scholarly journals Viral determinants in the NS3 helicase and 2K peptide that promote West Nile virus resistance to antiviral action of 2′,5′-oligoadenylate synthetase 1b

Virology ◽  
2010 ◽  
Vol 399 (1) ◽  
pp. 176-185 ◽  
Author(s):  
Eva Mertens ◽  
Anna Kajaste-Rudnitski ◽  
Shessy Torres ◽  
Anneke Funk ◽  
Marie-Pascale Frenkiel ◽  
...  
PLoS ONE ◽  
2014 ◽  
Vol 9 (3) ◽  
pp. e92545 ◽  
Author(s):  
Soumya Deo ◽  
Trushar R. Patel ◽  
Edis Dzananovic ◽  
Evan P. Booy ◽  
Khalid Zeid ◽  
...  

2003 ◽  
Vol 81 (3) ◽  
pp. 230-236 ◽  
Author(s):  
Marianne Lucas ◽  
Tomoji Mashimo ◽  
Marie‐Pascale Frenkiel ◽  
Dominique Simon‐Chazottes ◽  
Xavier Montagutelli ◽  
...  

2005 ◽  
Vol 281 (8) ◽  
pp. 4624-4637 ◽  
Author(s):  
Anna Kajaste-Rudnitski ◽  
Tomoji Mashimo ◽  
Marie-Pascale Frenkiel ◽  
Jean-Louis Guénet ◽  
Marianne Lucas ◽  
...  

Virology ◽  
2005 ◽  
Vol 331 (2) ◽  
pp. 457-470 ◽  
Author(s):  
Shannan L. Rossi ◽  
Qizu Zhao ◽  
Vivian K. O'Donnell ◽  
Peter W. Mason

2006 ◽  
Vol 80 (14) ◽  
pp. 7009-7019 ◽  
Author(s):  
Melanie A. Samuel ◽  
Kevin Whitby ◽  
Brian C. Keller ◽  
Anantha Marri ◽  
Winfried Barchet ◽  
...  

ABSTRACT West Nile virus (WNV) is a neurotropic, mosquito-borne flavivirus that can cause lethal meningoencephalitis. Type I interferon (IFN) plays a critical role in controlling WNV replication, spread, and tropism. In this study, we begin to examine the effector mechanisms by which type I IFN inhibits WNV infection. Mice lacking both the interferon-induced, double-stranded-RNA-activated protein kinase (PKR) and the endoribonuclease of the 2′,5′-oligoadenylate synthetase-RNase L system (PKR−/− × RL−/−) were highly susceptible to subcutaneous WNV infection, with a 90% mortality rate compared to the 30% mortality rate observed in congenic wild-type mice. PKR−/− × RL−/− mice had increased viral loads in their draining lymph nodes, sera, and spleens, which led to early viral entry into the central nervous system (CNS) and higher viral burden in neuronal tissues. Although mice lacking RNase L showed a higher CNS viral burden and an increased mortality, they were less susceptible than the PKR−/− × RL−/− mice; thus, we also infer an antiviral role for PKR in the control of WNV infection. Notably, a deficiency in both PKR and RNase L resulted in a decreased ability of type I IFN to inhibit WNV in primary macrophages and cortical neurons. In contrast, the peripheral neurons of the superior cervical ganglia of PKR−/− × RL−/− mice showed no deficiency in the IFN-mediated inhibition of WNV. Our data suggest that PKR and RNase L contribute to IFN-mediated protection in a cell-restricted manner and control WNV infection in peripheral tissues and some neuronal subtypes.


2015 ◽  
Vol 190 (2) ◽  
pp. 236-249 ◽  
Author(s):  
Soumya Deo ◽  
Trushar R. Patel ◽  
Grzegorz Chojnowski ◽  
Amit Koul ◽  
Edis Dzananovic ◽  
...  

Virology ◽  
2011 ◽  
Vol 417 (1) ◽  
pp. 147-153 ◽  
Author(s):  
Dominique Simon-Chazottes ◽  
Marie-Pascale Frenkiel ◽  
Xavier Montagutelli ◽  
Jean-Louis Guénet ◽  
Philippe Desprès ◽  
...  

2014 ◽  
Vol 95 (9) ◽  
pp. 1991-2003 ◽  
Author(s):  
Katherine L. Hussmann ◽  
Rianna Vandergaast ◽  
Kang Zheng ◽  
Lisa I. Hoover ◽  
Brenda L. Fredericksen

The molecular basis for the increased resistance of astrocytes to a non-neuropathogenic strain of West Nile virus (WNV), WNV-MAD78, compared with the neuropathogenic strain WNV-NY remains unclear. Here, we demonstrated that the reduced susceptibility of astrocytes to WNV-MAD78 is due to a combination of both cellular activities as well as viral determinants. Analyses of the viral particle indicated that astrocyte-derived WNV-MAD78 particles were less infectious than those of WNV-NY. Additionally, inhibition of cellular furin-like proteases increased WNV-MAD78 infectious particle production in astrocytes, suggesting that high levels of furin-like protease activity within these cells acted in a cell- and strain-specific manner to inhibit WNV-MAD78 replication. Moreover, analysis of recombinant viruses indicated that the structural proteins of WNV-MAD78 were responsible for decreased particle infectivity and the corresponding reduction in infectious particle production compared with WNV-NY. Thus, the composition of the WNV virion was also a major determinant for viral fitness within astrocytes and may contribute to WNV propagation within the central nervous system. Whether the WNV-MAD78 structural genes reduce virus replication and particle infectivity through the same mechanism as the cellular furin-like protease activity or whether these two determinants function through distinct pathways remains to be determined.


Sign in / Sign up

Export Citation Format

Share Document