12
IMMUNOLOGY 2017 © The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. IRE1a promotes viral infection by conferring resistance to apoptosis Susan L. Fink, 1,2,3 * Teshika R. Jayewickreme, 1 Ryan D. Molony, 1 Takao Iwawaki, 4 Charles S. Landis, 5 Brett D. Lindenbach, 6,7 Akiko Iwasaki 1,8 * The unfolded protein response (UPR) is an ancient cellular pathway that detects and alleviates protein-folding stresses. The UPR components X-box binding protein 1 (XBP1) and inositol-requiring enzyme 1a (IRE1a) promote type I interferon (IFN) responses. We found that Xbp1-deficient mouse embryonic fibroblasts and macrophages had impaired antiviral resistance. However, this was not because of a defect in type I IFN responses but rather an inability of Xbp1-deficient cells to undergo viral-induced apoptosis. The ability to undergo apoptosis limited infection in wild-type cells. Xbp1-deficient cells were generally resistant to the intrinsic pathway of apoptosis through an indirect mechanism involving activation of the nuclease IRE1a. We observed an IRE1a-dependent reduction in the abundance of the proapoptotic microRNA miR-125a and a corresponding increase in the amounts of the members of the antiapoptotic Bcl-2 family. The activation of IRE1a by the hepatitis C virus (HCV) protein NS4B in XBP1-proficient cells also conferred apoptosis resistance and promoted viral replication. Furthermore, we found evidence of IRE1a activation and decreased miR-125a abundance in liver biopsies from patients infected with HCV compared to those in the livers of healthy controls. Our results reveal a prosurvival role for IRE1a in virally infected cells and suggest a possible target for IFN-independent antiviral therapy. INTRODUCTION Great advances have been made in our understanding of the molecular definitions of pattern recognition receptors (PRRs) and the pathogen- associated molecular patterns (PAMPs) that cells use to distinguish viruses from self (1, 2). PRR engagement results in the transcription of the genes that encode the type I interferons (IFNs) IFN-a and IFN-b, which bind to the IFN-a/b receptor to induce the expression of hundreds of IFN-stimulated genes (ISGs). ISGs act in concert to block further viral replication and spread, as well as to support the activation of adaptive antiviral immunity (3). However, many viruses have evolved evasion mechanisms to limit PRR recognition and signal transduction, and PRR-independent mechanisms for innate sensing of viral infections remain unclear. Endoplasmic reticulum (ER) stress occurs during infection by vari- ous viruses, presumably due to the overwhelming synthesis of viral pro- teins (4). The unfolded protein response (UPR) is a ubiquitous cellular pathway to detect and alleviate ER stress. The UPR is initiated by three sensors that reside within the ER: protein kinase receptorlike ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 (IRE1) (57). IRE1, a highly conserved UPR sensor, oligo- merizes and autophosphorylates in response to ER stress, which activates its cytosolic ribonuclease (RNase) domain and initiates a nonconven- tional mRNA splicing reaction of Xbp1 mRNA (8). Once processed, the spliced Xbp1 mRNA encodes a transcription factor, which controls the expression of target genes. IRE1a targets other specific mRNAs, leading to their degradation in a process termed regulated IRE1- dependent decay (RIDD) (9, 10). ER stress synergistically enhances cytokine and IFN responses to PRR engagement through IRE1a and X-box binding protein 1 (XBP1) (1113). Specific activation of IRE1a also occurs during innate immune recognition of PAMPs by Toll-like receptors (TLRs) (11). In this setting, XBP1 promotes the production of inflammatory cytokines and IFN-b. Moreover, IRE1a generates ligands for RIG-Ilike receptors (RLRs) during the UPR (14), which are degraded by SKIV2L RNA exosomes to prevent in- appropriate activation of type I IFN responses (15). These observations prompted us to investigate the possible role of XBP1 in innate immune responses to viral infections, with the hypothesis that XBP1 could pro- mote IFN-mediated viral resistance. Here, we describe an unexpected role for XBP1 in antiviral resistance, not through enhancement of the IFN response but rather by modu- lating susceptibility to host cell apoptosis. Xbp1-deficient cells were resistant to apoptosis during infection with vesicular stomatitis virus (VSV) and herpes simplex virus (HSV), and failure to undergo cell death resulted in increased viral replication. Xbp1 deficiency results in activation of its upstream enzyme IRE1a, which degrades specific cytosolic RNA targets (16, 17). We found that apoptosis resistance in the Xbp1-deficient cells required IRE1a and its degradation of microRNA (miRNA), miR-125a. Notably, the hepatitis C virus (HCV) nonstructural protein 4B (NS4B), which stimulates IRE1a activation (18), promoted the survival of infected cells and viral replication. Moreover, liver biopsies from patients infected with HCV showed IRE1a activation and reduced miR-125a abundance compared to healthy controls. These findings highlight the role of UPR effectors in regulating IFN-independent mechanisms of innate antiviral resistance through the induction of apoptosis to limit viral infection. RESULTS Xbp1 deficiency impairs control of viral infection To determine the effect of Xbp1 deficiency on host defense against viral replication, we infected Xbp1 -/- mouse embryonic fibroblasts (MEFs) with an RNA virus, VSV, and a DNA virus, HSV. XBP1 deficiency was 1 Department of Immunobiology, Yale University, New Haven, CT 06520, USA. 2 De- partment of Laboratory Medicine, Yale University, New Haven, CT 06520, USA. 3 Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA. 4 Division of Cell Medicine, Department of Life Science, Medical Re- search Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan. 5 Divi- sion of Gastroenterology and Hepatology, Department of Medicine, University of Washington, Seattle, WA 98195, USA. 6 Department of Microbial Pathogenesis, Yale University, New Haven, CT 06520, USA. 7 Department of Comparative Medi- cine, Yale University, New Haven, CT 06520, USA. 8 Howard Hughes Medical Insti- tute, Chevy Chase, MD 20814, USA. *Corresponding author. Email: [email protected] (A.I.); [email protected] (S.L.F.) SCIENCE SIGNALING | RESEARCH ARTICLE Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017 1 of 11 on January 30, 2021 http://stke.sciencemag.org/ Downloaded from

IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

IMMUNOLOGY

1Department of Immunobiology, Yale University, New Haven, CT 06520, USA. 2De-partment of Laboratory Medicine, Yale University, New Haven, CT 06520, USA.3Department of Laboratory Medicine, University of Washington, Seattle, WA98195, USA. 4Division of Cell Medicine, Department of Life Science, Medical Re-search Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan. 5Divi-sion of Gastroenterology and Hepatology, Department of Medicine, Universityof Washington, Seattle, WA 98195, USA. 6Department of Microbial Pathogenesis,Yale University, New Haven, CT 06520, USA. 7Department of Comparative Medi-cine, Yale University, New Haven, CT 06520, USA. 8Howard Hughes Medical Insti-tute, Chevy Chase, MD 20814, USA.*Corresponding author. Email: [email protected] (A.I.); [email protected] (S.L.F.)

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

2017 © The Authors,

some rights reserved;

exclusive licensee

American Association

for the Advancement

of Science.

Dow

nloade

IRE1a promotes viral infection by conferring resistanceto apoptosisSusan L. Fink,1,2,3* Teshika R. Jayewickreme,1 Ryan D. Molony,1 Takao Iwawaki,4

Charles S. Landis,5 Brett D. Lindenbach,6,7 Akiko Iwasaki1,8*

The unfolded protein response (UPR) is an ancient cellular pathway that detects and alleviates protein-foldingstresses. The UPR components X-box binding protein 1 (XBP1) and inositol-requiring enzyme 1a (IRE1a) promotetype I interferon (IFN) responses. We found that Xbp1-deficient mouse embryonic fibroblasts and macrophageshad impaired antiviral resistance. However, this was not because of a defect in type I IFN responses but rather aninability of Xbp1-deficient cells to undergo viral-induced apoptosis. The ability to undergo apoptosis limited infectionin wild-type cells. Xbp1-deficient cells were generally resistant to the intrinsic pathway of apoptosis through anindirectmechanism involving activation of the nuclease IRE1a. We observed an IRE1a-dependent reduction in theabundance of the proapoptoticmicroRNAmiR-125a and a corresponding increase in the amounts of themembersof the antiapoptotic Bcl-2 family. The activation of IRE1a by the hepatitis C virus (HCV) protein NS4B in XBP1-proficientcells also conferred apoptosis resistance and promoted viral replication. Furthermore, we found evidence of IRE1aactivation and decreased miR-125a abundance in liver biopsies from patients infected with HCV compared to thosein the livers of healthy controls. Our results reveal a prosurvival role for IRE1a in virally infected cells and suggest apossible target for IFN-independent antiviral therapy.

d f

on January 30, 2021

http://stke.sciencemag.org/

rom

INTRODUCTIONGreat advances have been made in our understanding of the moleculardefinitions of pattern recognition receptors (PRRs) and the pathogen-associated molecular patterns (PAMPs) that cells use to distinguishviruses from self (1, 2). PRR engagement results in the transcription ofthe genes that encode the type I interferons (IFNs) IFN-a and IFN-b,which bind to the IFN-a/b receptor to induce the expression ofhundreds of IFN-stimulated genes (ISGs). ISGs act in concert to blockfurther viral replication and spread, as well as to support the activationof adaptive antiviral immunity (3). However,many viruses have evolvedevasion mechanisms to limit PRR recognition and signal transduction,andPRR-independentmechanisms for innate sensing of viral infectionsremain unclear.

Endoplasmic reticulum (ER) stress occurs during infection by vari-ous viruses, presumably due to the overwhelming synthesis of viral pro-teins (4). The unfolded protein response (UPR) is a ubiquitous cellularpathway to detect and alleviate ER stress. The UPR is initiated by threesensors that residewithin the ER: protein kinase receptor–like ERkinase(PERK), activating transcription factor 6 (ATF6), and inositol-requiringenzyme 1 (IRE1) (5–7). IRE1, a highly conserved UPR sensor, oligo-merizes and autophosphorylates in response to ER stress, which activatesits cytosolic ribonuclease (RNase) domain and initiates a nonconven-tional mRNA splicing reaction of Xbp1 mRNA (8). Once processed,the splicedXbp1mRNAencodes a transcription factor, which controlsthe expression of target genes. IRE1a targets other specific mRNAs,leading to their degradation in a process termed regulated IRE1-

dependent decay (RIDD) (9, 10). ER stress synergistically enhancescytokine and IFN responses to PRR engagement through IRE1aand X-box binding protein 1 (XBP1) (11–13). Specific activation ofIRE1a also occurs during innate immune recognition of PAMPs byToll-like receptors (TLRs) (11). In this setting, XBP1 promotes theproduction of inflammatory cytokines and IFN-b. Moreover, IRE1agenerates ligands for RIG-I–like receptors (RLRs) during the UPR(14), which are degraded by SKIV2L RNA exosomes to prevent in-appropriate activation of type I IFN responses (15). These observationsprompted us to investigate the possible role of XBP1 in innate immuneresponses to viral infections, with the hypothesis that XBP1 could pro-mote IFN-mediated viral resistance.

Here, we describe an unexpected role for XBP1 in antiviral resistance,not through enhancement of the IFN response but rather by modu-lating susceptibility to host cell apoptosis. Xbp1-deficient cells wereresistant to apoptosis during infection with vesicular stomatitis virus(VSV) and herpes simplex virus (HSV), and failure to undergo celldeath resulted in increased viral replication. Xbp1 deficiency resultsin activation of its upstream enzyme IRE1a, which degrades specificcytosolic RNA targets (16, 17). We found that apoptosis resistance intheXbp1-deficient cells required IRE1a and its degradationofmicroRNA(miRNA),miR-125a. Notably, the hepatitis C virus (HCV) nonstructuralprotein 4B (NS4B), which stimulates IRE1a activation (18), promotedthe survival of infected cells and viral replication.Moreover, liver biopsiesfrom patients infected with HCV showed IRE1a activation and reducedmiR-125a abundance compared to healthy controls. These findingshighlight the role of UPR effectors in regulating IFN-independentmechanisms of innate antiviral resistance through the induction ofapoptosis to limit viral infection.

RESULTSXbp1 deficiency impairs control of viral infectionTo determine the effect of Xbp1 deficiency on host defense against viralreplication, we infected Xbp1−/− mouse embryonic fibroblasts (MEFs)with an RNA virus, VSV, and a DNA virus, HSV. XBP1 deficiency was

1 of 11

Page 2: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

http://sD

ownloaded from

achieved through an insertion of the neomycin resistance gene intoparts of exons 1 and 2, as well as the intervening intron (19). Thisinsertion still enables Xbp1 mRNA splicing but results in a frameshiftof the remaining amino acids to prevent protein production. Comparedto wild-type (WT) MEFs, VSV replication was enhanced in Xbp1−/−

MEFs as determined by measuring VSV-G–green fluorescent protein(GFP) relative abundance by flow cytometry (Fig. 1, A and B) and byplaque assays of released virus from MEF culture medium (Fig. 1C).Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance (Fig. 1, Dand E) and viral titer in the supernatant (Fig. 1F).

To determine whether the impaired viral control in Xbp1−/− MEFsresulted from deficient IFN responses, we measured the expression ofgenes encoding type I IFNs and of an ISG,Mx1, in MEFs infected withVSV. Unexpectedly, we observed enhanced induction of Ifna4 andIfnb1 in Xbp1−/− MEFs infected with VSV compared to WT MEFs(fig. S1, A and B). Induction of Mx1 also increased in Xbp1−/− MEFs(fig. S1C). Moreover, Xbp1−/−MEFs were not impaired in IFN respon-siveness because pretreatment with IFN-b preventedVSV replication inXbp1−/− MEFs (fig. S1D). In contrast and consistent with previous re-ports (12, 13), the Xbp1-deficient MEF response to transfection withpolyinosinic:polycytidylic acid [poly(I:C)] [an MDA5 (melanomadifferentiation–associated protein 5) agonist] was impaired (fig. S1E),suggesting that the enhanced IFN response to VSV was specific to rep-licating virus. Together, these findings suggest that Xbp1 contributes toprotective antiviral responses independently of type I IFNs.

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

Xbp1 deficiency confers resistance to virus-triggeredcell deathViral infection often culminates in the death of infected host cells. Todetermine whether the phenotype we observed in the Xbp1-deficientMEFs was due to a difference in the death of infected cells, we evaluatedcell death and the abundance of virally encoded GFP after infection.During VSV infection, we found thatmost of theWT cells (~85%)weredead 24 hours after infection. In contrast, most Xbp1−/− MEFs (~74%)were resistant to cell death and accumulated higher amounts of viralprotein as determined by measurement of GFP abundance (Fig. 2, Aand B). Similarly, whereas a large proportion of HSV-infectedWT cellsunderwent cell death, infectedXbp1−/−MEFswere resistant to death (Fig.2, C and D). To determine whether acute ablation of Xbp1 expression

on January 30, 2021tke.sciencem

ag.org/

Xbp1–/–

WT

Mock

A VSV-GFP

Mock

D HSV-GFP

WT

VSV

GF

P M

FI

B

VSV

PF

U/m

l (lo

g)

C

Xbp1–/–

WTXbp1–/–

0

100

200

300

400

500

6

7

8******

GF

P M

FI

E

PF

U/m

l (lo

g)

F

WTXbp1–/–

****

0

50

100

150

200

250

5

6

7

8

GFP

% o

f M

ax%

of

Max

Fig. 1. Xbp1 deficiency enhances the susceptibility of MEFs to HSV and VSV.(A to F) WT and Xbp1−/− MEFs were infected with VSV-GFP at a multiplicity ofinfection (MOI) of 1 [(A) to (C)] or with HSV-1–GFP at an MOI of 10 [(D) to (F)].Twenty-four hours later, the extent of infection was determined by measuringthe relative abundance of GFP by flow cytometry. Data are from one experimentrepresentative of three independent experiments (A and D). The mean fluores-cence intensity (MFI) of GFP in the indicated cells was then determined. Data aremeans ± SD of three independent experiments (B and E). Viral titers in the cellculture medium were measured by plaque assay at 48 (C) and 72 (F) hours afterinfection. PFU, plaque-forming units. Data are means ± SD of three independentexperiments (C and F). *P < 0.05; ***P < 0.001 compared to WT, unpaired t test.

A

% D

ead

VSV

Xbp1–/–

–/–

WT

Mock

B

HSV

WTC

% D

ead

Mock HSV

D

Mock VSV

**

% V

iabi

lity

%V

iabi

lity

E

0

20

40

60 **F

0

20

40

60

80

100

120

Live

/dea

d

GFP

WT

Xbp1

Live

/dea

d

GFP

Xbp1

WT

Xbp1

WT

Xbp1Δ

0

20

40

60

80

100 **

0

20

40

60

80 **

–/–

–/–

Fig. 2. Xbp1-deficient cells are resistant to cell death during infection with VSVand HSV. (A to D) WT and Xbp1−/−MEFs were left uninfected (mock) or were infectedwith VSV-GFP [(A) and (B)] or HSV-1–GFP [(C) and (D)] for 24 hours. Cell deathwas thenassessed with a membrane-impermeant, amine-reactive fluorescent dye, which wasmeasured by flow cytometry. Data are from one experiment and are representativeof three experiments (A and C). The percentages of dead cells were then determined.Data are means ± SD of three independent experiments (B and D). (E and F) BMDMswere cultured from Xbp1flox/flox ESR Cre+ (Xbp1D) or Cre− littermate (WT) mice in thepresence of tamoxifen. Cells were infected with VSV-GFP at the indicated MOI for24 hours. Viability was then assessed by measuring 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) reduction. Data aremeans ± SD of three replicates and are representative of three experiments (E and F).**P < 0.01 compared to WT, unpaired t test.

2 of 11

Page 3: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

would have a similar effect, we treated WT MEFs with smallinterfering RNA (siRNA) targeting Xbp1. Xbp1 knockdown stronglysuppressed VSV-induced cell death and enhanced production ofvirally encoded GFP (fig. S2, A and B), consistent with the results ofexperiments with Xbp1−/− MEFs. Reconstitution of Xbp1−/− MEFswith plasmid-encoded Xbp1 restored VSV-induced cell death andrestricted the production of virally encoded GFP (fig. S2C). To deter-mine whether these findings extended to additional cell types, wecultured bone marrow–derived macrophages (BMDMs) from micewith a tamoxifen-inducible conditional Xbp1 deletion (Xbp1flox/flox

ESR Cre) (20). Xbp1D BMDMs were resistant to death during VSVinfection (Fig. 2, E and F, and fig. S2D), indicating that Xbp1 geneticdeficiency results in protection from cell death in fibroblasts andmacrophages. These results suggest that there should not be anXbp1-dependent antiviral phenotype against viruses that do nottrigger the death of infected cells. We found that infection with aVSV-Gpseudotyped lentivirus encodingGFPdid not result in host celldeath (fig. S3A). In this case, we did not observe enhanced productionof virally encoded GFP in Xbp1−/− MEFs (fig. S3, A and B). Thesefindings suggest that impaired control of VSV and HSV infection byXbp1−/− MEFs was directly related to their resistance to cell death.

Apoptosis limits the replication of VSV and HSVThe end point of cell death can result from numerous upstreamsignaling pathways. VSV infection induces apoptotic cell death inJurkat cells (21) and MCF-7 breast adenocarcinoma cells (22). WTMEFs infected with VSV demonstrated active caspase-3 (Fig. 3A), in-dicating apoptotic cell death in these cells. In contrast, Xbp1−/−MEFsfailed to activate caspase-3 during VSV infection (Fig. 3A). Similarly,HSV-infected WT MEFs contained active caspase-3, and Xbp1−/−

MEFs were resistant to the activation of this apoptotic effector (Fig.3B). These findings were not limited to MEFs because the activationof caspase-3 and caspase-7 also occurred in VSV-infected BMDMs andXbp1D BMDMs were resistant to VSV-induced caspase-3 activation(Fig. 3C).

Some viruses induce apoptosis as a means of viral transmission andavoidance of the immune system (23). In other cases, apoptosis isbeneficial for the host and limits viral replication. We observed de-creased abundance of virally encoded GFP in the population of deadcells during HSV infection of WT MEFs (Fig. 2C), suggesting that ap-optosis may limit viral replication. To test this hypothesis, we added acaspase inhibitor, carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (zVAD), to infected cells. We found that zVAD

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

prevented the death of VSV-infected MEFs (Fig. 3D) and led to theincreased abundance of virally encoded GFP (Fig. 3, D and E), pheno-copying the result obtained from experiments with Xbp1−/− MEFs.Similarly, inhibition of apoptosis with zVAD increased the abundanceof virally encodedGFP inHSV-infectedMEFs, to an extent similar tothat attained in Xbp1−/−MEFs (Fig. 3F). In addition, overproductionof the antiapoptotic protein BCL2 limited the death of VSV-infectedcells and led to the increased abundance of virally encoded GFP (fig.S3C). Together, these findings suggest that Xbp1-deficient cells areresistant to virus-induced apoptosis and that apoptosis directly limitsviral replication.

A

Mock MOI = 2 MOI = 0.4

Rel

ativ

e ca

spas

e-3

activ

ity

C *

*

DVSV

WT Xbp1–/– WT + zVAD

Mock

E VSV-GFP

Mock

F HSV-GFP

VSV

VSV HSV

Mock Mock

% o

f M

ax

% o

f M

ax

Active caspase-3 Active caspase-3

B

% o

f M

ax

% o

f M

ax

012345678

WTXbp1–/–

WTXbp1–/–

WT

Xbp1Δ

Live

/dea

d

GFP

WT

Xbp1–/– –/–

WT+ zVAD

WT Xbp1

WT+ zVAD

Fig. 3. Apoptosis induced by VSV and HSV limits viral infection. (A to B) WTand Xbp1−/− MEFs were left uninfected (mock) or were infected with VSV-GFP(A) or HSV-1–GFP (B) for 24 hours. Cells were then stained with an antibodyspecific for active (cleaved) caspase-3, which wasmeasured by flow cytometry. Dataare from one experiment and are representative of three experiments. (C) BMDMswere cultured from Xbp1flox/flox ESR Cre+ (Xbp1D) or Cre− littermate (WT) mice in thepresence of tamoxifen. Cells were infected with VSV-GFP at the indicated MOI for7 hours. Caspase-3 activity was then assessed by measuring fluorometric sub-strate cleavage and is shown relative to that in uninfected cells. Data are means ±SD of three replicates and are representative of three experiments. (D to F) MEFswere infected in the presence of zVAD to inhibit caspase activity. Twenty-four hoursafter infection, cell death was assessed with a membrane-impermeant, amine-reactive fluorescent dye, which was measured by flow cytometry. The extent ofinfection was determined by measuring the relative abundance of GFP by flow cy-tometry. Data are fromone experiment and are representative of three independentexperiments. *P < 0.01 compared to WT, unpaired t test.

3 of 11

Page 4: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

Resistance to virally induced apoptosis in Xpb1−/− cells isindependent of Beclin 1 and CHOPER stress is associatedwith autophagy,which regulates cell survival (24).In particular, XBP1 promotes transcription of the gene encoding theautophagy component, Beclin 1 (25). Consistent with these data, wefound decreased Beclin 1 in Xbp1-deficient cells (fig. S4A). However,knockdown of Beclin 1 with Beclin 1–specific siRNA did not affectVSV infection or the induction of cell death in either WT or Xbp1-deficient MEFs (fig. S4B). Our finding that Xbp1-deficient cells wereresistant to VSV- and HSV-induced apoptosis suggests the possibilitythat apoptosis during viral infection may directly result from XBP1-mediated transcriptional activity. During ER stress, XBP1 partially con-tributes to the production of the apoptosis mediator CHOP (C/EBPhomologous protein) (26), an induction process that may play a role invirus-induced apoptosis. As a functional control, transfection withChop-specific siRNA prevented the death of MEFs treated with theER stress–inducing agents tunicamycin and thapsigargin (fig. S5, A andB). In contrast, Chop knockdown did not prevent the death of VSV- orHSV-infected MEFs (fig. S5B). Further arguing against a direct role forXBP1-mediated transcriptional activity in virally induced cell death, wedid not observe Xbp1 splicing (fig. S6A) or induction of the expressionof the UPR-responsive genes Hspa5 [encoding BiP (binding immuno-globulin protein)] and Chop during VSV infection (fig. S6, B and C),consistent with published observations for VSV (27) and HSV (28, 29).

Xbp1-deficient cells are resistant to the intrinsic pathwayof apoptosisBecause we did not find evidence for XBP1-mediated transcriptionalactivity in promoting apoptosis during infection, we hypothesized thatXbp1-deficient cells may be inherently resistant to apoptosis in general.We therefore treated Xbp1−/−MEFs with a panel of apoptosis-inducingstimuli. Staurosporine and gliotoxin stimulate the intrinsic or mito-chondrial pathway of apoptosis (30–33), whereas ligation of the tumornecrosis factor (TNF) and Fas receptors initiates the extrinsic apoptoticpathway (34). Xbp1−/− MEFs were specifically resistant to stimuli thatinduced the intrinsic pathway of apoptosis, both as demonstrated byincreased viability (Fig. 4A) and impaired caspase-3 activation (Fig.4B). Consistent with previous studies demonstrating a protective roleof Xbp1 during ER stress (26, 35–37), Xbp1−/− MEFs were slightlymore susceptible to death induced by tunicamycin (Fig. 4A). In con-trast, there was no difference betweenWTandXbp1−/−MEFs in necrot-ic death induced by a high concentration of cycloheximide (Fig. 4A).These findings were further verified in experiments with Xbp1DBMDMs, which demonstrated resistance to staurosporine and the se-lective Bcl-2 inhibitor ABT-737 (38) but not to TNF-induced apoptosis(Fig. 4C). Thus, Xbp1 genetic deficiency results in specific protectionfrom the intrinsic pathway of apoptosis.

The resistance of Xbp1-deficient cells to apoptosis resultsfrom the activation of IRE1aXbp1 deficiency results in activation of its upstream enzyme IRE1a,which degrades specific cytosolic RNA targets (16, 17). Although theycannotmakeXBP1s protein,Xbp1-deficient cells transcribemRNA thatcontains the IRE1a cleavage sites. Consistent with previous reports, weobserved IRE1a activation in Xbp1-deficient cells, indicated by Xbp1mRNA splicing (fig. S7A). The magnitude of IRE1a activation inXbp1-deficient cells was not as robust as the response to canonicalUPR stimulation with thapsigargin, nor were classical RIDD substratesdiminished (fig. S7B), consistent with other studies (39).

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

To determine whether IRE1a was involved in resistance to apopto-sis, we knocked down Ire1awith siRNA (fig. S7C). As a functional con-trol, Ire1a-specific siRNA efficiently prevented Xbp1 mRNA splicing(fig. S7D). IRE1a knockdown in Xbp1−/− MEFs reversed resistance toVSV-induced cell death (Fig. 5A and fig. S7E). Expression of human

0

20

40

60

80

100

120

Intrinsic Extrinsic UPR Necrosis

% V

iabi

lity

A

Sts Glio TNF Fas TM CHX

***

Intrinsic Extrinsic

Intrinsic Extrinsic Necrosis

% C

aspa

se-3

act

ivity

B

Sts Glio Fas

* **

% V

iabi

lity

*

Sts ABT-737 TNF CHX TM %

Via

bilit

y

*

0

10

20

30

40

50

60

70

0

20

40

60

80

100

120

140

160

0

50

100

150

200

WT

Xbp1–/–

–/–

WT

Xbp1

WT

Xbp1Δ

C

Fig. 4. Xbp1-deficient cells are resistant to the intrinsic pathway of apopto-sis. (A and B) WT and Xbp1−/− MEFs were treated with staurosporine (sts), glio-toxin (glio), TNF and low-dose cycloheximide (TNF), Fas antibody and low-dosecycloheximide (Fas), and tunicamycin (TM) to induce the UPR, or high-dose cyclo-heximide (CHX). Twenty-four hours later, viability was assessed by measuring MTSreduction (A). Seven hours after treatment, caspase-3 activity was assessed bymeasuring fluorometric substrate cleavage and is shown relative to WT cells(B). Data are means ± SD of three replicates and are representative of threeexperiments. (C) BMDMs were cultured from Xbp1flox/flox ESR Cre+ (Xbp1D) orCre− littermate (WT) mice in the presence of tamoxifen and treated with the in-dicated inducers of cell death as described in (A). Twenty-four hours later, viabilitywas assessed by measuring MTS reduction. Data are means ± SD of three repli-cates and are representative of three experiments. *P < 0.05; **P < 0.001 com-pared to WT, unpaired t test.

4 of 11

Page 5: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

IRE1a, which was resistant to mouse Ire1a-specific siRNA, preventedthis reversal (fig. S7F). IRE1a knockdown alone was minimally cyto-toxic but restored caspase-3 activation in Xbp1−/− MEFs in responseto staurosporine (Fig. 5B) and sensitized these cells to staurosporine-induced cell death (Fig. 5C). To determine whether the RNase activityof IRE1a mediated resistance to apoptosis, we treated Xbp1-deficientcells with the selective IRE1a nuclease inhibitor 4m8C (8-formyl-7-hydroxy-4-methylcoumarin) (40). We found that 4m8C reversed the

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

resistance of Xbp1-deficient cells toapoptosis both during infectionwith VSV and during treatmentwith staurosporine (Fig. 5D). As anegative control, the structurally sim-ilar compound AMC (7-amino-4-methylcoumarin) had no effect atan equimolar concentration (fig.S2D). These findings indicate thatthe RNase activity of IRE1a contributesto resistance to the intrinsic pathwayof apoptosis observed in the settingof Xbp1 deficiency. We consistentlyobserved Xbp1 mRNA splicing to asmall extent in WT cells (fig. S7, Aand C), suggesting that IRE1a hassome basal activity (41), which couldregulate apoptotic responses in WTcells. Consistent with this, IRE1aknockdown in WT MEFs was nottoxic alone (Fig. 5, B, C, and E) butsensitized cells to VSV-induced celldeath and limited viral replication(Fig. 5, E and F).

IRE1a targets the proapoptoticmiRNA miR-125aStudies of coding genes targeted byIRE1a for RIDD have not revealedobvious candidates to explain our ob-served IRE1a-mediated resistance toapoptosis (9, 42). Thus, we focusedon miRNAs, which are also targetedby RIDD (43). To this end, we per-formed miRNA profiling of Xbp1-deficient cells and found fourmiRNAsthat were decreased in abundance inXbp1-deficient cells with active IRE1a(Fig. 6A). The miRNA miR-125awas represented twice among thesedifferentially expressed miRNAs.Quantitative polymerase chain re-action (PCR) analysis confirmed thedecrease in miR-125a abundance inXbp1-deficient cells, which was re-stored by reconstitution of the cellswith plasmid-encoded Xbp1 (fig.S8A). The miRNA miR-125a sensi-tizes cells to apoptosis and is thoughtto negatively regulate antiapoptoticBcl-2 family members, including B

cell lymphoma–extra large (Bcl-xL) and myeloid cell leukemia1 (Mcl-1) (44–46). In accordance with decreased miR-125a, we foundan increase in the abundances of antiapoptotic Bcl-xL andMcl-1 pro-teins in Xbp1-deficient cells (Fig. 6B). To test whether these effectswere dependent on IRE1a, we crossed Xbp1flox/flox × ESR Cre+ miceto Ern1flox/flox mice. BMDMs obtained from Xbp1flox/flox × Ern1flox/flox ×ESR Cre+ mice were treated with tamoxifen to generate XBP1D IRE1aDcells. We found that the protein amounts of both Bcl-xL and Mcl-1 in

VSV

Rel

ativ

e ca

spas

e-3

activ

ity

WT Xbp1–/– –/–WT Xbp1

Mock Staurosporine

0

10

20

30

*A B

% V

iabi

lity

C

0

20

40

60

80

100

120

0

10

20

30

WT Xbp1

Mock

*

% V

iabi

lity

D

WTXbp1Δ Xbp1Δ

+ 4µ8C%

Via

bilit

y

Mock VSV Staurosporine0

20

40

60

80

100

120

Ctrl siRNA Ire1α siRNA

Mock

VSV

E

GFP

% o

f M

ax

VSV ctrl siRNAVSV Ire1α siRNAMock

F

Live

/dea

d

GFP

Xbp1WTXbp1–/–

–/–

–/– Xbp1–/–

+Ire1α siRNA

No siRNA

Ctrl siRNAIre1α siRNA

No siRNA

Ctrl siRNAIre1α siRNA

WT

Staurosporine

** **** **

Live

/dea

d

Fig. 5. The resistance of Xbp1-deficient cells to apoptosis results from the activation of IRE1a. (A to F) Analysis ofthe effects of IRE1a knockdown on cell death. (A, E, and F) WT and Xbp1−/− MEFs were transfected with siRNA targetingIre1a or control siRNA (ctrl siRNA). Cells were then left uninfected (mock) or infected with VSV-GFP for 24 hours. Celldeath was then assessed with a membrane-impermeant, amine-reactive fluorescent dye, and the relative abundance ofGFP was measured by flow cytometry. Data are from one experiment and are representative of three (A) or two (E and F)independent experiments. (B and C) The indicated siRNA-transfected MEFs were left untreated (mock) or treated withstaurosporine. Seven hours later, caspase-3 activity was assessed by measuring fluorometric substrate cleavage and isshown relative to that in untreated WT cells (B). Twenty-four hours after treatment, viability was assessed by measuringMTS reduction (C). (D) BMDMs were cultured from Xbp1flox/flox ESR Cre+ (Xbp1D) or Cre− littermate (WT) mice in thepresence of tamoxifen and the IRE1a inhibitor 4m8C. Cells were then infected with VSV-GFP at an MOI of 2 or weretreated with staurosporine. Viability was assessed 24 hours later by measuring MTS reduction. Data are means ± SDof three replicates and are representative of three experiments (B, C, and D). *P < 0.01; **P < 0.001, unpaired t test.

5 of 11

Page 6: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

Fink et al., Sci. Signal. 10, eaai7814 (2017

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

-

)

.x

)-

-f

f

-s

-

r

--

,s--.-.

XBP1D IRE1aD BMDMs were reduced compared to those in XBP1Dcells, albeit not to the amount observed in the WT cells (Fig. 6B).These results indicated that antiapoptotic Bcl-xL and Mcl-1 proteinswere increased in abundance inXbp1-deficient cells, in amanner largelydependent on IRE1a. Consistent with our observation of sensitizationto apoptosis by IRE1a knockdown in WT cells (Fig. 5E), IRE1aD cellshad increased miR-125a abundance (fig. S8B). Inhibition of the RNaseactivity of IRE1a with the selective IRE1a nuclease inhibitor 4m8C wassufficient to increase the abundance of miR-125a (fig. S8B).

Finally, we wished to examine the extent to which miR-125a degra-dation by IRE1a was responsible for the prosurvival phenotype of theXbp1-deficient cells. Reconstitution of miR-125a with an miRNA mi-metic was sufficient to restore caspase-3 activation in Xbp1−/−MEFsin response to staurosporine (Fig. 6C) and to sensitizeXbp1-deficientcells to the intrinsic pathway of apoptosis (Fig. 6D and fig. S8C). Re-

) 6 June 2017

constitution with miR-125a also reversed the resistance of Xbp1−/−

MEFs to VSV-induced cell death (Fig. 6E). Finally, neutralizing miR-125a in WT cells with an miRNA hairpin inhibitor resulted in anantiapoptotic state resembling that observed in Xbp1-deficient cells(fig. S8D). Together, these findings suggest that the IRE1a-dependentdecrease in miR-125a abundance contributes to the resistance toapoptosis.

IRE1a activation during HCV infection mediates resistanceto apoptosisSome viruses encode genes that promote IRE1a activation. HCVNS4Bactivates IRE1a (18), and IRE1a activation is also seen inHCV-infectedcells (47). Curiously, in cells expressing NS4B, IRE1a splices Xbp1mRNA, but XBP1 targets are not transcribed (18), suggesting that thevirus uses IRE1a for another reason. Furthermore, HCV is suggested to

A B

Bcl-xL

Mcl-1

β-Actin

0.0001

0.0010

0.0100

0.1000

1.0000−3 − −2 1 0 1 2

miR-125a-3p

miR-125a-5p

miR-1224

miR-804

Log2 fold changeP

vDecreased in Xbp1Δ Increased in Xbp1Δ

miR-1902

C

WT + miR-ctrl

Xbp1Δ + miR-ctrl

WT + miR-125a

Xbp1Δ + miR-125a

% V

iabi

lity

D

0

25

50

75

100

125

Rel

ativ

e ca

spas

e-3

activ

ity

0

20

40

60

Mock Staurosporine Mock Staurosporine

** **** **

E miR-ctrl miR-125a

WT

Xbp1−/−

GFP

% o

f M

ax%

of

Max

WT + miR-ctrlWT + miR-125aMock

Xbp1− −/ + miR-ctrl Xbp1−/− + miR-125aMock

0.0

0.5

1.0

1.5

2.0

2.5

Bcl

-xL

(a.u

.)M

cl-1

(a.

u.)

0.0

0.5

1.0

1.5

2.0

2.5 *

*

Live

/dea

d

Fig. 6. IRE1a mediates reduction in proapoptotic miR-125a. (A) BMDMs werecultured from Xbp1flox/flox ESR Cre+ (Xbp1Dor Cre− littermate (WT) mice in the presenceof tamoxifen. Volcano plot demonstratingthe distribution of miRNAs between WTand Xbp1D BMDMs measured using theNanoString nCounter assay. Data are fromoneexperimentwithquadruplicate samples(B) BMDMs were cultured from Xbp1flox/flo

ESR Cre+ (Xbp1D), Xbp1flox/flox Ern1 flox/flox ESRCre+ (Xbp1D Ire1aD), or Cre− littermate (WTmice in the presence of tamoxifen. The relative abundances of Bcl-xL, Mcl-1, and b-actinin the cell lysates were determined by Western blotting and densitometry. The ratio oBcl-xL orMcl-1 tob-actin is shown, normalizedto that in WT cells. Data are means ± SD othree independent experiments. a.u., arbitraryunits. (C and D) WT and Xbp1−/− MEFs weretransfected with negative control miRNA mimetic (miR-ctrl) or a miR-125a mimetic. Cellwere left untreated (mock) or treated withstaurosporine. Sevenhours later, caspase-3activity was assessed bymeasuring fluorometricsubstrate cleavage and is shown relative tothat in untreated WT cells (C). Twenty-fouhours after treatment, viability was assessedby measuring MTS reduction (D). Data aremeans ± SD of three replicates and are representative of two experiments. (E) The indicated miRNA-transfected MEFs were infectedwithVSV-GFP for 24hours. Cell deathwas thenassessed with a membrane-impermeantamine-reactive fluorescent dye, which wameasured by flow cytometry. The extent of infection was determined by measuring the relative abundance of GFP by flow cytometryData are from one experiment and are representative of two independent experiments*P < 0.01; **P < 0.001, unpaired t test.

6 of 11

Page 7: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

cause resistance to the intrinsic path-way of apoptosis (48, 49), althoughthe mechanism of this effect remainsunknown.

Consistent with previous reports(18), we detected IRE1a activationas indicated by spliced XBP1 mRNAin cells transiently transfected with aplasmid encoding NS4B (Fig. 7A).The abundance of miR-125a was de-creased (fig. S8E), and NS4B expres-sion induced the IRE1a-dependentresistance to staurosporine (Fig. 7, Band C). We infected Huh-7.5 humanhepatoma cells with trans-packagedHCV replicons encoding Gaussialuciferase (Gluc), which enabled usto quantitate HCV replication overtime (50). IRE1a inhibition alonewas not cytotoxic, but it sensitizedHCV-infected cells to death (Fig. 7D).Furthermore, inhibition of IRE1adecreased the secretion of virally en-coded luciferase, a marker of viralreplication (Fig. 7E). These findingssuggest that IRE1a activation duringHCV infectionmaypromote viral rep-lication by inhibiting the death of theinfected cells. To determine whetherthese findings extended to humanHCV infection, we quantified splicedXBP1mRNA in the human liver tissueof patients infected with HCV. WedetectedHCV-associated IRE1a acti-vation, as indicated by an increase inspliced XBP1 mRNA abundance inliver tissue from HCV-infected pa-tients compared to that in the livertissue of HCV-negative controls (Fig.7F). In addition to IRE1a activation,HCV-infected patients also exhibitedstatistically significantly reducedmiR-125a abundance (Fig. 7G), sug-gesting that IRE1a activation duringhuman HCV infection may conferresistance to apoptosis.

DISCUSSIONHere, we examined the IRE1a-XBP1branch of the UPR in innate antiviraldefense.We uncovered an unexpectedrole for Xbp1 and IRE1a in modulat-ing susceptibility to the intrinsic

pathway of apoptosis, which is induced during VSV andHSV infectionand plays a critical role in limiting viral replication. Xbp1−/−MEFs wereprotected from virally induced cell death and, as a consequence, sus-tained more viral replication despite an increased IFN response com-pared to that of WT cells. In Xbp1−/− cells with active IRE1a, the

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

abundance of the proapoptotic miRNA miR-125a was decreased,conferring resistance to the intrinsic apoptotic pathway. IRE1a activa-tion by HCV NS4B in WT cells also conferred resistance to apoptosisand promoted viral replication. Therefore, Xbp1-deficient cells withactive IRE1a gained apoptosis resistance, which suggests that IRE1a

0.01

0.1

1

10

100

0.1

1

10

100

1000

A

XBP1u

XBP1s

B

NS4B-GFP+ IRE1α inhibitor

Staurosporine

Mock

Mock Staurosporine

GFP vectorNS4B-GFPNS4B-GFP + IRE1α inhibitor %

Dea

d ce

lls*

C

0

20

40

60

80

100

120

0

20

40

60

80

100

120

Mock HCV 24 h 48 h

No inhibitorIRE1αinhibitor

**DE

% V

iabi

lity

Sec

rete

d lu

cife

rase

(%

no

inhi

bito

r)

****

GFP vector NS4B-GFP

Uninfected

F

HCV

Spliced XBP1

***

Rel

ativ

e m

RN

A a

bund

ance

Uninfected

G

HCV

miR-125a

***R

elat

ive

mic

roR

NA

abun

danc

e

Live

/dea

d

GFP**

0

20

40

60

80

100

Fig. 7. IRE1a-mediated apoptosis resistance induced by HCV NS4B. (A) HeLa cells were transfected with constructsexpressing GFP alone (vector) or together with HCV NS4B. RNA was isolated 72 hours later. XBP1 mRNA maturation fromthe unspliced (u) to spliced (s) form was analyzed by reverse transcription PCR (RT-PCR). Data are from one experimentand are representative of two independent experiments. (B and C) Transfected cells were treated with IRE1a inhibitor 2for 24 hours, and staurosporine was then added. Twenty hours later, cell death was assessed with a membrane-impermeant,amine-reactive fluorescent dye, which was measured by flow cytometry. Data are from one experiment and are representativeof three independent experiments (B). The percentage of dead cells among transfected GFP-positive cells was calculated (C).Data are means ± SD of three independent experiments. (D and E) Huh-7.5 cells were infected with trans-packaged HCVencoding luciferase. Forty-eight hours later, viability was determined by measuring cellular adenosine triphosphate (D).Secretion of virally encoded luciferase was measured 24 and 48 hours after infection (E). Data are means ± SD of threereplicates and are representative of two experiments. (F and G) RNA was isolated from the liver tissue of HCV-infectedpatients (n = 11) and HCV-negative controls (n = 6). Expression of spliced XBP1 and miR-125a (relative to an internalcontrol) was determined by quantitative RT-PCR. Data are means ± SEM. *P < 0.05; **P < 0.01, unpaired t test. ***P < 0.01,Mann-Whitney test.

7 of 11

Page 8: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

knockdown or inhibition is a model for the loss of apoptosis resistance.Finally, we observed IRE1a activation and substantially less miR-125a abundance in liver biopsies from HCV-infected patients, sug-gesting the in vivo relevance of the survival strategy used by HCV.These results highlight a previously unappreciated role of the IRE1a-XBP1 axis in the regulation of apoptosis and its consequences in viralsusceptibility.

Previous studies showed that after the engagement of TLRs andRLRs, XBP1 plays an important role in enhancing cytokine and IFNproduction in macrophages and dendritic cells (11–13). Given theobservation that transfection with poly(I:C) induced less IFN produc-tion in Xbp1−/− MEFs compared to that in WT MEFs, our results areconsistent with these previous findings that Xbp1 promotes RLRsignaling for IFN production in MEFs. However, despite this impair-ment in IFN production downstream of RLRs, infected Xbp1-deficientMEFs produced large amounts of IFN.We speculate that the enhancedIFN response observed in Xbp1−/− MEFs may result from both pro-longed cellular survival and an accumulation of viral PAMPs. Apoptoticcaspases can cleave and inactivate signaling proteins important for theIFN response, suggesting that the apoptotic process directly antagonizesthe IFN response (51–54). In addition, IRE1a can cleave host RNA forRLR stimulation (15). Therefore, our results highlight a distinct conse-quence of IRE1a activation, wherebyXbp1 deficiency results in a robustprosurvival response, leading to prolonged RLR stimulation that miti-gates impairment inRLR signaling to generate enhanced IFN responses.The prosurvival signals induced through IRE1a activation are so dom-inant that they overcome ISG-mediated antiviral functions and enablevirus replication.

In addition to protection from virus-induced death, we found thatXbp1 deficiency conferred resistance to the intrinsic pathway of apopto-sis stimulated by various chemical inducers. IRE1a activation inXbp1-deficient cells contributed to this apoptosis resistance becausesiRNA-mediated knockdown of Ire1a expression or inhibition ofIRE1a nuclease function rendered the Xbp1-deficient cells susceptibleto apoptosis. RIDD targets both coding and noncoding RNAs, includ-ing miRNAs (42). We found that IRE1a reduced the amount of miR-125a, leading to the enhanced expression of its target genes. The targetsinclude genes encoding prosurvivalmembers of the Bcl-2 family (44–46).miRNA controls target genes at the transcriptional and transla-tional levels (55). We found that the prosurvival proteins Bcl-xLand Mcl-1 were present in increased amounts. The biological targetsand functions of the other miRNAs identified in this study, namelymiR-1224 andmiR-804, have not yet beenwell described. The relevanceof these other miRNAs in IRE1a-dependent phenotypes will be inves-tigated in future studies.

Our study revealed an unexpected role for IRE1a in controlling ap-optosis. Many studies of the UPR have been performed with high con-centrationsof pharmacological inducers ofER stress, such as tunicamycinand thapsigargin, which inevitably lead to cell death. In these experimen-tal settings of irremediable ER stress, variousmechanisms have been pro-posed for inducing apoptosis (56). The net effect of IRE1a activity inpromoting cell death compared to cell survival has been controversial,with some studies suggesting that IRE1a promotes cell survival (57–59),and others suggesting that IRE1a promotes cell death (60–62). Specif-ically, IRE1a was proposed to induce apoptosis by degrading miRNAsthat lead to increased caspase-2 abundance (60), although there areconflicting data challenging this observation (63). IRE1a has beenlinked to apoptosis in cells irreversibly damaged by the activation ofthe JNK (c-Jun N-terminal kinase)–ASK1 (apoptosis signal–regulating

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

kinase 1) pathway (64, 65). IRE1a appears to have opposing roles inregulating apoptosis: prosurvival in the absence of irreversible UPRbut proapoptotic under irreversible UPR. This double-edged natureof IRE1amay be important to consider for the use of IRE1a inhibitorsin the treatment of various human diseases (66).

We speculate that IRE1amay represent an ancient form of cell pro-tection that has been subverted by some viruses for their own replicativeadvantage. HCV and other members of the Flaviviridae family induceER stress and activate IRE1a. Our results suggest that HCV NS4Binduces IRE1a-dependent protection from apoptosis, which may favorthe development of chronic infection and hepatocellular carcinoma. Inthe setting of avian coronavirus infection, IRE1a also promotes cell sur-vival in association with JNK and Akt regulation (67). Influenza virusinduces only the IRE1a arm of the UPR (not ATF6 or PERK), andinhibitors of IRE1a reduce viral replication (68). Therefore, our findingspredict that the IRE1a-RIDD pathway could be exploited as a noveltarget of intervention against viral infections.

MATERIALS AND METHODSCells and virusesXbp1+/+ andXbp1−/−MEFswere gifts fromL.H.Glimcher (Weill CornellMedical College, New York, NY). H1-HeLa cells stably overexpressingthe antiapoptotic protein BCL2 have been previously described (69).MEFs and H1-HeLa cells were propagated in high-glucose Dulbecco’smodified Eagle’s medium (DMEM) (Gibco) and supplemented with10% heat-inactivated fetal bovine serum (FBS), 1%Hepes, and penicillin(100 U/ml) and streptomycin (100 mg/ml) (Gibco). Huh-7.5 cells werepropagated in high-glucose DMEM with 10% heat-inactivated FBS and1mMnonessential amino acids (Invitrogen).Xbp1flox/flox (20) (a gift fromL. H. Glimcher,Weill Cornell Medical College), Ern1flox/flox (70) [RIKENBioResource Center (BRC), Japan], and CAGGCre-ERTM (the JacksonLaboratory) mice were bred in the Yale animal facility. All proceduresperformed in this study complied with the federal guidelines and in-stitutional policies set by Yale Animal Care and Use Committee.BMDMs were prepared from 6- to 12-week-old male and female miceaccording to a previously describedmethod (71) and cultured in 0.2 mM4-hydroxytamoxifen (Sigma) during days 2 to 4 of differentiation to in-duce Cre-mediated recombination. The following genotype combina-tions of BMDMs were treated with tamoxifen to generate the cellsdescribed in Fig. 6: Xbp1flox/flox × ESR Cre+ (XBP1D), Xbp1flox/flox ×Ern1flox/flox × ESR Cre+ (XBP1D IRE1aD), or Xbp1flox/flox × ESR Cre−

(WT). VSV-G–GFPwas a gift from J. Rose (YaleUniversity, NewHaven,CT) and A. Geballe (Fred Hutchinson Cancer Research Center, Seattle,WA) and was maintained and titered in baby hamster kidney cells.HSV-1–GFP (72) was a gift fromP.Desai and S. Person (JohnsHopkinsUniversity, Baltimore, MD) and was maintained and titered in Verocells. VSV-G pseudotyped lentivirus was made by harvesting culturesupernatants of human embryonic kidney 293T cells transfected withplasmids encoding VSV-G, GFP, and Gag-Pol. Trans-packaged HCVreplicons were prepared by transfecting the JFH/Gluc replicon intoHuh-7.5[core-NS2] cells as previously described (50)

Infection and stimulation of cellsCells were incubated for 1 hour with VSV or HSV-1 in serum-free me-dium or pseudotyped virus in phosphate-buffered saline 0.1% bovineserum albumin, and then the inoculum was removed, and incubationcontinued in complete medium. Cells were incubated for 4 hourswith trans-packaged HCV, and then the inoculum was removed, and

8 of 11

Page 9: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

incubation continued in complete medium containing 60 mM IRE1Inhibitor II (Calbiochem). zVAD (Invivogen) was added at 20 to 100 mMafter removal of the viral inoculum. Poly(I:C) (1 mg/ml) was deliveredcomplexed to Lipofectamine 2000 (Invitrogen). Cells were treated with0.1 to 1 mM staurosporine (Enzo Life Sciences), 1 mM gliotoxin (Sig-ma), 10 mMABT-737 (Santa Cruz Biotechnology), TNF (50 ng/ml) +cycloheximide (0.1 mg/ml) (Abcam), antimouse CD95 (2 mg/ml) (BDPharmingen), Fas-activating antibody + cycloheximide (0.1 mg/ml),tunicamycin (10 to 100mg/ml) (Sigma), 1mMthapsigargin (Calbiochem),or cycloheximide (100 mg/ml) (high-dose CHX). Cells were treated withthe IRE1 inhibitor 4m8C (Calbiochem) or the structurally similar com-pound AMC (Sigma) at 25 mM for 3 days before infection or apoptosisinduction or 40 mM IRE1 Inhibitor II (Calbiochem) for 24 hours beforeapoptosis induction.

PlasmidspFLAG.XBP1u.CMV2 (Addgene plasmid #21832, from D. Ron) (73),the empty vector control c-Flag pcDNA3 (Addgeneplasmid#20011, fromS. Smale) (74), hIRE1a.pcD(Addgeneplasmid#21892, fromR.Kaufman),and hIRE1a wt (Addgene plasmid #20744, from F. Urano) (75) wereused. MEFs were transfected using TransIT-2020 (Mirus Bio). HCVNS4Bwas expressedwith an authenticN-terminal Ala residue by fusinga human ubiquitin gene to a codon-optimizedNS4B gene (76): Ubiquitinwas amplified by using primers YO-0905 (TTA ATT AAC GAG GATCCC GCC ACC ATG CAG ATC TTC GTG AAG AC) and YO-0928(TCG ATC AGG GCT GCT CTG CTG GCT CCA CCG CGG AGACGC AGC ACC); codon-optimized NS4B was amplified by usingprimers YO-0927 (GGT GCT GCG TCT CCG CGG TGG AGC CAGCAG AGC AGC CCT GAT CGA) and YO-0931 (GTT TAA ACTTAA CAA GGG ATG GGG CAG TCC T). Ubi-NS4B was thenassembled in secondary PCRs with primers YO-0905 and YO-0931,cloned into pCR2.1 (Invitrogen) for sequencing, and then subclonedinto pIRES2-EGFP (Clontech) by using the common Sac I and Pst Irestriction sites. H1-HeLa cells were transfected using TransIT-HeLa(Mirus Bio).

Expression analysisRNA isolated using the RNeasy kit (Qiagen) was used to synthesizecomplementary DNA (cDNA) using the iScript cDNA Synthesis Kit(Bio-Rad), and quantitative PCR was performed on a StratageneMx3000P or Bio-Rad CFX Connect using SYBR Green (Bio-Rad) withthe following primers (all primers listed in the 5′ to 3′ orientation):Ifna4, CTG CTA CTT GGA ATG CAA CTC (forward) and CAGTCT TGC CAG CAA GTT GG (reverse); Ifnb1, GCA CTG GGTGGA ATG AGA CTA TTG (forward) and TTC TGA GGC ATCAAC TGA CAG GTC (reverse); Mx1, AGT CCT TTC CAC AGGCAG AA (forward) and CAT TGA GAG AAA CTC ACC TAA GAAC (reverse); Xbp1s, GAG TCC GCA GCA GGT (forward) and GTGTCA GAG TCC ATG GGA (reverse); Hspa5 (Bip), TCA TCG GACGCA CTT GGA (forward) and CAA CCA CCT TGA ATG GCAAGA (reverse); Chop, GTC CCT AGC TTGGCT GACAGA (forward)and TGGAGAGCGAGGGCTTTG (reverse); Blos1, CAAGGAGCTGCA GGA GAA GA (forward) and GCC TGG TTG AAG TTC TCCAC(reverse);Pdgfrb, AACCCCCTTACAGCTGTCCT (forward) andTAATCCCGTCAGCATCTTCC (reverse); andhuman splicedXBP1,TGC TGA GTC CGC AGC AGG TG (forward) and GCT GGC AGGCTC TGG GGA AG (reverse). For analysis of Xbp1-splicing, primersflanking the spliced sequences in Xbp1 mRNA [ACA CGC TTG GGAATG GAC AC (forward) and CCA TGG GAA GAT GTT CTG GG

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

(reverse)] were used for PCR amplification, and products were separatedby electrophoresis through a 3% agarose gel and visualized by ethidiumbromide staining.

miRNA expression analysisRNA isolated using the miRNeasy Mini Kit (Qiagen) was subjected tomiRNA profiling using the nCounter mouse miRNA expression assayversion 1.5 (NanoString) according to the manufacturer’s protocol.Data were analyzed by using the nSolver software with normalizationto the geometric mean of the top 100 miRNAs as recommended by themanufacturer. Quantitative RT-PCR with the miRCURYUniversal RTmicroRNA qPCR system (Exiqon) was used to measure miR-125a-5p.Expression was calculated relative to the manufacturer’s suggested en-dogenous control (miR-103a-3p), with equivalent results also obtainedrelative to miR-16-5p.

Assessment of cell deathCells were stained with the LIVE/DEAD Fixable Far Red Dead CellStain Kit (Molecular Probes) and analyzed by flow cytometry on aBD FACSCalibur or BD LSRFortessa. Viability was also assessed bymeasuring MTS reduction using the CellTiter 96 AQueous One Solu-tion Cell Proliferation Assay (Promega). Cells were stained with theCaspase-3, Active Form, Apoptosis Kit (BD Pharmingen) and analyzedby flow cytometry on a BD FACSCalibur. Caspase-3/7 activity wasmeasured using the SensoLyteHomogeneousRh110Caspase-3/7AssayKit (AnaSpec).

Western blottingCell pellets were lysed in SDS sample buffer (Cell Signaling Technol-ogy). Proteinswere separated by SDS–polyacrylamide gel electrophoresis,transferred to polyvinylidene difluoride membranes, and incubated withantibodies against Mcl-1 (BioLegend, 613601), Bcl-xL (CellSignaling Technology, 54H6), or b-actin (Cell Signaling Technology,13E5).

Intracellular stainingCells were fixed in Cytofix/Cytoperm solution (BD) on ice, washedwithPerm/Wash buffer (BD), and stained withDyLight 550–conjugated an-tibody against Beclin 1 (Novus Biologicals, NB110-87318R) or an equalconcentration of isotype control antibody. Washed cells were analyzedby flow cytometry on a BD LSRFortessa flow cytometer.

siRNA, miR mimetics, and inhibitorsGene-specific siGENOME siRNA or siGENOME Non-TargetingsiRNA #4 (which targets firefly luciferasemRNA and has at least fourmismatches to all mouse genes) obtained fromDharmacon (ThermoFisher Scientific) was delivered complexed to Lipofectamine RNAiMAX(Invitrogen). After incubation for 48 hours, cells were replated at equaldensity before infection or apoptosis induction. miR mimetics, miRI-DIANmicroRNAhairpin inhibitor negative control #1, andmiRIDIANmiR-125a hairpin inhibitor were obtained from Dharmacon (ThermoFisher Scientific) and delivered complexed to Lipofectamine RNAiMAX(Invitrogen).

Luciferase assayConditioned cell culture medium was collected at 24 or 48 hours afterinfection, clarified by centrifugation, mixed with 1/4 volume 5× lysisbuffer (New England Biolabs), and assayed for luciferase activity(New England Biolabs).

9 of 11

Page 10: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

Liver specimensLiver samples from percutaneous biopsies of liver transplant recipientschronically infected with HCV or from HCV-negative control livertransplant recipients were obtained with the approval of the Universityof Washington Institutional Review Board. Tissue was archived inRNAlater and stored at −80°C. RNA was isolated using the miRNeasyMini Kit (Qiagen).

Statistical analysesSample size was chosen according to previous experience in similarexperiments; all samples were included in analysis. The unpairedStudent’s t test or the Mann-Whitney test was used for comparisonsbetween two groups. P values of less than 0.05 were considered statisti-cally significant.

http://sD

ownloaded from

SUPPLEMENTARY MATERIALSwww.sciencesignaling.org/cgi/content/full/10/482/eaai7814/DC1Fig. S1. Increased IFN and ISG expression during VSV infection of Xbp1-deficient MEFs.Fig. S2. Knockdown of Xbp1 with siRNA mimics Xbp1 deficiency, and reconstitution with Xbp1reverses resistance to cell death.Fig. S3. Xbp1 deficiency does not increase susceptibility to a noncytotoxic virus and mimicsBCL2 overexpression.Fig. S4. Resistance to virally induced apoptosis in Xpb1-deficient cells is independent of Beclin 1.Fig. S5. Death of VSV- and HSV-infected cells does not require Chop.Fig. S6. VSV infection does not activate the UPR.Fig. S7. The resistance of Xbp1-deficient cells to apoptosis results from the activation of IRE1a.Fig. S8. The miRNA miR-125a regulates resistance to apoptosis.

on January 30, 2021tke.sciencem

ag.org/

REFERENCES AND NOTES1. M. R. Thompson, J. J. Kaminski, E. A. Kurt-Jones, K. A. Fitzgerald, Pattern recognition

receptors and the innate immune response to viral infection. Viruses 3, 920–940 (2011).2. A. Pichlmair, C. Reis e Sousa, Innate recognition of viruses. Immunity 27, 370–383 (2007).3. W. M. Schneider, M. D. Chevillotte, C. M. Rice, Interferon-stimulated genes: A complex

web of host defenses. Annu. Rev. Immunol. 32, 513–545 (2014).4. L. Zhang, A. Wang, Virus-induced ER stress and the unfolded protein response. Front.

Plant Sci. 3, 293 (2012).5. D. Ron, P. Walter, Signal integration in the endoplasmic reticulum unfolded protein

response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).6. M. Schröder, R. J. Kaufman, The mammalian unfolded protein response. Annu. Rev.

Biochem. 74, 739–789 (2005).7. P. Walter, D. Ron, The unfolded protein response: From stress pathway to homeostatic

regulation. Science 334, 1081–1086 (2011).8. C. Hetz, F. Martinon, D. Rodriguez, L. H. Glimcher, The unfolded protein response:

Integrating stress signals through the stress sensor IRE1a. Physiol. Rev. 91, 1219–1243(2011).

9. J. Hollien, J. H. Lin, H. Li, N. Stevens, P. Walter, J. S. Weissman, Regulated Ire1-dependentdecay of messenger RNAs in mammalian cells. J. Cell Biol. 186, 323–331 (2009).

10. J. Hollien, J. S. Weissman, Decay of endoplasmic reticulum-localized mRNAs during theunfolded protein response. Science 313, 104–107 (2006).

11. F. Martinon, X. Chen, A.-H. Lee, L. H. Glimcher, TLR activation of the transcriptionfactor XBP1 regulates innate immune responses in macrophages. Nat. Immunol. 11,411–418 (2010).

12. F. Hu, X. Yu, H. Wang, D. Zuo, C. Guo, H. Yi, B. Tirosh, J. R. Subjeck, X. Qiu, X.-Y. Wang,ER stress and its regulator X-box-binding protein-1 enhance polyIC-induced innateimmune response in dendritic cells. Eur. J. Immunol. 41, 1086–1097 (2011).

13. J. A. Smith, M. J. Turner, M. L. DeLay, E. I. Klenk, D. P. Sowders, R. A. Colbert, Endoplasmicreticulum stress and the unfolded protein response are linked to synergistic IFN-binduction via X-box binding protein 1. Eur. J. Immunol. 38, 1194–1203 (2008).

14. J. A. Cho, A.-H. Lee, B. Platzer, B. C. S. Cross, B. M. Gardner, H. De Luca, P. Luong,H. P. Harding, L. H. Glimcher, P. Walter, E. Fiebiger, D. Ron, J. C. Kagan, W. I. Lencer, Theunfolded protein response element IRE1a senses bacterial proteins invading the ER toactivate RIG-I and innate immune signaling. Cell Host Microbe 13, 558–569 (2013).

15. S. C. Eckard, G. I. Rice, A. Fabre, C. Badens, E. E. Gray, J. L. Hartley, Y. J. Crow, D. B. Stetson,The SKIV2L RNA exosome limits activation of the RIG-I-like receptors. Nat. Immunol. 15,839–845 (2014).

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

16. K. Y. Hur, J.-S. So, V. Ruda, M. Frank-Kamenetsky, K. Fitzgerald, V. Koteliansky,T. Iwawaki, L. H. Glimcher, A.-H. Lee, IRE1a activation protects mice againstacetaminophen-induced hepatotoxicity. J. Exp. Med. 209, 307–318 (2012).

17. A. Kaser, A.-H. Lee, A. Franke, J. N. Glickman, S. Zeissig, H. Tilg, E. E. Nieuwenhuis,D. E. Higgins, S. Schreiber, L. H. Glimcher, R. S. Blumberg, XBP1 links ER stress tointestinal inflammation and confers genetic risk for human inflammatory boweldisease. Cell 134, 743–756 (2008).

18. Y. Zheng, B. Gao, L. Ye, L. Kong, W. Jing, X. Yang, Z. Wu, L. Ye, Hepatitis C virusnon-structural protein NS4B can modulate an unfolded protein response. J. Microbiol.43, 529–536 (2005).

19. A. M. Reimold, A. Etkin, I. Clauss, A. Perkins, D. S. Friend, J. Zhang, H. F. Horton, A. Scott,S. H. Orkin, M. C. Byrne, M. J. Grusby, L. H. Glimcher, An essential role in liverdevelopment for transcription factor XBP-1. Genes Dev. 14, 152–157 (2000).

20. C. Hetz, A.-H. Lee, D. Gonzalez-Romero, P. Thielen, J. Castilla, C. Soto, L. H. Glimcher,Unfolded protein response transcription factor XBP-1 does not influence prionreplication or pathogenesis. Proc. Natl. Acad. Sci. U.S.A. 105, 757–762 (2008).

21. J. A. Hobbs, R. H. Schloemer, G. Hommel-Berrey, Z. Brahmi, Caspase-3-like proteases areactivated by infection but are not required for replication of vesicular stomatitis virus.Virus Res. 80, 53–65 (2001).

22. J. A. Hobbs, G. Hommel-Berrey, Z. Brahmi, Requirement of caspase-3 for efficientapoptosis induction and caspase-7 activation but not viral replication or cell rounding incells infected with vesicular stomatitis virus. Hum. Immunol. 64, 82–92 (2003).

23. B. J. Thomson, Viruses and apoptosis. Int. J. Exp. Pathol. 82, 65–76 (2001).24. M. Høyer-Hansen, M. Jäättelä, Connecting endoplasmic reticulum stress to autophagy by

unfolded protein response and calcium. Cell Death Differ. 14, 1576–1582 (2007).25. A. Margariti, H. Li, T. Chen, D. Martin, G. Vizcay-Barrena, S. Alam, E. Karamariti, Q. Xiao,

A. Zampetaki, Z. Zhang, W. Wang, Z. Jiang, C. Gao, B. Ma, Y.-G. Chen, G. Cockerill, Y. Hu,Q. Xu, L. Zeng, XBP1 mRNA splicing triggers an autophagic response in endothelial cellsthrough BECLIN-1 transcriptional activation. J. Biol. Chem. 288, 859–872 (2013).

26. A.-H. Lee, N. N. Iwakoshi, L. H. Glimcher, XBP-1 regulates a subset of endoplasmicreticulum resident chaperone genes in the unfolded protein response. Mol. Cell. Biol. 23,7448–7459 (2003).

27. D. Baltzis, L.-K. Qu, S. Papadopoulou, J. D. Blais, J. C. Bell, N. Sonenberg, A. E. Koromilas,Resistance to vesicular stomatitis virus infection requires a functional cross talkbetween the eukaryotic translation initiation factor 2a kinases PERK and PKR.J. Virol. 78, 12747–12761 (2004).

28. H. F. Burnett, T. E. Audas, G. Liang, R. R. Lu, Herpes simplex virus-1 disarms the unfoldedprotein response in the early stages of infection. Cell Stress Chaperones 17, 473–483(2012).

29. M. Mulvey, C. Arias, I. Mohr, Maintenance of endoplasmic reticulum (ER) homeostasisin herpes simplex virus type 1-infected cells through the association of a viralglycoprotein with PERK, a cellular ER stress sensor. J. Virol. 81, 3377–3390 (2007).

30. A. Ganguly, S. Basu, P. Chakraborty, S. Chatterjee, A. Sarkar, M. Chatterjee,S. K. Choudhuri, Targeting mitochondrial cell death pathway to overcome drugresistance with a newly developed iron chelate. PLOS ONE 5, e11253 (2010).

31. A. T. Ho, Q. H. Li, R. Hakem, T. W. Mak, E. Zacksenhaus, Coupling of caspase-9 to Apaf1 inresponse to loss of pRb or cytotoxic drugs is cell-type-specific. EMBO J. 23, 460–472(2004).

32. V.-T. Nguyen, J. S. Lee, Z.-J. Qian, Y.-X. Li, K.-N. Kim, S.-J. Heo, Y.-J. Jeon, W. S. Park,I.-W. Choi, J.-Y. Je, W.-K. Jung, Gliotoxin isolated from marine fungus Aspergillus sp.induces apoptosis of human cervical cancer and chondrosarcoma cells. Mar. Drugs 12,69–87 (2014).

33. J. Pardo, C. Urban, E. M. Galvez, P. G. Ekert, U. Müller, J. Kwon-Chung, M. Lobigs,A. Mullbacher, R. Wallich, C. Borner, M. M. Simon, The mitochondrial protein Bak is pivotalfor gliotoxin-induced apoptosis and a critical host factor of Aspergillus fumigatusvirulence in mice. J. Cell Biol. 174, 509–519 (2006).

34. E. E. Varfolomeev, M. Schuchmann, V. Luria, N. Chiannilkulchai, J. S. Beckmann, I. L. Mett,D. Rebrikov, V. M. Brodianski, O. C. Kemper, O. Kollet, T. Lapidot, D. Soffer, T. Sobe,K. B. Avraham, T. Goncharov, H. Holtmann, P. Lonai, D. Wallach, Targeted disruption ofthe mouse Caspase 8 gene ablates cell death induction by the TNF receptors,Fas/Apo1, and DR3 and is lethal prenatally. Immunity 9, 267–276 (1998).

35. A. E. Byrd, I. V. Aragon, J. W. Brewer, MicroRNA-30c-2* limits expression of proadaptivefactor XBP1 in the unfolded protein response. J. Cell Biol. 196, 689–698 (2012).

36. L. Romero-Ramirez, H. Cao, D. Nelson, E. Hammond, A.-H. Lee, H. Yoshida, K. Mori,L. H. Glimcher, N. C. Denko, A. J. Giaccia, Q.-T. Le, A. C. Koong, XBP1 is essentialfor survival under hypoxic conditions and is required for tumor growth.Cancer Res. 64, 5943–5947 (2004).

37. A.-H. Lee, N. N. Iwakoshi, K. C. Anderson, L. H. Glimcher, Proteasome inhibitors disruptthe unfolded protein response in myeloma cells. Proc. Natl. Acad. Sci. U.S.A. 100,9946–9951 (2003).

38. T. Oltersdorf, S. W. Elmore, A. R. Shoemaker, R. C. Armstrong, D. J. Augeri, B. A. Belli,M. Bruncko, T. L. Deckwerth, J. Dinges, P. J. Hajduk, M. K. Joseph, S. Kitada, S. J. Korsmeyer,

10 of 11

Page 11: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

SC I ENCE S I GNAL ING | R E S EARCH ART I C L E

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from

A. R. Kunzer, A. Letai, C. Li, M. J. Mitten, D. G. Nettesheim, S. Ng, P. M. Nimmer,J. M. O’Connor, A. Oleksijew, A. M. Petros, J. C. Reed, W. Shen, S. K. Tahir, C. B. Thompson,K. J. Tomaselli, B. Wang, M. D. Wendt, H. Zhang, S. W. Fesik, S. H. Rosenberg, Aninhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435,677–681 (2005).

39. J. R. Cubillos-Ruiz, P. C. Silberman, M. R. Rutkowski, S. Chopra, A. Perales-Puchalt, M. Song,S. Zhang, S. E. Bettigole, D. Gupta, K. Holcomb, L. H. Ellenson, T. Caputo, A.-H. Lee,J. R. Conejo-Garcia, L. H. Glimcher, ER stress sensor XBP1 controls anti-tumor immunity bydisrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

40. B. C. Cross, P. J. Bond, P. G. Sadowski, B. K. Jha, J. Zak, J. M. Goodman, R. H. Silverman,T. A. Neubert, I. R. Baxendale, D. Ron, H. P. Harding, The molecular basis for selectiveinhibition of unconventional mRNA splicing by an IRE1-binding small molecule.Proc. Natl. Acad. Sci. U.S.A. 109, E869–E878 (2012).

41. D. Acosta-Alvear, Y. Zhou, A. Blais, M. Tsikitis, N. H. Lents, C. Arias, C. J. Lennon, Y. Kluger,B. D. Dynlacht, XBP1 controls diverse cell type- and condition-specific transcriptionalregulatory networks. Mol. Cell 27, 53–66 (2007).

42. D. S. Coelho, P. M. Domingos, Physiological roles of regulated Ire1 dependent decay.Front. Genet. 5, 76 (2014).

43. M. Maurel, E. Chevet, Endoplasmic reticulum stress signaling: The microRNA connection.Am. J. Physiol. Cell Physiol. 304, C1117–C1126 (2013).

44. Z. Tong, N. Liu, L. Lin, X. Guo, D. Yang, Q. Zhang, miR-125a-5p inhibits cell proliferationand induces apoptosis in colon cancer via targeting BCL2, BCL2L12 and MCL1.Biomed. Pharmacother. 75, 129–136 (2015).

45. D. Svensson, O. Gidlöf, K. M. Turczyńska, D. Erlinge, S. Albinsson, B.-O. Nilsson,Inhibition of microRNA-125a promotes human endothelial cell proliferation andviability through an antiapoptotic mechanism. J. Vasc. Res. 51, 239–245 (2014).

46. A. Balakrishnan, A. T. Stearns, P. J. Park, J. M. Dreyfuss, S. W. Ashley, D. B. Rhoads,A. Tavakkolizadeh, Upregulation of proapoptotic microRNA mir-125a after massive smallbowel resection in rats. Ann. Surg. 255, 747–753 (2012).

47. D. Sir, W.-l. Chen, J. Choi, T. Wakita, T. S. B. Yen, J.-h. Ou, Induction of incompleteautophagic response by hepatitis C virus via the unfolded protein response. Hepatology48, 1054–1061 (2008).

48. K. Machida, K. Tsukiyama-Kohara, E. Seike, S. Tone, F. Shibasaki, M. Shimizu, H. Takahashi,Y. Hayashi, N. Funata, C. Taya, H. Yonekawa, M. Kohara, Inhibition of cytochrome c releasein Fas-mediated signaling pathway in transgenic mice induced to express hepatitis Cviral proteins. J. Biol. Chem. 276, 12140–12146 (2001).

49. Y. Kamegaya, Y. Hiasa, L. Zukerberg, N. Fowler, J. T. Blackard, W. Lin, W. H. Choe,E. V. Schmidt, R. T. Chung, Hepatitis C virus acts as a tumor accelerator by blockingapoptosis in a mouse model of hepatocarcinogenesis. Hepatology 41, 660–667 (2005).

50. T. Phan, A. Kohlway, P. Dimberu, A. M. Pyle, B. D. Lindenbach, The acidic domain ofhepatitis C virus NS4A contributes to RNA replication and virus particle assembly. J. Virol.85, 1193–1204 (2011).

51. M. Rebsamen, E. Meylan, J. Curran, J. Tschopp, The antiviral adaptor proteins Cardif andTrif are processed and inactivated by caspases. Cell Death Differ. 15, 1804–1811 (2008).

52. I. Scott, K. L. Norris, The mitochondrial antiviral signaling protein, MAVS, is cleavedduring apoptosis. Biochem. Biophys. Res. Commun. 375, 101–106 (2008).

53. M. J. White, K. McArthur, D. Metcalf, R. M. Lane, J. C. Cambier, M. J. Herold, M. F. van Delft,S. Bedoui, G. Lessene, M. E. Ritchie, D. C. S. Huang, B. T. Kile, Apoptotic caspases suppressmtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 (2014).

54. A. Rongvaux, R. Jackson, C. C. D. Harman, T. Li, A. P. West, M. R. de Zoete, Y. Wu, B. Yordy,S. A. Lakhani, C.-Y. Kuan, T. Taniguchi, G. S. Shadel, Z. J. Chen, A. Iwasaki, R. A. Flavell,Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA.Cell 159, 1563–1577 (2014).

55. D. P. Bartel, MicroRNAs: Target recognition and regulatory functions. Cell 136, 215–233(2009).

56. Y. Chen, F. Brandizzi, IRE1: ER stress sensor and cell fate executor. Trends Cell Biol. 23,547–555 (2013).

57. J. H. Lin, H. Li, D. Yasumura, H. R. Cohen, C. Zhang, B. Panning, K. M. Shokat, M. M. LaVail,P. Walter, IRE1 signaling affects cell fate during the unfolded protein response.Science 318, 944–949 (2007).

58. J. H. Lin, H. Li, Y. Zhang, D. Ron, P. Walter, Divergent effects of PERK and IRE1 signaling oncell viability. PLOS ONE 4, e4170 (2009).

59. M. Lu, D. A. Lawrence, S. Marsters, D. Acosta-Alvear, P. Kimmig, A. S. Mendez, A. W. Paton,J. C. Paton, P. Walter, A. Ashkenazi, Opposing unfolded-protein-response signalsconverge on death receptor 5 to control apoptosis. Science 345, 98–101 (2014).

60. J.-P. Upton, L. Wang, D. Han, E. S. Wang, N. E. Huskey, L. Lim, M. Truitt, M. T. McManus,D. Ruggero, A. Goga, F. R. Papa, S. A. Oakes, IRE1a cleaves select microRNAs during ERstress to derepress translation of proapoptotic Caspase-2. Science 338, 818–822(2012).

61. D. Han, A. G. Lerner, L. Vande Walle, J.-P. Upton, W. Xu, A. Hagen, B. J. Backes, S. A. Oakes,F. R. Papa, IRE1a kinase activation modes control alternate endoribonucleaseoutputs to determine divergent cell fates. Cell 138, 562–575 (2009).

Fink et al., Sci. Signal. 10, eaai7814 (2017) 6 June 2017

62. A. G. Lerner, J.-P. Upton, P. V. K. Praveen, R. Ghosh, Y. Nakagawa, A. Igbaria, S. Shen,V. Nguyen, B. J. Backes, M. Heiman, N. Heintz, P. Greengard, S. Hui, Q. Tang, A. Trusina,S. A. Oakes, F. R. Papa, IRE1a induces thioredoxin-interacting protein to activate theNLRP3 inflammasome and promote programmed cell death under irremediable ERstress. Cell Metab. 16, 250–264 (2012).

63. J. J. Sandow, L. Dorstyn, L. A. O’Reilly, M. Tailler, S. Kumar, A. Strasser, P. G. Ekert,ER stress does not cause upregulation and activation of caspase-2 to initiateapoptosis. Cell Death Differ. 21, 475–480 (2014).

64. H. Nishitoh, A. Matsuzawa, K. Tobiume, K. Saegusa, K. Takeda, K. Inoue, S. Hori,A. Kakizuka, H. Ichijo, ASK1 is essential for endoplasmic reticulum stress-inducedneuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 16,1345–1355 (2002).

65. F. Urano, X. Wang, A. Bertolotti, Y. Zhang, P. Chung, H. P. Harding, D. Ron, Couplingof stress in the ER to activation of JNK protein kinases by transmembrane proteinkinase IRE1. Science 287, 664–666 (2000).

66. C. Hetz, E. Chevet, H. P. Harding, Targeting the unfolded protein response in disease.Nat. Rev. Drug Discov. 12, 703–719 (2013).

67. T. S. Fung, Y. Liao, D. X. Liu, The endoplasmic reticulum stress sensor IRE1a protects cellsfrom apoptosis induced by the coronavirus infectious bronchitis virus. J. Virol. 88,12752–12764 (2014).

68. I. H. Hassan, M. S. Zhang, L. S. Powers, J. Q. Shao, J. Baltrusaitis, D. T. Rutkowski, K. Legge,M. M. Monick, Influenza A viral replication is blocked by inhibition of the inositol-requiring enzyme 1 (IRE1) stress pathway. J. Biol. Chem. 287, 4679–4689 (2012).

69. E. F. Foxman, J. A. Storer, K. Vanaja, A. Levchenko, A. Iwasaki, Two interferon-independentdouble-stranded RNA-induced host defense strategies suppress the common coldvirus at warm temperature. Proc. Natl. Acad. Sci. U.S.A. 113, 8496–8501 (2016).

70. T. Iwawaki, R. Akai, S. Yamanaka, K. Kohno, Function of IRE1 a in the placenta is essentialfor placental development and embryonic viability. Proc. Natl. Acad. Sci. U.S.A. 106,16657–16662 (2009).

71. T. Ichinohe, H. K. Lee, Y. Ogura, R. Flavell, A. Iwasaki, Inflammasome recognition ofinfluenza virus is essential for adaptive immune responses. J. Exp. Med. 206, 79–87 (2009).

72. P. Desai, S. Person, Incorporation of the green fluorescent protein into the herpes simplexvirus type 1 capsid. J. Virol. 72, 7563–7568 (1998).

73. M. Calfon, H. Zeng, F. Urano, J. H. Till, S. R. Hubbard, H. P. Harding, S. G. Clark, D. Ron, IRE1couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415, 92–96 (2002).

74. S. Sanjabi, K. J. Williams, S. Saccani, L. Zhou, A. Hoffmann, G. Ghosh, S. Gerondakis,G. Natoli, S. T. Smale, A c-Rel subdomain responsible for enhanced DNA-binding affinityand selective gene activation. Genes Dev. 19, 2138–2151 (2005).

75. K. L. Lipson, R. Ghosh, F. Urano, The role of IRE1a in the degradation of insulin mRNA inpancreatic b-cells. PLOS ONE 3, e1648 (2008).

76. T. Kazakov, F. Yang, H. N. Ramanathan, A. Kohlway, M. S. Diamond, B. D. Lindenbach,Hepatitis C virus RNA replication depends on specific cis- and trans-acting activities ofviral nonstructural proteins. PLOS Pathog. 11, e1004817 (2015).

Acknowledgments: We thank H. Dong and K. Hayashi for technical support; B. Cookson andN. Kaminski for sharing laboratory facilities and equipment; and L. Glimcher, J. Rose, A. Geballe,P. Desai, S. Person, D. Ron, S. Smale, R. Kaufman, and F. Urano for providing us withvarious reagents. Funding: This work was supported by funding from the Howard HughesMedical Institute and NIH R01 AI054359 and AI064705 (to A.I.) and K08 AI119142 (to S.L.F.).S.L.F. was supported by NIH award T32 HL007974. B.D.L. was funded by R01 AI087925.Author contributions: S.L.F., T.R.J., R.D.M., T.I., C.S.L., B.D.L., and A.I. designed the experiments.S.L.F., T.R.J., R.D.M., and B.D.L. performed the experiments. S.L.F., T.R.J., R.D.M., and A.I.analyzed the data. S.L.F and A.I. wrote the manuscript. Competing interests: The authorsdeclare that they have no competing interests. Data and materials availability: The Xbp1+/+

and Xbp1−/− MEFs and Xbp1flox/flox mice require a materials transfer agreement (MTA) fromHarvard University. The Ern1flox/flox mice require an MTA from RIKEN BRC. The followingplasmids require an MTA from Addgene: pFLAG.XBP1u.CMV2, c-Flag pcDNA3, hIRE1a.pcD, andhIRE1a wt. The JFH-1 HCV replicon requires MTAs from Rockefeller University and ApathLLC (in the United States) or from the Tokyo Metropolitan Organization for Medical Researchand Toray Industries (elsewhere). The replicon was packaged by using the HCV strain J6structural genes, which were obtained under an MTA with the NIH. The Huh-7.5 cell linerequires an MTA from Washington University and Apath LLC.

Submitted 11 August 2016Accepted 19 May 2017Published 6 June 201710.1126/scisignal.aai7814

Citation: S. L. Fink, T. R. Jayewickreme, R. D. Molony, T. Iwawaki, C. S. Landis, B. D. Lindenbach,A. Iwasaki, IRE1a promotes viral infection by conferring resistance to apoptosis. Sci. Signal. 10,eaai7814 (2017).

11 of 11

Page 12: IRE1 promotes viral infection by conferring resistance to ...Similarly, Xbp1−/− MEFs also supported increased replication of HSV-1–GFP as indicated by measurement of GFP abundance

promotes viral infection by conferring resistance to apoptosisαIRE1

IwasakiSusan L. Fink, Teshika R. Jayewickreme, Ryan D. Molony, Takao Iwawaki, Charles S. Landis, Brett D. Lindenbach and Akiko

DOI: 10.1126/scisignal.aai7814 (482), eaai7814.10Sci. Signal. 

provide a potential therapeutic target. functions to enhance cell survival in response to viral infection and mayαTogether, these data suggest that IRE1

activation and decreased miR-125a abundance.αbiopsies from HCV-infected patients also showed increased IRE1cleaved the proapoptotic microRNA miR-125a, leading to decreased apoptosis and increased viral replication. Liver

, whichαcompromised in type I IFN production or responses. Instead, these cells showed increased activation of IRE1-deficient cells had defective antiviral responses to infection by hepatitis C virus (HCV); however, the cells were notXbp1

. found that et altranscription factor XBP1, which drives the expression of genes encoding type I interferons (IFNs). Fink generates theα mRNA by the nuclease IRE1Xbp1as that occurring during viral infection. During the UPR, processing of

The unfolded protein response (UPR) alleviates the cellular stress caused by the accumulation of proteins, suchResisting death by virus

ARTICLE TOOLS http://stke.sciencemag.org/content/10/482/eaai7814

MATERIALSSUPPLEMENTARY http://stke.sciencemag.org/content/suppl/2017/06/02/10.482.eaai7814.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/11/559/eaaw2150.fullhttp://stke.sciencemag.org/content/sigtrans/11/530/eaao4617.fullhttp://stm.sciencemag.org/content/scitransmed/7/282/282ra49.fullhttp://stke.sciencemag.org/content/sigtrans/10/486/eaao2378.fullhttp://science.sciencemag.org/content/sci/347/6228/1374.fullhttp://stke.sciencemag.org/content/sigtrans/10/470/eaal2323.fullhttp://stke.sciencemag.org/content/sigtrans/8/403/ra118.fullhttp://stke.sciencemag.org/content/sigtrans/9/458/ec295.abstract

REFERENCES

http://stke.sciencemag.org/content/10/482/eaai7814#BIBLThis article cites 76 articles, 30 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.Science SignalingYork Avenue NW, Washington, DC 20005. The title (ISSN 1937-9145) is published by the American Association for the Advancement of Science, 1200 NewScience Signaling

Copyright © 2017, American Association for the Advancement of Science

on January 30, 2021http://stke.sciencem

ag.org/D

ownloaded from