ArticlePDF Available

Deactivation of the antiviral state by rabies virus through targeting and accumulation of persistently phosphorylated STAT1

PLOS
PLOS Pathogens
Authors:

Abstract and Figures

Antagonism of the interferon (IFN)-mediated antiviral state is critical to infection by rabies virus (RABV) and other viruses, and involves interference in the IFN induction and signaling pathways in infected cells, as well as deactivation of the antiviral state in cells previously activated by IFN. The latter is required for viral spread in the host, but the precise mechanisms involved and roles in RABV pathogenesis are poorly defined. Here, we examined the capacity of attenuated and pathogenic strains of RABV that differ only in the IFN-antagonist P protein to overcome an established antiviral state. Importantly, P protein selectively targets IFN-activated phosphorylated STAT1 (pY-STAT1), providing a molecular tool to elucidate specific roles of pY-STAT1. We find that the extended antiviral state is dependent on a low level of pY-STAT1 that appears to persist at a steady state through ongoing phosphorylation/dephosphorylation cycles, following an initial IFN-induced peak. P protein of pathogenic RABV binds and progressively accumulates pY-STAT1 in inactive cytoplasmic complexes, enabling recovery of efficient viral replication over time. Thus, P protein-pY-STAT1 interaction contributes to ‘disarming’ of the antiviral state. P protein of the attenuated RABV is defective in this respect, such that replication remains suppressed over extended periods in cells pre-activated by IFN. These data provide new insights into the nature of the antiviral state, indicating key roles for residual pY-STAT1 signaling. They also elucidate mechanisms of viral deactivation of antiviral responses, including specialized functions of P protein in selective targeting and accumulation of pY-STAT1.
This content is subject to copyright.
RESEARCH ARTICLE
Deactivation of the antiviral state by rabies
virus through targeting and accumulation of
persistently phosphorylated STAT1
Gayathri Manokaran
1
, Michelle D. Audsley
1
, Haruka Funakoda
2
, Cassandra T. David
1
,
Katherine A. Garnham
1
, Stephen M. Rawlinson
1
, Celine Deffrasnes
1
, Naoto Ito
2‡
*,
Gregory W. MoseleyID
1‡
*
1Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia,
2Laboratory of Zoonotic Diseases, Joint Department of Veterinary Medicine, Faculty of Applied Biological
Sciences, Gifu University, Gifu, Japan
These authors contributed equally to this work.
NI and GWM also contributed equally to this work.
*naotoito@gifu-u.ac.jp (NI); greg.moseley@monash.edu (GWM)
Abstract
Antagonism of the interferon (IFN)-mediated antiviral state is critical to infection by rabies
virus (RABV) and other viruses, and involves interference in the IFN induction and signaling
pathways in infected cells, as well as deactivation of the antiviral state in cells previously
activated by IFN. The latter is required for viral spread in the host, but the precise mecha-
nisms involved and roles in RABV pathogenesis are poorly defined. Here, we examined the
capacity of attenuated and pathogenic strains of RABV that differ only in the IFN-antagonist
P protein to overcome an established antiviral state. Importantly, P protein selectively tar-
gets IFN-activated phosphorylated STAT1 (pY-STAT1), providing a molecular tool to eluci-
date specific roles of pY-STAT1. We find that the extended antiviral state is dependent on a
low level of pY-STAT1 that appears to persist at a steady state through ongoing phosphory-
lation/dephosphorylation cycles, following an initial IFN-induced peak. P protein of patho-
genic RABV binds and progressively accumulates pY-STAT1 in inactive cytoplasmic
complexes, enabling recovery of efficient viral replication over time. Thus, P protein-pY-
STAT1 interaction contributes to ‘disarming’ of the antiviral state. P protein of the attenuated
RABV is defective in this respect, such that replication remains suppressed over extended
periods in cells pre-activated by IFN. These data provide new insights into the nature of the
antiviral state, indicating key roles for residual pY-STAT1 signaling. They also elucidate
mechanisms of viral deactivation of antiviral responses, including specialized functions of P
protein in selective targeting and accumulation of pY-STAT1.
Author summary
Following viral infection, the host activates multiple antiviral defenses. The ability of
viruses to overcome these defenses is critical to disease. The earliest antiviral response
involves the production of interferon messenger molecules. Interferons act on infected
PLOS PATHOGENS
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 1 / 22
a1111111111
a1111111111
a1111111111
a1111111111
a1111111111
OPEN ACCESS
Citation: Manokaran G, Audsley MD, Funakoda H,
David CT, Garnham KA, Rawlinson SM, et al.
(2022) Deactivation of the antiviral state by rabies
virus through targeting and accumulation of
persistently phosphorylated STAT1. PLoS Pathog
18(5): e1010533. https://doi.org/10.1371/journal.
ppat.1010533
Editor: Matthias Johannes Schnell, Thomas
Jefferson University - Center City Campus: Thomas
Jefferson University, UNITED STATES
Received: November 4, 2021
Accepted: April 19, 2022
Published: May 16, 2022
Copyright: ©2022 Manokaran et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data Availability Statement: All relevant data are
within the manuscript and its Supporting
information files.
Funding: This research was supported by National
Health and Medical Research Council Australia
project grants 1125704 and 1160838 (G.W.M.)
https://www.nhmrc.gov.au/, Australian Research
Council discovery project grant DP150102569 and
DP210100998 (G.W.M) https://www.arc.gov.au/,
cells to inhibit viral proliferation, as well as on non-infected cells to establish an antiviral
state before infection and so limit viral spread through the host organism. Many strategies
used by viruses to overcome the former are well understood, but mechanisms important
to the latter, and their importance to disease, are less well defined. In this study, we inves-
tigated how rabies virus overcomes a pre-established antiviral state in target cells. We
found that the capacity to disable the antiviral state correlates with the ability to cause dis-
ease, and involves binding of a viral protein to cellular signaling proteins, which our data
indicate are responsible for the maintenance of a prolonged antiviral state. This advances
our understanding of antiviral responses, and identifies a key step in lethal infection by
rabies virus that causes approximately 60,000 human deaths per year. The findings may
contribute to new approaches for the development of vaccines or antivirals.
Introduction
The principal response of cells to viral infection is activation of the type-I interferon (IFN-I)
system. Following detection of virus, IFN-I is induced and released from infected cells and sig-
nals in autocrine (feedback to the infected cell) or paracrine (signaling to neighboring cells)
fashion, by binding to the type-I IFN receptor (IFNAR) [1,2]. This results in phosphorylation
of signal transducers and activators of transcription (STAT)1 and STAT2 [1,2] at conserved
tyrosines (Y701 in STAT1), generating pY-STATs. In a complex with IRF9, pY-STAT1/2 accu-
mulate in the nucleus and bind to IFN-stimulated response elements (ISREs), activating hun-
dreds of IFN-regulated genes (IRGs) including antiviral genes; this establishes an intrinsic
cellular antiviral state, and facilitates the development of an adaptive immune response [1,2].
Following IFN activation of cells, STAT1 phosphorylation occurs rapidly, reaching a peak
of pY-STAT1 within c. 30–60 min, during which pY-STAT1 accumulates in the nucleus to
activate IRGs. pY-STAT1-DNA binding and transcriptional activation is followed by release of
STATs from the DNA, dephosphorylation by nuclear phosphatases, and return to the cyto-
plasm [314]. Further phosphorylation/dephosphorylation cycles can support IRG regulation
until IFN signaling is turned off. Thus, STAT1 appears to dynamically monitor IFNAR activity
[15]. Peak pY-STAT1 levels and nuclear accumulation are typically lost within several hours,
which is associated with depleted IRG expression [3,510,12,13,1619]. Regulatory proteins
including phosphatases and SOCS proteins turn off the IFN response, with several of these
being IRGs that form negative feedback pathways [3,5].
These mechanisms cooperate to produce an appropriate response and avoid damage
through excessive IFN signaling/IRG activity [3,10]. Nevertheless, following initial stimulation,
the antiviral state can persist for extended periods, presumably to support viral clearance
[10,20]. pY-STAT1 is required to establish an antiviral state [21] but is rapidly depleted by
dephosphorylation, so the extended antiviral state is thought to involve unphosphorylated (U-
) STAT1. As STAT1 is an IRG [9,10], U-STAT1 levels are elevated following IFN stimulation,
and appear to induce prolonged expression of specific IRG subsets. This is proposed to main-
tain IRGs important to virus clearance, while depleting ‘early’ pY-STAT1-dependent IRGs that
are potentially damaging if expressed for extended periods [9,10].
Viral subversion of the IFN response is central to infection and pathogenesis [1,2,22] and is
mediated by viral IFN-antagonist proteins, which are often multifunctional and target several
stages of the response (IFN induction, IFN signaling and IRG effector functions) via numerous
mechanisms [2225]. The ability of many viruses to block IFN induction [22] raises the ques-
tion of why IFN signaling and effector stages are also inhibited. This likely relates to ‘leakiness’
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 2 / 22
and the Japan Society for the Promotion of Science
grant 18KK0192 (N.I.) https://www.jsps.go.jp/
english/index.html The funders had no role in study
design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing interests: I have read the journal’s
policy and the authors of this manuscript have the
following competing interests: G.W.M. holds a
patent application (PCT/AU2019/050908) and
Australian provisional patent (No. 201901137);
‘‘Novel Viruses’’ 2019.
in antagonism, so that some IFN is still released from infected cells, necessitating inhibition of
pY-STAT activated by autocrine signaling. Importantly, paracrine signaling also activates non-
infected cells to establish a protective antiviral state [1,2]. Furthermore, immune cells such as
plasmacytoid dendritic cells, which are often not productively infected, produce IFNs during
infections, resulting in paracrine signaling [2629]. Thus, progeny virus must overcome an
established antiviral state in target cells to spread within the host. However, analyses of viral
IFN antagonism often use systems in which infection or viral protein expression is established
prior to IFN treatment (e.g. [8,16,17,3034]); while this provides important information on
antagonistic mechanisms, it essentially mimics an autocrine response. As a result, for many
viruses, knowledge on the mechanisms used to overcome a preexisting antiviral state, and
their significance to pathogenesis, remains limited.
The main IFN antagonist of RABV is P protein [24], which can suppress IFN induction by
blocking IRF3/IRF7 phosphorylation [35,36], and can antagonize signaling by STATs via
physical interaction [8,16,17,25,3032,37,38]. P protein also interacts with the IRG PML (pro-
myelocytic leukemia) protein [39], and so may affect IRG functions. The P protein C-terminal
domain binds directly to STAT1 but, in contrast to many other antagonists [22], does not
affect STAT1 expression or tyrosine phosphorylation. Rather, P protein effects nuclear exclu-
sion of STAT1 via a strong nuclear export sequence (N-NES) in the N-terminus of full-length
P protein, and inhibits DNA interaction [8,16,17,25,30,4043]. Intriguingly, efficient P-pro-
tein-STAT1 interaction is dependent on IFN-treatment, such that P protein appears to antago-
nize pY-STAT1 selectively [16,30,32]. P protein-pY-STAT1 binding also inhibits
dephosphorylation so that in cells pre-infected by virus or expressing P protein, IFN treatment
causes a rapid cytoplasmic accumulation of pY-STAT1 that persists at high levels for extended
periods [8,16,17]. This is suggested to result from impaired nuclear accumulation and interac-
tion with nuclear phosphatases [8,16,17]. Thus, pY-STAT1 recycling is impaired as part of P
protein’s inhibitory mechanism [16].
The importance of P protein-mediated STAT1 antagonism in the pathogenesis of RABV
was revealed using the pathogenic Nishigahara (Ni) RABV strain that causes lethal infection in
mice [25,44,45], and an attenuated Ni-derived strain (Ni-CE), which is defective in antagonism
of STAT1 signaling, and non-pathogenic in mice. Substitution of the Ni P gene (which
encodes P protein) into Ni-CE generated the CE(NiP) strain, in which a pathogenic phenotype
was restored (Fig 1A), correlating with potent IFN/STAT-antagonistic function of Ni P protein
compared with Ni-CE P protein, in which antagonistic function is strongly impaired [24,45].
Ni-CE P protein can bind to STAT1, but is defective for nuclear export due to substitutions in
the N-NES, and so cannot efficiently inhibit pY-STAT1 nuclear localization [24,25]. Thus, P
protein is a pathogenesis factor, dependent on STAT1 antagonism. Importantly, RABV is neu-
rotropic and during infection of the CNS, astrocytes are the major IFN producers. As astro-
cytes are abortively infected they are not subject to efficient viral antagonism of IFN induction
[28], so the capacity of RABV to invade neurons pre-activated by paracrine signaling is
expected to be critical to disease. However, the potential importance of P protein/STAT1
antagonism in the disarming of an established antiviral state, and its relationship with patho-
genesis, has not been examined.
Here, we assessed the replication of CE(NiP) and Ni-CE in IFN pre-activated cells, finding
that, while replication is initially suppressed, CE(NiP), but not Ni-CE, progressively recovered,
indicating roles for P protein in overcoming an established antiviral state. Significant IFN sig-
naling was detectable >12 h post-IFN treatment, correlating with a persistent, low level of
pY-STAT1. The signaling was strongly inhibited by Ni P protein through progressive cyto-
plasmic accumulation of pY-STAT1, but Ni-CE P protein was defective for this antagonistic
function. Thus, it appears that following initial IFN activation, prolonged phosphorylation/
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 3 / 22
dephosphorylation cycling of STAT1 persists to maintain an antiviral state. By recruiting
pY-STAT1 into inactive complexes via P protein, pathogenic RABV progressively deactivates
the antiviral state. In contrast, Ni-CE P protein binds pY-STAT1, but does not effect
efficient cytoplasmic retention, resulting in prolonged signaling that correlates with viral
attenuation.
Fig 1. IFN sensitivity of Ni-CE and CE(NiP) viruses is dependent on MOI. (A,B) Schematic representation of genomes of Ni, Ni-CE and CE(NiP)
viruses with relative pathogenicity (A), and of viruses containing the luciferase (Luc) gene (B). (C) SK-N-SH cells infected with the indicated virus at
MOI of 0.01 or MOI of 2 were incubated in media containing IFN (+ IFN) or without IFN (No add.) at 500 U/ml for 3 or 2 days, respectively, before
collection of supernatant and titration (mean with SD, n = 3). , p 0.001; , p 0.0001.
https://doi.org/10.1371/journal.ppat.1010533.g001
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 4 / 22
Methods
Plasmids and cell culture
Constructs to express wild-type and mutated Ni and Ni-CE P proteins and CVS-N protein are
described elsewhere [8,16,25,46]. Plasmids used for the luciferase reporter gene assays were
pISRE-luc (Stratagene) and pRL-TK (Promega).
Human neuroblastoma SK-N-SH cells (ATCC number HTB-11) and mouse neuroblastoma
C1300 (NA) cells [47] were maintained in Eagle’s minimal essential medium supplemented
with 10% foetal calf serum. Lipofectamine 2000 (Invitrogen) was used for plasmid DNA trans-
fection into SK-N-SH cells. HEK293T and COS-7 cells were maintained in Dulbecco’s mini-
mal essential media supplemented with 10% foetal calf serum. Transfections were performed
using Lipofectamine 3000 (Invitrogen).
Viruses
RABV strains Ni and Ni-CE used in this study (Fig 1A) were previously recovered from cloned
cDNA [44,48]. The chimeric virus CE(NiP), which has the Ni P gene in the Ni-CE genome
background (Fig 1A) was generated by a reverse genetics approach [44]. Ni-CE and CE(NiP)
strains expressing firefly luciferase (Ni-CE-Luc and CE(NiP)-Luc, respectively) (Fig 1B) were
established in our previous study [49]. Stocks of all RABV strains were propagated in NA cells
and stored at -80˚C until use.
RABV infection assays
To assess the effect of multiplicity of infection (MOI) on the sensitivity of RABV to IFN,
SK-N-SH cells were inoculated with Ni-CE or CE(NiP) at MOI of 0.01 or MOI of 2 and then
were cultured in growth medium with or without 500 U/ml of human IFN-α2a (PBL Inter-
feron Source). Supernatant was collected at 3 or 2 days post-inoculation (dpi) for cultures
infected at MOI of 0.01 or MOI of 2, respectively, and stored at -80˚C. Virus titers were deter-
mined as reported previously [25].
To examine the effect of IFN treatment post-infection on RABV replication, SK-N-SH cells
were inoculated with Ni-CE-Luc or CE(NiP)-Luc at MOI of 3 and then cultured in growth
medium for 5 hours. At 6 hours post-inoculation (hpi), the medium was replaced with growth
medium with or without 500 U/ml of universal type I IFN (IFNα; PBL Interferon Source). The
cells were lysed at 12, 24 and 48 hpi and subjected to the luciferase assay using the Luciferase
Assay System (Promega) (see timeline in Fig 2A). To examine the effect of IFN treatment
before infection on RABV replication, SK-N-SH cells were incubated with or without 500 U/
ml of IFNαfor 12 hours before inoculation with Ni-CE-Luc or CE(NiP)-Luc at MOI of 3 for 1
h. After the inoculation, the cells were incubated again in growth medium for 12, 24 and 48
hpi (see timeline in Fig 2C); with the exception of the virus adsorption (for 1 h), the incubation
of cell samples in media either with or without 500U IFNαwas continuous throughout the
experiment (Fig 2C). Following incubation, cells were harvested for analysis by luciferase assay
(Fig 2C).
Luciferase reporter gene assays
To assess antagonism of IFN signalling induced following establishment of viral protein
expression (analogous to autocrine signalling following a viral infection), COS-7 cells were
transfected in triplicate with pRL-TK (40 ng), which expresses Renilla luciferase constitutively,
pISRE-luc (250 ng), which expresses firefly luciferase under the control of an ISRE-containing
promoter, and plasmid to express the indicated viral protein or empty vector (EV) (250 ng). 18
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 5 / 22
Fig 2. Ni-CE and CE(NiP) viruses are resistant to IFN signaling in infected cells but differin replication in cells
pre-activated by IFN. (A) and (C) show schematics of timelines for experiments shown in (B) and (D), respectively.
(A, B) SK-N-SH cells were infected with the indicated virus (MOI of 3), or mock infected, for 6 h before incubation in
media without (no add.) or with IFN (+ IFN) for the indicated times post-infection, and harvesting of cells for
measurement of luciferase activity. (C, D) SK-N-SH cells were incubated with or without IFN for 12 h before infection
with the indicated virus (MOI of 3), or mock infection, for the indicated times, and harvesting for measurement of
luciferase activity. Results of luciferase assays (B, D) show mean with SD (n = 3). n.s., p >0.05; , p 0.01.
https://doi.org/10.1371/journal.ppat.1010533.g002
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 6 / 22
h post-transfection, cells were incubated in media with or without 1000 U/ml IFNα(PBL
Interferon Source) for 12, 24 or 30 h. To assess IFN signalling activity and antagonism by viral
proteins expressed in cells following establishment of an antiviral state (analogous to infection
of cells following activation by paracrine signalling), COS-7 or HEK293T cells were treated
with 1000 U/ml IFNαfor 12 h before transfection as above and incubation for a further for 6,
12 or 18 h. Experiments using SK-N-SH cells to assess IFN signalling and antagonism by viral
proteins expressed following establishment of an antiviral state (analogous to paracrine signal-
ling) were performed similarly to those above except that cells were treated with or without
500 U/ml IFNα. Following the indicated incubation periods (see timelines in Figs 3A,4A and
4C), cells were lysed using passive lysis buffer (Promega) and lysates were subjected to a dual
luciferase assay as previously described [16,50]. Values for firefly luciferase activity were nor-
malised to those for Renilla luciferase by calculating the ratio of firefly to Renilla luminescence.
Western blot analysis and immunoprecipitation
To assess STAT1 phosphorylation/dephosphorylation following IFN treatment, non-trans-
fected COS-7 cells growing in 12 well plates were treated with 1000U IFNαfor periods from
0.5 to 60 h. To examine the effects of P protein expression on IFN-activated STAT1 phosphor-
ylation, COS-7 cells were incubated in media containing 1000U IFNα, or without IFN α, for
12 h before transfection with empty vector (EV), or with plasmid to express Ni or Ni-CE P
protein, Ni P protein mutated to substitute F209 and D235 for alanine (Ni-P mut), or CVS N
protein (1 μg plasmid per well), before incubation for periods from 6 h– 48h (see schematic
timeline in Fig 5B); the incubation in media either with or without IFNαwas continued
throughout the experiment. Following incubation, cells were washed with PBS and harvested
and lysed in lysis buffer (25mM Tris, 150mM NaCl, 0.2 mM EDTA, 0.5% IGEPAL; supple-
mented with cOmplete protease inhibitor and PhosSTOP). Cleared lysates were separated by
SDS-PAGE and transferred to a nitrocellulose membrane for immunoblotting analysis using
rabbit anti-pSTAT1 (CST #9167), rabbit anti-STAT1 (CST #14994), mouse anti-GFP (Abcam
Ab6556) or mouse anti-tubulin antibody (Sigma T8328), with either goat anti-rabbit HRP-
conjugated or goat anti-mouse HRP-conjugated secondary antibodies, before visualisation
using ECL Lightening Plus reagent.
To directly assess the interaction of viral proteins with pY-STAT1, COS-7 cells growing in 6
well plates were treated with 1000U IFNαfor 12 h before transfection to express the protein of
interest (3 μg plasmid per well). Cells were lysed 36 h post-transfection as above (see timeline
in Fig 5B). A portion of lysate was retained for analysis by western blotting and the remainder
was subjected to immunoprecipitation using GFP-Trap (Chromotek #gmta-20) according to
the manufacturer’s instructions. Immunoprecipitate and lysate were analysed by western blot-
ting as above.
Immunofluorescence staining and confocal laser scanning microscopy
(CLSM)
To assess pY-STAT1 activation and localization following IFN treatment, COS-7 cells growing
in a 12 well plate on coverslips were treated with 1000U IFNαfor periods from 0–48 h. To
assess effects of viral proteins on pY-STAT1 in cells pre-treated with IFN, cells were treated as
above for 12 h before transfection to express proteins of interest, and incubation for a further
12, 18, 24 or 36 h (see timeline in Fig 6B). Following incubations, cells were fixed in 4% formal-
dehyde (15 min, room temperature) before permeabilization in ice-cold 100% methanol (10
min, -20˚C). Cells were then blocked (1 x PBS (pH 7.4), 5% BSA, 0.3% Triton X-100) before
incubation with anti-pSTAT1 (CST #9167, 1/800 dilution in blocking buffer, 2 h, room
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 7 / 22
Fig 3. IFN/STAT1 signaling persists for over 12 h and is differentially inhibited by expression of Ni and Ni-CE P
proteins. (A) Timeline of experiments performed in (B); SK-N-SH cells were treated with or without IFN for 12 h
before transfection with plasmid to express Ni or Ni-CE P protein, or empty vector control (EV), together with
plasmids for the IFN/STAT1-dependent luciferase reporter assay; luciferase activity was measured 6 h or 12 h later. (B)
Results of luciferase assays show mean with SD (n = 3). n.s., p >0.05; , p 0.05; , p 0.01; , p 0.0001.
https://doi.org/10.1371/journal.ppat.1010533.g003
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 8 / 22
temperature) followed by anti-rabbit Alexa Fluor 568. Cells were mounted in Mowiol mount-
ing medium and imaged by CLSM using a Nikon C1 Inverted confocal microscope with 63x
oil objective (Monash Micro Imaging, Monash University). Digitised images were processed
using Fiji software (NIH).
Statistical analysis
Unpaired two-tailed Student’s t-test was performed using Prism software (version 8,
GraphPad).
Results
CE(NiP) but not Ni-CE virus can replicate in cells pre-activated by IFN
We previously found that infection of cells with Ni, CE(NiP) and Ni-CE virus at MOI of 0.01,
and incubation in media containing IFN following virus adsorption results in stronger inhibi-
tion of replication of Ni-CE virus than the viruses expressing Ni P protein [25]. Similar experi-
ments using CE(NiP) virus mutated to prevent P protein-STAT1 interaction indicated that
Fig 4. Ni-CE P protein is defective for antagonism of persistent IFN/STAT1 signaling. (A) Timeline of experiments performed in (B); COS-7 cells
were transfected with plasmids to express Ni P protein, Ni-CE P protein, or CVS-N protein (control) fused to the C-terminus of GFP, together with
plasmids for the IFN/STAT1-dependent luciferase reporter assay, followed by treatment with IFN at 18 h and harvesting of cell samples for luciferase
assays at the indicated times. (B) Results of dual luciferase assays, showing mean with SD (n = 3). (C) Timeline of experiments performed in (D) and
(E); COS-7 (D) or HEK293T (E) were incubated in media without or with IFN for 12 h before transfection with plasmids to express GFP-fused Ni or
Ni-CE P proteins or CVS-N protein, together with plasmids for the luciferase reporter assay; cells were then harvested at the indicated time points.
Results of luciferase assays using (D) COS-7 and (E) HEK293T cells showing mean with SD (n = 3). n.s., p >0.05; , p 0.05; , p 0.01; ,
p0.001; , p 0.0001.
https://doi.org/10.1371/journal.ppat.1010533.g004
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 9 / 22
Fig 5. P protein binds and accumulates residual pY-STAT1 in IFN pre-treated cells. (A) Non-transfected COS-7
cells were treated with IFN for the indicated periods before lysis and analysis by immunoblotting (IB) using the
indicated antibody. (B) Timeline of experiments shown in (C) and (D); COS-7 cells were incubated in media with or
without IFN and transfected 12 h later with empty vector (EV) or plasmids to express the indicated GFP-fused proteins
(C, D); cells were harvested at the indicated times for analysis of lysates by IB (blue arrows), or immunoprecipitation
for GFP (IP, red arrow). (C, D) IB analysis of cell lysates (C) and IPs (D) using the indicated antibodies.
https://doi.org/10.1371/journal.ppat.1010533.g005
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 10 / 22
infection at MOI of 0.001 followed by IFN treatment at 1 day post-infection (dpi) resulted in
substantially decreased titers of the mutant virus [32]. In both cases, the reduced resistance to
IFN correlated with attenuation, indicating that pathogenesis depends on IFN/STAT1 antago-
nism. However, since the MOIs used will result a mixed population of infected and non-
infected cells, the differences following IFN treatment could relate to suppression of replica-
tion in infected cells (analogous to autocrine signaling) and/or suppression of replication of
progeny virus following invasion of pre-activated cells (analogous to paracrine signaling).
Fig 6. Ni and Ni-CE P proteins cause accumulation of pY-STAT1 in different subcellularsites in IFN pre-treated cells. (A) Non-transfected COS-7
cells were treated with IFN for the indicated periods before fixation and immunostaining for pY-STAT1 and analysis by CLSM. (B) Timeline for
experiment shown in (C); COS-7 cells treated with IFN for 12 h were transfected with plasmids to express GFP-fused Ni P protein, Ni-CE P protein or
CVS-N protein, before fixation and immunostaining at the indicated times (blue arrows) for CLSM analysis. (C) Representative CLSM images are
shown; boxes in yellow are expanded in the zoom panel; identical microscope settings were used for all samples. Non-transfected cells or cells lacking
GFP expression (‘Cells’ panels in A, C) were visualized by enhancement of image brightness to detect auto-fluorescence. (D) Summary of results from
CLSM analysis showing the percentage of cells with detectable pY-STAT1 labelling; analysis included non-transfected cells (experiment shown in A),
and GFP-positive and GFP-negative cells in samples transfected with plasmid to express Ni P, Ni-CE P or N protein (experiment shown in C). Data
are from >15 fields of view (capturing >100 cells) for each condition; NT (non-transfected); , GFP not detectable. For values of 0%, pY-STAT1 was
not detected in 15 fields of view (>100 cells).
https://doi.org/10.1371/journal.ppat.1010533.g006
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 11 / 22
To investigate these possibilities, we examined the impact of MOI on replication following
IFN treatment by infecting SK-N-SH cells with Ni-CE or CE(NiP) virus at MOI of 0.01 or
MOI of 2. After virus adsorption, the cells were incubated with or without IFN (i.e. in media
containing recombinant IFN, or with no IFN added) (Fig 1C and 1D). In the MOI of 0.01 con-
dition, only a small proportion of cells will be infected before incubation with IFN, while in
the MOI of 2 condition, most or all cells will be infected. Thus, replication in the former condi-
tion involves (1) viral antagonism of IFN signaling in infected cells (mimicking autocrine sig-
naling) and (2) subversion of an antiviral state established in non-infected target cells before
invasion by progeny virus (mimicking paracrine signaling). In contrast, in the latter condition,
replication is primarily dependent on viral antagonism of IFN signaling in infected cells (auto-
crine signaling). By using viruses expressing P proteins with differing STAT1 antagonist func-
tion and pathogenicity (Fig 1A), this experiment also assessed the role of P protein in
replication under the different conditions, and its potential relationship to disease. Titers were
measured after 3 dpi (for MOI of 0.01) or 2 dpi (for MOI of 2); the measurement at 2 dpi for
the higher MOI is necessitated due to strong cytopathic effects of the viruses by 3 dpi.
In the MOI of 0.01 experiment, production for both viruses was inhibited by IFN, but Ni-
CE virus was substantially more sensitive to IFN than CE(NiP), indicated by a decrease in titer
of Ni-CE virus by 5.2 log following addition of IFN, whereas the titer of CE(NiP) was reduced
by only 3.0 log (Fig 1C). In contrast, in the MOI of 2 experiment, IFN had minimal effects on
the replication of either virus, with little difference observed between the different viruses
(titers reduced by only 1.1 and 0.7 log for Ni-CE and CE(NiP), respectively, following IFN
treatment) (Fig 1D). These data are consistent with RABV being relatively resistant to the pro-
duction of IFN-dependent antiviral mechanisms in cells where an active infection is estab-
lished, but susceptible to a pre-established antiviral state, such that spread into pre-activated
cells following low MOI infection is inhibited. Importantly, the greater sensitivity to IFN of
Ni-CE virus compared with CE(NiP) virus at low MOI (Fig 1C) suggested a major difference
in the capacity to surmount an established antiviral state, due to differing P protein function.
However, the less potent STAT1-antagonist function of Ni-CE P protein compared with Ni P
protein [25] appears sufficient to effectively suppress the establishment of an antiviral state by
IFN signaling in infected cells (Fig 1D).
To directly assess viral replication in cells treated with IFN pre- or post-infection, we used
CE(NiP) and Ni-CE virus engineered to express luciferase through insertion of a luciferase
gene in the G-L intergenic region [49] (Fig 1B), which enables measurement of primary and
secondary transcription. SK-N-SH cells were infected at high MOI (MOI of 3) for 6 h before
treatment with or without IFN by incubation in media containing 500 U/ml recombinant IFN
or media with no IFN added (Fig 2A and 2B); alternatively, cells were infected at MOI of 3 fol-
lowing a 12 h incubation in media with or without IFN (Fig 2C and 2D). The incubation of
cells either with or without IFN was continued for the remainder of the experiment. The first
condition, in which cells are infected before incubation with IFN, assesses effects of IFN on an
established infection (similar to autocrine signaling), and the second condition, in which cells
are incubated with IFN before infection, assesses effects of an established antiviral state on sub-
sequent infection (similar to infection of cells previously activated by paracrine signaling).
Consistent with the experiments using different MOIs (Fig 1C and 1D), treatment following
establishment of infection did not substantially affect transcription/replication and there was
no significant difference between the luciferase activity in Ni-CE and CE(NiP)-infected cells at
24 or 48 h post-infection (Fig 2B). In IFN pre-treated cells, there was a strong antiviral effect at
12 h post-infection, indicated by clearly reduced luciferase activity of both viruses compared to
that in non-treated cells (Fig 2D). However, luciferase activity increased in CE(NiP)-infected
cells at 24 h and 48 h. In contrast, there was no significant increase in luciferase activity in Ni-
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 12 / 22
CE-infected cells (Fig 2D). This indicated that IFN treatment induces a persistent antiviral
state that prevents replication by the attenuated Ni-CE virus for up to 60 h post-treatment
(infection for longer periods was not possible due to cytopathic effects of infection). While ini-
tially impeded by the antiviral state, CE(NiP) virus can progressively reverse this state,
enabling transcription/replication to recover (Fig 2D).
Taken together, these data indicate that the residual IFN antagonist activity of Ni-CE P pro-
tein is sufficient to suppress IFN signaling and the emergence of an antiviral state in pre-
infected cells, but not to reverse an established antiviral state in IFN-pretreated cells. Thus, dif-
ferences in P protein, which correlate with pathogenesis, relate to viral ‘disarming’ of the anti-
viral state.
IFN/STAT1 signaling persists over extended periods and is differentially
antagonized by Ni and Ni-CE P protein
The differing replication of CE(NiP) and Ni-CE in IFN pre-activated cells indicated that the
Ni and Ni-CE P proteins differ in a mechanism to inactivate the antiviral state. The capacity of
virus to replicate in IFN-activated cells is likely to depend at least in part on subversion of anti-
viral functions of certain IRGs [22]. However, while P protein interacts with PML and can
affect PML nuclear body structures [39], specific functions of P protein in antagonizing anti-
RABV effector functions of PML or other IRGs have not been reported, to our knowledge.
Since CE(NiP) and Ni-CE viruses, and P proteins thereof, differ in antagonism of IFN/
pY-STAT1 signaling in pre-infected cells or cells pre-transfected to express P protein [16,25],
we considered that this might be an important mechanism in disarming of a pre-established
antiviral state by RABV. Such a mechanism appears contrary to conventional understanding
that pY-STAT1 is dephosphorylated and inactivated within hours of IFN treatment, but
extended STAT1-dependent IRG transcription has been reported [9,10,20]. Thus, we hypothe-
sized that residual pY-STAT1 might support persistent signaling to maintain the antiviral
state, so that antagonism of pY-STAT1 would progressively disable the response, with the
activity of Ni P protein superior to that of Ni-CE P protein.
We examined IFN-I/STAT1/2 signaling using an ISRE-dependent luciferase reporter gene
assay as previously described [8,16,25,30,32]. We previously used ‘conventional’ assays where
cells are treated with IFN following transfection with luciferase plasmids and either co-trans-
fection with plasmids to express viral proteins, or infection by virus; this approach showed that
Ni-CE P protein is less able to inhibit IFN/STAT1 signaling than Ni-P protein, in transfection
and infection systems [25]. Importantly, for the current study we included a different approach
where cells were treated with IFN for 12 h before transfection of luciferase constructs and viral
protein expression constructs, and subsequent measurement of luciferase activity (Figs 3A and
4C). As the luciferase plasmids only report transcription after transfection, these assays will
only measure signaling persisting after 12 h IFN treatment. Because Ni-CE virus transcription/
replication (and, hence, P protein expression) is prevented by pre-treatment with IFN (Fig
2D), we used transfection rather than infection to compare effects of Ni-CE and Ni P protein
on persistent IFN/STAT signaling.
In SK-N-SH cells treated with IFN for 12 h before transfection for 6 h or 12 h, substantial
luciferase activity was detected compared with untreated cells, indicative of persistent signaling
(Fig 3B). At 6 h post-transfection, Ni P protein moderately suppressed IFN signaling, with
much greater inhibition apparent at 12 h post-transfection (c. 60% reduction), while Ni-CE P
protein had little to no effect (Fig 3B). As P protein selectively targets pY-STAT1, these data
suggest that the persistent signaling is due, at least in part, to pY-STAT1, which is strongly
antagonized by Ni P protein, with Ni-CE P protein partially defective [25]. Thus, the extended
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 13 / 22
antiviral state appears to involve prolonged pY-STAT1 signaling, with the differing recovery of
replication by CE(NiP) and Ni-CE viruses in IFN pre-activated cells (Fig 2D) relating to a dif-
fering capacity to antagonize this signaling.
To confirm these results and enable further mechanistic analysis, we repeated the analysis
using COS-7 cells transfected to express GFP-fused P proteins or GFP-fused to the nucleopro-
tein of challenge virus standard strain RABV (CVS N protein, which does not bind or antago-
nize to STAT1 and represents a standard control), as previously used for analysis of RABV/
STAT1 interactions [8,16,25,32,37]. Assays of cells transfected prior to IFN treatment (Fig 4A)
confirmed a substantial inhibition of IFN signaling by Ni P protein, with the antagonistic func-
tion of Ni-CE P protein clearly reduced (Fig 4 B).
In cells pretreated with IFN for 12 h before transfection (Fig 4C and 4D), persistent IFN/
STAT signaling was evident, and this was inhibited by Ni P protein at all times post-transfec-
tion, while antagonism by Ni-CE P protein was reduced (Fig 4D). Similar analysis of P proteins
in HEK293T cells also indicated persistent signaling, which was inhibited by Ni P protein,
with a significant impairment in antagonistic function of Ni-CE P protein (Fig 4C and 4E).
Thus, IFN/STAT1 signaling persists beyond the expected time for pY-STAT1 dephosphoryla-
tion/inactivation [3,5,19], and differential antagonism of this signaling by Ni and Ni-CE P pro-
teins correlates with the capacity of virus expressing these proteins to replicate in cells pre-
activated by IFN (Fig 2C and 2D).
Residual levels of pY-STAT1 persist over extended periods in IFN-activated
cells
The above data indicated that, following initial activation and dephosphorylation of STAT1, a
level of pY-STAT1-dependent signaling persists and is required to maintain an antiviral state.
Although previous studies indicate that pY-STAT1 is rapidly depleted following the activation
peak, a number of reports indicate persistence of low levels of pY-STAT1 exceeding those in
non-treated cells, after extended IFN treatment (>48 h; e.g. [4,9,11,14,51,52]). We thus
hypothesized that some pY-STAT1 is retained following depletion, providing signaling suffi-
cient to support ongoing antiviral functions. Western blot analysis of pY-STAT1 in lysates of
COS-7 cells indicated that, as expected, pY-STAT1 was at low or undetectable levels in
untreated cells, but was strongly induced after 0.5 h IFN treatment (Fig 5A) [4,79,11,15,16].
Consistent with reports that pY-STAT1 is dephosphorylated from c. 1–3 h post-IFN treatment
[4,7,9,11,15], pY-STAT1 was clearly reduced at 6 h post-IFN treatment and subsequent time
points (Fig 5A). Nevertheless, residual pY-STAT1 was detectable and persisted for at least 60 h
post-treatment (Fig 5A); the signal for STAT1 also appeared to increase at later time points,
possibly relating to the fact that STAT1 is an IRG and so inducible by IFN [10]. These data
indicate that the persistent signaling in reporter assays likely involves residual pY-STAT1, con-
sistent with the observed inhibitory effect of P protein, a selective pY-STAT1 antagonist.
Expression of P protein in cells pre-activated by IFN causes accumulation
of pY-STAT1
The above data are consistent with persistent IFN signaling in activated cells occurring
through dynamic phosphorylation/dephosphorylation cycling of a fraction of cellular STAT1
to maintain an extended antiviral state. The alternative possibility is that a small population of
pY-STAT1 is retained, but does not cycle dynamically and somehow maintains ongoing trans-
activation function. Because P protein inhibits STAT1 dephosphorylation [8,16,17], it provides
a tool to examine these possibilities and so elucidate mechanisms of the innate immune
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 14 / 22
response: specifically, if STAT1 is undergoing phosphorylation/dephosphorylation cycles,
inhibition of the latter by expression of P protein should result in accumulation of pY-STAT1.
To confirm that P protein interacts with the residual pY-STAT1 in cells following pro-
longed IFN treatment, we analyzed pY-STAT1 in cells treated with IFN for 12 h before trans-
fection to express P proteins and controls (Fig 5B); the controls included transfection with
empty vector (EV) or with plasmids to express CVS-N protein, or Ni-P protein mutated to dis-
able the STAT1-binding site (F209A/D235A, Ni-P mut [8]). Western blot analysis of cell
lysates indicated that at 6 h post-transfection, little P protein expression was evident and levels
of pY-STAT1 were similar in all IFN-treated samples (Fig 5C). However, at 12 h post-transfec-
tion and subsequent time points, P protein expression was detected, and correlated with accu-
mulation of pY-STAT1 in cells expressing Ni and Ni-CE P proteins (Fig 5C). These data are
consistent with the finding that both proteins bind to STAT1 [25], and we confirmed an inter-
action with pY-STAT1 at 48 h post-IFN treatment (36 h post-transfection) by immunoprecipi-
tation of P protein (Fig 5B and 5D). pY-STAT1 levels in cells expressing controls were clearly
reduced compared with those in cells expressing Ni or Ni-CE P proteins, and no interaction
with pY-STAT1 was evident for any of the controls.
These findings indicate that phosphorylation/dephosphorylation of STAT1 is ongoing at
steady state to maintain a low level of pY-STAT1. Binding by P protein recruits pY-STAT1
into inhibitory complexes, which are not efficiently dephosphorylated, while ongoing phos-
phorylation generates more pY-STATs that are subsequently recruited. Of note, although Ni-
CE P protein caused accumulation of pY-STAT1, the level of pY-STAT1 in lysates and amount
of pY-STAT1 co-precipitated by Ni-CE P protein was reduced compared with that observed
for Ni P protein. Thus, Ni-CE P protein is less able to retain pY-STAT1 in inactive complexes,
correlating with defective antagonism.
Ni and Ni-CE P protein accumulate pY-STAT1 in different subcellular
locations
To assess accumulation of pY-STAT1 in the cellular context, we treated COS-7 cells with or
without IFN for various time periods before fixation and immunostaining for pY-STAT1, and
analysis by CLSM (Fig 6). As expected, in non-transfected cells, pY-STAT1 was not detected in
untreated cells but became apparent in nuclei of c. 100% cells after 0.5 h IFN treatment; follow-
ing 12–48 h treatment, pY-STAT1 was depleted to below detectable levels (Fig 6A and 6D);
this is consistent with the induction and depletion of pY-STAT1 observed in cell lysates (Fig
5A). We next examined the accumulation of pY-STAT1 in cells treated for 12 h before trans-
fection to express Ni- or Ni-CE P proteins or CVS-N protein for different time periods (Fig
6B). In cells transfected to express Ni P protein, pY-STAT1 became detectable in c. 17% of
GFP-positive cells at 12 h post-transfection, and increased over subsequent time points to be
detectable in 63% of cells at 36 h post-transfection (Fig 6C and 6D). pY-STAT1 was only
detected in samples treated with IFN, and in these samples was observed only in cells express-
ing GFP-Ni-P protein and not in non-expressing cells in the same field of view (Fig 6C).
pY-STAT1 also accumulated in cells expressing Ni-CE P protein, but was less readily detected
(c. 6% and 47% of cells at 12 h and 36 h post-transfection, respectively) (Fig 6C and 6D).
Importantly, in Ni P protein-expressing cells with detectable pY-STAT1, the proteins coloca-
lized almost exclusively in the cytoplasm, whereas in Ni-CE P protein-expressing cells, signifi-
cant nuclear localization of pY-STAT1 was observed (Fig 6C, zoom panel). Thus, it appears
that both proteins interact with pY-STAT1 in IFN pre-activated cells and inhibit dephosphory-
lation, but Ni-CE P protein cannot prevent nuclear localization. This is consistent with results
from cells transfected with P protein or infected before IFN treatment [25], which indicated
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 15 / 22
that the differing antagonistic function of Ni and Ni-CE P proteins relates to differing subcel-
lular trafficking of P proteins and, hence, P protein-STAT1 complexes. The defect of Ni-CE P
protein in inhibiting STAT1 nuclear accumulation and, consequently, transactivation function
would be expected to increase dephosphorylation and recycling of STAT1, which is likely to
account for the apparent reduction in pY-STAT1 in cells expressing Ni-CE P protein com-
pared with Ni P protein (Fig 5C). Together, these defects in Ni-CE P protein result in impaired
inhibition of persistent IFN signaling, correlating with viral attenuation.
Discussion
In this study, we have characterized a mechanism by which RABV can invade and replicate
within cells that are in a pre-established antiviral state, finding that targeting of pY-STAT1 is a
critical element in inactivation of the antiviral condition. Central to this is the observation that
maintenance of the extended antiviral state following IFN activation is dependent on persistent
residual signaling by STAT1. This includes a significant contribution by a fraction of STAT1
that appears to be undergoing dynamic phosphorylation/dephosphorylation cycles, consistent
with ongoing active signaling, which results in a low steady-state level of pY-STAT1. The data
indicate that tyrosine phosphorylation of only a small proportion of STAT1 is required to sup-
port IRG transcription, based on the substantial effect of the pY-STAT1-antagonist P protein
on signaling in cells pre-activated by IFN (Fig 4D and 4E). Persistent pY-STAT1 stimulation is
possibly required to counteract cellular negative regulatory mechanisms [1,3], such that viral
inhibition of pY-STAT1 signaling results in a dominance of cellular countermeasures, ulti-
mately deactivating the antiviral state.
At the molecular level, it appears that, following infection of pre-activated cells, P protein of
pathogenic RABV specifically targets the small population of pY-STAT1 and inhibits signaling
and dephosphorylation by retaining pY-STAT1 in inhibitory cytoplasmic complexes. The
remaining non-phosphorylated STAT1 continues to be activated, and is then recruited by P
protein. This progressive accumulation and inhibition of STATs appears to tip the balance
between antiviral mechanisms and viral transcription, ultimately enabling the virus to over-
whelm the antiviral state (summarized in model shown in Fig 7). P protein of the attenuated
Ni-CE strain retains a capacity to bind and accumulate pY-STAT1, but not to effect strong
cytoplasmic arrest so that the antagonistic function is impaired. While the residual function of
Ni-CE P protein appears sufficient to prevent the activation of strong antiviral effects in cells
where infection is established prior to IFN stimulation (Fig 2B), it cannot efficiently surmount
a pre-established antiviral state (Fig 2D). The former condition is analogous to initial/early
infection of the host and autocrine signaling, where significant expression of Ni-CE P protein,
and possibly other viral proteins that contribute to immune subversion [46,53], may be estab-
lished before substantial activation of STAT1. The latter condition is analogous to a more
advanced infection, where paracrine signaling (including from non-infected immune cells),
has activated target cells prior to infection, with the weaker antagonistic function and limited
expression of Ni-CE P protein by invading virus unable to reverse the antiviral state. In con-
trast, Ni P protein, that is expressed by pathogenic CE(NiP) virus, is effective in both ‘auto-
crine’ and ‘paracrine’ conditions due to its greater antagonistic function. The capacity of
RABV to overcome an established antiviral state is likely to be particularly important to spread
in the central nervous system (CNS), consistent with the attenuation of Ni-CE virus.
Although we detected pY-STAT1 at late time points of IFN treatment, substantial dephos-
phorylation occurred following the initial pY-STAT1 peak (Fig 5A). The low expression of
pY-STAT1 after extended IFN treatment perhaps accounts for the lack of detection of this
‘late’ pY-STAT1 in some studies. IRG expression is also reported to be reduced following an
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 16 / 22
early wave of induction by IFN [5], so it has been assumed that, following initial activation,
pY-STAT1 signaling is lost. However, the antiviral response is clearly extended, and this has
been attributed to IFN-induced U-STATs [9,10,20]. Our new data support key roles for persis-
tent pY-STAT1 in maintaining the antiviral state, consistent with observations elsewhere of
prolonged pY-STAT1 expression [4,9,11,14,51,52]. By showing that a specific inhibitor of
pY-STAT1 (P protein) inhibits persistent signaling >12 h post-IFN treatment, and that this
correlates with a reversal of the antiviral state, our data support a substantial role of pY-STAT1,
distinct from U-STAT1.
This model, however, does not rule out effects of P protein on U-STAT1. Although the P
protein CTD binds to U-STAT1 in vitro [8,30], the interaction is of low affinity [8] and signifi-
cant co-precipitation of P protein and STAT1 from mammalian cells is dependent on IFN,
indicating minimal U-STAT1 binding [8,16,17]. Nevertheless, the progressive recruitment of
pY-STAT1 into inhibitory complexes will ultimately reduce the pool of ‘by-stander’ U-STAT1.
Assuming that inhibition of U-STAT1 is important, this raises the question of why RABV P
protein selectively targets pY-STAT1 rather than ‘total’ STAT1 (a mechanism used by a num-
ber of other viruses [22]). This may relate to the life cycle of RABV, where incubation can
extend to months before retrograde invasion of the CNS and, ultimately, anti-retrograde
spread for transmission to new hosts [54,55]. Damage to infected cells would limit retrograde
and anti-retrograde spread as well as potentially impairing further transmission by disabling
rabid animals; indeed, RABV infection is typically associated with limited sequelae [24]. Spe-
cific targeting of pY-STAT1 presumably ensures that STATs are inhibited only as required
(during or following IFN activation), minimizing any impact on potential homeostatic func-
tions of U-STATs. pY-STAT1 targeting also ensures efficient deployment of P protein, which
has multiple roles in infection [1,24]. Furthermore, while U-STATs activate certain IRG sub-
sets, pY-STATs are expected to have more potent function, and so may present more critical
targets. Following infection of an IFN-activated cell, P protein concentration will initially be
limiting, so high-affinity binding to a minor but critical pY-STAT1 subpopulation of cellular
STAT1 may ‘gain a foothold’ in immune subversion until increased P protein expression
Fig 7. Proposed model for deactivation of the antiviral state by RABV P protein. In non-activated cells (A), STAT1
is largely unphosphorylated (black boxes); following IFN activation, high levels of pY-STAT1 (red boxes) are induced
rapidly (B) and regulate the expression of IRGs, resulting in the establishment of an antiviral state (dark grey cell, C).
pY-STAT1 is dephosphorylated, but a low residual level of phosphorylation/dephosphorylation cycling and signaling
by pY-STAT1 persists, contributing to the maintenance of a prolonged antiviral state (C). Following infection by
RABV, P protein targets pY-STAT1 recruiting it to inactive cytoplasmic complexes (blue box, D); low levels of STAT1
activation continue, resulting in progressive accumulation (E) of pY-STAT1 by P protein, inhibiting persistent
signaling to ultimately inactivate the antiviral state (F).
https://doi.org/10.1371/journal.ppat.1010533.g007
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 17 / 22
permits more broad antagonism. Such selective mechanisms may be particularly important for
P proteins and other IFN antagonists that inhibit STATs via direct binding/sequestration, and
so have a defined stoichiometric interaction, in contrast to viral proteins that use enzymatic
mechanisms [22]. Finally, based on our observation that pY-STAT1 can interact with com-
plexes containing RABV P protein and N protein [56], and reports that STATs can accumulate
within viral replication bodies [1,5759], it is possible that binding/accumulation of pY-STATs
not only contributes to immune evasion, but also has distinct roles in replication.
Supporting information
S1 Data. Excel spreadsheet containing, in separate sheets, the underlying numerical data
for Fig panels 1C, 1D, 2B, 2D, 3B, 4B, 4D, and 4E.
(XLSX)
Acknowledgments
We acknowledge the facilities and technical assistance of the Monash Micro Imaging Facility
(Monash University).
Author Contributions
Conceptualization: Naoto Ito, Gregory W. Moseley.
Data curation: Naoto Ito, Gregory W. Moseley.
Formal analysis: Gayathri Manokaran, Michelle D. Audsley, Haruka Funakoda, Cassandra T.
David, Katherine A. Garnham, Stephen M. Rawlinson, Celine Deffrasnes, Naoto Ito, Greg-
ory W. Moseley.
Funding acquisition: Naoto Ito, Gregory W. Moseley.
Investigation: Gayathri Manokaran, Michelle D. Audsley, Haruka Funakoda, Cassandra T.
David, Katherine A. Garnham, Stephen M. Rawlinson, Celine Deffrasnes, Naoto Ito, Greg-
ory W. Moseley.
Methodology: Gayathri Manokaran, Michelle D. Audsley, Haruka Funakoda, Cassandra T.
David, Katherine A. Garnham, Stephen M. Rawlinson, Celine Deffrasnes, Naoto Ito, Greg-
ory W. Moseley.
Project administration: Naoto Ito, Gregory W. Moseley.
Resources: Naoto Ito, Gregory W. Moseley.
Supervision: Celine Deffrasnes, Naoto Ito, Gregory W. Moseley.
Validation: Gayathri Manokaran, Michelle D. Audsley, Cassandra T. David, Stephen M. Raw-
linson, Celine Deffrasnes, Naoto Ito, Gregory W. Moseley.
Visualization: Gayathri Manokaran, Michelle D. Audsley, Haruka Funakoda, Cassandra T.
David, Katherine A. Garnham, Stephen M. Rawlinson, Celine Deffrasnes, Naoto Ito, Greg-
ory W. Moseley.
Writing original draft: Naoto Ito, Gregory W. Moseley.
Writing review & editing: Michelle D. Audsley, Cassandra T. David, Stephen M. Rawlinson,
Celine Deffrasnes, Naoto Ito, Gregory W. Moseley.
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 18 / 22
References
1. Harrison AR, Moseley GW. The Dynamic Interface of Viruses with STATs. J Virol. 2020; 94(22). Epub
2020/08/28. https://doi.org/10.1128/JVI.00856-20 PMID: 32847860
2. Audsley MD, Moseley GW. Paramyxovirus evasion of innate immunity: Diverse strategies for common
targets. World journal of virology. 2013; 2(2):57–70. Epub 2013/11/01. https://doi.org/10.5501/wjv.v2.i2.
57 PMID: 24175230
3. Chen W, Daines MO, Khurana Hershey GK. Turning off signal transducer and activator of transcription
(STAT): the negative regulation of STAT signaling. J Allergy Clin Immunol. 2004; 114(3):476–89; quiz
90. Epub 2004/09/10. https://doi.org/10.1016/j.jaci.2004.06.042 PMID: 15356544.
4. Majoros A, Platanitis E, Szappanos D, Cheon H, Vogl C, Shukla P, et al. Response to interferons and
antibacterial innate immunity in the absence of tyrosine-phosphorylated STAT1. EMBO Rep. 2016; 17
(3):367–82. Epub 2016/02/18. https://doi.org/10.15252/embr.201540726 PMID: 26882544
5. David M, Grimley PM, Finbloom DS, Larner AC. A nuclear tyrosine phosphatase downregulates inter-
feron-induced gene expression. Mol Cell Biol. 1993; 13(12):7515–21. Epub 1993/12/01. https://doi.org/
10.1128/mcb.13.12.7515-7521.1993 PMID: 8246969
6. Reich NC, Liu L. Tracking STAT nuclear traffic. Nat Rev Immunol. 2006; 6(8):602–12. Epub 2006/07/
27. https://doi.org/10.1038/nri1885 PMID: 16868551.
7. Sohn SY, Hearing P. Adenovirus sequesters phosphorylated STAT1 at viral replication centers and
inhibits STAT dephosphorylation. J Virol. 2011; 85(15):7555–62. Epub 2011/05/20. https://doi.org/10.
1128/JVI.00513-11 PMID: 21593149
8. Hossain MA, Larrous F, Rawlinson SM, Zhan J, Sethi A, Ibrahim Y, et al. Structural Elucidation of Viral
Antagonism of Innate Immunity at the STAT1 Interface. Cell Rep. 2019; 29(7):1934–45 e8. Epub 2019/
11/14. https://doi.org/10.1016/j.celrep.2019.10.020 PMID: 31722208.
9. Cheon H, Stark GR. Unphosphorylated STAT1 prolongs the expression of interferon-induced immune
regulatory genes. Proc Natl Acad Sci U S A. 2009; 106(23):9373–8. Epub 2009/05/30. https://doi.org/
10.1073/pnas.0903487106 PMID: 19478064
10. Cheon H, Yang J, Stark GR. The functions of signal transducers and activators of transcriptions 1 and 3
as cytokine-inducible proteins. J Interferon Cytokine Res. 2011; 31(1):33–40. Epub 2010/12/21. https://
doi.org/10.1089/jir.2010.0100 PMID: 21166594
11. Morrow AN, Schmeisser H, Tsuno T, Zoon KC. A novel role for IFN-stimulated gene factor 3II in IFN-
gamma signaling and induction of antiviral activity in human cells. J Immunol. 2011; 186(3):1685–93.
Epub 2010/12/24. https://doi.org/10.4049/jimmunol.1001359 PMID: 21178011
12. Koster M, Hauser H. Dynamic redistribution of STAT1 protein in IFN signaling visualized by GFP fusion
proteins. Eur J Biochem. 1999; 260(1):137–44. Epub 1999/03/26. https://doi.org/10.1046/j.1432-1327.
1999.00149.x PMID: 10091593.
13. ten Hoeve J, de Jesus Ibarra-Sanchez M, Fu Y, Zhu W, Tremblay M, David M, et al. Identification of a
nuclear Stat1 protein tyrosine phosphatase. Mol Cell Biol. 2002; 22(16):5662–8. Epub 2002/07/26.
https://doi.org/10.1128/MCB.22.16.5662-5668.2002 PMID: 12138178
14. Steen HC, Kotredes KP, Nogusa S, Harris MY, Balachandran S, Gamero AM. Phosphorylation of
STAT2 on serine-734 negatively regulates the IFN-alpha-induced antiviral response. J Cell Sci. 2016;
129(22):4190–9. Epub 2016/11/02. https://doi.org/10.1242/jcs.185421 PMID: 27802159
15. Wiesauer I, Gaumannmuller C, Steinparzer I, Strobl B, Kovarik P. Promoter occupancy of STAT1 in
interferon responses is regulated by processive transcription. Mol Cell Biol. 2015; 35(4):716–27. Epub
2014/12/17. https://doi.org/10.1128/MCB.01097-14 PMID: 25512607
16. Wiltzer L, Larrous F, Oksayan S, Ito N, Marsh GA, Wang LF, et al. Conservation of a uniquemechanism
of immune evasion across the Lyssavirus genus. J Virol. 2012; 86(18):10194–9. Epub 2012/06/29.
https://doi.org/10.1128/JVI.01249-12 PMID: 22740405
17. Brzozka K, Finke S, Conzelmann KK. Inhibition of interferon signaling by rabies virus phosphoprotein P:
activation-dependent binding of STAT1 and STAT2. J Virol. 2006; 80(6):2675–83. Epub 2006/02/28.
https://doi.org/10.1128/JVI.80.6.2675-2683.2006 PMID: 16501077
18. McBride KM, McDonald C, Reich NC. Nuclear export signal located within theDNA-binding domain of
the STAT1transcription factor. EMBO J. 2000; 19(22):6196–206. Epub 2000/11/18. https://doi.org/10.
1093/emboj/19.22.6196 PMID: 11080165
19. Larner AC, Chaudhuri A, Darnell JE Jr. Transcriptional induction by interferon. New protein(s) deter-
mine the extent and length of the induction. J Biol Chem. 1986; 261(1):453–9. Epub 1986/01/05. PMID:
2934388.
20. Jaitin DA, Schreiber G. Upregulation of a small subset of genes drives type I interferon-induced antiviral
memory. J Interferon Cytokine Res. 2007; 27(8):653–64. Epub 2007/09/06. https://doi.org/10.1089/jir.
2006.0162 PMID: 17784817.
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 19 / 22
21. Horvath CM, Darnell JE Jr. The antiviral state induced by alpha interferon and gamma interferon
requires transcriptionally active Stat1 protein. J Virol. 1996; 70(1):647–50. Epub 1996/01/01. https://doi.
org/10.1128/JVI.70.1.647-650.1996 PMID: 8523587
22. Versteeg GA, Garcia-Sastre A. Viral tricks to grid-lock the type I interferon system. Current opinion in
microbiology. 2010; 13(4):508–16. Epub 2010/06/12. https://doi.org/10.1016/j.mib.2010.05.009 PMID:
20538505
23. Audsley MD, Jans DA, Moseley GW. Roles of nuclear trafficking in infection by cytoplasmic negative-
strand RNA viruses: paramyxoviruses and beyond. J Gen Virol. 2016; 97:2463–81. https://doi.org/10.
1099/jgv.0.000575 PMID: 27498841
24. Ito N, Moseley GW, Sugiyama M. The importance of immune evasion in the pathogenesis of rabies
virus. J Vet Med Sci. 2016; 78(7):1089–98. Epub 2016/04/05. https://doi.org/10.1292/jvms.16-0092
PMID: 27041139
25. Ito N, Moseley GW, Blondel D, Shimizu K, Rowe CL, Ito Y, et al. Role of interferon antagonist activity of
rabies virus phosphoprotein in viral pathogenicity. J Virol. 2010; 84(13):6699–710. Epub 2010/04/30.
https://doi.org/10.1128/JVI.00011-10 PMID: 20427527
26. Assil S, Coleon S, Dong C, Decembre E, Sherry L, Allatif O, et al. Plasmacytoid Dendritic Cells and
Infected Cells Form an Interferogenic Synapse Required for Antiviral Responses. Cell Host Microbe.
2019; 25(5):730–45 e6. Epub 2019/04/21. https://doi.org/10.1016/j.chom.2019.03.005 PMID:
31003939.
27. Kallfass C, Ackerman A, Lienenklaus S, Weiss S, Heimrich B, Staeheli P. Visualizing production of beta
interferon by astrocytes and microglia in brain of La Crosse virus-infected mice. J Virol. 2012; 86
(20):11223–30. Epub 2012/08/10. https://doi.org/10.1128/JVI.01093-12 PMID: 22875966
28. Pfefferkorn C, Kallfass C, Lienenklaus S, Spanier J, Kalinke U, Rieder M, et al. Abortively
Infected Astrocytes Appear To Represent the Main Source of Interferon Beta in the Virus-Infected
Brain. J Virol. 2016; 90(4):2031–8. Epub 2015/12/15. https://doi.org/10.1128/JVI.02979-15 PMID:
26656686
29. Ali S, Mann-Nuttel R, Schulze A, Richter L, Alferink J, Scheu S. Sources of Type I Interferons in Infec-
tious Immunity: Plasmacytoid Dendritic Cells Not Always in the Driver’s Seat. Front Immunol. 2019;
10:778. Epub 2019/04/30. https://doi.org/10.3389/fimmu.2019.00778 PMID: 31031767
30. Vidy A, Chelbi-Alix M, Blondel D. Rabies virus P protein interacts with STAT1 and inhibits interferon sig-
nal transduction pathways. J Virol. 2005; 79(22):14411–20. Epub 2005/10/29. https://doi.org/10.1128/
JVI.79.22.14411-14420.2005 PMID: 16254375
31. Vidy A, El Bougrini J, Chelbi-Alix MK, Blondel D. The nucleocytoplasmic rabies virus P protein counter-
acts interferon signaling by inhibiting both nuclear accumulation and DNA binding of STAT1. J Virol.
2007; 81(8):4255–63. Epub 2007/02/09. https://doi.org/10.1128/JVI.01930-06 PMID: 17287281
32. Wiltzer L, Okada K, Yamaoka S, Larrous F, Kuusisto HV, Sugiyama M, et al. Interaction of Rabies Virus
P-Protein With STAT Proteins is Critical to Lethal Rabies Disease. J Infect Dis. 2014; 209(11):1744–53.
Epub 2013/12/25. https://doi.org/10.1093/infdis/jit829 PMID: 24367042.
33. Miorin L, Kehrer T, Sanchez-Aparicio MT, Zhang K, Cohen P, Patel RS, et al. SARS-CoV-2 Orf6 hijacks
Nup98 to block STAT nuclear import and antagonize interferon signaling. Proc Natl Acad Sci U S A.
2020; 117(45):28344–54. Epub 2020/10/25. https://doi.org/10.1073/pnas.2016650117 PMID:
33097660
34. Xia H, Cao Z, Xie X, Zhang X, Chen JY, Wang H, et al. Evasion of Type I Interferon by SARS-CoV-2.
Cell Rep. 2020; 33(1):108234. Epub 2020/09/28. https://doi.org/10.1016/j.celrep.2020.108234 PMID:
32979938
35. Masatani T, Ozawa M, Yamada K, Ito N, Horie M, Matsuu A, et al. Contribution of the interaction
between the rabies virus P protein and I-kappa B kinase to the inhibition of type I IFN induction signal-
ling. J Gen Virol. 2016; 97(2):316–26. https://doi.org/10.1099/jgv.0.000362 PMID: 26647356.
36. Brzozka K, Finke S, Conzelmann KK. Identification of the rabies virus alpha/beta interferon
antagonist: phosphoprotein P interferes with phosphorylation of interferon regulatory factor 3. J Virol.
2005; 79(12):7673–81. Epub 2005/05/28. https://doi.org/10.1128/JVI.79.12.7673-7681.2005 PMID:
15919920
37. Harrison AR, Lieu KG, Larrous F, Ito N, Bourhy H, Moseley GW. Lyssavirus P-protein selectively targets
STAT3-STAT1 heterodimers to modulate cytokine signalling. PLoS Pathog. 2020; 16(9):e1008767.
Epub 2020/09/10. https://doi.org/10.1371/journal.ppat.1008767 PMID: 32903273
38. Lieu KG, Brice A, Wiltzer L, Hirst B, Jans DA, Blondel D, et al. The rabies virus interferon antagonist P
protein interacts with activated STAT3 and inhibits Gp130 receptor signaling. J Virol. 2013; 87
(14):8261–5. Epub 2013/05/24. https://doi.org/10.1128/JVI.00989-13 PMID: 23698294
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 20 / 22
39. Blondel D, Regad T, Poisson N, Pavie B, Harper F, Pandolfi PP, et al. Rabies virus P and small P prod-
ucts interact directly with PML and reorganize PML nuclear bodies. Oncogene. 2002; 21(52):7957–70.
Epub 2002/11/20. https://doi.org/10.1038/sj.onc.1205931 PMID: 12439746.
40. Brice A, Whelan DR, Ito N, Shimizu K, Wiltzer-Bach L, Lo CY, et al. Quantitative Analysis of the Microtu-
bule Interaction of Rabies Virus P3 Protein: Roles in Immune Evasion and Pathogenesis. Sci Rep.
2016; 6:33493. Epub 2016/09/22. https://doi.org/10.1038/srep33493 PMID: 27649849
41. Moseley GW, Lahaye X, Roth DM, Oksayan S, Filmer RP, Rowe CL, et al. Dual modes of rabies P-pro-
tein association with microtubules: a novel strategy to suppress the antiviral response. J Cell Sci. 2009;
122(Pt 20):3652–62. Epub 2009/09/24. https://doi.org/10.1242/jcs.045542 PMID: 19773364.
42. Pasdeloup D, Poisson N, Raux H, Gaudin Y, Ruigrok RW, Blondel D. Nucleocytoplasmic shuttling of
the rabies virus P protein requires a nuclear localization signal and a CRM1-dependent nuclear export
signal. Virology. 2005; 334(2):284–93. Epub 2005/03/23. https://doi.org/10.1016/j.virol.2005.02.005
PMID: 15780878.
43. Rowe CL, Wagstaff KM, Oksayan S, Glover DJ, Jans DA, Moseley GW. Nuclear Trafficking of the
Rabies Virus Interferon Antagonist P-Protein Is Regulated by an Importin-Binding Nuclear Localization
Sequence in the C-Terminal Domain. PloS one. 2016; 11(3):e0150477. https://doi.org/10.1371/journal.
pone.0150477 PMID: 26939125
44. Shimizu K, Ito N, Mita T, Yamada K, Hosokawa-Muto J, Sugiyama M, et al. Involvement of nucleopro-
tein, phosphoprotein, and matrix protein genes of rabies virus in virulence for adult mice. Virus Res.
2007; 123(2):154–60. Epub 2006/10/03. https://doi.org/10.1016/j.virusres.2006.08.011 PMID:
17010466.
45. Shimizu K, Ito N, Sugiyama M, Minamoto N. Sensitivity of rabies virus to type I interferon is determined
by the phosphoprotein gene. Microbiology and immunology. 2006; 50(12):975–8. Epub 2006/12/21.
https://doi.org/10.1111/j.1348-0421.2006.tb03875.x PMID: 17179666.
46. Masatani T, Ito N, Shimizu K, Ito Y, Nakagawa K, Sawaki Y, et al. Rabies virus nucleoprotein functions
to evade activation of the RIG-I-mediated antiviral response. J Virol. 2010; 84(8):4002–12. Epub 2010/
02/05. https://doi.org/10.1128/JVI.02220-09 PMID: 20130065
47. McMorris FA, Ruddle FH. Expression of neuronal phenotypes in neuroblastoma cell hybrids. Dev Biol.
1974; 39(2):226–46. Epub 1974/08/01. https://doi.org/10.1016/0012-1606(74)90237-1 PMID: 4851982.
48. Yamada K, Ito N, Takayama-Ito M, Sugiyama M, Minamoto N. Multigenic relation to the attenuation of
rabies virus. Microbiol Immunol. 2006; 50(1):25–32. Epub 2006/01/24. https://doi.org/10.1111/j.1348-
0421.2006.tb03767.x PMID: 16428870.
49. Yamaoka S, Ito N, Ohka S, Kaneda S, Nakamura H, Agari T, et al. Involvement of the rabies virus phos-
phoprotein gene in neuroinvasiveness. J Virol. 2013; 87(22):12327–38. Epub 2013/09/13. https://doi.
org/10.1128/JVI.02132-13 PMID: 24027304
50. Audsley MD, Marsh GA, Lieu KG, Tachedjian M, Joubert DA, Wang LF, et al. The immune evasion
function of J and Beilong virus V proteins is distinct from that of other paramyxoviruses, consistent with
their inclusion in the proposed genus Jeilongvirus. J Gen Virol. 2016; 97(3):581–92. Epub 2015/12/26.
https://doi.org/10.1099/jgv.0.000388 PMID: 26703878.
51. Su L, David M. Distinct mechanisms of STAT phosphorylation via the interferon-alpha/beta receptor.
Selective inhibition of STAT3 and STAT5 by piceatannol. J Biol Chem. 2000; 275(17):12661–6. Epub
2000/04/25. https://doi.org/10.1074/jbc.275.17.12661 PMID: 10777558.
52. Krejcova D, Prochazkova J, Kubala L, Pachernik J. Modulation of cell proliferation and differentiation of
human lung carcinoma cells by the interferon-alpha. Gen Physiol Biophys. 2009; 28(3):294–301. Epub
2009/12/29. https://doi.org/10.4149/gpb_2009_03_294 PMID: 20037195.
53. Ben Khalifa Y, Luco S, Besson B, Sonthonnax F, Archambaud M, Grimes JM, et al. The matrix protein of
rabies virus binds to RelAp43 to modulate NF-kappaB-dependent gene expression related to innate immu-
nity. Sci Rep. 2016; 6:39420. Epub 2016/12/22. https://doi.org/10.1038/srep39420 PMID: 28000711
54. Jackson AC. Chapter 9—Pathogenesis. In: Fooks AR, Jackson, A.C., editor. Rabies (Fourth Edition):
Academic Press; 2020. p. 303–45.
55. Jackson AC. Rabies virus infection: an update. J Neurovirol. 2003; 9(2):253–8. Epub 2003/04/23.
https://doi.org/10.1080/13550280390193975 PMID: 12707856.
56. Zhan J, Harrison AR, Portelli S, Nguyen TB, Kojima I, Zheng S, et al. Definition of the immune evasion-
replication interface of rabies virus P protein. PLoS Pathog. 2021; 17(7):e1009729. Epub 2021/07/09.
https://doi.org/10.1371/journal.ppat.1009729 PMID: 34237115
57. Harrison AR, Todd S, Dearnley M, David CT, Green D, Rawlinson SM, et al. Antagonism of STAT3 sig-
nalling by Ebola virus. PLoS Pathog. 2021; 17(6):e1009636. Epub 2021/06/25. https://doi.org/10.1371/
journal.ppat.1009636 PMID: 34166464
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 21 / 22
58. Ulane CM, Rodriguez JJ, Parisien JP, Horvath CM. STAT3 ubiquitylation and degradation by mumps
virus suppress cytokine and oncogene signaling. J Virol. 2003; 77(11):6385–93. Epub 2003/05/14.
https://doi.org/10.1128/jvi.77.11.6385-6393.2003 PMID: 12743296
59. Palosaari H, Parisien JP, Rodriguez JJ, Ulane CM, Horvath CM. STAT protein interference and sup-
pression of cytokine signal transduction by measles virus V protein. J Virol. 2003; 77(13):7635–44.
Epub 2003/06/14. https://doi.org/10.1128/jvi.77.13.7635-7644.2003 PMID: 12805463
PLOS PATHOGENS
Deactivation of the antiviral state by rabies virus
PLOS Pathogens | https://doi.org/10.1371/journal.ppat.1010533 May 16, 2022 22 / 22
... However, all isoforms contain a STAT1binding site in the C-terminal domain (CTD), which enables antagonism of IFN/STAT1 signaling (19,20). Studies to date have focused on P1 and P3 and indicate that antagonism involves several mechanisms deriving from distinct phenotypes of the isoforms (21)(22)(23)(24)(25). P1 is primarily cytoplasmic due to a strong N-terminal nuclear export sequence (N-NES), and so it effects nuclear exclusion/cytoplasmic accumulation of associated STAT1 to prevent ISG activation (24)(25)(26). Conservation of the STAT1-binding region and N-NES, and equivalent localization of P2 (27), suggest that P2 effects a similar mechanism but is "specialized" for antagonism due to a lack of L binding. Although this has not been directly assessed, it is consistent with studies using viruses with modified P1/P2 expression (28). ...
... IFN induces phosphorylation of STAT1 at residue Y 701 (pY-STAT1), promoting nuclear import and ISG activation before dephosphorylation, which becomes evident from ;0.5 h after IFN treatment and ultimately deactivates STAT1 (47)(48)(49). P1 proteins use an unusual mechanism of IFN antagonism that is conserved among proteins of different lyssaviruses, whereby P1 binds efficiently to pY-STAT1 and inhibits its dephosphorylation, thus retaining inactive pY-STAT1-P1 complexes in the cytoplasm (24)(25)(26)38). As a result, P1 expression or RABV infection results in accumulation of pY-STAT1 in IFN-treated cells (24)(25)(26)38). ...
... P1 proteins use an unusual mechanism of IFN antagonism that is conserved among proteins of different lyssaviruses, whereby P1 binds efficiently to pY-STAT1 and inhibits its dephosphorylation, thus retaining inactive pY-STAT1-P1 complexes in the cytoplasm (24)(25)(26)38). As a result, P1 expression or RABV infection results in accumulation of pY-STAT1 in IFN-treated cells (24)(25)(26)38). Dephosphorylation of pY-STAT1 follows nuclear entry and DNA binding (to activate transcription) followed by release from the DNA (16). ...
Article
Full-text available
Viral hijacking of microtubule (MT)-dependent transport is well understood, but several viruses also express discrete MT-associated proteins (vMAPs), potentially to modulate MT-dependent processes in the host cell. Specific roles for vMAP-MT interactions include subversion of antiviral responses by P3, an isoform of the P protein of rabies virus (RABV; genus Lyssavirus), which mediates MT-dependent antagonism of interferon (IFN)-dependent signal transducers and activators of transcription 1 (STAT1) signaling. P3 also undergoes nucleocytoplasmic trafficking and inhibits STAT1-DNA binding, indicative of intranuclear roles in a multipronged antagonistic strategy. MT association/STAT1 antagonist functions of P3 correlate with pathogenesis, indicating potential as therapeutic targets. However, key questions remain, including whether other P protein isoforms interact with MTs, the relationship of these interactions with pathogenesis, and the extent of conservation of P3-MT interactions between diverse pathogenic lyssaviruses. Using super-resolution microscopy, live-cell imaging, and immune signaling analyses, we find that multiple P protein isoforms associate with MTs and that association correlates with pathogenesis. Furthermore, P3 proteins from different lyssaviruses exhibit variation in intracellular localization phenotypes that are associated with STAT1 antagonist function, whereby P3-MT association is conserved among lyssaviruses of phylogroup I but not phylogroup II, while nucleocytoplasmic localization varies between P3 proteins of the same phylogroup within both phylogroup I and II. Nevertheless, the divergent P3 proteins retain significant IFN antagonist function, indicative of adaptation to favor different inhibitory mechanisms, with MT interaction important to phylogroup I viruses. IMPORTANCE Lyssaviruses, including rabies virus, cause rabies, a progressive encephalomyelitis that is almost invariably fatal. There are no effective antivirals for symptomatic infection, and effective application of current vaccines is limited in areas of endemicity, such that rabies causes ~59,000 deaths per year. Viral subversion of host cell functions, including antiviral immunity, is critical to disease, and isoforms of the lyssavirus P protein are central to the virus-host interface underpinning immune evasion. Here, we show that specific cellular interactions of P protein isoforms involved in immune evasion vary significantly between different lyssaviruses, indicative of distinct strategies to evade immune responses. These findings highlight the diversity of the virus-host interface, an important consideration in the development of pan-lyssavirus therapeutic approaches.
... Reporter assays for IFN induction and IFN signaling have been described previously (59,(82)(83)(84)(85). Briefly, to measure the activity of RIG-I and MDA5 activation of IFNβ induction pathways and effects thereon of expression of BFV nsp3, HEK-293T cells were co-transfec ted in triplicate with 40 ng pRL-TK (which expresses Renilla luciferase (RLuc) under the control of the constitutively active thymidine kinase promoter), 250 ng pGL3-IFNβ (in which firefly luciferase (Fluc) is under the control of the IFN-β promoter) and 250 ng plasmids to express the indicated GFP-fused nsP3 proteins or the control proteins ORF6-StrepII [expressing the SARS-CoV-2 ORF6 protein, a potent inhibitor of RIG-I/ MDA5-activated IFN induction, fused to StrepII tag (82)] or pEGFP-CVS-N [expressing the nucleoprotein (N protein) of rabies virus, which lacks intrinsic antagonist activity to the IFN-induction pathway (86)]. ...
Article
Full-text available
Barmah Forest virus (BFV) is a mosquito-borne virus that causes arthralgia with accompanying rash, fever, and myalgia in humans. The virus is mainly found in Australia and has caused outbreaks associated with significant health concerns. As the sole representative of the Barmah Forest complex within the genus Alphavirus, BFV is not closely related genetically to other alphaviruses. Notably, basic knowledge of BFV molecular virology has not been well studied due to a lack of critical investigative tools such as an infectious clone. Here we describe the construction of an infectious BFV cDNA clone based on Genbank sequence and demonstrate that the clone-derived virus has in vitro and in vivo properties similar to naturally occurring virus, BFV field isolate 2193 (BFV2193-FI). A substitution in nsP4, V1911D, which was identified in the Genbank reference sequence, was found to inhibit virus rescue and replication. T1325P substitution in nsP2 selected during virus passaging was shown to be an adaptive mutation, compensating for the inhibitory effect of nsP4-V1911D. The two mutations were associated with changes in viral non-structural polyprotein processing and type I interferon (IFN) induction. Interestingly, a nuclear localization signal, active in mammalian but not mosquito cells, was identified in nsP3. A point mutation abolishing nsP3 nuclear localization attenuated BFV replication. This effect was more prominent in the presence of type I interferon signaling, suggesting nsP3 nuclear localization might be associated with IFN antagonism. Furthermore, abolishing nsP3 nuclear localization reduced virus replication in mice but did not significantly affect disease. IMPORTANCE Barmah Forest virus (BFV) is Australia’s second most prevalent arbovirus, with approximately 1,000 cases reported annually. The clinical symptoms of BFV infection include rash, polyarthritis, arthralgia, and myalgia. As BFV is not closely related to other pathogenic alphaviruses or well-studied model viruses, our understanding of its molecular virology and mechanisms of pathogenesis is limited. There is also a lack of molecular tools essential for corresponding studies. Here we describe the construction of an infectious clone of BFV, variants harboring point mutations, and sequences encoding marker protein. In infected mammalian cells, nsP3 of BFV was located in the nuclei. This finding extends our understanding of the diverse mechanisms used by alphavirus replicase proteins to interact with host cells. Our novel observations highlight the complex synergy through which the viral replication machinery evolves to correct mutation errors within the viral genome.
Article
Rabies virus (RABV) is a neurotropic virus that causes fatal neuroinflammation in mammals. The insectivorous bat RABV strains are less pathogenic for mice than strains associated with other reservoirs. We characterized the tissue inflammatory response in the CNS of RABV isolated from insectivorous bats. Eptesicus furinalis (EPBRV)-infected mice had a robust inflammatory response and a greater amount of IL-1β, IL-6 and TNF-α, while Myotis nigricans (MNBRV)-infected mice showed a higher expression of IL-17 and greater activation of IFN-β. New approaches to understand the inflammatory response to different mechanisms of action may provide insights for the development of novel therapies for rabies.
Article
Full-text available
Rabies virus phosphoprotein (P protein) is a multifunctional protein that plays key roles in replication as the polymerase cofactor that binds to the complex of viral genomic RNA and the nucleoprotein (N protein), and in evading the innate immune response by binding to STAT transcription factors. These interactions are mediated by the C-terminal domain of P (PCTD). The colocation of these binding sites in the small globular PCTD raises the question of how these interactions underlying replication and immune evasion, central to viral infection, are coordinated and, potentially, coregulated. While direct data on the binding interface of the PCTD for STAT1 is available, the lack of direct structural data on the sites that bind N protein limits our understanding of this interaction hub. The PCTD was proposed to bind via two sites to a flexible loop of N protein (Npep) that is not visible in crystal structures, but no direct analysis of this interaction has been reported. Here we use Nuclear Magnetic Resonance, and molecular modelling to show N protein residues, Leu381, Asp383, Asp384 and phosphor-Ser389, are likely to bind to a ‘positive patch’ of the PCTD formed by Lys211, Lys214 and Arg260. Furthermore, in contrast to previous predictions we identify a single site of interaction on the PCTD by this Npep. Intriguingly, this site is proximal to the defined STAT1 binding site that includes Ile201 to Phe209. However, cell-based assays indicate that STAT1 and N protein do not compete for P protein. Thus, it appears that interactions critical to replication and immune evasion can occur simultaneously with the same molecules of P protein so that the binding of P protein to activated STAT1 can potentially occur without interrupting interactions involved in replication. These data suggest that replication complexes might be directly involved in STAT1 antagonism.
Article
Full-text available
Many viruses target signal transducers and activators of transcription (STAT) 1 and 2 to antagonise antiviral interferon signalling, but targeting of signalling by other STATs/cytokines, including STAT3/interleukin 6 that regulate processes important to Ebola virus (EBOV) haemorrhagic fever, is poorly defined. We report that EBOV potently inhibits STAT3 responses to interleukin-6 family cytokines, and that this is mediated by the interferon-antagonist VP24. Mechanistic analysis indicates that VP24 effects a unique strategy combining distinct karyopherin-dependent and karyopherin-independent mechanisms to antagonise STAT3-STAT1 heterodimers and STAT3 homodimers, respectively. This appears to reflect distinct mechanisms of nuclear trafficking of the STAT3 complexes, revealed for the first time by our analysis of VP24 function. These findings are consistent with major roles for global inhibition of STAT3 signalling in EBOV infection, and provide new insights into the molecular mechanisms of STAT3 nuclear trafficking, significant to pathogen-host interactions, cell physiology and pathologies such as cancer.
Article
Full-text available
Significance To successfully establish infection, viral pathogens have to overcome the interferon (IFN)-mediated antiviral response. Previous studies revealed that the viral accessory protein Orf6 of SARS-CoV and SARS-CoV-2 is able to inhibit STAT1 nuclear translocation to block IFN signaling. In this study, we report that Orf6 localizes at the nuclear pore complex (NPC) where it binds directly to the Nup98-Rae1 complex to target the nuclear import pathway and mediate this inhibition. A better understanding of the strategies used by viruses to subvert host immune responses is critical for the design of novel antivirals and vaccines.
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication and host immune response determine coronavirus disease 2019 (COVID-19), but studies evaluating viral evasion of immune response are lacking. Here, we use unbiased screening to identify SARS-CoV-2 proteins that antagonize type I interferon (IFN-I) response. We found three proteins that antagonize IFN-I production via distinct mechanisms: nonstructural protein 6 (nsp6) binds TANK binding kinase 1 (TBK1) to suppress interferon regulatory factor 3 (IRF3) phosphorylation, nsp13 binds and blocks TBK1 phosphorylation, and open reading frame 6 (ORF6) binds importin Karyopherin α 2 (KPNA2) to inhibit IRF3 nuclear translocation. We identify two sets of viral proteins that antagonize IFN-I signaling through blocking signal transducer and activator of transcription 1 (STAT1)/STAT2 phosphorylation or nuclear translocation. Remarkably, SARS-CoV-2 nsp1 and nsp6 suppress IFN-I signaling more efficiently than SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Thus, when treated with IFN-I, a SARS-CoV-2 replicon replicates to a higher level than chimeric replicons containing nsp1 or nsp6 from SARS-CoV or MERS-CoV. Altogether, the study provides insights on SARS-CoV-2 evasion of IFN-I response and its potential impact on viral transmission and pathogenesis.
Article
Full-text available
Many viruses target signal transducer and activator of transcription (STAT) 1 to antagonise antiviral interferon signalling, but targeting of STAT3, a pleiotropic molecule that mediates signalling by diverse cytokines, is poorly understood. Here, using lyssavirus infection, quantitative live cell imaging, innate immune signalling and protein interaction assays, and complementation/depletion of STAT expression, we show that STAT3 antagonism is conserved among P-proteins of diverse pathogenic lyssaviruses and correlates with pathogenesis. Importantly, P-protein targeting of STAT3 involves a highly selective mechanism whereby P-protein antagonises cytokine-activated STAT3-STAT1 heterodimers, but not STAT3 homodimers. RT-qPCR and reporter gene assays indicate that this results in specific modulation of interleukin-6-dependent pathways, effecting differential antagonism of target genes. These data provide novel insights into mechanisms by which viruses can modulate cellular function to support infection through discriminatory targeting of immune signalling complexes. The findings also highlight the potential application of selective interferon-antagonists as tools to delineate signalling by particular STAT complexes, significant not only to pathogen-host interactions but also cell physiology, development and cancer.
Article
Full-text available
Viruses commonly antagonise the antiviral type I interferon response by targeting signal transducers and activators of transcription (STAT) 1 and 2, key mediators of interferon signalling. Other STAT family members mediate signalling by diverse cytokines important to infection, but their relationship with viruses is more complex. Importantly, virus-STAT interaction can be antagonistic or stimulatory depending on diverse viral and cellular factors. While STAT antagonism can suppress immune pathways, many viruses promote activation of specific STATs to support viral gene expression and/or produce cellular conditions conducive to infection. It is also becoming increasingly clear that viruses can hijack non-canonical STAT functions to benefit infection. For a number of viruses, STAT function is dynamically modulated through infection as requirements for replication change. Given the critical role for STATs in infection by diverse viruses, the virus:STAT interface is an attractive target for the development of antivirals and live-attenuated viral vaccines. Here, we review current understanding of the complex and dynamic virus:STAT interface, and discuss how this relationship might be harnessed for medical applications.
Article
Full-text available
To evade immunity, many viruses express interferon antagonists that target STAT transcription factors as a major component of pathogenesis. Because of a lack of direct structural data, these interfaces are poorly understood. We report the structural analysis of full-length STAT1 binding to an interferon antagonist of a human pathogenic virus. The interface revealed by transferred cross-saturation NMR is complex, involving multiple regions in both the viral and cellular proteins. Molecular mapping analysis, combined with biophysical characterization and in vitro/in vivo functional assays, indicates that the interface is significant in disease caused by a pathogenic field-strain lyssavirus, with critical roles for contacts between the STAT1 coiled-coil/DNA-binding domains and specific regions within the viral protein. These data elucidate the potentially complex nature of IFN antagonist/STAT interactions, and the spatial relationship of protein interfaces that mediate immune evasion and replication, providing insight into how viruses can regulate these essential functions via single multifunctional proteins.
Article
Alpha and gamma interferons rapidly induce several early response genes in primary human diploid fibroblasts. The transcription rates of these genes are maximal after 1 h of interferon treatment and return to basal levels within 8 h. Three different interferon-activated DNA-binding complexes (ISGF3, GAF, and FcRF gamma) that are responsible for transcriptional activation of cellular genes have been characterized. Assembly of these complexes requires tyrosine phosphorylation of one or more of the protein components. In this report, we demonstrate that a nuclear tyrosine phosphatase is responsible for the deactivation of these interferon-regulated transcription factors and the subsequent transcriptional downregulation of the corresponding genes. Furthermore, tyrosine phosphorylation is required for nuclear localization of the 91-kDa protein that is part of all three interferon-induced transcription complexes. These results provide the first evidence for a nuclear tyrosine phosphatase activity as a mechanism of transcriptional regulation.
Chapter
Rabies virus is usually transmitted by an animal bite and the virus remains close to the site of the bite for a period of weeks during the incubation period. Subsequently, the virus spreads in peripheral or cranial nerves by retrograde fast axonal transport, and then spreads extensively in the central nervous system (CNS) by the same mechanism along neuroanatomical connections. The infection produces mild inflammatory changes in the CNS, but degenerative neuronal changes are not prominent. Rabies virus later spreads centrifugally to multiple organs along sensory and autonomic nerve pathways to salivary glands (in rabies vectors), skin (basis of skin biopsy as a diagnostic test), heart, gastrointestinal tract, adrenals, and other organs. Degeneration of neuronal processes has recently been recognized, which is related to oxidative stress. The blood-brain barrier plays a role in excluding immune effector cells from entering the CNS in street rabies virus infection. There is usually a fatal outcome in rabies in humans and animals.
Article
Type I interferon (IFN-I) is critical for antiviral defense, and plasmacytoid dendritic cells (pDCs) are a predominant source of IFN-I during virus infection. pDC-mediated antiviral responses are stimulated upon physical contact with infected cells, during which immunostimulatory viral RNA is transferred to pDCs, leading to IFN production via the nucleic acid sensor TLR7. Using dengue, hepatitis C, and Zika viruses, we demonstrate that the contact site of pDCs with infected cells is a specialized platform we term the interferogenic synapse, which enables viral RNA transfer and antiviral responses. This synapse is formed via α L β 2 integrin-ICAM-1 adhesion complexes and the recruitment of the actin network and endocytic machinery. TLR7 signaling in pDCs promotes interferogenic synapse establishment and provides feed-forward regulation, sustaining pDC contacts with infected cells. This interferogenic synapse may allow pDCs to scan infected cells and locally secrete IFN-I, thereby confining a potentially deleterious response. Plasmacytoid dendritic cells (pDCs) sense viruses by physically contacting infected cells. Assil et al. show that the contact site between pDCs and infected cells is a specialized platform, termed an interferogenic synapse, with polarized cellular components and machinery that efficiently transfer immunostimulatory RNA to induce antiviral responses in pDCs.