nodamura virus
Recently Published Documents


TOTAL DOCUMENTS

27
(FIVE YEARS 1)

H-INDEX

17
(FIVE YEARS 1)

mBio ◽  
2020 ◽  
Vol 11 (4) ◽  
Author(s):  
Qingxia Han ◽  
Gang Chen ◽  
Jinyan Wang ◽  
David Jee ◽  
Wan-Xiang Li ◽  
...  

ABSTRACT Distinct mammalian RNA viruses trigger Dicer-mediated production of virus-derived small-interfering RNAs (vsiRNA) and encode unrelated proteins to suppress vsiRNA biogenesis. However, the mechanism and function of the mammalian RNA interference (RNAi) response are poorly understood. Here, we characterized antiviral RNAi in a mouse model of infection with Nodamura virus (NoV), a mosquito-transmissible positive-strand RNA virus encoding a known double-stranded RNA (dsRNA)-binding viral suppressor of RNAi (VSR), the B2 protein. We show that inhibition of NoV RNA replication by antiviral RNAi in mouse embryonic fibroblasts (MEFs) requires Dicer-dependent vsiRNA biogenesis and Argonaute-2 slicer activity. We found that VSR-B2 of NoV enhances viral RNA replication in wild-type but not RNAi-defective MEFs such as Argonaute-2 catalytic-dead MEFs and Dicer or Argonaute-2 knockout MEFs, indicating that VSR-B2 acts mainly by suppressing antiviral RNAi in the differentiated murine cells. Consistently, VSR-B2 expression in MEFs has no detectable effect on the induction of interferon-stimulated genes or the activation of global RNA cleavages by RNase L. Moreover, we demonstrate that NoV infection of adult mice induces production of abundant vsiRNA active to guide RNA slicing by Argonaute-2. Notably, VSR-B2 suppresses the biogenesis of both vsiRNA and the slicing-competent vsiRNA-Argonaute-2 complex without detectable inhibition of Argonaute-2 slicing guided by endogenous microRNA, which dramatically enhances viral load and promotes lethal NoV infection in adult mice either intact or defective in the signaling by type I, II, and III interferons. Together, our findings suggest that the mouse RNAi response confers essential protective antiviral immunity in both the presence and absence of the interferon response. IMPORTANCE Innate immune sensing of viral nucleic acids in mammals triggers potent antiviral responses regulated by interferons known to antagonize the induction of RNA interference (RNAi) by synthetic long double-stranded RNA (dsRNA). Here, we show that Nodamura virus (NoV) infection in adult mice activates processing of the viral dsRNA replicative intermediates into small interfering RNAs (siRNAs) active to guide RNA slicing by Argonaute-2. Genetic studies demonstrate that NoV RNA replication in mouse embryonic fibroblasts is inhibited by the RNAi pathway and enhanced by the B2 viral RNAi suppressor only in RNAi-competent cells. When B2 is rendered nonexpressing or nonfunctional, the resulting mutant viruses become nonpathogenic and are cleared in adult mice either intact or defective in the signaling by type I, II, and III interferons. Our findings suggest that mouse antiviral RNAi is active and necessary for the in vivo defense against viral infection in both the presence and absence of the interferon response.


Author(s):  
Donald Bastin ◽  
Amelia S. Aitken ◽  
Adrian Pelin ◽  
Larissa A. Pikor ◽  
Mathieu J. F. Crupi ◽  
...  

2015 ◽  
Vol 59 (5) ◽  
pp. 299-304
Author(s):  
P. Shaik Syed Ali ◽  
Jasmine John ◽  
Manikandan Selvaraj ◽  
Teh Lay Kek ◽  
Mohd Zaki Salleh

2014 ◽  
Vol 20 (3) ◽  
Author(s):  
William J. Allen ◽  
Michael R. Wiley ◽  
Kevin M. Myles ◽  
Zach N. Adelman ◽  
David R. Bevan

2010 ◽  
Vol 150 (1-2) ◽  
pp. 12-21 ◽  
Author(s):  
John J. Rosskopf ◽  
John H. Upton ◽  
Lizette Rodarte ◽  
Tammy A. Romero ◽  
Ming-Ying Leung ◽  
...  

Biochemistry ◽  
2009 ◽  
Vol 48 (11) ◽  
pp. 2307-2309 ◽  
Author(s):  
Stephanie Körber ◽  
P. Shaik Syed Ali ◽  
Julian C.-H. Chen

2005 ◽  
Vol 79 (1) ◽  
pp. 495-502 ◽  
Author(s):  
B. Duane Price ◽  
Lance D. Eckerle ◽  
L. Andrew Ball ◽  
Kyle L. Johnson

ABSTRACT Nodamura virus (NoV) and Flock House virus (FHV) are members of the family Nodaviridae. The nodavirus genome is composed of two positive-sense RNA segments: RNA1 encodes the viral RNA-dependent RNA polymerase and RNA2 encodes the capsid protein precursor. A small subgenomic RNA3, which encodes nonstructural proteins B1 and B2, is transcribed from RNA1 during RNA replication. Previously, FHV was shown to replicate both of its genomic RNAs and to transcribe RNA3 in transiently transfected yeast cells. FHV RNAs and their derivatives could also be expressed from plasmids containing RNA polymerase II promoters. Here we show that all of these features can be recapitulated for NoV, the only nodavirus that productively infects mammals. Inducible plasmid-based systems were used to characterize the RNA replication requirements for NoV RNA1 and RNA2 in Saccharomyces cerevisiae. Induced NoV RNA1 replication was robust. Three previously described NoV RNA1 mutants behaved in yeast as they had in mammalian cells. Yeast colonies were selected from cells expressing NoV RNA1, and RNA2 replicons that encoded yeast nutritional markers, from plasmids. Unexpectedly, these NoV RNA replication-dependent yeast colonies were recovered at frequencies 104-fold lower than in the analogous FHV system. Molecular analysis revealed that some of the NoV RNA replication-dependent colonies contained mutations in the NoV B2 open reading frame in the replicating viral RNA. In addition, we found that NoV RNA1 could support limited replication of a deletion derivative of the heterologous FHV RNA2 that expressed the yeast HIS3 selectable marker, resulting in formation of HIS+ colonies.


2004 ◽  
Vol 78 (12) ◽  
pp. 6698-6704 ◽  
Author(s):  
Kyle L. Johnson ◽  
B. Duane Price ◽  
Lance D. Eckerle ◽  
L. Andrew Ball

ABSTRACT During infection of both vertebrate and invertebrate cell lines, the alphanodavirus Nodamura virus (NoV) expresses two nonstructural proteins of different lengths from the B2 open reading frame. The functions of these proteins have yet to be determined, but B2 of the related Flock House virus suppresses RNA interference both in Drosophila cells and in transgenic plants. To examine whether the NoV B2 proteins had similar functions, we compared the replication of wild-type NoV RNA with that of mutants unable to make the B2 proteins. We observed a defect in the accumulation of mutant viral RNA that varied in extent from negligible in some cell lines (e.g., baby hamster kidney cells) to severe in others (e.g., human HeLa and Drosophila DL-1 cells). These results are consistent with the notion that the NoV B2 proteins act to circumvent an innate antiviral response such as RNA interference that differs in efficacy among different host cells.


Virology ◽  
2003 ◽  
Vol 305 (2) ◽  
pp. 436-451 ◽  
Author(s):  
Kyle L. Johnson ◽  
B.Duane Price ◽  
L.Andrew Ball

Sign in / Sign up

Export Citation Format

Share Document