Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(8): e0136755, 2015.
Article in English | MEDLINE | ID: mdl-26317859

ABSTRACT

BACKGROUND: Inflammation is a key hallmark of ALI and is mediated through ungoverned cytokine signaling. One such cytokine, interleukin-1beta (IL-1ß) has been demonstrated to be the most bioactive cytokine in ALI patients. Macrophages are the key players responsible for IL-1ß secretion into the alveolar space. Following the binding of IL-1ß to its receptor, "activated" alveolar epithelial cells show enhanced barrier dysfunction, adhesion molecule expression, cytokine secretion, and leukocyte attachment. More importantly, it is an important communication molecule between the macrophage and alveolar epithelium. While the molecular determinants of this inflammatory event have been well documented, endogenous resolution processes that decrease IL-1ß secretion and resolve alveolar epithelial cell activation and tissue inflammation have not been well characterized. Lipid mediator Aspirin-Triggered Resolvin D1 (AT-RvD1) has demonstrated potent pro-resolutionary effects in vivo models of lung injury; however, the contribution of the alveoli to the protective benefits of this molecule has not been well documented. In this study, we demonstrate that AT-RvD1 treatment lead to a significant decrease in oxidant induced macrophage IL-1ß secretion and production, IL-1ß-mediated cytokine secretion, adhesion molecule expression, leukocyte adhesion and inflammatory signaling. METHODS: THP-1 macrophages were treated with hydrogen peroxide and extracellular ATP in the presence or absence of AT-RvD1 (1000-0.1 nM). A549 alveolar-like epithelial cells were treated with IL-1ß (10 ng/mL) in the presence or absence of AT-RvD1 (0.1 µM). Following treatment, cell lysate and cell culture supernatants were collected for Western blot, qPCR and ELISA analysis of pro-inflammatory molecules. Functional consequences of IL-1ß induced alveolar epithelial cell and macrophage activation were also measured following treatment with IL-1ß ± AT-RvD1. RESULTS: Results demonstrate that macrophages exposed to H2O2 and ATP in the presence of resolvins show decreased IL-1ß production and activity. A549 cells treated with IL-1ß in the presence of AT-RvD1 show a reduced level of proinflammatory cytokines IL-6 and IL-8. Further, IL-1ß-mediated adhesion molecule expression was also reduced with AT-RvD1 treatment, which was correlated with decreased leukocyte adhesion. AT-RvD1 treatment demonstrated reduced MAP-Kinase signaling. Taken together, our results demonstrate AT-RvD1 treatment reduced IL-1ß-mediated alveolar epithelial cell activation. This is a key step in unraveling the protective effects of resolvins, especially AT-RvD1, during injury.


Subject(s)
Acute Lung Injury/metabolism , Docosahexaenoic Acids/pharmacology , Epithelial Cells/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Models, Biological , Oxidative Stress/drug effects , Acute Lung Injury/pathology , Cell Line, Tumor , Epithelial Cells/pathology , Humans , Macrophages/pathology , Pulmonary Alveoli , Signal Transduction/drug effects
3.
Biochem Biophys Res Commun ; 426(2): 203-8, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22940131

ABSTRACT

Acute lung injury (ALI) is a devastating disease characterized by pulmonary edema. Removal of edema from the air spaces of lung is a critical function of the epithelial sodium channel (ENaC) in ALI. The molecular mechanisms behind resolution of pulmonary edema are incompletely understood. MicroRNA's (miRNA) are crucial gene regulators and are dysregulated in various diseases including ALI. Recent studies suggest that microRNA-16 (miR-16) targets serotonin transporter (SERT) involved in the serotonin (5-HT) transmitter system. Alterations in serotonin levels have been reported in various pulmonary diseases. However, the role of miR-16 on its target SERT, and ENaC, a key ion channel involved in the resolution of pulmonary edema, have not been studied. In the present study, the expression patterns of miR-16, SERT, ENaC and serotonin were investigated in mice exposed to room air and hyperoxia. The effects of miR-16 overexpression on ENaC, SERT, TGF-ß and Nedd4 in human alveolar epithelial cells were analyzed. miR-16 and ENaC were downregulated in mice exposed to hyperoxia. miR-16 downregulation in mouse lung was correlated with an increase in SERT expression and pulmonary edema. Overexpression of miR-16 in human alveolar epithelial cells (A549) suppressed SERT and increased ENaCß levels when compared to control-vector transfected cells. In addition, miR-16 over expression suppressed TGFß release, a critical inhibitor of ENaC. Interestingly Nedd4, a negative regulator of ENaC remained unaltered in miR-16 over expressed A549 cells when compared to controls. Taken together, our data suggests that miR-16 upregulates ENaC, a major sodium channel involved in resolution of pulmonary edema in ALI.


Subject(s)
Acute Lung Injury/metabolism , Epithelial Sodium Channels/metabolism , MicroRNAs/metabolism , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Aerobiosis , Animals , Cell Line, Tumor , Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Sodium Channels/genetics , Humans , Mice , MicroRNAs/genetics , Nedd4 Ubiquitin Protein Ligases , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Transforming Growth Factor alpha/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
PLoS One ; 7(5): e37712, 2012.
Article in English | MEDLINE | ID: mdl-22662197

ABSTRACT

INTRODUCTION: Pulmonary Arterial Hypertension (PAH) is a progressively devastating disease characterized by excessive proliferation of the Pulmonary Arterial Smooth Muscle Cells (PASMCs). Studies suggest that PAH and cancers share an apoptosis-resistant state featuring excessive cell proliferation. The proliferation of cancer cells is mediated by increased expression of Enhancer of Zeste Homolog 2 (EZH2), a mammalian histone methyltransferase that contributes to the epigenetic silencing of target genes. However, the role of EZH2 in PAH has not been studied. In this study, it is hypothesized that EZH2 could play a role in the proliferation of PASMCs. METHODS: In the present study, the expression patterns of EZH2 were investigated in normal and hypertensive mouse PASMCs. The effects of EZH2 overexpression on the proliferation of human PASMCs were tested. PASMCs were transfected with EZH2 or GFP using nucleofector system. After transfection, the cells were incubated for 48 hours at 37°C. Proliferation and cell cycle analysis were performed using flow cytometry. Apoptosis of PASMCs was determined using annexin V staining and cell migration was tested by wound healing assay. RESULTS: EZH2 protein expression in mouse PASMCs were correlated with an increase in right ventricular systolic pressure and Right Ventricular Hypertrophy (RVH). The overexpression of EZH2 in human PASMCs enhances proliferation, migration, and decrease in the rate of apoptosis when compared to GFP-transfected cells. In the G2/M phase of the EZH2 transfected cells, there was a 3.5 fold increase in proliferation, while there was a significant decrease in the rate of apoptosis of PASMCs, when compared to control. CONCLUSION: These findings suggest that EZH2 plays a role in the migration and proliferation of PASMCs, which is a major hallmark in PAH. It also suggests that EZH2 could play a role in the development of PAH and can serve as a potential target for new therapies for PAH.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Polycomb Repressive Complex 2/metabolism , Pulmonary Artery/metabolism , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Gene Expression , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypoxia , Mice , Mice, Inbred C57BL , Phenotype , Polycomb Repressive Complex 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...