Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
2.
Cancer Chemother Pharmacol ; 81(4): 705-716, 2018 04.
Article in English | MEDLINE | ID: mdl-29435611

ABSTRACT

PURPOSE: 17-oxo-DHA is an electrophilic keto-derivative of the omega-3 fatty acid docosahexaenoic acid (DHA) endogenously generated by cyclooxygenase-2 and a cellular dehydrogenase. 17-oxo-DHA displays anti-inflammatory and cytoprotective actions. DHA, alone or in combination with standard chemotherapy, displays antitumor activity. However, the effects of electrophilic keto-derivatives of DHA on cancer growth have never been evaluated. We investigated whether 17-oxo-DHA, alone or in combination with gemcitabine, displayed antitumor effects. Furthermore, we evaluated whether the enzyme 15-prostaglandin dehydrogenase (15-PGDH) was required for transducing the antitumor effects of DHA. METHODS: A panel of five histologically different human non-small cell lung cancer (NSCLC) cell lines was used. Cells were treated with 17-oxo-DHA and gemcitabine, alone or in combination, and apoptosis, proliferation, Fas and FasL expression (mRNA and protein) and active caspase-3/7 and -8 were assessed. Furthermore, an inhibitor of 15-PGDH was used to test the involvement of this enzyme in mediating the antitumor effects of DHA. RESULTS: 17-oxo-DHA (50 µM, 72 h) significantly reduced proliferation, increased cell apoptosis, Fas and FasL expression as well as active caspase-8 and -3/7. When 17-oxo-DHA was given in combination with gemcitabine, stronger effects were observed compared to gemcitabine alone. The enzyme 15-PGDH was required for DHA to promote its full anti-apoptotic effect suggesting that enzymatically generated keto-derivatives of DHA mediate its antitumor actions. CONCLUSIONS: Data herein provided, demonstrate that 17-oxo-DHA displays antitumor effects in NSCLC cell lines. Of note, the combination of 17-oxo-DHA plus gemcitabine, resulted in stronger anticancer effects compared to gemcitabine alone.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Deoxycytidine/analogs & derivatives , Fatty Acids, Omega-3/pharmacology , Lung Neoplasms/pathology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Therapy, Combination , Fatty Acids, Omega-3/chemistry , Humans , Lung Neoplasms/drug therapy , Tumor Cells, Cultured , Gemcitabine
3.
J Cell Mol Med ; 22(4): 2272-2282, 2018 04.
Article in English | MEDLINE | ID: mdl-29411515

ABSTRACT

Inflammation and ageing are intertwined in chronic obstructive pulmonary disease (COPD). The histone deacetylase SIRT1 and the related activation of FoxO3 protect from ageing and regulate inflammation. The role of SIRT1/FoxO3 in COPD is largely unknown. This study evaluated whether cigarette smoke, by modulating the SIRT1/FoxO3 axis, affects airway epithelial pro-inflammatory responses. Human bronchial epithelial cells (16HBE) and primary bronchial epithelial cells (PBECs) from COPD patients and controls were treated with/without cigarette smoke extract (CSE), Sirtinol or FoxO3 siRNA. SIRT1, FoxO3 and NF-κB nuclear accumulation, SIRT1 deacetylase activity, IL-8 and CCL20 expression/release and the release of 12 cytokines, neutrophil and lymphocyte chemotaxis were assessed. In PBECs, the constitutive FoxO3 expression was lower in patients with COPD than in controls. Furthermore, CSE reduced FoxO3 expression only in PBECs from controls. In 16HBE, CSE decreased SIRT1 activity and nuclear expression, enhanced NF-κB binding to the IL-8 gene promoter thus increasing IL-8 expression, decreased CCL20 expression, increased the neutrophil chemotaxis and decreased lymphocyte chemotaxis. Similarly, SIRT1 inhibition reduced FoxO3 expression and increased nuclear NF-κB. FoxO3 siRNA treatment increased IL-8 and decreased CCL20 expression in 16HBE. In conclusion, CSE impairs the function of SIRT1/FoxO3 axis in bronchial epithelium, dysregulating NF-κB activity and inducing pro-inflammatory responses.


Subject(s)
Forkhead Box Protein O3/genetics , Inflammation/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Sirtuin 1/genetics , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Chemokine CCL20/genetics , Cigarette Smoking/adverse effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Immunity, Cellular/drug effects , Inflammation/chemically induced , Inflammation/pathology , Interleukin-8/genetics , NF-kappa B/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Nicotiana/adverse effects , Nicotiana/chemistry
4.
J Cell Physiol ; 232(10): 2851-2859, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27925185

ABSTRACT

Histone deacetylase expression/activity may control inflammation, cell senescence, and responses to corticosteroids. Cigarette smoke exposure, increasing oxidative stress, may negatively affect deacetylase expression/activity. The effects of cigarette smoke extracts (CSE), carbocysteine, and beclomethasone dipropionate on chromatin remodeling processes in human bronchial epithelial cells are largely unknown. The present study was aimed to assess the effects of cigarette smoke, carbocysteine, and beclomethasone dipropionate on histone deacetylase 3 (HDAC3) expression/activity, N-CoR (nuclear receptor corepressor) expression, histone acetyltransferases (HAT) (p300/CBP) expression, p-CREB and IL-1 m-RNA expression, neutrophil chemotaxis. Increased p-CREB expression was observed in the bronchial epithelium of smokers. CSE increased p-CREB expression and decreased HDAC3 expression and activity and N-CoR m-RNA and protein expression. At the same time, CSE increased the expression of the HAT, p300/CBP. All these events increased acetylation processes within the cells and were associated to increased IL-1 m-RNA expression and neutrophil chemotaxis. The incubation of CSE exposed cells with carbocysteine and beclomethasone counteracted the effects of cigarette smoke on HDAC3 and N-CoR but not on p300/CBP. The increased deacetylation processes due to carbocysteine and beclomethasone dipropionate incubation is associated to reduced p-CREB, IL-1 m-RNA expression, neutrophil chemotaxis. These findings suggest a new role of combination therapy with carbocysteine and beclomethasone dipropionate in restoring deacetylation processes compromised by cigarette smoke exposure. J. Cell. Physiol. 232: 2851-2859, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Beclomethasone/pharmacology , Bronchi/drug effects , Carbocysteine/pharmacology , E1A-Associated p300 Protein/metabolism , Epithelial Cells/drug effects , Histone Deacetylases/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Smoke/adverse effects , Smoking/adverse effects , Acetylation , Bronchi/enzymology , Bronchi/pathology , Cell Line , Chemotaxis, Leukocyte/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoprotection , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Phosphorylation
5.
Sci Rep ; 6: 37625, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27883019

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by reduced lung function associated with increased local and systemic inflammatory markers, such as TNFα and IL-1ß. Glucocorticoids are used to treat this chronic disease, however their efficacy is low and new drugs are very much required. 17-oxo-DHA is a cyclooxygenase-2-dependent, electrophilic, α,ß-unsaturated keto-derivative of docosahexaenoic acid with anti-inflammatory properties. We evaluated the action of 17-oxo-DHA alone or in combination with the steroid fluticasone propionate (FP) in peripheral blood mononuclear cells (PBMCs) from COPD patients and healthy individuals exposed to lipopolysaccharide. We show that PBMCs from COPD patients released higher levels of TNFα and IL-1ß compared to controls. 17-oxo-DHA displayed strong anti-inflammatory effects. The addition of 17-oxo-DHA in combination with FP showed enhanced anti-inflammatory effects through the modulation of transcriptional and post-transcriptional mechanisms. 17-oxo-DHA, but not FP, was able to suppress the release of mature IL-1ß through inhibition of the NLRP3 inflammasome. Furthermore, 17-oxo-DHA inhibited inflammasome-dependent degradation of the glucocorticoid receptor (GR). Our findings suggest that 17-oxo-DHA in combination with FP or other steroids might achieve higher therapeutic efficacy than steroids alone. Combined treatment might be particularly relevant in those conditions where increased inflammasome activation may lead to GR degradation and steroid-unresponsive inflammation.


Subject(s)
Docosahexaenoic Acids/pharmacology , Fluticasone/pharmacology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Aged , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Case-Control Studies , Caspase 1/metabolism , Cell Line , Cell Separation , Docosahexaenoic Acids/therapeutic use , Enzyme Activation/drug effects , Female , Fluticasone/therapeutic use , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nigericin/pharmacology , Proteolysis/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptors, Glucocorticoid/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Mediators Inflamm ; 2016: 9063842, 2016.
Article in English | MEDLINE | ID: mdl-27298519

ABSTRACT

IL-17A is overexpressed in the lung during acute neutrophilic inflammation. Acetylcholine (ACh) increases IL-8 and Muc5AC production in airway epithelial cells. We aimed to characterize the involvement of nonneuronal components of cholinergic system on IL-8 and Muc5AC production in bronchial epithelial cells stimulated with IL-17A. Bronchial epithelial cells were stimulated with recombinant human IL-17A (rhIL-17A) to evaluate the ChAT expression, the ACh binding and production, the IL-8 release, and the Muc5AC production. Furthermore, the effectiveness of PD098,059 (inhibitor of MAPKK activation), Bay11-7082 (inhibitor of IkBα phosphorylation), Hemicholinium-3 (HCh-3) (choline uptake blocker), and Tiotropium bromide (Spiriva®) (anticholinergic drug) was tested in our in vitro model. We showed that rhIL-17A increased the expression of ChAT, the levels of ACh binding and production, and the IL-8 and Muc5AC production in stimulated bronchial epithelial cells compared with untreated cells. The pretreatment of the cells with PD098,059 and Bay11-7082 decreased the ChAT expression and the ACh production/binding, while HCh-3 and Tiotropium decreased the IL-8 and Muc5AC synthesis in bronchial epithelial cells stimulated with rhIL-17A. IL-17A is involved in the IL-8 and Muc5AC production promoting, via NFκB and ERK1/2 pathway activation, the synthesis of ChAT, and the related activity of autocrine ACh in bronchial epithelial cells.


Subject(s)
Acetylcholine/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interleukin-17/pharmacology , Interleukin-8/metabolism , Mucin 5AC/metabolism , NF-kappa B/metabolism , Autocrine Communication/drug effects , Bronchi/cytology , Cell Line , Flow Cytometry , Humans , MAP Kinase Signaling System/drug effects , Reverse Transcriptase Polymerase Chain Reaction
7.
Mediators Inflamm ; 2016: 8727289, 2016.
Article in English | MEDLINE | ID: mdl-27212811

ABSTRACT

IL-17A is involved in the activation of oxidative stress and inflammation in nasal epithelial cells. Hyaluronan (HA) in its high molecular weight form (HMW-HA) shows anti-inflammatory responses in contrast to low and medium molecular weight HA (LMW-HA and MMW-HA). The aim of this study was to investigate the pro- or anti-inflammatory biologic function of HA at different molecular weight in an in vitro model of nasal inflammation IL-17A mediated. We evaluated the ERK1/2 and IκBα phosphorylation, NF-κB signal pathway activation, ROS production, IL-8 and NOX-4 protein, and mRNA levels, in nasal epithelial cells RPMI 2650 stimulated with recombinant human (rh) IL-17A. Furthermore, the cells were treated with HMW-HA, MMW-HA, LMW-HA, and U0126. Our results showed that rhIL-17A increased the ERK1/2, IκBα phosphorylation and NF-κB signal pathway activation, ROS production, IL-8 and NOX-4 proteins, and mRNA levels. The addiction of HMW-HA or U0126 showed a significant downregulatory effect on inflammation due to the rhIL-17A stimulation in nasal epithelial cells. IL-17A is able to generate oxidative stress and inflammation via the activation of ERK1/2/NF-κB pathway in nasal epithelial cells. The HMW-HA might represent a coadjuvant of the classic anti-inflammatory/antioxidative treatment of nasal epithelial cells during IL-17A nasal inflammation.


Subject(s)
Epithelial Cells/metabolism , Hyaluronic Acid/metabolism , Inflammation/metabolism , Nasal Mucosa/cytology , Cell Line , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
8.
Life Sci ; 152: 107-16, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27038884

ABSTRACT

AIMS: IL-17A plays a key role in the persistence of airway inflammation, oxidative stress, and reduction of steroid-sensitivity in COPD. We studied the effect of IL-17A on chromatin remodeling and IL-8 production. MAIN METHODS: We measured the levels of IL-8 and IL-17A in induced sputum supernatants (ISS) from healthy controls (HCs), healthy smokers (HSs), and COPD patients by enzyme-linked immunosorbent assay (ELISA). A human bronchial epithelial cell line (16HBE) was stimulated with ISS from HCs, HSs, or COPD subjects. IL-8 was evaluated in 16HBE by Western blot and real-time polymerase chain reaction (PCR). Histone deacetylase 2 (HDAC2), acetyl histone H3 (Ac-His H3) (k9) and inhibitor kappa kinase alpha (IKKα) levels were evaluated in the nuclear extract by Western blot. Finally, we evaluated the effect of IL-17A depletion in ISS, the silencing of IKKα, and the anti-inflammatory effects of Tiotropium Spiriva® (100nM) on 16HBE. KEY FINDINGS: IL-8 and IL-17A levels were higher in ISS from COPD patients and HSs than from HCs. IL-8 protein and messenger RNA (mRNA) levels were increased in 16HBE stimulated with ISS from COPD patients compared with untreated cells. Furthermore, ISS from COPD patients reduced the nuclear levels of HDAC2 while increasing the activity of both Ac-His H3 (k9) and IKKα in stimulated 16HBE. IL-17A depletion in ISS and the IKKα silencing in 16HBE significantly increased the nuclear levels of HDAC2, reduced Ac-His H3 (k9), and promoted IL-8 synthesis in stimulated 16HBE. Tiotropium controls the proinflammatory activity generated by ISS from COPD patients in 16HBE. SIGNIFICANCE: IL-17A present in the airway of COPD patients, which induces chromatin remodeling, promotes the release of IL-8 in the bronchial epithelium. Tiotropium is able to control this proinflammatory activity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chromatin Assembly and Disassembly/drug effects , Epithelial Cells/metabolism , Interleukin-17/metabolism , Interleukin-8/metabolism , Tiotropium Bromide/pharmacology , Bronchi/cytology , Bronchi/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Histone Deacetylase 2/metabolism , Histones/metabolism , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Smoking/metabolism , Sputum/metabolism
9.
Article in English | MEDLINE | ID: mdl-26926362

ABSTRACT

We studied the role of PGE2, its biosynthetic enzymes and its receptors, in regulating the functions of lung fibroblasts through the production of Vascular Endothelial Growth Factor (VEGF) and Interleukin-8 (IL-8) in COPD subjects. Lung fibroblasts from Control (C) (n=6), Smoker (HS) (n=6) and COPD patients (n=8) were cultured, and basal PGE2, VEGF, and IL-8 measured in supernatants by ELISA. COX-1/COX-2 and EP receptors expression were assessed by western blot and by RT-PCR. Release of VEGF and IL-8 by human fetal lung fibroblasts (HFL-1; lung, diploid, human) was evaluated under different conditions. PGE2, VEGF, and IL-8 levels, COX-2, EP2, and EP4 protein expression and mRNA were increased in COPD when compared to Controls. Low concentrations of synthetic PGE2 increased the release of VEGF in HFL-1, but higher concentrations were needed to induce the release of IL-8. This effect was mimicked by an EP2 agonist and modulated by an EP4 antagonist. In the airways of COPD subjects, fibroblast-derived PGE2 may regulate angiogenesis and inflammation through the production of VEGF and IL-8 respectively, suggesting that the increase in expression of COX-2, EP2 and EP4 observed in COPD fibroblasts may contribute to steering the role of PGE2 from homeostatic to pro-inflammatory.


Subject(s)
Dinoprostone/pharmacology , Fibroblasts/drug effects , Interleukin-8/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Vascular Endothelial Growth Factor A/metabolism , Aged , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Humans , Interleukin-8/genetics , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Vascular Endothelial Growth Factor A/genetics
10.
Cytokine ; 76(2): 391-402, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26198032

ABSTRACT

Cigarette smoke is a risk factor for Chronic Obstructive Pulmonary Disease (COPD). Th-17 cytokines are involved in the pathogenesis of COPD. We aimed to evaluate the role of cigarette smoke on the expression of IL-17A, IL-17F and IL-17R in airways of COPD patients. Epithelial and subepithelial immunoreactivity for IL-17A, IL-17F and IL-17R was assessed in surgical specimens from COPD patients (n=15) and from healthy subjects (HC) (n=10) by immunohistochemistry. In vitro, human epithelial cell line 16HBE and A549 as well as PBMC from normal donors were stimulated with cigarette smoke extract (CSE) (0%, 2.5%, 5%, 10%) to evaluate the IL-17A, IL-17F and IL-17R expression by flow cytometry. Furthermore, rhIL-17A and CSE stimulation was evaluated on proliferation and apoptosis in 16HBE and in A549. In central and distal airways immunoreactivity for IL-17A, IL-17F and IL-17R significantly increased in the epithelium and IL-17A in the subepithelium from COPD than in HC. In distal airway, immunoreactivity for IL-17F increased in the subepithelium of COPD than in HC. IL-17A immunoreactivity positively correlate with IL-17R and total pack years in the epithelium from central and distal airways of COPD patients. In vitro, CSE stimulation significantly increased IL-17F and IL-17R in 16HBE (2.5%) and A549 (5%) while IL-17A and IL-17F in PBMC (10%). IL-17A and CSE stimulation, rather than CSE or rhIL-17A alone, significantly increased proliferation in 16HBE and apoptosis in A549. Cigarette smoke increases Th17 immunity in lung tissue of COPD patients, promoting the mechanism of proliferation and apoptosis in airway epithelial cells.


Subject(s)
Interleukin-17/metabolism , Lung/metabolism , Nicotiana , Receptors, Interleukin-17/metabolism , Smoke , Aged , Female , Humans , In Vitro Techniques , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism
11.
Mol Cell Biochem ; 396(1-2): 55-65, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25070832

ABSTRACT

Leptin is involved in the lung epithelial homeostasis. Its role in the nasal tract is largely unknown. Allergic rhinitis (AR) is induced by the allergen exposure leading to consequential structural abnormalities in the nasal epithelium. Topical corticosteroids are recommended as first-line therapy in AR. Parietaria pollen is one of the most important allergenic sources in the southern Europe. In vitro, in human nasal epithelial cell line RPMI 2650, we aimed to determine whether allergen stimulation acts on leptin/leptin receptor pathway and how fluticasone furoate (FF) influences this pathway. The effects of the major allergen recombinant Par j 1 (rPar j 1), of FF, of leptin, and of TGF-ß1 on cell proliferation, on leptin/leptin receptor expression and modulation (by clonogenic test, by RT-q-RT-PCR, by immunocytochemistry and by flow-cytometry), and on STAT-3 activation (assessing nuclear translocation by western blot analysis) were assessed. We found that rPar j 1 and TGF-ß1 significantly decreased cell proliferation and down-regulated the leptin/leptin receptor pathway, whereas FF and leptin reverted them, both alone and in combination. Furthermore, rPar j 1 reduced, while leptin and FF increased STAT-3 activation. In conclusion, FF and leptin itself are able to preserve nasal epithelial homeostasis restoring the leptin/leptin receptor pathway altered by rPar j 1 exposure.


Subject(s)
Androstadienes/pharmacology , Leptin/metabolism , Nasal Mucosa/metabolism , Receptors, Leptin/metabolism , Allergens/genetics , Allergens/pharmacology , Cell Line/drug effects , Cell Proliferation/drug effects , Homeostasis/drug effects , Humans , Nasal Mucosa/cytology , Nasal Mucosa/drug effects , Plant Proteins/genetics , Plant Proteins/pharmacology , Protein Transport/drug effects , Receptors, Leptin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Rhinitis, Allergic/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology
12.
Biochim Biophys Acta ; 1842(9): 1630-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24931101

ABSTRACT

Airway epithelium is a regulator of innate immune responses to a variety of insults including cigarette smoke. Cigarette smoke alters the expression and the activation of Toll Like Receptor 4 (TLR4), an innate immunity receptor. IL-33, an alarmin, increases innate immunity Th2 responses. The aims of this study were to explore whether mini-bronchoalveolar lavage (mini-BAL) or sera from smokers have altered concentrations of IL-33 and whether cigarette smoke extracts (CSE) alter both intracellular expression (mRNA and protein) and release of IL-33 in bronchial epithelial cells. The role of TLR4 in the expression of IL-33 was also explored. Mini-BALs, but not sera, from smokers show reduced concentrations of IL-33. The expression of IL-33 was increased also in bronchial epithelium from smokers. 20% CSE reduced IL-33 release but increased the mRNA for IL-33 by real time PCR and the intracellular expression of IL-33 in bronchial epithelial cells as confirmed by flow cytometry, immunocytochemistry and western blot analysis. The effect of CSE on IL-33 expression was also observed in primary bronchial epithelial cells. IL-33 expression was mainly concentrated within the cytoplasm of the cells. LPS, an agonist of TLR4, reduced IL-33 expression, and an inhibitor of TLR4 increased the intracellular expression of IL-33. In conclusion, the release of IL-33 is tightly controlled and, in smokers, an altered activation of TLR4 may lead to an increased intracellular expression of IL-33 with a limited IL-33 release.


Subject(s)
Bronchi/metabolism , Interleukins/metabolism , Respiratory Mucosa/metabolism , Smoke/adverse effects , Blotting, Western , Bronchi/drug effects , Bronchi/pathology , Bronchoalveolar Lavage , Cell Proliferation , Cells, Cultured , Flow Cytometry , Humans , Immunity, Innate/immunology , Immunoenzyme Techniques , Interleukin-33 , Lipopolysaccharides/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism
13.
Eur J Pharmacol ; 736: 35-43, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24797786

ABSTRACT

Cigarette smoke extract (CSE) affects the expression of Choline Acetyl-Transferase (ChAT), muscarinic acetylcholine receptors, and mucin production in bronchial epithelial cells. Mucin 5AC (MUC5AC), muscarinic acetylcholine receptor M3, ChAT expression, acetylcholine levels and acetylcholine binding were measured in a human pulmonary mucoepidermoid carcinoma cell line (H292) stimulated with CSE. We performed ChAT/RNA interference experiments in H292 cells stimulated with CSE to study the role of ChAT/acetylcholine in MUC5AC production. The effects of Hemicholinium-3 (HCh-3) (50 µM) (a potent and selective choline uptake blocker) and Tiotropium bromide (Spiriva(®)) (100 nM), alone or in combination with Salmeterol (SL) and Fluticasone propionate (FP), were tested in this model. MUC5AC, muscarinic acetylcholine receptor M3, ChAT, acetylcholine expression and acetylcholine binding significantly increased in H292 cells stimulated with CSE (5%) compared to untreated cells. HCh-3 reduced acetylcholine binding and MUC5AC production in H292 cells stimulated with CSE. ChAT/RNA interference eliminated the effect of CSE on MUC5AC production. FP reduced ChAT and acetylcholine binding in unstimulated cells, while showing a partial effect in CSE stimulated cells. SL increased the ChAT expression and acetylcholine binding in H292 cells stimulated with or without CSE. Tiotropium, alone or together with FP and SL, reduced acetylcholine binding and MUC5AC production in H292 cells stimulated with CSE. CSE affects the ChAT/acetylcholine expression, increasing MUC5AC production in H292 cells. Pharmacological treatment with anticholinergic drugs reduces the secretion of MUC5AC generated by autocrine acetylcholine activity in airway epithelial cells.


Subject(s)
Acetylcholine/metabolism , Choline O-Acetyltransferase/metabolism , Complex Mixtures/pharmacology , Mucin 5AC/metabolism , Nicotiana , Smoke , Albuterol/analogs & derivatives , Albuterol/pharmacology , Androstadienes/pharmacology , Bronchi/cytology , Bronchodilator Agents/pharmacology , Cell Line, Tumor , Choline O-Acetyltransferase/genetics , Cholinergic Antagonists/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fluticasone , Hemicholinium 3/pharmacology , Humans , Neurotransmitter Uptake Inhibitors/pharmacology , RNA Interference , Receptor, Muscarinic M3/metabolism , Salmeterol Xinafoate , Scopolamine Derivatives/pharmacology , Tiotropium Bromide
14.
Biochim Biophys Acta ; 1840(7): 2299-309, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24594225

ABSTRACT

BACKGROUND: 17-Oxo-DHA is an endogenous electrophilic derivative of the omega-3 fatty acid docosahexaenoic acid (DHA) which is generated in activated macrophages by the action of cyclooxygenase-2. METHODS: The ability of 17-oxo-DHA to control inflammation and oxidative stress was tested in human macrophages (THP-1) and bronchial epithelial cell line (16HBE) stimulated with cigarette smoke extract (CSE) and lipopolysaccharide (LPS). All data were further confirmed using primary bronchial epithelial cells, alveolar macrophages and peripheral blood mononuclear cells. RESULTS: 17-Oxo-DHA was a strong inducer of the anti-oxidant response promoting Nrf2 nuclear accumulation, leading to the expression of heme oxygenase 1 and more than doubling glutathione levels. This resulted in suppression of CSE-induced ROS generation in macrophages. In macrophages, 17-oxo-DHA potently suppressed TNFα release in response to LPS, CSE and IL-1ß acting at transcriptional level via a mechanism independent of Nrf2. Externally supplemented 17-oxo-DHA displayed the same effects in the presence of the Cox-inhibitor indomethacin. The non-electrophilic 17-oxo-DHA precursor DHA did not show any biological actions, indicating that the electrophilic moiety was required for this compound to become bioactive. CONCLUSIONS: 17-Oxo-DHA promotes cytoprotective actions both in immune and structural cells. In immune cells, 17-oxo-DHA is effective in contrasting CSE- and LPS-induced oxidative damage and inflammation acting via multiple independent pathways. GENERAL SIGNIFICANCE: Herein we provide insights on how the novel endogenous electrophilic DHA-derivative 17-oxo-DHA promotes anti-oxidant and anti-inflammatory actions. Data herein reported indicate that 17-oxo-DHA is an attractive lead compound for the development of new treatments for cigarette smoke-related airway inflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/metabolism , Docosahexaenoic Acids/pharmacology , Inflammation/drug therapy , Antioxidants/pharmacology , Cell Line , Cyclooxygenase 2/genetics , Docosahexaenoic Acids/analogs & derivatives , Epithelial Cells/drug effects , Humans , Inflammation/chemically induced , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/toxicity , Macrophages/drug effects , Smoking/adverse effects
15.
Immunology ; 141(2): 242-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24128051

ABSTRACT

Gemcitabine is a chemotherapy agent commonly used in the treatment of non-small cell lung cancer (NSCLC) that has been demonstrated to induce apoptosis in NSCLC cells by increasing functionally active Fas expression. The aim of this study was to evaluate the Fas/Fas ligand (FasL) system involvement in gemcitabine-induced lung cancer cell killing. NSCLC H292 cells were cultured in the presence or absence of gemcitabine. FasL mRNA and protein were evaluated by real-time PCR, and by Western blot and flow cytometry, respectively. Apoptosis of FasL-expressing cells was evaluated by flow cytometry, and caspase-8 and caspase-3 activation by Western blot and a colorimetric assay. Cytotoxicity of lymphokine-activated killer (LAK) cells and malignant pleural fluid lymphocytes against H292 cells was analysed in the presence or absence of the neutralizing anti-Fas ZB4 antibody, by flow cytometry. Gemcitabine increased FasL mRNA and total protein expression, the percentage of H292 cells bearing membrane-bound FasL (mFasL) and of mFasL-positive apoptotic H292 cells, as well as caspase-8 and caspase-3 cleavage. Moreover, gemcitabine increased CH11-induced caspase-8 and caspase-3 cleavage and proteolytic activity. Cytotoxicity of LAK cells and pleural fluid lymphocytes was increased against gemcitabine-treated H292 cells and was partially inhibited by ZB4 antibody. These results demonstrate that gemcitabine: (i) induces up-regulation of FasL in lung cancer cells triggering cell apoptosis via an autocrine/paracrine loop; (ii) induces a Fas-dependent apoptosis mediated by caspase-8 and caspase-3 activation; (iii) enhances the sensitivity of lung cancer cells to cytotoxic activity of LAK cells and malignant pleural fluid lymphocytes, partially via Fas/FasL pathway. Our data strongly suggest an active involvement of the Fas/FasL system in gemcitabine-induced lung cancer cell killing.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/analogs & derivatives , Fas Ligand Protein/physiology , Lung Neoplasms/drug therapy , fas Receptor/physiology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Deoxycytidine/pharmacology , Fas Ligand Protein/genetics , Humans , Killer Cells, Lymphokine-Activated/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Gemcitabine
16.
Toxicol Lett ; 223(2): 198-204, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24076166

ABSTRACT

Cigarette smoke represents the major risk factor for chronic obstructive pulmonary disease (COPD). Cigarette smoke extracts (CSE) alter TLR4 expression and activation in bronchial epithelial cells. Carbocysteine, an anti-oxidant and mucolytic agent, is effective in reducing the severity and the rate of exacerbations in COPD patients. The effects of carbocysteine on TLR4 expression and on the TLR4 activation downstream events are largely unknown. This study was aimed to explore whether carbocysteine, in a human bronchial epithelial cell line (16-HBE), counteracted some pro-inflammatory CSE-mediated effects. In particular, TLR4 expression, LPS binding, p21 (a senescence marker), IL-8 mRNA and release in CSE-stimulated 16-HBE as well as actin reorganization in neutrophils cultured with supernatants from bronchial epithelial cells which were stimulated with CSE and/or carbocysteine were assessed. TLR4 expression, LPS binding, and p21 expression were assessed by flow cytometry, IL-8 mRNA by Real Time PCR and IL-8 release by ELISA. Actin reorganization, a prerequisite for cell migration, was determined using Atto 488 phalloidin in neutrophils by flow cytometry and fluorescence microscopy. CSE increased: (1) TLR4, LPS binding and p21 expression; (2) IL-8 mRNA and IL-8 release due to IL-1 stimulation; (3) neutrophil migration. Carbocysteine in CSE stimulated bronchial epithelial cells, reduced: (1) TLR4, LPS binding and p21; (2) IL-8 mRNA and IL-8 release due to IL-1 stimulation; (3) neutrophil chemotactic migration. In conclusion, the present study provides compelling evidences that carbocysteine may contribute to control the inflammatory and senescence processes present in smokers.


Subject(s)
Antioxidants/pharmacology , Carbocysteine/pharmacology , Epithelial Cells/drug effects , Immunity, Innate/drug effects , Smoking/adverse effects , Aging/drug effects , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Bronchi/cytology , Bronchi/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Neutrophils/cytology , Neutrophils/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
17.
Biochim Biophys Acta ; 1832(12): 1949-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23811074

ABSTRACT

The induction of nitric oxide synthase (iNOS) expression via the signal transducer and activator of transcription 1 (STAT-1) is involved in the mechanism of oxidative/nitrosative stress. We investigated whether acetylcholine (ACh) generates oxidative/nitrosative stress in bronchial epithelial cells during airway inflammation of COPD and evaluated the effects of Tiotropium, a once-daily antimuscarinic drug, and Olodaterol, a long-acting ß2-agonist on these mechanisms. Human bronchial epithelial cells (16-HBE) were stimulated (4h, 37°C) with induced sputum supernatants (ISSs) from healthy controls (HC) (n=10), healthy smokers (HS) (n=10) or COPD patients (n=10), as well as with ACh (from 1µM to 100µM). The activation of STAT-1 pathway (STAT-1Ser727 and STAT-1Tyr701) and iNOS was evaluated in the cell lysates by Western blot analysis as well as nitrotyrosine levels by ELISA, while reactive oxygen species (ROS) were evaluated by flow cytometry. Finally, the effect of Tiotropium (Spiriva®) (100nM), alone or in combination with Olodaterol (1nM), was tested in this model. ISSs from COPD patients significantly increased the phosphorylation of STAT-1Ser727 and STAT-1Tyr701, iNOS and ROS/Nitrotyrosine when compared with ISSs from HC or HS subjects in 16-HBE cells. Furthermore, synthetic ACh increased all these parameters in stimulated 16HBE when compared with untreated cells. Tiotropium and Olodaterol reduced the oxidative/nitrosative stress generated by ACh and ISSs. We concluded that ACh mediated the oxidative/nitrosative stress involving the STAT-1 pathway activation in human bronchial epithelial cells during COPD. ß2-Long acting and antimuscarinic drugs, normally used in the treatment of COPD as bronchodilator, might be able to control these cellular events.


Subject(s)
Acetylcholine/pharmacology , Bronchi/drug effects , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Pulmonary Disease, Chronic Obstructive/pathology , STAT1 Transcription Factor/metabolism , Blotting, Western , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Cholinergic Agonists/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Male , Middle Aged , Nitric Oxide Synthase Type II/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism
18.
Biochim Biophys Acta ; 1822(7): 1079-89, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22440430

ABSTRACT

We quantified TGF-ß1 and acetylcholine (ACh) concentrations in induced sputum supernatants (ISSs) from 18 healthy controls (HC), 22 healthy smokers (HS) and 21 COPDs. ISSs from HC, HS and COPD as well as rhTGF-ß1 were also tested in neutrophil adhesion and in mAChR2, mAChR3 and ChAT expression experiments in human bronchial epithelial cells (16-HBE). Finally, we evaluated the effects of Olodaterol (a novel inhaled ß(2)-adrenoceptor agonist) and Tiotropium Spiriva®, alone or in combination, on neutrophil adhesion and mAChRs and ChAT expression in stimulated 16-HBE. The results showed that 1) TGF-ß1 and ACh concentrations are increased in ISSs from COPD in comparison to HC and HS, and TGF-ß1 in HS is higher than in HC; 2) ISSs from COPD and HS caused increased neutrophil adhesion to 16-HBE when compared to ISSs from HC. The effect of ISSs from COPD was significantly reduced by TGF-ß1 depletion or by the pretreatment with Olodaterol or Tiotropium alone or in combination, while the effect of ISSs from HS was significantly reduced by the pretreatment with Olodaterol alone; 3) mAChR2, mAChR3 and ChAT expression was increased in 16-HBE stimulated with ISSs from COPD and TGF-ß1 depletion significantly reduced this effect on mAChR3 and ChAT expression; 4) rhTGF-ß1 increased mAChR2, mAChR3 and ChAT expression in 16-HBE; 5) Olodaterol did not affect the expression of mAChRs and ChAT in 16-HBE. Our findings support the use of ß2 long-acting and anticholinergic drugs to control the bronchoconstriction and TGF-ß1-mediated neutrophilic inflammation in COPD.


Subject(s)
Acetylcholine/metabolism , Adrenergic beta-2 Receptor Agonists/therapeutic use , Cholinergic Antagonists/therapeutic use , Neutrophils/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Transforming Growth Factor beta1/metabolism , Acetylcholine/analysis , Adrenergic beta-2 Receptor Agonists/pharmacology , Aged , Analysis of Variance , Benzoxazines/pharmacology , Benzoxazines/therapeutic use , Bronchoconstriction/drug effects , Bronchodilator Agents/pharmacology , Bronchodilator Agents/therapeutic use , Case-Control Studies , Cell Adhesion/drug effects , Cell Line, Transformed , Choline O-Acetyltransferase/metabolism , Cholinergic Antagonists/pharmacology , Drug Therapy, Combination , Epithelial Cells/drug effects , Female , Flow Cytometry , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/metabolism , Scopolamine Derivatives/pharmacology , Scopolamine Derivatives/therapeutic use , Smoking/adverse effects , Sputum/chemistry , Tiotropium Bromide , Transforming Growth Factor beta1/analysis
19.
PLoS One ; 7(3): e33601, 2012.
Article in English | MEDLINE | ID: mdl-22438960

ABSTRACT

BACKGROUND: Altered pulmonary defenses in chronic obstructive pulmonary disease (COPD) may promote distal airways bacterial colonization. The expression/activation of Toll Like receptors (TLR) and beta 2 defensin (HBD2) release by epithelial cells crucially affect pulmonary defence mechanisms. METHODS: The epithelial expression of TLR4 and of HBD2 was assessed in surgical specimens from current smokers COPD (s-COPD; n = 17), ex-smokers COPD (ex-s-COPD; n = 8), smokers without COPD (S; n = 12), and from non-smoker non-COPD subjects (C; n = 13). RESULTS: In distal airways, s-COPD highly expressed TLR4 and HBD2. In central airways, S and s-COPD showed increased TLR4 expression. Lower HBD2 expression was observed in central airways of s-COPD when compared to S and to ex-s-COPD. s-COPD had a reduced HBD2 gene expression as demonstrated by real-time PCR on micro-dissected bronchial epithelial cells. Furthermore, HBD2 expression positively correlated with FEV1/FVC ratio and inversely correlated with the cigarette smoke exposure. In a bronchial epithelial cell line (16 HBE) IL-1ß significantly induced the HBD2 mRNA expression and cigarette smoke extracts significantly counteracted this IL-1 mediated effect reducing both the activation of NFkB pathway and the interaction between NFkB and HBD2 promoter. CONCLUSIONS: This study provides new insights on the possible mechanisms involved in the alteration of innate immunity mechanisms in COPD.


Subject(s)
Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Smoking/adverse effects , Smoking/metabolism , beta-Defensins/metabolism , Aged , Base Sequence , Bronchi/metabolism , Bronchi/pathology , Case-Control Studies , Cell Line , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Smoking/genetics , Toll-Like Receptor 4/metabolism , beta-Defensins/genetics
20.
Respir Med ; 105(10): 1491-500, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21612902

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterised by chronic inflammation in airways and lung parenchyma. CD8+ T-lymphocytes, crucial effector and regulatory cells in inflammation, are increased in the central and peripheral airways in COPD. The aim of this study was to assess the role of apoptosis in the accumulation of CD8+ T-lymphocytes within the airway wall in COPD. We examined the submucosa of transverse sections of central and peripheral airways from post-operative tissues from non-smokers (n = 16), smokers with normal lung function (n = 16), smokers with mild/moderate COPD (n = 16), and smokers with severe/very severe COPD (n = 9). TUNEL and immunohistochemistry techniques were used to identify apoptosis and cell phenotype, respectively. The percentage of apoptotic CD8+ T-lymphocytes was significantly lower (p < 0.0001) in smokers with mild/moderate COPD than in non-smokers, smokers with normal lung function, and smokers with severe/very severe COPD, and was positively related to values of FEV(1) and FEV(1)/FVC ratio, both in central and in peripheral airways. These data suggest that reduced apoptosis of CD8+ T-lymphocytes may be an important mechanism that contributes to the accumulation of these cells in the airway submucosa in smokers with mild/moderate COPD.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/pathology , Lung/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking/physiopathology , Analysis of Variance , CD8-Positive T-Lymphocytes/immunology , Female , Forced Expiratory Volume , Humans , Immunohistochemistry , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/immunology , Smoking/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...