Top Banner
Transferrin Receptor 1 Facilitates Poliovirus Permeation of Mouse Brain Capillary Endothelial Cells * Received for publication, September 8, 2015, and in revised form, November 17, 2015 Published, JBC Papers in Press, December 4, 2015, DOI 10.1074/jbc.M115.690941 Taketoshi Mizutani 1 , Aya Ishizaka, and Coh-ichi Nihei From the Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN), Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan As a possible route for invasion of the CNS, circulating polio- virus (PV) in the blood is believed to traverse the blood-brain barrier (BBB), resulting in paralytic poliomyelitis. However, the underlying mechanism is poorly understood. In this study, we demonstrated that mouse transferrin receptor 1 (mTfR1) is responsible for PV attachment to the cell surface, allowing inva- sion into the CNS via the BBB. PV interacts with the apical domain of mTfR1 on mouse brain capillary endothelial cells (MBEC4) in a dose-dependent manner via its capsid protein (VP1). We found that F-G, G-H, and H-I loops in VP1 are impor- tant for this binding. However, C-D, D-E, and E-F loops in VP1- fused Venus proteins efficiently penetrate MBEC4 cells. These results imply that the VP1 functional domain responsible for cell attachment is different from that involved in viral perme- ation of the brain capillary endothelium. We observed that co- treatment of MBEC4 cells with excess PV particles but not dex- tran resulted in blockage of transferrin transport into cells. Using the Transwell in vitro BBB model, transferrin co-treat- ment inhibited permeation of PV into MBEC4 cells and delayed further viral permeation via mTfR1 knockdown. With mTfR1 as a positive mediator of PV-host cell attachment and PV perme- ation of MBEC4 cells, our results indicate a novel role of TfR1 as a cellular receptor for human PV receptor/CD155-independent PV invasion of the CNS. Poliovirus (PV) 2 is an enterovirus belonging to the family Picornaviridae and is the causative agent of poliomyelitis (1, 2). Generally, PV enters the stomach via oral ingestion and invades the alimentary mucosa in an unidentified manner, and PV then proliferates in the alimentary mucosa (1, 2) and moves to the bloodstream. The circulating virus invades the CNS and repli- cates in motor neurons (MNs). Poliomyelitis is known to involve accumulated damage to the MNs by PV replication (3). The human PV receptor (hPVR/CD155) facilitates PV infection of cells; however, PV replication is restricted by host immune activities (e.g. IFN-/) (4 – 6). Although wild-type mice are not sensitive to PV (7), hPVR-expressing transgenic (Tg) mice were susceptible to PV via intravenous and intramuscular routes but not the oral route (7–12). Further, an IFN-/-deficient hPVR-Tg mouse was found to be susceptible to PV via the oral route (13). As a possible route for invasion of the CNS, PV enters the CNS via axonal transport through the skeletal muscle in an hPVR-dependent manner (14). Endocytic vesicles at the syn- apse take up intact PV, which is passively transported to the CNS. Interestingly, PV has been shown to invade the CNS via hPVR-independent axonal transport in hPVR-Tg and non-Tg mice (15), indicating that other unidentified pathways for PV transport may be present. Furthermore, we previously showed that PV promptly invades the CNS from the blood in non-Tg mice, which supports this speculation (16). In that study, intra- venously injected PV permeated the brain as fast as cationized rat serum albumin, which is BBB-permeable (16).Therefore, PV is thought to efficiently permeate the CNS by overcoming the BBB. The BBB is composed of a multilayer barrier composed of vascular endothelial cells with tight junctions filling the gaps between cells (17). Although the BBB was discovered over a century ago, its transport mechanisms are not fully understood. It restricts transport of substances between the CNS and blood by maintaining a strictly regulated microenvironment for high integrity neuronal response in the CNS (18, 19). Certain sub- stances are permitted transmission via the BBB from the blood- stream to the brain, facilitated by specific transporters on the cell membrane (e.g. glucose, amino acids, transferrin, and insu- lin) (20 –25). For example, transferrin is known to facilitate iron transport from the blood to the cells (26). Iron uptake increases transferrin affinity for the transferrin receptor on the cell mem- brane. The iron-transferrin complex is transported into the cells by receptor-mediated transcytosis, followed by the release of iron into the cytoplasm; transferrin then goes back to the outer cell membrane for recycling. This mechanism is some- times exploited by viruses for entry during infection (27–31). Given that transferrin receptor is a transporter in brain cap- illary endothelial cells and can be used as an entry receptor for several viral infections, we hypothesized that PV similarly invades the CNS via the BBB by using transferrin receptor as a vehicle. We examined this possibility in this study and demon- strated the interaction of PV with mouse transferrin receptor 1 (mTfR1) in vitro. Furthermore, we identified that VP1, a PV capsid protein, is responsible for the physical interaction * This work was supported by The Japan Society for the Promotion of Science KAKENHI Grants 25670221 and 15K14413 (to T. M.), 15K19117 (to A. I.), and 21790437 and 23659235 (to C. N.) and Japan Agency for Medical Research and Development Grant 15545492 (to T. M.). The authors declare that they have no conflicts of interest with the contents of this article. 1 To whom correspondence should be addressed: Lab. of Virology, Inst. of Microbial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN), Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan. Tel.: 81-3-3441-4173; Fax: 81-3-3441-7589; E-mail: [email protected]. 2 The abbreviations used are: PV, poliovirus; BBB, blood-brain barrier; mTfR1, mouse transferrin receptor 1; MN, motor neuron; hPVR, human PV recep- tor; AGMK, African green monkey kidney; Tg, transgenic; TEER, trans-endo- thelial electrical resistance; AD, apical domain. crossmark THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 291, NO. 6, pp. 2829 –2836, February 5, 2016 © 2016 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. FEBRUARY 5, 2016 • VOLUME 291 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 2829 by guest on January 2, 2020 http://www.jbc.org/ Downloaded from
9

Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

Sep 18, 2019

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

Transferrin Receptor 1 Facilitates Poliovirus Permeation ofMouse Brain Capillary Endothelial Cells*

Received for publication, September 8, 2015, and in revised form, November 17, 2015 Published, JBC Papers in Press, December 4, 2015, DOI 10.1074/jbc.M115.690941

Taketoshi Mizutani1, Aya Ishizaka, and Coh-ichi NiheiFrom the Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN), Tokyo, 3-14-23 Kamiosaki,Shinagawa-ku, Tokyo 141-0021, Japan

As a possible route for invasion of the CNS, circulating polio-virus (PV) in the blood is believed to traverse the blood-brainbarrier (BBB), resulting in paralytic poliomyelitis. However, theunderlying mechanism is poorly understood. In this study, wedemonstrated that mouse transferrin receptor 1 (mTfR1) isresponsible for PV attachment to the cell surface, allowing inva-sion into the CNS via the BBB. PV interacts with the apicaldomain of mTfR1 on mouse brain capillary endothelial cells(MBEC4) in a dose-dependent manner via its capsid protein(VP1). We found that F-G, G-H, and H-I loops in VP1 are impor-tant for this binding. However, C-D, D-E, and E-F loops in VP1-fused Venus proteins efficiently penetrate MBEC4 cells. Theseresults imply that the VP1 functional domain responsible forcell attachment is different from that involved in viral perme-ation of the brain capillary endothelium. We observed that co-treatment of MBEC4 cells with excess PV particles but not dex-tran resulted in blockage of transferrin transport into cells.Using the Transwell in vitro BBB model, transferrin co-treat-ment inhibited permeation of PV into MBEC4 cells and delayedfurther viral permeation via mTfR1 knockdown. With mTfR1 asa positive mediator of PV-host cell attachment and PV perme-ation of MBEC4 cells, our results indicate a novel role of TfR1 asa cellular receptor for human PV receptor/CD155-independentPV invasion of the CNS.

Poliovirus (PV)2 is an enterovirus belonging to the familyPicornaviridae and is the causative agent of poliomyelitis (1, 2).Generally, PV enters the stomach via oral ingestion and invadesthe alimentary mucosa in an unidentified manner, and PV thenproliferates in the alimentary mucosa (1, 2) and moves to thebloodstream. The circulating virus invades the CNS and repli-cates in motor neurons (MNs). Poliomyelitis is known toinvolve accumulated damage to the MNs by PV replication (3).The human PV receptor (hPVR/CD155) facilitates PV infection

of cells; however, PV replication is restricted by host immuneactivities (e.g. IFN-�/�) (4 – 6). Although wild-type mice are notsensitive to PV (7), hPVR-expressing transgenic (Tg) mice weresusceptible to PV via intravenous and intramuscular routes butnot the oral route (7–12). Further, an IFN-�/�-deficienthPVR-Tg mouse was found to be susceptible to PV via the oralroute (13).

As a possible route for invasion of the CNS, PV enters theCNS via axonal transport through the skeletal muscle in anhPVR-dependent manner (14). Endocytic vesicles at the syn-apse take up intact PV, which is passively transported to theCNS. Interestingly, PV has been shown to invade the CNS viahPVR-independent axonal transport in hPVR-Tg and non-Tgmice (15), indicating that other unidentified pathways for PVtransport may be present. Furthermore, we previously showedthat PV promptly invades the CNS from the blood in non-Tgmice, which supports this speculation (16). In that study, intra-venously injected PV permeated the brain as fast as cationizedrat serum albumin, which is BBB-permeable (16).Therefore, PVis thought to efficiently permeate the CNS by overcoming theBBB.

The BBB is composed of a multilayer barrier composed ofvascular endothelial cells with tight junctions filling the gapsbetween cells (17). Although the BBB was discovered over acentury ago, its transport mechanisms are not fully understood.It restricts transport of substances between the CNS and bloodby maintaining a strictly regulated microenvironment for highintegrity neuronal response in the CNS (18, 19). Certain sub-stances are permitted transmission via the BBB from the blood-stream to the brain, facilitated by specific transporters on thecell membrane (e.g. glucose, amino acids, transferrin, and insu-lin) (20 –25). For example, transferrin is known to facilitate irontransport from the blood to the cells (26). Iron uptake increasestransferrin affinity for the transferrin receptor on the cell mem-brane. The iron-transferrin complex is transported into thecells by receptor-mediated transcytosis, followed by the releaseof iron into the cytoplasm; transferrin then goes back to theouter cell membrane for recycling. This mechanism is some-times exploited by viruses for entry during infection (27–31).

Given that transferrin receptor is a transporter in brain cap-illary endothelial cells and can be used as an entry receptor forseveral viral infections, we hypothesized that PV similarlyinvades the CNS via the BBB by using transferrin receptor as avehicle. We examined this possibility in this study and demon-strated the interaction of PV with mouse transferrin receptor 1(mTfR1) in vitro. Furthermore, we identified that VP1, a PVcapsid protein, is responsible for the physical interaction

* This work was supported by The Japan Society for the Promotion of ScienceKAKENHI Grants 25670221 and 15K14413 (to T. M.), 15K19117 (to A. I.), and21790437 and 23659235 (to C. N.) and Japan Agency for Medical Researchand Development Grant 15545492 (to T. M.). The authors declare that theyhave no conflicts of interest with the contents of this article.

1 To whom correspondence should be addressed: Lab. of Virology, Inst. ofMicrobial Chemistry, Microbial Chemistry Research Foundation (BIKAKEN),Tokyo, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan. Tel.:81-3-3441-4173; Fax: 81-3-3441-7589; E-mail: [email protected].

2 The abbreviations used are: PV, poliovirus; BBB, blood-brain barrier; mTfR1,mouse transferrin receptor 1; MN, motor neuron; hPVR, human PV recep-tor; AGMK, African green monkey kidney; Tg, transgenic; TEER, trans-endo-thelial electrical resistance; AD, apical domain.

crossmarkTHE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 291, NO. 6, pp. 2829 –2836, February 5, 2016

© 2016 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

FEBRUARY 5, 2016 • VOLUME 291 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 2829

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

between PV and mTfR1. Co-incubation with transferrin orknockdown of mTfR1 resulted in delayed PV transcytosis viathe brain capillary endothelial cells in the BBB in vitro model.We identified the domain of VP1 responsible for attachment tomTfR1 and permeation of the brain capillary endothelial cells.In summary, we provide convincing evidence to support thedirect involvement of mTfR1 in PV permeation into the CNSvia BBB by using an in vitro model.

Experimental Procedures

Cell Culture—Mouse brain capillary endothelial cells(MBEC4) isolated from BALB/c mice brain cortices andimmortalized by SV40 transformation were maintained (32) inDMEM containing 10% fetal calf serum at 37 °C and 5% CO2.African green monkey kidney (AGMK) cells were grown inDMEM supplemented with 5% newborn calf serum.

Plasmid Construction—The pCold-GST-6His vector wasgenerated by inserting GST fragment (0.3 kb at DraIII-BamHIsites) in pCold-I vector (Takara Bio Ltd., Shiga, Japan), then aHis6 fragment was inserted between the XhoI and NotI restric-tion sites (33). To generate pCold-GST-6His-VP1, -VP2, and-VP3 vectors, all the coding fragments of capsid proteins (VP1,VP2, and VP3) were amplified by PCR, and these fragmentswere introduced at the BamHI/XhoI site in the pCold-GST-6His vector. The pCold-GST-Venus-6His-VP1 and each VP1loop peptide (cDNA) were constructed as follows. To generatethe pCold-GST-Venus-6His-empty vector, the PCR fragmentof 0.7-kb Venus cDNA was inserted into the BamHI/XhoIsite in the pCold-GST-6His vector. The linker sequence(SRG[SGGGG]2SSG) was introduced at XhoI sites in thepCold-GST-Venus-6His vector to generate pCold-GST-Ve-nus-L-6His. The whole cDNA or peptide sequences of VP1were inserted after linker sequence at the EcoRI/XhoI site inpCold-GST-Venus-L-6His. FLAG-mTfR1 and a series of itsdeletion mutants (FLAG-mTfR1– 609, -388 -186, PD2, andAD) were inserted into pTNT vector (Promega Corp., Madison,WI). FLAG-mTfR1 fragment was inserted into a pcDNA3.1-FLAG vector (34) to generate pcDNA3.1-FLAG-mTfR1. Shorthairpin RNAs were derived from mouse U6 promoter (mU6)on shRNA expressing retrovirus vector, pSSSP (35). Thisvector is self-inactivating and contains SV40 promoter-pu-romycin cassette. The mU6-driven shRNA cassette is intro-duced in the deleted U3 region of 3� long terminal repeat.The target sequence of short hairpin RNA used in this studyare listed below: shmTfR1, 5�-GAACCAGTTGTAAGAAC-AAT-G-3�, and shLacZ (control), 5�-GCAGTTATCTGGA-AGATCAGG-3�.

Antibodies—The following antibodies were used in thisstudy: rabbit anti-PV1/Mahoney hyperimmune serum was pre-pared by immunizing rabbits with purified PV1/Mahoney, anti-Flag (Wako Pure Chemical Industries Ltd., Osaka, Japan), anti-transferrin receptor (Zymed Laboratories Inc.. Inc., SanFrancisco, CA), and anti-�-actin (013-24553; Wako LTD.,Osaka, Japan). In Western blotting experiments, band imageswere analyzed using the LAS 4000 UV mini system (LifescienceTechnologies).

In Vitro Binding Experiments—GST fusion proteins wereexpressed in Escherichia coli Rosetta 2 (Novagen, Madison, WI)

via incubation of the cells with 0.1 mM isopropyl 1-thio-�-D-galactopyranoside overnight at 15 °C and then suspended inE. coli using His tag binding/wash buffer (20 mM Tris-HCl, pH8.0, 600 mM NaCl, 1 mM MgCl2, 10% glycerol, 0.1% NonidetP-40, 20 mM imidazole, and phosphatase inhibitors). E. coli wassonicated on ice using ELESTAIN035SD (ELECON Science.Corp, Chiba, Japan). Purification of GST-His-tagged proteinswas performed sequentially using Profinity IMAC nickel-charged resin (Bio-Rad) and glutathione-Sepharose 4B (GEHealthcare). The target proteins were eluted from these col-umns with His tag elution buffer (20 mM Tris-HCl, pH 8.0, 600mM NaCl, 1 mM MgCl2, 10% glycerol, 0.1% Nonidet P-40, 250mM imidazole, and protease inhibitors) and GST elution buffer(100 mM Tris-HCl, pH 8.0, 12 mM NaCl, 20 mM glutathione, andprotease inhibitors), respectively. [35S]Methionine-labeled full-length mTfR1 or its derivative constructs were translated usinga reticulocyte lysate system in vitro (Promega Corporation,Madison, WI), and incubated with PV particles or respectiveGST fusion proteins at 4 °C on a rotating platform for 2 h. Theprecipitated materials were washed for five times with TNEbuffer (10 mM Tris-HCl, pH 7.8, 150 mM NaCl, 1 mM EDTA, 1%Nonidet P-40, and protease inhibitors) and analyzed by SDS-PAGE followed by autoradiography.

Immunoprecipitation Assay—Immunoprecipitation assayswere performed as previously described (34). Briefly, cells werelysed with TNE buffer (10 mM Tris-HCl, pH 7.8, 150 mM NaCl,1 mM EDTA, 1% Nonidet P-40, and protease inhibitors), andcell lysates were incubated overnight on a rotating platform at4 °C with respective antibodies (5 �g each), which were previ-ously bound to Dynabeads� Protein G (Dynal, Oslo, Norway).After washing, precipitates were purified, subjected to SDS-PAGE, and analyzed by Western blotting or autoradiography.

PV Purification and Fluorescent Label of PV—PV was puri-fied by a protocol described previously (36). HeLa cells wereinfected with Mahoney virus at a multiplicity of infection of 10.The cells were harvested at 8 h postinfection, and the virus waspurified from cytoplasmic extracts of the infected cells by usingDEAE-Sepharose CL-6B, followed by sucrose density gradientand CsCl equilibrium centrifugation. Purified virus wasdesalted by gel filtration on a PD-10 column equilibrated withPBS(�). The PV concentration was determined by measuringthe absorbance at 260 nm; 1.0 optical density unit was regardedas equivalent to 9.4 � 1012 virions. Virus labeling was based ona protocol, which was described previously (13, 37). Briefly, PVwas labeled with Alexa Fluor-succinimidyl ester according tothe manufacturer’s instructions (Life Technologies). Thelabeled virus was purified on NAP5 columns (GE Healthcare),dialyzed against PBS(�) without loss of the specific infectivity.

Titration of Virus Infectivity—The numbers of plaque-form-ing unit in AGMK cells were determined by the plaque assay.For the measurement of plaque-forming unit, AGMK cells on6-cm dishes were inoculated with the viral suspension and thenincubated at 37 °C for 2–5 days for the observation of plaques.

Transwell in vitro blood brain barrier (BBB) permeationassay—Alexa 488-conjugated mouse transferrin was purchasedfrom Jackson ImmunoResearch Laboratories. FITC-labeleddextran (2,000 K) was purchased from Sigma-Aldrich. Mousebrain capillary endothelial cells (MBEC4) were seeded (5 � 104

Transferrin Receptor 1 Associates with VP1

2830 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 291 • NUMBER 6 • FEBRUARY 5, 2016

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

cells/well) into collagen-coated 24-well Transwell plates(#3496; Corning) and cultured for 48 h at 37 °C under 5% CO2(7). In vitro monolayer integrity was evaluated as trans-endo-thelial electrical resistance (TEER) using a Millicell� ERS vol-tohmmeter (Millipore, Bedford, MA). The average TEER valueof the cellular monolayer was 50 � 10 ��cm2 after backgroundsubtraction of the TEER value of a cell empty (blank) controlwell. Preparing 126.55 �g/ml purified PV particle, PV perme-ation was calculated as clearance volume in each time point asbelow: clearance volume (�l) � permeated apical side fluores-cent intensity (�g)/input PV fluorescent intensity volume(�g/�l).

FACS Analysis—MBEC4 cells were assessed by single-colorflow cytometric analysis. The following antibodies were usedfor the flow cytometric analysis: FITC anti-mouse CD71/TfR1antibody (113805) (BioLegend, San Diego, CA). Dead cells wereexcluded with the propidium iodide staining (Sigma-Aldrich).Samples were analyzed on iCyte flow cytometer (Sony Biotech-nology Inc., San Jose, CA).

Statistical Analyses—We performed t test. All statistical testswere two-sided. We considered p values less than 0.05 to bestatistically significant.

Results

PV Interacts with TfR1 on Mouse Brain Capillary EndothelialCells—To elucidate the interaction between PV particles andthe transferrin receptor, we generated mouse brain capillaryendothelial cell line (MBEC4) expressing FLAG-mTfR1.Because it is known that in PV-sensitive cells, the associationbetween PV capsid and hPVR promptly leads to PV capsid con-formational change followed by cell entry, we used PV-insensi-tive mouse cell lines to avoid this conformational change. We

conducted immunoprecipitation using anti-PV antibody ontotal lysate obtained from FLAG-mTfR1-MBEC4 cells exposedto PV for 1 h, followed by blotting with anti-FLAG antibody(Fig. 1A). FLAG-mTfR1 was clearly detected by Western blot-ting using an antibody against FLAG peptide. Next, we evalu-ated the interaction between PV and endogenous TfR1 onMBEC4 cells. The MBEC4 cells were then exposed to variousconcentrations of PV for 1 h, following which the supernatantwas discarded, and the whole cell extract was collected. Follow-ing immunoprecipitation using anti-PV antibody, endogenousmTfR1 was detected by Western blot using anti-mTfR1 anti-body in accordance with the PV dose (Fig. 1B). These resultssuggested that PV interacts with mTfR1 in vitro.

We speculated that if PV requires mTfR1 for cell permeation,PV might compete against transferrin during transport intocells. To examine this possibility, we assessed FITC-labeledtransferrin transport into MBEC4 cells in the presence of pre-treated PV or dextran. Because dextran is known to permeateinto arterial endothelial cells (39) through the intercellular gap(40) by virtue of its size (molecular mass, �15 kDa), dextran(molecular mass, 2 � 103 kDa) was used as a control for a dif-ferent pathway of permeation. It was presumably of a size sim-ilar to that of the PV particle, which is 8.6 � 103 kDa (41, 42).Co-treatment with purified PV effectively inhibited FITC-la-beled transferrin transport into MBEC4 cells (Fig. 1C). How-ever, an excess amount of dextran as compared with that of PVshowed no effect, even with increasing doses (Fig. 1D). Thisobservation confirmed the interaction between PV and mTfR1in vitro.

PV Interacts with mTfR1 through the Ectodomain—To iden-tify the domains of PV and mTfR1 that are responsible for their

PV 0 3,000 5,000 10,000 mTfR1

VP1VP2

MOI

α mTfR1

α PV

2.5 (%) Input

IP; α PV

IB

α mTfR1

IB:FLAG

Inpu

t (0.

1%)

IB: PV

FLAG−mTfR

←VP

←VP2

100-

75-37-

25-

PV

(- ) (+)

IP: PVA

B

Dextran 1mg / ml

PV

_

_ m.o.i. 5,000

C

D

667μg / ml

FIGURE 1. PV interacts with mTfR1 in vitro. A, immunoprecipitation assay for purified PV and Flag-mTfR1 interaction. MBEC4 cells were transfected withpcDNA3.1-Flag-mTfR1. After 48 h, PV was transduced in MBEC4 cells for 1 h, and the cell lysate was harvested for immunoprecipitation with anti-PV antibody.Immunoprecipitates were analyzed by Western blotting using anti-PV or FLAG antibodies. B, immunoprecipitation assay for purified PV and endogenous-mTfR1 interaction. PV was dose-dependently transduced in MBEC4 cell for 1 h at 37 °C, and the cell lysate was harvested for immunoprecipitation with theanti-PV antibody. Immunoprecipitates were analyzed by Western blotting using anti-PV or mTfR1 antibodies. C and D, PV inhibits transferrin transport inMBEC4 cells. Fluorescence images of transferrin penetration assay in MBEC4 cells are shown. Co-treatment with or without purified PV (C) and dextran (D) wasperformed as per the indicated ratio. FITC-labeled transferrin (10 �g/ml) was added, and the cells were incubated for 1 h, following which they were washedwith PBS and observed by fluorescence microscopy. Scale bars, 20 �m. IB, immunoblot; IP, immunoprecipitation; m.o.i., multiplicity of infection.

Transferrin Receptor 1 Associates with VP1

FEBRUARY 5, 2016 • VOLUME 291 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 2831

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

interaction, we identified the region of mTfR1 necessary for PVinteraction. Because mTfR1 contains three identical extracel-lular domains (Fig. 2A), we divided mTfR1 into four regions andgenerated in vitro translated 35S-labeled, FLAG-fused, and full-length mTfR1, as well as three derivative mTfR1 truncationsusing reticulocyte lysate (Fig. 2B). Subsequently, we tested theirinteraction with purified PV. The results showed that PV par-ticles directly interact with full-length mTfR1 (Fig. 2C). Eachtruncated mTfR1 (mTfR1– 609 and -388) also interacted withPV particles, except for the mTfR1–186 construct (Fig. 2C).According to the predicted structure of TfR1, the helicaldomain of TfR1 is folded into its homodimeric receptor (26).This hinders the natural interaction between PV and the helicaldomain. Together, these results indicated that the PV particlemay interact with the apical domain and/or secondary part ofprotease-like domain of mTfR1.

PV Efficiently Interacts with the Apical Domain of mTfR1—We further identified the mTfR1 domain that associates withthe PV particle. We constructed and generated in vitro trans-lated, 35S-labeled apical domain (AD) and second region of pro-tease-like domain (PD2) of mTfR1 (Fig. 2D). The result showedthat both mTfR1-AD and -PD2 are efficiently precipitated withPV particle (Fig. 2E), although the background signal wasslightly higher in the precipitation using mTfR1-PD2. Togetherwith the result shown in Fig. 2C, this result implied that theapical domain of mTfR1 is important for PV association. Giventhat the outer side of the PV capsid is composed of three poly-peptides (VP1, VP2, and VP3), these proteins are candidates forinteraction with mTfR1. To identify the PV capsid proteinresponsible for mTfR1 association, we constructed GST fusionVP1, VP2, and VP3 plasmids and generated the correspondingrecombinant proteins in E. coli. We then conducted GST pull-down assays using purified GST alone, GST-VP1, GST-VP2,and GST-VP3 and in vitro translated, 35S-labeled full-lengthmTfR1, as well as the three truncated proteins (Fig. 3A). VP1clearly bound to all mTfR1 constructs, except for the mTfR1–186 mutant (Fig. 3B). VP2 and VP3 showed weak or no bindingto any of the truncated mutants, although they bound to full-

length mTfR1. This result is possibly due to conformationalchanges by truncation. Together, these results suggested thatVP1 is the capsid protein with the highest affinity for mTfR1.

F-G, G-H, and H-I Loops on VP1 Are Responsible for Interac-tion with the Apical Domain of mTfR1—VP1 contains eightlarge �-sheet structures, and these antiparallel strands are com-pactly packed into the PV particle, similar to VP2 and VP3 (Fig.4A) (43). VP1 contains seven loop domains that are consideredas the outer exposed domains of the VP1 structure. To identifythe VP1 loop responsible for interaction with the AD or PD2domain of mTfR1, we constructed seven GST-Venus-fused

mTfR1

Protease-like domainApical domainHelical domain

Input( - +

PVA CB

388

609

Full

186

-CN-

TMD Ectodomain

1 124 763 (amino acid)

186 388 609

mTfR1 Full

mTfR1 609

mTfR1 388

mTfR1 186

-CN-

FLAG

mTfR1 AD

mTfR1 PD2

mTfR1 Full

mTfR1 609

mTfR1 388

mTfR1 186

75-

25-

48-

(KDa)

30-

20-

ADPD

2

(KDa)

Input( - +

PVD E

AD

PD2

FIGURE 2. PV interacts with the ectodomain of mTfR1 in vitro. A, a schematic representation of the mTfR1 protein and in vitro translated, FLAG-fused,full-length as well as truncated mTfR1 (mTfR1-full, -609, -388, and -186). Transmembrane domain (TMD) B, SDS gel electrophoresis of in vitro translatedFlag-fused full-length and truncated mTfR1 (mTfR1-full, -609, -388, and -186). C, immunoprecipitation assay for the interaction between purified PV and in vitrotranslated, FLAG-fused mTfR1, and its derivatives. D, SDS gel electrophoresis of in vitro translated Flag-fused apical and protease-like domain of mTfR1 (AD andPD2). E, immunoprecipitation assay for the purified poliovirus and in vitro translated 35S-labeled Flag-fused AD and PD2 of mTfR1 protein interaction.

Input GST VP1 VP2 VP3

GST-

75-GST VP1 VP2 VP3

GST-

63-

48-

35-

25-

M

mTfR1 Full

mTfR1 609

mTfR1 388

mTfR1 186

(KDa)

A

B

FIGURE 3. PV interacts with the apical domain of mTfR1 via the capsidprotein VP1. A and B, recombinant GST-VP1, VP2, and VP3 proteins werepurified from E. coli lysates and GST pulldown assay with in vitro translated ADand PD2 of mTfR1. GST pulldown materials were stained by Coomassie Blue.An asterisk indicates the nonspecific precipitated bands derived from reticu-locyte lysates (A). The precipitate of the GST pulldown assay was exposed toautoradiography after SDS-gel electrophoresis (B).

Transferrin Receptor 1 Associates with VP1

2832 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 291 • NUMBER 6 • FEBRUARY 5, 2016

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

proteins, each containing VP1 loops under Venus protein (Fig.4B and Table 1). We performed GST pulldown assays usingthese purified GST-Venus fusion proteins and in vitro trans-lated AD or PD2 of mTfR1 (Fig. 4C). F-G, G-H, and H-I loopsshowed GST-Venus-fused protein clearly bound to the AD ofmTfR1. GST-Venus-fused protein containing F-G loops wasthe only protein that interacted with PD2 of mTfR1 (Fig. 4C).These results showed that F-G, G-H, and H-I loops of VP1 arethe domains mainly responsible for the interaction between PVand mTfR1.

PV Permeates Mouse Brain Capillary Endothelial Cells andCompetes against Transferrin for Transport into Cells—Wedetermined whether PV has the ability to permeate brain cap-illary endothelial cells in vitro. We performed cell permeationassay and quantified the cell-permeating ratio into MBEC4 cellsusing purified GST-Venus-VP1 and the 7 GST-Venus-VP1-loop proteins (Fig. 4B). The Venus fusion proteins were incu-bated with MBEC4 cells for 24 h, followed by preparation of celllysate and measurement of fluorescence intensity of the perme-ated protein. As shown in Fig. 4D, full-length VP1 and B-Cloops of VP1 connected with GST-Venus proteins exhibitedmarginal permeation into MBEC4 cells compared with controlfusion protein. C-D, D-E, and E-F loops of VP1 connected withGST-Venus protein showed �2-fold permeable efficiency intoMBEC4 cells, whereas F-G, G-H, and H-I loops of VP1, respon-sible for mTfR1 attachment, showed poor permeation intoMBEC4 cells. These results suggest that the VP1 loops respon-sible for attachment to mTfR1 and permeation into MBEC4cells are indeed different.

Knockdown of Transferrin Receptor Prevents PV Transmis-sion in the in Vitro BBB Model—Our group previously observedthat circulating PV promptly invaded the brain from the bloodvia the BBB in a non-hPVR-Tg mouse model (16). To confirm

whether PV uses the transferrin receptor to invade the CNS, weconducted Transwell BBB in vitro model assay using MBEC4cells (38), wherein MBEC4 cells were confluently seeded on amicroporous membrane in the upper chamber of the Tran-swell. MBEC4 cells were exposed to fluorescein-labeled PVwith or without transferrin, and then fluorescence intensity ofthe medium collected from the lower chamber was assessed tomonitor PV that had permeated the MBEC4 cells (Fig. 5A).Because dextran is known to permeate into arterial endothelialcells (39) through the intercellular gap (40) by virtue of its size(molecular mass, �15 kDa), we used dextran as the permeationcontrol. To validate the Transwell BBB in vitro model assaysystem, we first compared the permeation ratio of FITC-labeledtransferrin to that of FITC-labeled dextran (70 kDa) as a similarsize control. The permeation ratio of transferrin was 2.5-foldhigher than that of dextran (Fig. 5B). Given that transferrinpermeates into cells via its association with the transferrinreceptor in the endocytic pathway, whereas dextran passivelypermeates through the intercellular gap, this result suggestedthat the assay system is functional. In a time course experiment,PV was shown to efficiently permeate through MBEC4 cellsinto the lower chamber (Fig. 5C). However, this PV transportwas delayed by the addition of transferrin (Fig. 5C). Because

FIGURE 4. Identification of VP1 loops responsible for interaction with mTfR1. A, a schematic representation of the capsid protein VP1. Arrows and curvedlines represent �-strands and loops of VP1, respectively. B, Coomassie Blue-stained SDS gel electrophoresis of GST-Venus-VP1 derivative constructs. C, GSTpulldown assay with GST-Venus-VP1 derivatives and in vitro translated AD and PD2 domains of mTfR1. D, quantitative analysis of selective permeability of eachGST-Venus-VP1 loop in MBEC4 cells. Relative fluorescence intensity of individual proteins against GST-Venus-empty protein was calculated in each imageobtained by fluorescence microscopy. Vec, vector.

TABLE 1Summary of the peptide sequence and length of VP1 loops

Loop Peptide sequence Length

amino acidsBC PASTTNKDKLF 11CD ITYKDTVQLRRKLEF 15DE TETNNGHALNQ 11EF PPGAPVPEKWDDYTWQTSSNP 21FG YGTAP 5GH HFYDGFSKVPLKDQSAALGDSLYGAASLNDFG 32HI VNDHNPTK 8

Transferrin Receptor 1 Associates with VP1

FEBRUARY 5, 2016 • VOLUME 291 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 2833

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

transferrin is known to interact with the protease-like domain(PD2) of TfR1 (44, 45), PV capsids and transferrin would com-pete for this TfR1 domain, which is in agreement with the resultshown in Fig. 2C. To validate whether unmodified PV particlesalso require mTfR1 to permeate through MBEC4 cells, we con-structed retroviral vector expressing shRNA against mTfR1(sh-mTfR1). Stable transductants were obtained using appro-priate antibiotics (Fig. 5D). We confirmed knockdown ofmTfR1 expression on these cells by flow cytometric analysis(Fig. 5E). Using mTfR1 or control knockdown MBEC4 cells, weperformed the Transwell BBB in vitro model assay. The numberof permeated PVs was estimated by colony forming assay inAGMK cells. From the results, PV permeation was delayed inmTfR1 knockdown MBEC4 cells as compared with controlMBEC4 cells (Fig. 5F). These results confirmed the positive roleof mTfR1 in PV permeation through brain capillary cells. Addi-tionally, the results showed that PV retains its infectivity aftertransmission through MBEC4 cells.

Discussion

Given that PV permeates the brain as fast as cationized ratalbumin with a high permeation rate in non-hPVR-Tg mice(16), it is believed that PV can directly invade the CNS via BBBtransmission in an hPVR-independent manner. In this study,we suggest a novel pathway where PV invades the CNS in anhPVR-independent manner and that mTfR1 is a key receptoron brain capillary endothelial cells, responsible for PV perme-ation of the CNS via the BBB. Transferrin receptor is known to

be a crucial molecule for cellular homeostasis during oxygen-ation (26). Because the CNS also requires active oxygenationand the antibody against the transferrin receptor stains braincapillary cells in the rat brain after intravenous administration(46, 47), transferrin receptor is a plausible candidate for thereceptor of PV permeation of the brain.

PV clearly interacted with mTfR1 proteins in vitro (Figs. 1and 2) in this study, and we found that VP1 is responsible forthis binding (Fig. 3B). VP1 is also known to play a role in theinteraction with hPVR/CD155. hPVR/CD155 inserts into thedeep pocket “canyon” on the surface of the PV particle, which iscomposed of capsid proteins (43). After the associationbetween the canyon and hPVR/CD155, the uncoating processpromptly occurs in the PV particle. For PV proliferation in thebrain after BBB transport, PV needs to retain its infectivity aftertranscytosis. The in vitro BBB model assay showed that perme-ated materials contained intact infectious particles (Fig. 5F).This result implies that PV permeates through brain capillarycells without loss of infectivity; a similar observation has beenpreviously noted for the brain of hPVR-Tg and non-Tg mice(16).

Our in vitro binding experiment showed that F-G, G-H, andH-I loops are the VP1 regions responsible for mTfR1 interac-tion (Fig. 4C). The three-dimensional prediction analysis for PVparticles showed that the G-H loop of VP1 faces the outer sideand is located near the canyon, although the F-G and H-I loopsof VP1 face the inside of the PV capsid. Considering this struc-

FIGURE 5. The permeation rate of PV in BBB in vitro assay is delayed by co-incubation with transferrin or mTfR1 knock down. A, a schematic represen-tation of in vitro BBB permeation assay using Transwells. MBEC4 cells are confluently seeded, and fluorescent PV was transduced into the upper chamber of theTranswell. Fluorescence intensity was measured by collecting medium from the lower chamber of the Transwell at indicated time points (min). B, validation ofthe in vitro BBB permeation assay. Quantitative analysis of fluorescently labeled transferrin (80 kDa) and dextran (70 kDa) permeating MBEC4 cells. Theclearance volume of each fluorescently labeled protein was calculated from the ratio of fluorescence intensity to input fluorescence. C, in vitro BBB permeationassay. Quantitative analysis of fluorescently labeled PV permeating MBEC4 cells. The clearance volume was calculated from the ratio of fluorescence intensityto input fluorescently labeled PV particles. D, expression of mTfR1 and �-actin proteins in MBEC4 cells stably transduced with retroviral vector expressingsh-mTfR1 or control LacZ shRNA (Con.). E, flow cytometric analysis of mTfR1 expression in sh-mTfR1 or control retrovirus vector-transduced MBEC4 cells. MouseTfR1 was stained with FITC-labeled anti-mTfR1. F, in vitro BBB permeation assay with knockdown mTfR1 and control MBEC4 cells. Quantitative analysis of PVpermeating through MBEC4 derivative cells at each indicated time point. The number of permeated PVs was estimated by colony forming assay using AGMKcells. **, p 0.01; *, p 0.05.

Transferrin Receptor 1 Associates with VP1

2834 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 291 • NUMBER 6 • FEBRUARY 5, 2016

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

tural analysis, G-H loop of VP1 is expected to be a probablecandidate for mTfR1 binding. Further analysis by inserting amutation at this loop on VP1 in PV particle or using the specificantibody against the identified loop of VP1 is an effectiveapproach for understanding this binding mechanism. Homol-ogy between the sequence of TfR1 in mice and humans is 77%,and both proteins play a role in iron delivery. Considering this,it is possible that PV also interacts with human TfR1 as a medi-ator for BBB permeation.

The in vitro BBB model using MBEC4 cells showed that invitro translated GST-Venus proteins fused with C-D, D-E, andE-F loops of VP1 permeate to a comparatively greater extentthan other loops (Fig. 4D). All of these loops face the outer sideof the PV particle, as revealed by the three-dimensional predic-tion analysis. This result shows that the VP1 loops responsiblefor mTfR1 binding and for permeation into MBEC4 cells areclearly separated. However, co-incubation with transferrin orknocking down of mTfR1 inhibited the transcytosis of PVthrough MBEC4 cells (Fig. 5); therefore, we speculated thatmTfR1 plays a role in attachment to the cell membrane, fol-lowed by PV permeation of the brain capillary endothelial cells.This also means that additional unidentified molecules wouldmediate this transcytosis. The expression of transferrin recep-tor is known to be ubiquitous, although the expression level iscomparatively higher in brain capillary endothelial cells. If thisPV permeation is a specific event in the BBB, the existence ofadditional molecules that are expressed only in brain capillaryendothelial cells is plausible. Therefore, further studies arerequired to identify these molecules to better understand theBBB permeation mechanism.

It has been shown that in the case of hPVR-dependent endo-cytosis, the route for PV transmission into the brain is viaperipheral MNs from the skeletal muscle (14). However, PV canslowly transmit via MNs in non-hPVR Tg mice in vivo as well,although PV-containing endocytic vesicles were not observedin MNs cultured in vitro (15). These observations indicate thatPV also potentially moves to the CNS by an unknown, hPVR-independent endocytic pathway (16). This discrepancybetween in vitro and in vivo results may be explained by thedecreased TfR1 expression in the extending neurites of MNs inculture (48). It cannot be excluded that PV uses transferrinreceptor as a positive mediator for transportation to the CNSfrom the skeletal muscles.

In this study, we showed that PV permeates mouse braincapillary endothelial cells through the BBB using the Transwellin vitro BBB model. Our results showed that mTfR1 is a positivemediator for BBB permeation of PV. To elucidate the completemechanism for BBB permeation of PV, identification of thecellular counterpart for C-D, D-E, and E-F loops of VP1 isrequired. This study contributes to understanding the mecha-nism of viral transmission into the CNS, indicating its impor-tance in the biology and pathogenesis of PV. Further, there is agrowing recognition of the importance to develop CNS drugdelivery systems. To this end, a BBB-permeable agent and/or abetter understanding of delivering drug agents to the CNS ishighly desired. Knowledge of the BBB permeation mechanismof PV will contribute to overcoming the challenge posed by thecurrent inability to transport drugs to the CNS via the BBB.

Author Contributions—T. M. planned the experiments and wrotethe manuscript. T. M. and A. I. performed the experiments and ana-lyzed the data. C. N. supplied plasmid materials and discussed theexperiments. All authors approved the final manuscript.

Acknowledgments—We were greatly saddened by the passing of Pro-fessor Akio Nomoto during the course of this work. He contributed toand deeply discussed all aspects of the data in this work. We thank Dr.S. Ohka and M. Sakai for analyzing BBB transport of PV in vitro. Wethank Editage for providing editorial assistance.

References1. Bordin, E. S. (1955) Ambiguity as a therapeutic variable. J. Consult. Psy-

chol. 19, 9 –152. Sabin, A. B. (1956) Pathogenesis of poliomyelitis; reappraisal in the light of

new data. Science 123, 1151–11573. Gromeier, M., and Wimmer, E. (1998) Mechanism of injury-provoked

poliomyelitis. J. Virol. 72, 5056 –50604. Muñoz, A., and Carrasco, L. (1984) Action of human lymphoblastoid in-

terferon on HeLa cells infected with RNA-containing animal viruses.J. Gen. Virol. 65, 377–390

5. Aoki, J., Koike, S., Ise, I., Sato-Yoshida, Y., and Nomoto, A. (1994) Aminoacid residues on human poliovirus receptor involved in interaction withpoliovirus. J. Biol. Chem. 269, 8431– 8438

6. Ida-Hosonuma, M., Iwasaki, T., Yoshikawa, T., Nagata, N., Sato, Y., Sata,T., Yoneyama, M., Fujita, T., Taya, C., Yonekawa, H., and Koike, S. (2005)The �/� interferon response controls tissue tropism and pathogenicity ofpoliovirus. J. Virol. 79, 4460 – 4469

7. Koike, S., Taya, C., Kurata, T., Abe, S., Ise, I., Yonekawa, H., and Nomoto,A. (1991) Transgenic mice susceptible to poliovirus. Proc. Natl. Acad. Sci.U.S.A. 88, 951–955

8. Horie, H., Koike, S., Kurata, T., Sato-Yoshida, Y., Ise, I., Ota, Y., Abe, S.,Hioki, K., Kato, H., and Taya, C. (1994) Transgenic mice carrying thehuman poliovirus receptor: new animal models for study of poliovirusneurovirulence. J. Virol. 68, 681– 688

9. Koike, S., Taya, C., Aoki, J., Matsuda, Y., Ise, I., Takeda, H., Matsuzaki, T.,Amanuma, H., Yonekawa, H., and Nomoto, A. (1994) Characterization ofthree different transgenic mouse lines that carry human poliovirus recep-tor gene–influence of the transgene expression on pathogenesis. Arch.Virol. 139, 351–363

10. Ren, R., and Racaniello, V. R. (1992) Human poliovirus receptor geneexpression and poliovirus tissue tropism in transgenic mice. J. Virol. 66,296 –304

11. Ren, R. B., Costantini, F., Gorgacz, E. J., Lee, J. J., and Racaniello, V. R.(1990) Transgenic mice expressing a human poliovirus receptor: a newmodel for poliomyelitis. Cell 63, 353–362

12. Zhang, S., and Racaniello, V. R. (1997) Expression of the poliovirus recep-tor in intestinal epithelial cells is not sufficient to permit poliovirus repli-cation in the mouse gut. J. Virol. 71, 4915– 4920

13. Ohka, S., Igarashi, H., Nagata, N., Sakai, M., Koike, S., Nochi, T., Kiyono,H., and Nomoto, A. (2007) Establishment of a poliovirus oral infectionsystem in human poliovirus receptor-expressing transgenic mice that aredeficient in �/� interferon receptor. J. Virol. 81, 7902–7912

14. Ohka, S., Yang, W. X., Terada, E., Iwasaki, K., and Nomoto, A. (1998)Retrograde transport of intact poliovirus through the axon via the fasttransport system. Virology 250, 67–75

15. Ohka, S., Sakai, M., Bohnert, S., Igarashi, H., Deinhardt, K., Schiavo, G.,and Nomoto, A. (2009) Receptor-dependent and -independent axonalretrograde transport of poliovirus in motor neurons. J. Virol. 83,4995–5004

16. Yang, W. X., Terasaki, T., Shiroki, K., Ohka, S., Aoki, J., Tanabe, S., No-mura, T., Terada, E., Sugiyama, Y., and Nomoto, A. (1997) Efficient deliv-ery of circulating poliovirus to the central nervous system independentlyof poliovirus receptor. Virology 229, 421– 428

17. Ballabh, P., Braun, A., and Nedergaard, M. (2004) The blood-brain barrier:

Transferrin Receptor 1 Associates with VP1

FEBRUARY 5, 2016 • VOLUME 291 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 2835

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

an overview: structure, regulation, and clinical implications. Neurobiol.Dis. 16, 1–13

18. Brightman, M. W. (1977) Morphology of blood-brain interfaces. Exp. EyeRes. 25, 1–25

19. Reese, T. S., and Karnovsky, M. J. (1967) Fine structural localization of ablood-brain barrier to exogenous peroxidase. J. Cell Biol. 34, 207–217

20. Shah, K., Desilva, S., and Abbruscato, T. (2012) The role of glucose trans-porters in brain disease: diabetes and Alzheimer’s Disease. Int. J. Mol. Sci.13, 12629 –12655

21. Pardridge, W. M., Eisenberg, J., and Yang, J. (1987) Human blood-brainbarrier transferrin receptor. Metabolism 36, 892– 895

22. Fishman, J. B., Rubin, J. B., Handrahan, J. V., Connor, J. R., and Fine, R. E.(1987) Receptor-mediated transcytosis of transferrin across the blood-brain barrier. J. Neurosci. Res. 18, 299 –304

23. Pardridge, W. M. (1986) Receptor-mediated peptide transport throughthe blood-brain barrier. Endocr. Rev. 7, 314 –330

24. Pardridge, W. M., Yang, J., and Eisenberg, J. (1985) Blood-brain barrierprotein phosphorylation and dephosphorylation. J. Neurochem. 45,1141–1147

25. Pardridge, W. M. (1983) Brain metabolism: a perspective from the blood-brain barrier. Physiol. Rev. 63, 1481–1535

26. Wang, J., and Pantopoulos, K. (2011) Regulation of cellular iron metabo-lism. Biochem. J. 434, 365–381

27. Ross, S. R., Schofield, J. J., Farr, C. J., and Bucan, M. (2002) Mouse trans-ferrin receptor 1 is the cell entry receptor for mouse mammary tumorvirus. Proc. Natl. Acad. Sci. U.S.A. 99, 12386 –12390

28. Flanagan, M. L., Oldenburg, J., Reignier, T., Holt, N., Hamilton, G. A.,Martin, V. K., and Cannon, P. M. (2008) New world clade B arenavirusescan use transferrin receptor 1 (TfR1)-dependent and -independent entrypathways, and glycoproteins from human pathogenic strains are associ-ated with the use of TfR1. J. Virol. 82, 938 –948

29. Martin, D. N., and Uprichard, S. L. (2013) Identification of transferrinreceptor 1 as a hepatitis C virus entry factor. Proc. Natl. Acad. Sci. U.S.A.110, 10777–10782

30. Palermo, L. M., Hueffer, K., and Parrish, C. R. (2003) Residues in the apicaldomain of the feline and canine transferrin receptors control host-specificbinding and cell infection of canine and feline parvoviruses. J. Virol. 77,8915– 8923

31. Radoshitzky, S. R., Abraham, J., Spiropoulou, C. F., Kuhn, J. H., Nguyen, D.,Li, W., Nagel, J., Schmidt, P. J., Nunberg, J. H., Andrews, N. C., Farzan, M.,and Choe, H. (2007) Transferrin receptor 1 is a cellular receptor for NewWorld haemorrhagic fever arenaviruses. Nature 446, 92–96

32. Tatsuta, T., Naito, M., Oh-hara, T., Sugawara, I., and Tsuruo, T. (1992)Functional involvement of P-glycoprotein in blood-brain barrier. J. Biol.Chem. 267, 20383–20391

33. Mizutani, T., Ishizaka, A., and Furuichi, Y. (2015) The Werner protein actsas a coactivator of nuclear factor �B (NF-�B) on HIV-1 and interleukin-8(IL-8) Promoters. J. Biol. Chem. 290, 18391–18399

34. Ishizaka, A., Mizutani, T., Kobayashi, K., Tando, T., Sakurai, K., Fujiwara,T., and Iba, H. (2012) Double plant homeodomain (PHD) finger proteinsDPF3a and -3b are required as transcriptional co-activators in SWI/SNFcomplex-dependent activation of NF-�B RelA/p50 heterodimer. J. Biol.Chem. 287, 11924 –11933

35. Mizutani, T., Ishizaka, A., Suzuki, Y., and Iba, H. (2014) 7SK small nuclearribonucleoprotein complex is recruited to the HIV-1 promoter via shortviral transcripts. FEBS Lett. 588, 1630 –1636

36. Kajigaya, S., Arakawa, H., Kuge, S., Koi, T., Imura, N., and Nomoto, A.(1985) Isolation and characterization of defective-interfering particles ofpoliovirus Sabin 1 strain. Virology 142, 307–316

37. Pelkmans, L., Kartenbeck, J., and Helenius, A. (2001) Caveolar endocytosisof simian virus 40 reveals a new two-step vesicular-transport pathway tothe ER. Nat. Cell Biol. 3, 473– 483

38. Dohgu, S., Kataoka, Y., Ikesue, H., Naito, M., Tsuruo, T., Oishi, R., andSawada, Y. (2000) Involvement of glial cells in cyclosporine-increased per-meability of brain endothelial cells. Cell. Mol. Neurobiol. 20, 781–786

39. Hashida, R., Anamizu, C., Yagyu-Mizuno, Y., Ohkuma, S., and Takano, T.(1986) Transcellular transport of fluorescein dextran through an arterialendothelial cell monolayer. Cell Struct. Funct. 11, 343–349

40. Choi, J. J., Wang, S., Tung, Y. S., Morrison, B., 3rd, and Konofagou, E. E.(2010) Molecules of various pharmacologically-relevant sizes can crossthe ultrasound-induced blood-brain barrier opening in vivo. UltrasoundMed. Biol. 36, 58 – 67

41. Putnak, J. R., and Phillips, B. A. (1981) Picornaviral structure and assembly.Microbiol. Rev. 45, 287–315

42. Scraba, D. G., Kay, C. M., and Colter, J. S. (1967) Physico-chemical studiesof three variants of Mengo virus and their constituent ribonucleates. J.Mol. Biol. 26, 67–79

43. Levy, H. C., Bostina, M., Filman, D. J., and Hogle, J. M. (2010) Catching avirus in the act of RNA release: a novel poliovirus uncoating intermediatecharacterized by cryo-electron microscopy. J. Virol. 84, 4426 – 4441

44. Cheng, Y., Zak, O., Aisen, P., Harrison, S. C., and Walz, T. (2004) Structureof the human transferrin receptor-transferrin complex. Cell 116, 565–576

45. Eckenroth, B. E., Steere, A. N., Chasteen, N. D., Everse, S. J., and Mason,A. B. (2011) How the binding of human transferrin primes the transferrinreceptor potentiating iron release at endosomal pH. Proc. Natl. Acad. Sci.U.S.A. 108, 13089 –13094

46. Friden, P. M., Walus, L. R., Musso, G. F., Taylor, M. A., Malfroy, B., andStarzyk, R. M. (1991) Anti-transferrin receptor antibody and antibody-drug conjugates cross the blood-brain barrier. Proc. Natl. Acad. Sci. U.S.A.88, 4771– 4775

47. Jefferies, W. A., Brandon, M. R., Hunt, S. V., Williams, A. F., Gatter, K. C.,and Mason, D. Y. (1984) Transferrin receptor on endothelium of braincapillaries. Nature 312, 162–163

48. Nakamura, Y., Nakamichi, N., Takarada, T., Ogita, K., and Yoneda, Y.(2012) Transferrin receptor-1 suppresses neurite outgrowth in neuroblas-toma Neuro2A cells. Neurochem. Int. 60, 448 – 457

Transferrin Receptor 1 Associates with VP1

2836 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 291 • NUMBER 6 • FEBRUARY 5, 2016

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: Transferrin Receptor 1 Facilitates Poliovirus Permeation ... · between PV and mTfR1. Co-incubation with transferrin or knockdown of mTfR1 resulted in delayed PV transcytosis via

Taketoshi Mizutani, Aya Ishizaka and Coh-ichi NiheiCapillary Endothelial Cells

Transferrin Receptor 1 Facilitates Poliovirus Permeation of Mouse Brain

doi: 10.1074/jbc.M115.690941 originally published online December 4, 20152016, 291:2829-2836.J. Biol. Chem. 

  10.1074/jbc.M115.690941Access 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/291/6/2829.full.html#ref-list-1

This article cites 48 references, 23 of which can be accessed free at

by guest on January 2, 2020http://w

ww

.jbc.org/D

ownloaded from