Abstract

The production of type I interferons (IFNs) in response to viral infections is critical for antiviral immunity. However, IFN production is transient, and continued expression can lead to inflammatory or autoimmune diseases. Thus, understanding the mechanisms underlying the negative regulation of IFN expression could lead to the development of novel therapeutic approaches to the treatment of these diseases. We report that the transcription factor IRF3 plays a central role in the negative regulation of interferon-β (IFNβ) expression during both acute and persistent (chronic) virus infections. We show that the degradation of IRF3 during acute infections, rather than the activation of transcriptional repressors, leads to the down regulation of IFNβ expression. We also show that the block to IFNβ expression in mouse embryonic fibroblasts that are persistently infected with Sendai virus (SeV) correlates with the absence of transcriptionally active IRF3. Remarkably, ongoing protein synthesis and viral replication are required to maintain repression of the IFNβ gene in persistently infected cells, as the gene can be activated by the protein synthesis inhibitor cycloheximide, or by the antiviral drug ribavirin. Finally, we show that the SeV V protein inhibits IRF3 activity in persistently infected cells. Thus, in conjunction with the known interference with STAT1 by the SeV C protein, both IFN activation and its signaling pathways are blocked in persistently infected cells. We conclude that the transcription factor IRF3 is targeted for turnover and inactivation through distinct mechanisms from both the host cells and virus, leading to the inhibition of IFNβ gene expression during acute and persistent viral infections. These observations show that IRF3 plays a critical role, not only in the activation of the IFNβ gene, but also in the controlling the duration of its expression. (284 words)

Highlights

  • Virus infection induces the transient expression of type I interferons (IFNs) in virtually every cell type [1]

  • Since we have established that the major block to IFNb expression is at the level of IRF3/7 protein (Fig. 3A), and IRF7 protein is too low to be detected in PI-mouse embryonic fibroblasts (MEF), we focused our attention on IRF3 regulation persistently infected MEFs (PI-MEFs)

  • Post-induction IFNb turn-off Here we show that inactivation of the transcription factor IRF3 plays a critical role in the negative regulation of IFNb expression during both acute and persistent Sendai virus infections

Read more

Summary

Introduction

Virus infection induces the transient expression of type I interferons (IFNs) in virtually every cell type [1]. Viral RNA is detected by RIGI and MDA5 in most cells [7] Both proteins undergo a conformational change upon binding to a 59-triphosphate panhandle RNA or long double stranded RNAs (dsRNAs) associated with virus infection and replication [8,9]. This conformational change leads to homodimerization of the RNA sensors, and signal transmission through a critical adaptor protein MAVS located on the mitochondrial membrane [10,11]. Chromatin remodeling factors and the basic transcription machinery are recruited to drive the expression of the gene [16]

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