scholarly journals Evasion of Antiviral Innate Immunity by Theiler's Virus L* Protein through Direct Inhibition of RNase L

2013 ◽  
Vol 9 (6) ◽  
pp. e1003474 ◽  
Author(s):  
Frédéric Sorgeloos ◽  
Babal Kant Jha ◽  
Robert H. Silverman ◽  
Thomas Michiels
2018 ◽  
Vol 14 (4) ◽  
pp. e1006989 ◽  
Author(s):  
Melissa Drappier ◽  
Babal Kant Jha ◽  
Sasha Stone ◽  
Ruth Elliott ◽  
Rong Zhang ◽  
...  
Keyword(s):  
Rnase L ◽  

Cytokine ◽  
2008 ◽  
Vol 43 (3) ◽  
pp. 278
Author(s):  
Robert H. Silverman ◽  
Beihua Dong ◽  
Michael Gale ◽  
Krishnamurthy Malathi

2011 ◽  
Vol 286 (30) ◽  
pp. 26319-26326 ◽  
Author(s):  
Babal Kant Jha ◽  
Irina Polyakova ◽  
Patricia Kessler ◽  
Beihua Dong ◽  
Benjamin Dickerman ◽  
...  

Nature ◽  
2007 ◽  
Vol 448 (7155) ◽  
pp. 816-819 ◽  
Author(s):  
Krishnamurthy Malathi ◽  
Beihua Dong ◽  
Michael Gale ◽  
Robert H. Silverman

2021 ◽  
Author(s):  
Abhishek Asthana ◽  
Christina Gaughan ◽  
Susan R. Weiss ◽  
Robert H Silverman

2′,5′-oligoadenylate(2-5A)-dependent endoribonuclease, RNase L, is a principal mediator of the interferon (IFN) antiviral response. Therefore, regulation of cellular levels of 2-5A is a key point of control in antiviral innate immunity. Cellular 2-5A levels are determined by IFN-inducible 2′,5′-oligoadenylate synthetases (OASs) and by enzymes that degrade 2-5A. Importantly, many coronaviruses and rotaviruses encode 2-5A degrading enzymes thereby antagonizing RNase L and its antiviral effects. A-kinase anchoring protein 7 (AKAP7), a mammalian counterpart, could possibly limit tissue damage from excessive or prolonged RNase L activation during viral infections or from self double-stranded-RNAs that activate OAS. We show these enzymes, members of the two-histidine-phosphoesterase (2H-PE) superfamily, constitute a sub-family referred here as 2′,5′-PEs. 2′,5′-PEs from mouse coronavirus (CoV) MHV (NS2), MERS-CoV (NS4b), group A rotavirus (VP3), and mouse (AKAP7) were investigated for their evolutionary relationships and activities. While there was no activity against 3′,5′-oligoribonucleotides, all cleaved 2′,5′-oligoadenylates efficiently, but with variable activity against other 2′,5′-oligonucleotides. The 2′,5′-PEs are shown to be metal ion-independent enzymes that cleave trimer 2-5A (2′,5′-p3A3) producing mono- or di-adenylates with 2′,3′-cyclic phosphate termini. Our results suggest that elimination of 2-5A might be the sole function of viral 2′,5′-PEs, thereby promoting viral escape from innate immunity by preventing or limiting the activation of RNase L.


2021 ◽  
Author(s):  
Yuan Chao Xue ◽  
Chen Seng Ng ◽  
Yasir Mohamud ◽  
Gabriel Fung ◽  
Huitao Liu ◽  
...  

During viral infection, the dynamic virus-host relationship is constantly in play. Many cellular proteins such as RNA-binding proteins (RBPs) have been shown to mediate antiviral responses during viral infection. Here we reported that the RBP, fused in sarcoma/translocated in liposarcoma (FUS/TLS), acts as a host restricting factor against the infection of coxsackievirus B3 (CVB3). Mechanistically, we found that deletion of FUS leads to increased viral RNA transcription and enhanced internal ribosome entry site (IRES)-driven translation, with no apparent impact on viral RNA stability. We further demonstrated that FUS physically interacts with viral genome, which may contribute to direct inhibition of viral RNA transcription/translation. Moreover, we identified a novel function for FUS in regulating host innate immune response. We showed that in the absence of FUS, gene expression of type I interferons and proinflammatory cytokines elicited by viral or bacterial infection is significantly impaired. Emerging evidence suggests a role for stress granules (SGs) in antiviral innate immunity. We further uncovered that knockout of FUS abolishes the ability to form SGs upon CVB3 infection or polyinosinic:polycytidylic acid (polyIC) treatment. Finally, we showed that, to avoid FUS-mediated antiviral response and innate immunity, CVB3 infection results in cytoplasmic mislocalization and cleavage of FUS through the enzymatic activity of viral proteases. Together, our findings in this study identified FUS as a novel host antiviral factor, which restricts CVB3 replication through direct inhibition of viral RNA transcription and protein translation and by regulating host antiviral innate immunity. IMPORTANCE Enteroviruses are common human pathogens, including those that cause myocarditis (coxsackievirus B3, CVB3), poliomyelitis (poliovirus) and hand, foot and mouse disease (enterovirus 71). Understanding the virus-host interaction is crucial for finding the treatment and prevention of these pathogens. In this study, we explored the interplay between host RNA binding protein FUS/TLS and CVB3 and reported that FUS/TLS restricts CVB3 replication through direct inhibition of viral RNA transcription/translation and by regulation of cellular antiviral innate immunity. To impede the antiviral role of FUS, CVB3 targets FUS for mislocalization and cleavage. Findings from this study provide novel insights into interactions between CVB3 and the FUS, which may lead to novel therapeutic interventions against enterovirus-induced diseases.


Sign in / Sign up

Export Citation Format

Share Document