Influenza A virus-induced autophagy contributes to enhancement of virus infectivity by SOD1 downregulation in alveolar epithelial cells

2018 ◽  
Vol 498 (4) ◽  
pp. 960-966 ◽  
Author(s):  
Kwang Il Jung ◽  
Chul Woong Pyo ◽  
Sang-Yun Choi
2009 ◽  
Vol 70 (12) ◽  
pp. 1016-1019 ◽  
Author(s):  
Fanny LeBouder ◽  
Khaled Khoufache ◽  
Catherine Menier ◽  
Yassmina Mandouri ◽  
Mahmoud Keffous ◽  
...  

2012 ◽  
Vol 13 (1) ◽  
pp. 43 ◽  
Author(s):  
Beata Kosmider ◽  
Elise M Messier ◽  
William J Janssen ◽  
Piruz Nahreini ◽  
Jieru Wang ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 509 ◽  
Author(s):  
Meenakshi Tiwary ◽  
Robert J. Rooney ◽  
Swantje Liedmann ◽  
Kim S. LeMessurier ◽  
Amali E. Samarasinghe

Eosinophils, previously considered terminally differentiated effector cells, have multifaceted functions in tissues. We previously found that allergic mice with eosinophil-rich inflammation were protected from severe influenza and discovered specialized antiviral effector functions for eosinophils including promoting cellular immunity during influenza. In this study, we hypothesized that eosinophil responses during the early phase of influenza contribute to host protection. Using in vitro and in vivo models, we found that eosinophils were rapidly and dynamically regulated upon influenza A virus (IAV) exposure to gain migratory capabilities to traffic to lymphoid organs after pulmonary infection. Eosinophils were capable of neutralizing virus upon contact and combinations of eosinophil granule proteins reduced virus infectivity through hemagglutinin inactivation. Bi-directional crosstalk between IAV-exposed epithelial cells and eosinophils occurred after IAV infection and cross-regulation promoted barrier responses to improve antiviral defenses in airway epithelial cells. Direct interactions between eosinophils and airway epithelial cells after IAV infection prevented virus-induced cytopathology in airway epithelial cells in vitro, and eosinophil recipient IAV-infected mice also maintained normal airway epithelial cell morphology. Our data suggest that eosinophils are important in the early phase of IAV infection providing immediate protection to the epithelial barrier until adaptive immune responses are deployed during influenza.


2003 ◽  
Vol 77 (7) ◽  
pp. 4104-4112 ◽  
Author(s):  
Shigefumi Okamoto ◽  
Shigetada Kawabata ◽  
Ichiro Nakagawa ◽  
Yoshinobu Okuno ◽  
Toshiyuki Goto ◽  
...  

ABSTRACT The apparent worldwide resurgence of invasive Streptococcus pyogenes infection in the last two decades remains unexplained. At present, animal models in which toxic shock-like syndrome or necrotizing fasciitis is induced after S. pyogenes infection are not well developed. We demonstrate here that infection with a nonlethal dose of influenza A virus 2 days before intranasal infection with a nonlethal dose of S. pyogenes strains led to a death rate of more than 90% in mice, 10% of which showed necrotizing fasciitis. Infection of lung alveolar epithelial cells by the influenza A virus resulted in viral hemagglutinin expression on the cell surface and promoted internalization of S. pyogenes. However, treatment with monoclonal antibodies to hemagglutinin markedly decreased this internalization. Our results indicate that prior infection with influenza A virus induces a lethal synergism, resulting in the induction of invasive S. pyogenes infection in mice.


Sign in / Sign up

Export Citation Format

Share Document