Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
Add more filters

Affiliation country
Publication year range
1.
Am J Pathol ; 193(6): 740-754, 2023 06.
Article in English | MEDLINE | ID: mdl-36965776

ABSTRACT

Acute lung injury (ALI) is a clinical syndrome characterized by a diffuse lung inflammation that commonly evolves into acute respiratory distress syndrome and respiratory failure. The lung microbiota is involved in the pathogenesis of ALI. Corisin, a proapoptotic peptide derived from the lung microbiota, plays a role in ALI and acute exacerbation of pulmonary fibrosis. Preventive therapeutic intervention with a monoclonal anticorisin antibody inhibits ALI in mice. However, whether inhibition of corisin with the antibody ameliorates established ALI is unknown. Here, the therapeutic effectiveness of the anticorisin antibody in already established ALI in mice was assessed. Lipopolysaccharide was used to induce ALI in mice. After causing ALI, the mice were treated with a neutralizing anticorisin antibody. Mice treated with the antibody showed significant improvement in lung radiological and histopathologic findings, decreased lung infiltration of inflammatory cells, reduced markers of lung tissue damage, and inflammatory cytokines in bronchoalveolar lavage fluid compared with untreated mice. In addition, the mice treated with anticorisin antibody showed significantly increased expression of antiapoptotic proteins with decreased caspase-3 activation in the lungs compared with control mice treated with an irrelevant antibody. In conclusion, these observations suggest that the inhibition of corisin is a novel and promising approach for treating established ALI.


Subject(s)
Acute Lung Injury , Pneumonia , Mice , Animals , Lung/pathology , Acute Lung Injury/pathology , Bronchoalveolar Lavage Fluid , Pneumonia/metabolism , Peptides/pharmacology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL
2.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38891998

ABSTRACT

Approximately 30% of steroid-resistant nephrotic syndromes are attributed to monogenic disorders that involve 27 genes. Mutations in KANK family members have also been linked to nephrotic syndrome; however, the precise mechanism remains elusive. To investigate this, podocyte-specific Kank1 knockout mice were generated to examine phenotypic changes. In the initial assessment under normal conditions, Kank1 knockout mice showed no significant differences in the urinary albumin-creatinine ratio, blood urea nitrogen, serum creatinine levels, or histological features compared to controls. However, following kidney injury with adriamycin, podocyte-specific Kank1 knockout mice exhibited a significantly higher albumin-creatinine ratio and a significantly greater sclerotic index than control mice. Electron microscopy revealed more extensive foot process effacement in the knockout mice than in control mice. In addition, KANK1-deficient human podocytes showed increased detachment and apoptosis following adriamycin exposure. These findings suggest that KANK1 may play a protective role in mitigating podocyte damage under pathological conditions.


Subject(s)
Cytoskeletal Proteins , Doxorubicin , Mice, Knockout , Podocytes , Animals , Humans , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/pathology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
3.
Int J Mol Sci ; 24(7)2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37047672

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive and fatal disease with a poor prognosis. Matrix metalloproteinase-2 is involved in the pathogenesis of organ fibrosis. The role of matrix metalloproteinase-2 in lung fibrosis is unclear. This study evaluated whether overexpression of matrix metalloproteinase-2 affects the development of pulmonary fibrosis. Lung fibrosis was induced by bleomycin in wild-type mice and transgenic mice overexpressing human matrix metalloproteinase-2. Mice expressing human matrix metalloproteinase-2 showed significantly decreased infiltration of inflammatory cells and inflammatory and fibrotic cytokines in the lungs compared to wild-type mice after induction of lung injury and fibrosis with bleomycin. The computed tomography score, Ashcroft score of fibrosis, and lung collagen deposition were significantly reduced in human matrix metalloproteinase transgenic mice compared to wild-type mice. The expression of anti-apoptotic genes was significantly increased, while caspase-3 activity was significantly reduced in the lungs of matrix metalloproteinase-2 transgenic mice compared to wild-type mice. Active matrix metalloproteinase-2 significantly decreased bleomycin-induced apoptosis in alveolar epithelial cells. Matrix metalloproteinase-2 appears to protect against pulmonary fibrosis by inhibiting apoptosis of lung epithelial cells.


Subject(s)
Idiopathic Pulmonary Fibrosis , Matrix Metalloproteinase 2 , Mice , Humans , Animals , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Lung/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Bleomycin/adverse effects , Mice, Transgenic , Fibrosis , Mice, Inbred C57BL
4.
Int J Mol Sci ; 23(22)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36430743

ABSTRACT

Diabetes mellitus is a global health problem. Diabetic nephropathy is a common complication of diabetes mellitus and the leading cause of end-stage renal disease. The clinical course, response to therapy, and prognosis of nephropathy are worse in diabetic than in non-diabetic patients. The role of transforming growth factorß1 in kidney fibrosis is undebatable. This study assessed whether the overexpression of transforming growth factorß1 is associated with insulin resistance and the rapid progression of transforming growth factorß1-mediated nephropathy under diabetic conditions. Diabetes mellitus was induced with streptozotocin in wild-type mice and transgenic mice with the kidney-specific overexpression of human transforming growth factorß1. Mice treated with saline were the controls. Glucose tolerance and kidney fibrosis were evaluated. The blood glucose levels, the values of the homeostasis model assessment for insulin resistance, and the area of kidney fibrosis were significantly increased, and the renal function was significantly impaired in the diabetic transforming growth factorß1 transgenic mice compared to the non-diabetic transgenic mice, diabetic wild-type mice, and non-diabetic mice. Transforming growth factorß1 impaired the regulatory effect of insulin on glucose in the hepatocyte and skeletal muscle cell lines. This study shows that transforming growth factorß1 overexpression is associated with insulin resistance and rapidly progressive kidney fibrosis under diabetic conditions in mice.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Insulin Resistance , Humans , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/drug therapy , Fibrosis , Kidney/metabolism , Mice, Transgenic , Glucose/metabolism , Diabetes Mellitus/pathology
5.
Molecules ; 27(24)2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36558053

ABSTRACT

Two compounds 1 and 2 were isolated from the culture broth of Lepista luscina. This is the first time that compound 1 was isolated from a natural source. The structure of compound 1 was identified via 1D and 2D NMR and HRESIMS data. Compounds 1 and 2 along with 8-nitrotryptanthrin (4) were evaluated for their biological activities using the A549 lung cancer cell line. As a result, 1 and 2 inhibited the expression of Axl and immune checkpoint molecules. In addition, compounds 1, 2 and 4 were tested for HIF inhibitory activity. Compound 2 demonstrated statistically significant HIF inhibitory effects on NIH3T3 cells and 1 and 2 against ARPE19 cells.


Subject(s)
Immune Checkpoint Proteins , Lung Neoplasms , Animals , Mice , Humans , NIH 3T3 Cells , Lung Neoplasms/metabolism , A549 Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Cell Line, Tumor
6.
Respirology ; 26(4): 342-351, 2021 04.
Article in English | MEDLINE | ID: mdl-33164314

ABSTRACT

BACKGROUND AND OBJECTIVE: Activation of the blood coagulation system is a common observation in inflammatory diseases. The role of coagulation in COPD is underexplored. METHODS: The study included 413 COPD patients and 49 controls from the 3-year Bergen COPD Cohort Study (BCCS). One hundred and forty-eight COPD patients were also examined during AECOPD. The plasma markers of coagulation activation, TAT complex, APC-PCI complex and D-dimer, were measured at baseline and during exacerbations by enzyme immunoassays. Differences in levels of the markers between stable COPD patients and controls, and between stable COPD and AECOPD were examined. The associations between coagulation markers and later AECOPD and mortality were examined by negative binomial and Cox regression analyses. RESULTS: TAT was significantly lower in stable COPD (1.03 ng/mL (0.76-1.44)) than in controls (1.28 (1.04-1.49), P = 0.002). During AECOPD, all markers were higher than in the stable state: TAT 2.56 versus 1.43 ng/mL, APC-PCI 489.3 versus 416.4 ng/mL and D-dimer 763.5 versus 479.7 ng/mL (P < 0.001 for all). Higher D-dimer in stable COPD predicted a higher mortality (HR: 1.60 (1.24-2.05), P < 0.001). Higher TAT was associated with both an increased risk of later exacerbations, with a yearly incidence rate ratio of 1.19 (1.04-1.37), and a faster time to the first exacerbation (HR: 1.25 (1.10-1.42), P = 0.001, all after adjustment). CONCLUSION: Activation of the coagulation system is increased during COPD exacerbations. Coagulation markers are potential predictors of later COPD exacerbations and mortality.


Subject(s)
Percutaneous Coronary Intervention , Pulmonary Disease, Chronic Obstructive , Blood Coagulation , Cohort Studies , Disease Progression , Humans
7.
Kidney Int ; 98(5): 1179-1192, 2020 11.
Article in English | MEDLINE | ID: mdl-33069430

ABSTRACT

Kidney fibrosis is the common consequence of chronic kidney diseases that inexorably progresses to end-stage kidney disease with organ failure treatable only with replacement therapy. Since transforming growth factor-ß1 is the main player in the pathogenesis of kidney fibrosis, we posed the hypothesis that recombinant thrombomodulin can ameliorate transforming growth factor-ß1-mediated progressive kidney fibrosis and failure. To interrogate our hypothesis, we generated a novel glomerulus-specific human transforming growth factor-ß1 transgenic mouse to evaluate the therapeutic effect of recombinant thrombomodulin. This transgenic mouse developed progressive glomerular sclerosis and tubulointerstitial fibrosis with kidney failure. Therapy with recombinant thrombomodulin for four weeks significantly inhibited kidney fibrosis and improved organ function compared to untreated transgenic mice. Treatment with recombinant thrombomodulin significantly inhibited apoptosis and mesenchymal differentiation of podocytes by interacting with the G-protein coupled receptor 15 to activate the Akt signaling pathway and to upregulate the expression of anti-apoptotic proteins including survivin. Thus, our study strongly suggests the potential therapeutic efficacy of recombinant thrombomodulin for the treatment of chronic kidney disease and subsequent organ failure.


Subject(s)
Renal Insufficiency, Chronic , Transforming Growth Factor beta1 , Fibrosis , GTP-Binding Proteins , Humans , Kidney/pathology , Proto-Oncogene Proteins c-akt , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Signal Transduction , Thrombomodulin/genetics , Transforming Growth Factor beta1/metabolism
8.
Allergy ; 75(9): 2267-2278, 2020 09.
Article in English | MEDLINE | ID: mdl-32145080

ABSTRACT

BACKGROUND: Bronchial asthma is a chronic disease characterized by inflammation, obstruction, and hyperresponsiveness of the airways. There is currently no curative therapy for asthma. Type 2 helper T cell response plays a critical role in the pathogenesis of the disease. Protein S is a glycoprotein endowed with anticoagulant, anti-inflammatory, and anti-apoptotic properties. Whether protein S can suppress bronchial asthma and be useful for its therapy is unknown. METHODS: To address this question here we compared the development of allergen-associated bronchial asthma between wild type and protein S-overexpressing transgenic mice. Mice were sensitized and challenged with ovalbumin. We also evaluated the circulating levels of total and active protein S in patients with bronchial asthma and healthy controls. RESULTS: The circulating level of total protein S and of its active form was significantly decreased in patients with bronchial asthma compared to controls. Allergic protein S transgenic mice showed a significant reduction of airway hyperresponsiveness, lung tissue inflammatory cell infiltration, lung levels of Th2 cytokines and IgE compared to their wild-type counterparts. Administration of exogenous human protein S also decreased airway hyperresponsiveness and Th2-mediated lung inflammation in allergic wild-type mice compared with their untreated mouse counterparts. Human protein S significantly shifted the Th1/Th2 balance to Th1 and promoted the secretion of Th1 cytokines (IL-12, tumor necrosis factor-α) from dendritic cells. CONCLUSIONS: These observations suggest the strong protective activity of protein S against the development of allergic bronchial asthma implicating its potential usefulness for the disease treatment.


Subject(s)
Asthma , Bronchial Hyperreactivity , Animals , Asthma/prevention & control , Cytokines , Disease Models, Animal , Humans , Immunoglobulin E , Lung , Mice , Mice, Inbred BALB C , Ovalbumin , Protein S , Th2 Cells
9.
Biosci Biotechnol Biochem ; 84(7): 1332-1338, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32200702

ABSTRACT

A novel compound, (R)-4-ethoxy-2-hydroxy-4-oxobutanoic acid (1), and six known compounds (2-7) were isolated from the fruiting bodies of the wild edible mushroom Leucopaxillus giganteus. The planar structure of 1 was determined by the interpretation of spectroscopic data analysis. The absolute configuration of 1 was determined by comparing specific rotation of the synthetic compounds. In the plant regulatory assay, the isolated compounds (1-7) and the chemically prepared compounds (8-10) were evaluated their biological activity against the lettuce (Lactuca sativa) growth. Compounds 1 and 3-10 showed the significant regulatory activity of lettuce growth. 1 showed the strongest inhibition activity among the all the compounds tested. In the lung cancer assay, all the compounds were assessed the mRNA expression of Axl and immune checkpoints (PD-L1, PD-L2) in the human A549 alveolar epithelial cell line by RT-PCR. Compounds 1-10 showed significant inhibition activity against Axl and/or immune checkpoint.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Agaricales/chemistry , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/metabolism , Plant Growth Regulators/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Fruiting Bodies, Fungal/chemistry , Humans , Lactuca/drug effects , Lactuca/growth & development , Lung Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
10.
Am J Pathol ; 188(5): 1195-1203, 2018 05.
Article in English | MEDLINE | ID: mdl-29454753

ABSTRACT

Protein S is a vitamin K-dependent glycoprotein produced mainly in the liver with anticoagulant, anti-inflammatory, immune-modulatory, and antiapoptotic properties. Protein S exacerbates acute liver injury by prolonging the survival of liver immune cells. However, the effect of protein S on chronic liver injury and fibrosis is unknown. Here, we investigated whether human protein S can affect chronic liver injury and fibrosis. Liver injury/fibrosis was induced by carbon tetrachloride injection in mice overexpressing human protein S and in wild-type mice. Human protein S transgenic mice receiving carbon tetrachloride showed significantly higher circulating levels of liver transaminases, increased liver expression of inflammatory cytokines, significantly more extended liver fibrosis, and areas with DNA breakage after chronic injury compared with wild-type mice. Wild-type mice infused with exogenous human protein S exhibited exacerbated liver injury and increased number of hepatic stellate cells compared with untreated mice. Human protein S inhibited apoptosis and increased Akt pathway activation in hepatic stellate cells. The antiapoptotic activity of protein S may play a role in chronic liver injury and subsequent liver fibrosis.


Subject(s)
End Stage Liver Disease/metabolism , Hepatic Stellate Cells/metabolism , Liver/metabolism , Protein S/metabolism , Animals , Apoptosis/physiology , Carbon Tetrachloride , End Stage Liver Disease/chemically induced , End Stage Liver Disease/pathology , Fibrosis/metabolism , Fibrosis/pathology , Hepatic Stellate Cells/pathology , Liver/pathology , Mice , Mice, Transgenic , Protein S/genetics , Signal Transduction/physiology
11.
Int J Mol Sci ; 20(5)2019 Mar 02.
Article in English | MEDLINE | ID: mdl-30832349

ABSTRACT

Acute lung injury is a fatal disease characterized by inflammatory cell infiltration, alveolar-capillary barrier disruption, protein-rich edema, and impairment of gas exchange. Protein S is a vitamin K-dependent glycoprotein that exerts anticoagulant, immunomodulatory, anti-inflammatory, anti-apoptotic, and neuroprotective effects. The aim of this study was to evaluate whether human protein S inhibits cell apoptosis in acute lung injury. Acute lung injury in human protein S transgenic and wild-type mice was induced by intratracheal instillation of lipopolysaccharide. The effect of human protein S on apoptosis of lung tissue cells was evaluated by Western blotting. Inflammatory cell infiltration, alveolar wall thickening, myeloperoxidase activity, and the expression of inflammatory cytokines were reduced in human protein S transgenic mice compared to the wild-type mice after lipopolysaccharide instillation. Apoptotic cells and caspase-3 activity were reduced while phosphorylation of extracellular signal-regulated kinase was enhanced in the lung tissue from human protein S transgenic mice compared to wild-type mice after lipopolysaccharide instillation. The results of this study suggest that human protein S is protective in lipopolysaccharide-induced acute lung injury by inhibiting apoptosis of lung cells.


Subject(s)
Acute Lung Injury/metabolism , Apoptosis , Protein S/metabolism , Acute Lung Injury/etiology , Animals , Humans , Lipopolysaccharides/toxicity , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Protein S/genetics
12.
Biochem Biophys Res Commun ; 497(1): 332-338, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29428733

ABSTRACT

Chronic obstructive pulmonary disease is the major growing cause of mortality and morbidity worldwide, and it is going to become the third most common cause of death by 2020. Chronic obstructive pulmonary disease is pathologically characterized by lung emphysema and small airway inflammation. Animal models are very important to get insights into the disease pathogenesis but current models of chronic obstructive pulmonary disease take a long time to develop. The need of a new model is compelling. In the present study we focus on the role of matrix metalloproteinases in the pathogenesis of chronic obstructive pulmonary disease and hypothesized that lung overexpression of latent matrix metalloproteinases-2 would allow the development of emphysema after short-term exposure to cigarette smoke extract inhalation. Human latent matrix metalloproteinases-2 transgenic mouse expressing high level of the protein in the lungs and wild type mouse were exposed to aerosolized cigarette smoke extract for two weeks. Transgenic mice showed significant lung emphysematous changes, increased infiltration of inflammatory cells and enhanced lung concentrations of inflammatory cytokines in the lungs compared to their wild type counterparts after inhalation of cigarette smoke extract. This novel mouse model will be a very useful tool for evaluating the mechanistic pathways and for development of novel therapies in cigarette smoke-associated lung emphysema.


Subject(s)
Environmental Exposure/adverse effects , Matrix Metalloproteinase 2/metabolism , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/etiology , Smoke/adverse effects , Tars/adverse effects , Tobacco Products/adverse effects , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation/drug effects
13.
Am J Pathol ; 187(10): 2312-2322, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28739343

ABSTRACT

Pulmonary fibrosis is the terminal stage of a group of idiopathic interstitial pneumonias, of which idiopathic pulmonary fibrosis is the most frequent and fatal form. Recent studies have shown that recombinant human thrombomodulin (rhTM) improves exacerbation and clinical outcome of idiopathic pulmonary fibrosis, but the mechanism remains unknown. This study evaluated the mechanistic pathways of the inhibitory activity of rhTM in pulmonary fibrosis. Transgenic mice overexpressing human transforming growth factor-ß1 that develop spontaneously pulmonary fibrosis, and wild-type mice treated with bleomycin were used as models of lung fibrosis. rhTM was administered to mice by i.p. injection or by the intranasal route. Therapy with rhTM significantly decreased the concentration of high mobility group box1, interferon-γ, and fibrinolytic markers, the expression of growth factors including transforming growth factor-ß1, and the degree of lung fibrosis. rhTM significantly suppressed apoptosis of lung epithelial cells in in vivo and in vitro experiments. The results of the present study demonstrated that rhTM can inhibit bleomycin-induced pulmonary fibrosis and transforming growth factor-ß1-driven exacerbation and progression of pulmonary fibrosis, and that apart from its well-recognized anticoagulant and anti-inflammatory properties, rhTM can also suppress apoptosis of lung epithelial cells.


Subject(s)
Apoptosis , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Thrombomodulin/therapeutic use , A549 Cells , Administration, Intranasal , Administration, Intravenous , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Disease Progression , Female , Humans , Injections, Intraperitoneal , Lung/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/complications , Pneumonia/drug therapy , Pneumonia/pathology , Pulmonary Fibrosis/complications , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombomodulin/administration & dosage , Thrombomodulin/blood , Transforming Growth Factor beta1/metabolism
14.
Int J Mol Sci ; 19(10)2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30257437

ABSTRACT

Crizotinib is highly effective against anaplastic lymphoma kinase-positive and c-ros oncogen1-positive non-small cell lung cancer. Renal dysfunction is associated with crizotinib therapy but the mechanism is unknown. Here, we report a case of anaplastic lymphoma kinase positive non-small cell lung cancer showing multiple cysts and dysfunction of the kidneys during crizotinib administration. We also present results demonstrating that long-term crizotinib treatment induces fibrosis and dysfunction of the kidneys by activating the tumor necrosis factor-α/nuclear factor-κB signaling pathway. In conclusion, this study shows the renal detrimental effects of crizotinib, suggesting the need of careful monitoring of renal function during crizotinib therapy.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Crizotinib/adverse effects , Kidney Diseases, Cystic/chemically induced , Kidney/drug effects , Lung Neoplasms/drug therapy , Aged , Animals , Antineoplastic Agents/therapeutic use , Crizotinib/therapeutic use , Female , Fibrosis , Humans , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/physiopathology , Mice
15.
Cytokine ; 90: 60-65, 2017 02.
Article in English | MEDLINE | ID: mdl-27776277

ABSTRACT

We investigated whether IL-33 is involved in mucus overproduction and goblet cell hyperplasia in eosinophilic chronic rhinosinusitis (ECRS). IL-33 mRNA was significantly higher in the eosinophilic CRS group than in the non-eosinophilic CRS group from human nasal polyps. IL-33 induced MUC5AC mRNA and MUC5AC protein, and also goblet cell hyperplasia at air liquid interface culture in human nasal epithelial cells. In addition to that, IL-33 induced MUC5B and FOXA3, and reduces FOXJmRNA. In conclusion, our present study demonstrated that the direct evidence of IL-33 which lead to increase mucin gene and protein expression, as well as goblet cell hyperplasia. This study provides novel insights into the role of IL-33 on mucus overproduction in eosinophilic inflammation of human airways.


Subject(s)
Gene Expression Regulation/immunology , Goblet Cells/immunology , Interleukin-33/immunology , Mucin 5AC/immunology , Mucin-5B/immunology , Rhinitis/immunology , Sinusitis/immunology , Female , Forkhead Transcription Factors/immunology , Goblet Cells/pathology , Hepatocyte Nuclear Factor 3-gamma/immunology , Humans , Hyperplasia , Male , Middle Aged
16.
Respir Res ; 18(1): 79, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28464879

ABSTRACT

BACKGROUND: Epithelial-mesenchymal transition is currently recognized as an important mechanism for the increased number of myofibroblasts in cancer and fibrotic diseases. We have already reported that epithelial-mesenchymal transition is involved in airway remodeling induced by eosinophils. Procaterol is a selective and full ß2 adrenergic agonist that is used as a rescue of asthmatic attack inhaler form and orally as a controller. In this study, we evaluated whether procaterol can suppress epithelial-mesenchymal transition of airway epithelial cells induced by eosinophils. METHODS: Epithelial-mesenchymal transition was assessed using a co-culture system of human bronchial epithelial cells and primary human eosinophils or an eosinophilic leukemia cell line. RESULTS: Procaterol significantly inhibited co-culture associated morphological changes of bronchial epithelial cells, decreased the expression of vimentin, and increased the expression of E-cadherin compared to control. Butoxamine, a specific ß2-adrenergic antagonist, significantly blocked changes induced by procaterol. In addition, procaterol inhibited the expression of adhesion molecules induced during the interaction between eosinophils and bronchial epithelial cells, suggesting the involvement of adhesion molecules in the process of epithelial-mesenchymal transition. Forskolin, a cyclic adenosine monophosphate-promoting agent, exhibits similar inhibitory activity of procaterol. CONCLUSIONS: Overall, these observations support the beneficial effect of procaterol on airway remodeling frequently associated with chronic obstructive pulmonary diseases.


Subject(s)
Eosinophils/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/physiology , Procaterol/administration & dosage , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Adrenergic beta-2 Receptor Agonists/administration & dosage , Bronchi/cytology , Bronchi/diagnostic imaging , Bronchi/physiology , Cell Line , Dose-Response Relationship, Drug , Eosinophils/cytology , Eosinophils/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Respiratory Mucosa/drug effects , Treatment Outcome
18.
Cell Immunol ; 293(1): 34-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25497974

ABSTRACT

Eosinophils and mast cells play critical roles in the pathogenesis of bronchial asthma. Activation of both cells leads to the release of pro-inflammatory mediators in the airway of asthmatic patients. Recently, we have shown that inhaled thrombomodulin inhibits allergic bronchial asthma in a mouse model. In the present study, we hypothesize that thrombomodulin can inhibit the activation of eosinophils and mast cells. The effect of thrombomodulin on the activation and release of inflammatory mediators from eosinophils and mast cells was evaluated. Thrombomodulin inhibited the eotaxin-induced chemotaxis, upregulation of CD11b and degranulation of eosinophils. Treatment with thrombomodulin also significantly suppressed the degranulation and synthesis of inflammatory cytokines and chemokines in eosinophils and mast cells. Mice treated with a low-dose of inhaled thrombomodulin have decreased number of eosinophils and activated mast cells and Th2 cytokines in the lungs compared to untreated mice. The results of this study suggest that thrombomodulin may modulate allergic responses by inhibiting the activation of both eosinophils and mast cells.


Subject(s)
Asthma/drug therapy , Eosinophils/drug effects , Mast Cells/drug effects , Thrombomodulin/administration & dosage , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Degranulation , Cell Line, Tumor , Chemokine CCL11/drug effects , Chemokine CCL11/metabolism , Chemotaxis/drug effects , Cytokines/biosynthesis , Cytokines/metabolism , Eosinophils/immunology , Eosinophils/pathology , Gene Expression , Humans , Mast Cells/immunology , Mast Cells/pathology , Mice , Ovalbumin , Primary Cell Culture , Recombinant Proteins/administration & dosage , Th1-Th2 Balance
19.
Biochem Biophys Res Commun ; 449(3): 351-6, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24845378

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a mechanism by which eosinophils can induce airway remodeling. Montelukast, an antagonist of the cysteinyl leukotriene receptor, can suppress airway remodeling in asthma. The purpose of this study was to evaluate whether montelukast can ameliorate airway remodeling by blocking EMT induced by eosinophils. EMT induced was assessed using a co-culture system of human bronchial epithelial cells and human eosinophils or the eosinophilic leukemia cell lines, Eol-1. Montelukast inhibited co-culture associated morphological changes of BEAS-2b cells, decreased the expression of vimentin and collagen I, and increased the expression of E-cadherin. Montelukast mitigated the rise of TGF-ß1 production and Smad3 phosphorylation. Co-culture of human eosinophils with BEAS-2B cells significantly enhanced the production of CysLTs compared with BEAS-2B cells or eosinophils alone. The increase of CysLTs was abolished by montelukast pre-treatment. Montelukast had similar effects when co-culture system of Eol-1 and BEAS-2B was used. This study showed that montelukast suppresses eosinophils-induced EMT of airway epithelial cells. This finding may explain the mechanism of montelukast-mediated amelioration of airway remodeling in bronchial asthma.


Subject(s)
Acetates/pharmacology , Airway Remodeling/drug effects , Bronchi/drug effects , Epithelial-Mesenchymal Transition/drug effects , Leukotriene Antagonists/pharmacology , Quinolines/pharmacology , Respiratory Mucosa/drug effects , Asthma/metabolism , Asthma/pathology , Bronchi/cytology , Bronchi/metabolism , Cell Line, Tumor , Coculture Techniques , Collagen Type I/metabolism , Cyclopropanes , Cysteine/antagonists & inhibitors , Eosinophils/physiology , Humans , Leukotrienes , Phosphorylation , Respiratory Mucosa/cytology , Smad3 Protein/metabolism , Sulfides , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism
20.
Allergol Int ; 63(1): 57-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24368584

ABSTRACT

BACKGROUND: Thrombomodulin treatment modulates the properties of dendritic cells (DCs) converting them from immunogenic to tolerogenic and inducing its own expression on DCs. Thrombomodulin binds to the inflammatory mediator, high mobility group protein B1 (HMGB1), antagonizing signalling through its receptor, receptor for advanced glycation end products (RAGE). METHODS: To test if soluble thrombomodulin could antagonize HMGB1 signaling via RAGE on DCs. DCs were prepared from mouse bone marrow cells or human monocytes. In some experiments dendritic cells were sorted into thrombomodulin+ and thrombomodulin- populations. Expression of surface maturation markers was determined by flow cytometry following treatment with thrombomodulin in the presence or absence of HMGB1. RESULTS: Thrombomodulin+ dendritic cells secrete less HMGB1 into the medium. HMGB1 reduces the effects of thrombomodulin on expression of DC maturation markers. Treatment with thrombomodulin reduces the expression of maturation markers such as CD80 and CD86 and increases the expression of thrombomodulin on the DC surface. Treatment of DCs with neutralizing anti-HMGB1 antibody acted synergistically with thrombomodulin in increasing thrombomodulin expression on DCs. Treatment with thrombomodulin can still reduce the expression of surface markers on DCs derived from mice that are deficient in RAGE showing that thrombomodulin can affect DCs by an alternative mechanism. CONCLUSIONS: The results of this study show that thrombomodulin modulates DCs both by antagonizing the interaction of HMGB1 with RAGE and by an independent mechanism.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , HMGB1 Protein/antagonists & inhibitors , Thrombomodulin/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Dendritic Cells/drug effects , Female , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL