scholarly journals The 2009 pandemic A/Wenshan/01/2009 H1N1 induces apoptotic cell death in human airway epithelial cells

2011 ◽  
Vol 3 (4) ◽  
pp. 221-229 ◽  
Author(s):  
Ning Yang ◽  
Xiaoxu Hong ◽  
Penghui Yang ◽  
Xiangwu Ju ◽  
Yuguo Wang ◽  
...  
PLoS ONE ◽  
2012 ◽  
Vol 7 (9) ◽  
pp. e46184 ◽  
Author(s):  
Penghui Yang ◽  
Jiejie Deng ◽  
Chenggang Li ◽  
Peirui Zhang ◽  
Li Xing ◽  
...  

2021 ◽  
Vol 55 (5) ◽  
pp. 590-604

BACKGROUND/AIMS: Oxidative stress and infections by Pseudomonas aeruginosa (P. aeruginosa) are prominent in lungs of patients suffering from cystic fibrosis (CF). METHODS: The present study examines effects of P. aeruginosa on lipid peroxidation in human and mouse lungs, and cell death induced by P. aeruginosa in human airway epithelial cells. The role of the Ca2+ activated Cl- channel TMEM16A, the phospholipid scramblase TMEM16F, and the CFTR Cl- channel for ferroptotic cell death is examined. RESULTS: Lipid peroxidation was detected in human CF lungs, which correlated with bacterial infection. In vivo inoculation with P. aeruginosa or Staphylococcus aureus (S. aureus) induced lipid peroxidation in lungs of mice lacking expression of CFTR, and in lungs of wild type animals. Incubation of CFBE human airway epithelial cells with P. aeruginosa induced an increase in reactive oxygen species (ROS), causing lipid peroxidation and cell death independent of expression of wt-CFTR or F508del-CFTR. Knockdown of TMEM16A attenuated P. aeruginosa induced cell death. Antioxidants such as coenzyme Q10 and idebenone as well as the inhibitor of ferroptosis, ferrostatin-1, inhibited P. aeruginosa-induced cell death. CFBE cells expressing wtCFTR, but not F508del-CFTR, activated a basal Cl- conductance upon exposure to P. aeruginosa, which was caused by an increase in intracellular basal Ca2+ concentrations and activation of Ca2+-dependent adenylate cyclase. CONCLUSION: The data suggest an intrinsic pro-inflammatory phenotype in CF epithelial cells, while ferroptosis is observed in both non-CF and CF epithelial cells upon infection with P. aeruginosa. CF cells fail to activate fluid secretion in response to infection with P. aeruginosa. The use of antioxidants and inhibitors of ferroptosis is proposed as a treatment of pneumonia caused by infection with P. aeruginosa.


2018 ◽  
Vol 102 ◽  
pp. 177-178
Author(s):  
Hrishikesh Kulkarni ◽  
Michelle Elvington ◽  
Yi-Chieh Perng ◽  
M. Kathryn Liszewski ◽  
Christopher Frakouh ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (4) ◽  
pp. 944
Author(s):  
Jung Yeon Hong ◽  
Mi Na Kim ◽  
Eun Gyul Kim ◽  
Jae Woo Lee ◽  
Hye Rin Kim ◽  
...  

Exposure to high oxygen concentrations leads to generation of excessive reactive oxygen species, causing cellular injury and multiple organ dysfunctions and is associated with a high mortality rate. Clusterin (CLU) is a heterodimeric glycoprotein that mediates several intracellular signaling pathways, including cell death and inflammation. However, the role of CLU in the pathogenesis of hyperoxic acute lung injury (HALI) is unknown. Wild-type (WT) and CLU-deficient mice and cultured human airway epithelial cells were used. Changes in cell death- and inflammation-related molecules with or without hyperoxia exposure in cells and animals were determined. Hyperoxia induced an increase in CLU expression in mouse lungs and human airway epithelial cells. Mice lacking CLU had increased HALI and mortality rate compared with WT mice. In vitro, CLU-disrupted cells showed enhanced release of cytochrome c, Bax translocation, cell death and inflammatory cytokine expression. However, treatment with recombinant CLU attenuated hyperoxia-induced apoptosis. Moreover, the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses revealed metabolic pathways, hematopoietic cell lineage, response to stress and localization and regulation of immune system that were differentially regulated between WT and CLU−/− mice. These results demonstrate that prolonged hyperoxia-induced lung injury is associated with CLU expression and that CLU replenishment may alleviate hyperoxia-induced cell death.


2019 ◽  
Vol 60 (2) ◽  
pp. 144-157 ◽  
Author(s):  
Hrishikesh S. Kulkarni ◽  
Michelle L. Elvington ◽  
Yi-Chieh Perng ◽  
M. Kathryn Liszewski ◽  
Derek E. Byers ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document