Diphenyleneiodonium enhances oxidative stress and inhibits Japanese encephalitis virus induced autophagy and ER stress pathways

2018 ◽  
Vol 502 (2) ◽  
pp. 232-237 ◽  
Author(s):  
Manish Sharma ◽  
Kiran Bala Sharma ◽  
Shailendra Chauhan ◽  
Sankar Bhattacharyya ◽  
Sudhanshu Vrati ◽  
...  
2019 ◽  
Vol 93 (17) ◽  
Author(s):  
Qianruo Wang ◽  
Xiu Xin ◽  
Ting Wang ◽  
Jiawu Wan ◽  
Yangtao Ou ◽  
...  

ABSTRACTAccumulated evidence demonstrates that Japanese encephalitis virus (JEV) infection triggers endoplasmic reticulum (ER) stress and neuron apoptosis. ER stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) has been reported to induce apoptosis under acute or prolonged ER stress. However, the precise role of PERK in JEV-induced apoptosis and encephalitis remains unknown. Here, we report that JEV infection activates the PERK-ATF4-CHOP apoptosis pathway bothin vitroandin vivo. PERK activation also promotes the formation of stress granule, which in turn represses JEV-induced apoptosis. However, PERK inhibitor reduces apoptosis, indicating that JEV-activated PERK predominantly induces apoptosis via the PERK-ATF4-CHOP apoptosis pathway. Among JEV proteins that have been reported to induce ER stress, only JEV NS4B can induce PERK activation. PERK has been reported to form an active molecule by dimerization. The coimmunoprecipitation assay shows that NS4B interacts with PERK. Moreover, glycerol gradient centrifugation shows that NS4B induces PERK dimerization. Both the LIG-FHA and the LIG-WD40 domains within NS4B are required to induce PERK dimerization, suggesting that JEV NS4B pulls two PERK molecules together by simultaneously interacting with them via different motifs. PERK deactivation reduces brain cell damage and encephalitis during JEV infection. Furthermore, expression of JEV NS4B is sufficient to induce encephalitis via PERK in mice, indicating that JEV activates PERK primarily via its NS4B to cause encephalitis. Taken together, our findings provide a novel insight into JEV-caused encephalitis.IMPORTANCEJapanese encephalitis virus (JEV) infection triggers endoplasmic reticulum (ER) stress and neuron apoptosis. ER stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) has been reported to induce apoptosis under acute or prolonged ER stress. However, whether the PERK pathway of ER stress response plays important roles in JEV-induced apoptosis and encephalitis remains unknown. Here, we found that JEV infection activates ER stress sensor PERK in neuronal cells and mouse brains. PERK activation induces apoptosis via the PERK-ATF4-CHOP apoptosis pathway upon JEV infection. Among the JEV proteins prM, E, NS1, NS2A, NS2B, and NS4B, only NS4B activates PERK. Moreover, activated PERK participates in apoptosis and encephalitis induced by JEV and NS4B. These findings provide a novel therapeutic approach for JEV-caused encephalitis.


2015 ◽  
Vol 161 (3) ◽  
pp. 699-703 ◽  
Author(s):  
Mingjie Huang ◽  
Ahui Xu ◽  
Xiaoyu Wu ◽  
Yanni Zhang ◽  
Yunli Guo ◽  
...  

2017 ◽  
Vol 98 (5) ◽  
pp. 1027-1039 ◽  
Author(s):  
Manish Sharma ◽  
Sankar Bhattacharyya ◽  
Kiran Bala Sharma ◽  
Shailendra Chauhan ◽  
Suramya Asthana ◽  
...  

Viruses ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 356 ◽  
Author(s):  
Zhao-Yang Wang ◽  
Zi-Da Zhen ◽  
Dong-Ying Fan ◽  
Pei-Gang Wang ◽  
Jing An

The Japanese encephalitis virus (JEV) is a Culex mosquito-borne flavivirus and is the pathogenic agent of Japanese encephalitis, which is the most important type of viral encephalitis in the world. Macrophages are a type of pivotal innate immunocyte that serve as sentinels and respond quickly to pathogen invasions. However, some viruses like JEV can hijack macrophages as a refuge for viral replication and immune escape. Despite their crucial involvement in early JEV infection, the transcriptomic landscapes of JEV-infected macrophages are void. Here, by using an in situ JEV infection model, we investigate the transcriptomic alteration of JEV-infected peritoneal macrophages. We found that, upon JEV infection, the macrophages underwent M1 polarization and showed the drastic activation of innate immune and inflammatory pathways. Interestingly, almost all the programmed cell death (PCD) pathways were activated, especially the apoptosis, pyroptosis, and necroptosis pathways, which were verified by the immunofluorescent staining of specific markers. Further transcriptomic analysis and TUNEL staining revealed that JEV infection caused apparent DNA damage. The transcriptomic analysis also revealed that JEV infection promoted ROS and RNS generation and caused oxidative stress, which activated multiple cell death pathways. Our work uncovers the pivotal pathogenic roles of oxidative stress and multiple PCD pathways in JEV infection, providing a novel perspective on JEV–host interactions.


2017 ◽  
Vol 8 (1) ◽  
pp. e2556-e2556 ◽  
Author(s):  
Sriparna Mukherjee ◽  
Noopur Singh ◽  
Nabonita Sengupta ◽  
Mahar Fatima ◽  
Pankaj Seth ◽  
...  

2011 ◽  
Vol 8 (1) ◽  
pp. 338
Author(s):  
Yi-Ping Wu ◽  
Chung-Ming Chang ◽  
Chun-Yu Hung ◽  
Meng-Chieh Tsai ◽  
Scott C Schuyler ◽  
...  

2011 ◽  
Vol 8 (1) ◽  
pp. 128 ◽  
Author(s):  
Yi-Ping Wu ◽  
Chung-Ming Chang ◽  
Chun-Yu Hung ◽  
Meng-Chieh Tsai ◽  
Scott C Schuyler ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document