13
Inhibition of Sphingosine Kinase by Bovine Viral Diarrhea Virus NS3 Is Crucial for Efficient Viral Replication and Cytopathogenesis * Received for publication, September 29, 2008, and in revised form, March 12, 2009 Published, JBC Papers in Press, March 17, 2009, DOI 10.1074/jbc.M807498200 Daisuke Yamane, Muhammad A. Zahoor, Yassir M. Mohamed, Walid Azab, Kentaro Kato, Yukinobu Tohya, and Hiroomi Akashi 1 From the Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid implicated in diverse cellular functions including survival, pro- liferation, tumorigenesis, inflammation, and immunity. Sphin- gosine kinase (SphK) contributes to these functions by convert- ing sphingosine to S1P. We report here that the nonstructural protein NS3 from bovine viral diarrhea virus (BVDV), a close relative of hepatitis C virus (HCV), binds to and inhibits the catalytic activity of SphK1 independently of its serine protease activity, whereas HCV NS3 does not affect SphK1 activity. Uncleaved NS2-3 from BVDV was also found to interact with and inhibit SphK1. We suspect that inhibition of SphK1 activity by BVDV NS3 and NS2-3 may benefit viral replication, because SphK1 inhibition by small interfering RNA, chemical inhibitor, or overexpression of catalytically inactive SphK1 results in enhanced viral replication, although the mechanisms by which SphK1 inhibition leads to enhanced viral replication remain unknown. A role of SphK1 inhibition in viral cytopathogenesis is also suggested as overexpression of SphK1 significantly atten- uates the induction of apoptosis in cells infected with cytopatho- genic BVDV. These findings suggest that SphK is targeted by this virus to regulate its catalytic activity. Bovine viral diarrhea virus (BVDV) 2 is an enveloped, posi- tive-sense single-stranded RNA virus classified in the genus Pestivirus of the family Flaviviridae. BVDV establishes persis- tent infections in cattle populations worldwide. Because BVDV shares virological and molecular properties with the Flaviviri- dae family member hepatitis C virus (HCV), which chronically infects an estimated 200 million patients worldwide (1), BVDV is regarded as a surrogate model for HCV (2). Both HCV and BVDV encode a single large precursor polyprotein that is pro- cessed by cellular and viral proteases into mature structural and nonstructural (NS) proteins. BVDV NS3 exhibits serine protease and helicase/ATPase activities that require its cofactor NS4A (3). NS3/4A protease is essential for generating mature NS proteins that are required for viral replication. HCV NS3/4A is well characterized and has been shown to suppress type-I interferons by cleaving the cel- lular interferon mediators IPS-1 and TRIF (4, 5). However, nei- ther interferon suppression nor cellular targets have been iden- tified for the BVDV NS3/4A protease (6). Lytic and persistent BVDV infections depend on the virus biotype. Cytopathogenic (CP) BVDV causes cytopathic effects via apoptosis, whereas noncytopathogenic (NCP) BVDV does not induce obvious changes in cell morphology and viability. These features are distinguished by NS2-3 processing differ- ences; free NS3 produced by NS2-3 cleavage is generated con- tinuously following CP BVDV infections, whereas NS3 is detected only until 9 h postinfection (p.i.) for NCP BVDV due to down-regulation of NS2-3 cleavage by this biotype (7). The CP biotype is characterized by dramatic up-regulation of viral RNA synthesis that could be correlated with the induction of cytopathic effect (7–9). Because free NS3, but not NS2-3, can form an active viral replicase complex with other NS proteins, increased viral RNA synthesis promoted through the release of free NS3 has been suggested to be a determinant of the charac- teristic lytic phenotype of CP BVDV infections (10). However, little is known about the regulation of cellular signaling by BVDV NS2-3, NS3, and NS3/4A, which is crucial for the con- trol of both viral replication and biotype. Recent studies on the mechanisms of viral replication revealed that HCV RNA synthesis occurs on a lipid raft mem- brane structure where the active viral replicase complex is found (11, 12). The significance of the lipid raft as a scaffold for viral replication is further demonstrated by the identification of a novel HCV replication inhibitor, NA255, which prevents the biosynthesis of sphingolipids, the major components of lipid rafts (13). Administration of NA255 results in disruption of the HCV replicase complexes from the lipid rafts. This report pro- poses that the interaction between HCV NS5B and sphingomy- elin on lipid rafts plays a crucial role for HCV RNA replication. Cellular sphingolipid metabolism is regulated by a large num- ber of converting enzymes that maintain a homeostasis (14) but * This work was supported in part by grants-in-aid from the Ministry of Agri- culture, Forestry and Fisheries of Japan, the Ministry of Education, Culture, Sports, Science and Technology, Japan, and the Japan Society for the Pro- motion of Science. 1 To whom correspondence should be addressed. Tel.: 81-3-5841-5396; Fax: 81-3-5841-8184; E-mail: [email protected]. 2 The abbreviations used are: BVDV, bovine viral diarrhea virus; HCV, hepatitis C virus; CP, cytopathogenic; NCP, noncytopathogenic; S1P, sphingosine 1-phosphate; SphK1, sphingosine kinase 1; m.o.i., multiplicity of infection; SKI, sphingosine kinase inhibitor; DMEM, Dulbecco’s modified Eagle’s medium; p.i., postinfection; siRNA, small interfering RNA; mAb, mono- clonal antibody; pAb, polyclonal antibody; GAPDH, glyceraldehyde-3- phosphate dehydrogenase; FCS, fetal calf serum; RT, reverse transcriptase; MDBK, Madin-Darby bovine kidney; HEK, human embryonic kidney; PBS, phosphate-buffered saline. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 284, NO. 20, pp. 13648 –13659, May 15, 2009 © 2009 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. 13648 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009 by guest on July 2, 2018 http://www.jbc.org/ Downloaded from

InhibitionofSphingosineKinasebyBovineViralDiarrhea ... · InhibitionofSphingosineKinasebyBovineViralDiarrhea VirusNS3IsCrucialforEfficientViralReplication ... using the SV Total RNA

Embed Size (px)

Citation preview

Inhibition of Sphingosine Kinase by Bovine Viral DiarrheaVirus NS3 Is Crucial for Efficient Viral Replicationand Cytopathogenesis*

Received for publication, September 29, 2008, and in revised form, March 12, 2009 Published, JBC Papers in Press, March 17, 2009, DOI 10.1074/jbc.M807498200

Daisuke Yamane, Muhammad A. Zahoor, Yassir M. Mohamed, Walid Azab, Kentaro Kato, Yukinobu Tohya,and Hiroomi Akashi1

From the Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, University of Tokyo,1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan

Sphingosine 1-phosphate (S1P) is a bioactive sphingolipidimplicated in diverse cellular functions including survival, pro-liferation, tumorigenesis, inflammation, and immunity. Sphin-gosine kinase (SphK) contributes to these functions by convert-ing sphingosine to S1P. We report here that the nonstructuralprotein NS3 from bovine viral diarrhea virus (BVDV), a closerelative of hepatitis C virus (HCV), binds to and inhibits thecatalytic activity of SphK1 independently of its serine proteaseactivity, whereas HCV NS3 does not affect SphK1 activity.Uncleaved NS2-3 from BVDV was also found to interact withand inhibit SphK1.We suspect that inhibition of SphK1 activityby BVDV NS3 and NS2-3 may benefit viral replication, becauseSphK1 inhibition by small interfering RNA, chemical inhibitor,or overexpression of catalytically inactive SphK1 results inenhanced viral replication, although the mechanisms by whichSphK1 inhibition leads to enhanced viral replication remainunknown. A role of SphK1 inhibition in viral cytopathogenesisis also suggested as overexpression of SphK1 significantly atten-uates the inductionof apoptosis in cells infectedwith cytopatho-genic BVDV. These findings suggest that SphK is targeted bythis virus to regulate its catalytic activity.

Bovine viral diarrhea virus (BVDV)2 is an enveloped, posi-tive-sense single-stranded RNA virus classified in the genusPestivirus of the family Flaviviridae. BVDV establishes persis-tent infections in cattle populations worldwide. Because BVDVshares virological and molecular properties with the Flaviviri-dae family member hepatitis C virus (HCV), which chronicallyinfects an estimated 200 million patients worldwide (1), BVDVis regarded as a surrogate model for HCV (2). Both HCV and

BVDV encode a single large precursor polyprotein that is pro-cessed by cellular and viral proteases intomature structural andnonstructural (NS) proteins.BVDV NS3 exhibits serine protease and helicase/ATPase

activities that require its cofactor NS4A (3). NS3/4A protease isessential for generating mature NS proteins that are requiredfor viral replication. HCVNS3/4A is well characterized and hasbeen shown to suppress type-I interferons by cleaving the cel-lular interferonmediators IPS-1 and TRIF (4, 5). However, nei-ther interferon suppression nor cellular targets have been iden-tified for the BVDV NS3/4A protease (6).Lytic and persistent BVDV infections depend on the virus

biotype. Cytopathogenic (CP) BVDV causes cytopathic effectsvia apoptosis, whereas noncytopathogenic (NCP) BVDV doesnot induce obvious changes in cell morphology and viability.These features are distinguished by NS2-3 processing differ-ences; free NS3 produced by NS2-3 cleavage is generated con-tinuously following CP BVDV infections, whereas NS3 isdetected only until�9 h postinfection (p.i.) forNCPBVDVdueto down-regulation of NS2-3 cleavage by this biotype (7). TheCP biotype is characterized by dramatic up-regulation of viralRNA synthesis that could be correlated with the induction ofcytopathic effect (7–9). Because free NS3, but not NS2-3, canform an active viral replicase complex with other NS proteins,increased viral RNA synthesis promoted through the release offree NS3 has been suggested to be a determinant of the charac-teristic lytic phenotype of CP BVDV infections (10). However,little is known about the regulation of cellular signaling byBVDV NS2-3, NS3, and NS3/4A, which is crucial for the con-trol of both viral replication and biotype.Recent studies on the mechanisms of viral replication

revealed that HCV RNA synthesis occurs on a lipid raft mem-brane structure where the active viral replicase complex isfound (11, 12). The significance of the lipid raft as a scaffold forviral replication is further demonstrated by the identification ofa novel HCV replication inhibitor, NA255, which prevents thebiosynthesis of sphingolipids, the major components of lipidrafts (13). Administration of NA255 results in disruption of theHCV replicase complexes from the lipid rafts. This report pro-poses that the interaction betweenHCVNS5B and sphingomy-elin on lipid rafts plays a crucial role for HCV RNA replication.Cellular sphingolipid metabolism is regulated by a large num-ber of converting enzymes thatmaintain a homeostasis (14) but

* This work was supported in part by grants-in-aid from the Ministry of Agri-culture, Forestry and Fisheries of Japan, the Ministry of Education, Culture,Sports, Science and Technology, Japan, and the Japan Society for the Pro-motion of Science.

1 To whom correspondence should be addressed. Tel.: 81-3-5841-5396; Fax:81-3-5841-8184; E-mail: [email protected].

2 The abbreviations used are: BVDV, bovine viral diarrhea virus; HCV, hepatitisC virus; CP, cytopathogenic; NCP, noncytopathogenic; S1P, sphingosine1-phosphate; SphK1, sphingosine kinase 1; m.o.i., multiplicity of infection;SKI, sphingosine kinase inhibitor; DMEM, Dulbecco’s modified Eagle’smedium; p.i., postinfection; siRNA, small interfering RNA; mAb, mono-clonal antibody; pAb, polyclonal antibody; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; FCS, fetal calf serum; RT, reverse transcriptase;MDBK, Madin-Darby bovine kidney; HEK, human embryonic kidney; PBS,phosphate-buffered saline.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 284, NO. 20, pp. 13648 –13659, May 15, 2009© 2009 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

13648 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

viral mechanisms that affect the sphingolipid metabolism tofacilitate viral replication have yet to be identified.In a search for potential host proteins that interact with

BVDV NS3, we identified sphingosine kinase 1 (SphK1) as abinding partner of NS3 using the yeast two-hybrid system.SphK1 is a lipid kinase that catalyzes the phosphorylation ofsphingosine to form sphingosine 1-phosphate (S1P), a bioactivesphingolipid implicated in diverse cellular functions, includingproliferation, survival, tumorigenesis, development, inflamma-tion, and immunity (14, 15). Here, we analyze the biologicalsignificance of the SphK1 interaction with NS3, NS2-3, andNS3/4A. Using purified recombinant SphK1 and NS3, SphKactivity was inhibited by NS3 in a dose-dependent manner,independently of its serine protease activity. The inhibitionappears to be specific for BVDVNS3 because HCVNS3 had noeffect on SphK activity. Using specific chemical inhibitors,small interfering RNA (siRNA), and a catalytically inactivemutant of SphK1,we investigated the significance of SphK inhi-bition in the viral replication. The present study is the firstreport demonstrating that SphK1 is targeted by a virus toinhibit its catalytic activity, and thismechanismmay contributeto the efficient replication and pathogenesis of BVDV.

EXPERIMENTAL PROCEDURES

Reagents and Antibodies—D-erythro-Sphingosine and sphin-gosine kinase inhibitor (SKI) (16) were purchased from Calbio-chem (La Jolla, CA). S1P was obtained from Cayman Chemical(Ann Arbor, MI). Anti-FLAGM2 monoclonal antibody (mAb;IgG1), anti-Myc mAb (IgG1), and isotype control IgG1 mAbwere from Sigma. Anti-BVDV NS3 (IgG2a) and anti-GAPDHmAbs were from TropBio (Townsville, Australia) and Ambion(Austin, TX), respectively. Rabbit polyclonal antibodies (pAbs)against calnexin and SphK1 were from Stressgen (Victoria,Canada) and Exalpha (Maynard,MA), respectively.Mouse pAbagainst junctional adhesion molecule 1 was described previ-ously (17). Goat anti-mouse IgG, IgG1, IgG2a, or anti-rabbitIgG antibodies conjugated with Alexa 488, Alexa 488, Alexa594, or Alexa 568, respectively, were from Invitrogen. RabbitpAb against HCV NS3 was kindly provided by Dr. MichinoriKohara (Tokyo Metropolitan Institute of Medical Science,Tokyo, Japan). Protein concentrations in samples were deter-mined with the Protein Quantification Kit-Rapid (Dojindo,Rockville, MD) using bovine serum albumin as a standard.Molybdenum blue spray was from Sigma. Recombinant humanSphK1 (hSphK1) was purchased from BPS Bioscience (SanDiego, CA).Cells and Viruses—MDBK, LB9.K, and human embryonic

kidney HEK293 cell lines were obtained from the AmericanTypeCultureCollection (ATCC) andmaintained inDulbecco’smodified Eagle’s medium (DMEM) supplemented with 5, 10,and 10% fetal calf serum (FCS), respectively, at 37 °C in ahumidified 5% CO2 atmosphere. MDBK and LB9.K cells wereconfirmed to be free of BVDV by reverse transcriptase-poly-merase chain reaction (RT-PCR). BVDV strains KS86-1cp,KS86-1ncp, and Nose have been described previously (18).Unless otherwise indicated, MDBK and LB9.K cells wereinfected with BVDV using a multiplicity of infection (m.o.i.) of5 for 1 h, washed twice with FCS-free DMEM, and incubated in

DMEM containing 5 or 10% FCS, respectively. End point viraltitrationwas performedwith four replicates onMDBKcells andthe 50% tissue culture infective dose (TCID50) determined asdescribed previously (7). The intracellular synthesis of virus-specific proteins at 72 h p.i. was detected by indirect immuno-fluorescence analysis using anti-BVDV NS3 mAb (TropBio)and a secondary fluorescein isothiocyanate-labeled antibody asdescribed below under “Immunofluorescence Microscopy.”RNA Extraction—Total RNA was extracted using the SV

Total RNA Isolation System (Promega, Madison, WI) accord-ing to the manufacturer’s protocol.Plasmids—The bovine SphK1 complementary DNA (cDNA;

GenBankTM data base accession number XM_870939.1) wasgenerated from total RNA extracted from MDBK cells by RT-PCR using primers that incorporated EcoRI and EcoRV sites atthe 5� and 3� ends, respectively. Mammalian expression vec-tor of N-terminal FLAG-tagged bovine SphK1, designatedpFlag-SphK1, was generated by cloning the SphK1 cDNAinto pFLAG-CMV2 (Sigma) using EcoRI and EcoRV sites.The fragments encoding a series of deletion mutants ofSphK1 were generated by PCR-mediated site mutagenesisusing pFlag-SphK1 as a template. The fragment of catalyticallyinactive SphK1G177D was generated by PCR-mediated mu-tagenesis using pFlag-SphK1 as a template with the mutagenicprimer 5�-TCGTGGATCAGCCCATCATCGGACATGACC-ACCAG-3� to substitute Gly177 to Asp.The mammalian expression vectors of BVDV NS proteins

were generated using SR� promoter vector, pME18S. TheMyctag sequence together with the multiple cloning site frompGBKT7 was amplified by PCR using pGBKT7-NS3 as a tem-plate and cloned betweenXhoI and PstI sites of pME18S vector.A DNA fragment encoding BVDV NS3, NS2-3, NS3/4A, andNS5Awas generated from the BVDVNose strain (genotype 1a;GenBank data base accession number AB078951) by Super-Script III One-Step RT-PCR System with Platinum Taq HighFidelity (Invitrogen) using primers that incorporated NdeI andPstI sites at the 5� and 3� ends, respectively. The fragments werethen cloned into pME18S-Myc using NdeI and PstI sites togenerate pME-NS3, pME-NS2-3, pME-NS3/4A, and pME-NS5A. A series of N-terminal deletion mutants of NS3 wasgenerated by PCR-mediated site mutagenesis using pME-NS3as a template. The fragment of serine protease-negative NS3/4AS2051A was generated by PCR-mediated mutagenesis usingpME-NS3/4A as a template with the mutagenic primer5�-AATATAGGCAGGCCCGCCCATCCCTTCAAGTT-3� tosubstitute Ser2051 to Ala. Hybrid cytomegalovirus enhancer/chicken �-actin (CAG) promoter-driven pME18S vectors,termed pCAG, encoding BVDV NS proteins were constructedby a replacement of the SR� promoter with the CAG promoterfragment from the pCAGGS vector using SspI andXhoI sites ofpME18S vectors. pEF-HCV NS3/4A, which contains the HCVNS3/4A cDNA of the HCV HCR6 strain (genotype 1b; Gen-Bankdata base accessionnumberAY045702) cloned into pEF-1vector (Invitrogen), was kindly provided by Dr. MichinoriKohara. All constructs were confirmed by sequencing with anABI PRISM 3150 genetic analyzer (Applied Biosystems, Tokyo,Japan).

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13649

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

Yeast Two-hybrid Screening—Potential interacting partnersof NS3 were sought using the yeast two-hybrid system accord-ing to the manufacturer’s manual for the MATCHMAKERLibrary Construction and Screening Kit (Clontech, Palo Alto,CA). The N-terminal domain of NS3 (amino acids 1889 to2032) derived from the BVDV Nose strain was amplified byRT-PCR using primers that incorporated NdeI and EcoRI sitesat the 5� and 3� ends, respectively, and cloned into NdeI andEcoRI sites of pGBKT7 in-frame with the Gal4 DNA-bindingdomain to express N-terminal Myc-tagged partial NS3, desig-nated pGBKT7-NS3. For the construction of theMDBK cDNAlibrary, first strand cDNAwas synthesized using random prim-ers from 0.6 �g of mRNA, which was purified from total RNAusing the oligotex-dT30�Super� mRNA Purification Kit(TaKaRa, Shiga, Japan), and double-stranded cDNA amplifiedby 22 cycles long distance (LD) PCR as described in manufac-turer’s protocol. Saccharomyces cerevisiae strain AH109 wastransformed with the bait plasmid pGBKT7-NS3, and selectedin synthetic medium lacking tryptophan. A positive clone har-boring pGBKT7-NS3was confirmed to expressN-terminal 142amino acids of theNS3 protein with anti-MycmAb byWesternblot analysis (data not shown). TheMDBKcell double-strandedcDNA library together with SmaI-linearized pGADT7-Rec(Clontech) was cotransformed in an AH109 clone harboringpGBKT7-NS3 to clone cDNA into the GAL4 AD expressionvector pGADT7-Rec by homologous recombination. Thetransformed yeast cells were grown on agar plates of syntheticmedium lacking histidine, leucine, and tryptophan containing20 �g/ml 5-bromo-4-chloro-3-indolyl-�-O-galactopyranoside(X-�-gal). A total of 145 clones were identified from 1 � 106colonies screened in the library. The insert DNA fragments ofisolated clones were amplified by PCR using LD-Insert Screen-ing Amplimer Sets (Clontech) according to the manufacturer’sprotocol, and then determined by sequencing.Transfection and Immunoprecipitation—LB9.K cells were

transiently transfected using Lipofectamine 2000 (Invitrogen)as described in the manufacturer’s protocol. Transfection effi-ciency of LB9.K cells was typically 80–90%. LB9.K cells wereseeded onto 6-well tissue culture plates 24 h before transfec-tion. Cells were then transfected with 4�g of plasmids per well.At 24 h post-transfection, cells were washed twice with ice-coldphosphate-buffered saline (PBS) and scraped into 0.2ml of lysisbuffer (50mMTris-HCl, pH 7.5, 150mMNaCl, 1mMEDTA, 1%Triton X-100, 20 mM sodium fluoride, 1 mM Na3VO4) supple-mented with Complete protease inhibitor mixture (RocheDiagnostics). The lysates equalized with the same amount ofproteins were immunoprecipitated with 3 �g of anti-FLAG,anti-Myc, anti-BVDVNS3, or controlmouse IgG1mAbs for 2 hat 4 °C, respectively. The immune complexes were precipitatedby incubationwith proteinG-Sepharose beads (GEHealthcare)for another 1 h. The agarose beads were washed four times with1 ml of wash buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1mM EDTA, 0.1% Triton X-100, 20 mM sodium fluoride, 1 mMNa3VO4). The immunoprecipitates were separated by SDS-PAGE and transferred to nitrocellulose membranes (Bio-Rad),probed with antibodies, and immunocomplexes detected byenhanced chemiluminescence (ECL). Antibodies used were:horseradish peroxidase-conjugated mAbs against FLAG

(1:1000 dilution; Sigma) and Myc (1:1000; Santa Cruz Biotech-nology), and a rabbit pAb against NS3 (1:3000). Images weretaken by LAS-4000mini image analyzer system (Fujifilm,Tokyo, Japan).Subcellular Fractionation—Cells were harvested into lysis

buffer lacking Triton X-100, sonicated, and centrifuged at1,000 � g for 10 min. Subcellular fractionation was performedby sequential centrifugation as described previously (19). Inbrief, postnuclear supernatants were centrifuged at 17,000 � gfor 15min to obtain the innermembrane fraction. The resultingsupernatants were centrifuged at 100,000 � g for 1 h to obtaincytosolic and pelleted plasma membrane fractions. The pelletcontaining inner or plasmamembrane was resuspended in lysisbuffer (volume comparable with supernatant) and sonicated.Sphingosine Kinase Assay—Sphingosine kinase activity was

determined as described previously (20). The labeled S1P wasseparated by TLC on Silica Gel G-60 (Whatman) with 1-buta-nol/ethanol/acetic acid/water (80:20:10:20, v/v) and visualizedby autoradiography. The radioactive spots corresponding toS1P were scraped and counted in a scintillation counter.Generation of Recombinant Bovine Sphingosine Kinase 1—

pFlag-SphK1 was transfected into HEK293 cells using Lipo-fectamine 2000 (Invitrogen) to express the Flag-SphK1, whichwas subsequently purified by binding to FLAG(M2)-Sepharose(Sigma), followed by elution with the FLAG peptide (0.2mg/ml). The eluted Flag-SphK1 was concentrated using anUltrafree-0.5 Centrifugal Filter Device (50,000 Da cutoff; Milli-pore, Billerica, MA) and diluted in the sphingosine kinasebuffer (20 mM Tris-HCl, pH 7.4, 20% glycerol, 1 mM �-mercap-toethanol, 1 mM EDTA, 15 mM sodium fluoride, 20 mMNa3VO4, and 0.5 mM 4-deoxypyridoxine) supplemented with aComplete protease inhibitormixture (RocheDiagnostics). Thisprocedure was repeated five times to reduce the concentrationof the FLAG peptide.Generation of Glutathione S-Transferase (GST) Fusion

Proteins—NS3, NS3/4A, and NS5A sequences of BVDV Nosestrain were amplified by PCR using plasmid pME-NS3/4A orpME-NS5Aas a template. ThePCRproduct ofNS3,NS3/4A, orNS5A was cloned into bacterial expression vector pGEX5X-2using SmaI andNotI sites (NS3 andNS3/4A) or EcoRI andNotIsites (NS5A). HCVNS3 and NS3/4A sequences were amplifiedby PCR using plasmid pEF-HCV NS3/4A as a template. ThePCR products were then cloned into pGEX5X-1 using EcoRIand XhoI sites. BVDV and HCVNS proteins were expressed inEscherichia coliBL21 as GST fusion proteins at theN terminus.Overnight cultures were grown with shaking at 37 °C in Luria-Bertani broth containing 50 �g/ml ampicillin and 20 �g/mlchloramphenicol. The culture was then diluted into freshLuria-Bertani broth containing 50 �g/ml ampicillin and 20�g/ml chloramphenicol, and grown with shaking at 37 °C to anA600 of 0.6–1.0. Expression of theGST fusion proteinswas theninduced with 1.2 mM isopropyl �-D-thiogalactopyranoside, andthe cultures were incubated with shaking at 37 °C for a further3 h. The bacterial cells were then harvested by centrifugation at6,000 � g for 10 min at 4 °C, resuspended in 10 ml of GST-soluble buffer (40 mM Tris-HCl, pH 7.5, 5 mM EDTA, 0.5%Triton X-100), and lysed by sonication. The lysates were mixedwell and centrifuged at 20,000� g for 20min at 4 °C. The result-

BVDV NS3 Inhibits Sphingosine Kinase

13650 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

ant clarified bacterial lysate was then incubated with GSH-Sepharose 4B for 2 h at 4 °C with constant mixing. Subse-quently, the GSH-Sepharose beads (with bound protein) werepelleted by centrifugation at 3,000 � g for 5 min at 4 °C andwashed five times inGST-soluble buffer. These beadswere theneither used directly in a pull-down assay, or the GST fusionproteins were eluted by incubationwith cold PBS containing 10mM GSH for 30 min with constant mixing. This elution proce-dure was repeated three times. Eluted proteins were concen-trated by using Ultrafree-0.5 Centrifugal Filter Device (10,000Da cutoff; Millipore).Immunofluorescence Microscopy—LB9.K cells were seeded

on an eight-well chamber slide (Nunc, Roskilde, Denmark) at2 � 104 per well 24 h before transfection. Nontransfected cellsor the cells transfected with Flag-SphK1 were inoculated withBVDV as described in the figure legends. At 18 h p.i., cells werewashed twice with PBS, fixed with PBS containing 4%paraformaldehyde, permeabilized with PBS containing 0.5%Triton X-100, and blocked with PBS containing 10% bovineserum albumin for 10 min. Nontransfected cells were thenincubated with anti-calnexin pAb and mouse anti-BVDV NS3mAb for 1 h followed by incubation with Alexa 568-conjugatedgoat anti-rabbit IgG andAlexa 488-conjugated goat anti-mouseIgG antibodies for 1 h at room temperature. Transfected cellswere double-stained with mouse anti-FLAG mAb (IgG1) andmouse anti-BVDV NS3 mAb (IgG2a) followed by Alexa 488-conjugated goat anti-mouse IgG1 and Alexa 594-conjugatedgoat anti-mouse IgG2a antibodies, or with mouse anti-FLAGmAb and anti-calnexin pAb followed by Alexa 488-conjugatedgoat anti-mouse IgG andAlexa 568-conjugated goat anti-rabbitIgG antibodies. Cells incubatedwith secondary antibodies werethen washed three times with PBS, mounted in Dako fluores-cent mounting medium (Dako Corporation, Carpinteria, CA),then sealed and observed under an LSM 510 microscope (CarlZeiss, Tokyo, Japan).Measurement of S1P Synthesis—LB9.K cells transiently

transfected with pCAG vectors encoding BVDVNS proteins orMDBK cells infected with BVDV were incubated for 4 h in

FIGURE 1. Specific interaction between SphK1 and BVDV NS3 or NS2-3.A, schematic representation of bovine SphK1. Functional domains (28, 37),diacylglycerol (DAG) catalytic domain, sphingosine-binding domain (SphBD),

and calmodulin-binding domain (CaM) are shown. Partial SphK1 cDNAsequence isolated from a positive clone is indicated by a two-headed arrow.B, LB9.K cells were transiently transfected with empty vector or SphK1 expres-sion vector (WT or G177D). At 24 h post-transfection, cells were harvested andcytosolic SphK activity measured as described under “Experimental Proce-dures.” Data in the graph are the mean � S.D. from three independent exper-iments, each performed in duplicate. C, shown on the lower panel is an auto-radiogram of a TLC plate used for separation of S1P. The arrowhead indicatesthe location of S1P visualized with molybdenum blue spray. The upper panelshows the expression of Flag-SphK1 by Western blotting with anti-FLAGmAb. D, Flag-SphK1 expression vector in combination with either an emptyvector, Myc-tagged BVDV NS5A, NS2-3, or NS3 were cotransfected into LB9.Kcells. Proteins immunoprecipitated (IP) with anti-Myc mAb (even lanes) or iso-type control IgG (Isotype Ct, odd lanes) were subjected to Western blotting(IB) using anti-FLAG mAb. E, 1 h after inoculation with either KS86-1cp orKS86-1ncp at an m.o.i. of 5, Flag-SphK1 expression vector was transfected intoLB9.K cells. At 24 h p.i., BVDV NS3 and/or NS2-3 protein was immunoprecipi-tated with isotype control IgG or anti-NS3 mAb as indicated. Immunoprecipi-tated proteins were subjected to Western blotting using anti-FLAG mAb.F, 24 h after infection with either KS86-1cp or KS86-1ncp at an m.o.i. of 5 inMDBK cells, NS3 and/or NS2-3 protein was immunoprecipitated with isotypecontrol IgG or anti-NS3 mAb as indicated. Immunoprecipitated proteins weresubjected to Western blotting using anti-SphK1 pAb. These data were repre-sentative of at least three independent experiments.

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13651

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

phosphate-free DMEM (Invitrogen), then labeled with freshphosphate-free DMEM containing [32P]orthophosphate (0.2mCi/ml) and incubated for 4 h at 37 °C in a humidified 5% CO2atmosphere. Cells were then scraped on ice into 400�l ofmeth-anol, 1 M NaCl, 5 M NaOH (100:100:3, v/v), then 200 �l ofchloroform added. Samples were vortexed thoroughly and cen-trifuged at 14,000 � g for 5 min. The upper aqueous phasecontaining S1P was transferred to a new tube, and acidifiedthrough addition of 20 �l of 1 M HCl and 400 �l of chloroform/methanol/HCl (100/200/1, v/v). Samples were vortexed thor-oughly and phases separated by addition of 120 �l of chloro-form and 120 �l of 2 M KCl. After centrifugation, the lowerorganic phase was dried under vacuum and resuspended inchloroform, and resolved by TLC as described above. Theradioactive spots corresponding to S1P were scraped from theplates and counted in a scintillation counter.RNA Interference—Duplex siRNAs were purchased from

Invitrogen. The siRNA sequence targeting SphK1 was 5�-GCAGUGGCCGCUUCUUUGAACUAUU-3� (sense) and 5�-AAUAGUUCAAAGAAGCGGCCACUGC-3� (antisense), corre-sponding to 634–658 relative to the first nucleotide of the startcodon. The sequence used for scrambled control siRNA was5�-GCAGGCCCGUUUCUUAGCAUUGAUU-3� (sense) and5�-AAUCAAUGCUAAGAAACGGGCCUGC-3� (antisense).LB9.K cells were transfected with 20 nM siRNAusing siLentfect(Bio-Rad) according to the manufacturer’s protocol.Quantitative Real-time RT-PCR—cDNA synthesis was per-

formed with the PrimeScript RT Reagent Kit (TaKaRa) accord-ing to the manufacturer’s protocol. GAPDH mRNA and viral

RNA were quantified using PowerSYBR Green PCR Master Mix(Applied Biosystems) as previouslydescribed (21).Apoptosis Assay—The DEVDase

activity assay was performed asdescribed previously (22) using Ac-DEVD-AMC as a substrate.

RESULTS

IdentificationofSphK1asaBindingPartner of BVDV NS3 and NS2-3—To identify potential cellular bind-ing partners of BVDV NS3, we con-ducted a yeast two-hybrid screenusing the N-terminal 142 aminoacids of NS3 as bait, and isolated acDNA clone encoding partialSphK1 from 1 of 145 total positivecolonies. The cDNA sequenceencoded 164 C-terminal aminoacids of SphK1 (Fig. 1A). Employingthe cDNA sequence for bovineSphK1 from theGenBank data base,specific primers were designed andused to clone a bovine SphK1 byRT-PCR cloning using total RNAisolated from MDBK cells. Subse-quently, a catalytically inactive form

of bovine SphK1 was constructed by substituting aspartic acid(D) for glycine (G) at position 177 in theATP-binding site of thediacylglycerol catalytic domain, according to the previous study(23). This SphK1G177D was used for further functional studies.To investigate whether the cloned bovine SphK1 encodes abona fide SphK, LB9.K cells were transiently transfected withexpression vectors containing FLAG-tagged SphK1. Similar tothe previous study (24), SphK activity in cell lysates from LB9.Kcells transiently transfected with SphK1 was increased �300-fold (Fig. 1, B andC). By comparison, expression of catalyticallyinactive SphK1G177D produced no detectable increase in SphKactivity. Western blot analysis using anti-FLAG antibodyrevealed a specific protein band with an apparent molecularmass consistent with the predicted size (�55 kDa) of FLAG-tagged SphK1, which was absent in vector-transfected cells(Fig. 1C).To confirm the interaction between NS3 and SphK1, we

cotransfected Flag-SphK1 andMyc-NS3 in LB9.K cells, immu-noprecipitated NS3 using an anti-Myc mAb, and determinedwhether SphK1 was coprecipitated with NS3 by Western blot-ting. We also cotransfected an empty vector or NS5A as a neg-ative control and uncleaved NS2-3. Both NS3 and NS2-3, butneither NS5A nor vector control, coprecipitated with SphK1(Fig. 1D). We attempted to express Myc-tagged NS2 in thesame way as above to demonstrate that NS2 does not mediatethe binding of NS2-3 to SphK1, but failed to express detectablelevels of NS2 protein, most likely due to its instability in LB9.Kcells (data not shown).

FIGURE 2. Colocalization of SphK1 with NS3 or NS2-3 in membrane fractions of mammalian cells. LB9.Kcells were transiently transfected with Flag-SphK1 expression vector. After 6 h, cells were inoculated with CPBVDV or NCP BVDV and further incubated for 18 h. Transfected cells were immunostained with (A) anti-FLAGmAb (green) and anti-BVDV NS3 mAb (red), (B) anti-BVDV NS3 mAb (green) and anti-calnexin pAb (red), and (C)anti-FLAG mAb (green) and anti-calnexin pAb (red). D, MDBK cells were mock-infected or infected with eitherKS86-1cp or KS86-1ncp at an m.o.i. of 5. At 20 h p.i., cells were lysed and subcellularly fractionated into cytosol(S), inner membrane (IM) (mitochondria, endoplasmic reticulum, and Golgi), and plasma membrane (PM), asdescribed under “Experimental Procedures.” Equal volumes of lysates were subjected to Western blotting withanti-SphK1 pAb and antibodies against GAPDH, Calnexin, or junctional adhesion molecule (JAM-1) as specificorganelle markers.

BVDV NS3 Inhibits Sphingosine Kinase

13652 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

To confirm the specific interaction of SphK1 with NS3and/or NS2-3 expressed in the context of BVDV infection, wetransfected an expression vector encoding Flag-SphK followingvirus infections, immunoprecipitated NS3 and NS2-3 withanti-NS3 mAb, and determined binding to SphK1 by Westernblotting. When expressed in the context of BVDV infection,Flag-SphK1 coprecipitated with NS3 and NS2-3 expressed inCP BVDV-infected cells as well as NS2-3 in NCP BVDV-in-fected cells (Fig. 1E). To further demonstrate whether NS3 andNS2-3 fromBVDV infection interacts with endogenous SphK1,we immunoprecipitated NS3 and NS2-3 with anti-NS3 mAbfrom the lysates of the BVDV-infected MDBK cells, and deter-mined binding to SphK1 byWestern blotting using anti-SphK1

pAb. As shown in Fig. 1F, specific interaction of endogenousSphK1 with NS3 and NS2-3 was detected.NS3 and NS2-3 Colocalize with SphK1 in Membrane

Fractions—Previous studies indicated that BVDV NS3 andNS2-3 are predominantly localized to the endoplasmic reticu-lum in cultured cells (25) and SphK1 is diffusely distributed inthe cytoplasm (19). To investigate the localization of these pro-teins in BVDV-infected LB9.K cells, we transfected the cellswith an expression vector encoding Flag-SphK1 and performedimmunostaining using anti-FLAG and anti-NS3 mAbs. Forpositive identification of the endoplasmic reticulum,we stainedcells with a calnexin pAb. The staining patterns of NS3 andNS2-3 (CP BVDV) or NS2-3 (NCP BVDV) overlapped with

FIGURE 3. Identification of the NS3-binding site in SphK1. A, schematic representation of SphK1 and its deletion mutants. Myc-NS3 expression vector incombination with expression vectors for Flag-SphK1 and its mutants were cotransfected into LB9.K cells. Proteins immunoprecipitated (IP) with anti-FLAG mAb(even lanes) or isotype control IgG (Isotype Ct; odd lanes) were subjected to Western blotting (IB) using anti-Myc mAb. B, to further determine the critical aminoacids of SphK1 for specific binding to NS3, deletion mutants ranging from the N-terminal 325–386 amino acids were immunoprecipitated with Flag-SphK1.C, according to the result obtained in B, deletion mutants ranging from the N-terminal 349 –361 amino acids were immunoprecipitated with Flag-SphK1. Thedata are representative of at least three independent experiments.

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13653

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

those of calnexin (Fig. 2B) and partially with those of SphK1(Fig. 2A). SphK1 was also partially colocalized with calnexin(Fig. 2C). We further examined the localization of endogenousSphK1 and NS3 by subcellular fractionation. The majority ofSphK1 and NS3 were found to localize in inner and plasmamembrane fractions in MDBK cells (Fig. 2D). Although previ-ous studies have shown that SphK1 in many different cell typesis mainly cytosolic (26, 27), these data suggest that bovineSphK1 localizes to both cytosolic and membrane fractions.Mapping the NS3-binding Domain on SphK1—Deletion

mutagenesis of SphK1was performed to identify theNS3-bind-ing domain on SphK1 (Fig. 3A). FLAG-tagged SphK1 mutantswere coexpressedwithMyc-NS3 and immunoprecipitatedwithanti-FLAGmAb.Myc-NS3 coprecipitatedwith SphK1mutantscontaining a region downstream of calmodulin, which con-sisted of amino acid residues 326–386. This region was alsopresent in the partial SphK1 cDNA sequence isolated from thepositive yeast colony in our yeast two-hybrid screen (Fig. 1A).FLAG-tagged SphK1 C-terminal deletion mutants of thisregion were constructed to further determine the NS3-bindingdomain. The SphK1 mutant consisting of amino acid residues1–361 coprecipitated with Myc-NS3 but the mutant with resi-dues 1–349 did not (Fig. 3B). Furthermore, the deletionmutantcontaining amino acids 1–361, but not 1–358, coprecipitatedwith NS3 (Fig. 3C), suggesting that amino acids 359–361 ofSphK1, or the protein folding affected by these residues, arecritical for its binding to NS3.Mapping the SphK1-binding Domain on NS3—Because the

N-terminal 142 amino acid residues of NS3 were sufficient tobind to SphK1 in our yeast two-hybrid screen, we constructedN-terminal deletionmutants of NS3 to determine the region ofNS3 that bind to SphK1 (Fig. 4A). Myc-tagged NS3 mutantswere coexpressed with Flag-SphK1 and immunoprecipitatedusing the anti-Myc mAb. Flag-SphK1 coimmunoprecipitatedwith the dN1mutant lacking amino acids 1–32, but notwith thedN2 mutant lacking amino acids 1–66 (Fig. 4, A and B). Asexpected from the yeast two-hybrid screen, theN1mutant con-taining NS3 amino acids 1–142 coimmunoprecipitated withSphK1, suggesting that amino acids 33–66 of NS3 are criticalfor its binding to SphK1.The Dose-dependent Inhibition of SphK1 Activity by NS3—

Weperformed in vitro SphK assays in the presence and absenceof NS3 to assess the functional significance of NS3 to SphK1.GST fusion proteins GST-NS3 and GST-NS5A, and GST alonewere expressed and purified from E. coli strain BL21 (Fig. 5A).We failed to express GST-NS2-3, most likely due to toxicity ofits highly hydrophobic NS2 domain (7). Recombinant bovineSphK1 (rSphK1) was purified from lysates of HEK293 cellstransiently transfected with the FLAG-tagged SphK1 expres-sion vector (Fig. 5A). Glutathione (GSH)-Sepharose beads wereused to bind to and precipitate GST-NS3, GST-NS5A, or GSTalone in the presence of rSphK1. The GST pull-down experi-ments confirmed a specific interaction between rSphK1 andGST-NS3 (Fig. 5B). Although GST-NS5A and GST aloneshowed no specific binding to rSphK1 (Fig. 5B) and had nosignificant effect on SphK1 activity, GST-NS3 inhibited SphK1catalytic activity dose-dependently with maximal inhibition

(�90%) attained using above a 5-fold molar excess of NS3 toSphK1 (Fig. 5C).Suppression of SphK1 Activity by NS3, NS2-3, and NS3/4A—

Uncleaved NS2-3 and NS3 are expressed in CP BVDV-infectedcells, whereas only NS2-3 is detectable in NCP BVDV-infectedcells after 9 h p.i. (7). Therefore, we assessed possible differ-ences in the regulation of SphK1 activity among NS3, NS2-3,and NS3/4A. Plasmids encoding BVDV NS3, NS2-3, andNS3/4A proteins were transiently transfected into LB9.K cells,and the endogenous SphK activity in the cell lysate was meas-ured using the in vitro SphK assay. Transfection with variousdoses of plasmid encoding NS3 exhibited a dose-dependentreduction in SphK activity by up to 30% (Fig. 5D). Cells trans-fected with plasmids encoding NS2-3 and NS3/4A showedendogenous SphK activity that was reduced by �15 and 30%,respectively (Fig. 5E). It seemed that the expression of theNS4Acofactor does not affect the inhibitory effect of NS3 on SphK1activity. To further determine whether inhibition of SphK1activity byNS3depends on its serine protease activity or not, weexamined the effect of the S2051A NS3/4Amutant, whose ser-ine protease activity is eliminated and the protease-dependentcleavage between NS3 and NS4A is deficient (3), on SphK1inhibition. As shown in Fig. 5E, S2051A mutant efficientlyinhibited endogenous SphK1 activity to the same extent aswild-type NS3/4A. In addition, SphK1 exhibited no putativeNS3/4A recognition sites (3) and no detectable degradationwhen coexpressed with Myc-NS3/4A (data not shown), sug-

FIGURE 4. Mapping of the SphK1-binding site on BVDV NS3. A, schematicrepresentation of NS3 and its deletion mutants. B, SphK1 expression vector incombination with expression vectors of Myc-NS3 and its deleted mutantswere cotransfected into LB9.K cells. Proteins immunoprecipitated (IP) withanti-Myc mAb (even lanes) or isotype control IgG (Isotype Ct; odd lanes) weresubjected to Western blotting (IB) using anti-FLAG mAb. Data shown in eachpanel are representative of at least three independent experiments.

BVDV NS3 Inhibits Sphingosine Kinase

13654 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

FIGURE 5. NS3 inhibits SphK1 activity in vitro. A, Coomassie Brilliant Blue-stained SDS-PAGE of the purified proteins as indicated above the lanes(Flag-SphK1 and GST-NS proteins). B, GST, GST-NS3, and GST-NS5A bound to GSH-Sepharose was incubated with 1 �M purified recombinant Flag-SphK1for 2 h. Beads were washed four times with wash buffer, then SphK1 associated with GST-NS3 was resolved by SDS-PAGE and detected by Westernblotting with anti-FLAG mAb (upper panel). GST-NS3 and GST-NS5A bound to GSH beads were visualized by blotting with anti-GST mAb (lower panel).C, the effect of GST-NS3, GST-NS5A, or GST control on the catalytic activity of purified recombinant bovine SphK1 (10 nM) was determined using the invitro SphK assay over a range of increasing concentrations of GST-NS proteins or GST. Shown on the left panels are autoradiograms of TLC plates usedfor separation of S1P. The arrowhead indicates the location of S1P. Data in a graph on the right panel represent the means � S.D. from three independentexperiments. D, increasing doses (0.25, 0.5, and 1 �g) of pCAG empty vector or pCAG vector encoding Myc-tagged NS3 were transfected into LB9.K cells.At 24 h post-transfection, cell lysates were subjected to the in vitro SphK assay as described under “Experimental Procedures.” E, pCAG vector encodingMyc-tagged NS3, NS2-3, NS3/4A, or the S2051A NS3/4A mutant, or empty vector was transfected into LB9.K cells. At 24 h post-transfection, cell lysateswere harvested and subjected to the in vitro SphK assay. Inset shows expression levels of NS proteins by Western blotting with anti-Myc mAb. F, LB9.Kcells transfected with empty vector or pCAG-NS3 were labeled with [32P]orthophosphate at 24 h post-transfection for 4 h, then harvested and analyzedfor endogenous S1P synthesis as described under “Experimental Procedures.” G, MDBK cells were mock-infected or infected with CP BVDV or NCP BVDVat an m.o.i. of 5. After 24 h p.i., cells were harvested into lysis buffer and immunoprecipitated by anti-NS3 mAb for 2 h followed by incubation with ProteinG-Sepharose beads for 1 h. Beads were then extensively washed, mixed with 10 nM purified recombinant bovine SphK1, and subjected to the in vitroSphK assay. Inset shows expression levels of NS3 and NS2-3 proteins bound to beads by Western blotting with anti-NS3 mAb. Data (mean � S.D.) shownin each panel are representative of at least three independent experiments. p values were determined for the experimental sample versus the control-treated sample using Student’s t test. **, p � 0.01; *, p � 0.05.

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13655

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

gesting that SphK1 inhibition by NS3 is independent of its ser-ine protease activity. In the cells transfected with the NS3expression plasmid, the endogenous S1P synthesis was reducedby �15% (Fig. 5F). The weaker inhibition of SphK1 by NS2-3appeared to be the result of lower expression levels of NS2-3relative toNS3 andNS3/4A (Fig. 5E, inset). However, the higherlevel of NS2-3 obtained from lysates of NCP BVDV-infectedMDBK cells by immunoprecipitation was shown to signifi-cantly inhibit SphK activity by �50%, similar to the inhibitionobserved with the NS2-3 and NS3 from CP BVDV-infectedMDBK cell lysates (Fig. 5G). These results suggest that SphK1activity is down-regulated by NS2-3 as well as NS3.Down-regulation of SphK1 Activity and S1P Synthesis in

BVDV-infected Cells—The kinetics of endogenous SphK1activity was examined in the context of BVDV infection. At 6and 12 h p.i., SphK activity in MDBK cells infected with bothBVDV biotypes was unaffected or slightly up-regulated. How-ever, at later than 18 h p.i., a time-dependent decrease in SphKactivity was observed, which culminated tomaximal reductionsof 30 and 50% at 48 h p.i. for cells infected with NCP BVDV andCP BVDV, respectively (Fig. 6A). No change in SphK1 mRNAlevels was observed inMDBK cells at 24 h p.i. with either BVDVbiotype (data not shown). Therefore, it appears unlikely thatSphK1 activity is down-regulated at the transcriptional level; itprobably occurs concomitantly with the accumulation of newlysynthesized NS3 and NS2-3 proteins in the cells. Cellular S1Psynthesis levels were reduced by 10 and 20% in CP BVDV- andNCP BVDV-infected cells, respectively, as SphK activitydecreased (Fig. 6B).Overexpression of SphK1 Reduced Apoptosis Induced by CP

BVDV Infection—Our previous studies showed that apoptoticcell death induced by CP BVDV infection is mediated throughactivation of DEVDase activity (9, 22). Because several previousstudies have demonstrated anti-apoptotic effects of SphK1 (24,28, 29), we tested whether inhibition of SphK1 by BVDV hadany influence in the context of apoptotic induction by overex-pression of SphK1 and catalytically inactive SphK1G177D. Over-expression of SphK, but not SphK1G177D, resulted in an �40%

reduction in DEVDase activity in CP BVDV-infected cells,whereas almost no reduction was observed inmock-infected orNCP BVDV-infected cells (Fig. 6C). Because overexpression ofSphK1hadno significant effect on viral replication (Fig. 7,E andF) but counteracted the activation of DEVDase, these data sug-gest that apoptotic cell death inCPBVDV-infected cellsmay beexaggerated by the inhibition of SphK.Inhibition of SphK1 Activity Significantly Enhanced Viral

Replication—To examine the influence of SphK activity onBVDV replication, we measured intracellular viral RNA levelsin the presence of SKI, whichwas identified as a highly selective,noncompetitive inhibitor of SphK (16). SKI treatmentincreased viral RNA levels of both CP BVDV and NCP BVDVinfections �2-fold in a dose-dependent manner (Fig. 7A). Tofurther analyze the effect of SphK1 inhibition on BVDV repli-cation more specifically, LB9.K cells were either depleted ofSphK1 by siRNA-mediated knockdown or by overexpression ofthe catalytically inactive SphK1G177D, and then infected withBVDV. Transfection with siRNA significantly inhibited endog-enous SphK1 expression levels (Fig. 7B) as well as endogenousSphK1 activity in LB9.K cells by �50% compared with levels inscrambled siRNA-transfected cells (Fig. 7C). Viral RNA levelsfrom both CP BVDV and NCP BVDV infections increased�2-fold compared with mock-treated or scrambled siRNA-transfected cells (Fig. 7D). Transfection with SphK1G177D alsoincreased viral RNA levels and titers concomitantly by over160% (Fig. 7, E and F). At 48 h p.i., the viral RNA level from CPBVDV infections was 2.5-fold in cells overexpressingSphK1G177D (data not shown). In contrast, transfection withwild-type SphK1, reaching a 300-fold increase in SphK activity,did not affect viral RNA levels and titers (Fig. 7, E and F). Wenext examined the effect of FCS addition on BVDV replicationas FCS is known to mediate SphK activation (30). FCS additionresulted in a dose-dependent increase in endogenous SphK1activity (Fig. 7G), and the viral RNA level was reduced by up to40% in the presence of 10% FCS and the effect was reversiblewith the addition of SKI (Fig. 7H). We also examined the effectof S1P, a metabolite of SphK, on viral RNA replication. How-

FIGURE 6. Kinetics of sphingosine kinase activity and the effect of SphK1 overexpression in BVDV-infected cells. A, MDBK cells infected with eitherKS86-1cp or KS86-1ncp at an m.o.i. of 5 were harvested at the indicated time points p.i., and SphK activity was determined by the in vitro SphK assay as describedunder “Experimental Procedures.” B, MDBK cells infected with CP BVDV or NCP BVDV were labeled with [32P]orthophosphate at 18 h p.i. for 4 h, harvested, andanalyzed for endogenous S1P synthesis as described under “Experimental Procedures.” Data are expressed as percent relative to the mock-infected samples.C, LB9.K cells were transiently transfected with empty vector, or SphK1 (WT) or SphK1G177D (G177D) expression vector. After 24 h, cells were infected asdescribed above in B. At 24 h p.i., cells were harvested and the cell lysates assessed for DEVDase activity. Data are expressed as percent relative to themock-infected samples transfected with empty vector. The data represent the mean � S.D. from at least three independent experiments. **, p � 0.01; *, p � 0.05.

BVDV NS3 Inhibits Sphingosine Kinase

13656 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

ever, addition of 1–10�MS1Pdid not affect the viral RNA levels(data not shown). These data collectively suggest that SphKinhibition may allow for enhanced BVDV replication, whereasit appears that S1P does not regulate BVDV replication.

HCV NS3 Does Not RegulateSphK1 Activity—Because BVDVNS3 was shown to inhibit bovineSphK1 activity, it was of interest todetermine whether NS3 fromHCV,a close relative of BVDV, regulatesthe catalytic activity of hSphK1. Wethus examined the effect of purifiedrecombinant HCV NS3 and NS3/4A expressed in E. coli (Fig. 8A) onthe catalytic hSphK1 activity invitro. Neither GST-HCV NS3 norNS3/4A had a significant effect onhSphK1 activity, even at 10-foldmolar excess to hSphK1 (Fig. 8B).Furthermore, transient transfectioninto HEK293 cells with pEF-HCVNS3 or NS3/4A did not affect theendogenous SphK1 activity, com-pared with empty vector-trans-fected cells (data not shown). Takentogether, hSphK1 activity appearsto be regulated neither by HCVNS3nor by HCV NS3/4A. In contrast,BVDV NS3 suppressed hSphK1activity efficiently by�80% at 5-foldmolar excess to hSphK1 (Fig. 8C).Accordingly, disruption of SphK1activity appears to be specific toBVDV NS3.

DISCUSSION

Sphingolipids are predominantcomponents of biological mem-branes that also play pivotal roles insignal transduction pathways. Inparticular, the sphingolipid S1P isan intracellular second messengerand an extracellular ligand withdiverse cellular functions includingsurvival, proliferation, differentia-tion, tumorigenesis, inflammation,and immunity (14, 15). Despite theprofound importance of S1P, noreports have described the directregulation of SphK activity bya virus, although some studiesshowed that SphK activity is up-reg-ulated after infection with humancytomegalovirus or respiratory syn-cytial virus, contributing to efficientviral replication or cell survival,respectively (31, 32). In the presentstudy, we demonstrated that BVDV

NS3 or NS2-3, but not HCVNS3, directly binds to and inhibitsthe catalytic activity of SphK1. Inhibition of SphK1 activity wasalso observed in the context of BVDV infection. This effectseems independent of additional cofactors or auxiliary proteins

FIGURE 7. Effects of SphK1 activity on BVDV replication. A, the effect of SphK1 inhibitor (SKI) on BVDV RNAreplication. After infection with either KS86-1cp or KS86-1ncp at an m.o.i. of 5 for 1 h, MDBK cells were treated with0, 1, and 5 �M SKI. After 48 h p.i., cells were harvested and the BVDV RNA level determined by real-time PCR analysis.B–D, SphK1 knockdown with siRNA enhanced BVDV replication. LB9.K cells were mock-transfected (�) or trans-fected with siRNA targeted to SphK1 (�) or scrambled control siRNA (scr.) at a final concentration of 20 nM. At 24 hpost-transfection, cells were mock-infected or infected with BVDV as described above. Cells were further incubatedfor 24 h, then harvested and assessed for (B) the endogenous SphK1 expression by Western blotting using anti-SphK1 pAb, (C) the endogenous SphK1 activity, and (D) the BVDV RNA level by real-time PCR analysis. E, LB9.K cellswere transiently transfected with empty vector or SphK1 expression vector (wild-type (WT) or G177D). After 24 h,cells were infected as described above. At 24 h p.i., cells were harvested and BVDV RNA level determined by real-timePCR analysis. At the same time, the viral titer (F) in the culture supernatant was assessed. G and H, the effect of SphK1activation by FCS on BVDV RNA replication. MDBK cells were infected as described above, and incubated with DMEMcontaining 0, 2, 5, or 10% FCS, or 10% FCS with 5 �M SKI. After 24 h p.i., cells were harvested and assessed for (G) theendogenous SphK1 activity and (H) the BVDV RNA level determined by real-time PCR analysis. The data areexpressed relative to the control treated data, which were set to 1.0, and represent the mean � S.D. from at leastthree independent experiments, each performed in duplicate. ��, p � 0.01; �, p � 0.05.

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13657

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

because the experiments were performed using purified pro-teins. Studies are currently underway to determine the aminoacid residues of NS3 responsible for SphK1 inhibition.

The involvement of sphingolipids in HCV replication hasbeen well described. Replicase complexes of HCV, a close rela-tive of BVDV, associate with lipid rafts enriched in sphingolip-ids and cholesterol via NS4B, NS5A, and NS5B (12). Althoughthe molecular mechanisms characterizing these raft-viral pro-tein interactions are still largely unknown, a direct associationofHCVNS5Bwith lipid rafts via its finger domain is consideredessential forHCV replication (13). In the presence of a chemicalinhibitor of sphingolipid biosynthesis, the association of NS5Bwith lipid rafts is disrupted and HCV replication is repressed.Because NS5B from HCV shares structural similarity with thatfrom BVDV (33), it is tempting to speculate that BVDV utilizessimilar cellular components for viral replication. Although themolecular mechanisms responsible for enhanced BVDV repli-cation by SphK inhibition are unknown, onemay postulate thatBVDV NS3 may alter the proportion of intracellular sphingo-lipid species through inhibition of SphK1 activity so they aremore suited for efficient viral replication. Although we showedthat HCV NS3 does not inhibit SphK1 activity in this study, itwill be important to determine whether SphK activity is regu-lated by HCV through other mechanisms or not.BVDV NS3 possesses serine protease and helicase activities

that are crucial for efficient viral replication.More importantly,NS3 is abundantly detected in cells infectedwithCPBVDV.Wethus hypothesized that an inhibitory interaction between NS3and SphK1may be a determinant for the induction of cell death.Unexpectedly, uncleaved NS2-3 also interacted with and cata-lytically inhibited SphK1, andNCPBVDV infection suppressedSphK activity. Because inhibitory regulation of SphK1 is con-served between both the CP and NCP biotypes, SphK1 inhibi-tion itself is not directly linked to the induction of cytopathiceffects by BVDV. Previously, we showed that viral cytopatho-genicity ismediated via up-regulation of DEVDase activity trig-gered by intracellular accumulation of double-stranded RNA, areplicative intermediate, resulting from increased viral RNAreplication (9). Therefore, an inhibitory interaction ofNS3withSphK1 seems to have an indirect role in virus-induced celldeath because inhibition of SphK1 activity enhances viral RNAreplication, which leads to an accumulation of double-strandedRNA. The expression of NS3 by itself has been suspected to becorrelated with induction of cell death because abundant levelsof NS3 are detected exclusively in CP BVDV-infected cells.Indeed, a previous study suggested that ectopic expression ofNS3 using an adenovirus-based vector is sufficient for theinduction of apoptosis (34).However, in this study,we observedno changes inmorphology orDEVDase activity in cells express-ing more abundant NS3 relative to CP BVDV-infected cellsusing the CAG promoter-driven expression vector (data notshown). Thus, it appears that cell death occurs via intracellularevents downstream of NS2-3 autoprocessing, presumably as aresult of enhanced RNA replication controlled by NS3 expres-sion (7–9).A pro-survival role of S1P has been demonstrated for cells

under stress induced by trophic deprivation or ceramide addi-tion (24). Additionally, overexpression of SphK1 protects manydifferent types of cells from apoptosis mediated by the corre-sponding stress induction (19, 24, 28). In the present study,reducedDEVDase activitywas observed after overexpression of

FIGURE 8. NS3 from BVDV, but not from HCV, inhibits catalytic activityof hSphK1 in vitro. A, Coomassie Brilliant Blue-stained SDS-PAGE of thepurified proteins as indicated above the lanes (left panel), and correspond-ing Western blotting analysis using anti-HCV NS3 pAb are shown (rightpanel). B, the effect of GST-HCV NS3, GST-HCV NS3/4A, or the GST controlon the catalytic activity of purified recombinant hSphK1 (10 nM) was deter-mined by an in vitro SphK assay over a range of increasing concentrationsof GST-HCV NS proteins or GST. Shown on the upper panel is an autoradio-gram of a TLC plate used for separation of S1P. Data in the graph on thelower panel represent the mean � S.D. from three independent experi-ments. C, the effect of GST-HCV NS3, GST-BVDV NS3, or the GST control onthe catalytic activity of purified hSphK1 (10 nM) was determined by an invitro SphK assay. GST-HCV NS3, GST-BVDV NS3, or GST was added tohSphK1 in 5-fold molar excess. Shown on the left panel is an autoradio-gram of a TLC plate used for separation of S1P. The arrowhead indicatesthe location of S1P. Data in a graph on the right panel are the mean � S.D.from three independent experiments. ��, p � 0.01.

BVDV NS3 Inhibits Sphingosine Kinase

13658 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 284 • NUMBER 20 • MAY 15, 2009

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

SphK inCPBVDV-infected cells. Note that oxidative and endo-plasmic reticulum stresses are common characteristics of CPBVDV infections (35, 36) directly linked to apoptosis. SphK1overexpression in CP BVDV-infected cells could function tocounteract these pro-apoptotic stresses, thereby causingreduced activation of DEVDase. The combined data suggestthat inhibitory interaction of NS3 with SphK1 may have a rolein enhancing BVDV-induced apoptosis.Interactions between S1P and its receptors are being increas-

ingly recognized as important in many aspects of immuneresponses, including lymphocyte chemotaxis. Given the impor-tance of S1P functions in immunity, BVDV-mediated down-regulation of SphK1 that catalyzes the formation of S1P mightpossibly contribute to BVDV pathogenesis. Further studies arerequired to analyze the details of SphK inhibition on lymphoidorgan or immune cells in cattle persistently infected withBVDV. Moreover, identification of sphingolipid species thatare crucial for viral replication or affected through SphK inhi-bition by NS3 or NS2-3 may provide new insights into themechanisms of BVDV pathogenesis.

Acknowledgment—We thank Dr. Michinori Kohara for providing thepEF-HCV NS3 and NS3/4A vectors and anti-HCV NS3 antibody.

REFERENCES1. Shepard, C. W., Finelli, L., and Alter, M. J. (2005) Lancet Infect. Dis. 5,

558–5672. Buckwold, V. E., Beer, B. E., andDonis, R. O. (2003)Antiviral Res. 60, 1–153. Xu, J., Mendez, E., Caron, P. R., Lin, C., Murcko, M. A., Collett, M. S., and

Rice, C. M. (1997) J. Virol. 71, 5312–53224. Li, K., Foy, E., Ferreon, J. C., Nakamura, M., Ferreon, A. C., Ikeda, M., Ray,

S. C., Gale, M., Jr., and Lemon, S. M. (2005) Proc. Natl. Acad. Sci. U. S. A.102, 2992–2997

5. Li, X. D., Sun, L., Seth, R. B., Pineda, G., and Chen, Z. J. (2005) Proc. Natl.Acad. Sci. U. S. A. 102, 17717–17722

6. Chen, Z., Benureau, Y., Rijnbrand, R., Yi, J., Wang, T.,Warter, L., Lanford,R. E., Weinman, S. A., Lemon, S. M., Martin, A., and Li, K. (2007) J. Virol.81, 964–976

7. Lackner, T., Muller, A., Pankraz, A., Becher, P., Thiel, H. J., Gorbalenya,A. E., and Tautz, N. (2004) J. Virol. 78, 10765–10775

8. Vassilev, V. B., and Donis, R. O. (2000) Virus Res. 69, 95–1079. Yamane, D., Kato, K., Tohya, Y., and Akashi, H. (2006) J. Gen. Virol. 87,

2961–297010. Lackner, T.,Muller, A., Konig,M., Thiel, H. J., andTautz,N. (2005) J. Virol.

79, 9746–975511. Aizaki, H., Lee, K. J., Sung, V.M., Ishiko, H., and Lai,M.M. (2004)Virology

324, 450–46112. Gao, L., Aizaki, H., He, J.W., and Lai,M.M. (2004) J. Virol. 78, 3480–3488

13. Sakamoto, H., Okamoto, K., Aoki, M., Kato, H., Katsume, A., Ohta, A.,Tsukuda, T., Shimma, N., Aoki, Y., Arisawa, M., Kohara, M., and Sudoh,M. (2005) Nat. Chem. Biol. 1, 333–337

14. Spiegel, S., and Milstien, S. (2003) Nat. Rev. Mol. Cell Biol. 4, 397–40715. Shida, D., Takabe, K., Kapitonov, D., Milstien, S., and Spiegel, S. (2008)

Curr. Drug Targets 9, 662–67316. French, K. J., Schrecengost, R. S., Lee, B. D., Zhuang, Y., Smith, S. N.,

Eberly, J. L., Yun, J. K., and Smith, C. D. (2003) Cancer Res. 63, 5962–596917. Makino, A., Shimojima,M.,Miyazawa, T., Kato, K., Tohya, Y., andAkashi,

H. (2006) J. Virol. 80, 4482–449018. Nagai, M., Sakoda, Y., Mori, M., Hayashi, M., Kida, H., and Akashi, H.

(2003) J. Gen. Virol. 84, 447–45219. Maceyka, M., Sankala, H., Hait, N. C., Le Stunff, H., Liu, H., Toman, R.,

Collier, C., Zhang, M., Satin, L. S., Merrill, A. H., Jr., Milstien, S., andSpiegel, S. (2005) J. Biol. Chem. 280, 37118–37129

20. Olivera, A., Kohama, T., Tu, Z., Milstien, S., and Spiegel, S. (1998) J. Biol.Chem. 273, 12576–12583

21. Yamane, D., Kato, K., Tohya, Y., andAkashi, H. (2008)Vet.Microbiol. 129,69–79

22. Yamane, D., Nagai, M., Ogawa, Y., Tohya, Y., and Akashi, H. (2005) Mi-crobes. Infect. 7, 1482–1491

23. Pitson, S.M.,Moretti, P. A., Zebol, J. R., Xia, P., Gamble, J. R., Vadas,M.A.,D’Andrea, R. J., and Wattenberg, B. W. (2000) J. Biol. Chem. 275,33945–33950

24. Olivera, A., Kohama, T., Edsall, L., Nava, V., Cuvillier, O., Poulton, S., andSpiegel, S. (1999) J. Cell Biol. 147, 545–558

25. Zhang, G., Flick-Smith, H., and McCauley, J. W. (2003) Virus Res. 97,89–102

26. Kihara, A., Anada, Y., and Igarashi, Y. (2006) J. Biol. Chem. 281,4532–4539

27. Pitson, S. M., Moretti, P. A., Zebol, J. R., Lynn, H. E., Xia, P., Vadas, M. A.,and Wattenberg, B. W. (2003) EMBO J. 22, 5491–5500

28. Taha, T. A., Hannun, Y. A., and Obeid, L. M. (2006) J. Biochem. Mol. Biol.39, 113–131

29. Xia, P.,Wang, L., Gamble, J. R., andVadas,M. A. (1999) J. Biol. Chem. 274,34499–34505

30. Olivera, A., and Spiegel, S. (1993) Nature 365, 557–56031. Machesky, N. J., Zhang, G., Raghavan, B., Zimmerman, P., Kelly, S. L.,

Merrill, A. H., Jr., Waldman, W. J., Van Brocklyn, J. R., and Trgovcich, J.(2008) J. Biol. Chem. 283, 26148–26160

32. Monick, M. M., Cameron, K., Powers, L. S., Butler, N. S., McCoy, D.,Mallampalli, R. K., andHunninghake, G.W. (2004)Am. J. Respir. CellMol.Biol. 30, 844–852

33. Choi, K. H., Groarke, J. M., Young, D. C., Kuhn, R. J., Smith, J. L., Pevear,D. C., and Rossmann, M. G. (2004) Proc. Natl. Acad. Sci. U. S. A. 101,4425–4430

34. St-Louis, M. C., Massie, B., and Archambault, D. (2005) Vet. Res. 36,213–227

35. Jordan, R., Wang, L., Graczyk, T. M., Block, T. M., and Romano, P. R.(2002) J. Virol. 76, 9588–9599

36. Schweizer, M., and Peterhans, E. (1999) J. Gen. Virol. 80, 1147–115537. Sutherland, C.M.,Moretti, P. A., Hewitt, N.M., Bagley, C. J., Vadas,M. A.,

and Pitson, S. M. (2006) J. Biol. Chem. 281, 11693–11701

BVDV NS3 Inhibits Sphingosine Kinase

MAY 15, 2009 • VOLUME 284 • NUMBER 20 JOURNAL OF BIOLOGICAL CHEMISTRY 13659

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from

Kato, Yukinobu Tohya and Hiroomi AkashiDaisuke Yamane, Muhammad A. Zahoor, Yassir M. Mohamed, Walid Azab, Kentaro

for Efficient Viral Replication and CytopathogenesisInhibition of Sphingosine Kinase by Bovine Viral Diarrhea Virus NS3 Is Crucial

doi: 10.1074/jbc.M807498200 originally published online March 17, 20092009, 284:13648-13659.J. Biol. Chem. 

  10.1074/jbc.M807498200Access 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

  http://www.jbc.org/content/284/20/13648.full.html#ref-list-1

This article cites 37 references, 20 of which can be accessed free at

by guest on July 2, 2018http://w

ww

.jbc.org/D

ownloaded from