host shutoff
Recently Published Documents


TOTAL DOCUMENTS

118
(FIVE YEARS 15)

H-INDEX

40
(FIVE YEARS 2)

2022 ◽  
Author(s):  
Emma Louise Wise ◽  
Jerzy Samolej ◽  
Gillian Elliott

Virion host shutoff (vhs) protein is an endoribonuclease encoded by herpes simplex virus 1 (HSV1). Vhs causes a number of changes to the infected cell environment that favour translation of late (L) virus proteins: cellular mRNAs are degraded, immediate-early (IE) and early (E) viral transcripts are sequestered in the nucleus with polyA binding protein (PABPC1), and dsRNA is degraded to help dampen the PKR-dependent stress response. To further our understanding of the cell biology of vhs, we constructed a virus expressing vhs tagged at its C-terminus with GFP. When first expressed, vhs-GFP localised to juxtanuclear clusters, and later it colocalised and interacted with its binding partner VP16, and was packaged into virions. Despite vhs-GFP maintaining activity when expressed in isolation, it failed to degrade mRNA or relocalise PABPC1 during infection, while viral transcript levels were similar to those seen for a vhs knockout virus. PKR phosphorylation was also enhanced in vhs-GFP infected cells, in line with a failure to degrade dsRNA. Nonetheless, mRNA FISH revealed that as in Wt but not Dvhs infection, IE and E, but not L transcripts were retained in the nucleus of vhs-GFP infected cells at late times. Moreover, a representative cellular transcript which is ordinarily highly susceptible to vhs degradation, was also retained in the nucleus. These results reveal that the vhs-induced nuclear retention of the infected cell transcriptome is dependent on vhs expression but not on its endoribonuclease activity, uncoupling these two functions of vhs.


mBio ◽  
2021 ◽  
Author(s):  
Wolfgang Nacken ◽  
André Schreiber ◽  
Dörthe Masemann ◽  
Stephan Ludwig

We investigated the role of IAV nonstructural protein 1 NS1 in host gene shutoff—a central feature of IAV replication. We demonstrate that the effector domain of NS1 alone mediates host gene shutoff by inhibition of host transcription and by deregulation of the polyadenylation (polyA) signal-mediated 3′ termination of host transcription.


2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Anita Nag ◽  
Yevgeniy Gerassimovich ◽  
Samantha Miladinovski‐Bengall

2021 ◽  
Vol 255 ◽  
pp. 108979
Author(s):  
Tianqiong He ◽  
Mingshu Wang ◽  
Anchun Cheng ◽  
Qiao Yang ◽  
Renyong Jia ◽  
...  

2021 ◽  
Author(s):  
Rachel Emily Levene ◽  
Shailab D. Shrestha ◽  
Marta Maria Gaglia

The influenza A endoribonuclease PA-X regulates virulence and transmission of the virus by reducing host gene expression and thus regulating immune responses to influenza A virus. Despite this key function in viral biology, the levels of PA-X protein remain markedly low during infection, and previous results suggest that these low levels are not solely the result of regulation of the level of translation and RNA stability. How PA-X is regulated post-translationally remains unknown. We now report that the PA-X protein is rapidly turned over. PA-X from multiple viral strains are short-lived, although the half-life of PA-X ranges from ∼30 minutes to ∼3.5 hours depending on the strain. Moreover, sequences in the variable PA-X C-terminal domain are primarily responsible for regulating PA-X half-life, although the N-terminal domain also accounts for some differences among strains. Interestingly, we find that the PA-X from the 2009 pandemic H1N1 strain has a longer half-life compared to the other variants we tested. This PA-X isoform has been reported to have a higher host shutoff activity, suggesting a role for protein turnover in regulating PA-X activity. Collectively, this study reveals a novel regulatory mechanism of PA-X protein levels that may impact host shutoff activity during influenza A virus infection. IMPORTANCE The PA-X protein from influenza A virus reduces host immune responses to infection through suppressing host gene expression, including genes encoding the antiviral response. Thus, it plays a central role in influenza A virus biology. Despite its key function, PA-X was only discovered in 2012 and much remains to be learned including how PA-X activity is regulated to promote optimal levels of viral infection. In this study, we reveal that PA-X protein levels are very low likely because of rapid turnover. We show that instability is a conserved property among PA-X variants from different strains of influenza A virus, but that the half-lives of PA-X variants differ. Moreover, the longer half-life of PA-X from the 2009 pandemic H1N1 strain correlates with its reported higher activity. Therefore, PA-X stability may be a way to regulate its activity and may contribute to the differential virulence of influenza A virus strains.


2020 ◽  
Author(s):  
Pnina Schlesinger ◽  
Niza Frenkel

Glioblastoma multiforme is the most prevalent and deadliest form of glioma and brain cancer, with a very poor prognosis. In an effort to develop an oncolytic viral vector for the treatment of Glioblastoma multiforme, we replaced the UL39 and UL40 genes encoding ribonucleotide reductase (RR) with green fluorescence protein and luciferase genes in wild type KOS and in the virion host shutoff mutant vhs-1, resulting in strains KOS-RR and Vhs-RR, respectively. KOS-RR and Vhs-RR caused death of infected U87 Glioblastoma multiforme cell cultures within one day after infection, whereas KOS and vhs-1-infected cells were more viable. All four viral strains caused apoptotic DNA laddering in infected H1299 lung cancer cells, while only Vhs-RR caused apoptosis in U87 cell cultures. Vhs-RR gave higher yields on U87 than on Vero cells, while it barely proliferated on non-dividing Goiter cells. These results indicate that Vhs-RR proliferates well in actively growing U87 Glioblastoma multiforme cells, causing their death in a mechanism involving apoptosis, while sparing non-dividing cells. Therefore, Vhs-RR is a promising candidate for oncolytic treatment of brain tumor malignancies.


2020 ◽  
Author(s):  
Rachel Emily Levene ◽  
Shailab D. Shrestha ◽  
Marta Maria Gaglia

ABSTRACTThe influenza A endoribonuclease PA-X regulates virulence and transmission of the virus by reducing host gene expression and thus regulating immune responses to influenza A virus. Despite this key function in viral biology, the levels of PA-X protein remain markedly low during infection, and previous results suggest that these low levels are not solely the result of regulation of the level of translation and RNA stability. How PA-X is regulated post-translationally remains unknown. We now report that the PA-X protein is rapidly turned over. PA-X from multiple viral strains are short-lived, although the half-life of PA-X ranges from ∼30 minutes to ∼3.5 hours depending on the strain. Moreover, sequences in the variable PA-X C-terminal domain are primarily responsible for regulating PA-X half-life, although the N-terminal domain also accounts for some differences among strains. Interestingly, we find that the PA-X from the 2009 pandemic H1N1 strain has a longer half-life compared to the other variants we tested. This PA-X isoform has been reported to have a higher host shutoff activity, suggesting a role for protein turnover in regulating PA-X activity. Collectively, this study reveals a novel regulatory mechanism of PA-X protein levels that may impact host shutoff activity during influenza A virus infection.IMPORTANCEThe PA-X protein from influenza A virus reduces host immune responses to infection through suppressing host gene expression, including genes encoding the antiviral response. Thus, it plays a central role in influenza A virus biology. Despite its key function, PA-X was only discovered in 2012 and much remains to be learned including how PA-X activity is regulated to promote optimal levels of viral infection. In this study, we reveal that PA-X protein levels are very low likely because of rapid turnover. We show that instability is a conserved property among PA-X variants from different strains of influenza A virus, but that the half-lives of PA-X variants differ. Moreover, the longer half-life of PA-X from the 2009 pandemic H1N1 strain correlates with its reported higher activity. Therefore, PA-X stability may be a way to regulate its activity and may contribute to the differential virulence of influenza A virus strains.


Pathogens ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 400 ◽  
Author(s):  
Pragyesh Dhungel ◽  
Fernando M. Cantu ◽  
Joshua A. Molina ◽  
Zhilong Yang

The synthesis of host cell proteins is adversely inhibited in many virus infections, whereas viral proteins are efficiently synthesized. This phenomenon leads to the accumulation of viral proteins concurrently with a profound decline in global host protein synthesis, a phenomenon often termed “host shutoff”. To induce host shutoff, a virus may target various steps of gene expression, as well as pre- and post-gene expression processes. During infection, vaccinia virus (VACV), the prototype poxvirus, targets all major processes of the central dogma of genetics, as well as pre-transcription and post-translation steps to hinder host cell protein production. In this article, we review the strategies used by VACV to induce host shutoff in the context of strategies employed by other viruses. We elaborate on how VACV induces host shutoff by targeting host cell DNA synthesis, RNA production and processing, mRNA translation, and protein degradation. We emphasize the topics on VACV’s approaches toward modulating mRNA processing, stability, and translation during infection. Finally, we propose avenues for future investigations, which will facilitate our understanding of poxvirus biology, as well as fundamental cellular gene expression and regulation mechanisms.


2020 ◽  
Vol 17 (1) ◽  
Author(s):  
Tianqiong He ◽  
Mingshu Wang ◽  
Anchun Cheng ◽  
Qiao Yang ◽  
Ying Wu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document