Abstract

ABSTRACTInfluenza A virus mRNAs are transcribed by the viral RNA-dependent RNA polymerase in the cell nucleus before being exported to the cytoplasm for translation. Segment 7 produces two major transcripts: an unspliced mRNA that encodes the M1 matrix protein and a spliced transcript that encodes the M2 ion channel. Export of both mRNAs is dependent on the cellular NXF1/TAP pathway, but it is unclear how they are recruited to the export machinery or how the intron-containing but unspliced M1 mRNA bypasses the normal quality-control checkpoints. Using fluorescent in situ hybridization to monitor segment 7 mRNA localization, we found that cytoplasmic accumulation of unspliced M1 mRNA was inefficient in the absence of NS1, both in the context of segment 7 RNPs reconstituted by plasmid transfection and in mutant virus-infected cells. This effect was independent of any major effect on steady-state levels of segment 7 mRNA or splicing but corresponded to a ∼5-fold reduction in the accumulation of M1. A similar defect in intronless hemagglutinin (HA) mRNA nuclear export was seen with an NS1 mutant virus. Efficient export of M1 mRNA required both an intact NS1 RNA-binding domain and effector domain. Furthermore, while wild-type NS1 interacted with cellular NXF1 and also increased the interaction of segment 7 mRNA with NXF1, mutant NS1 polypeptides unable to promote mRNA export did neither. Thus, we propose that NS1 facilitates late viral gene expression by acting as an adaptor between viral mRNAs and the cellular nuclear export machinery to promote their nuclear export.IMPORTANCE Influenza A virus is a major pathogen of a wide variety of mammalian and avian species that threatens public health and food security. A fuller understanding of the virus life cycle is important to aid control strategies. The virus has a small genome that encodes relatively few proteins that are often multifunctional. Here, we characterize a new function for the NS1 protein, showing that, as well as previously identified roles in antagonizing the innate immune defenses of the cell and directly upregulating translation of viral mRNAs, it also promotes the nuclear export of the viral late gene mRNAs by acting as an adaptor between the viral mRNAs and the cellular mRNA nuclear export machinery.

Highlights

  • Influenza A virus mRNAs are transcribed by the viral RNA-dependent RNA polymerase in the cell nucleus before being exported to the cytoplasm for translation

  • We characterize a novel functional role of NS1 in promoting the nuclear export of viral late gene mRNAs, in particular, M1 and HA mRNA. We base this conclusion on the notable difference in the bulk localizations of segment 7 mRNA between infected and RNP-transfected cells, a difference that could be obviated by the additional expression of NS1, supported further by the similar alterations in segment 7 mRNA localization seen in cells infected with NS1 mutant viruses

  • We found that NS1 increased the amount of segment 7 mRNA bound by NXF1 and that there was a good correlation between the ability of NS1 mutants to perform this function and to bind NXF1 itself, suggesting that NS1 acts as an adaptor jvi.asm.org protein providing a bridge between the viral transcription machinery and the cellular mRNA export pathway

Read more

Summary

Introduction

Influenza A virus mRNAs are transcribed by the viral RNA-dependent RNA polymerase in the cell nucleus before being exported to the cytoplasm for translation. Influenza A virus (IAV) has a genome constituted of eight single-stranded, negativesense RNA molecules, each separately encapsidated into viral ribonucleoprotein (vRNP) particles with one copy of the viral PB1-PB2-PA (3P) heterotrimeric RNA polymerase and multiple copies of a nucleoprotein (NP) [1] These RNPs are the templates for transcription and replication of the genome (the latter by a cRNA replicative intermediate), which occurs in the host cell nucleus. All mRNA transcripts start with a host-derived 7mGpppGm cap structure derived from cellular pre-mRNAs by the process of “cap-snatching” [1], which provides structural identity between the 5= ends of viral and cellular mRNAs

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