectable in the peripheral blood, but no significant changes were observed after treatment. After treatment with tiotropium bromide, the lung function was markedly improved; FEV(1), FEV(1)/Pre%, and FEV(1)/FVC were increased from (1.0 +/- 0.3) L, (35 +/- 10)% and (41 +/- 8)% to (1.1 +/- 0.3) L, (40 +/- 11)% and (45 +/- 11)%, respectively (t = 2.65, 2.56 and 2.37, respectively, all P < 0.05). By linear correlation analysis, the quantity of changes of CD(4)(+) T cells and CD(4)(+)CD(25)(+) T cells were negatively correlated with the rate of change in CD(8)(+)CD(25)(+)Treg (r = -0.61, P < 0.05 and r = -0.72, P < 0.01, respectively). CONCLUSIONS: in the peripheral blood of patients with stable COPD, there was expression of CD(8)(+)CD(25)(+)Treg and CD(4)(+)CD(25)(+)Treg. The muscarinic receptor antagonist tiotropium bromide, promoted the amplification of CD(4)(+), inhibited the expression of CD(25)(+), and enhanced the expression of CD(8)(+)Treg, suggesting that muscarinic receptor antagonist tiotropium bromide may possess immunomodulation function.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Muscarinic Antagonists/therapeutic use , Pulmonary Disease, Chronic Obstructive/metabolism , Scopolamine Derivatives/therapeutic use , T-Lymphocytes, Regulatory/metabolism , Aged , Bronchodilator Agents/therapeutic use , Female , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/drug therapy , Receptors, Muscarinic/metabolism , Tiotropium Bromide
14.
Biochim Biophys Acta Mol Cell Res ; 1867(11): 118806, 2020 11.
Article in English | MEDLINE | ID: mdl-32739525

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a specific form of chronic, progressive and fibrosing interstitial pneumonia of unknown cause. The main feature of IPF is a heterogeneous appearance with areas of sub-pleural fibrosis. However, the mechanism of sub-pleural fibrosis was poorly understood. In this study, our in vivo study revealed that pleural mesothelial cells (PMCs) migrated into lung parenchyma and localized alongside lung fibroblasts in sub-pleural area in mouse pulmonary fibrosis. Our in vitro study displayed that cultured-PMCs-medium induced lung fibroblasts transforming into myofibroblast, cultured-fibroblasts-medium promoted mesothelial-mesenchymal transition of PMCs. Furthermore, these changes in lung fibroblasts and PMCs were prevented by blocking TGF-ß1/Smad2/3 signaling with SB431542. TGF-ß1 neutralized antibody attenuated bleomycin-induced pulmonary fibrosis. Similar to TGF-ß1/Smad2/3 signaling, wnt/ß-catenin signaling was also activated in the process of PMCs crosstalk with lung fibroblasts. Moreover, inhibition of CD147 attenuated cultured-PMCs-medium induced collagen-I synthesis in lung fibroblasts. Blocking CD147 signaling also prevented bleomycin-induced pulmonary fibrosis. Our data indicated that crosstalk between PMC and lung fibroblast contributed to sub-pleural pulmonary fibrosis. TGF-ß1, Wnt/ß-catenin and CD147 signaling was involved in the underling mechanism.


Subject(s)
Epithelium/drug effects , Lung/metabolism , Pleura/drug effects , Pulmonary Fibrosis/genetics , Animals , Benzamides/pharmacology , Cell Movement/genetics , Dioxoles/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelium/pathology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Humans , Lung/drug effects , Lung/pathology , Mice , Pleura/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Smad2 Protein/genetics , Transforming Growth Factor beta1/genetics
15.
Zhonghua Jie He He Hu Xi Za Zhi ; 32(5): 335-41, 2009 May.
Article in Zh | MEDLINE | ID: mdl-19799066

ABSTRACT

OBJECTIVE: To investigate the relationship between clinical features of patients with A/H5N1 infection and their prognosis in mainland China. METHODS: This study included 28 human cases with A/H5N1 infection in mainland China from October 2005 to May 2008. Data were collected and reviewed from hospital medical records and publishied papers. A database was built by EPIDATA 3.02 and statistical analyses were performed with SPSS 13.0. RESULTS: The median age of the 28 cases was 29 years (range 6-62), and 15 were females. Ten patients survived, and 18 died. The typically clinical manifestations of human influenza A/H5N1 infection included fever and lower respiratory infection. The numbers of peripheral white blood cells, lymphocytes and platelets in the survival and non-survival groups were (4.01 +/- 1.86) x 10(9)/L vs (5.1 +/- 2.9) x 10(9)/L, (1.09 +/- 0.49) x 10(9)/L vs (0.98 +/- 0.44) x 10(9)/L, and (116 +/- 39) x 10(9)/L vs (101 +/- 40) x 10(9)/L, respectively; the differences were not statistically significant between the 2 groups (P>0.05). There was also no statistically significant difference in the increased serum enzymes, such as aspartate aminotransferase [(173 +/- 246) U/L vs (272 +/- 263) U/L], lactate dehydrogenase [(1016 +/- 568) U/L vs (1512 +/- 1052) U/L], creatine kinase [(1099 +/- 1590) U/L vs (2534 +/- 4281) U/L] and MB isoenzyme of creatine kinase [(28 +/- 30) U/L vs (125 +/- 197) U/L] (P>0.05) between the survival and the non-survival groups. However, there was a statistically significant difference in the number of patients with an initial LDH level more than 8 fold of the normal value between the survival and the non-survival groups (none vs 6, P<0.05). All of the 28 cases developed bilateral multiple infiltrates and consolidation in chest radiographs. Acute respiratory distress syndrome occurred in 22 cases, 17 of them died. All the 9 patients with acute kidney injury died. Ten patients received antiviral treatment with oseltamivir, and 6 of them survived. There was a statistical difference in the time of initiating oseltamivir treatment between the survival and the non-survival cases [(6.5 +/- 3.0) d vs (11.8 +/- 3.3) d, Z = 3.70, P<0.05]. Broad spectrum antibiotics and corticosteroids were administered in all of the 28 cases. There was no statistical difference between the survival and the non-survival groups regarding to the corticosteroid treatment (P>0.05). CONCLUSIONS: Initial LDH level reaching more than 8 fold of the normal value suggests a poor prognosis for human H5N1 infection. Patients complicated with either ARDS or acute kidney injury had a higher risk of death. Early administration of effective antiviral agents might improve the prognosis and decrease case fatality.


Subject(s)
Influenza, Human/epidemiology , Influenza, Human/therapy , Acute Kidney Injury/complications , Adolescent , Adult , Child , China/epidemiology , Female , Humans , Influenza A Virus, H5N1 Subtype , Influenza, Human/complications , Influenza, Human/diagnosis , Influenza, Human/enzymology , L-Lactate Dehydrogenase/analysis , Male , Middle Aged , Prognosis , Respiratory Distress Syndrome/complications , Treatment Outcome , Young Adult
16.
Toxicol Lett ; 303: 1-8, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30572104

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease that typically leads to respiratory failure and death. The cause of IPF is poorly understood. Although several environmental and occupational factors are considered as risk factors in IPF, cigarette smoking seems to be the most strongly associated risk factor. Here firstly, we treated mice with cigarette (16 mg tar, 1.0 mg nicotine in each cigarette) smoking and tried to explore the role of cigarette smoking in pulmonary fibrosis. Mice were continuously subjected to smoke for about 1 h each day (12 cigarettes per day, 5 days per week) during 40 days. Bleomycin was administrated by intraperitoneal injection at a dose of 40 mg/kg on days 1, 5, 8, 11 and 15. We found bleomycin induced pulmonary fibrosis in mice, and cigarette smoking augmented bleomycin-induced fibrosis reflected by both in fibrotic area and percentages of collagen in the lungs. Then we prepared and employed cigarette smoke extract (CSE) in cell models and found that CSE could induce the activation of p-Smad2/3 and p-Akt, as well as collagen-I synthesis and cell proliferation in lung fibroblasts and pleural mesothelial cells (PMCs). TGF-ß1 signaling mediated CSE-induced PMCs migration. Moreover, in vitro studies revealed that CSE had superimposed effect on bleomycin-induced activation of TGF-ß-Smad2/3 and -Akt signaling. TGF-ß-Smad2/3 and -Akt signaling were further augmented by cigarette smoking in the lung of bleomycin-treated mice. Taken together, these findings represent the first evidence that cigarette smoking aggravated bleomycin-induced pulmonary fibrosis via TGF-ß1 signaling.


Subject(s)
Bleomycin/toxicity , Cigarette Smoking/adverse effects , Idiopathic Pulmonary Fibrosis/pathology , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/metabolism , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/cytology , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Risk Factors , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
17.
Zhonghua Jie He He Hu Xi Za Zhi ; 31(12): 902-7, 2008 Dec.
Article in Zh | MEDLINE | ID: mdl-19134406

ABSTRACT

OBJECTIVE: To explore the anti-tumor immune responses of dendritic cells (DCs) loaded with intact wild-type p53 to mice challenged with tumor cells expressing p53 genes with mutations at different sites. METHODS: Ad-p53-DC immunization function was assessed by the expression of surface molecules and allogeneic MLR. DCs derived from bone marrow were transduced with adenovirus or a human wild-type p53 containing recombinant adenovirus (Ad-DC and Ad-p53-DC) and immunized C57BL/6 mice. Splenocytes were separated and cell cytotoxicity was measured against tumor cells expressing mutant p53 (MethA, D459 and P815) in a standard 6-h(51)Cr-release assay. Effector and target cells were incubated in the presence of anti-CD(4) or anti-CD(8) antibody. Ad-p53-DC was immunized in control Ad-DC before or after mice were challenged with either D459 tumor or with MethA sarcoma cells to observe whether immune response would provide tumor protection. RESULTS: Immunization with Ad-p53-DC developed significantly higher substantial CTL responses against Ad-p53-P815, D459 and MethA cells (effectors: target cells = 50:1), (27.8 +/- 3.4)%, (23.5 +/- 2.7)%, (58.3 +/- 9.2)% than with Ad-DC (9.3 +/- 1.8)%, (4.6 +/- 1.0)%, (23.5 +/- 3.7)% (t(d) = 5.79, 3.68, 5.02, all P < 0.05). In Ad-p53-DC immunized mice, anti-CD(8) antibody blocked the cytotoxicity against Ad-p53-P815 (26.7 +/- 2.8)% or D459 (6.1 +/- 1.2)%, but not anti-CD(4) antibody [(59.8 +/- 4.6)%, (18.9 +/- 2.4)%, t(d) = 8.79 or 9.18, all P < 0.05]. Ad-p53-DC immunization provided complete tumor protection in 80% of mice challenged with D459 and in 70% of mice challenged with MethA, while none protected in Ad-DC immunization group (chi(2) = 6.72, 5.86, all P < 0.05). Treated with Ad-p53-DC after D459 inoculation subcutaneously, mice were killed due to the bulky tumor more than 2 weeks later than the mice in the Ad-DC treatment group during 7 week observation (chi(2) = 9.48, P < 0.05). CONCLUSION: DCs transfected with 100 MOI Ad-p53 induced intense CTL responses against P815, D459 and MethA. This CTL response is mediated by CD(8)(+) T cells. Treatment with Ad-p53-DC significantly developed tumor immunology and slowed the growth of established tumors.


Subject(s)
Dendritic Cells/immunology , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Animals , Bone Marrow Cells , Cell Line, Tumor , Cytotoxicity, Immunologic , Genetic Vectors , Immunotherapy , Mice , Mice, Inbred C57BL , Transfection
18.
PLoS One ; 11(1): e0147232, 2016.
Article in English | MEDLINE | ID: mdl-26808506

ABSTRACT

BACKGROUND: CD8+ T cells (Cytotoxic T cells, Tc) are known to play a critical role in the pathogenesis of smoking related airway inflammation including chronic obstructive pulmonary disease (COPD). However, how cigarette smoke directly impacts systematic CD8+ T cell and regulatory T cell (Treg) subsets, especially by modulating muscarinic acetylcholine receptors (MRs), has yet to be well elucidated. METHODS: Circulating CD8+ Tc/Tregs in healthy nonsmokers (n = 15), healthy smokers (n = 15) and COPD patients (n = 18) were evaluated by flow cytometry after incubating with anti-CD3, anti-CD8, anti-CD25, anti-Foxp3 antibodies. Peripheral blood T cells (PBT cells) from healthy nonsmokers were cultured in the presence of cigarette smoke extract (CSE) alone or combined with MRs agonist/antagonist for 5 days. Proliferation and apoptosis were evaluated by flow cytometry using Ki-67/Annexin-V antibodies to measure the effects of CSE on the survival of CD8+ Tc/Tregs. RESULTS: While COPD patients have elevated circulating percentage of CD8+ T cells, healthy smokers have higher frequency of CD8+ Tregs. Elevated percentages of CD8+ T cells correlated inversely with declined FEV1 in COPD. CSE promoted the proliferation and inhibited the apoptosis of CD8+ T cells, while facilitated both the proliferation and apoptosis of CD8+ Tregs. Notably, the effects of CSE on CD8+ Tc/Tregs can be mostly simulated or attenuated by muscarine and atropine, the MR agonist and antagonist, respectively. However, neither muscarine nor atropine influenced the apoptosis of CD8+ Tregs. CONCLUSION: The results imply that cigarette smoking likely facilitates a proinflammatory state in smokers, which is partially mediated by MR dysfunction. The MR antagonist may be a beneficial drug candidate for cigarette smoke-induced chronic airway inflammation.


Subject(s)
Inflammation/etiology , Nicotiana , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, Muscarinic/physiology , Smoke/adverse effects , Smoking/immunology , T-Lymphocytes, Regulatory/drug effects , Tobacco Products , Apoptosis/drug effects , Atropine/pharmacology , Cell Division/drug effects , Female , Forced Expiratory Volume , Humans , Lymphocyte Activation/drug effects , Lymphocyte Count , Male , Middle Aged , Muscarine/pharmacology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology
19.
PeerJ ; 4: e2301, 2016.
Article in English | MEDLINE | ID: mdl-27547589

ABSTRACT

Background. CD8(+) T lymphocytes are known to play a critical role in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, systematic analyses of CD8(+) T cell (Cytotoxic T cells, Tc) subsets in COPD patients have yet to be well conducted. Methods. The whole Tc subsets, including Tc1/2/10/17, CD8(+) regulatory T cells (Tregs) and CD8(+) α7(+) T cells, were quantified by flow cytometry in peripheral blood from 24 stable COPD subjects (SCOPD), 14 patients during acute exacerbations (AECOPD), and 14 healthy nonsmokers (HN). Results. Acute exacerbations of COPD were accompanied by elevated levels of circulating CD8(+) T cells. Tc1 cells were increased in both SCOPD and AECOPD patients, whereas the percentage of Tc2 cells was decreased in SCOPD patients but remained normal in AECOPD patients. Tc17 cells were increased only in AECOPD patients, and the percentage of Tc10 cells was reduced in both SCOPD and AECOPD patients. The imbalances of pro/anti-inflammatory Tc subsets observed in COPD may be caused by the lack of Tc10 cells and the impaired anti-inflammatory capacity of CD8(+) Tregs. Conclusions. The imbalances between subsets of CD8(+) peripheral blood T cells contribute to the immune response dysfunction in COPD pathogenesis.

20.
Sheng Li Xue Bao ; 57(4): 493-7, 2005 Aug 25.
Article in Zh | MEDLINE | ID: mdl-16094498

ABSTRACT

To investigate the role of found in inflammatory zone 1 (FIZZ1) protein in the pathogenesis of experimental pulmonary fibrosis, 48 male Sprague-Dawley rats were randomly divided into two groups, the pulmonary fibrosis group and the control group. Rat pulmonary fibrosis was reproduced by an intratracheal injection of bleomycin (5 mg/kg body weight). Normal saline (1 ml/kg body weight) was given intratracheally injection in the control group. There were 24 rats in each group, and 6 animals were separately killed on the 7th, 14th, 21th and 28th day after treated with bleomycin or normal saline. Then the following tests were undertaken: (1) HE and Masson staining of lung section;(2) Determination of lung tissue hydroxyproline (HYP);(3) Immunohistochemical staining of protein of FIZZ1 in the lung;(4) In situ hybridization of FIZZ1 mRNA in the lung. The results showed: (1) There were full of inflammatory cells in the lung, the interval of alveoli enlarged and many alveolar spaces disappeared on the 7th day after treated with bleomycin in the fibrosis group. Collagen began to proliferate after 14 d. The pulmonary fibrosis was stably established on the 28th day, full of green fibers in the Masson staining of lung section. (2) The expression of FIZZ1 protein in the lung increased after 7 d in bleomycin-treated animals (3.013+/-0.326 vs 0.473+/-0.056, P<0.01 vs control), but was slightly decreased on the 14th day (2.124+/-0.197) and expensively decreased on the 21st day (1.760+/-0.105) and the 28th day (0.691+/-0.081). (3) The expression of FIZZ1 mRNA in the lung also increased after 7 d by treated with bleomycin (3.795+/-0.338 vs 0.678+/-0.087, P<0.01 vs control), but decreased on the 14th day (1.276+/-0.104) and further decreased on the 28th day (0.896+/-0.084). The expression of FIZZ1 protein and mRNA in fibrosis group was higher than that in the control group (P<0.05 or P<0.01). The results suggest that FIZZ1 protein and FIZZ1 mRNA are dynamically changed in the lung with experimental pulmonary fibrosis, which may contribute to the pathogenesis of pulmonary fibrosis.

SELECTION OF CITATIONS
SEARCH DETAIL