Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
3.
Eur J Pharmacol ; 890: 173654, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33068589

ABSTRACT

Tobacco smoke is the major risk factor for developing chronic obstructive pulmonary disease (COPD). Viral infection is a major cause of COPD exacerbation, which lacks effective drug treatments. In the present study, to mimic the pathogenesis of COPD, we employed a TLR3 ligand [Poly (I:C), PIC] to mimic viral infection to determine whether it enhances the effects of cigarette smoke (CS)-induced airway inflammation and remodeling. Our results showed that PIC enhanced the effects of cigarette smoke extract (CSE)-induced inflammatory cytokine IL-1ß, TNF-α and IL-8 mRNA expression and remodeling factor E-cadherin, α-SMA and TGF-ß1 mRNA expression with TLR3 upregulation and EGFR phosphorylation in pulmonary epithelial NCI-H292 cells. These responses were inhibited by a TLR3/dsRNA complex inhibitor (TLR3i) or a tyrosine kinase inhibitor icotinib (Ico). Similarly, in the PIC-enhanced CS-induced airway inflammation and remodeling mouse model, treatment with TLR3i or Ico reduced the mRNA and protein expression of the inflammatory cytokines IL-1ß and TNF-α and keratinocyte chemoattractant (KC) and the remodeling factors α-SMA, TGF-ß1, MMP-9 and MUC5AC, while increasing E-cadherin mRNA and protein expression. Furthermore, we found that TLRi and Ico can attenuate the airway hyperreactivity induced by PIC, which is enhanced by CS. Finally, PIC enhanced the effects of CS on TLR3 upregulation and EGFR phosphorylation and significantly increased Erk1/2 and P38 phosphorylation, whereas TLR3i and Ico markedly suppressed TLR3 upregulation and EGFR, Erk1/2 and P38 phosphorylation in the model. Our findings suggest that TLR3/EGFR may be a potential target for the treatment of airway inflammation and remodeling in COPD.


Subject(s)
Airway Remodeling/drug effects , Cigarette Smoking/adverse effects , Pneumonia/prevention & control , Poly I-C/toxicity , Protein Kinase Inhibitors/therapeutic use , Toll-Like Receptor 3/antagonists & inhibitors , Animals , Cell Line , ErbB Receptors/metabolism , Female , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred ICR , Pneumonia/chemically induced , Pneumonia/metabolism , Protein Kinase Inhibitors/pharmacology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Toll-Like Receptor 3/metabolism
4.
J Sci Food Agric ; 101(4): 1419-1427, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-32833246

ABSTRACT

BACKGROUND: Perilla seed oil (PSO) is the main constituent of perilla seeds currently being used in the food industry, however it also has great clinical potential in the regulation of lung function as a nutrition supplement because of the high content of α-linolenic acid (ALA). In this study, the pharmacological activities including anti-tussive, expectorant and anti-inflammatory effect of PSO were performed. Furthermore, the 90-day sub-chronic oral toxicity with a 30 day recovery period was evaluated in Wistar rats. RESULTS: The pharmacological studies demonstrated that PSO inhibited cough frequency induced by capsaicine in mice. PSO also inhibited the leukotriene B4 (LTB4) release from the calcium ionophore A23187-induced polymorphonuclear neutrophils (PMNs) to some extent. In this sub-chronic toxicity study, mortality, clinical signs, body weight, food consumption, hematology, serum biochemistry, urinalysis, organ weight, necropsy, and histopathology were used to evaluate the toxicity of PSO. Lower body weight and various negative impacts on liver related parameters without histopathological lesion were observed in the 16 g kg-1 groups. No clinically significant changes were discovered in the 4 g kg-1 group during the test period. CONCLUSION: In summary, PSO exhibited anti-tussive and anti-inflammatory activities in vivo and in vitro. These sub-chronic toxicity studies inferred that the 'no-observed adverse effect level' (NOAEL) of PSO in Wistar rats was determined to be 4 g kg-1 . These results may provide a safety profile and a valuable reference for the use of PSO. © 2020 Society of Chemical Industry.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Cough/drug therapy , alpha-Linolenic Acid/administration & dosage , Animals , Anti-Inflammatory Agents/adverse effects , Cough/immunology , Drug Evaluation, Preclinical , Female , Humans , Liver/drug effects , Male , Neutrophils/drug effects , Neutrophils/immunology , Plant Oils/administration & dosage , Plant Oils/adverse effects , Rats , Rats, Wistar , Toxicology , alpha-Linolenic Acid/adverse 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.
Food Funct ; 11(5): 4752-4764, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32420568

ABSTRACT

Ginger has been used as a flavoring agent and traditional medicine for a long time in Asian countries. Pharmacological studies showed that it has antiemetic, anti-inflammatory and analgesic effects, which is attributed to its chemical constituents. The aim of the present study is to investigate the anti-rheumatoid arthritis properties of cedrol (CE) found in ginger. In an in vivo anti-RA study, CE remarkably alleviated the paw swelling and arthritis score in CE-treated CIA mice compared with the model group. The neutrophil count and the productions of TNF-α and IL-1ß were inhibited, and the expressions of Rankl, Cox-1 and Cox-2 were down-regulated at the mRNA level. Radiologic evaluation, histopathological analysis and immunohistochemistry indicated that CE ameliorated inflammatory cell infiltration, synovial hyperplasia, and bone and cartilage damage, and exhibited an immunotherapeutic effect. The MTT assay demonstrated that CE (10-10-10-5 M) had no cytotoxicity on fibroblast-like synoviocytes (FLSs), and exhibited an inhibitory effect on the proliferation of LPS-induced FLSs at concentrations of 10-6 M and 10-5 M. Mechanism research showed that the suppressed expressions of pivotal inflammatory mediators including COX-1 and COX-2 subsequently reduced the production of PGE2, thereby causing the secretions of pro-inflammatory cytokines, ultimately attenuating the progression of inflammation. Meanwhile, the reduction in the mRNA levels of Mmp-13 and Mcp-1 responsible for osteoclastogenesis resistance was detected. This illustrated that CE showed anti-rheumatoid arthritis properties via blocking the phosphorylation of ERK/MAPK and p65/NF-κB signaling pathways in LPS-activated FLSs. The current research suggested that CE is an important functional component in ginger, which may be a promising candidate drug for RA therapy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Polycyclic Sesquiterpenes/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/chemically induced , Collagen , Disease Models, Animal , Lipopolysaccharides , Male , Mice , Mice, Inbred DBA , Phytotherapy , Polycyclic Sesquiterpenes/administration & dosage , Polycyclic Sesquiterpenes/pharmacology , Synoviocytes/drug effects
7.
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
8.
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
9.
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
10.
Mol Med Rep ; 19(3): 1728-1738, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30628641

ABSTRACT

The present study aimed to investigate the potent inhibitory effects and possible biochemical basis of the novel phosphodiesterase 4 (PDE4) inhibitor ciclamilast, which is a derivative of piclamilast (RP 73401), on PDE4 and allergic inflammation. Ciclamilast was orally administered to allergic rats, their lungs and bronchoalveolar lavage fluid (BALF) were harvested, and their levels of inflammation and goblet cell hyperplasia, particularly cAMP­PDE activity, and expression and distribution of PDE4 subtypes were determined. The results suggested that oral administration of ciclamilast significantly reduced the total leukocyte number and eosinophil number in BALF and suppressed lung histology changes, including the infiltration of inflammatory cells into the perivascular and peribronchial spaces, structural changes and goblet cell hyperplasia. For eosinophil infiltration, ciclamilast exhibited improved selectivity compared with piclamilast. Furthermore, ciclamilast significantly inhibited the upregulated activity of cAMP­PDE and showed improved selective inhibition of the protein expression of PDE4B than piclamilast in a dose­dependent manner. The mRNA expression of PDE4D was significantly increased in allergic rats, but PDE4B was not. PDE4B was mainly distributed in the cytoplasm, whereas PDE4D was mainly distributed in the cell membrane. The improved anti­inflammatory activity of ciclamilast compared with piclamilast may be due to its higher level of inhibition of the activity, mRNA and protein expression of PDE4, particularly its effect on PDE4B.


Subject(s)
Benzamides/administration & dosage , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Hypersensitivity/drug therapy , Inflammation/drug therapy , Pyridines/administration & dosage , Animals , Blood Cell Count , Bronchoalveolar Lavage Fluid , Cyclic AMP/genetics , Disease Models, Animal , Eosinophils/drug effects , Gene Expression Regulation/drug effects , Goblet Cells/drug effects , Goblet Cells/pathology , Humans , Hyperplasia/drug therapy , Hyperplasia/pathology , Hypersensitivity/genetics , Hypersensitivity/pathology , Inflammation/genetics , Inflammation/pathology , Leukocytes/drug effects , Lung/drug effects , Lung/pathology , Phosphodiesterase Inhibitors/administration & dosage , Rats
11.
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
12.
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
13.
J Int Med Res ; 46(4): 1349-1357, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29376467

ABSTRACT

Objective This study aimed to examine the mechanism of diaphragmatic dysfunction in sepsis due to severe acute pancreatitis (SAP) with intra-abdominal hypertension (IAH) in a rat model. Methods The rats were assigned at random to four groups: (1) control (n = 5), (2) SAP (n = 5), (3) SAP+IAH (n = 5), and (4) SAP+IAH+SS-31 (n = 5). Length and force output of the diaphragm were analysed in vivo. Histopathological examinations were performed by haematoxylin-eosin. Oxidative stress levels related to protease in diaphragmatic mitochondria were detected with a colorimetric technique. Results In the septic rat model due to SAP complicated by IAH, myofibres were increased. Muscle contractile function was significantly lower in the SAP+IAH group compared with the SAP and control groups. Glutathione peroxidase and superoxide dismutase levels were significantly lower and malondialdehyde levels were higher in the SAP and SAP+IAH groups compared with the control group. Notably, SS-31 could reverse atrophy of myofibres in SAP+IAH rats, as well as contractile dysfunction and mitochondrial dysfunction in the diaphragm. Conclusions Diaphragmatic structure and biomechanics are altered in septic rats due to SAP and IAH. This finding is mainly due to an increase in release of mitochondrial reactive oxygen species.


Subject(s)
Diaphragm/physiopathology , Intra-Abdominal Hypertension/complications , Intra-Abdominal Hypertension/physiopathology , Pancreatitis/complications , Pancreatitis/physiopathology , Sepsis/complications , Sepsis/physiopathology , Acute Disease , Animals , Citrate (si)-Synthase/metabolism , Diaphragm/pathology , Electron Transport Complex IV/metabolism , Glutathione Peroxidase/metabolism , Intra-Abdominal Hypertension/enzymology , Male , Malondialdehyde/metabolism , Mitochondria/metabolism , Muscle Contraction , Oxidative Stress , Pancreatitis/enzymology , Rats , Rats, Sprague-Dawley , Sepsis/enzymology , Superoxide Dismutase/metabolism
14.
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
15.
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
16.
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
17.
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
18.
Eur J Pharmacol ; 775: 138-48, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26872986

ABSTRACT

Ambroxol, a metabolite of bromhexine, is shown to exert several pharmacological activities, including secretolytic, anti-inflammatory and antioxidant actions. Oral and intravenous administration of ambroxol is useful for the airway inflammatory diseases. However, little is known about its potential in inhalation therapy for lipopolysaccharide (LPS)-induced mucous hypersecretion and inflammatory response. In the present study, we compared the pharmacological effects of ambroxol by inhalation with intravenous administration and preliminarily explored its mechanism of action. Our results demonstrated that ambroxol administered by inhalation inhibited MUC5AC expression, reduced glycosaminoglycan levels, enhanced the function of mucociliary clearance and promoted sputum excretion, suggesting that ambroxol increases expectoration of sputum by reducing its viscosity. Moreover, ambroxol significantly alleviated LPS-induced the influx of inflammatory cells and the extracellular signal-regulated kinase 1/2 (Erk 1/2) expression in lung tissues, and inhibited increases in the mRNA expression of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, CCL-2 (monocyte chemotactic protein-1), KC (keratinocyte cell protein) and interleukin (IL)-1ß in lung tissues. The secretolytic and anti-inflammatory effects of inhaled ambroxol at a dose of 7.5 mg/ml was comparable to that of ambroxol at 20 mg/ml i.v. and dexamethasone at 0.5 mg/kg i.p. In addition, we found that ambroxol dose-dependently inhibited LPS-induced increases in the mRNA expression of MUC5AC, TNF-α, and IL-1ß in human bronchial epithelial cell (NCI-H292) by inhibiting the Erk signaling pathway. These results demonstrate the beneficial effects of ambroxol in inhalation therapy for the airway inflammatory diseases.


Subject(s)
Acute Lung Injury/drug therapy , Ambroxol , Anti-Inflammatory Agents , Expectorants , MAP Kinase Signaling System/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Administration, Inhalation , Ambroxol/administration & dosage , Ambroxol/pharmacology , Ambroxol/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Cytokines/genetics , Expectorants/administration & dosage , Expectorants/pharmacology , Expectorants/therapeutic use , Female , Glycosaminoglycans/metabolism , Humans , Lipopolysaccharides , Lung/drug effects , Lung/metabolism , Mice, Inbred ICR , Mucin 5AC/genetics , Mucociliary Clearance/drug effects , Mucus/metabolism , RNA, Messenger/metabolism
19.
Pharm Biol ; 54(2): 347-53, 2016.
Article in English | MEDLINE | ID: mdl-25963228

ABSTRACT

CONTEXT: Cryptoporus volvatus (Peck) Hubb grows wild in China, and its fruiting bodies have been used traditionally to treat asthma and bronchitis. OBJECTIVES: This study evaluates the anti-inflammatory effect of Cryptoporus polysaccharides (CP) extracted from fruiting bodies of C. volvatus on lipopolysaccharide (LPS)-induced pro-inflammatory factors and the signaling pathways involved in human alveolar epithelial cells. MATERIALS AND METHODS: To evaluate the effects of CP on LPS-induced pro-inflammatory factors, A549 cells were pre-incubated with CP 1, 10, and 100 µg/ml for 1 h and then stimulated with LPS 10 µg/ml for 24 h. The expression of pro-inflammatory factors monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), Toll-like receptor 2 (TLR2), and phosphorylation of ERK1/2, p38, and NF-κB p65 were measured by q-PCR, ELISA, and western blotting. RESULTS: CP decreased LPS-induced mRNA expression of MCP-1, TNF-α, and IL-1ß (IC50 = 83.3, 85.2, and 91.6 µg/ml, respectively) and their correspondent protein expression (IC50 = 88.6, 76.4, and 81.6 µg/ml, respectively). Investigation of potential mechanisms indicated that CP 100 µg/ml reduced LPS-induced expression of TLR2 mRNA (66.9%, p < 0.01) and protein (63.2%, p < 0.01) that was a result of the decreased pro-inflammatory factors. LPS induction increased the expression of TLR2 and the phosphorylation of p38 and ERK1/2, NF-kB p65 concomitantly. CP 100 µg/ml inhibited the LPS-induced phosphorylation of the signaling proteins (p < 0.05). CONCLUSIONS: This suggests that CP pretreatment down-regulates LPS-mediated inflammation in lung epithelial cells. This study further confirmed that CP is a potential anti-inflammatory drug for the treatment of airway inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coriolaceae/chemistry , Cytokines/genetics , Epithelial Cells/drug effects , Fungal Polysaccharides/pharmacology , Pulmonary Alveoli/drug effects , Toll-Like Receptor 2/metabolism , Anti-Inflammatory Agents/isolation & purification , Blotting, Western , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cytokines/immunology , Dose-Response Relationship, Drug , Epithelial Cells/immunology , Fungal Polysaccharides/isolation & purification , Humans , Lipopolysaccharides/pharmacology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Signal Transduction , Time Factors
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...