Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Med Sci Monit ; 29: e942752, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37791420

ABSTRACT

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Qin Zhang, Xin-wei Dong, Jia-ying Xia, Ke-ying Xu, Zhe-rong Xu. Obestatin Plays Beneficial Role in Cardiomyocyte Injury Induced by Ischemia-Reperfusion In Vivo and In Vitro. Med Sci Monit, 2017; 23: 2127-2136. DOI: 10.12659/MSM.901361.

2.
Adv Mater ; 33(50): e2004542, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33829543

ABSTRACT

Inorganic-organic hybrid molecular multiferroic and magnetoelectric materials, similar to multiferroic oxide compounds, have recently attracted increasing attention because they exhibit diverse architectures, a flexible framework, fascinating physics, and potential magnetoelectric functionalities in novel multifunctional devices such as energy transformation devices, sensors, and information storage systems. Herein, the classification of multiferroicity and magnetoelectricity is briefly outlined and then the recent advances in the multiferroicity and magnetoelectricity of inorganic-organic hybrid molecular materials, particularly magnetoelectricity and the relevant magnetoelectric mechanisms and their categories are summarized. In addition, a personal perspective and an outlook are provided.

3.
Eur J Pharmacol ; 848: 55-61, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30707957

ABSTRACT

Various studies have shown that flavones have several pharmacological activities including anti-allergy activities. However, the bioavailability of oral flavones is very low, and whether inhaled administration can improve efficacy in respiratory disease models is unclear. In the present study, the anti-allergic activities of inhaling 5,7-dimethoxy-3,4'-dihydroxyflavone (MHF), a synthetic flavonoid, was investigated by comparison with disodium cromoglycate (DSCG) and nedocromil sodium (NS) in rat allergic models. In an anti-DNP-IgE-induced asthmatic model, inhaled MHF dose-dependently inhibited the increase in airway resistance after antigen challenge. In an ovalbumin (OVA)-induced asthmatic model, inhaled MHF showed significant suppression of airway hyperresponsiveness; a decrease in eosinophil and neutrophil counts, IL-4, IL-5 and leukotriene D4 in bronchoalveolar lavage fluid; a reduction in total IgE and OVA-specific IgE levels in serum; and suppression of eosinophil infiltration in lung tissue after antigen challenge. The efficacy of inhaled MHF was comparable to that of NS and DSCG. In conclusion, based on these findings, the report for the first time that that inhaled MHF may be a potential drug for the treatment of allergic asthma.


Subject(s)
Anti-Allergic Agents/administration & dosage , Asthma/drug therapy , Disease Models, Animal , Flavonoids/administration & dosage , Administration, Inhalation , Animals , Anti-Allergic Agents/chemistry , Asthma/chemically induced , Asthma/metabolism , Dose-Response Relationship, Drug , Female , Flavonoids/chemistry , Ovalbumin/toxicity , Rats , Rats, Sprague-Dawley
4.
Adv Mater ; 30(52): e1803716, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30370676

ABSTRACT

Magnetoelectric materials with a large magnetoelectric response, a low operating magnetic (or electric) field, and a room-temperature (or higher) operating temperature are of key importance for practical applications. However, such materials are extremely rare because a large magnetoelectric response often requires strong coupling between spins and electric dipoles. Herein, an example of a magnetoelectric composite is prepared by using a room-temperature multiaxial molecular-ionic ferroelectric, tetramethylammonium tetrachlorogallate(III) (1). Investigation of the magnetoelectric effect of the magnetoelectric laminate composite indicates that its room-temperature magnetoelectric voltage coefficient (αME ) is as high as 186 mV cm-1 Oe-1 at HDC = 275 Oe and at the HAC frequency of ≈39 kHz, providing a valid approach for the preparation of magnetoelectric materials and adding a new member to the magnetoelectric material family.

5.
Lung Cancer ; 122: 44-53, 2018 08.
Article in English | MEDLINE | ID: mdl-30032844

ABSTRACT

OBJECTIVES: Cigarette smoke (CS) is a major risk factor for the development of lung cancer and chronic obstructive pulmonary disease (COPD). Epithelial-mesenchymal transition (EMT) is found in invasive or metastatic phenotypes in lung cancer and COPD. MK-2206, a pan Akt inhibitor, has failed in clinical trials for solid tumors when administered alone at tolerated doses, but it has been shown to have synergistic effects when applied with certain molecular targeted agents. In this study, we investigated the working mechanism of MK-2206 in CS-induced pulmonary EMT both in vivo and in vitro. MATERIALS AND METHODS: The expression of Akt, epithelial-mesenchymal transition (EMT) markers and signaling proteins were analyzed by immunohistochemistry, real-time PCR and Western blot in cigarette smoke extract (CSE)-treated pulmonary epithelia and CS-treated lung tissues in mice. RESULTS AND CONCLUSION: We demonstrated that exposure of the epithelium to CSE and exposure of the mice to CS can induce EMT by activating the Akt signaling pathway. Intragastric application of MK-2206 at a low dose (50 mg/kg) reversed the changes of the key indicators of EMT in the lungs of CS-exposed mice, including TGF-ß1, α-SMA, vimentin, MMP-9, MMP-2, S100A4, collagen deposition, and E-cadherin. MK-2206 at a non-cytotoxic concentration (0.5 µM) or Akt knockdown consistently reversed the changes of the key indicators of EMT in the pulmonary epithelia. Moreover, we found that the effects of Akt inhibition or knockdown on the CS/CSE-induced EMT acted via the TGF-ß1/Akt/Smad/mTOR and Akt/P38 MAPK pathways. Taken together, our data offer a novel perspective on the molecular mechanism of Akt for CS-induced EMT. This finding may enhance the understanding of the mechanism behind the synergistic use of a low dose of MK-2206 to achieve antitumor efficacy with reduced adverse reactions in patients with lung cancer and COPD.


Subject(s)
Lung Neoplasms/metabolism , Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism , Animals , Cells, Cultured , Cigarette Smoking/adverse effects , Epithelial-Mesenchymal Transition , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology , Signal Transduction
6.
Eur J Pharmacol ; 824: 30-39, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29382534

ABSTRACT

Salvianolic acid B (SalB) is one of the main water-soluble composites from Chinese medicine Dansen (Radix miltiorrhiza). It is used for clinical treatment of various diseases including cardiovascular, lung, Liver, renal and cancers. However, the effects of SalB to allergy induced airway mucin hypersecretion, inflammation and hyperresponsiveness (AHR) remains not clear. Overproduction of airway MUC5AC is a central effector of inflammation that is strongly associated with AHR in asthmatic attack. In this study, we investigated the anti-asthmatic activity and mechanism of SalB in a murine model and human epithelial cells by monitoring changes in mucin expression and secretion, airway inflammation, AHR, and signaling pathways. SalB was administered by intragastric administration (i.g) daily for a week, starting at 21 days after sensitization of ovalbumin (OVA). All examinations were performed 24h after the last antigen challenge. We found that treatments with SalB significantly inhibited increase in the tracheobronchial secretion, glycosaminoglycan levels, interleukin (IL)-13, IL-4, and IL-5 cytokines mRNA and protein expression, and decrease in mucociliary clearance in lung tissues. Histological results demonstrated that SalB attenuated OVA-induced eosinophil infiltration, airway goblet cell hyperplasia, and MUC5AC and MUC5B mRNA and protein expression in lung tissues. SalB exhibited protective effects against AHR in OVA-challenged animals. In vitro, SalB significantly inhibited IL-13-induced MUC5AC and MUC5B mRNA and protein expression in human epithelial cells. These effects were blocked by SalB by downregulating the Erk1/2 and P38 signaling pathways. Taken together, these data indicate that treatment with SalB may improve AHR by inhibiting MUC5AC overproduction.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Benzofurans/pharmacology , MAP Kinase Signaling System/drug effects , Mucin 5AC/biosynthesis , Respiratory Hypersensitivity/drug therapy , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/metabolism , Benzofurans/therapeutic use , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Female , Gene Expression Regulation/drug effects , Glycosaminoglycans/metabolism , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Trachea/drug effects , Trachea/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Adv Mater ; 29(43)2017 Nov.
Article in English | MEDLINE | ID: mdl-28984995

ABSTRACT

A giant room-temperature magnetodielectric (MD) response upon the application of a small magnetic field is of fundamental importance for the practical application of a new generation of devices. Here, the giant room-temperature magnetodielectric response is demonstrated in the metal-organic framework (MOF) of [NH2 (CH3 )2 ]n [FeIII FeII(1-x) NiIIx (HCOO)6 ]n (x ≈ 0.63-0.69) (1) with its MD coefficient remaining between -20% and -24% in the 300-410 K temperature range, even at 0.1 T. Because a room-temperature magnetodielectric response has never been observed in MOFs, the present work not only provides a new type of magnetodielectric material but also takes a solid step toward the practical application of MOFs in a new generation of devices.

8.
Toxicology ; 389: 31-41, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28694203

ABSTRACT

Bleomycin (BLM) has potent tumor cell-killing properties that have given it an important place in cancer chemotherapy, but pulmonary toxicity is its major adverse effect. Soluble epoxide hydrolase (sEH) inhibitors have been reported to have protective effects in fibrosis models, but the effects of AUDA, an sEH inhibitor of BLM-induced pulmonary toxicity and fibrosis, remain to be researched. In this study, we assessed the effects of AUDA on the BLM-induced pulmonary fibrosis in a mouse model, and transforming growth factor (TGF)-ß1-induced epithelial proliferation and epithelial-mesenchymal transition (EMT) in vitro by monitoring changes in pulmonary function, inflammatory response, fibrotic remodeling, and signaling pathways. AUDA was administered by intragastric administration (i.g) daily for three weeks, starting at seven days after intratracheal instillation of BLM. All examinations were performed 24h after the last i.g. In vivo, AUDA significantly improved BLM-induced decline in lung function and body weight, and inhibited inflammatory cell accumulation and the mRNA and protein expression of interleukin (IL)-1ß, TGF-ß1, and matrix metalloproteinase 9 (MMP-9) in lung tissue. Moreover, AUDA attenuated BLM-induced deposition of collagen fibers, destruction of alveolar structures, and pulmonary parenchyma. Additionally, AUDA regulated the expression of α-smooth muscle actin (α-SMA) and E-cadherin by inhibiting the Smad3/p38 signaling pathway. In vitro, AUDA significantly inhibited TGF-ß1-induced epithelial cells and fibroblast proliferation, reduced sEH expression and α-SMA expression, and increased epoxyeicosatrienoic acid (EET) levels and E-cadherin expression in epithelial cells. These effects were blocked by AUDA by downregulating the Smad3 and p38 signaling pathways. Taken together, these data indicate that treatment with sEH inhibitors may improve BLM-induced pulmonary toxicity.


Subject(s)
Adamantane/analogs & derivatives , Bleomycin , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Lauric Acids/pharmacology , Lung/drug effects , Pulmonary Fibrosis/prevention & control , Smad3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adamantane/pharmacology , Animals , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Collagen/metabolism , Cytokines/metabolism , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Epoxide Hydrolases/metabolism , Female , Humans , Inflammation Mediators/metabolism , Lung/enzymology , Lung/pathology , Lung/physiopathology , Mice, Inbred ICR , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/prevention & control , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Time Factors
9.
Med Sci Monit ; 23: 2127-2136, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28472020

ABSTRACT

BACKGROUND Obestatin, primarily recognized as a peptide within the gastrointestinal system, has been shown to benefit the cardiovascular system. We designed this experiment to study the protective role and underlying mechanism of obestatin against ischemia-reperfusion(I/R) injury in myocardial cells. MATERIAL AND METHODS In an In vivo experiment, LAD was ligated for 0.5 h and then opened for reperfusion with obestatin for 24 h. Then, the infarction area was shown with TTC staining, and inflammation factors in serum were analyzed by qRT-PCR. In primary cultured cardiomyocytes, we measured the level of LDH, MDA, GSH, and SOD. Finally, we assessed cells apoptosis using flow cytometry and detected the concentrations of caspase-3, Bax, and Bcl-2 using Western blot analysis. RESULTS TTC staining showed that in the 3 obestatin groups, the infarct area became smaller with the increase of obestatin concentration. Obestatin also inhibited LDH expression in rat serum and decreased mRNA levels of TNF-α, IL-6, ICAM-1, and iNOS in rat cardiomyocytes after reperfusion. In primary cultured cardiomyocytes, obestatin decreased LDH content and increased GSH level after I/R injury. Obestatin was also found to antagonize the apoptosis of cardiomyocytes in a dose-dependent manner. Western blot analysis showed that obestatin downregulated the expression of caspase-3 and Bax and upregulated the expression of Bcl-2. CONCLUSIONS Obestatin can protect cardiomyocyte from I/R-induced injury in vitro and in vivo. This beneficial effect is closely related with its properties of anti-inflammation, anti-cytotoxicity, and anti-apoptosis. The protective effect of obestatin might be associated with activation of Bcl-2 and inhibition of caspase-3 and Bax.


Subject(s)
Ghrelin/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Animals , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Caspase 3/metabolism , Ghrelin/therapeutic use , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Ischemia/drug therapy , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type II/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism
10.
Toxicol Lett ; 273: 1-9, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28300665

ABSTRACT

Pulmonary fibrosis is common in a variety of inflammatory lung diseases, such as interstitial pneumonia, chronic obstructive pulmonary disease, and silicosis. There is currently no effective clinical drug treatment. It has been reported that grape seed extracts (GSE) has extensive pharmacological effects with minimal toxicity. Although it has been found that GSE can improve the lung collagen deposition and fibrosis pathology induced by bleomycin in rat, its effects on pulmonary function, inflammation, growth factors, matrix metalloproteinases and epithelial-mesenchymal transition remain to be researched. In the present study, we studied whether GSE provided protection against bleomycin (BLM)-induced mouse pulmonary fibrosis. ICR strain mice were treated with BLM in order to establish pulmonary fibrosis models. GSE was given daily via intragastric administration for three weeks starting at one day after intratracheal instillation. GSE at 50 or 100mg/kg significantly reduced BLM-induced inflammatory cells infiltration, proinflammatory factor protein expression, and hydroxyproline in lung tissues, and improved pulmonary function in mice. Additionally, treatment with GSE also significantly impaired BLM-induced increases in lung fibrotic marker expression (collagen type I alpha 1 and fibronectin 1) and decreases in an anti-fibrotic marker (E-cadherin). Further investigation indicated that the possible molecular targets of GSE are matrix metalloproteinases-9 (MMP-9) and TGF-ß1, given that treatment with GSE significantly prevented BLM-induced increases in MMP-9 and TGF-ß1 expression in the lungs. Together, these results suggest that supplementation with GSE may improve the quality of life of lung fibrosis patients by inhibiting MMP-9 and TGF-ß1 expression in the lungs.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Grape Seed Extract/therapeutic use , Lung/drug effects , Pulmonary Fibrosis/prevention & control , Animals , Anti-Inflammatory Agents/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Female , Grape Seed Extract/administration & dosage , Lung/immunology , Lung/pathology , Matrix Metalloproteinase 9/metabolism , Mice, Inbred ICR , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Respiratory Function Tests , Transforming Growth Factor beta/metabolism
11.
Int Immunopharmacol ; 33: 90-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26881857

ABSTRACT

Oral and injection administration of ambroxol has been clinically used to treat airway disease. However, little is known about its potentials in inhalation therapy. In present studies, we tested the effects of ambroxol by inhalation with intravenous administration, and explored the underlying working mechanism. The mice received 10 cigarettes exposure every day for 4 days. Inhaled solution of ambroxol was aerosolized 20 min before the exposure of cigarette smoke (CS). The effect of ambroxol on the expression of mucoprotein 5 AC (MUC5AC) and proinflammatory cytokines in NCI-H292 cells stimulated with cigarette smoke extract (CSE). Four days of daily inhalation of ambroxol at 3.75 or 7.5mg/ml for 20 min suppressed the accumulation of neutrophils and macrophages in the bronchoalveolar lavage fluid (BALF) and lung tissues, and inhibited increases in the mRNA and protein levels of tumor necrosis factor (TNF)-α, CCL-2 and KC, but not interleukin (IL)-1ß in the CS-exposed mice. Moreover, ambroxol at 3.75 or 7.5mg/ml facilitated airway mucosa cilia clearance, reduced glycosaminoglycans level in BALF and MUC5AC mRNA levels in lung tissues. The effects of ambroxol by inhalation at 7.5mg/ml was comparable to that of ambroxol at 20mg/kg i.v. and dexamethasone at 0.5mg/kg i.p. Using cultured lung epithelial cells, we demonstrated that pretreatment with ambroxol at 2 or 20 µM inhibited the CSE-induced up-regulation of MUC5AC, TNF-α, IL-1ß mRNA levels, which was through inhibiting Erk signaling pathway. Our results demonstrate the beneficial effects of ambroxol as an inhalation replace systemic administration for COPD therapy.


Subject(s)
Acute Lung Injury/drug therapy , Ambroxol/therapeutic use , Expectorants/therapeutic use , Macrophages/drug effects , Neutrophils/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Respiratory Mucosa/drug effects , Acute Lung Injury/chemically induced , Administration, Inhalation , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Macrophages/immunology , Mice , Mice, Inbred ICR , Mucin 5AC/genetics , Mucin 5AC/metabolism , Mucociliary Clearance/drug effects , Neutrophils/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Smoking/adverse effects
12.
Nutrients ; 7(8): 7085-105, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26305254

ABSTRACT

In this study, we examined the effects of apple polyphenols (APs) on hyperlipidemia, atherosclerosis, hepatic steatosis and endothelial function and investigated the potential mechanisms. ApoE(-/-) mice were fed a western-type diet and orally treated with APs (100 mg/kg) or atorvastatin (10 mg/kg) for 12 weeks. Hyperlipidemia and atherosclerosis in the aortic sinuses and, and hepatic lipidosis were measured. The treatment with APs or atorvastatin induced a remarkable reduction in the atherosclerotic lesions and hepatic steatosis and decreased the levels of low-density lipoprotein, triglyceride, CCL-2 and VCAM-1 levels in the plasma. Conversely, the APs significantly increased the plasma levels of high-density lipoprotein (HDL) cholesterol and markedly up-regulated the glutathione peroxidase (GPx), catalase (CAT) and superoxide dismutase (SOD) levels in liver tissues. Moreover, the APs treatment modulated lipid metabolism by up-regulating the transcription of associated hepatic genes including PPARα, while down-regulating the transcription of SCAP and its downstream genes associated with lipid synthesis in the liver. Histological assessment showed that the APs treatment also reduced the macrophage infiltration in the aortic root plaque and the inflammatory cells infiltrations to the liver tissues. Moreover, we confirmed that the APs treatment greatly reduced the ox-LDL-induced endothelial dysfunction and monocyte adhesion to rat aortic endothelial cells (RAECs). Mechanistically, the APs treatment suppressed the ROS/MAPK/NF-κB signaling pathway, and consequently, reduced CCL-2, ICAM-1 and VCAM-1 expression. Our results suggest that the APs are a beneficial nutritional supplement for the attenuation of atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Fatty Liver/prevention & control , Malus/chemistry , NF-kappa B/genetics , Polyphenols/pharmacology , Reactive Oxygen Species/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Survival/drug effects , Chemokine CCL2/blood , Chemokine CCL2/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/blood , Intercellular Adhesion Molecule-1/genetics , Lipoproteins, LDL/blood , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Cell Adhesion Molecule-1/blood , Vascular Cell Adhesion Molecule-1/genetics
13.
Eur J Pharmacol ; 761: 161-7, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26003274

ABSTRACT

Inhaled corticosteroid (ICS) therapy in combination with long-acting ß-adrenergic agonists (LABA) is the most important treatment for allergic asthma, although the mechanism still remains unclear. However, mast cells play a central role in the pathogenesis of asthma. In this study, we explored the sole or synergetic effects of des-ciclesonide (ICS) and formoterol (LABA) on the cytokines IL-4 and IL-13 and on histamine release from mast cells (RBL-2H3 cells). We found that des-ciclesonide (0.1, 1 and 10nM) and formoterol (0.1, 1 and 10µM) alone attenuated DNP-BSA-induced IL-4 and IL-13 production, respectively, in a concentration-dependent manner in DNP-IgE-sensitized mast cells. Des-ciclesonide (0.2nM) and formoterol (1µM) alone also reduced histamine production. However, the combination of des-ciclesonide (0.2nM) and formoterol (1µM) had a synergistic inhibition effect on IL-4 mRNA expression and protein production but not IL-13 and histamine release. The JNK inhibitor SP600125 (10µM) inhibited antigen-induced mRNA expression and protein production of IL-4. Des-ciclesonide and formoterol alone inhibited the activation of JNK in a concentration-dependent manner, and the combination of des-ciclesonide (0.2nM) and formoterol (1µM) exhibited greater inhibition effect compared with des-ciclesonide (0.2nM) or formoterol (1µM) alone. Taken together, these synergistic effects on mast cells might provide the rationale for the development of the most recent ICS/LABA combination approved for asthma therapy.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Dinitrophenols/immunology , Formoterol Fumarate/pharmacology , Immunoglobulin E/immunology , Interleukin-4/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mast Cells/drug effects , Pregnenediones/pharmacology , Serum Albumin, Bovine/immunology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Enzyme Activation , Gene Expression Regulation , Histamine Release/drug effects , Interleukin-13/metabolism , Interleukin-4/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mast Cells/enzymology , Mast Cells/immunology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects
14.
Article in English | MEDLINE | ID: mdl-25467970

ABSTRACT

In response to endothelial cell activation, arachidonic acid can be converted by cytochrome P450 (CYP) epoxygenases to epoxyeicosatrienoic acids (EETs), which have potent vasodilator and anti-inflammatory properties. In this study, we investigated the effects of exogenous EETs on cigarette smoke extract (CSE)-induced inflammation in human bronchial epithelial cells (NCI-H292). We found that CSE inhibited the expression of CYP2C8 and mildly stimulated the expression of epoxide hydrolase 2 (EPHX2) but did not change the expression of CYP2J2. Treatment with 11,12-EET or 14,15-EET attenuated the CSE-induced release of interleukin (IL)-8 by inhibiting the phosphorylation of p38 mitogen-activated protein kinases (MAPKs). Our results demonstrated that CSE may reduce the anti-inflammatory ability of epithelial cells themselves by lowering the EET level. EETs from pulmonary epithelial cells may play a critical protective role on epithelial cell injury.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Anti-Inflammatory Agents/pharmacology , Bronchi/cytology , Epithelial Cells/drug effects , Interleukin-8/genetics , Smoking/adverse effects , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Bronchi/drug effects , Cell Line , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Humans , Interleukin-8/metabolism , MAP Kinase Signaling System/drug effects , Vasodilator Agents/pharmacology
15.
Biochim Biophys Acta ; 1840(6): 1838-49, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24508121

ABSTRACT

BACKGROUND: Epithelial-mesenchymal transition (EMT) is the major pathophysiological process in lung fibrosis observed in chronic obstructive pulmonary disease (COPD) and lung cancer. Smoking is a risk factor for developing EMT, yet the mechanism remains largely unknown. In this study, we investigated the role of Rac1 in cigarette smoke (CS) induced EMT. METHODS: EMT was induced in mice and pulmonary epithelial cells by exposure of CS and cigarette smoke extract (CSE) respectively. RESULTS: Treatment of pulmonary epithelial cells with CSE elevated Rac1 expression associated with increased TGF-ß1 release. Blocking TGF-ß pathway restrained CSE-induced changes in EMT-related markers. Pharmacological inhibition or knockdown of Rac1 decreased the CSE exposure induced TGF-ß1 release and ameliorated CSE-induced EMT. In CS-exposed mice, pharmacological inhibition of Rac1 reduced TGF-ß1 release and prevented aberrations in expression of EMT markers, suggesting that Rac1 is a critical signaling molecule for induction of CS-stimulated EMT. Furthermore, Rac1 inhibition or knockdown abrogated CSE-induced Smad2 and Akt (PKB, protein kinase B) activation in pulmonary epithelial cells. Inhibition of Smad2, PI3K (phosphatidylinositol 3-kinase) or Akt suppressed CSE-induced changes in epithelial and mesenchymal marker expression. CONCLUSIONS AND GENERAL SIGNIFICANCE: Altogether, these data suggest that CS initiates EMT through Rac1/Smad2 and Rac1/PI3K/Akt signaling pathway. Our data provide new insights into the fundamental basis of EMT and suggest a possible new course of therapy for COPD and lung cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Neuropeptides/physiology , Nicotiana/adverse effects , Pulmonary Alveoli/pathology , Smoke/adverse effects , rac1 GTP-Binding Protein/physiology , Animals , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Smad2 Protein/physiology , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/biosynthesis
16.
Int Immunopharmacol ; 18(2): 358-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389380

ABSTRACT

Glycopyrronium bromide (GB) is a muscarinic receptor antagonist that has been used as a long-acting bronchodilator in chronic obstructive pulmonary disease (COPD) patients. The aim of this study was to investigate the anti-inflammatory activity of inhaled GB in a cigarette smoke-induced acute lung inflammation mouse model. We found that aerosol pre-treatment with GB suppresses the accumulation of neutrophils and macrophages in the bronchoalveolar lavage fluid (BALF) in cigarette smoke (CS)-exposed mice. GB at doses of 300 and 600 µg/ml significantly inhibited the CS-induced increases in the mRNA and protein expression levels of interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, monocyte chemotactic protein (MCP)-1 and transforming growth factor (TGF)-ß1 in lung tissues and the BALF. Moreover, GB at a dose of 600 µg/ml significantly inhibited the CS-induced changes in glutathione (GSH) and myeloperoxidase (MPO) activities in the BALF, decreased the CS-induced expression of matrix metalloproteinases (MMP)-9, and increased the CS-induced expression of tissue inhibitor of metalloproteinases (TIMP)-1, as determined through the immunohistochemical staining of lung tissue. Our results demonstrate the beneficial effects of inhaled GB on the inflammatory reaction in COPD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Glycopyrrolate/therapeutic use , Muscarinic Antagonists/therapeutic use , Pneumonia/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Glycopyrrolate/pharmacology , Lymphocytes/immunology , Macrophages/immunology , Matrix Metalloproteinase 9/immunology , Mice , Mice, Inbred ICR , Muscarinic Antagonists/pharmacology , Neutrophils/immunology , Oxidative Stress/drug effects , Pneumonia/chemically induced , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Smoke , Tissue Inhibitor of Metalloproteinase-1/immunology , Nicotiana
17.
Biochim Biophys Acta ; 1830(8): 4148-59, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583729

ABSTRACT

BACKGROUND: Ginseng is a traditional Chinese herb that has been used for thousands of years. In the present study, effects and mechanisms of AD-1 were evaluated for its development as a novel anti-lung cancer drug. METHODS: The cytotoxic activity was evaluated by MTT assay. Flow cytometry was employed to detect cell cycle, apoptosis and ROS. Western blot and immunohistochemistry were used to analyze signaling pathways. Lung cancer xenograft models were established by subcutaneous implantation of A549 or H292 cells into nude mice. RESULTS: AD-1 concentration-dependently reduces lung cancer cell viability without affecting normal human lung epithelial cell viability. In A549 and H292 lung cancer cells, AD-1 induces G0/G1 cell cycle arrest, apoptosis and ROS production. The apoptosis can be attenuated by a ROS scavenger - N-acetylcysteine (NAC). In addition, AD-1 up-regulates the expression of p38 and ERK phosphorylation. Addition of a p38 inhibitor SB203580, suppresses the AD-1-induced decrease in cell viability. Furthermore, genetic silencing of p38 attenuates the expression of p38 and decreases the AD-1-induced apoptosis. Treatment with NAC reduces AD-1-induced p38 phosphorylation, which indicates that ROS generation is involved in the AD-1-induced p38 activation. In mice, oral administration of AD-1 (10-40mg/kg) dose-dependently inhibited the growth of xenograft tumors without affecting body weight and decreases the expression of VEGF, MMP-9 and CD34 in tumor tissue. TUNEL staining confirms that the tumors from AD-1 treated mice exhibit a markedly higher apoptotic index. CONCLUSIONS AND GENERAL SIGNIFICANCE: These data support development of AD-1 as a potential agent for lung cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Ginsenosides/pharmacology , Lung Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Male , Mice
18.
Nutrition ; 29(1): 235-43, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22964088

ABSTRACT

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is a complex chronic inflammatory disease involving oxidative stress as well as a wide variety of cells activated from smoking cigarettes. There have been disappointingly few therapeutic advances in drug therapy for COPD. Plant polyphenols have been the topic of much research regarding their antioxidant activities and antiinflammatory and immunomodulatory effects. In the present study, we ask whether apple polyphenol provides protection against cigarette smoke (CS)-induced acute lung injury. METHODS: ICR mice were exposed to CS for 4 d with increasing exposure time for up to 6 h per day to elicit epithelial cells injury. One hour before smoke exposure, mice were treated with apple polyphenol (APP) by gavage; all examinations were performed 18 h after the last CS exposure. RESULTS: APP at 30, 100, or 300 mg not only significantly dose-dependently reduced the CS-induced accumulation of inflammatory cells and gene/protein expression of proinflammatory factors both in the lung and in bronchoalveolar lavage fluid, but also significantly reversed oxidative stress in the lungs. Additionally, treatment with APP also significantly regulated the CS-induced imbalance of matrix metalloproteinases-9/tissue inhibitor of metalloproteinase-1 expression in the lungs. To investigate further the possible signaling pathway of APP effects, we examined protein expression of p-P38 MAPK by immunohistochemistry that found treatment with APP significantly decreased the CS-induced increases of p-P38 expression in the lungs. CONCLUSION: Taken together, APP may be a potential dietary nutrient supplement agent to improve quality of life of COPD patients by inhibiting CS-exposed acute lung injury via P38 MAPK signaling pathway.


Subject(s)
Acute Lung Injury/prevention & control , Malus , Polyphenols/administration & dosage , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Chemokines/genetics , Cytokines/genetics , Dietary Supplements , Disease Models, Animal , Female , Gene Expression/drug effects , Humans , Malus/chemistry , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/prevention & control , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Smoking/adverse effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Immunol ; 189(6): 3159-67, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22891281

ABSTRACT

Cigarette smoke (CS), the major cause of chronic obstructive pulmonary disease, contains a variety of oxidative components that were implicated in the regulation of Src homology domain 2-containing protein tyrosine phosphatase 2 (Shp2) activity. However, the contribution of Shp2 enzyme to chronic obstructive pulmonary disease pathogenesis remains unclear. We investigated the role of Shp2 enzyme in blockading CS-induced pulmonary inflammation. Shp2 levels were assessed in vivo and in vitro. Mice (C57BL/6) or pulmonary epithelial cells (NCI-H292) were exposed to CS or cigarette smoke extract (CSE) to induce acute injury and inflammation. Lungs of smoking mice showed increased levels of Shp2, compared with those of controls. Treatment of lung epithelial cells with CSE showed elevated levels of Shp2 associated with the increased release of IL-8. Selective inhibition or knockdown of Shp2 resulted in decreased IL-8 release in response to CSE treatment in pulmonary epithelial cells. In comparison with CS-exposed wild-type mice, selective inhibition or conditional knockout of Shp2 in lung epithelia reduced IL-8 release and pulmonary inflammation in CS-exposed mice. In vitro biochemical data correlate CSE-mediated IL-8 release with Shp2-regulated epidermal growth factor receptor/Grb-2-associated binders/MAPK signaling. Our data suggest an important role for Shp2 in the pathological alteration associated with CS-mediated inflammation. Shp2 may be a potential target for therapeutic intervention for inflammation in CS-induced pulmonary diseases.


Subject(s)
Pneumonia/immunology , Pneumonia/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Smoking/adverse effects , Smoking/pathology , Tobacco Products/toxicity , Acute Disease , Animals , Cell Line , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-8/metabolism , Interleukin-8/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pneumonia/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Smoking/metabolism
20.
Vaccine ; 30(8): 1437-44, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22240342

ABSTRACT

Clinically, sublingual immunotherapy (SLIT) using allergen extracts effectively alleviates the symptoms of allergic rhinitis and asthma. We hypothesized that oral administration of a high-dose of allergen extracts imitates SLIT, which may prevent IgE-related responses in allergic diseases. In the present study, we investigated the effects of oral administration of allergen extracts from mugwort pollen (MP) on allergen-induced inflammation and airway hyperresponsiveness (AHR) in an allergic mouse model. After administration of MPdrop containing Art v 1 and Art v 4 extracts derived from MP specifically in MP-sensitized mice, the effects of MPdrop on AHR, inflammatory cell accumulation, cytokine production in the bronchoalveolar lavage fluid and lung tissue, and serum IgE and IgG levels were investigated. The results indicated that MPdrop not only prevented the AHR in response to methacholine in a dose-dependent manner but also significantly reduced the total serum and allergen-specific IgE levels. All of the maximal effects were achieved at a dose of 100µg/(kgd) and were comparable to the effects of dexamethasone at a dose of 0.5mg/(kgd). Furthermore, oral administration of MPdrop dose-dependently elevated allergen-specific serum IgG2a levels, reduced total and allergen-specific IgE levels and normalized the imbalance between the Th1 cytokine IL-12 and Th2 cytokines IL-4 and IL-5. Finally, oral administration of MPdrop significantly reduced goblet cell hyperplasia and eosinophilia in the MP-sensitized allergic mouse model. These data suggest that MPdrop effectively improves specific allergen-induced inflammation and AHR in MP-sensitized and -challenged mice and provides the rationale for clinical use of MPdrop in the specific allergen-induced asthma.


Subject(s)
Allergens/administration & dosage , Allergens/isolation & purification , Artemisia/chemistry , Desensitization, Immunologic/methods , Hypersensitivity/therapy , Pollen/chemistry , Administration, Oral , Animals , Artemisia/immunology , Disease Models, Animal , Female , Hypersensitivity/immunology , Hypersensitivity/pathology , Hypersensitivity/prevention & control , Immunoglobulin E/blood , Immunoglobulin G/blood , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred BALB C , Pollen/immunology , Respiratory System/immunology , Respiratory System/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...