You are on page 1of 14

crossmark

Mechanism of Cell Culture Adaptation of an Enteric Calicivirus, the


Porcine Sapovirus Cowden Strain
Zhongyan Lu,a Masaru Yokoyama,b Ning Chen,a,c Tomoichiro Oka,a,d Kwonil Jung,a Kyeong-Ok Chang,e Thavamathi Annamalai,a
Qiuhong Wang,a Linda J. Saifa
Food Animal Health Research Program, Ohio Agricultural Research and Development Center, College of Food, Agricultural, & Environmental Sciences, Department of
Veterinary Preventive Medicine, The Ohio State University, Wooster, Ohio, USAa; Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japanb;
Boehringer Ingelheim (China) Investment Co., Ltd., Shanghai, Chinac; Department of Virology II, National Institute of Infectious Disease, Tokyo, Japand; Department of
Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USAe

ABSTRACT

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


The porcine sapovirus (SaV) (PoSaV) Cowden strain is one of only a few culturable enteric caliciviruses. Compared to the wild-
type (WT) PoSaV Cowden strain, tissue culture-adapted (TC) PoSaV has two conserved amino acid substitutions in the RNA-
dependent RNA polymerase (RdRp) and six in the capsid protein (VP1). By using the reverse-genetics system, we identified that
4 amino acid substitutions in VP1 (residues 178, 289, 324, and 328), but not the substitutions in the RdRp region, were critical
for the cell culture adaptation of the PoSaV Cowden strain. The other two substitutions in VP1 (residues 291 and 295) reduced
virus replication in vitro. Three-dimensional (3D) structural analysis of VP1 showed that residue 178 was located near the dim-
er-dimer interface, which may affect VP1 assembly and oligomerization; residues 289, 291, 324, and 328 were located at protrud-
ing subdomain 2 (P2) of VP1, which may influence virus binding to cellular receptors; and residue 295 was located at the inter-
face of two monomeric VP1 proteins, which may influence VP1 dimerization. Although reversion of the mutation at residue 291
or 295 from that of the TC strain to that of the WT reduced virus replication in vitro, it enhanced virus replication in vivo, and
the revertants induced higher-level serum and mucosal antibody responses than those induced by the TC PoSaV Cowden strain.
Our findings reveal the molecular basis for PoSaV adaptation to cell culture. These findings may provide new, critical informa-
tion for the cell culture adaptation of other PoSaV strains and human SaVs or noroviruses.

IMPORTANCE
The tissue culture-adapted porcine sapovirus Cowden strain is one of only a few culturable enteric caliciviruses. We discovered
that 4 amino acid substitutions in VP1 (residues 178, 289, 324, and 328) were critical for its adaptation to LLC-PK cells. Two sub-
stitutions in VP1 (residues 291 and 295) reduced virus replication in vitro but enhanced virus replication and induced higher-
level serum and mucosal antibody responses in gnotobiotic pigs than those induced by the tissue culture-adapted strain. Struc-
tural modeling analysis of VP1 suggested that residue 178 may affect VP1 assembly and oligomerization; residues 289, 291, 324,
and 328 may influence virus binding to cellular receptors; and residue 295 may influence VP1 dimerization. Our findings will
provide new information for the cell culture adaptation of other sapoviruses and possibly noroviruses.

C aliciviruses, in the family Caliciviridae, are small, icosahedral,


and nonenveloped viruses with a diameter of 27 to 35 nm,
which have a positive-sense, single-stranded RNA genome of 6.5
cells in the presence of an intestinal content preparation from
uninfected Gn pigs (12, 14).
Gn pigs have been established as a relevant animal model be-
to 8.3 kb (1, 2). Caliciviruses have been classified into five genera cause of the similarity of anatomy, genetics, physiology, and im-
(Norovirus, Sapovirus, Vesivirus, Lagovirus, and Nebovirus) and munity with humans (15–17). PoSaV naturally infects pigs and
several proposed genera (3, 4). Among them, noroviruses (NoVs) causes mild-to-moderate gastroenteritis in Gn pigs (14, 18, 19),
and sapoviruses (SaVs) are the leading causes of gastroenteritis in thus mimicking SaV diarrhea in humans and providing an animal
humans of all ages. SaVs are often associated with sporadic, self- model suitable for studies of the replication and pathogenesis of
limiting gastroenteritis, the severity of which is reportedly milder enteric caliciviruses.
than that of NoVs (5, 6). However, SaVs also cause outbreaks
worldwide (7–10), and deaths associated with SaV infection have
been reported for long-term-care facilities (11). Received 1 September 2015 Accepted 8 November 2015
Because most enteric caliciviruses are unculturable, re- Accepted manuscript posted online 18 November 2015
search on pathogenesis and immunity, as well as the develop- Citation Lu Z, Yokoyama M, Chen N, Oka T, Jung K, Chang K-O, Annamalai T,
ment of antivirals, has been hampered. The porcine SaV Wang Q, Saif LJ. 2016. Mechanism of cell culture adaptation of an enteric
calicivirus, the porcine sapovirus Cowden strain. J Virol 90:1345–1358.
(PoSaV) Cowden strain, previously known as porcine enteric doi:10.1128/JVI.02197-15.
calicivirus (PEC), belongs to genogroup III (GIII) of the SaVs Editor: S. Schultz-Cherry
and is one of only a few culturable enteric caliciviruses (2, 12, Address correspondence to Qiuhong Wang, wang.655@osu.edu, or Linda J. Saif,
13). The PoSaV Cowden strain was adapted to a porcine kidney saif.2@osu.edu.
cell line (LLC-PK) after passage of the virus in gnotobiotic Copyright © 2016, American Society for Microbiology. All Rights Reserved.
(Gn) pigs, followed by 20 passages in primary porcine kidney

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1345


Lu et al.

The genome of PoSaV is composed of two open reading genomic organization and mapping of the mutations are illustrated (Fig.
frames (ORFs). ORF1 encodes a polyprotein that is processed 1). TC-WTVP1 was generated by replacing a partial VP1 fragment (nucle-
into several nonstructural proteins (NSs) and the major struc- otide [nt] positions 5227 to 6060 and amino acid positions 30 to 308) of
tural protein VP1 by a viral protease. ORF2 encodes a small pCV4A with the corresponding sequence fragment of the WT PoSaV
structural protein, VP2 (20). VP1 is divided into two domains: Cowden strain. Briefly, the WT PoSaV Cowden VP1 fragment containing
two ApaI restriction enzyme sites (nucleotide positions 5227 to 5232 and
a shell (S) domain (amino acid positions 3 to 216) and a pro-
6055 to 6060) was reverse transcribed by using SuperScript III reverse
truding (P) domain (amino acid positions 217 to 544) (21).
transcriptase (Life Technologies, NY, USA) and amplified by PCR with
The P domain is further divided into the P1 (amino acid posi- primers ApaI-F and ApaI-R using PrimeStar HS high-fidelity DNA poly-
tions 217 to 272 and 425 to 544) and P2 (amino acid positions merase (Clontech Laboratories Inc., CA, USA). The amplicons were di-
273 to 424) subdomains (21, 22). gested by the ApaI restriction enzyme and cloned into the ApaI-digested
Reverse-genetics systems are important tools to rescue uncul- pCV4A plasmid.
turable viruses and to study virus replication mechanisms. A re- Full-length cDNA clones of TC-WTRdRp, VP1 of the TC strain with
verse-genetics system, pCV4A, that was constructed for the PoSaV an S-to-C mutation at position 178 (TCVP1-S178C), TCVP1-H289Y,
Cowden strain contained the full-length genomic cDNA of the TCVP1-D291N, TCVP1-R295K, TCVP1-I324M¡M324I, and TCVP1-

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


tissue culture-adapted (TC) PoSaV Cowden strain, directly down- G328E¡E328G were generated based on the pCV4A backbone by using
stream from the T7 RNA polymerase promoter (20). Infectious a QuikChange II XL site-directed mutagenesis kit (Agilent Technologies,
TC PoSaV particles were rescued after transfection of LLC-PK TX, USA) according to the manufacturer’s instructions (Fig. 1). For ex-
ample, TC-WTRdRp was generated when the two RNA-dependent RNA
cells with the in vitro-transcribed and -capped PoSaV genomic
polymerase (RdRp) amino acid residues at positions 1252 and 1379 of
RNA (20).
pCV4A were mutated from the TC to the WT residues (H1252Y and
In this study, we investigated the genetic basis of cell culture K1379R). Three full-length cDNA clones of chimeric genomes (TC-
adaptation of the PoSaV Cowden strain by comparative sequence WTVP1-C178S, TC-WTVP1-Y289H, and TC-WTVP1-C178S&Y289H)
analyses of the genomes of different passages of wild-type (WT) were generated based on the TC-WTVP1 backbone, whose residues at
and TC PoSaVs and the generation of a series of PoSaV mutants amino acid positions 324 and 328 in VP1 were of the TC type. Full-length
using the reverse-genetics system. We further investigated their cDNA clones of TCVP1-I324M and TCVP1-G328E were generated by di-
differences in replication both in vivo and in vitro as well as their gestion of pCV4A with the EcoRI restriction enzyme (nucleotide posi-
putative structural differences. To our knowledge, our studies are tions 4582 to 6877) and replacement with I324M or G328E engineered
the first to identify which amino acid residues are critical for the PCR products. Using TCVP1-I324M as an example, two fragments were
cell culture adaptation of a SaV. This study provides new informa- PCR amplified with primers EcoRI-F and 6111-R and with primers
tion on cell culture adaptation of SaVs that may be applicable to 6111-F and EcoRI-R, respectively, using pCV4A as the template. The PCR
product containing the I324M mutation was assembled by overlap PCR
other human SaVs or to NoVs.
with primers EcoRI-F and EcoRI-R, using the two fragments as the tem-
MATERIALS AND METHODS plates. The overlap PCR products containing the I324M mutation were
Cells and viruses. The LLC-PK cell line (ATCC CL-101) and a human digested with the EcoRI restriction enzyme and inserted into EcoRI re-
embryonic kidney cell line, HEK 293T/17 (ATCC CRL-11268), were ob- striction enzyme-digested pCV4A. The recombinant plasmid was trans-
tained from the American Type Culture Collection (ATCC). LLC-PK cells formed and amplified in competent Escherichia coli 10-beta cells (New
were passaged and maintained as previously described (20, 23). HEK England BioLabs Inc., MA, USA).
293T/17 cells and a baby hamster kidney cell line (BHK-T7) stably ex- In vitro transcription and capping of viral genomic RNA. In vitro
pressing T7 RNA polymerase were cultured in Dulbecco’s modified Ea- transcription and capping of viral genomic RNA were performed accord-
gle’s medium (DMEM; Life Technologies, NY, USA) with 10% fetal bo- ing to the manufacturer’s instructions. Briefly, the reverse-genetics plas-
vine serum (FBS; Thermo Scientific, MA, USA), 1% nonessential amino mid DNA was extracted from E. coli, linearized by NotI restriction enzyme
acids (NEAA; Invitrogen, NY, USA), and 1% antibiotic-antimycotic (In- digestion, and purified by phenol-chloroform extraction twice. Subse-
vitrogen, NY, USA). quently, genomic RNA was transcribed in vitro from the linearized plas-
Two passage levels of the WT PoSaV Cowden strain (Gn pig passage mid by using a MEGAscript T7 transcription kit (Life Technologies, NY,
level 5 [I-1113] and level 13 [R418]) from the small intestinal contents USA). The reaction mixture was treated with DNase, and the RNA was
(SICs) or large intestinal contents (LICs) of Gn pigs were used for se- purified with an RNeasy minikit (Qiagen, CA, USA) and analyzed by
quencing. TC PoSaV was propagated in LLC-PK cells (TC PoSaV-2010; agarose gel (1%) electrophoresis under denaturing conditions with form-
cell culture passage level 30) with 50 ␮M glycochenodeoxycholic acid aldehyde. The transcribed RNA was capped by using the ScriptCap m7G
(GCDCA; Sigma-Aldrich, MO, USA) as previously described (24). capping system (Cellscript, WI, USA), followed by RNA purification us-
Sequence analyses. The genomes of TC PoSaV-2010 (passage level 30) ing the RNeasy minikit. The RNA transcripts were suspended in RNase-
and WT PoSaV I-1113 (Gn pig passage level 5) and the VP1 region of WT free water to a final concentration of 500 ng/␮l for transfection.
PoSaV R418 (Gn pig passage level 13) were sequenced by the primer Transfection of BHK-T7 cells or HEK 293T/17 cells to rescue infec-
walking method based on the TC PoSaV genome (GenBank accession no. tious viruses. The purified RNA transcripts were transfected into 1-day-
AF182760), as previously described (25). The 5= and 3= ends were deter- old HEK 293T/17 or BHK-T7 cells (⬃50 to 70% confluent) in 24-well cell
mined by using 5=-rapid amplification of cDNA ends (RACE) and 3=- culture plates with Lipofectamine 2000 (Invitrogen, NY, USA). Briefly,
RACE methods. Sequence editing and assembly were performed by using 1-day-old HEK 293T/17 or BHK-T7 cells were washed with Opti-MEM I
the Lasergene software package (v10; DNASTAR Inc., WI, USA). Multi- (Invitrogen, NY, USA). Approximately 1.5 ␮g capped RNA and 4 ␮l Li-
ple-sequence alignment was done with ClustalW using the DNA Data pofectamine 2000 were diluted in 50 ␮l Opti-MEM I separately and incu-
Bank of Japan (DDBJ) (http://www.ddbj.nig.ac.jp). bated at room temperature for 5 min. The RNA and the Lipofectamine
Generation of full-length cDNA clones of PoSaV WT and TC chime- 2000 solution were then mixed (total volume of 100 ␮l) and incubated at
ric genomes, mutants, and revertant mutant strains. Plasmid pCV4A room temperature for 20 min before being added to HEK 293T/17 or
containing the full-length cDNA of TC PoSaV (TC PoSaV-2005; cell cul- BHK-T7 cell monolayers. After 6 h of incubation at 37°C, the supernatant
ture passage level 27) was provided by Kyeong-Ok Chang (20). The prim- was replaced with DMEM (10% FBS, 1% NEAA, and 1% antibiotic-anti-
ers for the generation of these chimeric clones are listed in Table 1. The mycotic). After 1 day of incubation at 37°C, HEK 293T/17 or BHK-T7 cell

1346 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

TABLE 1 Primers for generation of chimeric PoSaV clones


Primer Sequencea Location in PEC genome (positions) Orientation
ApaI-F 5=-GAGTCCAGACCAGTCCAGCCAGC 5203–5225 Forward
ApaI-R 5=-TGGGTAGTGGTTGATGATGTTG 6090–6069 Reverse
TC-3763-CTF 5=-CGTGAATGACCCAAGGTACCCCTTCTCACAACA 3747–3779 Forward
TC-3763-CTR 5=-TGTTGTGAGAAGGGGTACCTTGGGTCATTCACG 3779–3747 Reverse
TC-4145-AGF 5=-AGAAAAGAATGACCAAGGCAAAAGACGCCTGCTGTG 4122–4157 Forward
TC-4145-AGR 5=-CACAGCAGGCGTCTTTTGCCTTGGTCATTCTTTTCT 4157–4122 Reverse
TC-5671-ATF 5=-TTGGTGGGGCTATAGCATGTTTGGCACTTTACGTG 5654–5688 Forward
TC-5671-ATR 5=-CACGTAAAGTGCCAAACATGCTATAGCCCCACCAA 5688–5654 Reverse
TC-6004-CTF 5=-CCCGTGTCAATGGAAAGTACACTGACAACACAGGT 5987–6021 Forward
TC-6004-CTR 5=-ACCTGTGTTGTCAGTGTACTTTCCATTGACACGGG 6021–5987 Reverse
TC-6010-GAF 5=-CCCGTGTCAATGGAAAGCACACTAACAACACAGGTA 5987–6022 Forward
TC-6010-GAR 5=-TACCTGTGTTGTTAGTGTGCTTTCCATTGACACGGG 6022–5987 Reverse

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


TC-6023-GAF 5=-AGCACACTGACAACACAGGTAAGGCAGTGTTTCA 6002–6035 Forward
TC-6023-GAR 5=-TGAAACACTGCCTTACCTGTGTTGTCAGTGTGCT 6035–6002 Reverse
WT-5671-TAF 5=-TTGGTGGGGCTATAGCAAGTTTGGCACTTTACGTG 5654–5688 Forward
WT-5671-TAR 5=-CACGTAAAGTGCCAAACTTGCTATAGCCCCACCAA 5688–5654 Reverse
WT-6004-TCF 5=-CCCGTGTCAATGGAAAGCACACTAACAACACAGGT 5987–6021 Forward
WT-6004-TCR 5=-ACCTGTGTTGTTAGTGTGCTTTCCATTGACACGGG 6021–5987 Reverse
EcoRI-F 5=-GAGGCCTACGAGGAATTCAAG 4570–4590 Forward
EcoRI-R 5=-GAGCCTGATTAAAAGAATTCATAATA 6891–6866 Reverse
6111-F 5=-CAACAATGTTCAACACAGGAAC 6104–6125 Forward
6111-R 5=-GTTGAACATTGTTGATGCAGC 6117–6097 Reverse
6122-F 5=-CAACACAGAAACTGCCGTAAATG 6114–6138 Forward
6122-R 5=-GGCAGTTTCTGTGTTGAATATTG 6129–6107 Reverse
6111-6122-F 5=-CAATGTTCAACACAGAAACTGCC 6107–6129 Forward
6111-6122-R 5=-CAGTTTCTGTGTTGAACATTGTTG 6127–6104 Reverse
QCback-F 5=-GGCTGCATCAACAATATTCAACACAGGAACTGCC 6096–6129 Forward
QCback-R 5=-GGCAGTTCCTGTGTTGAATATTGTTGATGCAGCC 6129–6096 Reverse
a
Boldface and underlined nucleotides are the mutation sites; underlined sequences are EcoRI digestion sites.

lysates were harvested by freezing and thawing once, followed by centrif- IF and IHC staining for detection of VP1 proteins in cell culture.
ugation at 2,095 ⫻ g for 5 min to remove cell debris. The cell lysates were Immunofluorescence (IF) staining was performed for the detection of
used to inoculate LLC-PK cells to generate virus pools. VP1 protein in mutant-infected cells (20), while IHC staining was per-
Recovery of progeny virus in LLC-PK cells. The LLC-PK cell mono- formed for virus infectivity titration (23). Briefly, cell monolayers were
layers in 6-well plates were washed with minimum essential medium fixed with 10% neutral formalin buffer at room temperature for 30 min,
(MEM) and inoculated with HEK 293T/17 or BHK-T7 cell lysates in the and the fixed cells were then permeabilized with 1% Triton X-100 in PBS
presence of 50 ␮M GCDCA. Cytopathic effects (CPEs) were monitored at room temperature for 10 min. Gn pig hyperimmune serum to the WT
daily. Infected cells were incubated for up to 6 days postinoculation, be- PoSaV Cowden strain was used as a primary antibody (18). Fluorescein
fore harvesting of the first passage of each mutant. Each mutation was isothiocyanate (FITC)-conjugated goat anti-swine IgG(H⫹L) serum
confirmed by reverse transcription-PCR (RT-PCR) with primer sets cov- (KPL, MD, USA) or horseradish peroxidase (HRP)-conjugated goat anti-
ering the mutated region, followed by sequence analysis. swine IgG(H⫹L) serum (KPL, MD, USA) was used as a secondary anti-
Plaque assays. Tenfold serially diluted samples were inoculated into body. The IF signal was observed by using an IX70 fluorescence micro-
wells of 6-well cell culture plates. After incubation at 37°C for 1.5 h with scope (Olympus, PA, USA). For IHC, cells were stained with the substrate
rocking, the inoculum was removed, and the cell monolayer was overlaid 3-amino-9-ethylcarbazole (AEC) (Sigma-Aldrich, MO, USA) at room
with 0.85% low-melting-temperature agarose (Sigma-Aldrich, MO, USA) temperature for at least 2 h and observed by using light microscopy.
in MEM supplemented with 50 ␮M GCDCA (20). After plaques formed Three-dimensional structural analyses of the VP1 proteins of WT
(5 days postinoculation), cell monolayers were stained with 1 ml of a and TC PoSaVs. The three-dimensional (3D) structure of a SaV VP1
0.03% neutral red–phosphate-buffered saline (PBS) solution for 30 min at protein is not available in the database. The VP1 protein of WT PoSaV
37°C. The solution was removed, and the plaques were counted and ob- shares higher sequence identity (38%) with that of feline calicivirus (FCV)
served under a microscope. The plaque sizes were quantified by using the (PDB accession no. 3M8L) than with those of San Miguel sea lion virus
Icy bioimage program (26). (SMSV) (34%) (PDB accession no. 2GH8) and recombinant Norwalk
Growth kinetics test for progeny mutants in LLC-PK cells. A growth virus (rNV) (29%) (PDB accession no. 1IHM). When the sequence iden-
kinetics curve for each mutant was determined by collecting cell lysates at tity is 30 to 50%, the obtained model tends to have ⬃90% of the main
different postinoculation time points. LLC-PK cells in 6-well plates were chain modeled with a 1.5-Å root mean square error (27). Therefore, the
incubated with each mutant virus at a multiplicity of infection (MOI) of VP1 dimer structural models of WT and TC PoSaVs were constructed
0.01 for 1 h. The inoculum was removed, and the plate was washed once based on the crystal structure of the FCV VP1 protein at a resolution of
before the addition of maintenance MEM in the presence of 50 ␮M 3.40 Å by the homology modeling method using “MOE-Align” and
GCDCA. Supernatants and cell lysates were collected at 24, 48, 72, and 96 “MOE-Homology” in the Molecular Operating Environment (MOE)
h postinoculation (hpi) after three cycles of freezing and thawing. Virus (version 2014-09; Chemical Computing Group Inc., Quebec, Canada).
infectivity titers were determined in LLC-PK cells as the 50% tissue cul- Twenty-five intermediate models were obtained from one homology
ture infectious dose (TCID50) by immunohistochemistry (IHC) staining modeling with the MOE, among which the intermediate models with the
using 96-well plates, as described below (23). best scores were selected according to the generalized Born (GB)/volume

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1347


Lu et al.

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest

FIG 1 Diagrams of constructions of TC and WT PoSaVs and the mutants derived from the pCV4A backbone. The 8 amino acid (aa) residues, at positions 1252
and 1379 in the ORF1 polyprotein and at positions 178, 289, 291, 295, 324, and 328 in VP1, that differed between the WT (red) and TC (green) PoSaVs are
indicated in boldface and italic type for each mutant. The TCVP1-D291N and TCVP1-R295K mutants were tested in Gn pigs with WT and TC strains as controls.
The TC-WTVP1-C178S&Y289H mutant was not tested (NT) due to the 2-log10-lower virus infectivity titer that could not be equalized by further concentration.
Replication of the mutants in cell culture or in Gn pigs is noted.

1348 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

TABLE 2 Experimental design for inoculation of Gn pigs with WT or TC PoSaV Cowden or mutants
No. of Gn pigs euthanized at acute
Groupa Oral inoculum No. of Gn pigs Titer (log10 GE/ml in MEM [5 ml/pig])b infection phase (PIDs 5–7)
1 TC Cowden strain 9 11.5 3
2 TCVP1-R295K mutant 7 11.5 2
3 TCVP1-D291N mutant 9 11.5 3
4 WT Cowden strain PS799 7 11.5 3
5 MEM 4 2
a
All pigs were 4 to 7 days of age at inoculation.
b
A RT-qPCR titer of 11.5 log10 GE/ml is equivalent to ⬃7.0 log10 TCID50/ml by an infectivity assay.

integral (VI) scoring function. The final 3D models were thermodynam- Gn pigs and experimental design. Gn pigs were derived and main-
ically simulated by energy minimization using the AMBER10 extended tained as previously described (18, 32). A total of 36 Gn pigs were assigned

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


Huckel theory (EHT) force field combined with the GB model of aqueous to five groups (Table 2) and inoculated orally with (i) TC PoSaV (cell
solvation implemented in the MOE (28–30). Physically unacceptable local culture passage level 30) (n ⫽ 9), (ii) TCVP1-R295K (n ⫽ 7), (iii) TCVP1-
structures of the optimized 3D models were further refined on the basis of D291N (n ⫽ 9), (iv) WT PoSaV (PS799) (Gn pig passage level 13) (n ⫽ 7),
evaluation using the Ramachandran plot in the MOE. The structures of or (v) MEM (n ⫽ 4). TC PoSaV and the TCVP1-D291N and TCVP1-R295K
WT PoSaV VP1 dimers were generated from the monomeric structures by mutants were harvested from cell culture and concentrated to ⬃7.0 log10
the MOE on the basis of the assembly information for the FCV VP1 crystal TCID50/ml (equivalent to a real-time quantitative RT-PCR [RT-qPCR]
structures. The quality of the models was assessed by using the 3D struc- titer of ⬃11.5 log10 genome equivalents [GE]/ml) by ultracentrifugation
ture evaluation program Verify3D (31). at 126,000 ⫻ g for 1.5 h at 4°C. The WT PoSaV Cowden strain inoculated
Prediction of effects of point mutations on stability of PoSaV. The into Gn pigs was filtered through 0.22-␮m-pore-size filters prior to inoc-
change in the stability of the WT PoSaV VP1 protein by each mutation was ulation. Each pig was inoculated orally with a 5-ml inoculum containing
analyzed by using the Protein Design application in the MOE (version TC PoSaV or mutant PoSaVs (⬃7.0 log10 TCID50/ml) or containing WT
2014-09). The structure of WT PoSaV VP1 was constructed as described PoSaV with a RT-qPCR titer of ⬃11.5 log10 GE/ml.
above. The single point mutations in VP1 were generated individually, All inoculated Gn pigs were observed daily for clinical signs. Their
and ensembles of protein conformations were generated by using the feces were scored as 0 for normal, 1 for pasty, 2 for semiliquid, and 3 for
LowMode molecular dynamics (MD) module with Boltzmann distribu- liquid, with fecal scores of ⱖ2 indicating diarrhea (18). Rectal swabs (RSs)
tion in the MOE to calculate average stability. The stability scores in the were collected daily for titration of virus shedding. Blood was collected for
structures refined by energy minimization were obtained by using the serum from each Gn pig before inoculation; at postinoculation days
stability scoring function of the Protein Design application in the MOE. (PIDs) 1, 3, 6, 9, 16, 23, and 27; and at euthanasia. Two to three Gn pigs of

TABLE 3 Summary of amino acid substitutions in the genomes from different passages of the WT or TC PoSaV Cowden strain
WT-PoSaV sequence in Gn pigs at passage: TC-PoSaV sequence in LLC-PK cells at passage:
Amino acid Nucleotide
Gene positionc position 5 b a
5 (I-1113 ) 13 (R418 ) a a
13 (PS499 ) 20b 27 30
NS1 17 59 TTT TTT TTT TTT TTT TCT (S17F) TTT
18 62 GAC GGC (G18D) GGC GGC GAC GGC GGC
24 80 CCA CCA CCA CCA CCA CCA CTA (L24P)
29 94 GCG GCG GCG GCG GCG GCG ACG (T29A)

NS3 356 1075 ATT ATT ATT ATT ATT GTT (V356I) ATT
367 1109 AAG AAG AAG AAG AAG AGG (R367K) AAG

NS4 733 2207 GGC GAC/GGC GGC GGC GGC (D733G) GGC GGC

NS7 1252 3763 TAC TAC TAC TAC CAC (Y1252H) CAC CAC
1379 4145 AGA AGA AGA AGA AAA (R1379K) AAA AAA
1392 4185 ATA ATA ATG/ATA ATA ATA(M1392I) ATA ATA

VP1 75 5362 ACA ACA ACA ACA ACA ACA GCA (A1785T)
178 5671 TGT TGT TGT TGT AGT (C178S) AGT AGT
289 6004 TAC TAC TAC TAC CAC (Y289H) CAC CAC
291 6010 AAC AAC AAC AAC GAC (N291D) GAC GAC
295 6023 AAG AAG AAG AAG AGG (K295R) AGG AGG
324 6111 ATA ATG ATG ATG ATA (M324I) ATA ATA
328 6122 GGA GAA GAA GAA GGA (E328G) GGA GGA

ORF2 27 6851 CAT CAT CAT CAT CAT CAT CAA (E27H)
35 6873 AAT AAT AAT AAT AAT AAT GAT (D35N)
a
Sample identification based on the labeling system in the laboratory.
b
See reference 25.
c
Boldface numbers refer to consensus amino acid substitutions among different passages of TC compared with WT PoSaV.

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1349


Lu et al.

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


FIG 2 Growth kinetics and representative plaques of TC-pCV4A (TC) and the culturable mutants TC-WTRdRp, TCVP1-D291N (291), TCVP1-R295K (295), and
TC-WTVP1-C178S&Y289H (Double) in LLC-PK cells. (A) The plaque sizes of the TCVP1-D291N, TCVP1-R295K, and TC-WTVP1-C178S&Y289H mutants are
smaller than those of TC-pCV4Aand TC-WTRdRp in LLC-PK cells. (B) LLC-PK cells were inoculated with each virus at a MOI of 0.01. Cell lysates were collected
at 24, 48, 72, and 96 hpi for titration of infectious virus. TC-pCV4A and TC-WTRdRp replicated in LLC-PK cells to significantly higher titers (72 hpi) than did the
culturable mutants (P ⬍ 0.05 by one-way ANOVA followed by Duncan’s multiple-range test on log10-transformed titers).

each group were euthanized at the acute phase of infection: on the next virus-like particles (VLPs) (33) at 4°C overnight, followed by incubation
day after an increase of the fecal viral RNA titer or on the day following the with Alexa Fluor 488-conjugated goat anti-guinea pig IgG(H⫹L) serum
onset of clinical signs. The remaining pigs were euthanized after fecal virus (Life Technologies, NY, USA) at room temperature for 1 h. Thereafter,
shedding was no longer detected at the termination of the experiment. tissue sections were counterstained with 4=,6-diamidino-2-phenylindole
Animal care and use for these studies were approved by the Institutional (DAPI) for nuclei and examined by using fluorescence microscopy.
Animal Care and Use Committee (IACUC) at The Ohio State University. Detection of PoSaV RNA in rectal swabs. RSs were collected, sus-
Mucosal antibody samples were collected at necropsy by scraping the pended in 4 ml of MEM, and centrifuged at 2,095 ⫻ g for 30 min. The
mucosa from the ileum and centrifuging the sample at 2,095 ⫻ g for 20 supernatant was collected as a 10% fecal suspension and stored at ⫺20°C
min at 4°C. until RNA extraction. Total RNA was extracted from 50-␮l RS suspen-
Histopathological examination. At necropsy, blood, SICs, and LICs sions by using a MagMax RNA extraction kit (Life Technologies, NY,
were collected from each Gn pig. Fresh duodenum; proximal, mid-, and USA) according to the manufacturer’s instructions. The virus titer was
distal jejunum; ileum; colon; cecum; liver; spleen; lung; and kidney spec- determined by using One-Step TaqMan SaV-specific RT-qPCR as de-
imens were collected and immersed immediately in 10% neutral buffered scribed previously (23).
formalin (NBF). Tissues fixed in 10% NBF were trimmed, embedded in Detection of PoSaV-specific antibodies in serum and mucosal sam-
paraffin, sectioned at 4 ␮m, stained with Harris’ hematoxylin and alco- ples by an enzyme-linked immunosorbent assay (ELISA). A recombi-
holic eosin Y solution (H&E; Sigma-Aldrich, MO, USA), and examined nant baculovirus expressing PoSaV VP1 was generated as previously de-
for histopathology microscopically. scribed and used to infect Sf9 cells for VLP production (33). Briefly,
Duodenum; proximal, mid-, and distal jejunum; ileum; colon; and purified PoSaV VLPs were used as antigens to coat Nunc 96-well plates
cecum specimens were collected in duplicate, immersed in a sucrose so- (MaxSorp surface; Thermo Scientific, MA, USA) at 4°C overnight at a
lution (130 mM Na2HPO4, 30 mM KH2PO4, 10% [wt/vol] sucrose, and final concentration of 2 ␮g/ml (100 ng/well) in 0.05 M carbonate buffer
0.01% sodium azide [pH 7.2]) on ice, embedded in a optimum-cutting- (pH 9.6). The plates were blocked with 2% nonfat dry milk (NFDM) in
temperature (OCT) compound (Sakura, PA, USA), stored at ⫺20°C over- PBST at 37°C for 1 h. After being washed three times with PBST, serum
night, and then sectioned at 4 to 7 ␮m in a cryostat microtome. To detect samples were 4-fold serially diluted in PBST containing 2% NFDM and
the PoSaV antigen in tissues, frozen sections were fixed with acetone for 20 added to the wells. The plates were incubated at 37°C for 1 h and washed
min at ⫺20°C, followed by washing with PBS twice for 5 min each. Tissue with PBST three times. HRP-conjugated goat anti-swine IgG(H⫹L) se-
sections were then blocked with 5% normal goat serum in 0.01 M PBS– rum (KPL, MD, USA) diluted 1:3,000 or HRP-conjugated goat anti-swine
0.05% Tween 20 (PBST) (pH 7.2) for 20 min. After blocking, the tissue IgA serum (AbD Serotec, NC, USA) diluted 1:5,000 in PBST containing
sections were incubated with hyperimmune guinea pig serum to PoSaV 2% NFDM was added to wells, followed by incubation at 37°C for 1 h.

1350 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


FIG 3 Superimposition of the modeled structures of PoSaV Cowden and the template FCV structure and the 3D models of WT and TC PoSaV VP1s. (A) Superim-
position of the modeled structures (WT PoSaV Cowden on the left and TC PoSaV Cowden on the right) and the template structure (FCV VP1 [PDB accession no.
3M8L]) by homology modeling. The blue ribbon structure denotes WT PoSaV Cowden VP1, the cyan ribbon structure denotes TC PoSaV Cowden VP1, and the
magenta ribbon structure denotes the template structure of FCV VP1. (B) Amino acid residue 178 was located at the dimer-dimer interface in the S domain; residues 289,
291, 324, and 328 were located in the P2 region; and residue 295 was located on the interface of two monomeric VP1 proteins in the P2 region.

After the plates were washed three times with PBST, the substrate were determined by using the Reed-Muench method (34) and expressed
3,3=,5,5=-tetramethylbenzidine (TMB; KPL, MD, USA) was added to each as the reciprocal of the highest serum dilution that inhibited PoSaV infec-
well for color development at room temperature. An equal volume of 1 M tion in 50% of wells.
phosphoric acid was added to terminate the reactions after 5 min of incu- Statistical analysis. One-way analysis of variance (ANOVA) fol-
bation at room temperature. The absorbance at 450 nm was measured by lowed by Duncan’s multiple-range test was used to assess differences
using a spectrometer (SpectraMax 430 PC; Molecular Devices LLC, CA, in plaque sizes, mean durations of virus shedding, and log-trans-
USA). The antibody titer was determined as the reciprocal of the highest formed titers (including antibody titer, VN antibody titer, and viral
serum dilution with an absorbance value greater than or equal to the mean RNA titer) among groups. One-way ANOVA was used to assess ratios
absorbance of a series of negative-control serum samples plus 3 times the of villus height to crypt depth (VH/CD ratios) and the mean numbers
standard deviation (SD) of the negative controls (33). of antigen-positive cells per villus. A significance level of a P value of
Virus neutralization test. Serum samples were tested for virus neu- ⬍0.05 was used for all comparisons.
tralization (VN) antibodies to PoSaV by a 50% cell culture infectivity Nucleotide sequence accession numbers. The genomes of WT PoSaV
reduction test. A total of 100 TCID50/well of TC PoSaV was incubated I-1113 (Gn pig passage level 5) and TC PoSaV-2010 (passage level 30)
with an equivalent volume of 4-fold serially diluted serum samples at 37°C were deposited in GenBank under accession numbers KT922087 and
for 1 h before application to cell monolayers. Quadruplicate wells were KT922088, respectively. The VP1 region of WT PoSaV R418 (Gn pig
used for each serum dilution. Nonneutralized PoSaV was detected on passage level 13) was deposited in GenBank under accession number
LLC-PK cells by IHC staining as described above. The VN antibody titers KT945132.

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1351


Lu et al.

RESULTS
Consistent mutations occur in the RdRp and VP1 regions at dif-
ferent passages of TC PoSaV compared to WT PoSaV. To inves-
tigate which genes were critical for PoSaV adaptation to cell cul-
ture, the genome of TC PoSaV at passage level 30 in LLC-PK cells
was sequenced in this study and compared with those of WT
PoSaV at pig passage levels 5 and 13, TC PoSaV at cell culture
passage level 20 (25), and the infectious clone pCV4A carrying the
cDNA of TC PoSaV-2005 (cell culture passage level 27) (20). Two
and six conserved amino acid mutations were observed in the
RdRp (residues 1252 and 1379) and VP1 of (residues 178, 289,
291, 295, 324, and 328) regions TC PoSaV (Fig. 1 and Table 3),
respectively.
The VP1 region is critical for cell culture adaptation of

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


PoSaV. To examine whether the RdRp or the VP1 region was
critical for PoSaV adaptation to cells, we engineered two chimeric FIG 4 Changes in ⌬G for the C178S, Y289H, N291D, K295R, M324I, and
genomes (Fig. 1) based on the previously established PoSaV re- E328G point mutations. Mutations C178S, Y289H, N291D, and E328G de-
verse-genetics system, pCV4A: (i) TC-WTRdRp, whose amino acid creased thermodynamic stability, whereas K295R and M324I compensated for
residues 1252 and 1379 of the polyprotein (RdRp region) were the changes.
mutated from the TC to the WT phenotype (H1252Y and
K1379R), and (ii) TC-WTVP1, whose partial VP1 region (nt 5227
stitutions D291N, R295K, and TC-WTVP1-C178S&Y289H can al-
to 6060 and amino acids 30 to 308, excluding amino acid residues
ter viral replication in LLC-PK cell cultures, the growth kinetics
324 and 328) was replaced with the corresponding WT fragment.
and plaque sizes of TCVP1-D291N, TCVP1-R295K, and TC-
The capped genomic RNA transcripts were transfected into
WTVP1-C178S&Y289H were compared to those of TC-pCV4A in
BHK-T7 cells. After infection of LLC-PK cells with BHK-T7 cell
LLC-PK cells (Fig. 2). The TCVP1-D291N (0.041 ⫾ 0.011 mm2) and
lysates, the PoSaV VP1 proteins were detected exclusively by an
TCVP1-R295K (0.047 ⫾ 0.016 mm2) viruses and especially the TC-
immunofluorescence assay (IFA) in LLC-PK cells inoculated with
WTVP1-C178S&Y289H (0.018 ⫾ 0.003 mm2) virus formed signif-
TC-WTRdRp- but not TC-WTVP1-transfected products. We fur-
icantly smaller (P ⬍ 0.05) plaques than those formed by TC-
ther compared the growth kinetics and plaque sizes of the TC-
pCV4A (0.146 ⫾ 0.066 mm2) and TC-WTRdRp (0.168 ⫾ 0.052
WTRdRp virus to those of the TC-pCV4A virus in LLC-PK cells (Fig. mm2) (Fig. 2A). The infectious titers of TCVP1-D291N, TCVP1-
2). TC-WTRdRp had plaque sizes (0.168 ⫾ 0.052 mm2) similar to R295K, and TC-WTVP1-C178S&Y289H increased postinocula-
those of TC-pCV4A (0.146 ⫾ 0.066 mm2) and had growth kinetics tion. However, the peak titers of TC-pCV4A (7.1 ⫾ 0.2 log10
similar to those of TC-pCV4A in LLC-PK cells, showing increasing TCID50/ml) and TC-WTRdRp (7.0 ⫾ 0.0 log10 TCID50/ml) were
titers between 0 and 72 hpi with similar peak titers (7.1 ⫾ 0.2 log10 the highest, followed by TCVP1-R295K (6.6 ⫾ 0.0 log10 TCID50/
TCID50/ml for TC-pCV4A and 7.0 ⫾ 0.0 log10 TCID50/ml for TC- ml), TCVP1-D291N (6.4 ⫾ 0.0 log10 TCID50/ml), and TC-WTVP1-
WTRdRp). These results suggested that the VP1 region, but not the C178S&Y289H (4.9 ⫾ 0.1 log10 TCID50/ml) (Fig. 2B). These data
RdRp region, was critical for cell culture adaptation of PoSaV. led us to conclude that the D291N and R295K amino acid substi-
Four (residues 178, 289, 324, and 328) of the six amino acid tutions in the VP1 region reduced PoSaV replication in LLC-PK
residues in the VP1 region were essential for PoSaV adaptation to cells.
LLC-PK cells. To address which of the individual mutations in the Comparative structural analyses of the VP1 proteins of TC
VP1 region was essential for PoSaV adaptation, each of the sites and WT PoSaVs predicted the location and potential function of
was mutated to the WT sequence individually (Fig. 1): TCVP1- amino acid residues. The 3D structural models of both the TC
S178C, TCVP1-H289Y, TCVP1-D291N, TCVP1-R295K, TCVP1- and WT PoSaV Cowden strains matched the template structure
I324M, and TCVP1-G328E. The TCVP1-D291N and TCVP1-R295K (FCV VP1 [PDB accession no. 3M8L]) (Fig. 3A). Subsequently,
mutants replicated in LLC-PK cells. No infectious virus was res- 3D structural analysis was performed to examine whether the
cued from LLC-PK cells infected with the transfection lysates of residue changes in VP1 between the WT and TC PoSaVs re-
TCVP1-S178C, TCVP1-H289Y, TCVP1-I324M, or TCVP1-G328E in- sulted in structural changes. Amino acid residue 178 was lo-
fectious clones. Furthermore, when the point mutations of TC- cated in the S domain near the dimer-dimer interface (Fig. 3B).
WTVP1 were mutated back to TC sequences, the double mutant C178 of the WT was exposed, but S178 of the TC strain was
(TC-WTVP1-C178S&Y289H, carrying TC amino acids at residues hidden. Amino acid residues 289, 291, 324, and 328 in PoSaV
324 and 328), instead of the individual point mutants (TC- were located at the P2 region, while amino acid 295 was located
WTVP1-C178S and TC-WTVP1-Y289H), was rescued in LLC-PK at the interface of two monomeric VP1 proteins forming a
cells. Infectious virus was rescued from infectious clones of the dimer at P2 (Fig. 3B).
TCVP1-I324M¡M324I and TCVP1-G328E¡E328G back muta- To investigate whether the mutations at residues 178, 289, 291,
tions. These results indicate that the 4 amino acids at residues 178, 295, 324, and 328 influenced the stability of WT PoSaV, we ana-
289, 324, and 328 in the VP1 region are essential for PoSaV adap- lyzed the changes in thermodynamic stability by the mutations
tation to LLC-PK cells. using the Protein Design application in the MOE. A positive num-
Single amino acid substitutions in the VP1 region alter PoSaV ber indicated decreased stability, while a negative number indi-
growth kinetics in vitro. To examine whether the amino acid sub- cated increased stability. The changes in ⌬G caused by the C178S,

1352 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


FIG 5 Geometric mean titers (GMT) of viral RNA shedding of TCVP1-D291N and TCVP1-R295K compared with those of TC-pCV4A and the WT PoSaV
Cowden strain in Gn pigs on various PIDs. Gn pigs were inoculated with the corresponding virus inoculum. Rectal swabs were collected daily for titration of viral
RNA shedding. The WT PoSaV Cowden strain had the highest peak titer and longest duration of viral RNA shedding among the experimental groups. The TC
PoSaV Cowden strain had the lowest peak titer and shortest duration of shedding among the experimental groups. Peak viral RNA shedding titers and durations
of shedding in TCVP1-D291N- and TCVP1-R295K-inoculated Gn pigs were intermediate between the WT and TC groups.

Y289H, N291D, K295R, M324I, and E328G point mutations were Clinical signs and histopathological lesions are observed ex-
0.54 ⫾ 0.04, 4.51 ⫾ 0.02, 1.80 ⫾ 0.24, ⫺1.43 ⫾ 0.31, ⫺0.42 ⫾ clusively in WT PoSaV-infected Gn pigs. Moderate diarrhea (fe-
0.15, and 1.86 ⫾ 0.37 kcal/mol, respectively (Fig. 4). The C178S, cal score of 2) was observed in three of seven (43%) WT PoSaV-
Y289H, N291D, and E328G mutations decreased the stability, inoculated Gn pigs. Diarrhea developed by PIDs 3 to 12 and
whereas K295R and M324I, especially K295R, compensated for persisted for 2 to 16 days. No diarrhea or other clinical signs were
the changes. observed in TC PoSaV Cowden-, TCVP1-D291N-, TCVP1-R295K-,
Single amino acid substitutions in the VP1 region alter or mock-inoculated Gn pigs.
PoSaV replication in pigs. To investigate whether the amino acid Microscopically, histopathological lesions were not observed
substitutions from the TC to the WT sequence in VP1 could re- in organs from the TC PoSaV-, TCVP1-D291N-, or TCVP1-R295K-
store the virulence of WT PoSaV in vivo, we performed pathogen- inoculated or control Gn pigs. WT PoSaV-inoculated Gn pigs
esis studies in Gn pigs. Among the experimental groups, the WT euthanized at the acute phase of infection exhibited mild-to-mod-
PoSaV-inoculated Gn pigs had a significantly longer duration of erate, diffuse, and atrophic enteritis, demonstrating shortened
viral RNA shedding (30.3 ⫾ 3.8 days) and higher peak viral RNA and blunt villi from the duodenum to the midjejunum of the small
titers (10.8 ⫾ 0.4 log10 GE/g) than those of the other three inocu- intestine (Fig. 6A to C). Duodenal, proximal, and midjejunal tis-
lated Gn pig groups. Relative to the mutants, the TCVP1-D291N- sues showed moderate, diffuse villous atrophy (Fig. 6A to C). The
inoculated Gn pigs had a longer duration of viral RNA shedding VH/CD ratios for duodenal and midjejunal tissues were 3.54 ⫾
(23.2 ⫾ 3.4 days) and a higher peak RNA titer (8.6 ⫾ 0.8 log10 1.06 and 3.98 ⫾ 1.84, respectively, significantly lower than those
GE/g) than those of the TCVP1-R295K- or TC PoSaV Cowden for control pigs (5.42 ⫾ 2.26 and 5.95 ⫾ 2.25, respectively) (Table
strain-inoculated Gn pigs (Fig. 5 and Table 4). 5). Interestingly, the VH/CD ratio for jejunal tissues from the

TABLE 4 Comparative viral RNA shedding parameters for TC, WT, and mutant PoSaV Cowden strains
Groupa No. of Gn pigs Time of onset (PIDs)b Duration (days) (SD)c Peak titer (log10 GE/ml) (SD)c Time of peak titer (PIDs)d
a a
TC 6 1–4 19.8 (2.6) 7.7 (0.4) 3–14
295 5 1–4 20.8 (3.4)a 7.9 (1.0)a 5–15
291 6 1–6 23.2 (3.4)b 8.6 (0.8)b 3–19
WT 4 1–3 30.3 (3.8)c 10.8 (0.4)c 6–10
NC 2 NAe NA NA NA
a
TC, 295, 291, WT, and NC refer to the TC PoSaV Cowden strain, TCVP1-R295K, TCVP1-D291N, the WT PoSaV Cowden strain, and the negative control, respectively.
b
The onset of RNA shedding refers to the PIDs when fecal viral RNA was first detected by RT-qPCR.
c
Superscript letters denote significant differences among the groups (determined by one-way ANOVA followed by Duncan’s multiple-range test).
d
Time of peak titer refers to PIDs that have the highest viral RNA titers by RT-qPCR.
e
NA, not applicable.

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1353


Lu et al.

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


FIG 6 Histopathological examination of small intestinal samples from WT PoSaV Cowden- or mock (negative-control [NC])-infected Gn pigs. Shown are H&E
staining (left) and IF staining (right) of samples collected from different regions of the small intestine. (A to C) Samples from WT PoSaV Cowden-inoculated Gn
pigs at PID 5, showing fusion, shortening (arrows), or blunting (arrowhead) of villi in the duodenum (A), proximal jejunum (B), and midjejunum (C). (D to F)
Samples from mock-inoculated Gn pigs at PID 5, showing normal villi in the duodenum (D), proximal jejunum (E), and midjejunum (F). Intestinal samples were
collected in duplicate for IF staining and for H&E staining. Bar, 50 ␮m.

TCVP1-D291N-inoculated Gn pigs was statistically lower than titrated (Fig. 8A to C). From PIDs 6 to 27, Gn pigs inoculated with
those for the TCVP1-R295K- and TC PoSaV Cowden-inoculated the WT had the highest IgG, IgA, and VN serum antibody titers.
and control Gn pigs (Table 5), indicating more severe villous at- Gn pigs inoculated with TCVP1-D291N had higher serum IgG,
rophy in the jejuna of TCVP1-D291N-inoculated Gn pigs than in IgA, and VN antibody titers than did those inoculated with TC or
TCVP1-R295K- and TC PoSaV-inoculated and control Gn pigs. TCVP1-R295K mutant PoSaV at PIDs 6 to 27. Gn pigs inoculated
Virus VP1 antigens were detected in the frozen tissues of WT with the WT also had the highest IgA mucosal antibody titers at
PoSaV-inoculated Gn pigs by IF staining. Most antigen-positive PID 27 (Fig. 8D). Gn pigs inoculated with TCVP1-D291N and TC
cells were distributed in the mature enterocytes lining the intesti- PoSaV had significantly higher IgA mucosal antibody titers than
nal villi of the midjejunum and, to a lesser extent, in the duode- did those inoculated with TCVP1-R295K (P ⬍ 0.05 by one-way
num and proximal jejunum (Fig. 6). Antigen-positive cells were ANOVA followed by Duncan’s multiple-range test on log4-trans-
rarely detected in the distal jejunum and ileum. The mean num- formed titers).
bers of antigen-positive cells per villus differed significantly
among different regions of the small intestine, with most of the DISCUSSION
positive epithelial cells being observed in the duodenum to midje- The genomes of the WT (Gn pig passage level 5) and TC PoSaV
junum, with an increasing trend from duodenum (1.18 ⫾ 0.55 Cowden strains (cell culture passage level 20) were reported in
cells) to proximal jejunum (2.45 ⫾ 0.42 cells) and to midjejunum 1999 (25). It was also reported that the WT PoSaV Cowden strain
(3.78 ⫾ 1.30 cells) (Fig. 7). Antigen-positive cells were not ob- grew in porcine primary kidney cell culture after 13 passages in Gn
served in any tissue sections from TC PoSaV-, TCVP1-D291N-, or pigs but only with mock Gn pig intestinal contents in medium
TCVP1-R295K-inoculated Gn pigs. (14). Therefore, we selected one Gn pig intestinal sample from
IgG, IgA, and VN antibody titers in pig serum samples were both the 5th and the 13th passages of WT PoSaV for genomic
sequence analysis. Besides the reported 6 amino acid substitutions
between WT and TC PoSaVs (25), we identified two additional
TABLE 5 Ratios of villus length to crypt depth in different regions of substitutions at amino acid residues 324 and 328 of VP1. We com-
small intestines of Gn pigs pared seven genomes from different passages of the WT and TC
Avg VL/CD ratio (SD)b PoSaV Cowden strains (Table 3). Compared to the WT PoSaV
Cowden strain, 8 of 19 amino acid mutations were conserved
Groupa Duodenum Jejunum Ileum
among all TC PoSaV genomes. The differences between the re-
TC 5.21 (0.86)a 5.93 (1.60)a 4.68 (0.52) ported WT PoSaV Cowden genome and our results are likely due
295 5.97 (1.15)a 6.44 (0.78)a 5.26 (0.66) to the intestinal content samples from different pigs and the dif-
291 5.32 (0.94)a 5.09 (1.47)b 5.20 (0.76)
ferent passage levels tested. Because the two newly identified sites
WT 3.54 (1.06)b 3.98 (1.84)c 5.23 (1.20)
NC 5.42 (2.26)a 5.95 (2.25)a 6.57 (0.33)
(residues 324 and 328 in the VP1 region) were conserved in the
a
newly sequenced genomes from the 5th and 13th Gn pig passages
TC, 295, 291, WT, and NC refer to the TC PoSaV Cowden strain, TCVP1-R295K,
TCVP1-D291N, the WT PoSaV Cowden strain, and the negative control, respectively.
of WT PoSaV, both sites were also included in this study. The
b
Superscript letters denote significant differences among the groups not sharing the establishment of the reverse-genetics system pCV4A for the
same letter (determined by one-way ANOVA). PoSaV Cowden strain provided an important tool to study

1354 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


FIG 7 Distribution of antigen-positive cells per villus in different regions of the small intestine of WT PoSaV-inoculated pigs. Mean numbers of antigen-positive
cells per villus were significantly different among the duodenum, proximal jejunum, and midjejunum of the small intestine (P ⬍ 0.05 by one-way ANOVA).

the molecular mechanisms of PoSaV cell culture adaptation (20). cessfully rescued from LLC-PK cells by using the one-step method
In this study, we generated a series of PoSaV mutants to test which (20, 35). However, for unknown reasons, we failed to rescue in-
of the 8 amino acids were critical for cell culture adaptation. In fectious virus. Because infectious murine norovirus (MNV) was
previous reports, a one-step in vitro transcription-and-capping rescued from RAW 264.7 cells by using two-step in vitro transcrip-
procedure was used to generate infectious PoSaV genomic RNA tion followed by capping (36), we tried the two-step method and
for transfection (20). Infectious PoSaV Cowden virions were suc- rescued infectious PoSaV from LLC-PK cells. The genome-linked

FIG 8 Serum and mucosal antibody responses in Gn pigs. (A) Geometric mean titers (GMT) of IgG antibodies to the corresponding PoSaV strains or mock (NC)
in serum samples from Gn pigs. (B) Geometric mean titers of IgA antibodies to the corresponding PoSaV strains or mock infection in serum samples of Gn pigs.
(C) Geometric mean titers of virus-neutralizing antibodies to the corresponding PoSaV strains or mock in serum samples of Gn pigs. (D) Geometric mean titers
of IgA antibodies to the corresponding PoSaV strains in ileal mucosal samples from Gn pigs. Serum samples were collected from each Gn pig before inoculation
and at PIDs 1, 3, 6, 9, 16, 23, and 27. Ileal mucosal samples were collected at PID 5 and PID 27. Data points marked with different letters at each day differed
significantly (P ⬍ 0.05 by one-way ANOVA followed by Duncan’s multiple-range test on log10-transformed titers).

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1355


Lu et al.

virus protein (VPg) is encoded by the NS5 gene and is covalently gesting that the two amino acid substitutions in the P2 subdomain
linked to the 5= end of the SaV genomic RNA (20, 37, 38). In our of VP1 affected PoSaV virulence in the host and could be better
system, the cap structure analog m7G(5=)ppp(5=)G was added to candidates than the TC PoSaV Cowden strain for PoSaV vaccine
the 5= end of the SaV genomic RNA transcripts to simulate SaV development.
VPg (36, 39). The first calicivirus structure was reported in 1994 for a pri-
Among the mutants, TCVP1-S178C, TCVP1-H289Y, TCVP1- mate calicivirus (42). Currently, structures of VLPs or virion par-
I324M, and TCVP1-G328E could not be recovered in 4 to 5 repeats ticles of caliciviruses have been determined for GI.1 and GII.10
of the experiment. Whether virus particles were formed was un- human NoVs (43, 44), GV MNV (45, 46), San Miguel sea lion
known. However, HEK 293T/17 cells transfected with in vitro- virus (47), Tulane virus (48), rabbit hemorrhagic disease virus
transcribed and -capped RNAs of TCVP1-S178C, TCVP1-H289Y, (49), and FCV (50). Therefore, structures are available for all clas-
TCVP1-I324M, and TCVP1-G328E showed several VP1-positive sified genera of the family Caliciviridae, except for the genera Sa-
cells by IHC staining using hyperimmune serum against the VLPs povirus and Nebovirus. Chen et al. reported that sapovirus showed
of the PoSaV Cowden strain (data not shown). This suggests that more structural similarity to vesivirus (22). The FCV VP1 protein
the defect may occur at any step post-VP1 expression, such as has the closest phylogenetic relatedness (37% amino acid identity)

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


virion assembly and binding to the receptors on LLC-PK cells or to that of the PoSaV Cowden strain among the existing calicivirus
entry into LLC-PK cells. TCVP1-D291N, TCVP1-R295K, and TC- VP1 structures. To understand the location of the mutation sites
WTVP1-C178S&Y289H were culturable in the LLC-PK cell line, based on structural predictions, we performed 3D structure mod-
although the titer of the TC-WTVP1-C178S&Y289H strain was 2 eling and analysis of the VP1 protein of the PoSaV Cowden strain
log10 units lower than those of the other culturable strains. In the using FCV VP1 as the template. In a previous study, the complete
in vitro study, both the TCVP1-D291N and TCVP1-R295K mutants removal of the P domain of recombinant Norwalk virus-like par-
showed reduced replication compared with that of TC PoSaV. ticles resulted in the formation of smooth particles, which dem-
However, in our Gn pig study, both the TCVP1-D291N and TCVP1- onstrated that the S domain is sufficient for assembly of the capsid
R295K mutants had relatively higher peak viral RNA titers, a lon- (51). Since the P2 region of caliciviruses is the most protruding
ger duration of fecal viral RNA shedding, and higher-level serum part of VP1 and is highly variable, it has been considered respon-
and mucosal antibody responses than those of TC PoSaV but sible for binding to host receptors (52). A recent study indicated
lower than those of WT PoSaV. This indicates that the replication that the ␣2,3- and ␣2,6-linked sialic acids on O-linked glycopro-
efficiencies of PoSaV mutants were discordant in vitro and in vivo. teins are receptors on LLC-PK cells for the PoSaV Cowden strain
In vitro, peak virus infectivity titers (highest to lowest) were TC (53). In our study, based on their locations in the structural
PoSaV ⬎ TCVP1-R295K ⬎ TCVP1-D291N ⬎ TC-WTVP1- model, amino acid position 178 was located in the S domain,
C178S&Y289H; in vivo, peak viral RNA titers (highest to lowest) which may influence VP1 oligomerization, virion assembly, and
were WT PoSaV ⬎ TCVP1-D291N ⬎ TCVP1-R295K ⬎ TC PoSaV. stability. Amino acid positions 289, 291, 324, and 328 were located
Because the critical mutation sites are all located in VP1, different in the P2 region, which affects binding to the receptors on LLC-PK
receptors were probably used by the PoSaV Cowden strain in vivo cells and may impair virus replication. Amino acid position 295
to infect small intestinal epithelial cells in pigs and in vitro to infect was located in the P2 region at the interface of two monomeric
porcine kidney LLC-PK cells. VP1 proteins, which may influence VP1 dimerization.
Three passages of MNV1 in the macrophage cell line RAW The changes in thermodynamic stability caused by the muta-
264.7 resulted in a total of 3 amino acid substitutions, which in- tions indicate whether or not the protein structure can be main-
cluded V716I and H845R in the 3A-like protease (NS4) region and tained. The overall change in thermodynamic stability is the sum
E296K in the P2 region of VP1. Only V716I and E296K were sus- of each estimated value. Decreasing the stability is a disadvantage
pected to be related to decreased virulence in mice and increased to maintain the structure, whereas increasing the stability is an
titers in RAW 264.7 cells (40, 41). Using reverse genetics, the advantage. Therefore, mutations that decrease stability are critical
K296E but not the I716V back mutation restored the virulence of for function, e.g., cell culture adaptation, rather than structure.
the MNV1 revertant in mice (41). Therefore, a single amino acid The mutations that increase stability then play a role in the com-
substitution in the P2 region of VP1 of a norovirus may affect its pensation for decreasing stability. In this study, thermodynamic
virulence in the host and replication efficacy in cell culture. A stability analysis indicated that C178S, Y289H, N291D, and
previous study concluded that the TC PoSaV Cowden strain was E328G decrease stability, while K295R and M324I increase stabil-
attenuated in vivo compared with the WT PoSaV Cowden strain ity. These results suggest that C178S, Y289H, N291D, and E328G
after oral inoculation of Gn pigs with a TC PoSaV Cowden strain would provide essential functions for cell culture adaptation,
cell culture supernatant (cell culture passage 20) or a filtrate of the whereas K295R and M324I would compensate for the decreased
WT PoSaV Cowden strain (18). In our study, we concentrated the stability of C178S, Y289H, N291D, and E328G.
TC PoSaV Cowden strain, TCVP1-D291N, and TCVP1-R295K, We compared 12 WT GIII PoSaV VP1 protein sequences avail-
which were 1-log10-higher doses than those used in the previous able in GenBank (http://www.ncbi.nlm.nih.gov/nucleotide/). We
study. However, clinical signs were observed exclusively in the WT found that C178 in the S domain is conserved among all 12 WT
PoSaV-infected pigs but not in the TC PoSaV Cowden strain-, GIII PoSaVs (WT PoSaV Cowden [GenBank accession no.
TCVP1-D291N-, or TCVP1-R295K-infected Gn pigs. These results KT922087], SaV1-CHN [accession no. ACP43737], HW20-
indicated that even at a 1-log10-higher inoculation dose, the TC KOR [accession no. ADN84680], ID3-HUN [accession no.
PoSaV Cowden strain did not cause diarrhea in Gn pigs. Although ABD38714], LL14-US [accession no. AAR37376], MM280-US
the TCVP1-D291N and TCVP1-R295K revertants did not cause di- [accession no. AAX32888], JJ259-US [accession no. AAX37311],
arrhea in Gn pigs, they replicated more efficiently and induced QW270-US [accession no. AAX37314], PES-VENEZ [accession
relatively higher-level immune responses than TC PoSaV, sug- no. AAY88248], PoS6-HUN [accession no. ACS68238], PoS9-

1356 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3


Mutations in VP1 Make Porcine Sapovirus Grow In Vitro

HUN [accession no. ACS68240], and s20-JAP [accession no. acute gastroenteritis of young children in the community. J Infect Dis
BAE94661]), suggesting that C178S may be the most critical mu- 181(Suppl 2):S288 –S294. http://dx.doi.org/10.1086/315590.
6. Sakai Y, Nakata S, Honma S, Tatsumi M, Numata-Kinoshita K, Chiba
tation during PoSaV Cowden strain tissue culture adaptation.
S. 2001. Clinical severity of Norwalk virus and Sapporo virus gastroenteri-
In this study, WT PoSaV antigen was observed in epithelial tis in children in Hokkaido, Japan. Pediatr Infect Dis J 20:849 – 853. http:
cells of the Gn pig small intestine from the duodenum to midje- //dx.doi.org/10.1097/00006454-200109000-00005.
junum but rarely in the distal jejunum or ileum. We confirmed the 7. Hansman GS, Saito H, Shibata C, Ishizuka S, Oseto M, Oka T, Takeda
region of PoSaV infection in Gn pigs, as reported previously (18). N. 2007. Outbreak of gastroenteritis due to sapovirus. J Clin Microbiol
45:1347–1349. http://dx.doi.org/10.1128/JCM.01854-06.
Also, as previously reported, morphological alterations in duode-
8. Cubitt WD, Pead PJ, Saeed AA. 1981. A new serotype of calicivirus
nal and jejunal villi were observed in WT PoSaV Cowden-infected associated with an outbreak of gastroenteritis in a residential home for the
Gn pigs (18, 54). IF staining of small and large intestinal impres- elderly. J Clin Pathol 34:924 –926. http://dx.doi.org/10.1136/jcp.34.8.924.
sion smears as well as villus length measurement also confirmed 9. Svraka S, Vennema H, van der Veer B, Hedlund KO, Thorhagen M,
that the small intestine was the major infection site (18, 54). Our Siebenga J, Duizer E, Koopmans M. 2010. Epidemiology and genotype
analysis of emerging sapovirus-associated infections across Europe. J Clin
data support results of previous studies of the pathogenesis of the Microbiol 48:2191–2198. http://dx.doi.org/10.1128/JCM.02427-09.
WT PoSaV Cowden strain in Gn pigs. 10. Kobayashi S, Fujiwara N, Yasui Y, Yamashita T, Hiramatsu R, Mina-

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


This study demonstrated that cell culture adaptation of the gawa H. 2012. A foodborne outbreak of sapovirus linked to catered box
PoSaV Cowden strain is due to amino acid substitutions in the lunches in Japan. Arch Virol 157:1995–1997. http://dx.doi.org/10.1007
VP1 region. The single-revertant mutation (from the TC to /s00705-012-1394-8.
11. Lee LE, Cebelinski EA, Fuller C, Keene WE, Smith K, Vinje J, Besser
the WT phenotype) at certain positions, positions 291 and 295, in JM. 2012. Sapovirus outbreaks in long-term care facilities, Oregon and
the VP1 region reduced virus replication in vitro but partially re- Minnesota, USA, 2002-2009. Emerg Infect Dis 18:873– 876. http://dx.doi
gained PoSaV replication efficiency in vivo. The genetic basis de- .org/10.3201/eid1805.111843.
lineated for cell culture adaptation of PoSaV may provide new 12. Parwani AV, Flynn WT, Gadfield KL, Saif LJ. 1991. Serial propagation of
critical information for the rescue of other uncultivable PoSaVs porcine enteric calicivirus in a continuous cell line. Effect of medium
supplementation with intestinal contents or enzymes. Arch Virol 120:
and human SaVs. In future studies, we plan to construct reverse- 115–122.
genetics systems for selected PoSaV and human SaV strains by 13. Karst SM, Wobus CE, Lay M, Davidson J, Virgin HW, IV. 2003.
introducing site-directed mutations at structurally corresponding STAT1-dependent innate immunity to a Norwalk-like virus. Science 299:
positions in the VP1 protein to rescue such unculturable SaVs. 1575–1578. http://dx.doi.org/10.1126/science.1077905.
14. Flynn WT, Saif LJ. 1988. Serial propagation of porcine enteric calici-
virus-like virus in primary porcine kidney cell cultures. J Clin Micro-
ACKNOWLEDGMENTS biol 26:206 –212.
We acknowledge Juliette Hanson, Jeff Ogg, Andrew Wright, Ronna 15. Saif LJ, Ward LA, Yuan L, Rosen BI, To TL. 1996. The gnotobiotic piglet
Wood, and Megan Strother for assistance with animal care and Chun- as a model for studies of disease pathogenesis and immunity to human
Ming Lin, Xiaohong Wang, and Susan Sommer-Wagner for technical rotaviruses. Arch Virol Suppl 12:153–161.
16. Meurens F, Summerfield A, Nauwynck H, Saif L, Gerdts V. 2012. The
assistance.
pig: a model for human infectious diseases. Trends Microbiol 20:50 –57.
Zhongyan Lu was supported by a scholarship provided by the China http://dx.doi.org/10.1016/j.tim.2011.11.002.
Scholarship Council. Salaries and research support were provided by state 17. Hart EA, Caccamo M, Harrow JL, Humphray SJ, Gilbert JG, Trevanion
and federal funds provided to the Ohio Agricultural Research and Devel- S, Hubbard T, Rogers J, Rothschild MF. 2007. Lessons learned from the
opment Center (OARDC), The Ohio State University. initial sequencing of the pig genome: comparative analysis of an 8 Mb
The funders had no role in study design, data collection and interpre- region of pig chromosome 17. Genome Biol 8:R168. http://dx.doi.org/10
tation, or the decision to submit the work for publication. .1186/gb-2007-8-8-r168.
18. Guo M, Hayes J, Cho KO, Parwani AV, Lucas LM, Saif LJ. 2001.
Comparative pathogenesis of tissue culture-adapted and wild-type
FUNDING INFORMATION Cowden porcine enteric calicivirus (PEC) in gnotobiotic pigs and induc-
HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID) tion of diarrhea by intravenous inoculation of wild-type PEC. J Virol 75:
provided funding to Linda J. Saif under grant numbers R01 AI49742-04 9239 –9251. http://dx.doi.org/10.1128/JVI.75.19.9239-9251.2001.
and R21 AI081009-2. HHS | NIH | National Institute of Allergy and In- 19. Saif LJ, Bohl EH, Theil KW, Cross RF, House JA. 1980. Rotavirus-like,
fectious Diseases (NIAID) provided funding to Kyeong-Ok Chang under calicivirus-like, and 23-nm virus-like particles associated with diarrhea in
grant number U01AI08001. China Scholarship Council provided funding young pigs. J Clin Microbiol 12:105–111.
to Zhongyan Lu. HHS | NIH |National Institute of Allergy and Infectious 20. Chang KO, Sosnovtsev SV, Belliot G, Wang Q, Saif LJ, Green KY. 2005.
Diseases (NIAID) provided funding to Qiuhong Wang under grant num- Reverse genetics system for porcine enteric calicivirus, a prototype sapo-
ber R21 AI081009-2. virus in the Caliciviridae. J Virol 79:1409 –1416. http://dx.doi.org/10.1128
/JVI.79.3.1409-1416.2005.
21. Ryu MS, Jung EH, Cho KO, Kang SY. 2012. Expression of porcine
REFERENCES sapovirus VP1 gene and VP1 specific monoclonal antibody production.
1. Clarke IN, Lambden PR. 1997. The molecular biology of caliciviruses. J Hybridoma (Larchmt) 31:155–162. http://dx.doi.org/10.1089/hyb.2011
Gen Virol 78(Part 2):291–301. .0112.
2. Farkas T, Sestak K, Wei C, Jiang X. 2008. Characterization of a rhesus 22. Chen R, Neill JD, Noel JS, Hutson AM, Glass RI, Estes MK, Prasad BV.
monkey calicivirus representing a new genus of Caliciviridae. J Virol 82: 2004. Inter- and intragenus structural variations in caliciviruses and their
5408 –5416. http://dx.doi.org/10.1128/JVI.00070-08. functional implications. J Virol 78:6469 – 6479. http://dx.doi.org/10.1128
3. King AMQ, Adams MJ, Carstens EB, Lefkowitz EJ. 2012. Virus taxon- /JVI.78.12.6469-6479.2004.
omy. Classification and nomenclature of viruses. Ninth report of the In- 23. Wang Q, Zhang Z, Saif LJ. 2012. Stability of and attachment to lettuce
ternational Committee on Taxonomy of Viruses. Elsevier Academic Press, by a culturable porcine sapovirus surrogate for human caliciviruses.
San Diego, CA. Appl Environ Microbiol 78:3932–3940. http://dx.doi.org/10.1128
4. Oka T, Wang Q, Katayama K, Saif LJ. 2015. Comprehensive review of /AEM.06600-11.
human sapoviruses. Clin Microbiol Rev 28:32–53. http://dx.doi.org/10 24. Chang KO, Sosnovtsev SV, Belliot G, Kim Y, Saif LJ, Green KY. 2004.
.1128/CMR.00011-14. Bile acids are essential for porcine enteric calicivirus replication in associ-
5. Pang XL, Honma S, Nakata S, Vesikari T. 2000. Human caliciviruses in ation with down-regulation of signal transducer and activator of tran-

February 2016 Volume 90 Number 3 Journal of Virology jvi.asm.org 1357


Lu et al.

scription 1. Proc Natl Acad Sci U S A 101:8733– 8738. http://dx.doi.org/10 cells and macrophages. PLoS Biol 2:e432. http://dx.doi.org/10.1371
.1073/pnas.0401126101. /journal.pbio.0020432.
25. Guo M, Chang KO, Hardy ME, Zhang Q, Parwani AV, Saif LJ. 1999. 41. Bailey D, Thackray LB, Goodfellow IG. 2008. A single amino acid
Molecular characterization of a porcine enteric calicivirus genetically re- substitution in the murine norovirus capsid protein is sufficient for
lated to Sapporo-like human caliciviruses. J Virol 73:9625–9631. attenuation in vivo. J Virol 82:7725–7728. http://dx.doi.org/10.1128
26. de Chaumont F, Dallongeville S, Chenouard N, Herve N, Pop S, /JVI.00237-08.
Provoost T, Meas-Yedid V, Pankajakshan P, Lecomte T, Le Montagner 42. Prasad BV, Matson DO, Smith AW. 1994. Three-dimensional structure
Y, Lagache T, Dufour A, Olivo-Marin JC. 2012. Icy: an open bioimage of calicivirus. J Mol Biol 240:256 –264. http://dx.doi.org/10.1006/jmbi
informatics platform for extended reproducible research. Nat Methods .1994.1439.
9:690 – 696. http://dx.doi.org/10.1038/nmeth.2075. 43. Prasad BV, Hardy ME, Dokland T, Bella J, Rossmann MG, Estes MK.
27. Baker D, Sali A. 2001. Protein structure prediction and structural genom- 1999. X-ray crystallographic structure of the Norwalk virus capsid. Science
ics. Science 294:93–96. http://dx.doi.org/10.1126/science.1065659. 286:287–290. http://dx.doi.org/10.1126/science.286.5438.287.
28. Case DA, Darden TA, Cheatham TEI, Simmerling CL, Wang J, Duke 44. Hansman GS, Taylor DW, McLellan JS, Smith TJ, Georgiev I, Tame
RE, Luo R, Crowley M, Walker RC, Zhang W, Merz KM, Wang B, JR, Park SY, Yamazaki M, Gondaira F, Miki M, Katayama K, Murata
Hayik S, Roitberg A, Seabra G, Kolossváry I, Wong KF, Paesani F, K, Kwong PD. 2012. Structural basis for broad detection of genogroup
Vanicek J, Wu X, Brozell SR, Steinbrecher T, Gohlke H, Yang L, Tan C, II noroviruses by a monoclonal antibody that binds to a site occluded
Mongan J, Hornak V, Cui G, Mathews DH, Seetin MG, Sagui C, Babin

Downloaded from http://jvi.asm.org/ on February 18, 2021 by guest


in the viral particle. J Virol 86:3635–3646. http://dx.doi.org/10.1128
V, Kollman PA. 2008. AMBER 10. University of California, San Fran- /JVI.06868-11.
cisco, CA. 45. Katpally U, Voss NR, Cavazza T, Taube S, Rubin JR, Young VL,
29. Gerber PR, Muller K. 1995. MAB, a generally applicable molecular force Stuckey J, Ward VK, Virgin HW, IV, Wobus CE, Smith TJ. 2010.
field for structure modelling in medicinal chemistry. J Comput Aided Mol High-resolution cryo-electron microscopy structures of murine norovi-
Des 9:251–268. http://dx.doi.org/10.1007/BF00124456. rus 1 and rabbit hemorrhagic disease virus reveal marked flexibility in the
30. Summa CM, Levitt M. 2007. Near-native structure refinement using in receptor binding domains. J Virol 84:5836 –5841. http://dx.doi.org/10
vacuo energy minimization. Proc Natl Acad Sci U S A 104:3177–3182. .1128/JVI.00314-10.
http://dx.doi.org/10.1073/pnas.0611593104. 46. Katpally U, Wobus CE, Dryden K, Virgin HW, IV, Smith TJ. 2008.
31. Eisenberg D, Luthy R, Bowie JU. 1997. VERIFY3D: assessment of protein Structure of antibody-neutralized murine norovirus and unexpected dif-
models with three-dimensional profiles. Methods Enzymol 277:396 – 404.
ferences from viruslike particles. J Virol 82:2079 –2088. http://dx.doi.org
http://dx.doi.org/10.1016/S0076-6879(97)77022-8.
/10.1128/JVI.02200-07.
32. Meyer RC, Bohl EH, Kohler EM. 1964. Procurement and maintenance of
47. Chen R, Neill JD, Estes MK, Prasad BV. 2006. X-ray structure of a native
germ-free Seine for microbiological investigations. Appl Microbiol 12:
calicivirus: structural insights into antigenic diversity and host specificity.
295–300.
Proc Natl Acad Sci U S A 103:8048 – 8053. http://dx.doi.org/10.1073/pnas
33. Guo M, Qian Y, Chang KO, Saif LJ. 2001. Expression and self-assembly
.0600421103.
in baculovirus of porcine enteric calicivirus capsids into virus-like parti-
cles and their use in an enzyme-linked immunosorbent assay for antibody 48. Yu G, Zhang D, Guo F, Tan M, Jiang X, Jiang W. 2013. Cryo-EM
detection in swine. J Clin Microbiol 39:1487–1493. http://dx.doi.org/10 structure of a novel calicivirus, Tulane virus. PLoS One 8:e59817. http://dx
.1128/JCM.39.4.1487-1493.2001. .doi.org/10.1371/journal.pone.0059817.
34. Reed LJ, Muench H. 1938. A simple method of estimating fifty per cent 49. Hu Z, Tian X, Zhai Y, Xu W, Zheng D, Sun F. 2010. Cryo-electron
endpoints. Am J Epidemiol 27:493– 497. microscopy reconstructions of two types of wild rabbit hemorrhagic dis-
35. Shivanna V, Kim Y, Chang KO. 2014. The crucial role of bile acids in the ease viruses characterized the structural features of Lagovirus. Protein Cell
entry of porcine enteric calicivirus. Virology 456 – 457:268 –278. http://dx 1:48 –58. http://dx.doi.org/10.1007/s13238-010-0007-0.
.doi.org/10.1016/j.virol.2014.04.002. 50. Ossiboff RJ, Zhou Y, Lightfoot PJ, Prasad BV, Parker JS. 2010. Con-
36. Yunus MA, Chung LM, Chaudhry Y, Bailey D, Goodfellow I. 2010. formational changes in the capsid of a calicivirus upon interaction with its
Development of an optimized RNA-based murine norovirus reverse ge- functional receptor. J Virol 84:5550 –5564. http://dx.doi.org/10.1128/JVI
netics system. J Virol Methods 169:112–118. http://dx.doi.org/10.1016/j .02371-09.
.jviromet.2010.07.006. 51. Bertolotti-Ciarlet A, White LJ, Chen R, Prasad BV, Estes MK. 2002.
37. Chaudhry Y, Nayak A, Bordeleau ME, Tanaka J, Pelletier J, Belsham GJ, Structural requirements for the assembly of Norwalk virus-like particles. J
Roberts LO, Goodfellow IG. 2006. Caliciviruses differ in their functional Virol 76:4044 – 4055. http://dx.doi.org/10.1128/JVI.76.8.4044-4055.2002.
requirements for eIF4F components. J Biol Chem 281:25315–25325. http: 52. Bu W, Mamedova A, Tan M, Xia M, Jiang X, Hegde RS. 2008. Structural
//dx.doi.org/10.1074/jbc.M602230200. basis for the receptor binding specificity of Norwalk virus. J Virol 82:
38. Hosmillo M, Chaudhry Y, Kim DS, Goodfellow I, Cho KO. 2014. 5340 –5347. http://dx.doi.org/10.1128/JVI.00135-08.
Sapovirus translation requires an interaction between VPg and the cap 53. Kim DS, Hosmillo M, Alfajaro MM, Kim JY, Park JG, Son KY, Ryu EH,
binding protein eIF4E. J Virol 88:12213–12221. http://dx.doi.org/10.1128 Sorgeloos F, Kwon HJ, Park SJ, Lee WS, Cho D, Kwon J, Choi JS, Kang
/JVI.01650-14. MI, Goodfellow I, Cho KO. 2014. Both alpha2,3- and alpha2,6-linked
39. Hinnebusch AG. 2014. The scanning mechanism of eukaryotic transla- sialic acids on O-linked glycoproteins act as functional receptors for por-
tion initiation. Annu Rev Biochem 83:779 – 812. http://dx.doi.org/10.1146 cine sapovirus. PLoS Pathog 10:e1004172. http://dx.doi.org/10.1371
/annurev-biochem-060713-035802. /journal.ppat.1004172.
40. Wobus CE, Karst SM, Thackray LB, Chang KO, Sosnovtsev SV, 54. Flynn WT, Saif LJ, Moorhead PD. 1988. Pathogenesis of porcine enteric
Belliot G, Krug A, Mackenzie JM, Green KY, Virgin HW. 2004. calicivirus-like virus in four-day-old gnotobiotic pigs. Am J Vet Res 49:
Replication of norovirus in cell culture reveals a tropism for dendritic 819 – 825.

1358 jvi.asm.org Journal of Virology February 2016 Volume 90 Number 3

You might also like