Poster Presentation Lorne Infection and Immunity 2019

RNA stability in alphavirus infected cells (#168)

Damien Phan 1
  1. The Peter Doherty Institute for Infection and Immunity, Melbourne

Preliminary data shows that the alphavirus Semliki Forest virus (SFV) replicates less well in RNase-L knockout (KO) mouse embryonic fibroblasts (MEFs) than in wild-type cells. This is surprising, as RNase-L is generally considered to be an antiviral protein, degrading RNA in response to viral infection (1).

One explanation involves the interaction of alphaviruses with host RNA-binding proteins. Studies performed primarily with the alphavirus Sindbis virus (SINV) have shown the RNA binding protein HuR binds with high affinity to alphavirus RNA through sequence elements in the 3’ untranslated region (UTR), resulting in increased stability of viral transcripts (2, 3). Stability of some host transcripts is also reduced during infection (4). Tristetraprolin (TTP) is another RNA-binding protein that binds competitively with HuR and promotes RNA degradation (5). TTP has been found to recruit RNase L to cellular transcripts under mitogen stimulation, resulting in the degradation of these transcripts (6).

We hypothesise that in SFV infected cells, HuR is sequestered to viral RNA. This protects viral RNA from TTP binding, and therefore degradation by RNase L. Cellular transcripts with no HuR bound would remain vulnerable to TTP binding and RNase L degradation, leading to reduction of cellular transcripts, and reduced competition for translation.

Consistent with studies in SINV, we have shown using immunofluorescent microscopy that HuR relocalises from the nucleus to the cytoplasm during SFV infection. We will determine the roles of HuR, TTP and RNase L in SFV infection using siRNA knockdown of expression of these genes. The effect of RNase L on host transcript levels during SFV infection will be determined using whole-transcriptome sequencing of SFV infected WT and RNase L KO cells.

  1. Silverman RH. 2007. Viral encounters with 2',5'-oligoadenylate synthetase and RNase L during the interferon antiviral response. J Virol 81:12720-12729.
  2. Sokoloski KJ, Dickson AM, Chaskey EL, Garneau NL, Wilusz CJ, Wilusz J. 2010. Sindbis virus usurps the cellular HuR protein to stabilize its transcripts and promote productive infections in mammalian and mosquito cells. Cell Host & Microbe 8:196-207.
  3. Dickson AM, Anderson JR, Barnhart MD, Sokoloski KJ, Oko L, Opyrchal M, Galanis E, Wilusz CJ, Morrison TE, Wilusz J. 2012. Dephosphorylation of HuR protein during alphavirus infection is associated with HuR relocalization to the cytoplasm. J Biol Chem 287:36229-36238.
  4. Barnhart MD, Moon SL, Emch AW, Wilusz CJ, Wilusz J. 2013. Changes in cellular mRNA stability, splicing, and polyadenylation through HuR protein sequestration by a cytoplasmic RNA virus. Cell Rep 5:909-917.
  5. Wu X, Brewer G. 2012. The regulation of mRNA stability in mammalian cells: 2.0. Gene 500:10-21.
  6. Brennan-Laun SE, Li XL, Ezelle HJ, Venkataraman T, Blackshear PJ, Wilson GM, Hassel BA. 2014. RNase L attenuates mitogen-stimulated gene expression via transcriptional and post-transcriptional mechanisms to limit the proliferative response. J Biol Chem 289:33629-33643.