Enhancing Expression of The Classical Swine Fever Virus Glycoprotein E2 in Yeast and Its Application To A Blocking Elisa

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Journal of Biotechnology 174 (2014) 1–6

Contents lists available at ScienceDirect

Journal of Biotechnology
journal homepage: www.elsevier.com/locate/jbiotec

Enhancing expression of the classical swine fever virus glycoprotein


E2 in yeast and its application to a blocking ELISA
Chih-Yuan Cheng a,1 , Ching-Wei Wu a,1 , Guang-Jan Lin a , Wei-Cheng Lee b ,
Maw-Sheng Chien b,∗∗ , Chienjin Huang a,∗
a
Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo Kuang Road,
Taichung 40227, Taiwan, ROC
b
Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 40227,
Taiwan, ROC

a r t i c l e i n f o a b s t r a c t

Article history: Classical swine fever virus (CSFV) infection is a severe swine disease, often causing large economic losses.
Received 15 November 2013 A Pichia pastoris yeast-expressed CSFV glycoprotein E2 (yE2) has been shown to induce a protective
Received in revised form immune response against the virus. To improve the expression level of yE2, the first codon of E2 gene,
20 December 2013
Arg (CGG), which is the least used in P. pastoris, was optimized to the most favorite codon AGA. The
Accepted 8 January 2014
yield of E2 protein was remarkably increased in the codon optimized strain (N342). Three truncated E2
Available online 24 January 2014
subunits encoding the N-terminal 330 (N330), 301 (N301), and 190 (N190) residues, respectively, were
also constructed. The immunogenicity of each recombinant E2 subunits was confirmed by immunization
Keywords:
Classical swine fever virus
of pigs, and all immunized groups demonstrated high neutralizing antibody titers after boost immu-
Subunit marker vaccine nization, which lasted for a long period of time. In addition, a monoclonal antibody (MAb), 1B6, specific
Codon optimization to yE2, was generated and shown to recognize CSFV-infected cells. A panel of swine sera were tested
Monoclonal antibody by peroxidase-conjugated MAb 1B6-based blocking enzyme-linked immunosorbent assay (ELISA) using
Blocking ELISA N330 as coated antigen, and the assay demonstrated high sensitivity and specificity. The recombinant
yE2 subunits may provide potential subunit vaccine candidates and useful diagnostic reagents for CSFV
with easy manipulation and low cost.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction can also protect pigs against CSF in different ways (Bouma et al.,
2000; Dong and Chen, 2007; Hulst et al., 1993). Currently, commer-
Classical swine fever virus (CSFV) is a small enveloped virus cially available CSF E2 subunit vaccines have been developed using
belonging to the genus Pestivirus of the Flaviviridae family the baculovirus expression system (Bouma et al., 1999; de Smit
(Lindenbach et al., 2007). CSFV infection results in a highly conta- et al., 2001; Moormann et al., 2000), but the required insect cell cul-
gious and severe disease characterized by fever (CSF), often causing ture is costly and the purification procedures are time consuming.
large economic losses in the pig industry worldwide. The CSFV Recently, we constructed a recombinant E2 protein using the yeast
genome is a positive sense single-stranded RNA comprising a single, Pichia pastoris secreted expression system. This yeast-expressed E2
long, open-reading-frame (ORF) that encodes a polyprotein. This (yE2) could induce a protective immune response against lethal
polyprotein is co- and post-translationlly processed by cellular and challenge infection (Lin et al., 2012, 2009). The advantages of this
viral protease to yield all final viral proteins. Structural proteins eukaryotic expression system include simple manipulation, easy
include a nucleocapsid protein C and three envelope glycoproteins, purification, and less cost (Cereghino and Cregg, 2000). However,
Erns , E1, and E2 (Rümenapf et al., 1993). several genetic and physiological factors determine the productiv-
The glycoprotein E2 represents the most significant target for ity of a recombinant system (Hohenblum et al., 2003). Synonymous
inducing the production of neutralizing antibodies in the host, and codon usage bias differences, one major factor among others, sig-
nificantly affects heterologous gene expression (Sinclair and Choy,
2002; Su et al., 2007). To improve the expression yield of yE2, the
first codon CGG (Arg) of the E2 gene, which is the least used in P.
∗ Corresponding author. Tel.: +886 4 22853906; fax: +886 4 22851741.
∗∗ Co-corresponding author. Tel.: +886 4 22851940.
pastoris, was optimized to the most favorite codon AGA, and sev-
E-mail addresses: mschien@dragon.nchu.edu.tw (M.-S. Chien),
eral truncated mutants were also constructed and evaluated for
cjhuang@dragon.nchu.edu.tw, jin827.huang@gmail.com (C. Huang). their immunogenicities in pigs. The expressed E2 recombinant pro-
1
These authors contributed equally to the article. tein was further utilized as an antigen to generate a monoclonal

0168-1656/$ – see front matter © 2014 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.jbiotec.2014.01.007
2 C.-Y. Cheng et al. / Journal of Biotechnology 174 (2014) 1–6

Table 1 PVDF transfer membrane (NEN) using semi-dry transfer cell (Bio-
Sequences of oligonucleotides used for cloning the defined coding region of the CSFV
Rad) according to the manufacturer’s instruction. The membrane
E2 protein.
was then treated sequentially with blocking solution (PBS contain-
Oligonucleotide Sequence in 5 –3 directiona Restriction site ing 5% non-fat skim milk), with 1000-fold dilution of monoclonal
yE2N1F TTATCGATTAGACTAGCCTGCAAGG ClaI antibody (MAb) WH303 specific to CSFV E2 (Veterinary Laboratory
yE2N190F GTATCGATTAGACTAGCCTGCAAG ClaI Agency), and 5000-fold dilution of anti-mouse IgG goat antibody
yE2N342R CGCTCTAGAAATTCTGCGAAGTAAT XbaI conjugated to horseradish peroxidase (Zymed). Finally, the mem-
yE2N330R CGCTCTAGATCCAGGTCAAACCAGT XbaI
brane was soaked in a chromogen/substrate solution (TMB single
yE2N301R CGCTCTAGAGTTTTTGCGTAATTGA XbaI
yE2N190R CGGAATTCTTTCACACATGTCCAG EcoRI solution, Zymed) for color development.
a
The sequences recognized by the restriction enzyme are boxed and the first
codon of the E2 gene was optimized to AGA and is underlined. 2.4. Immunization of pigs

Fifteen 6-week-old specific-pathogen-free (SPF) piglets were


antibody specific to CSFV E2 and establish a blocking ELISA for randomly allotted to four E2 subunit immunized groups and a
detecting swine antibodies against E2. control group (n = 3 for each group). Each piglet was immunized
intramuscularly into the neck region with one dose of vaccine twice
2. Materials and methods at 3-week intervals. Each dose of vaccine contained 300 ␮g total
secreted proteins of each expressed E2 variants, including the N342,
2.1. Construction of recombinant expression plasmids N330, N301, and N190 groups or normal saline (the control group)
in a 1:1 water-in-oil emulsion with the adjuvant ISA563 (SEPPIC).
Plasmid pGAPZ␣C/E2dc containing the CSFV E2 gene encod-
ing the amino acid (a.a.) residues 1–342 without the C-terminal 2.5. Serological examination
transmembrane region of vaccine LPC strain was constructed
previously (Lin et al., 2009). The defined coding region corre- Serum blood samples were collected from each pig before vacci-
sponding to the a.a. residues 1–342, 1–330, 1–301, and 1–190 nation and at biweekly intervals after vaccination. Serum samples
of E2 was amplified by polymerase chain reaction (PCR) using were tested for CSFV E2-specific antibodies with a commercial
the specific primer pair yE2N1F/yE2N342R, yE2N1F/yE2N330R, E2 blocking ELISA (HerdChek CSFV Antibody Test Kit, IDEXX Lab-
yE2N1F/yE2N301R, and yE2N190F/yE2N190R, with the first argi- oratories) according to the manufacturer’s instruction. The CSFV
nine codon changed to AGA, respectively (Table 1). The PCR reaction neutralizing antibody titers in serum were determined by the
was carried out as described previously (Lin et al., 2009). The serum neutralization test as described previously (Lin et al., 2009).
amplified E2 gene fragment was gel-purified and then treated with Replicates of two-fold dilutions of heat-inactivated (30 min, 56 ◦ C)
appropriate restriction enzymes for cloning into the yeast expres- serum samples (50 ␮l) were mixed with 200 TCID50 CSFV LPC strain
sion vector pGAPZ␣C (Invitrogen) to construct the expression (50 ␮l) in a microtitration plate, and incubated at 37 ◦ C for 1 h.
plasmid pGAPZ␣C/E2N342, pGAPZ␣C/E2N330, pGAPZ␣C/E2N301, After adding 1 × 104 PK-15 cells (100 ␮l) and incubating at 37 ◦ C
and pGAPZ␣C/E2N190, respectively. The recombinant E2 proteins for 72 h, the cells were subjected to immunofluorescence staining
were expressed as fusion to a C-terminal peptide containing the with the MAb WH303. Neutralizing antibody titers were presented
myc epitope and a polyhistidine tag. The accuracy of the ORF of the as the reciprocal of the highest dilution that completely inhibited
E2 coding sequences was confirmed by DNA sequencing. replication of the virus.

2.2. Expression of E2 protein in Pichia pastoris 2.6. Preparation of the monoclonal antibody against E2

Recombinant expression plasmids were transformed into P. pas- Five 6-week-old female BALB/c mice were immunized subcuta-
toris SMD1168 competent cells using a Pichia EasyCompTM Kit neously with 100 ␮l of a mixture of 50 ␮g purified N342 and an
(Invitrogen) according to the manufacturer’s instructions. Trans- equal volume of Freund’s complete adjuvant (Sigma). A similar
formed cells were then plated onto yeast extract peptone dextrose immunization was given 2 weeks later using Freund’s incom-
(YPD; 1% yeast extract, 2% peptone, 2% glucose) agar containing plete adjuvant. After an additional 3 weeks, the immunized mice
100 ␮g/ml Zeocin (Invitrogen) and incubated at 30 ◦ C for 2–3 days intraperitoneally received a final booster with 200 ␮l of 30 ␮g puri-
until single colonies were formed. A single colony of recombinant fied N342 without adjuvant. The fusion was carried out 5 days later
yeast was inoculated in 5 ml YPD medium and incubated at 30 ◦ C according to the methods described previously (Huang et al., 1994).
in a shaking incubator (250 rpm) overnight. Then 0.1 ml of the The hybridoma secreting specific antibody to E2 was selected
overnight culture was transferred to 50 ml fresh YPD medium in by ELISA and further characterized by indirect immunofluores-
a 250 ml baffled flask and continuously incubated for 4 days. The cence (IIF) analysis with CSFV-infected PK-15 cells (ATCC-CCL33)
supernatants were clarified by centrifugation (20 min, 12,000 × g, as described previously (Wu et al., 2013). The subclass of the MAb
4 ◦ C) and the secreted protein was concentrated by ultrafiltration was determined by Mouse MonoAb-ID kit (Zymed).
using Centricon YM-10 or 30 (Millipore) filter devices, followed
by dialysis against phosphate-buffered saline (PBS). The protein 2.7. Preparation of horseradish peroxidase (HRP) linked MAb
concentration was determined by a Bradford protein assay kit against E2 conjugate
(Bio-Rad).
The MAb was purified by the Protein A/G affinity column (Pierce)
2.3. Western blotting analysis followed by conjugating with HRP using SureLINKTM Activated HRP
(KPL) according to the manufacturer’s instruction.
Yeast-secreted proteins were treated in equal volumes of the
unreduced 2× sample buffer (125 mM Tris–Cl [pH 6.8], 20% glyc- 2.8. Blocking ELISA for detecting antibody against E2
erol, 4% SDS, 0.25% bromophenol blue). Proteins were separated
by 12% sodium dodecyl sulfate-polyacrylamide gel electrophore- ELISA plates (Corning) were coated at 4 ◦ C overnight with 50 ␮l
sis (SDS-PAGE) and transferred by electroblotting onto PolyScreen of 1 ␮g/ml purified N330 in coating buffer (carbonate buffer, pH
C.-Y. Cheng et al. / Journal of Biotechnology 174 (2014) 1–6 3

Fig. 1. Schematic diagram of the expressed coding regions of E2 recombinant subunits. Bars represent expressed coding sequences. The amino acid residue numbers at both
termini and the first codon for arginine are indicated. Potential glycosylation sites are indicated by an asterisk, and the epitope recognized by the MAb WH303 is indicated
by a gray box. The conformational antigenic domains B ± C and D ± A characterized by van Rijn et al. (1994) are also showed.

9.6). The plate was then thoroughly washed with PBS contain- the yeast P. pastoris using recombinant expression plasmids, includ-
ing 0.05% Tween-20 (PBST) and blocked with PBS containing 3% ing pGAPZ␣C/E2N342, pGAPZ␣C/E2N330, pGAPZ␣C/E2N301, and
bovine serum albumin (BSA) 37 ◦ C for 1 h. After washing, each well pGAPZ␣C/E2N190 for expressing different E2 recombinant pro-
received 50 ␮l of 2-fold dilution of tested swine serum in dilu- teins. The yeast ␣-factor signal sequence efficiently channels into
tion buffer (PBS containing 1% BSA) and was incubated at 37 ◦ C for a secretion pathway. Schematic diagrams of N342, N330, N301,
1 h. Subsequently, the plate was thoroughly washed with PBST and and N190 are shown in Fig. 1. Expressed recombinant E2 pro-
each well received 50 ␮l of 500-fold dilution of HRP-MAb anti-E2 teins were analyzed by Western blotting analysis with MAb,
conjugate in dilution buffer at 37 ◦ C for 45 min. Finally, the plate WH303, specific to E2 (Fig. 2A) N342 and N330 could form
was washed with PBST three times and PBS twice. Then, 100 ␮l of homodimers about 110 kDa in size while N301 and N190 pre-
freshly prepared chromogen/substrate solution (ABTS single solu- dominantly exhibited monomers about 52 and 43 kDa in size,
tion, Zymed) was added into each well and the plate was incubated respectively. The expression level of N342 was further compared
at room temperature for 15 min. The optical density (OD) of each with yE2 at 24 h-interval for 4 days. Remarkable increases of
well was read at 405 nm using a microplate reader (MRXII, Dynex). yield in N342 were revealed during the entire expression course
Each sample was analyzed in duplicate, and the mean OD value (Fig. 2B).
of each tested sample (ODTEST ) and that of the negative control
(ODNEG ) were calculated. The inhibition percentage of each sample 3.2. Immunization of pigs
was calculated according to the following formula:
ODNEG − ODTEST To evaluate the immunogenicity of various E2 recombinant
Blocking % = × 100 proteins, three 6-week-old SPF piglets of each group were immu-
ODNEG
nized intramuscularly with 300 ␮g of each total secreted protein.
All recombinant E2-immunized pigs showed strong antibody
3. Results responses and seroconverted to CSFV-E2-specific antibody after
booster vaccination, as determined by a commercial E2-blocking
3.1. Expression of CSFV E2 protein variants in Pichia pastoris ELISA test (40%), while no antibody was detected in the control
pigs (Fig. 3A) All of the N342, N330, N301, and N190 groups could
Various coding regions of the CSFV E2 gene with the first argi- mount anamnestic responses following booster vaccination with
nine codon optimized to AGA were introduced into the genome of average neutralizing antibody titers of 1:1789, 1:1448, 1:1708, and

Fig. 2. Characterization of recombinant E2 proteins by Western blotting analysis. The culture supernatant of each recombinant E2 proteins including N190, N301, N330 and
N342 were harvested at 96 h (A). For comparison of the expression levels between the yE2 and codon optimized strain (N342), the culture supernatants of N342 and yE2
were harvested at 24 h, 48 h, 72 h, and 96 h, respectively (B). The expressed E2 proteins were separated by 12% SDS-PAGE in the absence of ␤-mercaptoethanol followed by
Western blotting analysis with the monoclonal antibody (WH303) specific to E2. The expected E2 protein is indicated by an arrow.
4 C.-Y. Cheng et al. / Journal of Biotechnology 174 (2014) 1–6

Fig. 5. Reactivities of swine sera to yE2 (N330) in the blocking ELISA (A) and the
Fig. 3. Time course of ELISA antibody development (A) and neutralizing antibody comparison to a commercial ELISA kit (B).
(B) of variant E2 recombinant proteins immunized pigs after vaccination. All of the
pigs received a booster immunization (↓) at 3 weeks post-immunization.
3.4. Development of a blocking ELISA for detecting antibody
against E2
1:2572, respectively that lasted for at least 12 weeks with titers
above the protective titer of 1:32 (Fig. 3B). The optimal concentration of antigen coated on the plate was
determined by checkerboard titration. The best positive/negative
(P/N) ratio was obtained when the N330 was diluted at a concen-
3.3. Preparation of MAb specific to the CSFV E2 tration of 1 ␮g/ml, the tested swine serum was diluted at 1:2, and
the HRP-anti-E2 (1B6) conjugate was diluted at 1:500 (data not
A stable hybridoma secreting antibody reacting specifically with shown). A panel of swine serum samples collected from the Ani-
CSFV-infected cells in IIF (Fig. 4A) and yE2 subunits in Western blot- mal Disease Diagnostic Center, National Chung Hsing University
ting analysis (Fig. 4B) was selected and cloned. This MAb, 1B6, was (NCHU) were determined by the commercial E2 blocking ELISA kit
determined to be the IgG1 subclass type, and also demonstrated (IDEXX). Fifty-two sera were tested by N330 and MAb 1B6-based
neutralizing antibody activity to all three different genotypes blocking ELISA and the results expressed as blocking % are shown in
of CSFV infection, including strain LPC, 96TD and 94.4, in the Fig. 5A The blocking percentages of 17 negative sera ranged from
neutralization assay (data not shown). The MAb was purified 16.8% to 33.1% with an average of 23.9%, while 35 positive sera
and then coupled with HRP to generate the HRP-anti-E2 (1B6) ranged from 43.3% to 69.9% with an average of 60%. When the block-
conjugate. ing percentage cut-off value was set at 40%, 17 negative sera were

Fig. 4. Characterization of the specificity of the MAb 1B6 by IIF (A) and Western blotting analysis (B). (A) Normal (PK-15) or CSFV-infected PK-15 (CSFV/PK-15) cells were
subjected to immunofluorescence staining with the MAb 1B6. (B) Yeast-expressed E2 subunit N190 (lane 1) and N330 (lane 2), and the wild-type yeast-secreted protein
(lane 3) were separated by 12% SDS-PAGE followed by Western blotting analysis with 1B6.
C.-Y. Cheng et al. / Journal of Biotechnology 174 (2014) 1–6 5

determined as negative and resulted in a specificity of 100%, and status of animals that have been vaccinated or CSFV infected (König
all the positive sera were positive with the corresponding sensi- et al., 1995; Weiland et al., 1992). Currently, commercially available
tivities of 100%. When the cut-off value was set more restricted diagnostic ELISA kits mostly use baculovirus-expressed recombi-
at 45% or 50%, the sensitivities were slightly decreased to 94.3% nant E2 or Erns as ELISA antigens (Moormann et al., 2000; Windisch
(33/35) and 91.4% (32/35), respectively. A correlation coefficient et al., 1996). However, the cost of insect cell culture and time-
between our established blocking ELISA (NCHU) and the commer- consumption to purify the expressed product are major concerns. A
cial ELISA kit (IDEXX) was determined using the other 40 selected more economical yeast-expressed Erns (yErns )-based indirect sand-
serum samples with even distribution of blocking percentages wich ELISA was developed in a previous study and demonstrates
determined by IDEXX ELISA. A regression line was plotted between high sensitivity and specificity (Wu et al., 2011). In the present
the blocking percentages of the corresponding serum sample using study, a MAb specific to E2 (1B6)- and yE2 subunit (N330)-based
Microsoft Excel. High correlation (R = 0.9250) between two ELISAs blocking ELISA was established. This assay demonstrates a high sen-
was demonstrated as shown in Fig. 5B. sitivity and specificity, and high correlation with a commercial CSFV
E2 blocking ELISA kit. It may offer a useful and inexpensive method
for routine diagnosis of swine antibody to E2.
4. Discussion
Conflict of interest
The yeast P. pastoris has been used as an efficient system for
recombinant protein production (Cereghino and Cregg, 2000). Sev- None declared.
eral factors such as gene dosage, promoter activity, codon usage of
the expressed gene, and protein processing and folding may influ- Acknowledgement
ence the expression levels of heterologous proteins (Hohenblum
et al., 2004; Outchkourov et al., 2002). Low yield of human endo- This work was supported in part by grant NSC101-2313-B-005-
statin in P. pastoris has been attributed to translational inefficiencies 012-MY3 from the National Science Council, Taiwan, Republic of
and synonymous codon usage bias which is the more probable China.
cause of the translational barriers (Sinclair and Choy, 2002; Su et al.,
2007). According to Zhang et al. (1991), there are eight low-usage References
codons including AGG (Arg), CGA (Arg), CGG (Arg), CGC (Arg), CCG
(Pro), CUC (Leu), and GCG (Ala) in yeast. Among those synonymous Bouma, A., de Smit, A.J., de Kluijver, E.P., Terpstra, C., Moormann, R.J.M., 1999. Efficacy
and stability of a subunit vaccine based on glycoprotein E2 of classical swine
codons for Arg, CGG is the least-used (2%), while AGA is the most
fever virus. Vet. Microbiol. 66, 101–114.
favored (54%). Few codons of the CSFV E2 gene are low-usage in Bouma, A., De Smit, A.J., De Jong, M.C.M., De Kluijver, E.P., Moormann, R.J.M., 2000.
yeast, though it was successfully expressed in our previous study Determination of the onset of the herd-immunity induced by the E2 sub-unit
(Lin et al., 2009). To improve the yield of yE2, the first codon (CGG) vaccine against classical swine fever virus. Vaccine 18, 1374–1381.
Cereghino, J.L., Cregg, J.M., 2000. Heterologous protein expression in the methy-
of E2 gene was optimized to AGA, and shorter coding regions were lotrophic yeast Pichia pastoris. FEMS Microbiol. Rev. 24, 45–66.
also constructed. The expression level of yE2 increased remark- de Smit, A.J., Bouma, A., de Kluijver, E.P., Terpstra, C., Moormann, R.J.M., 2001. Dura-
ably after the first codon changed (Fig. 2B), while all the truncated tion of the protection of an E2 subunit marker vaccine against classical swine
fever after a single vaccination. Vet. Microbiol. 78, 307–317.
subunits demonstrated higher yields especially for N330 (Fig. 2A). Dong, X.N., Chen, Y.H., 2007. Marker vaccine strategies and candidate CSFV marker
Successful translation passed the first codon of the heterologous vaccines. Vaccine 25, 205–230.
gene with high frequency and certainly circumvented the bottle- Hohenblum, H., Borth, N., Mattanovich, D., 2003. Assessing viability and cell-
associated product of recombinant protein producing Pichia pastoris with flow
neck of the P. pastoris expression system. More rare-usage codons cytometry. J. Biotechnol. 102, 281–290.
of E2 have now been altered and the expression levels are currently Hohenblum, H., Gasser, B., Maurer, M., Borth, N., Mattanovich, D., 2004. Effects of
under study. gene dosage, promoters, and substrates on unfolded protein stress of recombi-
nant Pichia pastoris. Biotechnol. Bioeng. 85, 367–375.
The yeast-expressed E2 (yE2) is capable of inducing a complete
Huang, C., Chien, M.S., Landolt, M., Winton, J., 1994. Characterizatrion of the infec-
protective immune response and preventing horizontal transmis- tious hematopoietic necrosis virus glycoprotein using neutralizing monoclonal
sion of CSFV, and appears to be a potential subunit marker vaccine antibodies. Dis. Aquat. Org. 18, 29–35.
Hulst, M.M., Westra, D.F., Wensvoort, G., Moormann, R.J.M., 1993. Glycoprotein E1
(Lin et al., 2012). The immunogenicity of each truncated yE2 recom-
of hog cholera virus expressed in insect cells protects swine from hog cholera.
binant proteins was evaluated by the immunization of pigs. All J. Virol. 67, 5435–5442.
of the recombinant yE2 subunit-immunized pigs, including N342, König, M., Lengsfeld, T., Pauly, T., Stark, R., Thiel, H.J., 1995. Classical swine fever virus
N330, N301 and N190 groups, could mount anamnestic responses independent induction of protective immunity by two structural glycoproteins.
J. Virol. 69, 6479–6486.
following booster immunization with high neutralizing antibody Lindenbach, B.D., Thiel, H.J., Rice, C.M., 2007. Flaviviridae: the viruses and their repli-
titers of an average above 1:1448 (Fig. 3B), that reveals much higher cation. In: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology. , 5th ed. Lippincott
efficacy than yE2-induced averaged neutralizing titer of 1:132 in Williams & Wilkins, Philadelphia, pp. 1101–1152.
Lin, G.J., Liu, T.Y., Tseng, Y.Y., Chen, Z.W., You, C.C., Hsuan, S.L., Chien, M.S., Huang,
the previous study (Lin et al., 2009). A quantitative assay for CSFV C., 2009. Yeast expressed classical swine fever virus glycoprotein E2 induces a
E2 glycoprotein is currently under construction. In addition, N301 protective immune response. Vet. Microbiol. 139, 369–374.
and N190 are present predominantly monomer forms (Fig. 2A), Lin, G.J., Deng, M.C., Chen, Z.W., Liu, T.Y., Wu, C.W., Cheng, C.Y., Chien, M.S., Huang,
C., 2012. Yeast expressed classical swine fever E2 subunit vaccine candidate
suggesting C-terminal truncation hampers formation of E2 homod- provides complete protection against lethal challenge infection and prevents
imer, and the region of a.a. residues 302–330 is indispensible for horizontal virus transmission. Vaccine 30, 2336–2341.
dimerization. Moreover, the N-terminal 190 residues, which con- Moormann, R.J.M., Bouma, A., Kramps, J.A., Terpstra, C., De Smit, H.J., 2000. Devel-
opment of a classical swine fever subunit marker vaccine and companion
sist of major conformational antigenic domains, B ± C and D ± A,
diagnostic test. Vet. Microbiol. 73, 209–219.
(van Rijn et al., 1994) retain correct immunogenicity that is capa- Outchkourov, N.S., Stiekema, W.J., Jongsma, M.A., 2002. Optimization of the expres-
ble of inducing a high neutralizing antibody response. Therefore, sion of equistatin in Pichia pastoris. Protein Expr. Purif. 1, 18–24.
Rümenapf, T., Unger, G., Strauss, J.H., Thiel, H.J., 1993. Processing of the envelope
those recombinant yE2 subunits represent a potential subunit E2
glycoproteins of pestiviruses. J. Virol. 67, 3288–3294.
marker vaccine candidate with advantages of easy manipulation Sinclair, G., Choy, F.Y., 2002. Synonymous codon usage bias and the expression of
and low cost. human glucocerebrosidase in the methylotrophic yeast, Pichia pastoris. Protein
CSF is an economically important swine disease that has Expr. Purif. 1, 96–105.
Su, C., Duan, X., Wang, X., Wang, C., Cao, R., Zhou, B., Chen, P., 2007. Heterologous
received widespread attention. Detecting antibodies to Erns and expression of FMDV immunodominant epitopes and HSP70 in P. pastoris and the
E2 in swine serum provides a direct measure of the immune subsequent immune response in mice. Vet. Microbiol. 124, 256–263.
6 C.-Y. Cheng et al. / Journal of Biotechnology 174 (2014) 1–6

van Rijn, P.A., Miedema, G.K.W., Wensvoort, G., van Gennip, H.G.P., Moormann, Wu, C.W., Chien, M.S., Liu, T.Y., Lin, G.J., Lee, W.C., Huang, C., 2011. Characterization
R.J.M., 1994. Antigenic structure of envelope glycoprotein E1 of hog cholera of the monoclonal antibody against classical swine fever virus glycoprotein Erns
virus. J. Virol. 68, 3934–3942. and its application to an indirect sandwich ELISA. Appl. Microbiol. Biotechnol.
Weiland, E., Ahl, R., Stark, R., Weiland, F., Thiel, H.J., 1992. A second envelop glyco- 92, 815–821.
protein mediates neutralization of a Pestivirus, hog cholera virus. J. Virol. 66, Wu, C.W., Chien, M.S., Huang, C., 2013. Characterization of the swine U6 promoter
3677–3682. for short hairpin RNA expression and its application to inhibition of virus repli-
Windisch, J.M., Schneider, R., Stark, R., Weiland, E., Meyers, G., Thiel, H.J., 1996. RNase cation. J. Biotechnol. 168, 78–84.
of classical swine fever virus: biochemical characterization and inhibition by Zhang, S., Zubay, G., Goldman, E., 1991. Low-usage codons in Escherichia coli, yeast,
virus-neutralizing monoclonal antibodies. J. Virol. 70, 352–358. fruit fly and primates. Gene 105, 61–72.

You might also like