9
Hsp90 inhibitors reduce inuenza virus replication in cell culture Geoffrey Chase a, 1 , Tao Deng b,c, 1 , Ervin Fodor b , Bo Wah Leung b , Daniel Mayer a , Martin Schwemmle a, , George Brownlee b, a Department of Virology, Institute for Medical Microbiology and Hygiene, University of Freiburg, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany b Chemical Pathology Unit, Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, UK c College of Life Sciences, Nankai University, Tianjin, China abstract article info Article history: Received 12 March 2008 Returned to author for revision 15 April 2008 Accepted 30 April 2008 Keywords: Inuenza viruses replication RNA polymerase Hsp90 Geldanamycin 17 AAG Antiviral agents The viral RNA polymerase complex of inuenza A virus consists of three subunits PB1, PB2 and PA. Recently, the cellular chaperone Hsp90 was shown to play a role in nuclear import and assembly of the trimeric polymerase complex by binding to PB1 and PB2. Here we show that Hsp90 inhibitors, geldanamycin or its derivative 17-AAG, delay the growth of inuenza virus in cell culture resulting in a 12 log reduction in viral titre early in infection. We suggest that this is caused by the reduced half-life of PB1 and PB2 and inhibition of nuclear import of PB1 and PA which lead to reduction in viral RNP assembly. Hsp90 inhibitors may represent a new class of antiviral compounds against inuenza viruses. © 2008 Elsevier Inc. All rights reserved. Introduction The RNA-dependent RNA polymerase of the inuenza A virus (FluA) is a heterotrimeric complex consisting of the PB1, PB2, and PA subunits. The trimeric complex catalyzes viral RNA transcription and replication in the nucleus of infected cells. Recently, we (Deng et al., 2005; Mayer et al., 2007) and others (Kawaguchi and Nagata, 2007; Momose et al., 2002) have identied several cellular proteins that associate with the trimeric polymerase complex or its individual subunits. One of these proteins, heat shock protein 90 (Hsp90), was reported to bind to the PB1 and PB2 polymerase subunits (Naito et al., 2007). It has been suggested that Hsp90 is involved in the assembly and nuclear transport of viral RNA polymerase subunits, possibly as a molecular chaperone for the polymerase subunits prior to the formation of a mature ternary polymerase complex (Naito et al., 2007). Hsp90 is an atypical member of the family of heat shock-induced chaperones (Pearl and Prodromou, 2006; Whitesell and Lindquist, 2005). It binds to a specic set of client proteins, including intracellular receptors and signal transduction proteins, and facilitates their folding presumably through domain rearrangement (Pearl and Prodromou, 2006; Whitesell and Lindquist, 2005). This chaperone activity is dependent on various co-chaperones and ATP (Whitesell and Lindquist, 2005). Hsp90 activity can be inhibited by drugs which compete for its N-terminal ATP-binding site. These drugs include natural products (radicicol, geldanamycin) and semi-synthetic or synthetic compounds [e.g. 17-allylamino-demethoxygeldanamycin (17-AAG), a geldanamycin (GA) analogue] (Whitesell and Lindquist, 2005). In this study, we report that Hsp90 inhibitors GA and 17-AAG delay viral growth in cell culture. We observed a reduction of viral transcription and replication with GA treatment in a concentration dependent manner in virus infected cells. By studying the ribonu- cleoprotein complex (RNP) formation, nuclear transport and protein stability of polymerase subunits in transiently-transfected cells, we suggest that inhibition of virus growth was due to both a reduced half-life of PB1 and PB2 and an inhibition of nuclear import of PB1 and PA. This leads to a reduction in viral RNP assembly. This conclusion is consistent with the observation that reduced amounts of viral polymerase complex were detected with the GA treatment in the nucleus of infected cells. Results No cytopathic effects were observed with the Hsp90 inhibitors, GA and 17-AAG, at the concentrations used in this study We rst systematically analyzed the toxicity of both Hsp90 inhibitors (GA and 17-AAG) by the MTT toxicity assay (see Materials and methods) in the three different cell lines (MDCK, 293T and A549) Virology 377 (2008) 431439 Corresponding authors. G. Brownlee is to be contacted at Fax: +44 1865 275556. M. Schwemmle, Fax: +49 761 203 6639. E-mail addresses: [email protected] (M. Schwemmle), [email protected] (G. Brownlee). 1 Contributed equally. 0042-6822/$ see front matter © 2008 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2008.04.040 Contents lists available at ScienceDirect Virology journal homepage: www.elsevier.com/locate/yviro

Hsp90 inhibitors reduce influenza virus replication in cell culture

Embed Size (px)

Citation preview

Page 1: Hsp90 inhibitors reduce influenza virus replication in cell culture

Virology 377 (2008) 431–439

Contents lists available at ScienceDirect

Virology

j ourna l homepage: www.e lsev ie r.com/ locate /yv i ro

Hsp90 inhibitors reduce influenza virus replication in cell culture

Geoffrey Chase a,1, Tao Deng b,c,1, Ervin Fodor b, Bo Wah Leung b, Daniel Mayer a,Martin Schwemmle a,⁎, George Brownlee b,⁎a Department of Virology, Institute for Medical Microbiology and Hygiene, University of Freiburg, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germanyb Chemical Pathology Unit, Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, UKc College of Life Sciences, Nankai University, Tianjin, China

⁎ Corresponding authors. G. Brownlee is to be contaM. Schwemmle, Fax: +49 761 203 6639.

E-mail addresses: [email protected]@path.ox.ac.uk (G. Brownlee).

1 Contributed equally.

0042-6822/$ – see front matter © 2008 Elsevier Inc. Aldoi:10.1016/j.virol.2008.04.040

a b s t r a c t

a r t i c l e i n f o

Article history:

The viral RNA polymerase c Received 12 March 2008Returned to author for revision15 April 2008Accepted 30 April 2008

Keywords:Influenza viruses replicationRNA polymeraseHsp90Geldanamycin17 AAGAntiviral agents

omplex of influenza A virus consists of three subunits PB1, PB2 and PA. Recently,the cellular chaperone Hsp90 was shown to play a role in nuclear import and assembly of the trimericpolymerase complex by binding to PB1 and PB2. Here we show that Hsp90 inhibitors, geldanamycin or itsderivative 17-AAG, delay the growth of influenza virus in cell culture resulting in a 1–2 log reduction in viraltitre early in infection. We suggest that this is caused by the reduced half-life of PB1 and PB2 and inhibition ofnuclear import of PB1 and PA which lead to reduction in viral RNP assembly. Hsp90 inhibitors may representa new class of antiviral compounds against influenza viruses.

© 2008 Elsevier Inc. All rights reserved.

Introduction

TheRNA-dependent RNApolymerase of the influenzaAvirus (FluA)is a heterotrimeric complex consisting of the PB1, PB2, and PA subunits.The trimeric complex catalyzes viral RNA transcription and replicationin the nucleus of infected cells. Recently, we (Deng et al., 2005; Mayeret al., 2007) and others (Kawaguchi and Nagata, 2007; Momose et al.,2002) have identified several cellular proteins that associate with thetrimeric polymerase complex or its individual subunits. One of theseproteins, heat shock protein 90 (Hsp90), was reported to bind to thePB1 and PB2 polymerase subunits (Naito et al., 2007). It has beensuggested thatHsp90 is involved in the assembly andnuclear transportof viral RNA polymerase subunits, possibly as a molecular chaperonefor the polymerase subunits prior to the formation of a mature ternarypolymerase complex (Naito et al., 2007).

Hsp90 is an atypical member of the family of heat shock-inducedchaperones (Pearl and Prodromou, 2006; Whitesell and Lindquist,2005). It binds to a specific set of client proteins, including intracellularreceptors and signal transduction proteins, and facilitates their foldingpresumably through domain rearrangement (Pearl and Prodromou,2006; Whitesell and Lindquist, 2005). This chaperone activity is

cted at Fax: +44 1865 275556.

burg.de (M. Schwemmle),

l rights reserved.

dependent on various co-chaperones andATP (Whitesell and Lindquist,2005). Hsp90 activity can be inhibited by drugs which compete for itsN-terminal ATP-binding site. These drugs include natural products(radicicol, geldanamycin) and semi-synthetic or synthetic compounds[e.g.17-allylamino-demethoxygeldanamycin (17-AAG), a geldanamycin(GA) analogue] (Whitesell and Lindquist, 2005).

In this study, we report that Hsp90 inhibitors GA and 17-AAG delayviral growth in cell culture. We observed a reduction of viraltranscription and replication with GA treatment in a concentrationdependent manner in virus infected cells. By studying the ribonu-cleoprotein complex (RNP) formation, nuclear transport and proteinstability of polymerase subunits in transiently-transfected cells, wesuggest that inhibition of virus growth was due to both a reducedhalf-life of PB1 and PB2 and an inhibition of nuclear import of PB1 andPA. This leads to a reduction in viral RNP assembly. This conclusion isconsistent with the observation that reduced amounts of viralpolymerase complex were detected with the GA treatment in thenucleus of infected cells.

Results

No cytopathic effects were observed with the Hsp90 inhibitors, GA and17-AAG, at the concentrations used in this study

We first systematically analyzed the toxicity of both Hsp90inhibitors (GA and 17-AAG) by the MTT toxicity assay (see Materialsand methods) in the three different cell lines (MDCK, 293T and A549)

Page 2: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 1. Lack of toxicity of GA and 17-AAG. MDCK (A), 293T (B), or A549 (C) cells wereseeded in 96-well plates. Cells were then treated with the concentrations of inhibitorsor solvent, as shown, for 24 h, and metabolic activity was measured using an MTT kit asdescribed in Materials and methods. Hydrogen peroxide (H2O2) was used as a control toinduce cell death. The absorbance at 570 nm of DMSO-treated cells was arbitrarily set to100%. Values represent the mean of three independent experiments, and error barsshow the standard deviation of the mean.

432 G. Chase et al. / Virology 377 (2008) 431–439

which were used in this study. The MTT assay measures the metabolicactivity of cultured cells using a colorimetric substrate, which is awell-known cytotoxicity marker (Mosmann, 1983). As shown in Fig. 1,no significant toxicity was observed at concentrations of GA from 200to 800 nM and at concentrations of 17-AAG from 200 to 1000 nM in allthree cells lines. Therefore, we performed future experiments with GAat concentrations no higher than 800 nM, and with 17-AAG at con-centrations no higher than 1000 nM.

Hsp90 inhibitors delay viral growth

We next tested whether the Hsp90 inhibitors, GA and 17-AAG, wereable to block viral multiplication in cell culture. MDCK cells were infectedwith influenza A virus (A/WSN/33) at a multiplicity of infection (m.o.i.) of0.01. One hour post infection (p.i.), cells were washed and incubated forfurther 23 hwithmedia containing solvent (DMSO) or various amounts ofHsp90 inhibitors (GA and17-AAG). Viral titres were determined by plaqueassay (Cooper, 1967). Fig. 2A showed that the inhibition was mostpronounced at 800 nMGA and at 1 μM17-AAG. The kinetics of inhibitionof virus multiplication with 800 nM GA (upper panel) and with 1 μM 17-AAG treatment (lower panel) is shown in Fig. 2B. Similar results wereobtained using several other FluA strains including a distantly relatedH7N7 strain (SC35M, data not shown).We conclude that Hsp90 inhibitorsinhibit viral growth at early time points.

Hsp90 inhibitors delay accumulation of viral mRNA, cRNA and vRNA

To determine whether viral transcription and/or replication isaffected by Hsp90 inhibitors, 293T cells were infected with A/WSN/33virus (m.o.i.=6) in the presence of GA or a DMSO control. Cells wereharvested at 3, 6 and 9 h p.i. and total RNA was isolated (see Materialsand methods). The levels of the three species of viral RNAs, mRNA, cRNAand vRNA, synthesized from the neuraminidase (NA) segment and thehost cell derived 5S rRNA (Hara et al., 2006). were determined byprimer extension (Fodor et al., 2002; Vreede et al., 2004) (Fig. 3A) andquantitated (Figs. 3B–D). Increasing amounts of GAup to 400 nM resultedin adelayed synthesis ofNAmRNA, cRNAandvRNA. Indetail, at 3hp.i.,weobserved adecrease in all threeRNAspecies. At later timepoints, however,therewas a reduction of cRNA (Fig. 3C) and vRNA levels (Fig. 3D) but not ofmRNA levels, although thepeak levels ofmRNAweredelayed compared tothe DMSO control (Fig. 3B, see Discussion).

Accumulation of viral RNAs is inhibited by Hsp90 inhibitors in a RNPreconstitution assay

A preliminary experiment, using a FluA minireplicon assay with afirefly luciferase reporter gene to test the effect of Hsp90 inhibitors on thereconstitutedviral polymerase in293Tcells (Ghanemet al., 2007), showedthat 400 nM GA significantly reduced reporter activity (data not shown).Next we performed a RNP reconstitution experiment to test the effect ofGAon individual viral RNA species. 293Tcellswere transfectedwith Pol II-driven pcDNA expression plasmids coding for PB1, PB2, PA and NP (Fodoret al., 2002) and a Pol I-driven expressionplasmid coding for the influenzaA virus NA vRNA segment (Pleschka et al., 1996) in the presence ofincreasing amounts of GA. The three species of viral RNAs were analyzedby primer extension at 24 h post-transfection (Fodor et al 2002). Fig. 3A(raw data) and B (quantitated) showed that increasing amounts of GAresulted in a dose-dependent reduction in all three viral RNA species —specifically mRNA, cRNA and vRNA, suggesting that accumulation of viralRNAs is dependent on Hsp90 function.

GA inhibits RNP formation

The decrease in all three viral RNA species observed in Fig. 4A mayresult from a decrease in RNP formation in GA treated cells. To directlytest this hypothesis, we reconstituted a viral RNP containing TAP-taggedNP in 293T cells with GA treatment (200 nM and 400 nM) or DMSOcontrol and performed TAP purification (Deng et al., 2005) at differenttimes post transfection (13 and 20 h). This procedure is known to bespecific for the TAP tag since no detectable nonspecific pull-down ofindividual polymerase subunits by IgG Sepharose was observed before(Deng et al., 2005). Moreover it is known that TAP-tagged NP retainssignificant replication and transcription activities compared towild typein reconstituted RNP when assayed by primer extension (Vreede andBrownlee, 2007). After TAP purification of the RNPs, the association of

Page 3: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 2.Hsp90 inhibitors delay growth of influenza A virus in cell culture. A) Effect of Hsp90 inhibitors (GA and 17-AAG at various concentrations) on virus titres in MDCK cells infectedwith influenza A virus (A/WSN/33) at m.o.i.=0.01. B) Effect of GA (800 nM) and 17-AAG (1000 nM) on kinetics of viral growth. MDCK cells were infected and subsequently incubatedwith GA or 17-AAG or solvent (DMSO). Viral titres were determined by plaque assay at 24 h (A) or the indicated time points (B) p.i. Values represent the mean of three independentexperiments, and error bars show the standard deviation of the mean.

433G. Chase et al. / Virology 377 (2008) 431–439

viral polymerase with the RNP complexes was determined by Westernblot analysis.We used PA, rather than PB1 and PB2 antibody to estimatethe level of the reconstituted RNP complex because known specificinteractions between free NP and either PB1 or PB2 precludes usingthese antibodies (Biswas et al., 1998; Medcalf et al., 1999). As shown inFig. 4C (ii), there is a significant reduction in the amount of co-purifiedPA in cells treated with GA compared to untreated cells, despite similaramounts of purified TAP-NP (iii) and similar amounts of PA (i) in thecrude cell extracts from both treated and untreated cells. We alsoanalysed the activity of the purified RNPs in an in vitro ApG-primedtranscription assay (see Materials and methods) in the absence of UTPgiving rise to a 14 nt long partial transcript from endogenous, viral NAtemplate present in the RNP complex. Any cRNP purified with the vRNPwould not be transcribed in the ApG-primed transcription assay in theabsence of UTP, because UTP is required at nucleotide 3 immediatelyfollowing the ApG primer (Deng et al., 2006). As expected, lower yieldsof transcripts were produced with TAP-purified vRNPs from GA-treatedcells, compared to vRNPs from untreated cells [Fig. 4C (iv)]. Somewhatsurprisingly the inhibition by GA was apparently more pronounced inthe purified RNPs when assayed for enzymatic activity by ApG-primedtranscription [Fig. 4C (iv)] than when mRNA was assayed in recon-stituted RNP by primer extension (Fig. 4B). The reasons for this areunknown but may reflect the different assays used (mRNA levels versusprotein activity) or the slightly different times of transfection. However,ApG-primed transcription of purified vRNPs in vitro is not blocked byGA (Fig. 4D). Overall, these results indicate that formation of activevRNPs is hindered by treatment with GA, but GA does not inhibit thepreformed complex.

In order to exclude the possibility that the effect of GA observed(Figs. 4A–C) was caused by GA directly inhibiting host Pol I or Pol II,293T cells were transfected with either pPol I-NA (Fig. 4E), or the PolII-driven expression plasmid, pcDNA-NA (Fig. 4F). The pPOl I- and PolII transcripts were then analysed by primer extension (see Materialsand methods) at different time post-transfection (13 and 20 h).

Quantitation of the three experiments indicates no inhibition of Pol Ior Pol II transcription.

Hsp90 inhibitors enhance the degradation of PB1 and PB2

It has been shown that GA and 17-AAG can prevent the release ofclient proteins from Hsp90, which eventually leads to their degrada-tion by the proteasome (Pearl and Prodromou, 2006). The simplestexplanation of the observed inhibition of FluA by GA or 17-AAG is thusan enhanced degradation of viral proteins which happen to be Hsp90clients. To test this hypothesis, we performed pulse-chase experi-ments as shown in Fig. 5 (upper panel). Preliminary experiments hadshown that individually expressed polymerase PB1 and PB2 subunitshad a very short half life (b2 h); therefore, a short, 1 h chase wasoptimal for our experiments. Quantitation showed that PB2 ispreferentially degraded with increasing concentrations of inhibitor,and PB1 is degraded only at the highest concentration of inhibitor,whereas PA was unaffected (Fig. 5). This selectivity for PB1 and PB2,but not PA, correlates with the observation that both PB1 and PB2 bindto Hsp90 [(Naito et al., 2007), data not shown]. Therefore, these resultssupport the hypothesis that PB1 and PB2 are client proteins of Hsp90.Thus Hsp90 inhibitors can enhance the degradation of PB1 and PB2.

GA treatment reduces nuclear accumulation of PA and PB1, but not PB2

Recently it has been reported that Hsp90 may be important for thenuclear import of influenza polymerase subunits (Naito et al., 2007).Wethus analyzed the intracellular localization of the three individuallyexpressed subunits in 293T cells, in the presence or absence of 500 nMGAusingGFP fusionproteinswhichdistribute similarly to their untaggedcounterparts (Fodor and Smith, 2004). We observed a significantcytoplasmic retention of PB1, and to our surprise also PA (see below),but not PB2 (Fig. 6). The finding that PB2 localizationwas not affected byGA was unexpected since PB2 is an Hsp90 client protein. We can only

Page 4: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 3. Effect of GA on accumulation of viral RNAs in influenza A virus (A/WSN/33) infected 293T cells. A) Primer extension assays of vRNA, mRNA, and cRNA of NA segment at varioustime points post-infection with DMSO, 200 nM or 400 nM GA. A longer exposure of the cRNA signal is shown below. B) Quantitation of viral mRNAs in GA-treated infected cells. Theradioactive signals corresponding to the viral mRNAs and 5S rRNA obtained in Awere quantified by phosphorimage analysis. Values were first standardized to the 5S rRNA signal. Thethree experiments were then normalized to one another by correcting for the different phosphoimaging exposure times in three different experiments. C) Quantitation of viral cRNAsin GA-treated infected cells. D) Quantitation of viral vRNAs in GA-treated infected cells. Error bars in B, C and D are standard deviations of three independent experiments.

434 G. Chase et al. / Virology 377 (2008) 431–439

speculate that the quantitation method we used (Fig. 6) was insuffi-ciently sensitive to detect a small increase in the levels of cytoplasmicPB2 that would be expected in the presence of GA. The difficulty inestimating cytoplasmic PB2 levels is compounded by the fact thatrelatively low levels of PB2 are present in the cytosol, because of itsefficient nuclear localization compared to PB1 and PA (Fodor and Smith,2004). We also tested the effect of GA on intracellular localization of the3P complex by co-transfecting three pcDNA plasmids expressing PB1-GFP, PA, and PB2 in the presence or absence of 500 nMGA.We observedonly nuclear localization of the complex, as assayed by the GFP signal(data not shown) in both treated and, as previously reported (Fodor andSmith, 2004), in untreated cells. However, the GFP signal was generallylower in cells treatedwith 500 nMGA than those treatedwith theDMSOcontrol (data not shown).

GA treatment reduces nuclear levels of 3P in infected cells

Since we have observed that GA treatment inhibits RNP formation(Fig. 4), nuclear import of individually expressed PB1 and PA (Fig. 6),and enhances the degradation of PB1 and PB2 (Fig. 5), we wereinterested to test the intracellular localization of the viral polymerasecomplex in infected cells. A549 cells were infected with influenza Avirus (A/WSN/33) at a multiplicity of infection of 2. Cells wereincubated with media containing solvent (DMSO) and with either200 nM or 400 nM GA. At 9 h post-infection, the localization of thepolymerase complexes was examined by indirect immunofluores-cence (Fig. 7) (see Materials and methods). Consistent with the

nuclear localization of the 3P complex in transfected cells (see above),the PA subunit of the polymerase complex localized in the nucleus ofthe infected A549 cells at 9 h post-infection in cells with orwithout GAtreatment. However, significantly, the levels of PA in the nucleus werereduced in a dose-dependent manner with the GA treatment(compare Fig. 7 upper, middle and lower panels). The low levels ofPA and its ability to form a complex initially with PB1 in the cytosoland subsequently with PB2 in the cell nucleus (Deng et al., 2005;Fodor and Smith, 2004) might exclude detectable cytoplasmiclocalization in the presence of GA in these infection experiments,even though this was observed with individually transfected PB1 andPA (Fig. 6, above).

Discussion

The aim of this workwas to test whether Hsp90 inhibitors could beused as influenza antivirals since Hsp90 was known to play a role inthe nuclear import and assembly of the trimeric polymerase complex(Naito et al., 2007). Here, we provide evidence that GA and 17-AAGimpair influenza viral growth by reducing the levels of ribonucleo-protein complexes (Figs. 3 and 7). We speculate that the decrease inthe amounts of RNPs is due to selective degradation of the PB1 andPB2 subunits in the presence of Hsp90 inhibitors (Fig. 5). Thisobservation correlates with our finding that less polymerase complexis associated with TAP-NP in GA-treated cells (Fig. 4C), which suggeststhat there are fewer trimeric complexes in the cell nucleus which arecompetent to form ribonucleoprotein complexes. Moreover the

Page 5: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 4. Hsp90 inhibitors reduce RNP formation. A) GA reduces accumulation of viral RNAs in a RNP reconstitution assay. The positions of transcription products derived from NAmRNA, cRNA, vRNA, and 5S rRNA, used as a control, are indicated on the right. A typical result of four experiments, harvested 24 h post-transfection, is shown. B) Quantitation of viralRNAs in the presence of GA in the RNP reconstitution assay. An average of four experiments is shownwith standard deviations. Values were standardized to the 5S rRNA signal, andvalues were expressed as % of the control values set to 100% (samples treated with DMSO). Values significantly different from 100% (Pb0.05, Student's t test) are indicated by anasterisk (⁎). C) GA inhibits RNP formation. The amount of PA in crude lysate (i) and in RNP (ii) was examined byWestern blot using a polyclonal anti-PA antibody (Hara et al., 2006).The amount of TAP-NP (N-terminally tagged) present in the purified samplewas examined byWestern blotswith anti-NP antibody (iii). The activity of the purified RNPswas analysedin vitro using an ApG-primed transcription assay (iv). The reaction contains [α-32P]-GTP,(iv). The 14 nt long transcription product (TP) is indicated on the right. D) GA does not inhibitpreformed RNP. An in vitro ApG-primed transcription assay (see iv above) using RNPs purified from the DMSO control sample [same sample used in Fig. 4C (iv) lane 4] was performedin the presence of increasing concentrations of GA. E) GA does not inhibit Pol I transcription. Quantitation of 3 experiments analysing a Pol I NA transcript with DMSO, 200 nM or400 nM GA treatment at 13 h and 20 h post-transfection by primer extension analysis. F) GA does not inhibit Pol II transcription. Quantitation of three experiments analyzing a Pol IINA transcript with DMSO, 200 nM GA or 400 nM GA treatment at 13 h and 20 h post-transfection by primer extension analysis.

435G. Chase et al. / Virology 377 (2008) 431–439

cytoplasmic retention of both PA and PB1 in GA-treated cells (Fig. 6)could cause further reduction in the amount of RNPs available for viraltranscription and replication in the nucleus. Although there is noevidence for an interaction between PA and Hsp90 [(Naito et al., 2007),data not shown] it is possible that Hsp90 is required for a host functionsupporting PAnuclear accumulation. However, we have been unable to

detect significantly less formation of recombinant trimeric polymeraseisolated by transient (37 h) transfection of GA-treated (either at 200 or400 nM) 293T cells, using PB1, PA and a PB2-TAP tagged constructs ofinfluenza A/WSN/33 virus, by IgG affinity methods (Deng et al., 2005)(data not shown). Our failure to showsignificant differences in trimericpolymerase levels in the presence of GA suggests that the expected

Page 6: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 5.Hsp90 inhibitors induce enhanced degradation of PB1 and PB2. Upper panel: 105 293Tcells were transfected with 2 μg pCA expression vectors coding either for HA-tagged PB1,PB2 or PA and pulse-chased with 35S-labelled amino acids (cysteine andmethionine) as indicated in the presence of increasing concentrations of 17-AAG. Cell extracts were obtainedfollowed by immunoprecipitation of the individual subunits using agarose beads coupled to an anti-HA antibody. Lower panel: Protein levels from three independent experimentswere quantitated. Values are expressed as % of control (DMSO) levels. Error bars are the standard deviations; asterisks (⁎) indicate values significantly different from 100% (Pb0.05,Student's t test).

436 G. Chase et al. / Virology 377 (2008) 431–439

reduction in trimeric polymerase may either be too small to detectunder the experimental conditions used or that other unknown effectsof GA may interfere with RNP assembly.

GA delays the accumulation of all 3 viral species (mRNA, cRNA andvRNA), although the kinetics differ for the accumulation of mRNAcompared to that of cRNA and vRNA (Fig. 3). Specifically, in virus-

Fig. 6. GA treatment reduces nuclear accumulation of PA and PB1, but not PB2. 293T cells weGFP or PB2-GFP (Fodor and Smith, 2004) in the presence of 500 nM GA. At 16 h post transfec(lower panels). Cells were scored for the localization pattern of the GFP-tagged polymerase sucytoplasmic. The number of cells showing each localization patternwas expressed as % of thepatterns from three independent transfections with standard deviations is shown. Student'ssignificantly different from the control values of DMSO-treated cells. Values that significant

infected cells, the delay in mRNA synthesis is less marked than that ofvRNA and cRNA suggesting that GA inhibits replication more thantranscription. A possible explanation of this observation is as follows:early after infection GA limits the amount of available influenzapolymerase thereby inhibiting the synthesis ofmRNA, cRNA and vRNA.At later times, vRNA and cRNA accumulation aremore dependent than

re transfected with plasmids expressing GFP-tagged polymerase subunits PA-GFP, PB1-tion, cells were fixed with paraformaldehyde and analyzed by fluorescence microscopybunit: N, predominantly nuclear; N/C, distributed throughout the cell; C, predominantlytotal cell number (approximately 500 cells) (upper panels). The average % of localizationt test was performed to assess whether the values obtained in the presence of GAwerely differ from each other (Pb0.01) are indicated by an asterisk (⁎).

Page 7: Hsp90 inhibitors reduce influenza virus replication in cell culture

Fig. 7. GA treatment reduces nuclear accumulation of PA in infected cells. A) A549 cells were infected with influenza A/WSN/33 viruses at an m.o.i of 2 and were treated with DMSO,200 nM or 400 nM GA. At 9 h post-infection, cells were fixed and stained with a rabbit polyclonal anti-PA primary antibody and a Cy3-conjugated anti-rabbit secondary antibody.Images were taken using confocal microscopy under 20× magnification with identical settings. B) Images of 200 nM GA sample were taken under 63× magnification with identicalsetting to show the nuclear signals.

437G. Chase et al. / Virology 377 (2008) 431–439

mRNA on the levels of 3P and NP because these viral proteins areneeded to stabilize RNP (Vreede et al., 2004), whereas mRNA isstabilized by host proteins. Alternatively, we speculate that, at latertime points, the inhibition of host Pol II by the influenza polymerase(Engelhardt et al., 2005; Rodriguez et al., 2007) may be less markedbecause of the limited amounts of influenza polymerase in thepresence of GA. Thus more host capped primers could be availablefor the synthesis of viralmRNA, allowing its continued synthesis late ininfection. Overall our results are consistent with a previous studywhich showed that GA can cause reduction in the levels of both PB2and vRNA in FluA-infected cells (Dalton et al., 2006).

There is ample evidence that Hsp90 inhibitors impair the growthof many other viruses, including poliovirus (Geller et al., 2007),vesicular stomatitis virus (VSV) (Connor et al., 2007) and hepatitis Cvirus (Nakagawa et al., 2007). In the case of VSV, another negative-strand RNA virus, GA caused a similar delay in viral multiplication asobserved here with FluA, although at early time points p.i. there was agreater reduction in titres with VSV than FluA (Connor et al., 2007).However, a direct comparison of titres is not possible due to thedifferent concentrations of GA used in the two studies. In all cases, theinhibition was paralleled by enhanced degradation of viral proteinswhich bind to Hsp90, such as polymerases (Connor et al., 2007) and

Page 8: Hsp90 inhibitors reduce influenza virus replication in cell culture

438 G. Chase et al. / Virology 377 (2008) 431–439

capsid proteins (Geller et al., 2007). However, these studies, includingours, cannot formally exclude the possibility that these effects are dueto the inhibition of Hsp90-dependent cellular functions that canindirectly influence viral replication. Moreover, Hsp90 inhibitors couldhave additional unrecognized effects. Nonetheless, as recently shownfor poliovirus infection (Geller et al., 2007), such low-toxicityinhibitors may provide a new tool to interfere with viral infection.Unlike existing fluA antivirals (e.g. zanamivir, oseltamivir, amanta-dine) which are designed to target influenza viral proteins (Palese andShaw, 2006), Hsp90 inhibitors, such as geldanamycin, 17-AAG orimproved more active derivatives, targeting a host factor may providea novel antiviral strategy not readily susceptible to the development ofdrug resistance.

Materials and methods

Plasmids

The plasmids encoding PB1, PB2, PA and NP of influenza virus A/WSN/33 (pcDNA-PB1, pcDNA-PA, pcDNA-PB1, pcDNA-PB2, pcDNA-NP,pcDNA-TAP-NP pcDNA-PB1-GFP, pcDNA-PB2-GFP and pcDNA PA-GFP)have been described previously (Deng et al., 2005; Engelhardt et al.,2005; Fodor et al., 2002; Fodor and Smith, 2004). pPOLI-NA has alsobeen described (Pleschka et al., 1996). pcDNA-NA was derived frompPOLI-NA by restriction with NgoMIV followed by the insertion of theNA open reading frame of the NA gene of A/WSN/33 virus into the AgeIsite of pcDNA3A (Fodor et al., 2002). pCA-PA-HA, pCA-PB1-HA, andpCA-PB2-HA have also been described previously (Ghanem et al.,2007; Mayer et al., 2007).

MTT assay

The 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-mide (MTT) assay measures the metabolic activity of cultured cellsusing a colorimetric substrate (Mosmann, 1983). Cells were seededinto 96-well plates and treated with GA (Invivogen) or 17-AAG(Biomol). 24 h after treatment, cells were assayed for metabolicactivity using the MTT Cell Proliferation Kit I (Roche) according tomanufacturer's instructions.

Analysis of vRNA, mRNA and cRNA by primer extension assays

Primer extension analysis of neuraminidase (NA)-specific viralRNAs was performed as described previously (Fodor et al., 2002;Vreede et al., 2004). Transcription products were analyzed on 6%polyacrylamide gels containing 7 M urea in Tris-borate-EDTA bufferand were detected by autoradiography. The expected sizes of thetranscription products were 129 nucleotides (nt) (vRNA), 169 to 177 nt(mRNA), and 160 nt (cRNA).

Purification of reconstituted RNPs

Human embryonic kidney 293Tcells were transfectedwith pcDNA-PA, pcDNA-PB1, pcDNA-PB2, pcDNA-TAP-NP and pPOLI-NA for 48 h toexpress influenza RNA polymerase subunits, viral RNA template andTAP-tagged NP. The RNP complexes were purified by using the TAPmethod as described previously (Deng et al., 2005; Fodor and Smith,2004; Puig et al., 2001). Briefly, cell lysates prepared from transfected293T cells were incubated with IgG-Sepharose and bound proteinsreleased by cleavage with tobacco etch virus (TEV) protease (Invitro-gen). Purified protein samples were analyzed byWestern blotting. TheApG-primed transcription assay [modified from (Fodor et al., 2002)] ofpurified RNPs was performed in the presence of [α-32P]-GTP, ATP, CTP,but in the absence of UTP, giving rise to a 14 nt long partial transcript(AGCGAAAGCAGGAG) of endogenous, viral NA vRNA template presentin the RNPcomplex. Transcriptswere analysedby 16%PAGE in7Murea.

Pulse-chase experiments

Transfected 293T cells were starved by incubation for 15 min withDMEM without cysteine or methionine, then pulse-labelled 1.5 h inthe same medium containing 60 μCi/ml 35S-cysteine/methionine(Amersham). Cells were then chasedwith complete DMEM containingGA or 17-AAG for 1 h before lysis in RIPA buffer (50 mM Tris–HCl pH8.0, 150 mM NaCl, 1% Nonidet P-40, 0.1% SDS, 0.25% sodiumdeoxycholate) and incubation with anti-HA agarose (Sigma) for 2 hat 4 °C. Beads were washed 4 times with RIPA buffer, resuspended inloading buffer, separated by SDS-PAGE and labelled proteins visua-lized by autoradiography and quantitated.

Localization of polymerase subunits in transfected cells

293T cells grown on 22×22 mm coverslips in six-well plates, weretransfected with plasmids expressing PB1-GFP, PA-GFP and PB2-GFP,individually, in the presence or absence of 500 nM GA. At 16 h post-transfection, cells were fixed with 4% paraformaldehyde in 250 mMHEPES (pH 7.5), and coverslips were mounted in Mowiol containing4′,6′-diamidino-2-phenylindole. Cells were viewed by using a ZeissAxioplan microscope with a Zeiss×63 oil immersion objective with anumerical aperture of 1.25, and the images were processed by usingAdobe Photoshop 7.0. For numerical analysis of polymerase distribu-tion in transfected cells, 500 (for individually expressed PB1-GFP, PA-GFP and PB2-GFP) fluorescent cells per coverslip were examined andscored according to whether the observed fluorescence was pre-dominantly cytoplasmic, nuclear or present throughout the cell.Percentages of cells were calculated and expressed as an average fromthree independent experiments.

Indirect immunofluorescence

A549 cells grown on 13 mm coverslips in 24-well plates wereinfected at an m.o.i of 2 in the presence DMSO or 200 nM or 400 nMGA. At 6 and 9 h post-infection, the cells were fixed with 2%paraformaldehyde in phosphate-buffered saline (PBS). Followingfixation, the cells were permeabilized with 0.1% Triton X-100 in PBSfor 10 min at room temperature, blocked and incubated with a rabbitpolyclonal PA antibody in PBS containing 3% bovine serum albumin for1 h at room temperature. The cells were further incubated with Cy3-conjugated anti-rabbit secondary antibody (Jackson ImmunoRe-search) at a 1/400 dilution at room temperature for 1 h. Coverslipswere mounted in Mowiol. Cells were viewed by confocal microscopywith a 20× or 63× objectives, and the images were processed by LMSimage browser.

Acknowledgments

We thank Dr T. Toyoda (Shanghai) for providing rabbit anti-PAantibody. This study was supported by the Deutsche Forschungsge-meinschaft (DFG), the Bundesministerium für Bildung und Forschung(BMBF, FluResearchNet), the European Commission (FLUINNATE) andMRCgrants (G9523972,G9826944,G9901312,G117/457andG0700848).

References

Biswas, S.K., Boutz, P.L., Nayak, D.P., 1998. Influenza virus nucleoprotein interacts withinfluenza virus polymerase proteins. J. Virol. 72, 5493–5501.

Connor, J.H., McKenzie, M.O., Parks, G.D., Lyles, D.S., 2007. Antiviral activity and RNApolymerase degradation following Hsp90 inhibition in a range of negative strandviruses. Virology 362, 109–119.

Cooper, P., 1967. The plaque assay of animal viruses. In: Maramorosch, K., Koprowski, H.(Eds.), Methods in Virology, vol. III. Academic Press, New York, pp. 243–311.

Dalton, R.M., Mullin, A.E., Amorim, M.J., Medcalf, E., Tiley, L.S., Digard, P., 2006.Temperature sensitive influenza A virus genome replication results from lowthermal stability of polymerase-cRNA complexes. Virol. J. 3, 58.

Deng, T., Sharps, J., Fodor, E., Brownlee, G.G., 2005. In vitro assembly of PB2 with a PB1-

Page 9: Hsp90 inhibitors reduce influenza virus replication in cell culture

439G. Chase et al. / Virology 377 (2008) 431–439

PA dimer supports a new model of assembly of influenza A virus polymerasesubunits into a functional trimeric complex. J. Virol. 79, 8669–8674.

Deng, T., Vreede, F.T., Brownlee, G.G., 2006. Different de novo initiation strategies areused by influenza virus RNA polymerase on its cRNA and viral RNA promotersduring viral RNA replication. J. Virol. 80, 2337–2348.

Engelhardt, O.G., Smith, M., Fodor, E., 2005. Association of the influenza A virus RNA-dependent RNA polymerasewith cellular RNA polymerase II. J. Virol. 79, 5812–5818.

Fodor, E., Smith, M., 2004. The PA subunit is required for efficient nuclear accumulationof the PB1 subunit of the influenza A virus RNA polymerase complex. J. Virol. 78,9144–9153.

Fodor, E., Crow, M., Mingay, L.J., Deng, T., Sharps, J., Fechter, P., Brownlee, G.G., 2002. Asingle amino acidmutation in the PA subunit of the influenza virus RNA polymeraseinhibits endonucleolytic cleavage of capped RNAs. J. Virol. 76, 8989–9001.

Geller, R., Vignuzzi, M., Andino, R., Frydman, J., 2007. Evolutionary constraints onchaperone-mediated folding provide an antiviral approach refractory to develop-ment of drug resistance. Genes Dev. 21, 195–205.

Ghanem, A., Mayer, D., Chase, G., Tegge, W., Frank, R., Kochs, G., Garcia-Sastre, A.,Schwemmle, M., 2007. Peptide-mediated interference with influenza A virus poly-merase. J. Virol. 81, 7801–7804.

Hara, K., Schmidt, F.I., Crow, M., Brownlee, G.G., 2006. Amino acid residues in theN-terminal region of the PA subunit of influenza A virus RNA polymerase play acritical role in protein stability, endonuclease activity, cap binding, and virion RNApromoter binding. J. Virol. 80, 7789–7798.

Kawaguchi, A., Nagata, K., 2007. De novo replication of the influenzaRNA genome isregulated by DNA replicative helicase, MCM. EMBO J. 26, 4566–4575.

Mayer, D., Molawi, K., Martinez-Sobrido, L., Ghanem, A., Thomas, S., Baginsky, S.,Grossmann, J., Garcia-Sastre, A., Schwemmle, M., 2007. Identification of cellularinteraction partners of the influenza virus ribonucleoprotein complex and poly-merase complex using proteomic-based approaches. J. Proteome Res. 6, 672–682.

Medcalf, L., Poole, E., Elton, D., Digard, P., 1999. Temperature-sensitive lesions in twoinfluenza A viruses defective for replicative transcription disrupt RNA binding bythe nucleoprotein. J. Virol. 73, 7349–7356.

Momose, F., Naito, T., Yano, K., Sugimoto, S., Morikawa, Y., Nagata, K., 2002. Identificationof Hsp90 as a stimulatory host factor involved in influenza virus RNA synthesis.J. Biol. Chem. 277, 45306–45314.

Mosmann, T., 1983. Rapid colorimetric assay for cellular growth and survival:application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63.

Naito, T., Momose, F., Kawaguchi, A., Nagata, K., 2007. Involvement of Hsp90 in assemblyand nuclear import of influenza virus RNA polymerase subunits. J. Virol. 81,1339–1349.

Nakagawa, S., Umehara, T., Matsuda, C., Kuge, S., Sudoh, M., Kohara, M., 2007. Hsp90inhibitors suppress HCV replication in replicon cells and humanized liver mice.Biochem. Biophys. Res. Commun. 353, 882–888.

Palese, P., Shaw, M.L., 2006. Orthomyxoviridae: the viruses and their replication, In:Knipe, D.M., Howley, P.M. (Eds.), Fifth ed. Fields Virology, vol. 2. Lippincott, Williams& Wilkins, Philadelphia, pp. 1647–1689.

Pearl, L.H., Prodromou, C., 2006. Structure and mechanism of the Hsp90 molecularchaperone machinery. Ann. Rev. Biochem. 75, 271–294.

Pleschka, S., Jaskunas, S.R., Engelhardt, O.G., Zurcher, T., Palese, P., Garcia-Sastre, A.,1996.A plasmid-based reverse genetics system for influenza A virus. J. Virol. 70,4188–4192.

Puig, O., Caspary, F., Rigaut, G., Rutz, B., Bouveret, E., Bragado-Nilsson, E., Wilm, M.,Seraphin, B., 2001. The tandem affinity purification (TAP) method: a generalprocedure of protein complex purification. Methods 24, 218–229.

Rodriguez, A., Perez-Gonzalez, A., Nieto, A., 2007. Influenza virus infection causesspecific degradation of the largest subunit of cellular RNA polymerase II. J. Virol. 81,5315–5324.

Vreede, F.T., Brownlee, G.G., 2007. Influenza virion-derived viral ribonucleoproteinssynthesize both mRNA and cRNA in vitro. J. Virol. 81, 2196–2204.

Vreede, F.T., Jung, T.E., Brownlee, G.G., 2004. Model suggesting that replication ofinfluenza virus is regulated by stabilization of replicative intermediates. J. Virol. 78,9568–9572.

Whitesell, L., Lindquist, S.L., 2005. HSP90 and the chaperoning of cancer. Nat. Rev.,Cancer 5, 761–772.