Encephalomyocarditis Virus Disrupts Stress Granules, the ...

12
Encephalomyocarditis Virus Disrupts Stress Granules, the Critical Platform for Triggering Antiviral Innate Immune Responses Chen Seng Ng, a,b Michihiko Jogi, a,b,c Ji-Seung Yoo, a,b Koji Onomoto, a,b,c Satoshi Koike, d Takuya Iwasaki, d Mitsutoshi Yoneyama, a,b,c Hiroki Kato, a,b Takashi Fujita a,b Laboratory of Molecular Genetics, Institute for Virus Research, Kyoto University, Kyoto, Japan a ; Laboratory of Molecular Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan b ; Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan c ; Neurovirology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan d In response to stress, cells induce ribonucleoprotein aggregates, termed stress granules (SGs). SGs are transient loci containing translation-stalled mRNA, which is eventually degraded or recycled for translation. Infection of some viruses, including influ- enza A virus with a deletion of nonstructural protein 1 (IAVNS1), induces SG-like protein aggregates. Previously, we showed that IAVNS1-induced SGs are required for efficient induction of type I interferon (IFN). Here, we investigated SG formation by different viruses using green fluorescent protein (GFP)-tagged Ras-Gap SH3 domain binding protein 1 (GFP-G3BP1) as an SG probe. HeLa cells stably expressing GFP-G3BP1 were infected with different viruses, and GFP fluorescence was monitored live with time-lapse microscopy. SG formations by different viruses was classified into 4 different patterns: no SG formation, stable SG formation, transient SG formation, and alternate SG formation. We focused on encephalomyocarditis virus (EMCV) infec- tion, which exhibited transient SG formation. We found that EMCV disrupts SGs by cleavage of G3BP1 at late stages of infection (>8 h) through a mechanism similar to that used by poliovirus. Expression of a G3BP1 mutant that is resistant to the cleavage conferred persistent formation of SGs as well as an enhanced induction of IFN and other cytokines at late stages of infection. Additionally, knockdown of endogenous G3BP1 blocked SG formation with an attenuated induction of IFN and potentiated vi- ral replication. Taken together, our findings suggest a critical role of SGs as an antiviral platform and shed light on one of the mechanisms by which a virus interferes with host stress and subsequent antiviral responses. I n eukaryotic cells, viral infections induce several responses. Cel- lular pathogen recognition receptors such as RIG-I-like recep- tors (RLRs) and Toll-like receptors recognize specific pathogen- associated molecular patterns and activate the transcription of hundreds of genes, including interferons (IFNs), inflammatory cytokines, and antiviral proteins. Secreted IFNs, in turn, activate a secondary JAK-STAT signaling cascade, which culminates in the activation of various interferon-stimulated genes (ISGs) (1, 2). A representative ISG, protein kinase RNA activated (PKR), acts as an antiviral protein by inducing the blockade of viral translation (3– 5). PKR is also known to be associated with the cellular stress responses. Virus infection results in the accumulation of double- stranded RNA (dsRNA), thereby activating PKR and phosphory- lation of the subunit of eukaryotic initiation factor 2 (eIF2), leading to the formation of stress granules (SGs) (6, 7). Several studies have reported the interaction between viruses and SGs, especially the effects of specific types of viruses on the fate of SG formation and how viruses modulate stress granule assembly (8– 11). Recently, we reported that RLR recruitment to SGs during SG formation is critical for RLR-mediated signaling and that non- structural protein 1 of influenza A virus (IAV) blocks RLR signal- ing by inhibiting SGs and the antiviral response (12). Accumulat- ing evidence suggests that viruses have evolved strategies to prevent SG formation. These results suggest that virus-induced SGs potentially serve as platforms for antiviral activity; however, the underlying molecular mechanism still remains to be eluci- dated. In the present study, we aim to delineate the physiological im- pact of stress granule formation and its viral modulation. We em- ployed an enhanced green fluorescent protein (EGFP)-tagged stress granule marker, Ras-Gap SH3 domain binding protein 1 (G3BP1), to probe the subcellular distribution of virus-induced SGs (13, 14). This system allows us to monitor SGs in an individ- ual virus-infected cell. Infection with RNA and DNA viruses dis- played three distinct patterns: stable, transient, and alternate for- mation of SGs. We focused on encephalomyocarditis virus (EMCV), which exhibited transient formation of SGs. We show that EMCV disrupts SGs through G3BP1 cleavage. Furthermore, we found that EMCV-induced SGs are required for efficient acti- vation of IFN and cytokine genes. We propose a new antiviral concept highlighting the potential cross talk of virus-induced stress responses and activation of the IFN signaling cascade. This may provide new insight into understanding the mechanism by which antiviral genes are regulated. MATERIALS AND METHODS Plasmid constructs. The stress granule marker constructs pEGFP-C1- G3BP1 (NCBI RefSeq accession no. NM_005754) was a kind gift from Jamal Tazi (Institute de Génétique Moléculaire de Montpellier, France). The pEGFP-C1-G3BP1 Q325E mutant construct was generated by site- directed mutagenesis with a KOD-Plus mutagenesis kit (Toyobo, Japan) using primers containing the desired mutation according to manufactur- er’s instructions and were completely sequenced by using an ABI Prism Received 22 November 2012 Accepted 6 June 2013 Published ahead of print 19 June 2013 Address correspondence to Takashi Fujita, [email protected]. Supplemental material for this article may be found at http://dx.doi.org/10.1128 /JVI.03248-12. Copyright © 2013, American Society for Microbiology. All Rights Reserved. doi:10.1128/JVI.03248-12 September 2013 Volume 87 Number 17 Journal of Virology p. 9511–9522 jvi.asm.org 9511 on April 9, 2016 by PENN STATE UNIV http://jvi.asm.org/ Downloaded from

Transcript of Encephalomyocarditis Virus Disrupts Stress Granules, the ...

Encephalomyocarditis Virus Disrupts Stress Granules, the CriticalPlatform for Triggering Antiviral Innate Immune Responses

Chen Seng Ng,a,b Michihiko Jogi,a,b,c Ji-Seung Yoo,a,b Koji Onomoto,a,b,c Satoshi Koike,d Takuya Iwasaki,d Mitsutoshi Yoneyama,a,b,c

Hiroki Kato,a,b Takashi Fujitaa,b

Laboratory of Molecular Genetics, Institute for Virus Research, Kyoto University, Kyoto, Japana; Laboratory of Molecular Cell Biology, Graduate School of Biostudies, KyotoUniversity, Kyoto, Japanb; Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japanc; Neurovirology Project, TokyoMetropolitan Institute of Medical Science, Tokyo, Japand

In response to stress, cells induce ribonucleoprotein aggregates, termed stress granules (SGs). SGs are transient loci containingtranslation-stalled mRNA, which is eventually degraded or recycled for translation. Infection of some viruses, including influ-enza A virus with a deletion of nonstructural protein 1 (IAV�NS1), induces SG-like protein aggregates. Previously, we showedthat IAV�NS1-induced SGs are required for efficient induction of type I interferon (IFN). Here, we investigated SG formation bydifferent viruses using green fluorescent protein (GFP)-tagged Ras-Gap SH3 domain binding protein 1 (GFP-G3BP1) as an SGprobe. HeLa cells stably expressing GFP-G3BP1 were infected with different viruses, and GFP fluorescence was monitored livewith time-lapse microscopy. SG formations by different viruses was classified into 4 different patterns: no SG formation, stableSG formation, transient SG formation, and alternate SG formation. We focused on encephalomyocarditis virus (EMCV) infec-tion, which exhibited transient SG formation. We found that EMCV disrupts SGs by cleavage of G3BP1 at late stages of infection(>8 h) through a mechanism similar to that used by poliovirus. Expression of a G3BP1 mutant that is resistant to the cleavageconferred persistent formation of SGs as well as an enhanced induction of IFN and other cytokines at late stages of infection.Additionally, knockdown of endogenous G3BP1 blocked SG formation with an attenuated induction of IFN and potentiated vi-ral replication. Taken together, our findings suggest a critical role of SGs as an antiviral platform and shed light on one of themechanisms by which a virus interferes with host stress and subsequent antiviral responses.

In eukaryotic cells, viral infections induce several responses. Cel-lular pathogen recognition receptors such as RIG-I-like recep-

tors (RLRs) and Toll-like receptors recognize specific pathogen-associated molecular patterns and activate the transcription ofhundreds of genes, including interferons (IFNs), inflammatorycytokines, and antiviral proteins. Secreted IFNs, in turn, activate asecondary JAK-STAT signaling cascade, which culminates in theactivation of various interferon-stimulated genes (ISGs) (1, 2). Arepresentative ISG, protein kinase RNA activated (PKR), acts as anantiviral protein by inducing the blockade of viral translation (3–5). PKR is also known to be associated with the cellular stressresponses. Virus infection results in the accumulation of double-stranded RNA (dsRNA), thereby activating PKR and phosphory-lation of the � subunit of eukaryotic initiation factor 2 (eIF2�),leading to the formation of stress granules (SGs) (6, 7). Severalstudies have reported the interaction between viruses and SGs,especially the effects of specific types of viruses on the fate of SGformation and how viruses modulate stress granule assembly (8–11). Recently, we reported that RLR recruitment to SGs during SGformation is critical for RLR-mediated signaling and that non-structural protein 1 of influenza A virus (IAV) blocks RLR signal-ing by inhibiting SGs and the antiviral response (12). Accumulat-ing evidence suggests that viruses have evolved strategies toprevent SG formation. These results suggest that virus-inducedSGs potentially serve as platforms for antiviral activity; however,the underlying molecular mechanism still remains to be eluci-dated.

In the present study, we aim to delineate the physiological im-pact of stress granule formation and its viral modulation. We em-ployed an enhanced green fluorescent protein (EGFP)-taggedstress granule marker, Ras-Gap SH3 domain binding protein 1

(G3BP1), to probe the subcellular distribution of virus-inducedSGs (13, 14). This system allows us to monitor SGs in an individ-ual virus-infected cell. Infection with RNA and DNA viruses dis-played three distinct patterns: stable, transient, and alternate for-mation of SGs. We focused on encephalomyocarditis virus(EMCV), which exhibited transient formation of SGs. We showthat EMCV disrupts SGs through G3BP1 cleavage. Furthermore,we found that EMCV-induced SGs are required for efficient acti-vation of IFN and cytokine genes. We propose a new antiviralconcept highlighting the potential cross talk of virus-inducedstress responses and activation of the IFN signaling cascade. Thismay provide new insight into understanding the mechanism bywhich antiviral genes are regulated.

MATERIALS AND METHODSPlasmid constructs. The stress granule marker constructs pEGFP-C1-G3BP1 (NCBI RefSeq accession no. NM_005754) was a kind gift fromJamal Tazi (Institute de Génétique Moléculaire de Montpellier, France).The pEGFP-C1-G3BP1 Q325E mutant construct was generated by site-directed mutagenesis with a KOD-Plus mutagenesis kit (Toyobo, Japan)using primers containing the desired mutation according to manufactur-er’s instructions and were completely sequenced by using an ABI Prism

Received 22 November 2012 Accepted 6 June 2013

Published ahead of print 19 June 2013

Address correspondence to Takashi Fujita, [email protected].

Supplemental material for this article may be found at http://dx.doi.org/10.1128/JVI.03248-12.

Copyright © 2013, American Society for Microbiology. All Rights Reserved.

doi:10.1128/JVI.03248-12

September 2013 Volume 87 Number 17 Journal of Virology p. 9511–9522 jvi.asm.org 9511

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

DNA sequencer to verify the presence of the mutation. This plasmid con-tained a single-point amino acid substitution at position 325 (from glu-tamine to glutamate), which is resistant to cleavage by 3CPRO of poliovirus(PolioV) (15). Expression vectors for EMCV pFirefly-leader and pFirefly-3C proteases were described previously (16).

Viruses. PolioV (Mahoney strain), vesicular stomatitis virus (VSV)(Indiana strain), EMCV, adenoviruses (type 5), Sindbis virus (SINV), andTheiler’s murine encephalomyelitis virus (TMEV) (GDVII strain) wereprepared by infecting BHK cells at a multiplicity of infection (MOI) of 1.Cell culture medium was collected after confirming cytopathic effectsfollowing infection. Medium containing newly produced viruses was cen-trifuged at 1,500 rpm for 5 min to pellet the cell debris, and supernatantscontaining viruses were collected and stored at �80°C. The viral titer wasassessed by a plaque assay on L929 cells, as previously described (17).Newcastle disease virus (NDV) (Miyadera strain), Sendai virus (SeV)(Cantell), and influenza A virus with a deletion of the NS1 gene(IAV�NS1) (strain A/Puerto Rico 8/34) (18, 19) were propagated in theallantoic cavities of embryonated chicken eggs, and stocks were thenstored at �80°C.

Generation of stable HeLa cells and general cell culture conditions.Cell lines were maintained in Dulbecco’s modified Eagle’s medium(DMEM) supplemented with 10% heat-inactivated fetal bovine serum(Nacalai Tesque, Japan) and penicillin-streptomycin (100 U/ml and 100�g/ml, respectively; Nacalai Tesque, Japan). To generate HeLa cells stablyexpressing the EGFP-G3BP1 wild type (wt) and the EGFP-G3BP1 Q325Emutant, pEGFP-C1-G3BP1 and pEGFP-C1-G3BP1 Q325E mutant ex-pression constructs were linearized by using the restriction enzyme ApaLI

(TaKaRa, Japan). The linearized plasmids were then transfected intoHeLa cells by using FuGENE6 (Promega, USA) according to manufactur-er’s recommendations. Transformants were selected by including 1mg/ml of G418 in the culture medium. Individual colonies were isolatedand characterized.

Live-cell imaging and immunofluorescence microscopy. For thelive-cell imaging analysis, HeLa cells stably expressing EGFP-G3BP1(HeLa/G-G3BP cells) were seeded into a 12-well plate and incubated at37°C. After 24 h, cells were washed with DMEM (10% fetal bovine serumand 1% penicillin-streptomycin) for several rounds. Cells were then in-fected with various types of RNA and DNA viruses. After 1 h of infection,virus was removed and replaced with 1.0 ml of DMEM imaging medium(4,500 mg/liter D-glucose and L-glutamine, 25 mM HEPES buffer, nosodium pyruvate, and phenol red; Invitrogen). Imaging was immediatelyinitiated every 10 min. Live cells were maintained on the microscope stageat 37°C with 5% carbon dioxide in a humidity-controlled chamber. Im-ages were taken by using Biophotonics-ImageJ software. All imaging wasperformed by using a Leica CTR 6500 instrument.

For the immunofluorescence analysis, cells were seeded into either a12-well plate or an 8-well chamber slide and incubated at 37°C. After 24 h,cells were subjected to various treatments, such as plasmid transfection orvirus infection. Cells were then rinsed in phosphate-buffered saline (PBS)several times, fixed with 4% paraformaldehyde solution for 10 min atroom temperature, washed with PBS for two additional rounds, perme-abilized with acetone-methanol (1:1) for 1 min, and blocked with phos-phate-buffered saline containing 0.1% Tween 20 (PBST) and containingbovine serum albumin (BSA) (5.0 mg/ml) for 1 h at 4°C. Cells were then

FIG 1 Characterization of HeLa/G-G3BP cells. (A) HeLa/G-G3BP1 clone 12 was mock treated or stimulated, as indicated. DAPI, 4=,6-diamidino-2-phenylin-dole. (B and C) Cells were fixed and examined for GFP fluorescence. Four independent HeLa/G-G3BP cell clones were stimulated by arsenite (B) or by infectionwith NDV (C), and the percentage of GFP speckle-positive cells was determined. (D) Parental HeLa cells and HeLa/G-G3BP1 clones were infected with NDV for12 h, and the IFN-� gene expression level was determined by RT-qPCR. (Error bars indicate standard deviations of duplicates [n � 2].)

Ng et al.

9512 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

incubated with primary antibody followed by fluorophore-conjugatedsecondary antibodies (Invitrogen) for 1 h at 4°C. Cells were washed withPBST extensively and mounted. All images were obtained by using a LeicaCTR 6500 instrument.

siRNA-directed gene silencing. The small interfering RNA (siRNA)universal negative control and siRNAs targeting the stress granule markerG3BP1 (50 nM) and the dsRNA protein kinase PKR were purchased fromInvitrogen and transfected by using either Lipofectamine 2000 (Invitro-gen) or RNAiMax (Invitrogen) according to the manufacturer’s recom-mendations. The sequences of siRNAs are as follows: sense sequence5=-CGG AUU AGC GAC AAA UUU AUU-3= and antisense sequence 5=-UAA AUU UGU CGC UAA UCC GUU-3= for RIG-I, sense sequence 5=-UUU ACU UCA CGC UCC GCC UUC UCG U-3= and antisense sequence5=-ACG AGA AGG CGG AGCGUGAAGUAA A-3= for PKR#1, sense se-quence 5=-AUG UCA GGA AGG UCA AAU CUG GGU G-3= and anti-sense sequence 5=-CAC CCA GAU UUG ACC UUC CUG ACA U-3= forPKR#2 (#n, designated siRNA number), and sense sequence 5=-UAAUUU CCC ACC ACU GUU AAU GCG C-3= and antisense sequence 5=-GCGCAUUAACAGUGGUGGGAAAUUA-3= for G3BP1. At 48 h post-transfection, cells were subjected to viral infection or other treatments. Aspecific antibody for G3BP1 (Santa Cruz) was used to monitor the knock-down efficiency.

RNA analysis. RNA was harvested from cells by using TRIzol (Invit-rogen) according to the manufacturer’s instructions. ContaminatingDNA was then eliminated by using recombinant DNase I (10 units/�l;Roche) according to the manufacturer’s protocol. Treated samples werepurified by phenol-chloroform extraction. Five hundred nanograms ofpurified RNA was used as a template to synthesize cDNA by using a HighCapacity cDNA reverse transcription kit (Applied Biosystems), as speci-fied by the manufacturer, with the following cycles: 25°C for 10 s, 37°C for2 h, and 85°C for 10 s. The concentration of cDNA was quantified by theuse of a spectrophotometer, and the final concentration was adjusted to 1�g/�l. cDNA samples were then subjected to either standard PCR orreal-time quantitative PCR (RT-qPCR) analysis with specific probes fromthe TaqMan gene expression assay (Applied Biosystems). Quantificationof EMCV RNA was performed by using SYBR master mix (Applied Bio-systems) with specific primers targeting the EMCV capsid coding region.Standard PCR was performed with cDNA samples together with a mastermix containing 1� PCR buffer, 2.5 mM each deoxynucleoside triphos-phates (dNTPs), 0.2 units of Ex Taq polymerase, and 1.0 �M both forwardand reverse primers. PCR buffer, dNTPs, and Ex Taq polymerase werepurchased from TaKaRa, Japan. Primers were all customized and pur-chased from Invitrogen. PCR was performed with a 50-�l reaction mix-

FIG 2 Three major forms of virus-induced stress granule distribution patterns in HeLa/G-G3BP cells infected with different viruses. (A to K) HeLa/G-G3BP cellswere infected with NDV (A), IAV (B), IAV�NS1 (C), EMCV (D), SINV (E), PolioV (F), SeV (G), VSV (H), adenovirus type 5 with an E1A deletion(Adeno5�E1A) (I), wild-type adenovirus type 5 (Adeno5WT) (J), and TMEV (K) for approximately 9 to 12 h, and SG formation was monitored and quantifiedas described in Materials and Methods. (Error bars indicate standard deviations of triplicates [n � 3].) N.D., not detectable; ��, P � 0.005; �, P � 0.05. (L to N)Representative cell images taken at the indicated times after infection for stable (NDV) (L), transient (SINV) (M), and alternating (Adeno5�E1A) (N) SGformation. (O) Wild-type HeLa cells were mock infected or infected for 4 or 12 h and fixed to examine the localization of endogenous G3BP1 by immunostaining.

Loss of Stress Granules Impairs Interferon Signals

September 2013 Volume 87 Number 17 jvi.asm.org 9513

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

ture with an initial annealing temperature of 56°C to 60°C. PCR productswere analyzed by agarose gel electrophoresis.

Western blotting. Cells were collected in ice-cold PBS by using ascraper. Cells were collected by centrifugation and lysed by NP-40 buffer(50 mM Tris [pH 8.0], 150 mM NaCl, 1% [vol/vol] NP-40, 1 nM vana-date, 1 mM leupeptin, and phenylmethanesulfonyl fluoride), followed bycentrifugation at 15,000 rpm for 10 min and ultracentrifugation at100,000 rpm for 5 min. The supernatant was mixed with an equal volumeof 2� SDS buffer, boiled for 5 min, separated by SDS-PAGE (30 �g/lane),

and transferred onto a nitrocellulose membrane. The membranes wereincubated in blocking buffer (PBS, 5% [wt/vol] dry milk powder) for 30min at room temperature, followed by incubation with primary antibodydiluted in blocking buffer at 4°C overnight. Membranes were washedextensively with TBST (Tris-buffered saline, 0.1% Tween 20), followed byincubation with a conjugated secondary antibody for 1 h at room temper-ature. The proteins were visualized by using alkaline phosphatase buffercontaining 5’-bromo-4-chloro-3’-indolylphosphate (BCIP)-Nitro BlueTetrazolium (NBT) (Promega) color development substrate (100 mM

FIG 3 EMCV infection results in the cleavage of G3BP1. (A) Immunoblotting (IB) showing the kinetics of G3BP1 cleavage in EMCV-infected HeLa/G-G3BP1cells. N.C., negative control. (B) HeLa cells stably expressing FLAG-G3BP1 Q325E protein were infected with EMCV, and the G3BP1 Q325E protein level wasmonitored by immunoblotting. (C) Western blot analysis of HeLa/G-G3BP1 cells infected with EMCV. Lysates were prepared at the indicated time points afterinfection and subjected to immunoblotting with the indicated antibodies. FL, full-length; n.s., not significant. (D, left) HeLa cells were transiently transfected withan empty vector or the expression vector for leader or 3C and analyzed for endogenous G3BP1 by Western blotting. (Right) HeLa/G-G3BP1 and HeLa/G-G3BP1Q325E cells were transiently transfected with an empty vector or the expression vector for leader or 3C and analyzed by Western blotting using anti-GFP.MW, molecular weight (in thousands); p.h.i., hour postinfection; cp, cleavage fragment.

FIG 4 HeLa/G-G3BPQ325E cells display stable formation of SGs induced by EMCV infection. (A and B) Both HeLa/G-G3BP1 (A) and HeLa/G-G3BP1Q325E(B) cells were infected with EMCV. GFP fluorescence images of these cells taken every 40 min are shown. (C) Quantitative analysis of SG formation pattern ofHeLa/G-G3BP1Q325E cells infected with EMCV. (Error bars indicate standard deviations of triplicates [n � 3].) N.D., not detectable; ��, P � 0.005.

Ng et al.

9514 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

Tris-HCl [pH 9.0], 150 mM NaCl, 1 mM MgCl2, 66 �l of NBT [50 mg/ml], and 33 �l of BCIP [50 mg/ml]).

Antibodies. The antibodies used in this study include mouse mono-clonal anti-green fluorescent protein (GFP) (1:1,000 dilution) (MBL),goat polyclonal anti-G3BP1 (1:500) (catalog no. sc-70283; Santa Cruz),mouse monoclonal anti-G3BP1 (1:1,000) (catalog no. sc-365338; SantaCruz), rabbit polyclonal anti-PKR (1:1,000) (catalog no. sc-709; Santa Cruz),rabbit polyclonal anti-TIA-1/R (1:1,000) (catalog no. sc-48371; Santa Cruz),goat polyclonal anti-TIAR (1:1,000) (catalog no. sc-1749; Santa Cruz), rabbitpolyclonal anti-HuR (1:1,000) (catalog no. sc-365816; Santa Cruz), andpropidium iodide (PI) (1:2,000 in PBST) (Miltenyi Biotec). The RIG-Iantibodies were generated by immunizing a rabbit with a synthetic pep-tide corresponding to amino acids 793 to 807 of RIG-I and MDA5. Mousemonoclonal anti-poly(A) binding protein (PABP) (1:1,000) (catalognumber ab6125; Abcam), rabbit monoclonal antiactin (1:5,000)(Poly6221; BioLegend), mouse anti-FLAG (1:1,000) (Sigma-Aldrich),and rabbit monoclonal anti-phospho-PKR pT446 (1:1,000) (EpitomicsInc.) antibodies were also used. Anti-EMCV polyclonal antibody was ob-tained by immunizing a rabbit with purified EMCV virions. Anti-MDA5polyclonal antibody was obtained by immunizing a rat with recombinantMDA5 (produced in insect cells), which was preactivated with RNA li-gands.

Quantification of the distribution pattern of virus-induced SGs. SGformation was quantified visually by using eyesight counting. The totalnumber of cells displaying each unique distribution pattern in each loca-tion was recorded, and the percentage of each pattern was calculated. Asfor the fixed cells, 10 pictures were taken randomly at different locations.Cells displaying SG foci were quantified manually. Graphs display theaverage percentages of replicates (at least 20 times).

RESULTSCharacterization of HeLa cells stably expressing an SG marker,G3BP1. To monitor SGs in living cells, we generated HeLa/G-G3BP cells (Fig. 1). Constitutive aggregation of intrinsic SG com-ponents has been reported to lead to a severe stall in protein syn-

thesis and eventual apoptosis (14, 20). All the HeLa/G-G3BPclones displayed uniform and high-level GFP expression, andtheir growth rate was comparable to that of parental cells (ourunpublished observations). It has been well documented thatG3BP1 accumulates in SG foci in response to arsenite treatment(oxidative stress) and virus infection (12, 13). HeLa/G-G3BPclone 12 was treated with arsenite or infected with Newcastle dis-ease virus (NDV) or influenza A virus (IAV) with an NS1 deletion(IAV�NS1), and GFP localization was then examined by confocalmicroscopy. As shown in Fig. 1A, a speckle-like localization ofGFP was induced by these stimuli. Other clones also exhibitedsimilar speckle formation after arsenite treatment or NDV infec-tion (Fig. 1B and C). We confirmed that other SG components,TIA-1, TIAR, HuR, and eIF3, colocalized with the GFP speckles(our unpublished observations). These results indicate thatEGFP-G3BP1 acts as a suitable probe for virus-induced SGs.However, since transient overexpression of G3BP1 results in SGformation without external stress (13), we tested if the HeLa/G-G3BP clones would exhibit a normal antiviral response. As shownin Fig. 1D, all clones exhibited induction of IFN-� mRNA com-parable to that exhibited by parental cells. We chose clone 18 forfurther analyses.

G3BP1 exhibits three redistribution patterns after infectionwith both RNA and DNA viruses. To examine the dynamics ofcytoplasmic SGs induced by viral infection, the cells were infectedwith different viruses, as shown in Fig. 2, and monitored live fordistribution of GFP fluorescence (representative results are shownin Movies S1 to S9 in the supplemental material). Cells infectedwith SeV, IAV, VSV, and TMEV did not show SG formation (8).Other viruses induced SGs, typically forming a large number ofsmall granules at around 5 h postinfection and gradually fusing toeach other. SG formation was quantified (Fig. 2A to K) and clas-

FIG 5 Cleavage or knockdown of G3BP1 results in enhanced EMCV replication. (A) HeLa/G-G3BP1 and HeLa/G-G3BP1Q325E cells were infected with EMCV.(Top) Total RNA was harvested at 12 h postinfection, and EMCV RNA was quantified by qPCR. (Bottom) The culture supernatant was subjected to plaquetitration. (B) HeLa cells were transfected with either control siRNA or siRNA that targeted G3BP1. (Top) After 48 h, G3BP1 was detected by Western blotting(left) or by staining using anti-G3BP1 antibody (right). (Bottom, left) To investigate the effect of G3BP1 knockdown on viral replication, the cells were infectedwith EMCV for 12 h, total RNA was extracted, and EMCV RNA was quantified by qPCR. (Right) The culture supernatant was analyzed to determine viral titers.

Loss of Stress Granules Impairs Interferon Signals

September 2013 Volume 87 Number 17 jvi.asm.org 9515

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

FIG 6 Inhibition of G3BP1 in EMCV-infected cells results in sustained cytokine/chemokine mRNA accumulation. HeLa/G-G3BP1 and HeLa/G-G3BP1Q325Ecells were infected with EMCV. (A) Culture supernatant was subjected to an enzyme-linked immunosorbent assay for human IFN-� (hIFN-�). (B to E) TotalRNA was harvested at the indicated time points. mRNA levels for IFN-� (B), CXCL10 (C), IL-6 (D), and RANTES (E) were determined by RT-qPCR. (F, left)Both HeLa/G-G3BP and HeLa/G-G3BPQ325E cells were infected with IAV�NS1, and the IFN-� mRNA level was quantified as described above. (Right) Lysatesof IAV�NS1-infected HeLa/G-G3BP1 cells were examined for cleavage of G3BP1 by Western blotting. Data depicted are representative of two independentexperiments. (Error bars indicate standard deviations of duplicates.) ��, P � 0.005; �, P � 0.05.

Ng et al.

9516 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

sified into three predominant patterns, stable formation (Fig. 2L),transient formation (Fig. 2M), and alternating formation (Fig.2N), within a single cell. NDV, IAV�NS1, and adenovirus type 5displayed stable formation of SGs (see Movies S1 to S3 in thesupplemental material). Whereas SINV, EMCV, and PolioV in-duced foci at around 5 to 6 h postinfection, the foci disappearedthereafter (transient formation) (Fig. 2D to F; see also Movies S4to S6 in the supplemental material). Interestingly, adenovirus type5 with an E1A deletion exhibited multiple rounds of formationand disappearance of SGs (alternate formation) in the majority ofcells (Fig. 2I; see also Movie S7 in the supplemental material). Asimilar oscillation of SGs in cells infected with hepatitis C virus(HCV) and treated with IFN was reported previously (21). Col-lectively, these live-cell-imaging analyses demonstrated that viralinfections trigger host stress responses; however, different virusesinduce distinct response patterns, presumably through specificunderlying mechanisms. The observed SG formation patterns areunlikely to be due to G3BP1 overexpression, because wt HeLa cellsexhibited transient SG formation upon EMCV infection whenendogenous G3BP1 was used as a marker (Fig. 2O).

EMCV infection results in the cleavage of G3BP1. We focusedon the mechanism of transient formation of SGs by EMCV be-cause PolioV has been reported to inhibit SG formation by cleav-age of G3BP1 (15). We examined if EGFP-G3BP1 is cleaved byEMCV by Western blotting. The EGFP-G3BP1 fusion protein isdetected as a polypeptide of 96 kDa, and EMCV infection resultedin the appearance of an 80-kDa GFP-containing protein at 6 hpostinfection, and nearly complete cleavage of EGFP-G3BP1 oc-curred at 10 h postinfection (Fig. 3A). Because the fusion proteincontains an EGFP moiety at the N terminus of G3BP, the cleavage

of G3BP1 is likely to occur at the C-terminal region of G3BP1. Weverified the cleavage site by using an antibody detecting the C-ter-minal epitope of G3BP1 (see Fig. S1A and S1B in the supplementalmaterial). Because the mapped cleavage site was close to that ofPolioV and cleavage by PolioV is prevented by an amino acidsubstitution within G3BP1 (Q325E) (15), we therefore examinedthis mutant for cleavage by EMCV (Fig. 3B). We found that theG3BP1 Q325E mutant was resistant to cleavage by EMCV, sug-gesting a common cleavage mechanism. To examine whether thedisruption of SGs by EMCV is due solely to cleavage of G3BP1, weexamined other SG components, such as PABP, TIA-1/R, HuR,and PKR, which are also essential for SG formation. Figure 3Cshows that the levels of SG components, with the exception ofG3BP1, did not change upon EMCV infection and that G3BP1cleavage coincided with the detection of EMCV proteins. Expres-sion of EMCV 3C protease but not leader protein by transfectionwas sufficient to reproduce G3BP1 cleavage at Q325 (Fig. 3D),strongly suggesting that the cleavage is mediated by 3C protease.We next examined SG formation of HeLa/G-G3BPQ325E cells. Insharp contrast to cells expressing wild-type G3BP1 (see Movie S6in the supplemental material), HeLa/G-G3BPQ325E cells exhib-ited stable formation of SGs, as judged by single-cell imaging (Fig.4A and B; see also Movie S8 in the supplemental material) and

FIG 7 IFN production and cytokine gene activation in HeLa/G-G3BP andHeLa/G-G3BPQ325E cells at the early phase. HeLa/G-G3BP1 and HeLa/G-G3BP1Q325E cells were mock treated or infected with EMCV for the indicatedtimes. Total RNA was extracted, and mRNA levels for IFN-� (A), CXCL10 (B),IL-6 (C), and RANTES (D) were quantified by RT-qPCR.

FIG 8 Knockdown of G3BP1 attenuates EMCV-induced cytokine/chemokinegene activation. HeLa cells were transfected with either control siRNA orsiRNA that targeted G3BP1. After 48 h of incubation, cells were infected withEMCV for 12 h, and total RNA was collected as indicated. mRNAs levels forIFN-� (A), RANTES (B), CXCL10 (C), and IL-6 (D) were determined byRT-qPCR. Data are representative of two independent experiments. (Errorbars indicate standard deviations of duplicates [n � 2].) �, P � 0.05.

Loss of Stress Granules Impairs Interferon Signals

September 2013 Volume 87 Number 17 jvi.asm.org 9517

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

quantification (Fig. 4C). These results suggest that EMCV disruptsSGs by cleavage of G3BP1 through a mechanism similar to that ofPolioV.

G3BP1 negatively regulates EMCV replication. To examinethe impact of SG disruption on EMCV replication, we infectedboth HeLa/G-G3BP and HeLa/G-G3BPQ325E cells with EMCVand analyzed viral replication by RT-qPCR (Fig. 5A). The EMCVRNA level recovered from HeLa/G-G3BP cells was 6-fold higherthan that recovered from HeLa/G-G3BPQ325E cells. Similarly, asignificantly lower viral yield was observed for cells expressingG3BP1 Q325E, suggesting that SG formation is critical for sup-pressing EMCV replication. To further confirm the involvementof G3BP1, we depleted endogenous G3BP1 by siRNA-mediatedknockdown (Fig. 5B) and examined its effect on EMCV replica-tion. G3BP1 knockdown caused increased EMCV replication, asjudged by the approximately 5-fold augmentation of viral RNAand viral yield (Fig. 5B). These results suggest that G3BP1 is in-volved in the negative regulation of EMCV.

G3BP1 is critical for EMCV-induced interferon and cytokinegene activation. Based on the above-described findings, we nextasked how G3BP1 exerts its antiviral role. The type I interferonsystem constitutes major innate antiviral responses; therefore, weexamined EMCV-induced IFN-� gene activation in HeLa/G-G3BP and HeLa/G-G3BPQ325E cells (Fig. 6). In HeLa/G-G3BPcells, IFN-� mRNA accumulated at 4 h postinfection, followed bya gradual decrease. However, IFN-� mRNA levels persisted in

HeLa/G-G3BPQ325E cells after 8 h postinfection (Fig. 6B). Inagreement with these results, the amount of IFN-� protein re-leased into the culture medium at 24 h was significantly aug-mented by the Q325E mutation (Fig. 6A). A similar enhancementof cytokine mRNA was observed for CXCL10, interleukin-6 (IL-6), and RANTES (Fig. 6C to E). We investigated gene activation atearly time points between 0 and 4 h and observed similar activa-tion kinetics between HeLa/G-G3BP and HeLa/G-G3BPQ325Ecells (Fig. 7), suggesting that the reduced gene activation of HeLa/G-G3BP cells is due to G3BP1 cleavage. The Q325E mutation didnot affect IFN-� gene induction in the case of IAV�NS1, whichdid not cause G3BP1 cleavage (Fig. 6F). Next, we examined theeffects of depletion of endogenous G3BP1 on cytokine gene acti-vation. As expected, knockdown of endogenous G3BP1 attenu-ated the expression of IFN-� and other cytokine genes (Fig. 8A toD). These results strongly suggest that G3BP cleavage leads toattenuation of antiviral cytokine induction.

It has been well documented that MDA5 senses EMCV in-fection (22–25) and that virus- and oxidative stress-inducedSGs recruit RIG-I, MDA5, and LGP2 (12). Therefore, we hy-pothesized that EMCV-induced SG regulates IFN-� gene acti-vation by facilitating MDA5 activation. We examined MDA5localization in EMCV-infected HeLa cells by immunostaining.MDA5 displayed relocalization to speckle-like granules uponEMCV infection (Fig. 9A). The speckles also contained endog-enous G3BP1 (Fig. 9A) and TIAR (Fig. 9B). Interestingly, PI, a

FIG 9 EMCV infection recruits MDA5 into SGs. HeLa cells were mock treated or infected with EMCV (MOI of 10) and fixed. The cells were stained for MDA5,G3BP1, and PI (A) or MDA5, TIAR, and PI (B).

Ng et al.

9518 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

dye that binds to dsDNA and dsRNA, stains cytoplasmic speck-les found only in virus-infected cells, and the dsRNA specklescolocalized with G3BP1 and TIAR. These observations suggestthat EMCV infection induces SGs, which recruit SG compo-nents, MDA5, and EMCV dsRNA.

PKR is essential for SG formation and IFN induction inEMCV infection. Various types of viruses were shown to induceSG formation through PKR activation (26–28). We thereforeexamined whether EMCV induces SG formation in a PKR-dependent manner. Endogenous PKR expression was effi-ciently downregulated by siRNA (Fig. 10A). Under these con-ditions, SG formation by EMCV was decreased significantly(Fig. 10A). We next asked whether cleavage of G3BP1 results inPKR dephosphorylation. Immunoblot analyses showed thatPKR was autophosphorylated at 4 h postinfection; however, at12 h, when G3BP1 cleavage was nearly complete, PKR phos-phorylation was undetectable (Fig. 10B, lane 3), suggesting thatG3BP1 cleavage resulted in PKR dephosphorylation. Finally,we examined whether the final outcome of signaling, IFN-�gene expression, was dependent on PKR. In PKR knockdowncells, the induction of IFN-� mRNA by EMCV was significantlydecreased compared to that in control cells (Fig. 10C). Wefurther confirmed previous reports that IFN induction bypoly(I·C) or IAV�NS1 infection was PKR dependent (12).From the data presented above, we concluded that the loss of

PKR impaired EMCV-induced SG formation, leading to a re-duction of IFN-� gene activation.

DISCUSSION

Viral infection causes stress in host cells, resulting in SG forma-tion. To date, both pro- and antiviral roles have been described forvirus-induced SGs (28–30), and this issue remains controversial.

In this study, we demonstrated that SGs are potentially involved inmediating virus-triggered IFN responses. It was reported previouslythat PolioV 3C protease cleaves G3BP1 at residue Q325, resulting inthe disruption of SGs (15). This observation indicates not only thatG3BP1 is a component of SGs but also that its inactivation by cleavagecauses the disruption of SGs. Here, we show that EMCV shares G3BPcleavage activity with specificity identical to that of PolioV 3C, requir-ing intact Q325. Interestingly, coxsackievirus also disrupts SGs (31)by a similar mechanism (G. Fung, C. S. Ng, J. Zhang, J. Shi, J. Wong,P. Piesik, L. Han, F. Chu, J. Jagdeo, E. Jan, T. Fujita, and H. Luo,unpublished observation), suggesting that this strategy is shared bysome picornaviruses to evade immune responses. At the early phaseof EMCV infection, cleavage of G3BP1 was not evident. However, at4 h postinfection, cleavage was detectable, and at 10 h, cleavagereached completion, suggesting that the accumulation of 3C is nec-essary for the disruption. We observed that stable expression of theG3BP1 Q325E mutant blocked the disassembly of SGs as well as en-hanced IFN-� production at a late phase of infection. Furthermore,

FIG 10 Involvement of PKR in EMCV-induced SG and IFN-� gene activation. (A) Knockdown of PKR expression results in reduced SGs. (Left) HeLa cellstransfected with siRNA targeting PKR for 48 h were examined for PKR expression by Western blotting. (Middle) The cells were infected with EMCV for 6 h andstained for endogenous G3BP1. (Right) SG-containing cells were quantified. (B) HeLa cells infected with EMCV for 0, 4, and 12 h were analyzed for G3BP1,phospho-PKR, EMCV proteins, and actin by immunoblotting. (C) HeLa cells transfected with siRNA targeting PKR for 48 h were mock treated, transfected withpoly(I·C), or infected with IAV�NS1 or EMCV. After 12 h, IFN mRNA was quantified by RT-qPCR. ��, P � 0.005; �, P � 0.05.

Loss of Stress Granules Impairs Interferon Signals

September 2013 Volume 87 Number 17 jvi.asm.org 9519

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

knockdown experiments showed that G3BP1 is necessary for efficientactivation of the IFN-� gene, particularly in the later stages of infec-tion. Although it was reported previously that PolioV 3C cleavesRIG-I and MDA5 (32) and that EMCV cleaves RIG-I (33), we did notobserve these cleavages, even under conditions in which G3BP1 wascleaved by EMCV or PolioV (Fig. 11). Taken together, we concludethat G3BP1 is a physiological regulator of IFN-� gene inductionthrough the formation of SGs, which recruit the RNA sensor MDA5.In addition, the persistent activation of the IFN-� gene at late timepoints is likely due to the increase of the local concentrations of bothMDA5 and its ligands within the condensed granules.

Collectively, the data presented above strongly suggest that 3Cprotease of EMCV acts as a critical factor for evading host IFN pro-duction to ensure efficient replication. It was demonstrated previ-ously that PKR plays a critical role in dsRNA- or IAV�NS1-inducedSG formation and subsequent IFN-� gene activation (12). Our ob-servation that PKR is required for efficient IFN gene activation byEMCV suggests that PKR is responsible for initiating SG formation(Fig. 10).

Considering that the assembly of SGs is a part of the antiviralresponse of the host, it is plausible that viruses evolve strategies toblock it. Indeed, IAV, SeV, and TMEV do not induce SG (Fig. 2),and it was reported previously that leader RNA, NS1, and leader

protein are responsible for inhibition, respectively (8, 12, 34). Al-though TMEV belongs to the Picornaviridae, its mechanism of SGinhibition appeared to be distinct from those of EMCV andPolioV. TMEV and mengovirus inhibit SG by the action of leaderprotein (8, 31). We found that 3C but not the leader protein ofEMCV inhibits SG formation (Fig. 12). It is tempting to speculatethat leader proteins of TMEV and mengovirus inhibit IFN pro-duction (35, 36) through the blockade of SG formation, whereRLR and viral RNA efficiently interact, as one of the mechanisms.Interestingly, although the leader protein of EMCV did not affectSGs, it inhibited IFN gene activation (Fig. 12), suggesting thatleaders of different cardioviruses are functionally equivalent (37,38) but with distinct modes of action. Therefore, these virusesencode multiple inhibitory proteins to efficiently manipulate hostimmune responses. EMCV and SINV induced SGs at early timepoints after infection, but SG formation was disrupted later. Asimilar phenomenon was reported previously for West Nile anddengue viruses by monitoring TIA-1/R as an SG marker (29). Inthe case of EMCV and PolioV, G3BP1 cleavage by viral 3C pro-tease is responsible for the disassembly of SGs. Therefore, activemechanisms for the disruption of SGs by SINV, West Nile virus,and dengue virus have been suggested, although the underlyingmechanisms remain to be determined. In addition to transient

FIG 11 RIG-I was not cleaved after EMCV or PolioV infection. (A) HeLa cells were either mock treated or infected with EMCV for the indicated times. RIG-Iwas detected by Western blotting. (B) HeLa cells were mock treated or infected with PolioV for 9 h. G3BP1 (left) and RIG-I (right) were examined by Westernblotting. CTD, C-terminal domain; shRIG-I, short-hairpin RIG-I.

Ng et al.

9520 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

formation of SGs, some viruses exhibited alternating formationsof SGs; SGs were formed at an early stage and then disappearedand re-formed at a later stage. This alternating pattern is alsodependent on the cell lines used (our unpublished observations),suggesting that the pattern of SG formation is determined by a

dynamic balance between the host antiviral response and the viralinhibitory mechanism (21). Such a host mechanism could be atherapeutic target to enhance host defense against viruses.

Here, we provide evidence that EMCV-induced SGs are in-volved in regulating IFN-� gene expression. Thus, virus-induced

FIG 12 EMCV 3C but not leader inhibits SG. (A) HeLa/G-G3BP1 and HeLa/G-G3BP1Q325E cells were transiently transfected with an empty vector or theexpression vector for leader or 3C for 48 h. Cells were treated with 0.5 mM sodium arsenite for 30 min, fixed, and stained for TIAR, an SG marker. (B) HeLa cellswere transiently transfected with and empty vector or the expression vector for leader or 3C (0 �g, 2 �g, and 4 �g) for 48 h. Cells were mock treated or transfectedwith long poly(I·C) (2 �g/�l) for 12 h. Total RNA was collected, and the mRNA level for IFN-� was determined by RT-qPCR. Data are representative of threeindependent experiments. (Error bars indicate standard deviations of duplicates [n � 3].)

Loss of Stress Granules Impairs Interferon Signals

September 2013 Volume 87 Number 17 jvi.asm.org 9521

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from

SGs might play dual roles: (i) suppressing viral replicationthrough an inhibition of viral protein synthesis and (ii) serving asa platform to facilitate IFN-� production.

ACKNOWLEDGMENTS

We thank Jamal Tazi for providing pEGFP-G3BP1, A. C. Palmenberg forexpression vectors for EMCV leader and 3C proteins, and Gabriel Fung(University of British Columbia) and Peter Gee (Kyoto University) forproofreading the manuscript.

This research was supported by grants from the Ministry of Education,Culture, Sports, Science and Technology (MEXT) of Japan (innovativeareas, infection competency [no. 24115004] and scientific research A [no.23249023]); the Ministry of Health, Labor and Welfare of Japan; the Ue-hara Memorial Foundation; the Mochida Memorial Foundation for Med-ical and Pharmaceutical Research; the Takeda Science Foundation; theNaito Foundation; and Nippon Boehringer Ingelheim. C.S.N. is a recipi-ent of a Monbukagakusho fellowship from the MEXT. We declare that wehave no conflicts of interest.

REFERENCES1. Meylan E, Tschopp J, Karin M. 2006. Intracellular pattern recognition

receptors in the host response. Nature 442:39 – 44.2. Darnell JE, Jr, Kerr IM, Stark GR. 1994. Jak-STAT pathways and tran-

scriptional activation in response to IFNs and other extracellular signalingproteins. Science 264:1415–1421.

3. Garcia MA, Gil J, Ventoso GI, Guerra S, Domingo E, Rivas C, EstebanM. 2006. Impact of protein kinase PKR in cell biology: from antiviral toantiproliferative action. Microbiol. Mol. Biol. Rev. 70:1032–1060.

4. Der S, Lau AS. 1995. Involvement of the double-stranded-RNA-dependent kinase PKR in interferon expression and interferon-mediatedantiviral activity. Proc. Natl. Acad. Sci. U. S. A. 92:8841– 8845.

5. Terenzi F, DeVeer M, Ying H, Restifo NP, Williams BR, Silverman RH.1999. The antiviral enzymes PKR and RNase L suppress gene expressionfrom viral and non-viral based vectors. Nucleic Acids Res. 27:4369 – 4375.

6. McInerney GM, Kedersha NL, Kaufman RJ, Anderson P, Liljeström P.2005. Importance of eIF2� phosphorylation and stress granule assemblyin alphavirus translation regulation. Mol. Biol. Cell 16:3753–3763.

7. White JP, Lloyd RE. 2012. Regulation of stress granules in virus systems.Trends Microbiol. 20:175–183.

8. Borghese F, Michiels T. 2011. The leader protein of cardioviruses inhibitsstress granule assembly. J. Virol. 85:9614 –9622.

9. Katoh H, Okamoto T, Fukuhara T, Kamban H, Morita E, Mori Y,Kamitani W, Matsuura Y. 2013. Japanese encephalitis virus core proteininhibits stress granule formation through an interaction with Caprin-1and facilitates viral propagation. J. Virol. 87:489 –502.

10. Okonski KM, Samuel CE. 2013. Stress granule formation induced bymeasles virus is protein kinase PKR dependent and impaired by RNAadenosine deaminase ADAR1. J. Virol. 87:756 –766.

11. Dinh PX, Beura LK, Das PB, Panda D, Das A, Pattnaik AK. 2013.Induction of stress granule-like structures in vesicular stomatitis virus-infected cells. J. Virol. 87:372–383.

12. Onomoto K, Jogi M, Yoo JS, Narita R, Morimoto S, Takemura A,Sambhara S, Kawaguchi A, Osari S, Nagata K, Matsumiya T, Namiki H,Yoneyama M, Fujita T. 2012. Critical role of an antiviral stress granulecontaining RIG-I and PKR in viral detection and innate immunity. PLoSOne 7:e43031. doi:10.1371/journal.pone.0043031.

13. Tourrière H, Chebli K, Zekri L, Courselaud B, Blanchard JM, BertrandE, Tazi J. 2003. The RasGAP-associated endoribonuclease G3BP assem-bles stress granules. J. Cell Biol. 160:823– 831.

14. Kedersha N, Tisdale S, Hickman T, Anderson P. 2008. Real-time andquantitative imaging of mammalian stress granules and processing bodies.Methods Enzymol. 448:521–552.

15. White JP, Cardenas AM, Marissen WE, Lloyd RE. 2007. Inhibition ofcytoplasmic mRNA stress granule formation by a viral proteinase. CellHost Microbe 15:295–305.

16. Porter FW, Bochkov YA, Albee AJ, Wiese C, Palmenberg AC. 2006. Apicornavirus protein interacts with Ran-GTPase and disrupts nucleocyto-plasmic transport. Proc. Natl. Acad. Sci. U. S. A. 103:12417–12422.

17. Ouda R, Onomoto K, Takahasi K, Edwards MR, Kato H, Yoneyama M,Fujita T. 2011. Retinoic acid-inducible gene I-inducible miR-23b inhibits

infections by minor group rhinoviruses through down-regulation of thevery low density lipoprotein receptor. J. Biol. Chem. 286:26210 –26219.

18. Garcia-Sastre A, Egorov A, Matassov D, Brandt S, Levy DE, Durbin JE,Palese P, Muster T. 1998. Influenza A virus lacking the NS1 gene repli-cates in interferon-deficient systems. Virology 252:324 –330.

19. Tisoncik JR, Billharz R, Burmakina S, Belisie SE, Proll SC, Korth MJ,Garcia-Sastre A, Katze MG. 2011. The NS1 protein influenza A virussuppresses interferon-regulated activation of antigen-presentation andimmune-proteasome pathways. J. Gen. Virol. 92:2093–2104.

20. Kharraz Y, Salmand PA, Camus A, Auriol J, Gueydan C, Kruys V,Morello D. 2010. Impaired embryonic development in mice overexpress-ing the RNA-binding TIAR. PLoS One 5:e11352. doi:10.1371/journal.pone.0011352.

21. Ruggieri A, Dazert E, Metz P, Hofmann S, Bergeest JP, Mazur J,Bankhead P, Hiet MS, Kallis S, Alvisi G, Samuel CE, Lohmann V,Kaderali L, Rohr K, Frese M, Stoecklin G, Bartenschlager R. 2012.Dynamic oscillation of translation and stress granule formation mark thecellular response to virus infection. Cell Host Microbe 12:71– 85.

22. Kato H, Sato S, Yoneyama M, Yamamoto M, Uematsu S, Matsui K,Tsujimura T, Takeda K, Fujita T, Takeuchi O, Akira S. 2005. Cell type-specific involvement of RIG-I in antiviral response. Immunity 23:19–28.

23. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K,Uematsu S, Jung A, Kawai T, Ishii KJ, Yamaguchi O, Otsu K, TsujimuraT, Koh CS, Reis e Sousa C, Matsuura Y, Fujita T, Akira S. 2006.Differential roles of MDA5 and RIG-I helicases in the recognition of RNAviruses. Nature 441:101–105.

24. Ng CS, Kato H, Fujita T. 2012. Recognition of viruses in the cytoplasm byRLRs and other helicases— how conformational changes, mitochondrialdynamics and ubiquitination control innate immune responses. Int. Im-munol. 24:739 –749.

25. Feng Q, Hato SV, Langereis MA, Zoll J, Virgen-Slane R, Peisley A, HurS, Semler BL, van Rij RP, van Kuppeveld FJ. 2012. MDA5 detects thedouble-stranded RNA replicative form in picornavirus-infected cells. CellRep. 2:1187–1196.

26. Lindquist ME, Mainou BA, Dermody TS, Crowe JE, Jr. 2011. Activation ofprotein kinase R is required for induction of stress granules by respiratorysyncytial virus but dispensable for viral replication. Virology 413:103–110.

27. Khaperskyy DA, Hatchette TF, McCormick C. 2012. Influenza A virusinhibits cytoplasmic stress granule formation. FASEB J. 26:1629 –1639.

28. Simpson-Holley M, Kedersha N, Dower K, Rubins KH, Anderson P,Hensley LE, Connor JH. 2011. Formation of antiviral cytoplasmic gran-ules during orthopoxvirus infection. J. Virol. 85:1581–1593.

29. Emara MM, Brinton MA. 2007. Interaction of TIA-1/TIAR with West Nileand dengue virus products in infected cells interferes with stress granule for-mation and processing body. Proc. Natl. Acad. Sci. U. S. A. 104:9041–9046.

30. Qin Q, Hastings C, Miller CL. 2009. Mammalian orthoreovirus particlesinduce and are recruited into stress granules at early times postinfection. J.Virol. 83:11090 –11101.

31. Langereis MA, Feng Q, van Kuppeveld FJ. 27 March 2013. MDA5localizes to stress granules but this localization is not required for theinduction of type I interferon. J. Virol. doi:10.1128/JVI.03213-12.

32. Barral PM, Morrison JM, Drahos J, Gupta P, Sarker D, Fisher PB,Racaniello VR. 2007. MDA-5 is cleaved in poliovirus-infected cells. J.Virol. 81:3677–3684.

33. Barral PM, Sarkar D, Fisher PB, Racaniello VR. 2009. RIG-I is cleavedduring picornavirus infection. Virology 391:171–176.

34. Iseni F, Garcin D, Nishio M, Kedersha N, Anderson P, Kolakofsky D.2002. Sendai virus trailer RNA binds TIAR, a cellular protein involved invirus-induced apoptosis. EMBO J. 21:5141–5150.

35. Ricour C, Delhaye S, Hato SV, Olenyik TD, Michel B, van KuppeveldFJ, Gustin KE, Michiels T. 2009. Inhibition of mRNA export anddimerization of interferon regulatory factor 3 by Theiler’s virus leaderprotein. J. Gen. Virol. 90:177–186.

36. Delhaye S, van Pesch V, Michiels T. 2004. The leader protein of Theiler’svirus interferes with nucleocytoplasmic trafficking of cellular proteins. J.Virol. 78:4357– 4362.

37. Paul S, Michiels T. 2006. Cardiovirus leader proteins are functionallyinterchangeable and have evolved to adapt to virus replication fitness. J.Gen. Virol. 87:1237–1246.

38. Hato SV, Ricour C, Schulte BM, Lanke KH, de Bruijni M, Zoll J, MelchersWJ, Michiels T, van Kuppeveld FJ. 2007. The mengovirus leader proteinblocks interferon-alpha/beta gene transcription and inhibits activation of in-terferon regulatory factor 3. Cell. Microbiol. 9:2921–2930.

Ng et al.

9522 jvi.asm.org Journal of Virology

on April 9, 2016 by P

EN

N S

TA

TE

UN

IVhttp://jvi.asm

.org/D

ownloaded from