Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
2.
Eur J Pharmacol ; 955: 175902, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37422119

ABSTRACT

Allergic rhinitis (AR) is a nasal mucosal disease with sneezing and nasal itching as the main symptoms. Although AR treatment continues to improve, there remains a lack of effective drugs. There are still controversies regarding whether anticholinergic drugs can effectively and safely relieve the symptoms of AR and reduce inflammation in the nasal mucosa. Here, we synthesized 101BHG-D01, which is a novel anticholinergic drug that mainly targets the M3 receptor and may reduce the adverse effects of other anticholinergic drugs on the heart. We evaluated the effects of 101BHG-D01 on AR and investigated the potential molecular mechanism of anticholinergic therapy for AR. We found that 101BHG-D01 effectively alleviated AR symptoms, reduced the infiltration of inflammatory cells and attenuated the expression of inflammatory factors (IL-4, IL-5, IL-13, etc.) in various AR animal models. In addition, 101BHG-D01 reduced the activation of mast cells and the release of histamine from rat peritoneal mesothelial cells (RPMCs) challenged by IgE. Moreover, 101BHG-D01 reduced the expression of MUC5AC in IL-13-challenged rat nasal epithelial cells (RNECs) and human nasal epithelial cells (HNEpCs). Furthermore, IL-13 stimulation significantly increased JAK1 and STAT6 phosphorylation, which was suppressed by 101BHG-D01. We demonstrated that 101BHG-D01 reduced mucus secretion and inflammatory cell infiltration in the nasal mucosa, which may occur through a reduction in activation of the JAK1-STAT6 signaling pathway, indicating that 101BHG-D01 is a potent and safe anticholinergic therapy for AR.


Subject(s)
Interleukin-13 , Rhinitis, Allergic , Humans , Rats , Animals , Mice , Interleukin-13/metabolism , Immunoglobulin E , Disease Models, Animal , Rhinitis, Allergic/drug therapy , Rhinitis, Allergic/metabolism , Nasal Mucosa , Mice, Inbred BALB C , Ovalbumin/pharmacology , Cytokines/metabolism
4.
Article in English | MEDLINE | ID: mdl-34403817

ABSTRACT

The anthropogenic-induced cadmium (Cd) pollution poses great threats to human health and wildlife survival. Birds also suffer from Cd contamination and Cd exerts negative impacts on multiple organs in birds. However, its toxic effects on cardiac organ of birds are still unclear. In this study, one-week old male Japanese quails were exposed to 15, 30, 60 and 75 mg/kg Cd for 5 weeks when birds in control group reached sex maturity. The results showed that Cd could cause microstructural damages including congestion and myocardial fiberolysis. Ultrastructural analysis also showed myocardial muscle fiber disarrangement and rupture as well as mitochondrial swelling, vacuolation and membrane lysis in Cd concentration groups. Moreover, Cd induced oxidative stress in the heart by decreasing antioxidant enzyme activities of catalase (CAT), glutathione peroxidase (GPX), total antioxidant capacity (T-AOC), superoxide dismutase (SOD) while increasing oxidative biomarkers such as malondialdehyde (MDA), inducible nitric oxide synthase (iNOS), and content of nitric oxide (NO). In addition, mRNA expression levels of genes involved in muscle fiber formation signaling pathway such as Follistatin (FST), paired box 3 (PAX3), myogenic differentiation 1 (MYoD1) and SRY-box transcription factor 6 (SOX6), were down-regulated by Cd exposure. Furthermore, PI3K/Akt/mTOR signaling pathway were disrupted by Cd exposure implying energy supply deficiency in the heart. We concluded that Cd caused cardiac dysfunction by inducing heart underdevelopment, histopathological injury, oxidative stress and myocardial muscle fiber formation disruption.


Subject(s)
Cadmium/toxicity , Cardiotoxicity/etiology , Heart/drug effects , Muscle Fibers, Skeletal/drug effects , Myocardium/pathology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cardiotoxicity/pathology , Coturnix , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Myocardium/metabolism , Oxidative Stress/drug effects
5.
Respir Res ; 21(1): 161, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32586329

ABSTRACT

Cigarette smoke (CS) is a major risk factor for the development of lung cancer and chronic obstructive pulmonary disease (COPD). Epithelial-mesenchymal transition (EMT) commonly coexists in lung cancer and COPD. CS triggers many factors including matrix metalloproteinases (MMPs) production, contributing to EMT progression in the lungs. Here, how Shp2 signaling regulates the CS-induced MMP-9 production and EMT progression were investigated in mouse lungs and in pulmonary epithelial cell cultures (NCI-H292) found CS induced MMP-9 production, EMT progression (increased vimentin and α-SMA; decreased E-cadherin) and collagen deposition in lung tissues; cigarette smoke extract (CSE) induced MMP-9 production and EMT-related phenotypes in NCI-H292 cells, which were partially prevented by Shp2 KO/KD or Shp2 inhibition. The CSE exposure induced EMT phenotypes were suppressed by MMP-9 inhibition. Recombinant MMP-9 induced EMT, which was prevented by MMP-9 inhibition or Shp2 KD/inhibition. Mechanistically, CS and CSE exposure resulted in ERK1/2, JNK and Smad2/3 phosphorylation, which were suppressed by Shp2 KO/KD/inhibition. Consequentially, the CSE exposure-induced MMP-9 production and EMT progression were suppressed by ERK1/2, JNK and Smad2/3 inhibitors. Thus, CS induced MMP-9 production and EMT resulted from activation of Shp2/ERK1/2/JNK/Smad2/3 signaling pathways. Our study contributes to the underlying mechanisms of pulmonary epithelial structural changes in response to CS, which may provide novel therapeutic solutions for treating associated diseases, such as COPD and lung cancer.


Subject(s)
Cigarette Smoking/metabolism , Epithelial-Mesenchymal Transition/physiology , Matrix Metalloproteinase 9/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Animals , Cell Line, Tumor , Cigarette Smoking/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Inhalation Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology
6.
Respir Res ; 21(1): 22, 2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31931796

ABSTRACT

Airway remodeling consists of the structural changes of airway walls, which is often considered the result of longstanding airway inflammation, but it may be present to an equivalent degree in the airways of children with asthma, raising the need for early and specific therapeutic interventions. The arachidonic acid cytochrome P-450 (CYP) pathway has thus far received relatively little attention in its relation to asthma. In this study, we studied the inhibition of soluble epoxide hydrolase (sEH) on airway remodeling and hyperresponsiveness (AHR) in a chronic asthmatic model which long-term exposure to antigen over a period of 12 weeks. The expression of sEH and CYP2J2, the level of 14, 15-epoxyeicosatrienoic acids (EETs), airway remodeling, hyperresponsiveness and inflammation were analyzed to determine the inhibition of sEH. The intragastric administration of 3 or 10 mg/kg ZDHXB-101, which is a structural derivative of natural product honokiol and a novel soluble epoxide hydrolase (sEH) inhibitor, daily for 9 weeks significantly increased the level of 14, 15-EETs by inhibiting the expression of sEH and increasing the expression of CYP2J2 in lung tissues. ZDHXB-101 reduced the expression of remodeling-related markers such as interleukin (IL)-13, IL-17, MMP-9 N-cadherin, α-smooth muscle actin, S100A4, Twist, goblet cell metaplasia, and collagen deposition in the lung tissue or in bronchoalveolar lavage fluid. Moreover, ZDHXB-101 alleviated AHR, which is an indicator that is used to evaluate the airway remodeling function. The inhibitory effects of ZDHXB-101 were demonstrated to be related to its direct inhibition of the extracellular signal-regulated kinase (Erk1/2) phosphorylation, as well as inhibition of c-Jun N-terminal kinases (JNK) and the signal transducer and activator of transcription-3 (STAT3) signal transduction. These findings first revealed the anti-remodeling potential of ZDHXB-101 lead in chronic airway disease.


Subject(s)
Airway Remodeling/drug effects , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/metabolism , MAP Kinase Signaling System/drug effects , STAT3 Transcription Factor/metabolism , Airway Remodeling/physiology , Animals , Anti-Asthmatic Agents/chemistry , Anti-Asthmatic Agents/pharmacology , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Epoxide Hydrolases/metabolism , Female , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred ICR , STAT3 Transcription Factor/antagonists & inhibitors
7.
Eur J Pharmacol ; 868: 172874, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31866410

ABSTRACT

Airway remodeling in asthma is difficult to treat because of its complex pathophysiology that involves proinflammatory cytokines, as well as the arachidonic acid cytochrome P-450 (CYP) pathway; however, it has received little attention. In this study, we assessed the efficacy of a soluble epoxide hydrolase (sEH) on airway remodeling in a mouse model of chronic asthma. The expression of sEH and CYP2J2 and the level of 14,15-epoxyeicosatrienoic acid (14,15-EET), airway remodeling and hyperresponsiveness (AHR) were analyzed to determine the level of sEH inhibition. AUDA, a sEH inhibitor, was given daily for 9 weeks orally, which significantly increased the level of 14,15-EET by inhibiting the expression of sEH and increasing the expression of CYP2J2 in lung tissues. The inhibition of sEH reduced the expression of remodeling-related molecular markers, such as interleukin (IL)-13, IL-17, matrix metalloproteinase 9, N-cadherin, α-smooth muscle actin (α-SMA), S100A4, Twist, epithelial goblet cell metaplasia, and collagen deposition in bronchoalveolar lavage fluid (BAL fluid) and lung tissues. Moreover, remodeling-related eosinophil accumulation in the BAL fluid and infiltration into the lung tissue were improved by AUDA. Finally, AUDA alleviated AHR, which is a functional indicator of airway remodeling. The effect of AUDA on airway remodeling was related to the downregulation of extracellular-regulated protein kinases (Erk1/2), c-Jun N-terminal kinases (JNK) and signal transducer and activator of transcription 3 (STAT3). To our knowledge, this is the first report to demonstrate that inhibition of sEH exerts significant protective effects on airway remodeling in asthma.


Subject(s)
Adamantane/analogs & derivatives , Airway Remodeling/drug effects , Asthma/drug therapy , Epoxide Hydrolases/antagonists & inhibitors , Lauric Acids/pharmacology , Lung/drug effects , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/blood , Adamantane/pharmacology , Adamantane/therapeutic use , Airway Remodeling/immunology , Animals , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Epoxide Hydrolases/metabolism , Female , Humans , Lauric Acids/therapeutic use , Lung/immunology , Lung/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
8.
Ecotoxicol Environ Saf ; 183: 109588, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31450035

ABSTRACT

Lead (Pb) is one of the most hazardous metals to human and wildlife and it also has multiple negative impacts on birds. However, its influences on bird gut morphology and intestinal microbiota were still unclear. We used female Japanese quails (Coturnix japonica) to examine the effects of chronic lead exposure (0, 50 ppm and 1000 ppm) on cecal histology, microbial communities and immune function. The results showed 50 ppm lead exposure caused subtle damages of cecum cell structure. However, 1000 ppm lead exposure caused severe cecum histopathological changes characterized by mucosa abscission, Lieberkühn glands destruction and lymphocyte proliferation. Moreover, both lead concentrations induced ultrastructural damages featured by nucleus pyknosis, mitochondrial vacuolation and microvilli contraction. Meanwhile, microbial community structure, species diversity, taxonomic compositions and taxa abundance in the cecum were affected by lead exposure. Furthermore, the mRNA relative expression of immunity-related genes such as interleukin 2 (IL-2) and gamma interferon (IFN-γ) was significantly downregulated while that of interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and natural killer kappa B (NF-κB) was significantly upregulated in the cecum of 50 and 1000 ppm lead exposure groups. We concluded that lead exposure may cause gut health impairment of female Japanese quails by inducing cecal histopathological changes, microbiota dysbiosis and cecal immune disorder.


Subject(s)
Cecum/drug effects , Coturnix/immunology , Coturnix/microbiology , Dysbiosis/chemically induced , Environmental Exposure , Gastrointestinal Microbiome/drug effects , Lead/toxicity , Animals , Cecum/immunology , Cecum/microbiology , Cecum/pathology , Cytokines/genetics , Disease Models, Animal , Female
9.
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
10.
Brain Res ; 1687: 104-116, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29510141

ABSTRACT

Extra-retinal, non-pineal, encephalic photoreceptors (EP) play important roles in mediating development of the reproductive system by the annual change in day length (photoperiodic gonadal response - PGR) in birds. However, the distribution of rhodopsin-like EPs and their functional daily, circadian and seasonal changes are still unclear in the avian brain. This study identifies two novel groups of rhodopsin-immunoreactive cells in the nucleus paraventricularis magnocellularis (PVN) of the hypothalamus and in the medial basal hypothalamus (MBH) in a seasonally breeding species, Gambel's white-crowned sparrow (Zonotrichia leucophrys gambelii). In the PVN, rhodopsin-ir cell number showed both daily and circadian changes with more labeled cells apparent in the night phase in photosensitive birds, while only circadian changes were observed involving fewer labeled cells in the night phase in photorefractory birds. Single long day photo-stimulation significantly decreased the rhodopsin-ir cell number only in photosensitive birds, coincident with a rise in plasma levels of luteinizing hormone (LH). In the MBH, rhodopsin-ir cell number did not show daily, circadian or single long day induced changes in either photoperiodic states. But, overall these rhodopsin expressing neurons significantly increased from photosensitive to photorefractory states. In the median eminence (ME), more intense rhodopsin-ir was detected in photorefractory birds compared to photosensitive birds. For expression of GnRH and vasoactive intestinal polypeptide (VIP), seasonal differences were found with opposite relationships, consistent with previous studies. Our results suggest different roles of the two groups of rhodopsin-like EPs in the regulation of PGR in white-crowned sparrows.


Subject(s)
Circadian Rhythm , Hypothalamus, Middle/cytology , Intralaminar Thalamic Nuclei/cytology , Photoreceptor Cells/metabolism , Rhodopsin/metabolism , Seasons , Animals , Gonadotropin-Releasing Hormone/metabolism , Sparrows/physiology , Vasoactive Intestinal Peptide/metabolism
11.
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
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1778-1788, 2017 07.
Article in English | MEDLINE | ID: mdl-28428003

ABSTRACT

Cigarette smoke (CS) is a major risk factor for the development of chronic obstructive pulmonary disease (COPD). Our previous studies have indicated that Rac1 is involved in lipopolysaccharide-induced pulmonary injury and CS-mediated epithelial-mesenchymal transition. However, the contribution of Rac1 activity to CS-induced lung inflammation remains not fully clear. In this study, we investigated the regulation of Rac1 in CS-induced pulmonary inflammation. Mice or 16HBE cells were exposed to CS or cigarette smoke extract (CSE) to induce acute inflammation. The lungs of mice exposed to CS showed an increase in the release of interleukin-6 (IL-6) and keratinocyte-derived chemokine (KC), as well as an accumulation of inflammatory cells, indicating high Rac1 activity. The exposure of 16HBE cells to CSE resulted in elevated Rac1 levels, as well as increased release of IL-6 and interleukin-8 (IL-8). Selective inhibition of Rac1 ameliorated the release of IL-6 and KC as well as inflammation in the lungs of CS-exposed mice. Histological assessment showed that treatment with a Rac1 inhibitor, NSC23766, led to a decrease in CD68 and CD11b positive cells and the infiltration of neutrophils and macrophages into the alveolar spaces. Selective inhibition or knockdown of Rac1 decreased IL-6 and IL-8 release in 16HBE cells induced by CSE, which correlated with CSE-induced Rac1-regulated Erk1/2 mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription-3 (STAT3) signaling. Our data suggest an important role for Rac1 in the pathological alterations associated with CS-mediated inflammation. Rac1 may be a promising therapeutic target for the treatment of CS-induced pulmonary inflammation.


Subject(s)
Cigarette Smoking/adverse effects , Lung/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Neuropeptides/metabolism , Pneumonia/metabolism , STAT3 Transcription Factor/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cigarette Smoking/genetics , Cigarette Smoking/metabolism , Cytokines/genetics , Cytokines/metabolism , Inflammation/etiology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lung/pathology , Mice , Mitogen-Activated Protein Kinase 3/genetics , Neuropeptides/genetics , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/pathology , STAT3 Transcription Factor/genetics , rac1 GTP-Binding Protein/genetics
13.
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
14.
PLoS One ; 10(6): e0128278, 2015.
Article in English | MEDLINE | ID: mdl-26035589

ABSTRACT

Cytochrome P-450 epoxygenase (EPOX)-derived epoxyeicosatrienoic acids (EETs), 5-lipoxygenase (5-LO), and leukotriene B4 (LTB4), the product of 5-LO, all play a pivotal role in the vascular inflammatory process. We have previously shown that EETs can alleviate oxidized low-density lipoprotein (ox-LDL)-induced endothelial inflammation in primary rat pulmonary artery endothelial cells (RPAECs). Here, we investigated whether ox-LDL can promote LTB4 production through the 5-LO pathway. We further explored how exogenous EETs influence ox-LDL-induced LTB4 production and activity. We found that treatment with ox-LDL increased the production of LTB4 and further led to the expression and release of both monocyte chemoattractant protein-1 (MCP-1/CCL2) and intercellular adhesion molecule-1 (ICAM-1). All of the above ox-LDL-induced changes were attenuated by the presence of 11,12-EET and 14,15-EET, as these molecules inhibited the 5-LO pathway. Furthermore, the LTB4 receptor 1 (BLT1 receptor) antagonist U75302 attenuated ox-LDL-induced ICAM-1 and MCP-1/CCL2 expression and production, whereas LY255283, a LTB4 receptor 2 (BLT2 receptor) antagonist, produced no such effects. Moreover, in RPAECs, we demonstrated that the increased expression of 5-LO and BLT1 following ox-LDL treatment resulted from the activation of nuclear factor-κB (NF-κB) via the p38 mitogen-activated protein kinase (MAPK) pathway. Our results indicated that EETs suppress ox-LDL-induced LTB4 production and subsequent inflammatory responses by downregulating the 5-LO/BLT1 receptor pathway, in which p38 MAPK phosphorylation activates NF-κB. These results suggest that the metabolism of arachidonic acid via the 5-LO and EPOX pathways may present a mutual constraint on the physiological regulation of vascular endothelial cells.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonate 5-Lipoxygenase/chemistry , Leukotriene B4/metabolism , Lipoproteins, LDL/pharmacology , Pulmonary Artery/metabolism , Receptors, Leukotriene B4/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Blotting, Western , Cells, Cultured , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Phosphorylation/drug effects , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Leukotriene B4/genetics , Receptors, Leukotriene B4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vasodilator Agents/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
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
16.
Drug Dev Res ; 76(3): 123-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25958838

ABSTRACT

Bencycloquidium bromide (BCQB), a novel M3 receptor antagonist, alleviates airway hyperresponsiveness, inflammation, and airway remodeling in a murine model of asthma. The aim of this study was to investigate the anti-inflammatory activity of inhaled BCQB in a cigarette smoke (CS)-induced model of acute lung inflammation. Mice exposed to CS developed chronic obstructive pulmonary disease (COPD). Inhalation of BCQB suppressed the accumulation of neutrophils and macrophages in airways and lung and also inhibited the CS-induced increases in mRNA levels of keratinocyte-derived chemokine, monocyte chemotactic protein-1, tumor necrosis factor-alpha, and interleukin-1ß in lung and protein expression levels in bronchoalveolar lavage fluid. Moreover, BCQB (300 µg/ml) inhibited the CS-induced changes in superoxide dismutase and myeloperoxidase activities in the lungs. Our study suggests that BCQB might be a potential therapy for inflammation in CS-induced pulmonary diseases, including COPD.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Disease Models, Animal , Nicotiana/adverse effects , Pneumonia/drug therapy , Receptor, Muscarinic M3/antagonists & inhibitors , Smoke/adverse effects , Administration, Inhalation , Animals , Dose-Response Relationship, Drug , Female , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred ICR , Pneumonia/metabolism , Pneumonia/pathology , Smoking/adverse effects , Smoking/metabolism , Smoking/pathology , Treatment Outcome
17.
Asia Pac J Clin Oncol ; 11(3): 253-61, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25866140

ABSTRACT

AIM: This systematic review and meta-analysis was performed to assess the efficacy and side effects between single-agent and doublet chemotherapy in first-line treatment of advanced non-small cell lung cancer with performance status 2 (PS2). METHODS: We searched for randomized controlled trials in online electronic databases and extracted data from eligible studies for meta-analysis. Pooled hazard ratio (HR) for overall survival (OS), pooled risk difference (RD) for 1-year survival, pooled risk ratio (RR) for objective response rate (ORR) and adverse effects were calculated using a fixed-effect model. RESULTS: Six trials with 386 participants in the single-agent group and 389 participants in the doublet group were included in this review. Compared with single-agent chemotherapy, doublet significantly improved OS (HR 0.72; 95% CI 0.61-0.84; P < 0.0001) and significantly increased 1-year survival rate (RD -0.09; 95% CI -0.14 to -0.03; P = 0.004) and ORR (RR 0.4; 95% CI 0.30-0.69; P = 0.0002). Doublet chemotherapy also significantly increased the risk of grade 3/4 neutropenia (RR = 4.97; 95% CI 2.93-8.43; P < 0.00001), thrombocytopenia (RR = 10.29; 95% CI 3.80-27.85; P < 0.00001) and anemia (RR = 2.50; 95% CI 1.27-4.90; P = 0.008). CONCLUSIONS: Our study implies that carboplatin-containing doublet chemotherapy improved OS, 1-year survival rate and ORR, but increased the risk of grade 3/4 hematotoxicity. Carboplatin-containing doublet may well be superior to non-carboplatin-containing treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Survival Rate
18.
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
19.
Toxicol Mech Methods ; 24(8): 575-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25134437

ABSTRACT

Cigarette smoke contains reactive oxygen (ROS) that can cause oxidative stress. It increases the number of apoptotic and necrotic lung cells and further induces the development of chronic airway disease. In this study, we investigated the effects of cigarette smoke extract (CSE) on apoptosis in human bronchial epithelial cells (BEAS-2B). CSE exposure induced ROS generation and p38 mitogen-activated protein kinase (MAPK) activation that are associated with the activation of apoptosis-regulating signal kinase 1 (ASK-1). N-acetylcysteine (a general antioxidant) attenuated the CSE-induced ASK-1 and p38 MAPK activation and cell apoptosis, suggesting a triggering role of ROS in ASK-1/p38 MAPK activation during apoptotic progression. In contrast, the inhibition and knockdown of p38 attenuated the expression of anti-oxidant master NF-E2-related factor 2 (Nrf-2) and CSE-induced apoptosis, suggesting that p38 MAPK modulates Nrf-2 expression and presumably prevents cell apoptosis. Taken together, the data presented in this manuscript demonstrate that the ROS-dependent ASK-1/p38 signaling cascade regulates CSE-induced BEAS-2B cell apoptosis. In addition, anti-oxidative Nrf-2 is also up-regulated by the ROS/p38 signaling cascade in this progression.


Subject(s)
Apoptosis/drug effects , Bronchi/drug effects , Gene Expression Regulation/drug effects , NF-E2-Related Factor 2/agonists , Respiratory Mucosa/drug effects , Smoking/adverse effects , Up-Regulation/drug effects , Acetylcysteine/pharmacology , Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Bronchi/enzymology , Bronchi/metabolism , Cell Line , Complex Mixtures/antagonists & inhibitors , Complex Mixtures/toxicity , Enzyme Activation/drug effects , Gene Silencing , Humans , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/chemistry , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Respiratory Mucosa/enzymology , Respiratory Mucosa/metabolism , Smoke , Tobacco Products , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL