You are on page 1of 10

Hsp70 Protein Positively Regulates Rabies

Virus Infection
Xavier Lahaye, Aurore Vidy, Baptiste Fouquet and Danielle
Blondel
J. Virol. 2012, 86(9):4743. DOI: 10.1128/JVI.06501-11.
Published Ahead of Print 15 February 2012.

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
Updated information and services can be found at:
http://jvi.asm.org/content/86/9/4743

These include:
REFERENCES This article cites 42 articles, 21 of which can be accessed free
at: http://jvi.asm.org/content/86/9/4743#ref-list-1

CONTENT ALERTS Receive: RSS Feeds, eTOCs, free email alerts (when new
articles cite this article), more»

Information about commercial reprint orders: http://journals.asm.org/site/misc/reprints.xhtml


To subscribe to to another ASM Journal go to: http://journals.asm.org/site/subscriptions/
Hsp70 Protein Positively Regulates Rabies Virus Infection
Xavier Lahaye,* Aurore Vidy,* Baptiste Fouquet, and Danielle Blondel
CNRS, UPR 3296 Virologie Moléculaire et Structurale, Gif sur Yvette, France

The Hsp70 chaperone plays a central role in multiple processes within cells, including protein translation, folding, intracellular
trafficking, and degradation. This protein is implicated in the replication of numerous viruses. We have shown that rabies virus
infection induced the cellular expression of Hsp70, which accumulated in Negri body-like structures, where viral transcription
and replication take place. In addition, Hsp70 is present in both nucleocapsids purified from infected cells and in purified viri-
ons. Hsp70 has been shown to interact with the nucleoprotein N. The downregulation of Hsp70, using specific chaperone inhibi-

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
tors, such as quercetin or RNA interference, resulted in a significant decrease of the amount of viral mRNAs, viral proteins, and
virus particles. These results indicate that Hsp70 has a proviral function during rabies virus infection and suggest that Hsp70 is
involved in at least one stage(s) of the viral life cycle, such as viral transcription, translation, and/or production. The mechanism
by which Hsp70 controls viral infection will be discussed.

I n eukaryotic cells, the posttranslational folding of proteins and


quality control are performed by molecular chaperones and heat
shock proteins (HSPs). The ubiquitous chaperone family of the
was found to have both positive and negative regulatory effects on
the RNA replication of flock house virus, a nodavirus (39), high-
lighting the complexity of this particular virus-chaperone interac-
70-kDa HSPs plays a central role in protein homeostasis and pro- tion.
tection against proteotoxic stresses by preventing protein misfold- Rabies virus, the prototype of the Lyssavirus genus that belongs
ing and aggregation by directing damaged protein to the ubiqui- to the Rhabdoviridae family (Mononegavirales order), causes a fa-
tin-proteasome system for degradation (4, 15, 34, 40). Hsp70 tal disease that is associated with intense viral replication in the
chaperones not only survey the folding status of proteins as part of central nervous system. Its single-stranded negative-sense RNA
the quality control function, which is very important under stress genome (⬃12 kb), encoding five viral proteins, is encapsidated by
conditions, but also are involved in the regulation of fundamental the nucleoprotein (N; 40 kDa) to form the nucleocapsid that is
cellular processes, such as signal transduction, cell cycle regula- associated with the RNA-dependent RNA polymerase L (220 kDa)
tion, apoptosis, and innate immunity (23, 25). and its cofactor, phosphoprotein (P; 33 kDa). Inside the viral par-
Viral infection depends on the successful recruitment of host ticle, the nucleocapsid has a tightly coiled helical structure that is
cellular factors at different steps of the viral life cycle: genome associated with the matrix protein (M; 22 kDa) and is surrounded
replication, viral protein synthesis, viral assembly, and counterde- by a membrane containing a unique glycoprotein (G; 62 kDa).
fense against cell apoptosis and innate immunity. During viral The virus enters the host cell through the endosomal transport
infection, large amounts of viral proteins are synthesized and pro- pathway via a low-pH-induced membrane fusion process cata-
tein folding can become a limiting step. Therefore, on the one lyzed by G (14). The nucleocapsid released into the cytoplasm
hand viruses need cellular chaperones for their own protein fold- serves as a template for transcription and replication processes
ing processes; on the other hand, as chaperones are involved in the that are catalyzed by the L-P polymerase complex (for a review see
regulation of fundamental cellular processes, viruses have to in- reference 1). It has been shown recently that rabies virus transcrip-
teract with them. Hsp70 chaperone, the major inducible heat tion and replication take place within Negri body-like (NBL)
shock protein, is frequently recruited by viruses. Research during structures, which are inclusion bodies formed during viral infec-
the past 30 years has revealed the involvement of Hsp70 in all steps tion (20). During transcription, a positive-stranded leader RNA
of viral life cycle replication and for viruses from numerous fam- and five capped and polyadenylated mRNAs are synthesized. The
ilies of diverse orders (24, 28). Several DNA viruses, such as her- replication process yields nucleocapsids containing full-length
pesvirus (HSV1) (10, 38), vaccinia virus (18), adenovirus type 5 antigenome-sense RNA, which in turn serve as templates for the
(24), and simian virus 40 (SV40) (27, 31), induce the specific synthesis of genome-sense RNA. During their synthesis, both the
expression of Hsp70 (33). In most cases this induction has a pro- nascent antigenome and the genome are encapsidated by N pro-
viral effect. Evidence is growing that the Hsp70 induction is also
important for the replication of many positive-strand RNA and
negative-strand RNA viruses (28). In the case of measles virus, Received 6 October 2011 Accepted 9 February 2012
Hsp70 interacts with nucleocapsids (42) and induces the in- Published ahead of print 15 February 2012
creased expression of viral genes (5). Nevertheless, in some cases Address correspondence to Danielle Blondel, blondel@vms.cnrs-gif.fr.
Hsp70 was found to inhibit viral infection. An increase of its ex- * Present address: X. Lahaye, Institut Curie, Centre de Recherche, Human Innate
pression confers to cells a protection against rotavirus (2), vesic- Immunity, Paris, France; A. Vidy, Max von Pettenkofer Institute für Virologie,
ular stomatitis virus (9), respiratory syncytial virus (41), and in- Ludwig Maximilians University, Munich, Germany.
fluenza virus (16) infections. In the latter case, it has been reported X.L. and A.V. contributed equally to this article.
that Hsp70, which interacts with the ribonucleoprotein complex, Copyright © 2012, American Society for Microbiology. All Rights Reserved.
interferes with the polymerase activity and negatively regulates doi:10.1128/JVI.06501-11
viral transcription and replication (16, 22). Interestingly, Hsp70

0022-538X/12/$12.00 Journal of Virology p. 4743– 4751 jvi.asm.org 4743


Lahaye et al.

tein. This encapsidation is regulated by the P protein, which plays Treatment of cells with drugs. Cells were kept in DMEM containing
the role of chaperone, preventing the N protein from binding to 25 to 200 ␮M quercetin for 1 h before virus inoculation and during infec-
cellular RNA and from aggregating (32). The neosynthesized tion. As all drugs were reconstituted in dimethyl sulfoxide (DMSO), un-
genomic RNA either serves as a template for secondary transcrip- treated cells were maintained in medium containing the same concentra-
tion or is transported to the cell membrane and assembles with the tion of DMSO. The expression of Hsp70 was induced by a heat shock (HS)
at 45°C for 20 min, and then cells were reincubated at 37°C.
M and G proteins for the budding of neosynthesized virions.
siRNA experiments. The siRNA-Hsp70 sequence of 21 nucleotides
We have previously shown that Hsp70 protein accumulates in
(nt) was chosen based on the rules proposed by Elbashir et al. (12) and was
the NBL structures, which are sites of rabies virus transcription designed to interfere with the two mRNAs encoding Hsp70 (GenBank
and replication (20). In addition, Hsp70 has been previously accession number NM_005345 for HSPA1A and BC057397 for HSPA1B)
shown to be incorporated in purified rabies virus (37). Therefore, but not with the two Hsc70 mRNA variants. The sequences were the
we investigated the role of this chaperone during viral infection. following: siRNA-Hsp70 sense, 5=-CACGGCAACCUGGAGAUCA-de-
We report here that rabies virus infection induces the expression oxythymidine (dTdT)-3=; siRNA-Hsp70 antisense, 5=-UGAUCUCCACC
of Hsp70. We also show that Hsp70 interacts with the nucleopro- UUGCCGUG-dTdT-3=; siRNA-scrambled sense, 5=-ACAAUUCCGGGG

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
tein N, the major component of NBL structures, and that Hsp70 GAGAGAG-dTdT-3=; and siRNA-scrambled antisense, 5=-CUCUCUCC
downregulation results in the inhibition of different steps of the CCCGGAAUUGA-dTdT-3=. Transfections were obtained using an
viral cycle. The inhibition of viral protein synthesis and of viral Invitrogen Lipofectamine kit. BSR cells (in serum-free DMEM) were
production can reflect the inhibition of viral RNA synthesis, but it transfected with 400 pmol of scrambled siRNA or 200 to 400 pmol of
siRNA-Hsp70. After 6 h at 37°C, the medium was completed with 5%
also could be due to a specific role of Hsp70 in these further steps.
FCS. After 48 h at 37°C, it was possible to induce a heat shock and to infect
MATERIALS AND METHODS cells. After incubation, the culture supernatant was collected to quantify
viral production. The cell layer was recovered to obtain total extract for
Virus stock and cell infection. BSR cells, cloned from BHK 21 (baby Western blot analysis.
hamster kidney) cells, were grown in Dulbecco’s modified Eagle medium
Detection of viral mRNA and Hsp70 mRNA by RT-PCR. The pres-
(DMEM) supplemented with 10% fetal bovine serum (FBS). Human neu-
ence of Hsp70 mRNA or rabies N and P mRNA was determined by RT-
roblastoma SK-N-BE cell lines were grown in RPMI 1640 plus L-glu-
PCR. Total cellular RNA was isolated from cell culture using an RNA kit
tamine (Gibco) supplemented with 15% fetal calf serum (FCS).
according to the manufacturer’s instructions (RNA NOW). Oligo(dT)
The CVS strain of rabies virus was grown in BSR cells. Virus titers were
primer was used for the synthesis of the first strand of cDNA. The
determined by standard plaque assays of BSR cells.
expression of Hsp70 and rabies genes was carried out by the PCR
Antibodies and drugs. The mouse polyclonal anti-P antibody has
amplification of the Hsp70 or rabies N and P genes using the following
been described previously, and the anti-P monoclonal antibodies (MAb)
primers: Hsp70 sense (Hsp70-A), 5=-GCCGGATCCATATGGCCAAAG
25C2 and 30F2 have been previously described (36). The monoclonal
CC-3=; Hsp70 antisense (XhoI-B), 5=-GCCCTCGAGCTAATCTACCTC
anti-N MAb 62B5 was produced in mice immunized with the virus. Rab-
CT-3=; CVS-N sense (N-A), 5=-GCCGGATCCATGGATGCCGACAAG
bit polyclonal anti-P and anti-M antibodies were obtained by the repeated
injection of purified recombinant protein produced in Escherichia coli. A-3=; CVS-N antisense (N-B), 5=-GCCGGATCCTTATGAGTCACTCG
The mouse anti-Hsp70 MAb (SPA-810) and rabbit polyclonal anti-Hsp70 A-3=; CVS-P sense (P-A), 5=-GCCGCTAGCATGAGCAAGATCTTTGT
(SPA-812) antibody were obtained from Stressgen, and the anti-␣-tubu- T-3=; and CVS-P antisense (P-B), 5=-CTCGAGTTAGCAGGATGTATAG
lin MAb (N356) was from Amersham. Quercetin (Qct; Q4951) and the CG-3=. The PCRs were performed using three different volumes of RT
rabbit polyclonal anti-actin (20–33) antibody were obtained from Sigma. mixture (cDNA; 1, 3, and 5 ␮l). The PCR products were electrophoresed
The mouse anti-green fluorescent protein (GFP) MAb (11814460001) in a 1.2% agarose gel and visualized with ethidium bromide.
was purchased from Roche. Northern blot analysis. RNA was isolated from cells with the RNA
Plasmids and siRNA. The plasmids pCDM8-P, encoding the P pro- NOW kit (Ozyme). Total RNA was separated on a 1.5% agarose gel under
tein (CVS strain), and pCDM8-N, encoding the N protein (CVS strain), denaturing conditions and blotted onto nylon membranes (Roche Mo-
have been described previously (6). The plasmid pRL-TK was described lecular Biochemicals). Hybridizations were performed with digoxigenin
elsewhere (11, 13) and was a generous gift of D. Garcin (University of (DIG)-labeled oligonucleotides recognizing the rabies virus (RAV) N or P
Geneva School of Medicine). The plasmids pTit-N, pTit-P, and pTit-L gene sequence and by incubation with anti-DIG antibody conjugated to
were described previously (7) and were obtained from K. K. Conzelmann alkaline phosphatase, followed by CDP Star analysis.
(Ludwig-Maximilians University of Munich). The plasmid pDImut (21) Minireplicon system assay and luciferase assay. The minireplicon
was provided by N. Tordo (Institut Pasteur). The plasmid pCMV-Hsp70, system assay was slightly modified from Le Mercier et al. (21). BSR cells
encoding the Hsp70 protein, and pCMV-Neo were kindly provided by G. were grown in a 12-well plate (3 ⫻ 105 cells per well) in DMEM supple-
Trugnan (2). The siRNA-Hsp70 duplex and scrambled siRNA were pur- mented with 5% FCS and incubated for 24 h at 37°C (in 5% CO2).
chased from Eurogentec. The cells were transfected with pTit-N (1.2 ␮g), pTit-P (1.2 ␮g),
Cell infection and transfection. BSR cells were grown on glass cover- pTit-L (0.3 ␮g), pT7 (1 ␮g; to allow the cytoplasmic expression of the T7
slips in 6-well plates and were infected with rabies virus at various multi- RNA polymerase), pRL-TK (0.3 ␮g), and pDImut (0.75 ␮g) using the
plicities of infection (MOI) and at various times postinfection (p.i.). BSR FuGENE 6 reagent as described by the manufacturer (number
cells grown to 90% confluence in 60-mm-diameter dishes also were trans- 11814443001 from Roche). The N, P, and L proteins and the RNA mini-
fected with 2 ␮g of plasmid using Lipofectamine 2000 as described by the genome formed a functional RNP template resulting in luciferase gene
manufacturer (Invitrogen). transcription, thus the amount of luciferase was related to the transcrip-
Purification of virus and nucleocapsids. Purified virus was obtained tional activity of the reconstituted RNP. The inhibition of Hsp70 expres-
as follows. Virions were pelleted through a cushion of 25% glycerol TNE sion was obtained by siRNA-Hsp70 transfection 48 h before minireplicon
(10 mM Tris, pH 7.5, 1 mM EDTA, 50 mM NaCl) in TD (0.8 mM Tris, pH transfection, and the overexpression of Hsp70 was induced by transfec-
7.4, 150 mM NaCl, 5 mM KCl, 0.7 mm Na2HPO4, 10 mM EDTA) and tion with pCMV-Hsp70 (1.2 ␮g). In the absence of pCMV-Hsp70, all cells
further purified by centrifugation in a sucrose gradient (10 to 40%, wt/ were transfected with pCMV-Neo. Thirty-six hours after the transfection
vol) in TD. of the minireplicon system, the measurement of Firefly and Renilla lucif-
Rabies virus nucleocapsids were isolated from infected BSR cells as erase activities were performed according to the manufacturer’s protocol
described previously (17). (dual-luciferase assay system from Promega). Relative expression levels

4744 jvi.asm.org Journal of Virology


Role of Hsp70 in Rabies Infection

were calculated by dividing the Firefly luciferase values by those of the


Renilla luciferase.
Pulse-chase experiments and immunoprecipitation. At 16 h postin-
fection, the culture medium was replaced with methionine- and cysteine-
free medium, and the culture was incubated for 1 h. The cells then were
labeled with 2 ml of prewarmed medium containing 20 ␮Ci of [35S]
methionine and [35S]cysteine (0.8 Mbq) (express protein labeling mix;
Perkin Elmer) for 5 min. Prewarmed medium containing 5 mM cold
methionine and 1 mM cold cysteine then was added for the chase period.
The cells were washed in cold PBS and lysed on ice in 500 ␮l of buffer
containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 5 mM EDTA, 0.5%
NP-40, and an antiprotease cocktail (2 ␮g of leupeptin per ml, 2 ␮g of
antipain per ml, 2 ␮g pepstatin per ml, 2 ␮g of chymostatin per ml, and 16
␮g of aprotinin per ml). Nuclei were eliminated from the lysate by cen-

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
trifugation at 12,000 ⫻ g for 2 min at 4°C. The cytoplasmic fractions were
incubated for 2 h at 4°C with specific antibodies (anti-P, anti-N, or anti-
M). Immune complexes were precipitated by incubation with protein
A-Sepharose for 1 h at 4°C, washed three times, denatured in Laemmli
buffer, and analyzed by SDS-PAGE. The quantification of radioactivity
was performed by a phosphorimager.
Coimmunoprecipitation. Cells were harvested by being scraped into
cold phosphate-buffered saline (PBS), and cell extracts were prepared as
described above. The cytoplasmic fraction was incubated overnight at 4°C
with specific antibodies (anti-Hsp70 or anti-N). Immune complexes were
precipitated by incubation with protein A-Sepharose for 1 h at 4°C, FIG 1 Hsp70 interacts with the rabies virus N protein. (A) Hsp70 is detected
washed three times, and denatured in Laemmli buffer. Immunoprecipi- in purified virus and in purified nucleocapsid. Purified virus (5 ␮g) and puri-
tated proteins were analyzed by Western immunoblotting using different fied nucleocapsid (5 ␮g), prepared as described in Materials and Methods,
antibodies. were analyzed by SDS-PAGE followed by Coomassie blue staining (lanes 1 and
Western blot analysis. Cells were washed and resuspended in PBS, 4) and Western blotting using a mixture of anti-N, anti-P, and anti-M anti-
lysed in hot Laemmli sample buffer, and boiled for 5 min. About 20 ␮g of bodies (lanes 2 and 5) and a polyclonal anti-Hsp70 (lanes 3 and 6). (B) BSR
protein was analyzed on a 10% SDS-PAGE gel and transferred onto a cells were cotransfected with plasmids expressing Hsp70 and rabies N protein.
nitrocellulose membrane. The proteins were blocked on the membranes Two days after transfection, cells were harvested and lysed. Cell lysate was
incubated with the irrelevant polyclonal anti-GFP antibody (lanes 2 and 6),
with 10% skimmed milk in PTBS for 2 h and incubated overnight at 4°C with the mouse MAb anti-N antibody (lane 3), or with the polyclonal anti-
with rabbit polyclonal anti-Hsp70, anti-P, anti-N, anti-actin, or anti-tu- Hsp70 antibody (lane 5). Immune complexes were analyzed by Western im-
bulin antibody. The blots then were washed extensively in PBS-Tween and munoblotting using anti-Hsp 70 (lanes 2 and 3) and anti-N (lanes 5 and 6)
incubated for 1 h with the appropriate peroxidase-coupled secondary antibodies. Direct cellular extracts of transfected cells (input) also were ana-
antibodies (Amersham). All of the blots then were subjected to chemilu- lyzed by WB with anti-Hsp70 (lane 1) and anti-N (lane 4). Bands correspond-
minescence (ECL; Amersham). Protein spot levels were determined by ing to Hsp70, N, and heavy-chain IgG are indicated.
using ImageJ quantification software.
Immunoaffinity columns and proteomic analysis. (i) Immunoaffin-
ity columns. Monoclonal anti-N antibodies (62B5 and 64B6), which were Research Unit of Biochemistry and Structure Protein, INRA, Jouy en Jo-
previously described (36), were first purified by binding to protein A sas, France).
Sepharose, eluted with 100 mM glycine (pH 3), and immediately read-
justed to neutral pH by adding Tris (pH 8). RESULTS
Second, the purified antibodies were applied on a protein A-Sepharose
Hsp70 interacts with the nucleoprotein. Hsp70 has been shown
column equilibrated in Tris M, pH 8. Protein A was then washed and
resuspended in 0.2 M borate buffer, pH 9.
to be present in rabies virions (37) and within NBL structures
The cross-linking reaction takes place in the presence of 20 mM di- containing the viral L, N, and P proteins and viral RNAs (20, 26).
methyl pimelimidate during 30 min at room temperature under gentle We confirmed here that Hsp70 was also incorporated in purified
rotation. Protein A then is washed and resuspended with 0.2 M ethanol- virions (Fig. 1A, lanes 1 to 3). NBL structures are sites of viral
amine (pH 8) for 2 h. After the removal of ethanolamine, the free anti- transcription and replication (20). To identify the viral partner(s)
bodies (not cross-linked to protein A) were eluted with 100 mM glycine of Hsp70, we first investigated whether Hsp70 protein was present
buffer (pH 3). in the nucleocapsids composed of the viral RNA tightly associated
(ii) Purification of viral and cellular proteins associated with N. Cy- with the N protein. Rabies nucleocapsids were purified from in-
toplasmic fractions of uninfected or infected SK-N-BE cells (1 ⫻ 108 cells fected cells by cesium chloride gradient and analyzed by Western
at an MOI of 10) were loaded separately on the anti-N immunoaffinity blotting with anti-N and anti-Hsp70 antibodies. Hsp70 was found
columns. The columns then were washed extensively with PBS, and the to be associated with the nucleocapsids containing exclusively the
proteins bound to the column were eluted using 100 mM glycine buffer
N protein (Fig. 1A, lanes 4 to 6). This result suggests that Hsp70
(pH 3).
(iii) Analysis of proteins bound to columns of anti-N. The eluates
binds to the N protein.
were resolved on SDS-PAGE, and the proteins were revealed using silver To examine whether Hsp70 interacts with the N protein in
nitrate (silver stain plus kit; Bio-Rad) or colloidal Coomassie blue. Unique rabies-infected cells, two types of experiments were conducted. In
protein bands that had been compared to proper controls were the first one, an immunoaffinity column immobilized with anti-N
trypsinized for proteomic analysis by matrix-assisted laser desorption MAbs was used to identify cellular factors interacting with the N
ionization–time of flight (MALDI-TOF; Platform of Proteomics in the protein. The cytoplasmic fraction of human neuroblastoma SK-

May 2012 Volume 86 Number 9 jvi.asm.org 4745


Lahaye et al.

N-BE-infected cells was loaded on an anti-N immunoaffinity col-


umn. In parallel, uninfected cell extracts or infected cell extracts
incubated without antibodies were used as controls. The proteins
retained on the column were eluted, analyzed by SDS-PAGE, and
identified by MALDI-TOF proteomic analysis after excision from
the gels. Among the cellular proteins, the chaperone Hsp70 was
identified as a cellular partner of the N protein (data not shown).
As expected, the viral phosphoprotein P was also detected as a viral
partner of N. In the second experiment, BSR cells were cotrans-
fected by two plasmids individually expressing Hsp70 and N pro-
tein. Cell extracts were immunoprecipitated with a mouse mono-
clonal anti-N antibody. The proteins present in the immune
complexes then were analyzed by Western blotting with anti-

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
Hsp70 antibodies. Hsp70 was detected in the immunoprecipitates
obtained with the anti-N antibody but not with the unrelated
anti-GFP antibody, indicating the specificity of the interaction
(Fig. 1B, lanes 2 and 3). The reverse experiment showed that the N
protein also could be coimmunoprecipitated with Hsp70 (Fig. 1B,
lane 5). Thus, all of these data indicated that Hsp70 interacts with
the viral N protein in the absence of other viral proteins, and this
supports the idea that this interaction results in the localization of
Hsp70 in NBL structures (20).
Rabies infection induces Hsp70 expression. We then investi-
gated the effect of viral infection on the expression of Hsp70. In-
fected cells were analyzed at different times postinfection (p.i.) by
Western blotting. Hsp70 was detected at early times postinfection
(4 h p.i.), whereas no detectable trace was observed in uninfected
cells (Fig. 2A). The Hsp70 level was enhanced during viral infec-
tion (Fig. 2A). This was confirmed by the confocal immunofluo-
rescence analysis of infected cells, which showed that Hsp70 levels
FIG 2 Expression of Hsp70 during rabies virus infection. (A) BSR cells were
increased in the NBL structures concomitantly with those of viral uninfected (Ni) or infected at an MOI of 1. At different times p.i. as indicated,
N and P proteins (Fig. 2B, 16 h p.i.) before spreading throughout cell extracts were analyzed by Western blotting using anti-Hsp70, anti-N, an-
the cytoplasm (Fig. 2B, 24 h p.i.). These results indicated that ti-P, and anti-actin antibodies. (B) BSR cells were infected as described for
Hsp70 is induced by rabies infection. panel A. Double-immunofluorescence staining was performed with anti-
Hsp70 and anti-P antibodies. The scale bars correspond to 20 ␮m.
Modulation of cellular Hsp70 and effect on rabies virus in-
fection. It is not known whether the induction of Hsp70 expres-
sion is the result of a cellular stress response due to viral infection
or if the protein is induced specifically by some viral mechanism with the progression of infection (Fig. 4A, 16 to 20 h p.i.). In
because it would play a role during the viral life cycle. To analyze contrast, the inhibition of Hsp70 (Qct) resulted in a drastic reduc-
the function of this Hsp70 induction during rabies infection, dif- tion of the level of P protein at 12 and 16 h p.i. (factors of 5 and 20,
ferent experiments were performed to modulate the intracellular respectively). The accumulation of the other viral proteins was
level of Hsp70. also inhibited (data not shown). This inhibition was not due to
First, we determined the conditions to modulate the Hsp70 drug toxicity, as the number of infected BSR cells obtained by
expression in noninfected cells. To induce the expression of counting the fluorescent cells was unchanged in the absence of
Hsp70 protein, BSR cells were subjected to heat shock (HS) for 20 Hsp70 (data not shown). This drastic effect on the viral protein
min at 45°C, and then the cells were reincubated at 37°C for dif- level was attenuated at later stages of infection concomitantly with
ferent times. The Western blotting indicated that the induction of the induced Hsp70 expression (Fig. 4A). To investigate whether
the expression of Hsp70 was maximal at 6 h postshock and was the inhibition of viral protein accumulation was due to the inhi-
maintained thereafter (Fig. 3A). In contrast, treatment with an bition of viral protein translation or to an increase of viral protein
inhibitor of Hsp synthesis, quercetin (Qct), before HS inhibited degradation, we analyzed the effect of Qct on viral translation by
the synthesis of Hsp70 proteins. The application of 100 ␮M Qct pulse-chase experiments with [35S]methionine/cysteine. Infected
resulted in a drastic inhibition of Hsp70 expression (Fig. 3B) with- cells treated with Qct were labeled for 5 min and chased for peri-
out causing visible cell damage to the cell (data not shown). Thus, ods of up to 60 min. Cells then were lysed and immunoprecipi-
the following experiments were performed at 6 h after HS or with tated with anti-N and anti-P or anti-M antibodies. The immuno-
100 ␮M Qct to induce or inhibit Hsp70 expression, respectively. precipitates were analyzed by SDS-PAGE. Treatment with Qct
We first analyzed the effect of the modulation of Hsp70 on the dramatically affected viral protein synthesis (Fig. 4B). To analyze
synthesis of viral proteins during viral infection (Fig. 4A and B). the effect of Qct on cellular protein synthesis, mock-infected cells
When Hsp70 was overexpressed (HS), we observed an increase of left untreated or treated with Qct were labeled for 5 min and an-
P protein synthesis (factor of 4 at 12 h p.i.) compared to control alyzed directly by SDS-PAGE. In contrast to viral proteins, cellular
cells (Fig. 4A), although this increase became less pronounced protein synthesis did not seem to be affected (Fig. 4B, lower, lanes

4746 jvi.asm.org Journal of Virology


Role of Hsp70 in Rabies Infection

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
FIG 3 Modulation of Hsp70 expression in the absence of infection. (A) Uninfected BSR cells were incubated for 20 min at 45°C and then at 37°C for various times
after heat shock (HS) as indicated. Cell extracts were analyzed by Western blotting using anti-Hsp70 and anti-tubulin antibodies (used as a loading control). The
level of Hsp70 was quantified by immunoblot scanning and normalized with respect to the amount of tubulin (lower panel). (B) Uninfected BSR cells were
untreated (0) or were treated with different concentrations of quercetin (Qct) (as indicated) for 1 h before HS at 45°C for 20 min in the presence of quercetin.
After HS, the cells were incubated at 37°C for 6 h. Cell extracts were analyzed by Western blotting using anti-Hsp70 and anti-tubulin antibodies. The level of
Hsp70 was quantified by immunoblot scanning and normalized with respect to the amount of tubulin (lower panel).

17 and 18). This indicates that the drug led to the inhibition of lanes 4 and 5 versus lane 3, and B). The decrease of the amount of
viral protein translation but did not affect protein stability. viral N and P proteins was maintained in HS conditions between a
We next studied the effect of Hsp70 on viral production. Sim- factor of 2 to 4 depending on siRNA-Hsp70 quantities (Fig. 6A,
ilar experiments were performed and supernatants were used for lanes 9 and 10 versus lane 8, and B). As expected, the N and P
viral titration (Fig. 5A and B). When Hsp70 was overexpressed protein levels were similar in the presence of scrambled siRNA
(HS), viral production at 16 h p.i. was slightly higher (factor 2) (Fig. 6A, lanes 2 and 3 versus 7 and 8, and B). These results con-
than that in normal conditions (Fig. 5A and B). In contrast, the firmed that the specific inhibition of Hsp70 synthesis led to a re-
inhibition of Hsp70 (with Qct) induced a dose-dependent inhibi- duction of viral protein levels and indicated that Hsp70 has a
tion of the viral production during the viral cycle (Fig. 5A). Viral proviral role.
yield was 10- to 20-fold and up to 100-fold lower than that in The effect of siRNA-Hsp70 on viral production was also ana-
control cells in the presence of 100 and 200 mM Qct, respectively lyzed in the same conditions as those described above. The siRNA-
(Fig. 5A and B). These data indicated that the reduction of Hsp70 Hsp70 treatment resulted in an inhibition of the viral production
significantly impairs viral protein synthesis and virus production, of 10- to 15-fold compared to control and scrambled siRNA,
suggesting that Hsp70 has a proviral effect during rabies infection. whatever the dose of siRNA and conditions (Fig. 6C). These re-
However, the overexpression of Hsp70 in infected cells has at most sults confirmed that Hsp70 plays a positive role in rabies infection.
a small effect on viral life cycle. This could be explained by the fact Effect of Hsp70 on viral RNA synthesis. We next tested
that Hsp70 is efficiently induced by rabies infection, and this in- whether the inhibition of viral protein synthesis (observed in the
duction would be sufficient to support the viral life cycle. presence of Qct or in siRNA experiments) could reflect an inhibi-
Specific inhibition of Hsp70 expression by siRNA and effect tion of viral transcription. First, we analyzed the effect of Qct on
on viral infection. As quercetin is not a specific inhibitor of Hsp70 viral mRNA synthesis. Total RNAs were extracted from untreated
but inhibits the synthesis of other heat shock proteins, we per- infected cells, cells treated with Qct, or cells submitted to HS and
formed silencing experiments to investigate more specifically the were analyzed for the presence of the viral N mRNA by Northern
function of Hsp70 during rabies viral infection. BSR cells were blot analysis. In the presence of Qct, the amount of N mRNA was
transfected with two different concentrations of Hsp70 siRNA reduced by a factor 5 (Fig. 7A), indicating that Qct impaired viral
(200 and 400 pmol) for 48 h, and then cells were infected under transcription. However, the induction of Hsp70 expression (HS)
normal (control) or heat shock conditions. Hsp70 was efficiently did not result in an increase of the amount of rabies N mRNA
downregulated, as shown in HS-treated cells (Fig. 6A, lanes 9 and (Fig. 7A).
10) compared to control and scrambled siRNA (Fig. 6A, lanes 6 to We then tried to confirm these effects on viral RNA synthesis in
8). In the absence of HS, the induction of Hsp70 by viral infection the presence of a more specific Hsp70 depletion. As Northern
(Fig. 6A, lanes 2 and 3) was also inhibited after Hsp70 silencing blotting is not the most sensitive method, we first used RT-PCR to
(Fig. 6A, lanes 4 and 5). These observations were confirmed by the detect the viral mRNA of N and P proteins and the mRNA of
quantification of the cellular Hsp70 protein level (Fig. 6B). Under Hsp70 in infected cells after Hsp70 silencing experiments and in
these conditions, the knockdown of Hsp70 expression was associ- cells infected under normal (control) or heat shock (HS) condi-
ated with a significant reduction of the amounts of viral N and P tions (Fig. 7B). Increasing amounts of RT reactions (1, 3, and 5 ␮l)
proteins of 3- to 8-fold depending on the siRNA dose (Fig. 6A, were used for PCR amplification. As expected, HS or/and rabies

May 2012 Volume 86 Number 9 jvi.asm.org 4747


Lahaye et al.

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
FIG 4 Effect of modulation of Hsp70 on viral protein synthesis. (A) BSR cells were left untreated (DMSO) or were treated with quercetin (Qct; 100 ␮M) for 1
h and infected (MOI of 1) in the presence of Qct, or cells were submitted to heat shock (HS) (after 1 h of DMSO treatment) and infected (MOI of 1) 6 h after HS.
Cells were lysed at different times p.i. as indicated, and cell extracts were analyzed by Western blotting using anti-Hsp70, anti-P, and anti-tubulin antibodies. The
levels of P and Hsp70 proteins present in different conditions (DMSO, Qct, and HS) were quantified by Western blot scanning and normalized with respect to
the amount of tubulin (expressed in logarithmic scale) (lower panel). An arbitrary level of 100 is applied to P and Hsp70 levels (*) in a classical infection (DMSO)
for each hour (p.i.) to allow comparisons. (B) BSR cells were left untreated (⫺) or were treated with 100 ␮M Qct (⫹) for 1 h and infected (MOI of 1) in the
presence of Qct. After 16 h p.i., cell were pulse labeled with [35S]methionine and [35S]cysteine for 5 min and then chased in cold medium for the times indicated
before lysis. The lysates were immunoprecipitated with anti-N and anti-P antibodies (upper panel) or anti-M antibodies (lower panel). The immunoprecipitates
were analyzed by SDS-PAGE followed by autoradiography. Mock-infected cells were labeled for 5 min (without chase), and cell extracts (CE) were directly
analyzed by SDS-PAGE followed by autoradiography (lower panel, lanes 17 and 18).

virus infection induced the expression of Hsp70 mRNA (Fig. 7A, (Fig. 7C). Taken together, these data suggested that Hsp70 plays a
lanes 1 to 3 and 7 to 9) and Hsp70 was efficiently downregulated in minor role even in viral transcription. However, whether this role
the presence of siRNA-Hsp70 compared to control and scrambled is sufficient to explain further effects on viral protein synthesis and
siRNA (Fig. 7B, lanes 4 to 6 and 10 to 12). Hsp70 induction re- viral replication should be further investigated.
sulted in a slight increase of the amount of viral mRNA of N and P
(Fig. 7B, lanes 7, 8, and 9 versus lanes 1, 2, and 3); however, Hsp70 DISCUSSION
silencing did not result in a decrease of the amount of viral mRNA In this study, we have investigated the role of Hsp70 during rabies
by this method (Fig. 7B, lanes 10, 11, and 12 versus lanes 4, 5, virus infection. Coimmunoprecipitation and proteomic analyses
and 6). indicated that Hsp70 interacts with N. However, we cannot ex-
We then used a more quantitative assay, the luciferase reporter clude that this interaction is indirect and mediated by one or more
system in a minireplicon system, that allows the reconstitution of other factors. Whatever the case, this interaction probably results
a functional rabies virus RNP. The luciferase gene reporter is in the presence of Hsp70 in purified nucleocapsids, within NBL
framed by 3= leader and 5= trailer sequences and is under the con- structures, and in purified rabies particles. In addition, we have
trol of rabies transcription signals (21). The production of lucif- shown that rabies infection induces an increase of cellular Hsp70
erase activity is the result of the encapsidation of the minigenome levels as soon as 4 h p.i. in BSR cells and in neuroblastoma cells
followed by its transcription by the L-P complex and by its repli- (data not shown). The increase in Hsp70 levels following viral
cation in the presence of N protein. The siRNA-Hsp70-treated infection has been widely observed. This can be explained by dif-
BSR cells were cotransfected with plasmids encoding L, N, and P ferent mechanisms. The induction may occur indirectly through
proteins and the minigenome. As expected, luciferase activity the production of a large number of unfolded proteins during
background was obtained in the absence of the L protein (Fig. 7C). viral infection. Second, the viruses could interfere with stress sig-
The overexpression of Hsp70 induced a slight stimulation of the nal transduction pathways independently of the folding status of
luciferase activity (to about 20%) compared to that of the control proteins. Indeed, Hsp70, which is involved in many cellular pro-
(Fig. 7C). Similarly, the Hsp70 silencing weakly reduced the lucif- cesses, such as cell cycle, apoptosis, and cellular innate immunity
erase activity (to about 20 to 40%) in a dose-dependent manner. pathways, is frequently diverted by viruses for their benefit (23,
We ensured that the amounts of viral N and P proteins expressed 25). In the case of rabies virus, Hsp70 enrichment might simply be
from the plasmids were unchanged in the different conditions correlated with excess amounts of viral proteins (particularly N

4748 jvi.asm.org Journal of Virology


Role of Hsp70 in Rabies Infection

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
FIG 5 Effect of Qct treatment on viral production. BSR cells were infected at
an MOI of 1 without any treatment (DMSO) or in the presence of Qct as
indicated or were subjected to HS treatments as described in the text. Culture
media were collected at 16 h p.i. (A) or at different times pi (B), and viral titers
were determined as described in Materials and Methods. P values (**, P ⬍
0.001; ***, P ⬍ 0.0001) were calculated using the Student’s test.

and P); nevertheless, the recruitment of Hsp70 inside the NBLs,


which are sites of viral transcription and replication, in the puri-
fied nucleocapsids and in the purified virions suggests that Hsp70 FIG 6 Effect of specific inhibition of Hsp70 by siRNA interference on rabies
plays an active role in the viral life cycle. replication. (A) BSR cells were not transfected (Control) or were transfected
with scrambled siRNA (Scr) or siRNA-Hsp70 (Hsp) (two concentrations as
The downregulation of Hsp70, using specific chaperons inhib-
indicated). After 48 h, cells were submitted to HS (⫹HS) or not (⫺HS), and 6
itors such as quercetin or RNA interference, significantly reduced h later cells were infected at an MOI of 3 for 16 h. Cell extracts were analyzed by
the intracellular level of viral proteins and viral production. The Western blotting using anti-Hsp70, anti-N, anti-P, and anti-tubulin antibod-
reduction of viral protein accumulation appeared to be the result ies. (B) Quantitative analysis of cellular P, N, and Hsp70 protein levels in cells
of an inhibition of viral translation more than protein instability, treated as described for panel A. The resulting Western blots (three indepen-
dent experiments) were quantified using immunoblot scanning and normal-
as shown by pulse-chase labeling (in the presence of Qct). Viral ized with respect to the amount of tubulin. The amounts of N (black histo-
transcription was also decreased. Interestingly, the inhibition at grams), P (gray histograms), and Hsp70 (white histograms) were measured,
different steps of the viral cycle (transcription, translation, and and an arbitrary level of 100 is applied to N, P, and Hsp70 levels in a classical
viral production) was more drastic in the presence of Qct than infection (lane 2 of panel A) for comparisons. P values (***, P ⬍ 0.0001) were
calculated using the Student’s test. (C). Culture media of infected cells treated
siRNA, suggesting that other chaperones are involved in the rabies
as described for panel A were collected at 16 h p.i., and viral titers were deter-
virus infection cycle. Unexpectedly, the overexpression of Hsp70 mined as described in Materials and Methods. Viral titers represent averages
following heat shock treatment or pCMV-Hsp70 transfection re- from three independent experiments. Error bars indicate the standard devia-
sulted in a slight increase of viral protein synthesis or viral pro- tions.
duction. This is likely because infected cells already expressed a
sufficiently high level of Hsp70, and the overexpression before the
infection did not have much effect. The siRNA-induced reduction tion. Indeed, the resulting inhibition in viral protein synthesis and
of viral yield may be due to a decrease of the amount of viral in viral production could be a direct consequence of the viral
particles released, but it could well be related to the reduced infec- transcription inhibition, but it could also be amplified by an
tivity of the progeny virions lacking Hsp70. Hsp70-specific role in these further steps of the viral infection.
All of these data indicate that Hsp70 has a proviral effect on Interestingly, the potential role of Hsp70 in viral RNA synthe-
rabies virus replication by acting during at least one step of the sis is consistent with the interaction of Hsp70 with N and its asso-
viral cycle. As these steps are connected, it is difficult to clearly ciation with the NBLs, which are sites of viral transcription and
determine the mechanism by which Hsp 70 controls viral infec- replication, suggesting a possible interaction with the polymerase

May 2012 Volume 86 Number 9 jvi.asm.org 4749


Lahaye et al.

complex. Indeed, previous studies of three paramyxoviruses, mea-


sles virus, canine distemper virus, and respiratory syncytial virus,
have provided evidence for an interaction between the N protein
and Hsp70 (3, 29, 30). In these cases, Hsp70 has been shown to
stimulate viral polymerase in vitro activity, but it is not clear
whether Hsp70 acts directly as a cofactor of the enzymatic process
or whether it assists the folding of the N protein in an active form.
Interestingly, the positive effect of Hsp70 on viral infection is not
a common property of negative-strand RNA viruses belonging to
the same order, highlighting the complexity of the virus-Hsp con-
nections.
Other chaperones could play a role in rabies viral RNA synthe-
sis. For example, Hsp60 has been detected in the transcriptase

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
complex of VSV, although its function has not been investigated
so far (35), and Hsp40 has been shown to enhance the binding of
Hsp70 with the N protein of measles virus (8). More recently, it
has been reported that Hsp70 is associated with complexes formed
between Hsp40 and the viral protein Nef in HIV-1-infected cells
and that the differential expression of Hsp40 and Hsp70 recipro-
cally regulates viral gene expression and replication (19).
In conclusion, these data support a positive regulatory role for
Hsp70 in the rabies virus infection cycle and suggest that Hsp70,
consistently with its multifunctional role, participates at the dif-
ferent stages of the viral life cycle, such as the transcriptional
and/or translational level and/or viral assembly and budding.
ACKNOWLEDGMENTS
We are grateful to Yves Gaudin for careful reading of the manuscript. We
acknowledge Noël Tordo (Unit Antiviral Strategies, Institut Pasteur,
France) for providing the rabies minireplicon system. We thank the Plat-
form of Proteomics (Unités de Recherches de Biochimie et Structures des
Protéines, Jouy en Josas, France). Confocal microscopy was performed at
the Plate-forme Imagerie et Biologie Cellulaire of the CNRS campus,
which is supported by the Institut Fédératif de Recherche 87 La Plante et
Son Environnement and the ASTRE program of the Conseil Général de
l’Essonne.
We acknowledge support from the CNRS.
FIG 7 Effect of Hsp70 on viral RNA synthesis. (A) Effect of Quercetin on viral
RNA by Northern blotting. BSR cells were left untreated (C) or were treated with REFERENCES
quercetin (Qct; 100 ␮M) for 1 h and infected (MOI of 1) in the presence of Qct, or
cells were submitted to heat shock (HS) (after 1 h of DMSO treatment) and in- 1. Albertini AA, Ruigrok RW, Blondel D. 2011. Rabies virus transcription
fected (MOI of 1) 6 h after HS. Total RNA was extracted from cells, and samples and replication. Adv. Virus Res. 79:1–22.
(10 ␮g of RNA per lane) were analyzed for the presence of rabies N mRNA and 2. Broquet AH, et al. 2007. Hsp70 negatively controls rotavirus protein
GAPDH (glyceraldehyde-3-phosphate dehydrogenase) mRNA, as described in bioavailability in caco-2 cells infected by the rotavirus RF strain. J. Virol.
Materials and Methods. (B) Detection of viral RNA by RT-PCR. BSR cells were left 81:1297–1304.
untreated (Control) or were treated with scrambled siRNA or siRNA-Hsp70 (two 3. Brown G, et al. 2005. Evidence for an association between heat shock
concentrations as indicated). After 48 h, cells were submitted to HS (⫹) or not protein 70 and the respiratory syncytial virus polymerase complex within
(⫺), and 6 h later cells were infected by CVS at an MOI of 3 for 16 h and total RNA lipid-raft membranes during virus infection. Virology 338:69 – 80.
was extracted. The presence of Hsp70 mRNA or rabies N and P mRNA was deter- 4. Bukau B, Deuerling E, Pfund C, Craig EA. 2000. Getting newly synthe-
mined by RT-PCR as described in Materials and Methods. The PCRs were per- sized proteins into shape. Cell 101:119 –122.
formed using three different volumes of RT mixture (cDNA; 1, 3, and 5 ␮l) and 5. Carsillo T, Traylor Z, Choi C, Niewiesk S, Oglesbee M. 2006. hsp72, a
were analyzed on a 1.2% agarose gel with ethidium bromide. (C) Luciferase ex- host determinant of measles virus neurovirulence. J. Virol. 80:11031–
pression from a rabies minireplicon system. BSR cells were left untreated (control), 11039.
were treated with scrambled siRNA or siRNA-Hsp70 (different concentrations), 6. Chenik M, Chebli K, Gaudin Y, Blondel D. 1994. In vivo interaction of
or were transfected by a plasmid encoding Hsp70 (pCMV-Hsp70). After 48 h, cells rabies virus phosphoprotein (P) and nucleoprotein (N): existence of two
were transfected with plasmids encoding L, N, and P proteins, T7 polymerase, and N-binding sites on P protein. J. Gen. Virol. 75:2889 –2896.
the plasmid (pRL-TK) encoding Renilla luciferase in the presence of a rabies mini- 7. Conzelmann KK. 1998. Nonsegmented negative-strand RNA viruses: ge-
replicon system plasmid for the expression of Firefly luciferase. The L plasmid was netics and manipulation of viral genomes. Annu. Rev. Genet. 32:123–162.
missing from one experiment, and this served as a negative control (without L). 8. Couturier M, et al. 2010. High affinity binding between Hsp70 and the
After 36 h, cells extracts were analyzed for luciferase expression. Data represent the C-terminal domain of the measles virus nucleoprotein requires an Hsp40
separate assays for Firefly luciferase normalized to the expression of Renilla lucif- co-chaperone. J. Mol. Recognit. 23:301–315.
erase and are expressed as percentages of the control (*). Error bars indicate stan- 9. De Marco A, Santoro MG. 1993. Antiviral effect of short hyperthermic
dard deviations (from three independent experiments). In the bottom panel of B, treatment at specific stages of vesicular stomatitis virus replication cycle. J.
the expression of N, P, and tubulin (Tub) present in the cell extracts of control (C), Gen. Virol. 74:1685–1690.
scramble (Sc), and siRNA Hsp was analyzed by Western blot analysis with anti-N, 10. Diaz-Latoud C, et al. 1997. Herpes simplex virus Us11 protein enhances
anti-P, and anti-tubulin antibodies. recovery of protein synthesis and survival in heat shock treated HeLa cells.
Cell Stress Chaperones 2:119 –131.

4750 jvi.asm.org Journal of Virology


Role of Hsp70 in Rabies Infection

11. Didcock L, Young DF, Goodbourn S, Randall RE. 1999. The V protein formation of nonestablished rat embryo fibroblasts. J. Virol. 61:2648 –
of simian virus 5 inhibits interferon signaling by targeting STAT1 for 2654.
proteasome-mediated degradation. J. Virol. 73:9928 –9933. 28. Nagy PD, Wang RY, Pogany J, Hafren A, Makinen K. 2011. Emerging
12. Elbashir SM, Harborth J, Weber K, Tuschl T. 2002. Analysis of gene picture of host chaperone and cyclophilin roles in RNA virus replication.
function in somatic mammalian cells using small interfering RNAs. Meth- Virology 411:374 –382.
ods 26:199 –213. 29. Oglesbee M, Ringler S, Krakowka S. 1990. Interaction of canine distem-
13. Garcin D, Curran J, Kolakofsky D. 2000. Sendai virus C proteins must per virus nucleocapsid variants with 70K heat-shock proteins. J. Gen. Vi-
interact directly with cellular components to interfere with interferon ac- rol. 71:1585–1590.
tion. J. Virol. 74:8823– 8830. 30. Oglesbee MJ, Kenney H, Kenney T, Krakowka S. 1993. Enhanced
14. Gaudin Y. 2000. Rabies virus-induced membrane fusion pathway. J. Cell production of morbillivirus gene-specific RNAs following induction of
Biol. 150:601– 612. the cellular stress response in stable persistent infection. Virology 192:
15. Hartl FU, Hayer-Hartl M. 2002. Molecular chaperones in the cytosol: 556 –567.
from nascent chain to folded protein. Science 295:1852–1858. 31. Omar RA, Lanks KW. 1984. Heat shock protein synthesis and cell survival
16. Hirayama E, Atagi H, Hiraki A, Kim J. 2004. Heat shock protein 70 is in clones of normal and simian virus 40-transformed mouse embryo cells.
related to thermal inhibition of nuclear export of the influenza virus ribo- Cancer Res. 44:3976 –3982.

Downloaded from http://jvi.asm.org/ on May 11, 2014 by UCSF Library & CKM
nucleoprotein complex. J. Virol. 78:1263–1270. 32. Peluso RW, Moyer SA. 1988. Viral proteins required for the in vitro
17. Iseni F, Barge A, Baudin F, Blondel D, Ruigrok RW. 1998. Character- replication of vesicular stomatitis virus defective interfering particle ge-
ization of rabies virus nucleocapsids and recombinant nucleocapsid-like nome RNA. Virology 162:369 –376.
structures. J. Gen. Virol. 79:2909 –2919. 33. Phillips B, Abravaya K, Morimoto RI. 1991. Analysis of the specificity
and mechanism of transcriptional activation of the human hsp70 gene
18. Jindal S, Young RA. 1992. Vaccinia virus infection induces a stress re-
during infection by DNA viruses. J. Virol. 65:5680 –5692.
sponse that leads to association of Hsp70 with viral proteins. J. Virol.
34. Pratt WB, Toft DO. 2003. Regulation of signaling protein function and
66:5357–5362.
trafficking by the hsp90/hsp70-based chaperone machinery. Exp. Biol.
19. Kumar M, et al. 2011. Reciprocal regulation of human immunodeficiency
Med. 228:111–133.
virus-1 gene expression and replication by heat shock proteins 40 and 70.
35. Qanungo KR, Shaji D, Mathur M, Banerjee AK. 2004. Two RNA poly-
J. Mol. Biol. 410:944 –958. merase complexes from vesicular stomatitis virus-infected cells that carry
20. Lahaye X, et al. 2009. Functional characterization of Negri bodies (NBs) out transcription and replication of genome RNA. Proc. Natl. Acad. Sci.
in rabies virus infected cells: evidence that NBs are sites of viral transcrip- U. S. A. 101:5952–5957.
tion and replication. J. Virol. 83:7948 –7958. 36. Raux H, Iseni F, Lafay F, Blondel D. 1997. Mapping of monoclonal
21. Le Mercier P, Jacob Y, Tanner K, Tordo N. 2002. A novel expression antibody epitopes of the rabies virus P protein. J. Gen. Virol. 78:119 –124.
cassette of lyssavirus shows that the distantly related Mokola virus can 37. Sagara J, Kawai A. 1992. Identification of heat shock protein 70 in the
rescue a defective rabies virus genome. J. Virol. 76:2024 –2027. rabies virion. Virology 190:845– 848.
22. Li G, Zhang J, Tong X, Liu W, Ye X. 2011. Heat shock protein 70 inhibits 38. Santoro MG. 1994. Heat shock proteins and virus replication: hsp70s as
the activity of influenza a virus ribonucleoprotein and blocks the replica- mediators of the antiviral effects of prostaglandins. Experientia 50:1039 –
tion of virus in vitro and in vivo. PLoS One 6:e16546. 1047.
23. Mayer MP. 2010. Gymnastics of molecular chaperones. Mol. Cell 39:321– 39. Weeks SA, et al. 2010. A targeted analysis of cellular chaperones reveals
331. contrasting roles for heat shock protein 70 in flock house virus RNA rep-
24. Mayer MP. 2005. Recruitment of Hsp70 chaperones: a crucial part of viral lication. J. Virol. 84:330 –339.
survival strategies. Rev. Physiol. Biochem. Pharmacol. 153:1– 46. 40. Young JC, Barral JM, Ulrich Hartl F. 2003. More than folding: localized
25. Mayer MP, Bukau B. 2005. Hsp70 chaperones: cellular functions and functions of cytosolic chaperones. Trends Biochem. Sci. 28:541–547.
molecular mechanism. Cell Mol. Life Sci. 62:670 – 684. 41. Zeng R, Zhang Z, Mei X, Gong W, Wei L. 2008. Protective effect of a
26. Menager P, et al. 2009. Toll-like receptor 3 (TLR3) plays a major role in RSV subunit vaccine candidate G1F/M2 was enhanced by a HSP70-Like
the formation of rabies virus Negri bodies. PLoS Pathog. 5:e1000315. protein in mice. Biochem. Biophys. Res. Commun. 377:495– 499.
27. Michalovitz D, Fischer-Fantuzzi L, Vesco C, Pipas JM, Oren M. 1987. 42. Zhang X, et al. 2005. Hsp72 recognizes a P binding motif in the measles
Activated Ha-ras can cooperate with defective simian virus 40 in the trans- virus N protein C-terminus. Virology 337:162–174.

May 2012 Volume 86 Number 9 jvi.asm.org 4751

You might also like