Abstract

Virus infection leads to activation of the interferon (IFN)-induced endoribonuclease RNase L, which results in degradation of viral and cellular RNAs. Both cellular and viral RNA cleavage products of RNase L bind pattern recognition receptors (PRRs), like retinoic acid-inducible I (Rig-I) and melanoma differentiation-associated protein 5 (MDA5), to further amplify IFN production and antiviral response. Although much is known about the mechanics of ligand binding and PRR activation, how cells coordinate RNA sensing with signaling response and interferon production remains unclear. We show that RNA cleavage products of RNase L activity induce the formation of antiviral stress granules (avSGs) by regulating activation of double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) and recruit the antiviral proteins Rig-I, PKR, OAS, and RNase L to avSGs. Biochemical analysis of purified avSGs showed interaction of a key stress granule protein, G3BP1, with only PKR and Rig-I and not with OAS or RNase L. AvSG assembly during RNase L activation is required for IRF3-mediated IFN production, but not IFN signaling or proinflammatory cytokine induction. Consequently, cells lacking avSG formation or RNase L signaling produced less IFN and showed higher susceptibility during Sendai virus infection, demonstrating the importance of avSGs in RNase L-mediated host defense. We propose a role during viral infection for RNase L-cleaved RNAs in inducing avSGs containing antiviral proteins to provide a platform for efficient interaction of RNA ligands with pattern recognition receptors to enhance IFN production to mount an effective antiviral response.IMPORTANCE Double-stranded RNAs produced during viral infections serve as pathogen-associated molecular patterns (PAMPs) and bind pattern recognition receptors to stimulate IFN production. RNase L is an IFN-regulated endoribonuclease that is activated in virus-infected cells and cleaves single-stranded viral and cellular RNAs. The RNase L-cleaved dsRNAs signal to Rig-like helicases to amplify IFN production. This study identifies a novel role of antiviral stress granules induced by RNase L as an antiviral signaling hub to coordinate the RNA ligands with cognate receptors to mount an effective host response during viral infections.

Highlights

  • Virus infection leads to activation of the interferon (IFN)-induced endoribonuclease RNase L, which results in degradation of viral and cellular RNAs

  • These results suggest that direct activation of RNase L by 2-5A induces the formation of antiviral stress granules (avSGs) and that Rig-I, protein kinase R (PKR), oligoadenylate synthetase (OAS), and RNase L are recruited to these avSGs

  • Previous studies showed that RNase L cleavage products amplify IFN-␤ production through Rig-I and or melanoma differentiationassociated protein 5 (MDA5) via the MAVS (IPS-1) signaling pathway to sustain the antiviral response, but how the cells coordinate RNA sensing with the signaling response remains unclear [46]

Read more

Summary

Introduction

Virus infection leads to activation of the interferon (IFN)-induced endoribonuclease RNase L, which results in degradation of viral and cellular RNAs. We propose a role during viral infection for RNase L-cleaved RNAs in inducing avSGs containing antiviral proteins to provide a platform for efficient interaction of RNA ligands with pattern recognition receptors to enhance IFN production to mount an effective antiviral response. IMPORTANCE Double-stranded RNAs produced during viral infections serve as pathogen-associated molecular patterns (PAMPs) and bind pattern recognition receptors to stimulate IFN production. IFN produced by virus-infected cells acts in autocrine and paracrine ways by binding to cell surface receptors (IFN-␣/␤ receptor [IFNAR]) to induce expression of antiviral IFN-stimulated genes (ISGs), including retinoic acid-inducible I (Rig-I), melanoma differentiation-associated protein 5 (MDA5), 2=-5=-oligoadenylate synthetase (OAS), RNase L, double-stranded RNA (dsRNA)-dependent protein kinase R (PKR), and interferon-induced proteins with tetratricopeptide repeats (IFIT), to perpetuate antiviral signaling [3, 4]. Phosphorylated eIF2␣ (P-eIF2␣) represses translation and causes aggregation of stalled translation preinitiation complexes containing mRNAs, initiation factors, small ribosomal subunits, and RNA-binding proteins, together with the Ras-GAP SH3 domain binding protein (G3BP) and T-cell-restricted intracellular antigen 1 (TIA 1), into stress granules (SGs) [25]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call