Gallotannin Inhibits the Expression of Chemokines and Inflammatory Cytokines in A549 Cells

2005 ◽  
Vol 68 (3) ◽  
pp. 895-904 ◽  
Author(s):  
Katalin Erdèlyi ◽  
Andrea Kiss ◽  
Edina Bakondi ◽  
Péter Bai ◽  
Csaba Szabó ◽  
...  
2020 ◽  
Author(s):  
Ling Zheng ◽  
Jun Fei ◽  
Zheng Xu ◽  
Chun-Mei Feng ◽  
Se-Ruo Li ◽  
...  

Abstract Background and Objectives Limited studies suggested that calprotectin may take part in the pathophysiology of community-acquired pneumonia (CAP). Nevertheless, there is no clinical study to analyze the role of S100A8 in CAP patients. The objective of this study was to analyze the association of serum S100A8 with the severity of CAP based on a cross-sectional study. Methods Entire 200 CAP patients and 100 normal subjects were recruited. Demographic data, clinical information and serum were collected on admission. Serum S100A8 and inflammatory cytokines were detected. Results Serum S100A8 was increased in CAP patients on admission. Serum S100A8 was gradually increased in parallel with the CAP severity scores. Serum S100A8 was positively correlated with CAP severity scores (CURB-65, CRB-65, PSI, CURXO and SMART-COP), blood routine parameters (WBC, neutrophil-lymphocyte ratio and monocyte-lymphocyte ratio) and inflammatory cytokines (TNFα, IL-1β and CRP). Furtherly, univariate and multivariate logistical regression analysis revealed that there was a positive association between serum S100A8 with CRB-65, PSI and CURXO. Moreover, the predictive capacity of serum S100A8 was performed by receiver operating characteristic area under the curve (AUC) analysis. The AUCs of S100A8 for CAP and CAP severity were 0.855 and 0.893, respectively. Mechanistic analysis found that S100A8 knockdown alleviated streptococcus pneumoniae-evoked inflammatory cytokines in A549 cells. Conclusion Serum S100A8 on admission was positively associated with the severity of CAP. S100A8 knockdown alleviates streptococcus pneumoniae- evoked inflammatory cytokines in A549 cells, indicating that S100A8 may exert an important role in the pathophysiology of CAP and be an early serum diagnostic biomarker for CAP.


Immunobiology ◽  
1998 ◽  
Vol 199 (1) ◽  
pp. 14-22 ◽  
Author(s):  
Michael Stoeck ◽  
Wolfgang Kromer ◽  
Volker Gekeler

2021 ◽  
Vol 27 ◽  
Author(s):  
Vesara A. Gatera ◽  
Ronny Lesmana ◽  
Ida Musfiroh ◽  
Raden Tina D. Judistiani ◽  
Budi Setiabudiawan ◽  
...  

2019 ◽  
Vol 2019 ◽  
pp. 1-10 ◽  
Author(s):  
Qian-Lu Wang ◽  
Lei Yang ◽  
Yue Peng ◽  
Min Gao ◽  
Ming-Shi Yang ◽  
...  

Objectives. To investigate the protective effect of ginsenoside Rg1 on relieving sepsis-induced lung inflammation and injury in vivo and in vitro. Methods. Cultured human pulmonary epithelial cell line A549 was challenged with LPS to induce cell injury, and CLP mouse model was generated to mimic clinical condition of systemic sepsis. Rg1 was applied to cells or animals at indicated dosage. Apoptosis of cultured cells was quantified by flow cytometry, along with ELISA for inflammatory cytokines in supernatant. For septic mice, lung tissue pathology was examined, plus ELISA assay for serum cytokines. Western blotting was used to examine the activation of inflammatory pathways and ER stress marker proteins in both cells and mouse lung tissues. Reactive oxygen species (ROS) level was quantified by DCFDA kit. Results. Ginsenoside Rg1 treatment remarkably suppressed apoptosis rate of LPS-induced A549 cells, relieved mouse lung tissue damage, and elevated survival rate. Rg1 treatment also rescued cells from LPS-induced intracellular ROS. In both A549 cells and mouse lung tissues, further study showed that Rg1 perfusion significantly suppressed the secretion of inflammatory cytokines including tumor necrosis factor- (TNF-) alpha and interleukin- (IL-) 6 and relieved cells from ER stress as supported by decreased expression of marker proteins via upregulating sirtuin 1 (SIRT1). Conclusion. Our results showed that ginsenoside Rg1 treatment effectively relieved sepsis-induced lung injury in vitro and in vivo, mainly via upregulating SIRT1 to relieve ER stress and inflammation. These findings provide new insights for unrevealing potential candidate for severe sepsis accompanied with lung injury.


2011 ◽  
Vol 300 (4) ◽  
pp. L548-L559 ◽  
Author(s):  
Stephen T. Buckley ◽  
Carlos Medina ◽  
Michael Kasper ◽  
Carsten Ehrhardt

Fibrosis of the lung is characterized by the accumulation of myofibroblasts, a key mediator in the fibrogenic reaction. Cumulative evidence indicates that epithelial-mesenchymal transition (EMT), a process whereby epithelial cells become mesenchyme-like, is an important contributing source for the myofibroblast population. Underlying this phenotypical change is a dramatic alteration in cellular structure. The receptor for advanced glycation end-products (RAGE) has been suggested to maintain lung homeostasis by mediating cell adhesion, while the family of ezrin/radixin/moesin (ERM) proteins, on the other hand, serve as an important cross-linker between the plasma membrane and cytoskeleton. In the present investigation, we tested the hypothesis that RAGE and ERM interact and play a key role in regulating EMT-associated structural changes in alveolar epithelial cells. Exposure of A549 cells to inflammatory cytokines resulted in phosphorylation and redistribution of ERM to the cell periphery and localization with EMT-related actin stress fibers. Simultaneously, blockade of Rho kinase (ROCK) signaling attenuated these cytokine-induced structural changes. Additionally, RAGE expression was diminished after cytokine stimulation, with release of its soluble isoform via a matrix metalloproteinase (MMP)-9-dependent mechanism. Immunofluorescence microscopy and coimmunoprecipitation revealed association between ERM and RAGE under basal conditions, which was disrupted when challenged with inflammatory cytokines, as ERM in its activated state complexed with membrane-linked CD44. Dual-fluorescence immunohistochemistry of patient idiopathic pulmonary fibrosis (IPF) tissues highlighted marked diminution of RAGE in fibrotic samples, together with enhanced levels of CD44 and double-positive cells for CD44 and phospho (p)ERM. These data suggest that dysregulation of the ERM-RAGE complex might be an important step in rearrangement of the actin cytoskeleton during proinflammatory cytokine-induced EMT of human alveolar epithelial cells.


2020 ◽  
Vol 11 ◽  
Author(s):  
Lei Zhang ◽  
Wen-Xu Chen ◽  
Ling-Li Li ◽  
Yu-Zhu Cao ◽  
Ya-Di Geng ◽  
...  

Background: Targeting inflammatory microenvironment is a promising anti-tumor strategy. Paeonol is a phenolic compound with effective anti-inflammatory and anti-tumor properties. However, the effects of paeonol on non-small cell carcinoma (NSCLC) have not been fully investigated. Here, we evaluated the effects of paeonol on proliferation and metastasis of NSCLC and elucidated the underlying mechanisms.Methods: The effects of paeonol on inflammatory cytokines were determined by cell proliferation and ELISA assays. Assays of wound healing, single cell migration and perforation invasion were used to evaluate migration and invasion of NSCLC cells. Expression of marker proteins in epithelial-mesenchymal transition (EMT) and matrix metalloproteinase (MMP) family enzymes were detected by Western blot assays. Nude mouse A549 cells transplantation tumor model was used to study the anti-lung cancer effects of paeonol in vivo. TUNEL stanining were used to detect the apoptosis of tumor cells in A549 lung cancer mice, and Ki67 analysis was used to detect the proliferation of tumor cells in A549 lung cancer mice. Immunohistochemistry was used to detect the effects of paeonol on signaling molecules in tumor tissues.Results: Paeonol inhibited A549 cancer cell migration and invasion in vitro. Paeonol inhibited secreaion of inflammatory cytokines in A549 cells, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, and transforming growth factor (TGF)-β. Paeonol altered the expression of marker proteins involved in EMT and MMP family enzymes. In addition, paeonol inhibited the transcriptional activity of nuclear factor-κB (NF-κB) and phosphorylation of signal transducers and activators of transcription 3 (STAT3). Paeonol inhibited the growth of A549 cells transplanted tumors in nude mice.Conclusion: Paeonol potently inhibited NSCLC cell growth, migration and invasion associated with disruption of STAT3 and NF-κB pathways, suggesting that it could be a promising anti-metastatic candidate for tumor chemotherapy.


2020 ◽  
Author(s):  
Ye Chen ◽  
Chao Zhang ◽  
Changxue Xiao ◽  
Xiao-dong Li ◽  
Zhi-li Hu ◽  
...  

Abstract Objective: To investigate lncRNAs and their roles in regulating the inflammatory response under treatment of Dexamethasone (Dex) in asthma.Methods: IL-1 beta (10 ng/ml) and LPS (1 μg/ml) was used to induce an inflammatory cell model with A549 cells, and the results showed that IL-1 beta performed better against LPS. Dex with different concentration was used to attenuate inflammation by IL-1 beta, and its effect was assessed by RT-PCR to detect the inflammatory related mRNA, including IKbeta-alpha, IKKbeta, IL-6, IL-8, and TNF-alpha and ELISA to detect the inflammatory cytokines TNF-alpha, IL-6 and IL-8. RT-PCR was used to quantify levels of lncRNAs, including lncMALAT1, lncHotair, lncH19, and lncNeat1. lncH19 was most closely correlated with the inflammatory response, which was induced by IL-1beta and attenuated by Dex, Among the lncRNAs, the level of lncH19 exhibited the highest increase following treatment with 1 μM and 10 μM Dex. Therefore, lncH19 was selected for further function study. lncH19 expression was inhibited by shRNA transduced by lentivirus. Cell assays for cell proliferation and apoptosis as well as RT-PCR, western blot, and ELISA for inflammatory related genes were conducted to confirm the functions of lncH19. Predicted target miRNAs of lncH19 included the following: hsa-miR-346, hsa-miR-324-3p, hsa-miR-18a-3p, hsa-miR-18b-5p, hsa-miR-146b-3p, hsa-miR-19b-3p, and hsa-miR-19a-3p. Following estimation by RT-PCR, hsa-miR-346, hsa-miR-18a-3p, hsa-miR-324-3p showed consistent patterns in A549 NC and A549 shlncH19. miRNA inhibitor was transfected into A549 NC and A549 shlncH19 cells, and expression levels were determined by RT-PCR. hsa-miR-324-3p was inhibited the most relative to hsa-miR-346 and hsa-miR-18a-3p and was subjected to further function study. RT-PCR, ELISA and Western Blot for inflammatory related genes detection were conducted to validate the functions of the target hsa-miR-324-3p. Results: Dex with 1μM and 10 μM were shown to be effective in attenuating the inflammatory response. During this process, lncH19 significantly increased in expression (P < 0.05). Dex with 1μM was for further study. Under IL-1 beta treatment with or without Dex, the inhibition of lncH19 lead to an increase cell proliferation, a decrease in cell apoptosis, an increase in the protein level of inflammatory-related genes, and the phosphorylation of P65, ICAM-1, VCAM-1, and inflammatory cytokines. Following prediction of the targets of lncH19 and validation by RT-PCR, miR-346, miR-18a-3p, and miR-324-3p were found to be negatively correlated to lncH19. Additionally, Dex increased the expression of lncH19, but the expression of the aforementioned miRNAs was reduced. Among miRNAs, miR-324-3p was the most markedly down-regulated following treatment of miRNA inhibitors. The MTS assay and cell apoptosis assay showed that the miR-324-3p inhibitor inhibited cell proliferation and induced cell apoptosis, thereby significantly attenuating the inflammatory response, which reversed the effect of lncH19 in regulating cell proliferation and the secretion of inflammatory cytokines (P < 0.05). Therefore, lncH19 might regulate miR-324-3p during Dex treatment in inflammatory response of asthma.Conclusion: Dex can attenuate the inflammatory response in asthma via regulation of the lncH19/miR-324-3p cascade.


2020 ◽  
Author(s):  
Ye Chen ◽  
Chao Zhang ◽  
Changxue Xiao ◽  
Xiao-dong Li ◽  
Zhi-li Hu ◽  
...  

Abstract Objective: To investigate lncRNAs and their roles in regulating the pulmonary inflammatory response under treatment of Dexamethasone (Dex).Methods: IL-1β (10 ng/mL) and LPS (1 μg/mL) was used to induce an inflammatory cell model with A549 cells, and the results showed that IL-1β performed better against LPS. Dex with different concentration was used to attenuate inflammation by IL-1β, and its effect was assessed by RT-PCR to detect the inflammatory related mRNA, including IKβ-α, IKKβ, IL-6, IL-8, and TNF-α. And ELISA to detect the inflammatory cytokines TNF-α, IL-6 and IL-8. RT-PCR was used to quantify levels of lncRNAs, including lncMALAT1, lncHotair, lncH19, and lncNeat1. LncH19 was most closely correlated with the inflammatory response, which was induced by IL-1β and attenuated by Dex. Among the lncRNAs, the level of lncH19 exhibited the highest increase following treatment with 1 μM and 10 μM Dex. Therefore, lncH19 was selected for further function study. LncH19 expression was inhibited by shRNA transduced by lentivirus. Cell assays for cell proliferation and apoptosis as well as RT-PCR, western blot, and ELISA for inflammatory related genes were conducted to confirm the functions of lncH19. Predicted target miRNAs of lncH19 included the following: hsa-miR-346, hsa-miR-324-3p, hsa-miR-18a-3p, hsa-miR-18b-5p, hsa-miR-146b-3p, hsa-miR-19b-3p and hsa-miR-19a-3p. Following estimation by RT-PCR, hsa-miR-346, hsa-miR-18a-3p and hsa-miR-324-3p showed consistent patterns in A549 NC and A549 shlncH19. miRNA inhibitor was transfected into A549 NC and A549 shlncH19cells, and expression levels were determined by RT-PCR. Hsa-miR-324-3p was inhibited the most relative to hsa-miR-346 and hsa-miR-18a-3p and was subjected to further function study. RT-PCR, ELISA and western blotting for inflammatory related genes detection were conducted to validate the functions of the target hsa-miR-324-3p.Results: Dex with 1 μM and 10 μM were shown to be effective in attenuating the inflammatory response. During this process, lncH19 significantly increased in expression (P < 0.05). Dex with 1 μM was for further study. Under IL-1β treatment with or without Dex, the inhibition of lncH19 lead to an increase cell proliferation, a decrease in cell apoptosis, an increase in the protein level of inflammatory-related genes, the phosphorylation of P65, ICAM-1 and VCAM-1, and inflammatory cytokines. Following prediction of the targets of lncH19 and validation by RT-PCR, miR-346, miR-18a-3p and miR-324-3p were found to be negatively correlated to lncH19. Additionally, Dex increased the expression of lncH19, but the expression of the miRNAs was reduced. Among miRNAs, miR-324-3p was the most markedly down-regulated following treatment of miRNA inhibitors. The MTS assay and cell apoptosis assay showed that the miR-324-3p inhibitor inhibited cell proliferation and induced cell apoptosis, thereby significantly attenuating the inflammatory response, which reversed the effect of lncH19 in regulating cell proliferation and the secretion of inflammatory cytokines (P < 0.05). Therefore, lncH19 might regulate miR-324-3p during Dex treatment in pulmonary inflammatory response.Conclusion: Dex can attenuate the pulmonary inflammatory response via regulation of the lncH19/miR-324-3p cascade.


Sign in / Sign up

Export Citation Format

Share Document