IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

41
IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING TO THE PHOSPHORYLATION OF INTERFERON REGULATORY FACTOR 3 Marc. J. Servant 1,2 , Benjamin ten Oever 1,2 , Cecile LePage 1,2 , Lucia Conti 5 , Sandra Gessani 5 , Ilkka Julkunen 4 , Rongtuan Lin 1,2 and John Hiscott 1-3* Terry Fox Molecular Oncology Group, Lady Davis Institute for Medical Research 1 , and Departments of Microbiology & Immunology 3 and Medicine 2 , McGill University, Montreal, Canada H3T 1E2 and National Public Health Institute, Department of Virology, Mannerheimintie 166, FIN-00300 Helsinki, Finland 4 And Istituto Superiore di Sanita 5 00161 Rome, Italy Suggested running title: Distinct pathways leading to IRF-3 phosphorylation * To whom reprint requests should be addressed at: Dr. John Hiscott Lady Davis Institute for Medical Research 3755 Cote Ste. Catherine Montreal, Quebec, Canada H3T 1E2 Telephone: (514) 340-8222 Ext. 5265 Fax: (514) 340-7576 E-mail: [email protected] Copyright 2000 by The American Society for Biochemistry and Molecular Biology, Inc. JBC Papers in Press. Published on October 16, 2000 as Manuscript M007790200 by guest on March 22, 2018 http://www.jbc.org/ Downloaded from

Transcript of IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

Page 1: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING TO THEPHOSPHORYLATION OF INTERFERON REGULATORY FACTOR 3

Marc. J. Servant1,2, Benjamin ten Oever1,2, Cecile LePage1,2, Lucia Conti5, Sandra Gessani5,Ilkka Julkunen4, Rongtuan Lin1,2 and John Hiscott1-3*

Terry Fox Molecular Oncology Group, Lady Davis Institute for Medical Research1, andDepartments of Microbiology & Immunology3 and Medicine2,

McGill University, Montreal, Canada H3T 1E2and

National Public Health Institute, Department of Virology, Mannerheimintie 166, FIN-00300Helsinki, Finland4

AndIstituto Superiore di Sanita5

00161 Rome, Italy

Suggested running title: Distinct pathways leading to IRF-3 phosphorylation

* To whom reprint requests should be addressed at:

Dr. John HiscottLady Davis Institute for Medical Research3755 Cote Ste. CatherineMontreal, Quebec, Canada H3T 1E2Telephone: (514) 340-8222 Ext. 5265Fax: (514) 340-7576E-mail: [email protected]

Copyright 2000 by The American Society for Biochemistry and Molecular Biology, Inc.

JBC Papers in Press. Published on October 16, 2000 as Manuscript M007790200 by guest on M

arch 22, 2018http://w

ww

.jbc.org/D

ownloaded from

Page 2: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

2

ABSTRACT

Infection of host cells by viruses leads to the activation of multiple signaling pathways, resulting in

the expression of host genes involved in the establishment of the antiviral state. Among the

transcription factors mediating the immediate response to virus is interferon regulatory factor-3

(IRF-3) which is posttranslationally modified as a result of virus infection. Phosphorylation of

latent cytoplasmic IRF-3 on serine and threonine residues in the C-terminal region leads to

dimerization, cytoplasmic to nuclear translocation, association with the p300/CBP coactivator and

stimulation of DNA binding and transcriptional activities. We now demonstrate that IRF-3 is a

phosphoprotein that is uniquely activated via virus dependent C-terminal phosphorylation.

Paramyxoviridae including measles virus and rhabdoviridae – vesicular stomatitis virus – are potent

inducers of a unique virus-activated kinase activity. In contrast, stress-inducers, growth factors,

DNA-damaging agents and cytokines do not induce C-terminal IRF-3 phosphorylation,

translocation or transactivation, but rather activate a MAPKKK-related signaling pathway that

results in N-terminal IRF-3 phosphorylation. The failure of numerous well-characterized

pharmacological inhibitors to abrogate virus-induced IRF-3 phosphorylation suggests the

involvement of a novel kinase activity in IRF-3 regulation by viruses.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

3

INTRODUCTION

Virus infection of mammalian cells triggers multiple signal transduction cascades involved in the

activation of a diverse set of immunoregulatory genes and proteins that together create the antiviral

state, an intracellular environment that antagonizes virus replication. The type I interferon (IFN)1

family is essential to the development of the antiviral state and the IFN gene family represents one

of the best characterized models of virus inducible gene activation (1). Once produced, these

secreted proteins induce gene expression in neighbouring cells through cell surface cytokine

receptors and the JAK-STAT signaling pathways. STAT1/2 heterodimers, in conjunction with

interferon-stimulated gene factor 3γ (ISGF3γ) bind to interferon-stimulated response elements

(ISRE) found in numerous IFN-induced genes such as 2'-5' oligoadenylate synthase and the

double stranded RNA (dsRNA) activated kinase (PKR), resulting in the induction of proteins

which impair viral gene expression and replication (1). Molecular regulation of IFN gene

transcription is tightly regulated by extra- and intracellular signals induced at the site of infection.

One of the best characterized models of such regulation is the virus-inducible promoter/enhancer of

the IFN-β gene (2-4). This promoter includes an overlapping set of regulatory elements designated

positive regulatory domains (PRDs) I to IV, which interact with several signal-responsive

transcription factors including NF-κB (p50-p65), ATF-2/c-Jun heterodimers and interferon

regulatory factors (IRF) that bind to PRD II, PRD IV and PRD I-III, respectively. Together with

the chromatin associated HMG I(Y) proteins, these transcription factors form a stereospecific

transcriptional enhancer complex, termed the enhanceosome (2-4) that stimulates the high level,

transient activation of IFN-β transcription.

The pathways involved in NF-κB and ATF-2/c-Jun activation have been well characterized.

Following viral infection, treatment with proinflamatory stimuli like tumor necrosis factor (TNF)-

α, interleukin-1 (IL-I), or exposure to dsRNA, these transcriptions factors are activated through

stimulation of distinct kinase cascades. In nonstimulated cells, the NF-κB factors are retained in the

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

4

cytoplasm in association with inhibitory subunits - IκBs; virus-induced phosphorylation at

conserved N-terminal residues is accomplished by the IκB kinase (IKK) complex. Phosphorylation

triggers a signal that induces ubiquitin-dependent degradation of IκB, and subsequent nuclear

translocation of the NF-κB dimers (reviewed in (5)). The rate limiting step in this process is the

activation of IKK which is composed of two catalytic subunits IKKα and β and one regulatory

subunit IKKγ/NEMO. Numerous studies now suggest that the IKKβ catalytic subunit is required

for IKK and NF-κB activation by TNF-α, IL-I, lipopolysaccharide (LPS), dsRNA and viral

infection (6-10). Unlike NF-κB, the heterodimers ATF-2/c-Jun are expressed as nuclear proteins

that are activated by phosphorylation of their activation domains by c-Jun amino terminal kinases

(JNKs) which are downstream of a well defined stress-activated kinase cascade (11)

The pathway(s) regulating IRF-3 phosphorylation and activation are also the focus of considerable

investigation. IRF-3 belongs to the family of IRFs which include IRF-1 to IRF-7, interferon

consensus sequence-binding protein (IRF-8), and ISGF3γ (IRF-9) (12). IRF-3 is expressed

constitutively in a variety of tissues, and the relative levels of IRF-3 mRNA do not change in virus-

infected or IFN-treated cells. IRF-3 demonstrates a unique response to viral infection.

Phosphorylation of latent cytoplasmic IRF-3 on serine and threonine residues in the C-terminal

region leads to a conformational change, dimerization, cytoplasmic to nuclear translocation,

association with the p300/CBP coactivator, stimulation of DNA binding and transcriptional

activities (3,13-17). Activated IRF-3 can in turn induce a specific subset of type 1 IFN genes in

response to viral infection including IFN-β and human IFN α1 (murine α4), as well as the CC-

chemokine RANTES and the Interleukin-15 (15,18-22). As with NF-κB activation, the rate

limiting step in this process is C-terminal phosphorylation of IRF-3 by an uncharacterized virus

activated kinase (VAK) activity.

Previous studies have demonstrated that treatment with dsRNA was sufficient to trigger the nuclear

accumulation of IRF-3 (17) and the formation of an IRF-3 containing DNA binding complex

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

5

(3,16). Recent studies also suggest that phosphorylation and activation of IRF-3 is not restricted to

viral infection, since LPS, DNA-damaging and stress-inducing agents all stimulate nuclear

accumulation of IRF-3, DNA-binding activity and transactivation (23-25). Using a variety of

pharmacological and molecular approaches, we now demonstrate that IRF-3 is uniquely activated

via C-terminal virus dependent phosphorylation. In addition to Sendai virus and Newcastle Disease

virus (NDV), measles virus (MeV) and vesicular stomatitis virus (VSV) are also identified as potent

inducers of VAK activity. In contrast, exposure of cells to stress-inducers, growth factors, DNA-

damaging agents and cytokines including doxorubicin and TNF-α, resulted in N-terminal

phosphorylation but not C-terminal IRF-3 phosphorylation by a mitogen-activated protein kinase

kinase kinase (MAPKKK)-related signaling pathway. N-terminal phosphorylation was not

sufficient to promote nuclear translocation, transactivation or degradation of IRF-3. The fact that

numerous well characterized pharmacological inhibitors failed to block VAK activity suggests the

involvement of a novel kinase in IRF-3 regulation by viruses.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

6

MATERIALS AND METHODS

Reagents. PDTC, sorbitol, LPS and ribavirin were purchased from Sigma and disolved in

distilled water or phosphate-buffered saline (PBS). All other pharmacological inhibitors were from

Calbiochem or Biomol and resuspended in dimethyl sulfoxyde (DMSO) or ethanol. Recombinant

macrophage inflammatory protein (MIP)1α, MIP1β and RANTES were from R&D System.

Pertussis Toxin, epiderma growth factor (EGF), platelet-derived growth factor-BB (PDGF-BB),

Insulin and thrombin were kind gifts from Dr. Sylvain Meloche.

Plasmid constructions and Mutagenesis. CMVBL-IRF-3wt, -IRF-3 5A, -IRF-3 5D; pFlag-

IRF-3 1-240, the reporter plasmids containing two PRD II sites, pGL3-P2(2)tk-LUC and the IFNβ

promoter, pGL3-IFN-β-LUC were described previously (13,19,26). The κB-mutated RANTES

promoter, pGL3-κBm-RANTES-LUC, was prepared by cloning the BglII-SalI fragment (-397 to

+5; filled in with the Klenow enzyme) from the κBm-RANTES-CAT reporter plasmid (19) into the

NheI site (filled in with the Klenow enzyme) of the pGL3-basic vector. The expression constructs

encoding different C-terminal IRF-3 truncations, pFlag-IRF-3 1-198, 1-186, 1-174 and 1-150 were

generated by overlap PCR mutagenesis using Vent DNA polymerase. Constructs encoding for

MAPKKKs, PCDNA3-MEKK1-HA and pRK5-MYC-Cot, were kind gifts from Drs Richard

Gaynor and Warner Greene respectively.

Cell Culture. The rtTA-Jurkat, rtTA-Jurkat IRF-3wt and rtTA-Jurkat IRF-3-5D were described

previously (27). Human embryonic kidney (HEK) 293 cells and Hela cells were grown in αMEM

and Dulbecco’s MEM respectively supplemented with 10% fetal bovine serum (FBS), glutamine,

and antibiotics. The monocytic cell line U937 was cultured in RPMI supplemented with 5% FBS.

The human bronchial lung carcinoma cell line A549 was purchase from ATCC (CCL-185) and

cultured in F12K supplemented with 10% FBS. Extracts of primary monocytes uninfected or

infected with NDV were a kind gift of Dr. Sandra Gessani, ISS, Rome.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

7

Transfections and luciferase assays. All transfections were carried out on subconfluent

HEK 293 cells grown in 60 mm petri dishes or 24 well plates (luciferase assay). 5 µg of DNA

constructs (per 60 mm dish) or 10 ng of pRLTK reporter (Renilla luciferase for internal control),

100 ng of pGL3 reporter (firefly luciferase, experimental reporter) and 250-500 ng of expression

plasmids (24 well plate) were introduced into target cells by calcium phosphate coprecipitation

method. At 24h posttransfection, cells were infected with Sendai virus for 12h (80

hemagglutinating units (HAU)/ml) or treated with the different inducers for the indicated times. At

36h, cells were collected, washed in ice-cold PBS and assayed for reporter gene activities

(Promega); whole cell extracts (WCE) were prepared in NP-40 lysis buffer (50 mM Tris pH 7.4;

150 mM NaCl, 30 mM NaF, 5 mM EDTA, 10% glycerol, 1.0 mM Na3VO4, 40 mM β-

glycerophosphate, 10-4M phenylmethylsulfonyl fluoride (PMSF), 5µg/ml of each leupeptin,

pepstatin and aprotinin and 1% NP-40) and stored at -80°C.

Immunoblot analysis. To verify the state of phosphorylation of IRF-3 and to confirm the

expression of the transgenes, WCE (30 to 60 µg) were subjected to electrophoresis on 7.5, 10 or

12% acrylamide gels. Proteins were electrophoretically transferred to Hybond-C nitrocellulose

membranes (Nycomed Amersham, Inc.) in 25 mM Tris, 192 mM glycine and 20% methanol. The

membranes were blocked in TBS containing 5% nonfat dry milk and 0.1% Tween 20 for 1h at

25°C before incubation for 1.5h at 25°C with anti-IRF-3 (Santa Cruz), anti-Flag M2 (Sigma), anti-

Myc (Santa Cruz), anti-IκBα (Santa Cruz) and anti-IκBα Ser32 phosphospecific antibody (New

England Biolab) (1:1000 to1:3000) in blocking solution. After washing four times in TBS, 0.1%

Tween 20, the membranes were incubated for 1h with HRP-conjugated goat anti-rabbit or anti-

mouse IgG (1: 10000) in blocking solution. Immunoreactive bands were visualized by enhanced

chemiluminescence (Nycomed Amersham, Inc).

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

8

For coprecipitation studies, WCE (200-1000 µg) were incubated with 1 µg anti-CBP antibody A-

22 (Santa Cruz) cross-linked to 30 µl of protein A-Sepharose beads for 3 h at 4°C (Pharmacia

Biotech). The beads were washed five times with NP-40 lysis buffer, resuspended in denaturating

sample buffer, and the eluted IRF-3 proteins associated with CBP were analyzed by

immunoblotting.

Cytoplasmic and nuclear extracts preparations. To examine subcellular localization of the

IRF-3 protein, nuclear and cytoplasmic extracts were prepated from Hela cells after treatment with

different inducers for 8 h. The cells were washed in buffer A (10 mM HEPES, pH 7.9, 1.5 mM

MgCl2, 10 mM KCl, 0.5 mM dithiothreitol (DTT), 0.5 mM PMSF) and were resuspended in buffer

A containing 0.1% NP-40. The cells were then chilled on ice for 10 min before centrifugation at

10.000 x g. This procedure was performed twice to remove cytoplasmic contaminants in the

nuclear extracts. After centrifugation, supernatants were kept as cytoplasmic extracts. The pellet

were then resuspended in buffer B (20 mM HEPES, pH 7.9, 25% glycerol, 0.42 M NaCl, 1.5 mM

MgCl2, 0.2 mM EDTA, 0.5 mM DTT, 0.5 mM PMSF, 5 µg/ml of each leupeptin, pepstatin,

aprotinin, spermine and spermidine). Samples were incubated on ice for 15 min before being

centrifuged at 10,000 x g. Nuclear extract supernatants were diluted with buffer C (20 mM

HEPES, pH 7.9, 20% glycerol, 0.2 mM EDTA, 50 mM KCl, 0.5 mM DTT, 0.5 mM PMSF).

Equivalent amounts of nuclear and cytoplasmic extracts (20 µg) were subjected to SDS-PAGE in a

10% polyacrylamide gel. Proteins were electrophoretically transferred to Hybond-C nitrocellulose

membranes which were probed with IRF-3 antiboby as described earlier.

Phosphatase treatment. HEK 293 cells were left untransfected or transfected with expression

plasmids encoding wild-type or mutated forms of IRF-3. At 36h posttransfection, cells were

stimulated and WCE were prepared. Endogenous IRF-3 (400 µg) or overexpressed IRF-3 (150 µg)

proteins were immunoprecipitated with anti-IRF-3 antibody (Santa Cruz) or anti-Flag antibody

(Sigma) cross-linked to 30 µl of protein G-sepharose beads for 4 h at 4°C. Precipitates were

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

9

washed two times in NP-40 lysis buffer followed by two washes in phosphatase buffer (50 mM

Tris pH 9.0, 1 mM MgCl2, 0.1 mM ZnCl2, 1 mM spermidine, 0.5 mM PMSF, 5 µg/ml aprotinin

and 5 µg/ml leupeptin). The phosphatase treatment was started by resuspending the beads in a total

volume of 40 µl of phosphatase assay buffer containing 5 U of calf intestine alkaline phosphatase

(CIP; Pharmacia) in the absence or presence of a phosphatase inhibitor (PI) mix containing (final

concentration) 10 mM NaF, 1.5 mM Na2MoO4, 1 mM β-glycerophosphate, 0.4 mM Na3VO4, and

0.1 µg of okadaic acid per ml. The reactions were incubated at 37°C for 2 h and stopped by

washing the beads once with NP-40 lysis buffer and addition of 50 µl of 2X denaturating sample

buffer. The samples were resolved by SDS-gel electrophoresis and analyzed by immunobloting

using anti-IRF-3 and anti-Flag antibodies.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

10

RESULTS

Multiple forms of IRF-3 phosphoprotein. C-terminal phosphorylation of IRF-3 following

paramyxovirus infection is a prerequisite for its nuclear translocation, association with CBP/p300

coactivators and transcriptional activation (13,16,17,28). VAK activity is relatively easy to detect

in extracts from virus infected cells, since phosphorylated IRF-3 migrates slower in SDS-PAGE

than non-phosphorylated IRF-3 (13,16,17), a phenomenon observed with many phosphoproteins.

To characterize the different forms of phosphorylated IRF-3 in virus-infected cells, IRF-3 specific

immunoblotting was used to reveal two forms of IRF-3 (designated forms I and II) in uninfected

HEK 293, U937, Jurkat cells (Fig. 1B, lane 1, 3 and 7). These forms were also present in human

epithelial Hela cells, human bronchial epithelial A549 cells, primary human monocytes (see Figs. 3

and 6) and freshly isolated primary B cells (data not shown). Sendai virus infection resulted in the

appearance of two slowly migrating forms of IRF-3 (forms III and IV) in HEK 293, U937, and

IRF-3 expressing Jurkat cells (Fig. 1B, lanes 2,4,5,6,and 8). Forms III and IV represent IRF-3

phosphorylated at a cluster of serines near the C terminal end of the protein ((13) and see Fig. 4C).

In addition, a net decrease in the amount of IRF-3 was observed between 4 and 12h after virus

infection of U937 cells, supporting the idea that C-terminally phosphorylated IRF-3 is subject to

proteasome-mediated degradation (13). Overexpression of the constitutively active form of IRF-

3(5D) (13,19,26,27) in Jurkat cells demonstrated that the phosphomimetic form migrated slower in

SDS-PAGE than endogenous IRF-3 protein, at a position similar to form IV observed in cells

infected with Sendai virus (Fig. 1B, lane 9). This initial experiment, while largely confirming

previous observations, nevertheless clearly demonstrates that multiple forms of IRF-3

phosphoprotein exist in unstimulated and virus infected cells.

Phosphatase treatment of immunoprecipitated IRF-3 isolated from cells overexpressing IRF-3wt

revealed that form II was also a phosphoprotein (Fig. 1C, lanes 4-6). Phosphatase treatment

resulted in the disappearance of form II from the extract and an increase in non-phosphorylated

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

11

IRF-3 (form I) (Fig. 1C, compare lanes 4 and 5), an effect that was blocked by addition of

phosphatase inhibitors (Fig. 1C, lane 6). Interestingly, IRF-3(5A) and IRF-3(5D), in which the

five phosphoacceptor sites in the C-terminus were mutated to alanine (5A) and aspartic acid (5D)

(see Fig.1A), were still expressed as two forms, form II (5A, lane 7) and form V (5D, lane 10) (see

also Fig. 1B, lane 9). These forms remained sensitive to phosphatase treatment (Fig. 1C, lanes 8

and 11) but were present when phosphatase inhibitors were used (lanes 9 and 12). Also, when

endogenous IRF-3 was immunoprecipitated from Sendai virus infected HEK 293, forms III and IV

were readily detected (Fig. 1C, lane 13); CIP treatment resulted in the conversion of form III and

IV to forms I and II (Fig. 1C, lane 14), an effect that was also blocked by phosphatase inhibitors

(Fig. 1C, lane 15). Based on these preliminary observations, it appeared that multiple forms of

IRF-3 phosphoprotein could be detected and basal IRF-3 phosphorylation - represented by form II

and form V (for IRF-3(5D)) - did not occur at the C-terminal phosphoacceptor sites implicated in

IRF-3 activation.

Pharmacological inhibitors fail to block VAK activity. In the effort to identify the

pathway(s) activated by viral infection and implicated in IRF-3 phosphorylation, the effect of well

characterized pharmacological inhibitors on IRF-3 phosphorylation following Sendai virus infection

was examined (Fig. 2 and Table 1). Use of specific pharmacological inhibitors that targetted

MEK1/2 (PD98059), p38α and β2 (SB203580), PI3-kinase (Wortmannin and LY294002) and

mTOR/FRAP (rapamycin) (29-35) did not affect the generation of the two hyperphosphorylated

forms of IRF-3 (III and IV) by Sendai virus (Fig 2A, lanes 3-7). Pretreatment of cells with the

intracellular calcium chelating agent BAPTA-AM (Fig. 2A, lane 10 and Table I) did however induce

a shift from form I to form II and also completely blocked virus-induced IRF-3 phosphorylation,

suggesting that a calcium dependent pathway may be upstream of IRF-3 activation (Fig.2A, lane

11). Many other pharmacological inhibitors also failed to block IRF-3 phosphorylation (Table I).

Interestingly, ribavirin, a selective inhibitor of the RNA polymerase of paramyxoviruses (36) had a

dose-dependent inhibitory effect on IRF-3 phosphorylation (Fig. 2B, lanes 3-8 and Table I),

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

12

possibly due to its ability to inhibit the replication of Sendai virus (data not shown). Furthermore,

UV-treated Sendai virus was unable to induce C-terminal IRF-3 phosphorylation (Fig. 2C),

suggesting that complete IRF-3 activation through C-terminal phosphorylation requires replication

competent virus.

Activation of IRF-3 is restricted to viral infection. The antagonizing effect of ribavirin

and UV-treatment on virus-dependent IRF-3 activation (Fig. 2BC) indicated that C-terminal

phosphorylation may be specific to virus infection. Viruses from different families were tested for

their capacity to induce IRF-3 phosphorylation and activation. Two paramyxoviridae family

members, MeV and NDV and one rhabdoviridae family member, VSV were also able to induce the

generation of form III and IV in HEK 293 cells, primary monocytes and human bronchial epithelial

A549 cells respectively (Fig. 3A, lanes 2, 4, 7). These viruses resulted in a phosphorylation-

dependent degradation of IRF-3 which was no longer detected in primary monocytes after infection

with NDV for 18h (Fig. 3A, lane 5, and data not shown). Induction of IRF-3 forms III and IV by

MeV infection also resulted in transactivation of IFN-β and NF-κB mutated RANTES promoters

(κBm-RANTES) (Fig. 3B).

Stress-inducers, DNA-damaging agents, growth factors and NF- B inducers

stimulate N-terminal IRF-3 phosphorylation. Recent studies showed that DNA-damaging

agents and stress-inducers activated IRF-3 in Hela cells (23,25). To determine which forms of IRF-

3 were activated by this diverse array of agents, HEK 293 cells were induced with Sendai virus,

stress-inducing stimuli sorbitol and anisomycin, DNA-damaging agent doxorubicin and the growth

factor/NF-κB inducer phorbol 12-myristate 13-acetate (PMA) (Fig. 4A). Treatment with

anisomycin, sorbitol, doxorubicin, PMA and also epidermal growth factor (EGF, data not shown)

resulted in the accumulation of form II without the generation of form III and IV (Fig. 4A, lanes 7-

18), as observed with Sendai virus (Fig. 4A, lanes 4-6). The conversion of form I to form II using

growth factor , stress and DNA damaging agents was sensitive to CIP treatment and, as shown

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

13

above, CIP was sensitive to phosphatase inhibitors (Fig. 4A, lanes 2,5,8,11,14,17), indicating that

the phosphorylation elicited by these agents was distinct from the virus-induced phosphorylation.

Previous studies demonstrated also that dsRNA treatment was sufficient to trigger the nuclear

accumulation of IRF-3 and the formation of a functionally active IRF-3 containing DRAF complex

(3,16,17). LPS treatment of U373 astrocytoma cells was also shown to induce IRF-3 nuclear

translocation and DNA binding activity (24). Surprisingly dsRNA treatment of HEK 293 cells and

LPS treatment of U937 and Hela cells did not induce any phosphorylation of IRF-3, as detected by

immunoblot analysis (data not shown). Other cytokines and growth factors such as CC-chemokines

(MIP1-α, MIP1-β and RANTES) thrombin, insulin, platelet-derived-growth factor-BB also had

also no effect on IRF-3 phosphorylation (data not shown).

DNA damaging agents activate the classical stress pathway MKK4/SEK1 and JNK (37-39);

furthermore, the catalytic activity of MKK4/SEK1 is regulated by MAPKKK family members of

which MEKK1 is the best described member (40). Anisomycin, EGF and hyperosmolarity are also

good inducers of MEKK1 activity (data not shown and (41-43) Therefore, the effect of

overexpressing MEKK1 on IRF-3 phosphorylation was examined. Figure 4B demonstrates that

Flag-tagged IRF-3 was expressed as non-phosphorylated form I and phosphorylated form II (Fig.

4B, lane 1). Importantly, coexpression of MAPKKKs MEKK1 and Cot, a member of the

MAPKKK family recently implicated in NF-κB activation following TCR engagement (44),

induced the accumulation of form II in transfected cells (Fig. 4B, lanes 4 and 7). This form

represented the phosphatase sensitive form of IRF-3 (Fig. 4B, lanes 5,6,8 and 9) as observed

above with stress-inducers and DNA-damaging agents (Fig. 4A). In contrast to Sendai virus

infection (see Fig. 1B, lanes 5-6), no degradation of IRF-3 was observed in cells overexpressing

MEKK1/Cot or treated with stress-inducers and DNA damaging agents after 16 to 24 hours of

treatment (data not shown).

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 14: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

14

Partial mapping of the region of IRF-3 phosphorylated by these agents or by MAPKKKs

overexpression revealed that phosphorylation did not occur in the C-terminal region (Fig. 4C).

Overexpression of IRF3wt showed the accumulation of form II following sorbitol treatment or

when MEKK1 and Cot were co-transfected (Fig.4C, lanes 8,12 and 16). However, when the Ser-

Thr cluster at aa396-aa405 was mutated to Ala (5A), the shift from form I to form II still occured

under the same conditions (Fig. 4C, lanes 9, 14 and 17). Overexpression of IRF-3wt and 5A

showed that the cluster of serine residues in the C-terminal region was essential for Sendai virus-

induced generation of form III and IV (Fig. 4C, compare lanes 4 and 6). In addition, no

accumulation of form II was observed in Sendai virus-infected cells overexpressing IRF-3 (5A)

(Fig. 4C, lane 6). Therefore an independent pathway leading to IRF-3 phosphorylation - distinct

from the virus inducible C-terminal specific pathway - appears to be stimulated by stress-inducers,

DNA-damaging agents, and growth factors.

To further pinpoint the region of IRF-3 targeted for phosphorylation by stress-inducers and DNA

damaging agents, a series of IRF-3 deletion mutants were evaluated. As illustrated in Fig. 5A,

truncation of full length IRF-3 to a protein of 240aa or 198aa did not alter the generation of forms I

and II (Fig. 5A, lanes 4-6); however truncation to a protein of 186aa resulted in a single form of

IRF-3 (Fig. 5A, lane 3), indicating that the modification occurred between aa186-aa198. With IRF-

3(1-198), anisomycin resulted in the conversion of form I to form II (Fig.5B, compare lanes 1 and

13); CIP treatment reverted form II to form I in a manner that was sensitive to phosphatase

inhibitors (Fig. 5B, lanes 14 and 15). The 150aa, 174aa and 186aa IRF-3 truncations were

expressed as a single form in both control and anisomycin treated cells and were insensitive to

phosphatase (Fig. 5B, lanes 4-12 and 16-24). As shown above for full length IRF-3, stress

inducers, DNA damaging agents and growth factors such as doxorubicin and PMA as well as

MEKK1 and Cot1 overexpression resulted in the complete or partial conversion of Flag-tagged

IRF-3 (1-198) from form I to form II (Fig. 5C), thus indicating that the phosphorylation site was

located between aa186 and aa198aa. Analysis of this region of IRF-3 revealed a single potential site

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 15: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

15

of Ser phosphorylation located within the sequence 186-GPSENPLKRLLVP-198. In addition,

Sendai virus infection did not induce accumulation of form II (Fig. 5C lane 2 and Fig. 4C, lanes 5-

6), suggesting that the modification of IRF-3 by stress-inducers and DNA-damaging agent was not

used by virus to induce the activated forms of IRF-3 (form III and IV).

N-terminal phosphorylation does not alter IRF-3 function. To examine the functional

consequences of N-terminal phosphorylation on IRF-3 activity, cells were stimulated with stress-

inducers and evaluated for IRF-3 functions such as CBP association, nuclear accumulation of IRF-

3, DNA binding and transactivation activity. Figure 6A shows that PMA, doxorubicin, stress-

inducers such as anisomycin, sorbitol and NaCl, and TNF-α induced a shift from form I to form II

(Fig. 6A, lanes 3-8) without inducing the slowly migrating forms of IRF-3 observed when cells

were infected by Sendai virus (Fig. 6A, lane 2). The effect of TNF-α on the conversion of form I

to form II was transient, with maximal conversion to form II occurring after 30 min and returning

to equal proportions of form I and II after 2h (RL, data not shown).

Since association of IRF-3 with CBP coactivator is a critical step in IRF-3 activation

(13,16,17,28), the relationship between the conversion of form I to form II and association with

CBP coactivator was evaluated. Co-immunoprecipitation analysis demonstrated that in Hela, HEK

293 and U937 cells, the association between IRF-3 and CBP was only detected in Sendai infected

cells when forms III and IV are present (Fig. 6B, lane 2); similarly when cytoplasmic and nuclear

partitioning was evaluated, only virus-induced IRF-3 translocated from the cytoplasm to the

nucleus of Hela cells (Fig. 6C, lanes 2 and 7).

Next, the effect of DNA damaging and stress-inducing agents on the transactivating potential of

IRF-3 was measured using a reporter gene assay with the IRF-3 responsive κBm-RANTES-LUC

(19). Sendai virus infection resulted in a 25-fold induction of RANTES activity in HEK 293 cells

(Fig. 6D). Virus-inducible expression of the RANTES promoter was inhibited by cotransfection

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 16: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

16

with a dominant-negative mutant of IRF-3 (∆NIRF-3) (19,27), demonstrating that the inducibility

of the RANTES promoter was essentially dependent on IRF-3 activation (Fig. 6D) (19). Under the

same conditions, stimulation for up to 15h with NF-κB inducers (PMA and TNF-α at 100 ng/ml),

DNA-damaging agent (doxorubicin) and stress-inducers (LPS at 100 µg/ml, anisomycin, and

sorbitol) failed to stimulate RANTES activity. Moreover, a pretreatment of cells for 1h with 10

µg/ml LPS did not affect Sendai virus-induced RANTES activity (Fig. 6D). Cotransfection with a

MEKK1 expression construct also had no effect on RANTES activity, whereas both TNF-

α treatment and MEKK1 stimulated NF-κB dependent LUC activity 7- and 12-fold respectively.

These experiments demonstrate that stress inducing agents, DNA damaging agents and cytokines

such as doxorubicin and TNF-α and growth factors stimulate a MAPKKK-related pathway that

phosphorylates IRF-3 in the N-terminal part of the protein. However, this phosphorylation event

appears to have no readily discernible consequence on IRF-3 translocation, association with CBP

coactivator, DNA binding activity or transactivation.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 17: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

17

DISCUSSION

In the present study, we describe a series of pharmacological and molecular experiments designed

to further characterize the signaling pathway(s) leading to IRF-3 phosphorylation and activation

following virus infection or treatment with a variety of activating agents. The results demonstrate

that IRF-3 phosphoprotein exists as two forms in uninfected cells: form I represents

nonphosphorylated IRF-3, while form II represents a basally phosphorylated form of IRF-3 that is

sensitive to phosphatase treatment. Based on mapping studies using IRF-3 deletions and point

mutations, basal phosphorylation of IRF-3 does not occur within the previously characterized

cluster of serine residues at the C terminus of IRF-3 (13,17). Rather, basal phosphorylation

appears to map to the N-terminal domain of IRF-3 between aa186 and aa198. Treatment with

stress-inducers, DNA-damaging agents, cytokines, and growth factors, does not induce C-terminal

IRF-3 phosphorylation, translocation or transactivation but rather activates a MAPKKK-related

signaling pathway that increases the proportion of N-terminally phosphorylated IRF-3. Following

viral infection, two additional slowly migrating forms of IRF-3 are detected - designated form III

and IV - that are sensitive to phosphatase treatment and represent C-terminal phosphorylation of

IRF-3. Only forms III and IV translocate to the nucleus of virus infected cells, and only C

terminally phosphorylated IRF-3 possesses DNA binding potential, CBP coactivator association

and transcriptional activity. Several well characterized, specific pharmacological inhibitors failed to

block virus-induced C-terminal phosphorylation, thus apparently ruling out many known signaling

pathways in the virus activation cascade. Furthermore, in vitro kinase assays demonstrated that

extracellular-activated kinases (ERK 1/2), JNK, p38α, IKKα/β and PKR were unable to

phosphorylate the C-terminal end of IRF-3 (data not shown). Full activation of IRF-3 appears to be

restricted to viral infection including paramyxoviruses (MeV, Sendai, NDV) and rhabdoviruses

(VSV) which are potent inducers of VAK activity. Our data thus provide evidence of an

uncharacterized virus-regulated kinase pathway involved in C-terminal IRF-3 phosphorylation and

activation.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 18: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

18

The results of this study contradict a number of recent investigations demonstrating that stress

inducers and DNA damaging agents functionally activate IRF-3. Navarro and David (24) reported

that LPS treatment of human U373 astrocytoma cells resulted in IRF-3 activation, via a Toll-

receptor and p38 dependent pathway. The authors demonstrated nuclear translocation and DNA

binding of IRF-3 but did not examine the phosphorylation state of IRF-3 or the functional activity

of the LPS-induced complex. Also the IRF-3-DNA complex that was identified migrated rapidly in

EMSA at a position consistent with a complex that did not include CBP/p300 coactivator. The

functionally active complex contains minimally IRF-3, CBP/p300 and DNA, resulting in a high

molecular weight virus-induced complex (VIC) (45) or virus-activated factor (VAF) (3). In light of

the present findings, an interpretation consistent with these observations is that LPS-induced IRF-3

phosphorylation occurs in the N-terminal domain. In U373 cells, LPS appears to be sufficient to

induce translocation of IRF-3 into the nucleus followed by enhanced IRF-3 DNA binding (24).

However, because of the absence of C-terminal phosphorylation, IRF-3 was unable to engage

CBP/p300 coactivator. Furthermore, in our hands with several cell types, LPS was unable to

induce functional IRF-3 activation (data not shown).

Kim et al, in a series of recent papers (23,46), demonstrated that stress inducers and genotoxic

agents such as DNA damaging agents doxorubicin and UV radiation stimulated IRF-3 (and IRF-7)

phosphorylation, nuclear translocation, CBP association and transcriptional activation of an IRF-3

responsive promoter. These experiments raise the exciting possibility that IRF-3 activation may be

central to the innate host response to environmental stress. However, the analysis of IRF-3

phosphorylation by Kim et al was not resolved sufficiently to delineate the different IRF-3

phosphorylated forms. Furthermore, the construct used to measure IRF-3 functional activity

consisted of an artificial construct containing five Gal4 binding sites to measure the activity of a

Gal4-IRF-3 fusion construct. As detailed in the present manuscript, DNA damaging agents did

stimulate IRF-3 phosphorylation at the N-terminal site but failed to induce nuclear accumulation

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 19: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

19

through CBP association or transcriptional activation of a natural IRF-3 responsive promoter –

RANTES, even using the identical HeLa cell model. At this stage, we believe that overexpression of

IRF-3 coupled with a sensitive but artificial transcriptional readout may lead to IRF-3 activation in

response to genotoxic stress.

Interestingly, a small molecule CG18 that stimulates MEKK1 activity was used to activate the

stress-mediated signaling pathway and was shown to stimulate the formation of the IFN-β

enhanceosome (25). All the enhancer binding activities – ATF.c-Jun, IRF-3 and NF-κB were

activated. MEKK1 activated IRF-3 through the JNK pathway but not through p38 or IKK

pathways. These experiments imply that MEKK1 can induce IRF-3 and ATF2/c-Jun through the

JNK pathway and NF-κB through the IKK pathway, resulting in the integration of multiple signal

transduction pathways leading to the proper assembly of the IFN-β enhanceosome. The

phosphorylation sites targeted by the MEKK1-related pathway are distinct from the C-terminal

sites, since the IRF-3(5A) protein was still phosphorylated in response to CG18 and MEKK1.

The IRF-3 function regulated by N-terminal phosphorylation remains to be elucidated. However,

based on the present study, several scenarios are possible. N-terminal phosphorylation by the

stress-induced pathway may alter IRF-3 conformation, thus making the C-terminal Ser-Thr cluster

more accessible to VAK (Fig. 7, pathway #1). This possibility was also proposed by Kim et al

(25). The two step mechanism is however questionable since viral infection did not induce N-

terminal phosphorylation of IRF-3 (Figs. 4C and 5C), indicating that VAK activity does not require

this modification to activate IRF-3. Another possibility is that N-terminal phosphorylation may

control IRF-3 activity at a step preceding nuclear translocation, such as relief of autoinhibition or

dimerization (Fig. 7, pathway #2). Finally, the possibility exists also that N-terminal

phosphorylation has no major effect on IRF-3 activity as a transcription factor, but may rather be

involved in a distinct function of IRF-3 based on the observation that IRF-3 interacts with

regulatory proteins that are not involved in transcription control (data not shown).

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 20: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

20

Finally, these experiments demonstrate for the first time that replication competent virus is required

for full activation of IRF-3 since UV-inactivation or ribavirin inhibition of virus replication blocked

IRF-3 activity. Furthermore, the paramyxovirus MeV and the rhabdovirus VSV may be added to

the growing list of viruses capable of activating IRF-3 function. Interestingly, influenza virus (as

well as other viruses of different classes) was unable to activate IRF-3 (BTO, data not shown).

Consistent with this observation, a recent study has demonstrated that the influenza virus NS1

protein, a dsRNA binding protein, specifically inhibited IRF-3 (47), although the mechanism of

inhibition remains to be elucidated. These studies demonstrate that, as with many other viruses, the

ability to interfere with the IFN antiviral cascade may contribute significantly to the virulence and

pathogenicity of viral infection.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 21: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

21

ACKNOWLEDGEMENTS

The authors wish to thank Drs. Paula Pitha, Brian Ward, Warner Greene, Richard Gaynor and

Sylvain Meloche for reagents used in this study and members of the Molecular Oncology Group,

Lady Davis Institute for helpful discussions. This research was supported by grants from the

Medical Research Council of Canada and the Cancer Research Society Inc. MJS was supported by

a MRC Fellowship, RL in part by a Fraser Monat McPherson Fellowship from McGill University

and JH by a MRC Senior Scientist award.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 22: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

22

REFERENCES

1. Stark, G. R., Kerr, I. M., Williams, B. R. G., Silverman, R. H., and Schreiber, R. D.

(1998) Annual Review of Biochemistry 67, 227-264

2. Kim, T. K., and Maniatis, T. (1997) Molecular Cell 1 , 119-129

3. Wathelet, M. G., Lin, C. H., Parakh, B. S., Ronco, L. V., Howley, P. M., and Maniatis,

T. (1998) Molecular Cell 1 , 507-518

4. Falvo, J. V., Parekh, B. S., Lin, C. H., Fraenkel, E., and Maniatis, T. (2000) Molecular

and Cellular Biology 20, 4814-4825

5. DeLuca, C., Kwon, H. J., Pelletier, N., Wainberg, M. A., and Hiscott, J. (1998) Virology

244, 27-38

6. Chu, W.-M., Ostertag, D., Li, Z.-W., Chang, L., Chen, Y., Hu, Y., Williams, B.,

Perrault, J., and Karin, M. (1999) Immunity 11, 721-731

7. Delhase, M., Hayakawa, M., Chen, Y., and Karin, M. (1999) Science 284, 309-313

8. Li, Q., Van Antwerp, D., Mercurio, F., Lee, K. F., and Verma, I. M. (1999) Science

284, 321-325

9. Li, Z. W., Chu, W., Hu, Y., Delhase, M., Deerinck, T., Ellisman, M., Johnson, R., and

Karin, M. (1999) Journal of Experimental Medicine 189, 1839-1845

10. Tanaka, M., Fuentes, M. E., Yamaguchi, K., Durin, M. H., Dalrymple, S. A., Hardy, K.

L., and Goeddel, D. V. (1999) Immunity 10, 421-429

11. Davis, R. J. (1999) Biochem. Soc. Symp. 64, 1-12

12. Mamane, Y., Heylbroeck, C., Genin, P., Algarte, M., Servant, M. J., LePage, C.,

DeLuca, C., Kwon, H., Lin, R., and Hiscott, J. (1999) Gene 237(1), 1-14

13. Lin, R., Heylbroeck, C., Pitha, P. M., and Hiscott, J. (1998) Molecular and Cellular

Biology 18, 2986-2996

14. Navarro, L., Mowen, K., Rodems, S., Weaver, B., Reich, N., Spector, D., and David,

M. (1998) Molecular and Cellular Biology 18, 3796-3802

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 23: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

23

15. Sato, M., Tanaka, N., Hata, N., Oda, E., and Taniguchi, T. (1998) FEBS Letters 425,

112-116

16. Weaver, B. K., Kumar, K. P., and Reich, N. C. (1998) Molecular and Cellular Biology

18, 1359-1368

17. Yoneyama, M., Suhara, W., Fukuhara, Y., Fukada, M., Nishida, E., and Fujita, T.

(1998) EMBO Journal 17, 1087-1095

18. Juang, Y. T., Lowther, W., Kellum, M., Au, W. C., Lin, R., Hiscott, J., and Pitha, P.

M. (1998) Proceedings of the National Academy of Sciences USA 95, 9837-9842

19. Lin, R., Heylbroeck, C., Genin, P., Pitha, P., and Hiscott, J. (1999) Molecular and

Cellular Biology 19, 959-966

20. Marié, I., Durbin, J. E., and Levy, D. E. (1998) EMBO Journal 17, 6660-6669

21. Schafer, S. L., Lin, R., Moore, P. A., Hiscott, J., and Pitha, P. M. (1998) Journal of

Biological Chemistry 273, 2714-2720

22. Azimi, N., Tagaya, Y., Mariner, J., and Waldmann, T. A. (2000) Journal of Virology 74,

7338-7348

23. Kim, T., Kim, T., Song, Y.-H., Min, I. M., Yim, J., and Kim, T. K. (1999) Journal of

Biological Chemistry 274, 30686-30689

24. Navarro, L., and David, M. (1999) Journal of Biological Chemistry 274, 35535-35538

25. Kim, T., Kim, T. Y., Lee, W. G., Yim, J., and Kim, T. K. (2000) Journal of Biological

Chemistry 275, 16910-16917

26. Lin, R., Mamane, Y., and Hiscott, J. (1999) Mol.Cell.Biol. 19, 2465-2474

27. Heylbroeck, C., Balachandra, S., Servant, M. J., Deluca, C., Barber, G., Lin, R., and

Hiscott, J. (2000) Journal of Virology 74, 3781-3792

28. Kumar, K. P., McBride, K. M., Weaver, B. K., Dingwall, C., and Reich, N. C. (2000)

Molecular and Cellular Biology 20, 4159-4168

29. Alessi, D. R., Cuenda, A., Cohen, P., Dudley, D. T., and Saltiel, A. R. (1995) J Biol

Chem 270(46), 27489-94

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 24: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

24

30. Pang, L., Sawada, T., Decker, S. J., and Saltiel, A. R. (1995) J Biol Chem 270(23),

13585-8

31. Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J., and Saltiel, A. R. (1995) Proc Natl

Acad Sci U S A 92(17), 7686-9

32. Gould, G. W., Cuenda, A., Thomson, F. J., and Cohen, P. (1995) Biochem J 311(Pt 3) ,

735-8

33. Yano, H., Nakanishi, S., Kimura, K., Hanai, N., Saitoh, Y., Fukui, Y., Nonomura, Y.,

and Matsuda, Y. (1993) J Biol Chem 268(34), 25846-56

34. Sanchez-Margalet, V., Goldfine, I. D., Vlahos, C. J., and Sung, C. K. (1994) Biochem

Biophys Res Commun 204(2), 446-52

35. Brown, E. J., Albers, M. W., Shin, T. B., Ichikawa, K., Keith, C. T., Lane, W. S., and

Schreiber, S. L. (1994) Nature 369(6483), 756-8

36. Patterson, J. L., and Fernandez-Larsson, R. (1990) Rev. Infect. Dis. 12, 1139-1146

37. Ip, Y. T., and Davis, R. J. (1998) Curr. Opin. Cell Biol. 10, 205-219

38. Kyriakis, J. M., and Avruch, J. (1996) J. Biol. Chem. 271, 24313-24316

39. Sanchez-Perez, I., and Perona, R. (1999) FEBS Lett. 453, 151-158

40. Fanger, G. R., Gerwins, P., Widmann, C., Jarpe, M. B., and Johnson, G. L. (1997)

Curr. Opin. Genet. Dev. 7 , 67-74

41. Widmann, C., Gerwins, P., Lassignal Johnson, N., Jarpe, M. B., and Johnson, G. L.

(1998) Molecular and cellular Biology 18, 2416-2429

42. Yujiri, T., sather, S., Fanger, G. R., and Johnson, G. L. (1998) Science 282, 1911-1914

43. Fanger, G. R., Johnson, N. L., and Johnson, G. L. (1997) EMBO J. 16, 4961-4972

44. Lin, X., Cunningham, E. T. J., Mu, Y., Geleziunas, R., and Greene, W. C. (1999)

Immunity 10, 271-280

45. Genin, P., Braganca, J., Darracq, N., Doly, J., and Civas, A. (1995) Nucleic Acids Res.

23, 5055-5063

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 25: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

25

46. Kim, T. K., Kim, T., Kim, T. Y., Lee, W. G., and Yim, J. (2000) Cancer Research 60,

1153-1156

47. Talon, J., Horvath, C. M., Polley, R., Basler, C. F., Muster, T., Palese, P., and Garcia-

Sastre, A. (2000) Journal of Virology 74, 7989-7996

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 26: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

26

FOOTNOTES

1The abbreviations used are: IRF: interferon regulatory factor, IFN: interferon, PRD: positif

regulatory domain, IKK: IκB kinase, dsRNA: double stranded RNA, LPS: lipopolysaccharide,

EGF: epidermal growth factor, MAPKKK: mitogen-activated protein kinase kinase kinase, PMA:

phorbol 12-,myristate 13-acetate, VAK: virus-activated kinase, HEK: human embryonic kydney,

CIP: calf intestine alkaline phosphatase, TNF: tumor necrosis factor, MeV: measle virus, NDV:

newcastle disease virus, VSV: vesicular stomatitis virus.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 27: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

27

FIGURE LEGENDS

Figure 1. Multiple forms of IRF-3 phosphoprotein. (A) Schematic representation of

IRF-3. The DNA binding domain, the NES element, the proline rich region and the C-terminal IRF

association domain are indicated. The region between aa382 and aa414 are expanded below the

schematic. The amino acids targeted for alanine or aspartic acid substitutions are shown in large

letters. The point mutations are indicated below the sequence: 5A, S396A, S398A, S402A, T404A,

S405A; 5D, S396D, S398D, S402D, T404D, S405D. cDNAs encoding for IRF-3 lacking the C-

terminal region (IRF-3 1-240, 1-198, 1-186, 1-174 and 1-150) and the DNA binding domain

(∆NIRF-3 (133-427)) are also shown. (B) Phosphorylation of IRF-3 in HEK 293, U937 and

rtTA-Jurkat, rtTA-Jurkat IRF-3wt and rtTA-Jurkat IRF-3 5D cells. Jurkat cells were induced with

Dox (1 µg/ml) for 16 h. Then HEK 293, U937, and rtTA-Jurkat IRF3wt were infected with Sendai

virus (80 HAU/ml) for 4, 8 or 12h or left uninfected (-). Endogenous IRF-3 proteins were detected

in whole cell extracts (55 µg) by immunoblotting using anti-IRF-3 antibody (Santa Cruz). (C)

Forms II, III, and IV are sensitive to phosphatase treatment. HEK 293 cells were transfected with

vector alone pBSCMV (pBS) or constructs encoding for IRF-3(wt), IRF-3(5A) and IRF-3(5D) or

left untransfected (Sendai virus). At 36h posttransfection or 8h after infection with Sendai virus (80

HAU/ml), whole cell extracts were prepared and subjected to immunoprecipitation using IRF-3

antibody covalently linked to protein A-sepharose beads. Immunoprecipitated IRF-3 was then used

in a phosphatase assay as described in Materials and Methods. The resulting immunoprecipitated

proteins were resolved by 7.5% SDS-PAGE. IRF-3 phosphorylated forms were analyzed by

immunoblotting using anti-IRF-3 antibody (Santa Cruz). CIP: calf intestinal phosphatase; PI:

phosphatase inhibitors. Lanes 13 to 15 are derived from the experiment shown in Figure 4A.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 28: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

28

Figure 2. Pharmacological inhibitors fail to block VAK activity. (A and B) HEK

293 cells were pretreated with different pharmacological inhibitors for 30min and then were left

untreated (-) or infected with Sendai virus (80 HAU/ml) for 8h (+) in the continuous presence of

inhibitors. Whole cell extracts (75 µg) were prepared from infected and control cells and were

analyzed for the presence of phosphorylated forms of IRF-3 by immunoblotting with anti-IRF-3

antibody. The concentration of inhibitors used were: PD98059, 50 µM; SB203580, 30 µM;

Wortmannin, 100 nM; LY294002, 50 µM; rapamycin, 15 ng/ml; BAPTA-AM, 15 µM; dimethyl

sulfoxide (DMSO), 0.1%; ribavirin (Riba), 250-1000 µg/ml. (C) HEK 293 cells were left

untreated (-) or infected with UV-treated virus (80 HAU/ml) or untreated virus (40 and 80

HAU/ml) for 8 h. Whole cell extracts and immunoblotting were preformed as described above.

Figure 3. Activation of IRF-3 is restricted to virus infections. (A) Phosphorylation of

IRF-3. Whole cell extracts (75 µg), prepared from HEK 293 cells, freshly isolated primary

monocytes and A549 cells uninfected (-) or infected with MeV (MOI of 1.0), NDV (100 HAU/ml)

and VSV (MOI of 10) for different time points, were resolved by 7.5% SDS-PAGE and transferred

to nitrocellulose. IRF-3 was analyzed by immunoblotting for the presence of phosphorylated IRF-

3 forms (II to IV) with anti-IRF-3 antibody. (B) Transactivation of PRD I-III- and ISRE-

containing promoters. HEK 293 cells were transiently transfected with reporter constructs

containing IFN-β enhancer (IFN-β-LUC) and the κB-mutated RANTES promoter (κBm-

RANTES-LUC). At 24h posttransfection, cells were treated as indicated in the legend and LUC

activity was analyzed 12h later. Relative LUC activity was measured as fold activation as described

in materials and methods. Each value represents the mean ± S.E. of triplicate determinations. The

data are representative of at least two different experiments with similar results. The concentration

of viruses used was: Sendai virus, 80 HAU/ml; Measles virus, MOI of 1.0.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 29: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

29

Figure 4. Stress-inducers, DNA-damaging agent, and NF- B inducers stimulate

N-terminal IRF-3 phosphorylation. (A and B) Phosphatase treatment. HEK 293 cells were

left untreated (-) or treated for 8 h with the indicated agents (A) or cotransfected with Flag-IRF3wt

and two MAPKKKs, MEKK1 and Cot (B). At 36h posttransfection or following different

treatments, whole cell extracts were prepared and subjected to immunoprecipitation using IRF-3

antibody covalently linked to protein A-sepharose beads or Flag antibody immobilized onto protein-

G sepharose beads. The immunoprecipitated IRF-3 proteins were then used in a phosphatase assay,

as described in Materials and Methods. The resulting immunoprecipitated proteins were resolved by

7.5% SDS-PAGE and transferred to nitrocellulose. Phosphorylation of IRF-3 was analyzed by

immunoblotting with anti-IRF-3 antibody. CIP: calf intestinal phosphatase; PI: phosphatase

inhibitors. (C) Phosphorylation or IRF-3 by stress-inducers does not occur in the C-terminal end

of the protein. HEK 293 cells were transfected with vector alone pBSCMV or constructs encoding

for IRF-3wt (WT) and IRF-3 5A (5A) or co-transfected with filling vector (-) or MEKK1 and Cot

expression plasmids (+). At 30h posttransfection, where indicated, cells were left untreated (-) or

treated (+) for 8h with Sendai virus (80 HAU/ml) or sorbitol (0.3 M). Whole cell extracts (30 µg)

were analyzed for IRF-3 phosphorylation by immunoblotting with anti-IRF-3 antibody.

Figure 5. Mapping the site of N-terminal phosphorylation of IRF-3. (A) C-terminal

IRF-3 truncation results in the expression of one form of IRF-3. HEK 293 cells were transfected

with various IRF-3 expression plasmids as indicated above the lanes. At 30 h posttransfection,

whole cell extract were prepared (20 µg) and analyzed for IRF-3 expression using anti-Flag

antibody. Arrows show the two forms of IRF-3, asterics show the expression of only one form of

IRF-3 following truncation (B) . HEK 293 cells were transfected with constructs as indicated in

(A). At 36h posttransfection or 3 h after treatment with 1 µM anisomycin, whole cell extracts were

prepared and subjected to immunoprecipitation using Flag antibody covalently linked to protein G-

sepharose beads. Immunoprecipitated IRF-3 was then used in a phosphatase assay as described in

Materials and Methods. The resulting immunoprecipitated proteins were resolved by 12% SDS-

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 30: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

30

PAGE. IRF-3 phosphorylated forms were analyzed by immunoblotting using anti-Flag antibody.

CIP: calf intestinal phosphatase; PI: phosphatase inhibitors. (C) HEK 293 cells were transfected

with Flag-tagged truncated version of IRF-3 (Flag-IRF-3 (1-198)) or cotransfected with filling

vectors (pCDNA3 and pRK5) or Cot and MEKK1 expression plasmids where indicated. At 30h

posttransfection, Flag-IRF-3 (1-198)-transfected cells were stimulated for 8 h with Sendai virus (80

HAU/ml), PMA (100 ng/ml); Sorbitol (0.3 M); Doxorubicin (1 µg/ml) and Anisomycin (1 µM). 30

µg of whole cell extracts were then resolved by SDS-gel electrophoresis on 12% acrylamide gel and

transferred to nitrocellulose membrane. Phosphorylation of Flag-IRF-3 was analyzed by

immunoblotting with anti-Flag antibody.

Figure 6. N-terminal phosphorylation does not alter IRF-3 subcellular localization

or function. (A) IRF-3 phosphorylation. Whole cell extracts prepared from HEK 293 cells

untreated (-) or treated with different agents or infected with Sendai virus (80 HAU/ml) for 8h

(except for TNF-α-treated cells where a 30 min stimulation is shown) were prepared. Protein

extracts (75 µg) were analyzed by immunoblotting for the presence of phosphorylated IRF-3 (II to

IV) with anti-IRF-3 antibody. The concentration of agents used were: sorbitol, 0.3 M; NaCl, 0.25

M; anisomycin, 1 µM; doxorubicin, 1 µg/ml; PMA, 100 ng/ml; TNF- α, 25 ng/ml. (B) Interaction

between IRF-3 and CBP coactivator. Hela, HEK 293 and U937 cells were treated as described in

A. Whole cell extracts (500 µg) were immunoprecipitated with anti-CBP antibody A22, covalently

linked to protein A-sepharose beads. The immunoprecipitated proteins were resolved by SDS-gel

electrophoresis on 7.5% acrylamide gel and transferred to nitrocellulose membrane. The membrane

was probed with anti-IRF-3 antibody. As indicated, only form III and IV were found to bind CBP.

Lane 10: WCE (30 µg) prepared from uninfected HEK 293 cells were used to show the position of

forms I and II. The concentration of agents used are described in A. LPS: 10 µg/ml. (C)

Cytoplasmic to nuclear translocation of IRF-3. Hela cells were treated as indicated in Figure 6 A

and B. Cytoplasmic and nuclear fractions were prepared as described in Materials and Methods

section. Each isolated fraction was subjected to 10% SDS-PAGE, transferred to nitrocellulose

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 31: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

31

membrane and probed with anti-IRF-3 antibody. Lower panels, membranes were stripped and

reblotted with an anti-α-actin antibody. (D) Transactivation of ISRE and PRD II containing

promoters. HEK 293 cells were transfected with the κB-mutated RANTES promoter (κBm-

RANTES-LUC) or P2(2)tk-LUC reporter plasmids and the MEKK1 (250 ng) or ∆NIRF-3 (500

ng) expression plasmids when indicated. At 24h posttransfection, cells were treated as indicated

below the bar graph and LUC activity was analyzed 12h later. Relative LUC activity was measured

as fold activation. Each value represents the mean ± SE. of triplicate determinations. The data are

representative of at least three different experiments with similar results. The concentration of

agents used were: Sendai virus, 80 HAU/ml; PMA, 100 ng/ml; LPS, 10 or100 µg/ml; TNF-α, 10

or 100 ng/ml; doxorubicin, 1 µg/ml, anisomycin,1µM and sorbitol, 0.20 M.

Figure 7 . Schematic representation of IRF-3 activation following N- and C-

terminal phosphorylation. In uninfected cells, intramolecular association between the C

terminus and the DBD maintains IRF-3 in a latent state in the cytoplasm by masking both DBD and

IAD regions of the protein (form I). Basal activities of both N-terminal kinase and phosphatase may

affect the overall ratio between IRF-3 form I and II. Treatment of cells with stress-inducers, DNA

damaging agents and growth factors activates a MAPKKK-related pathway involved in the positive

regulation of the N-terminal kinase, resulting in an increase in form II. N-terminal phosphorylation

may induce a conformational change that reveals phosphoacceptor sites for VAK in the C-terminal

end of IRF-3 (pathway #1). C-terminal phosphorylation by VAK then relieves the intramolecular

association between DBD and IAD leading to homodimerization of IRF-3. C-terminal autoinhibition

could also be relieved through N-terminal phosphorylation (pathway #2) resulting in

homodimerization of IRF-3 before C-terminal phosphorylation by VAK. IRF-3 can then

accumulate in the nucleus and activate genes through DNA binding and CBP association.

Ultimately, IRF-3 is degraded by the proteasome pathway. DBD, DNA binding domain, IAD, IRF

association domain.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 32: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

32

TABLE LEGEND

Table I. List of pharmacological inhibitors, their cellular targets and effects on Sendai virus-

induced IRF-3 phosphorylation in HEK 293 cells.

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 33: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

A

N C1

414

IRF Association DomainDNA Binding NES

382 - GGA SS LENTVDLHI SNSHPL SLTSDQYKAYLQD -

5A/5D

(385/386) (396/398) (402/404/405)

NIRF-3 (133-427)

1-1981-186

1-1741-150

1-240

Pro

- 8 - 4 8 12Sendai virus (h)

B

C

293 U937 JurkatrtT

AIR

F-3 W

TIR

F-3- 5

DI

III

IV

+ + + + + + + ++ + + +

CIPPI

WT 5A 5D

+ ++

IV/5D

III

- 8 -

II

I

IIIIV/5D

II

I

III

IV

II

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

1 2 3 4 5 6 7 8 9

Figure 1Servant et al

pBS Sendai virus

V

V

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 34: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

Sendai virus(8 hours)

- 40 80 80

UVnon-treated

IIIIV

II

ISendai virus

(8 hours)- + + + + + +

PD98

059

SB20

3580

DM

SOD

MSO

Wor

tman

nin

LY29

4002

Rap

amyc

in

A

B

C

II-IVI

- + - + - + - +Sendai virus ( 8 hours)

250 R

iba

500 R

iba

1000

Rib

a

ddH

2O

II-IVI

- + +

BAPT

A-A

MBA

PTA

-AM

DM

SOD

MSO

-

1 2 3 4 5 6 7 8 9 10 11

1 2 3 4 5 6 7 8

1 2 3 4

Figure 2Servant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 35: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

A

B

uninfectedMeasles virusSendai virus

Bm-RANTES-LUC

Rel

ativ

e L

UC

act

ivit

y

20

0

10

30

IFN- -LUC

- 18 h - 6 h 18 h - 24 h

MeV NDV VSV

293primary

monocytes A549

III

IV

II

I

1 2 3 4 5 6 7

Figure 3Servant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 36: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

III

+ + + + + ++ + +

CIP

PI

pcDNA3 MEKK1 Cot

Flag-IRF-3 WTB

III

- Sendai

virus

Anisomyc

in

PMA

Doxor

ubicin

Sorbito

l

+ + + + + + + + + + + ++ + + + + +

CIP

PI

A

IIIIV

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18

1 2 3 4 5 6 7 8 9

Sendai virusSorbitol

MEKK1 + +

Cot + +

I

-- --

IIIIIIV

+ + +- - -

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 175AWTpB

SCM

V

WT 5A WT 5A WT 5ApBSC

MV

pBSC

MV

+ + +

C

Figure 4Servant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 37: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

Flag-IRF-3: 150 174 186 198 240 427

1 2 3 4 5 6

**

*

IgG

CIPPI

+ ++

+ ++

+ ++

+ ++

+ ++

+ ++

+ ++

+ ++

control Anisomycin

Flag-IRF-3:1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

198 186 174 150 198 186 174 150

Flag-IRF-3 (I) Flag-IRF-3 (II)

Flag-IRF-3 (1-198)

Send

ai v

irus

PMA

Sorb

itol

Dox

orub

icin

Ani

som

ycin

pCD

NA

3

pRK

5-C

ot-

pRK

5

pCD

NA

3-M

EKK

1

1 2 3 4 5 6 7 8 9 10

A

B

C

Figure 5Servant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 38: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

A

-

Sorb

itol

TNF-

Dox

orub

icin

Ani

som

ycin

Send

ai v

irus

NaC

l

IVIII II

I

PMA

1 2 3 4 5 6 7 8

B

III-IV

-

Send

ai v

irus

Sorb

itol

PMA

Dox

orub

icin

LPS

Ani

som

ycin

TNF-

293

U937

IIIIV

III

Hela

IIIIV

1 2 3 4 5 6 7 8 9

10

Nac

l

C

-actin

IIIIV

III

Send

ai v

irus

Sorb

itol

Dox

orub

icin

Send

ai v

irus

Sorb

itol

Dox

orub

icin- -

cytoplasmic nuclear1 2 3 4 5 6 7 8 9 10

PMA

PMA

D

-

Sorb

itol

PMA

Send

ai v

irus

LPS

100

LPS

10 +

Sen

dai v

irus

Dox

orub

icin

Bm-RANTES-LUC

Rel

ativ

e L

UC

act

ivit

y

20

0

10

30

Send

ai v

irus

+

NIR

F-3

MEK

K1

P2(2

)tk-L

UC

MEK

K1

Ani

som

ycin

Figure 6Servant et al

TNF-

100

TNF-

10

IgG

IgG

IgG

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 39: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

IAD

DBD

P

P

P

IADDBD I

P

IAD

DBDII

P

N-terminalN-terminalkinasekinase

N-terminalN-terminalphosphatasephosphatase

III/IV

VAK

stress-inducersDNA damaging agents

growth factors

P

DBDP

P P

P

DBDP

PP

IAD

IAD

CBP association/genes activation/

IRF-3 degradation

MAPKKK 1

2 DBDP

DBDP

IAD

IAD

VAK

Figure 7Servant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 40: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

Pharmacological inhibitors Target

Inhibition of IRF-3 phosphorylation

-PI3-kinaseWortmannin (100 nM)

-PI3-kinaseLY294002 (50 µM)

-mTOR/FRAPRapamycin (10-25 ng/ml)

tyrosine kinasesGenistein (25-100 µM)

p38 SB203580 (10-50 µM) -CAM kinase IIKN93 (10 µM) -

Actinomycin D (5 µg/ml) DNA -

-K-252a (200 nM) Ser/Thr kinases

MEK1,2PD 98059 (50-100 µM) -

c, -nPKCsGF109203X (0.01-10 µM) -

JAK-2AG490 (50 µM) -

IKKNaSal (20 mM) -

cPKCsGö6976 (0.1-10 µM) -

Ribavirin (0.25-1 mg/ml)RNA polymerase of

paramyxoviruses +

ser/thr kinases

tyrosine kinases

phosphatidylinositol kinase

mRNA and protein synthesis

calcium chelator +BAPTA-AM (15 µM)

-Reactive Oxygene SpeciesPDTC (100 µM)

-Pertussis Toxin (100 ng/ml) Gi/Go pathways

-Cytochalasin D (1µM) actin filaments

-Okadaic acid (25-100 nM) PP-1/PP-2A phosphatases

miscellaneous

dual specificity kinase

-H-7 (1 µM) Ser/Thr kinases

-

Table IServant et al

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Page 41: IDENTIFICATION OF DISTINCT SIGNALING PATHWAYS LEADING ...

Julkunen, Rongtuan Lin and John HiscottMarc J. Servant, Benjamin ten Oever, Cecile LePage, Lucia Conti, Sandra Gessani, Ilkka

interferon regulatory factor 3Identification of distinct signaling pathways leading to the phosphorylation of

published online October 16, 2000J. Biol. Chem. 

  10.1074/jbc.M007790200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

by guest on March 22, 2018

http://ww

w.jbc.org/

Dow

nloaded from