The proteins NPRO and ERNS are unique for the genus Pestivirus, but only NPRO has been demonstrated to be nonessential for in vitro growth. While this was also speculated for ERNS, it has always been previously shown that pestivirus replicons with deletions of the structural proteins ERNS, E1, or E2 did not produce any infectious progeny virus in susceptible host cells. Here, we demonstrated for the first time that BuPV ERNS is dispensable for the generation of infectious virus particles but still important for efficient passaging. The ERNS-defective BuPV particles showed clearly limited growth in cell culture but were capable of several rounds of infection, expression of foreign genes, and highly efficient trans-complementation to rescue virus replicon particles (VRPs). The noncytopathic characteristics and the absence of preexisting immunity to BuPV in human populations and livestock also provide a significant benefit for a possible use, e.g., as a vector vaccine platform.
KEYWORDS: Bungowannah virus, ERNS, pestivirus, foreign gene expression, replicon, viral vector, virus replicon particles
ABSTRACT
Autonomously replicating subgenomic Bungowannah virus (BuPV) RNAs (BuPV replicons) with deletions of the genome regions encoding the structural proteins C, ERNS, E1, and E2 were constructed on the basis of an infectious cDNA clone of BuPV. Nanoluciferase (Nluc) insertion was used to compare the replication efficiencies of all constructs after electroporation of in vitro-transcribed RNA from the different clones. Deletion of C, E1, E2, or the complete structural protein genome region (C-ERNS-E1-E2) prevented the production of infectious progeny virus, whereas deletion of ERNS still allowed the generation of infectious particles. However, those ΔERNS viral particles were defective in virus assembly and/or egress and could not be further propagated for more than three additional passages in porcine SK-6 cells. These “defective-in-third-cycle” BuPV ΔERNS mutants were subsequently used to express the classical swine fever virus envelope protein E2, the N-terminal domain of the Schmallenberg virus Gc protein, and the receptor binding domain of the Middle East respiratory syndrome coronavirus spike protein. The constructs could be efficiently complemented and further passaged in SK-6 cells constitutively expressing the BuPV ERNS protein. Importantly, BuPVs are able to infect a wide variety of target cell lines, allowing expression in a very wide host spectrum. Therefore, we suggest that packaged BuPV ΔERNS replicon particles have potential as broad-spectrum viral vectors.
IMPORTANCE The proteins NPRO and ERNS are unique for the genus Pestivirus, but only NPRO has been demonstrated to be nonessential for in vitro growth. While this was also speculated for ERNS, it has always been previously shown that pestivirus replicons with deletions of the structural proteins ERNS, E1, or E2 did not produce any infectious progeny virus in susceptible host cells. Here, we demonstrated for the first time that BuPV ERNS is dispensable for the generation of infectious virus particles but still important for efficient passaging. The ERNS-defective BuPV particles showed clearly limited growth in cell culture but were capable of several rounds of infection, expression of foreign genes, and highly efficient trans-complementation to rescue virus replicon particles (VRPs). The noncytopathic characteristics and the absence of preexisting immunity to BuPV in human populations and livestock also provide a significant benefit for a possible use, e.g., as a vector vaccine platform.
INTRODUCTION
Bungowannah virus (BuPV), a member of the species Pestivirus F of the genus Pestivirus within the family Flaviviridae (1), is associated with the porcine myocarditis syndrome (PMC) in young pigs (2, 3). The first outbreak of BuPV was detected in Australia in New South Wales in 2003, but until now, the virus had not spread to any other region in Australia or to other countries worldwide (4, 5). Bungowannah virus is genetically and antigenically very divergent from the four classical pestivirus species bovine viral diarrhea virus 1 (BVDV-1; only recently reclassified as Pestivirus A), bovine viral diarrhea virus 2 (BVDV-2; Pestivirus B), border disease virus (BDV; Pestivirus D), and classical swine fever virus (CSFV; Pestivirus C). The atypical pestivirus BuPV is mostly related to the porcine lateral shaking inducing neurodegenerative agent (LINDA) virus, which was identified in Austria in 2015 (6), and the most recently in toothed whales detected Phocoena pestivirus (PhoPeV) (7). Moreover, the in vitro cell tropism of BuPV differs markedly from that of other pestiviruses. With the exception of CSFV, which can only infect cells of swine origin, pestiviruses are able to infect a variety of cell cultures derived from cloven-hoofed animals (8). However, the broadest in vitro cell tropism was described for BuPV. Besides bovine, ovine, and porcine cells, cell lines of human, mouse, bat, and monkey origin are also susceptible to BuPV (9).
Pestivirus particles are small, enveloped virions. The genome is a positive-sense single-stranded RNA (ssRNA) of about 11.5 to 13 kb that consists of a single open reading frame (ORF) flanked by nontranslated regions (NTRs) at both genome ends (10–15). The 5′ and 3′ NTRs are highly structured and required for RNA replication and translation (16–18). The 5′ NTR contains an internal ribosomal entry site (IRES) for cap-independent translation initiation (19, 20). The 3′ NTR exhibits highly variable as well as highly conserved regions (17).
The ORF encodes a single polyprotein which is co- and posttranslationally cleaved by both cellular and viral proteases into the mature structural proteins capsid (C), ERNS, E1, and E2 and the nonstructural proteins NPRO, p7, NS2-NS3 (NS2, NS3), NS4A, NS4B, NS5A, and NS5B (10, 12, 21, 22). The nonstructural proteins NS3, NS4A, NS4B, NS5A, and NS5B are essential components of the pestiviral replication machinery (23–25). The positively charged capsid protein forms the viral capsid and is responsible for the packaging of the viral RNA, probably by a histone-like protein-RNA interaction (26). The viral envelope is composed of the three glycoproteins ERNS, E1, and E2 embedded in a lipid membrane of host cellular origin. In virus particles and pestivirus-infected cells, disulfide-linked E2 and ERNS homodimers and E1-E2 heterodimers can be detected (21, 23, 27–29). The envelope protein ERNS is like the nonstructural protein NPRO unique to pestiviruses. NPRO is described as an interferon antagonist in host cells and is not essential for virus replication in cell culture (30, 31). However, while the function of ERNS in the pestiviral life cycle is not fully understood, it has been shown that ERNS is involved in virus entry by binding to glucosaminoglycans (32), that the uptake of ERNS bound to cells is powered by clathrin-dependent endocytosis, and that ERNS also acts as interferon antagonist (33). Unlike the other pestiviral glycoproteins E1 and E2, ERNS lacks a typical membrane anchor but exhibits a long amphipathic α-helix (34, 35). A very special feature of ERNS is its intrinsic RNase activity, which is an important virulence factor (36–39).
Autonomously replicating subgenomic pestivirus RNAs (replicons) were originally identified as defective interfering RNAs that could be packaged by a helper virus. Reverse genetics systems were used to generate first self-replicating RNAs analogous to the defective interfering RNAs in vitro (25, 40, 41). Pestivirus replicons became important tools to study molecular mechanisms of pestivirus replication, assembly, and egress (42, 43). So, it became clear that neither the structural proteins C, ERNS, E1, and E2 nor the nonstructural proteins p7 and NS2 are required for RNA replication but are indispensable for the generation of infectious virus particles (25, 42, 44). Interestingly, viable CSFV mutants with deletions within the capsid protein encoding the genomic region and a second-site mutation in the C-terminal region of NS3 could be rescued (45). Replicons expressing heterologous genes, e.g., the green fluorescent protein (GFP) or a luciferase reporter gene, have also been used for antiviral drug screening and small interfering RNA (siRNA) testing (46–50). Packaging of replicons, generated by trans-complementation in cell lines providing the deleted genes, has been shown to have a high potential for the development of safe and efficacious marker vaccines (43, 51, 52). Furthermore, replicons expressing homologous genes have been successfully used as nontransmissible vaccine vectors (53–55).
In the present study, we constructed BuPV replicons with deletions within the regions encoding the structural proteins C, ERNS, E1, and E2 and compared their replication efficiencies. All generated mutants were able to replicate autonomously and, with exception of the mutants with ERNS deletions, did not produce any detectable infectious virus progeny. Here, we could for the first time show that the deletion of BuPV ERNS does not prevent the generation of infectious virus particles. However, the virus particles could not be propagated in porcine SK-6 cells efficiently, and after several passages, infectious virus was no longer detectable. Replicons with ERNS deletions were able to express foreign genes, as shown exemplarily for CSFV envelope protein E2, an immunogenic domain of Schmallenberg virus (SBV) Gc, and the immunogenic receptor binding domain (RBD) of the Middle East respiratory syndrome coronavirus (MERS-CoV) spike protein, and could be trans-complemented in a newly established cell line constitutively expressing the BuPV ERNS protein.
RESULTS
Construction and characterization of BuPV replicons.
The BuPV replicons were constructed analogously to efficiently replicating BVDV and CSFV replicons (43, 51, 56) by using the BuPV plasmid pA/BV as the template. The different replicons exhibited either a deletion of 123 bases within the capsid protein (BVΔC, amino acids [aa] 208 to 248), a deletion of 456 bases within E1 (BVΔE1, aa 506 to 657), a deletion of 468 bases within ERNS (BVΔERNS_156, aa 328 to 483), a complete deletion of E2 (BVΔE2, aa 695 to 1071), or a complete deletion of the structural protein-encoding region (BVΔC-E2; aa 208 to 1071) (Fig. 1A). At 72 h posttransfection (hpt) of in vitro-transcribed RNA of the full-length BuPV plasmid pA/BV or the replicon constructs, the expression of nonstructural protein NS3 could be detected by immunofluorescence (IF) staining, indicating that all in vitro-synthesized RNAs were able to replicate autonomously after electroporation into cells. No apparent differences in fluorescence intensity were observed between the different replicon constructs after electroporation. Expression of the deleted structural proteins was not detected (BVΔE2, negative in E2 staining; BVΔERNS, negative in ERNS staining; BVΔC-E2, negative in E2 and ERNS staining) (Fig. 1B). Because BuPV-C- and BuPV-E1-specific antibodies are not available, the absence of the capsid protein C in BVΔC and of the E1 protein in BVΔE1 RNA-electroporated cells could not be confirmed by staining or Western blotting but could be shown by reverse transcription PCR (RT-PCR) analysis of the RNAs. In order to examine whether recombinant virus was generated, fresh SK-6 cells were inoculated with filtered supernatants of the electroporated cells sampled at 72 hpt. To exclude a carryover event from electroporated cells or cell fragments, the supernatants were centrifuged and filtered after harvesting to remove residual cells from the supernatants. As expected, infectious virus progeny could not be recovered from supernatants of cells electroporated with RNA of the constructs BVΔC, BVΔE1, BVΔE2, and BVΔC-E2.
FIG 1.
(A) Schematic representation of the infectious BuPV full-length cDNA backbone clone pA/BV and the replicon constructs generated in this study. Light gray boxes represent the nonstructural proteins, and dark gray boxes represent the structural proteins. Deletions are represented by horizontal dotted lines. Lines at the left and right ends represent NTRs. (B) IF analysis of SK-6 cells electroporated with in vitro-transcribed RNA of the full-length clone pA/BV and replicon constructs BVΔC, BVΔERNS_156, BVΔE2, BVΔE1, and BVΔC-E2 or inoculated with transfection supernatants (first passage) using a pan-pestivirus NS3-specific antibody (anti-NS3), BuPV-specific monoclonal E2 antibody (anti-E2), and BuPV ERNS monoclonal antibody (anti-ERNS) at 72 hpt. Mock- and pA/BV-electroporated cells were used as controls. Bar, 100 μm.
Surprisingly, in cells inoculated with supernatants of BVΔERNS_156 RNA-electroporated cells, a large number of BuPV protein-expressing foci could be observed, indicating that infectious recombinant virions (BVΔERNS_156) could be rescued, and this could not be observed for any other constructed replicons. To further exclude any carryover event, the passaging of BVΔERNS_156 RNA-electroporated cells onto fresh SK-6 cells was repeated, but with an extensive washing step after 2 h. Again, BuPV protein-expressing foci could be observed (Fig. 2). However, the rescued virions showed a clear growth defect and could not efficiently be further propagated.
FIG 2.
BVΔERNS_156 RNA-electroporated cells were centrifuged, filtered, and passaged onto fresh SK-6 cells; the cells were then incubated for 72 h (A) or the medium was removed after 2 h, the cells were washed with phosphate-buffered saline, fresh DMEM was added, and then the cells were incubated for 72 h (B). After the incubation period, the cells were fixed and stained using an anti-NS3-specific MAb.
Comparison of the replication efficiencies of the BuPV replicons.
In order to compare the replication efficiencies of the BuPV replicons, corresponding constructs expressing the nanoluciferase reporter gene between NPRO and C (pA/BV_Nluc, BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE2_Nluc, BVΔE1_Nluc, BVΔC-E2_Nluc) were generated. An insertion of the first 21 nucleotides (nt) of the capsid gene upstream and a short GSG linker (9 nt) as well as the teschovirus 2Apro sequence (59 nt) downstream of the Nluc coding region facilitates the correct cleavage of Nluc from the viral polyprotein by the autoproteolytic activity of NPRO and teschovirus 2Apro (Fig. 3A). A replication-incompetent construct (pA/BV_GAA_Nluc) was generated by mutation of the GDD RNA polymerase motif in NS5B to GAA (Fig. 3A) and served as a negative control in the Nluc assays.
FIG 3.
(A) Schematic representation of cDNA constructs with an Nluc reporter gene insertion, which is indicated by hatched boxes. 7 aa capsid, 7 N-terminal amino acids of BuPV capsid protein; Teschovirus 2Apro, teschovirus 2A protease. The mutated nucleotides introduced into the NS5B gene of the RNA-dependent RNA polymerase-defective construct BV_GAA are shown in capital letters. (B) Growth characteristics of pA/BV, BV_wt, and pA/BV_Nluc in SK-6 cells. All represented viruses replicate efficiently, and comparable virus titers could be determined at the depicted hours postinfection. (C) Nluc activity in SK-6 cells electroporated with in vitro-transcribed RNA of the replicon constructs BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE1_Nluc, BVΔE2_Nluc, and BVΔC-E2_Nluc or the full-length clone BV_Nluc and the replication-incompetent cDNA clone BV_GAA_Nluc at 4, 24, 48, and 72 hpt, normalized to BV_GAA_Nluc. Three technical and biological replicates were measured for each construct; error bars represent standard deviations, and a horizontal dotted line represents the dynamic range of the luminometer.
Electroporation of SK-6 cells with in vitro-transcribed RNA from the full-length cDNA clone pA/BV_Nluc and the replicons with Nluc insertions (BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE2_Nluc, BVΔE1_Nluc, BVΔC-E2_Nluc) resulted in RNA replication and production of viral proteins comparable to that of the corresponding constructs without the Nluc gene. To determine whether the Nluc insertion had an effect on the virus replication, growth kinetic studies were performed with the wild-type BuPV (BV_wt) and the recombinant viruses pA/BV and pA/BV_Nluc in SK-6 cells. Growth kinetics studies demonstrated comparable replication efficiencies of BV_wt and the recombinant viruses pA/BV and pA/BV_Nluc, and virus titers of about 106.75 to 107.525 50% tissue culture infective dose (TCID50)/ml could be measured at 72 h postinfection (hpi) (Fig. 3B). Furthermore, Nano-Glo luciferase assays demonstrated the expression of biologically active Nluc from all replication-competent constructs with Nluc insertions.
In order to quantify the Nluc expression as a measure of replication efficiency of the different replicon constructs, supernatants of electroporated cells were removed at 4, 24, 48, and 72 hpt, and cells were assayed for Nluc activity. Figure 2C shows the values normalized to the replication-incompetent GAA mutant. In parallel, we monitored the consistent expression of NS3 in all samples by IF staining (data not shown).
Nluc activity was initially detected at 4 hpt, and at this time point, no significant differences could be shown for the electroporated replicons and the control.
At 24 hpt, maximum relative light unit (RLU) values of around 6 × 106 could be estimated for the replicons BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE2_Nluc, and BVΔC-E2_Nluc. BVΔE1_Nluc replicated on a slightly reduced level, with maximal values of 3.6 × 106 RLU (Fig. 3C). Virus production could already be detected at 24 h after electroporation of pA/BV_Nluc, and the maximum Nluc values of pA/BV_Nluc were higher at this and the following time points than for the replicons. With the exception of construct BVΔE1_Nluc, there were no significant differences in RNA replication of the deletion replicons over the investigated time (Table 1).
TABLE 1.
Significance analysis of replication efficiencies of the BuPV replicons expressing nanoluciferase and pA/BV_Nluca
| Time point and comparison | Significant? | Summaryb | Adjusted P value |
|---|---|---|---|
| 4 h | |||
| BVΔC_Nluc vs BVΔERNS_156__Nluc | No | NS | >0.9999 |
| BVΔC_Nluc vs BVΔE1_Nluc | No | NS | >0.9999 |
| BVΔC_Nluc vs BVΔE2_Nluc | No | NS | >0.9999 |
| BVΔC_Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔC_Nluc vs pA/BV_Nluc | No | NS | >0.9999 |
| BVΔERNS_156__Nluc vs B VΔE1_Nluc | No | NS | >0.9999 |
| BVΔERNS_156_Nluc vs BVΔE2_Nluc | No | NS | >0.9999 |
| BVΔERNS_156__Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔERNS_156__Nluc vs pA/BV_Nluc | No | NS | >0.9999 |
| BVΔE1_Nluc vs BVΔE2_Nluc | No | NS | >0.9999 |
| BVΔE1_Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔE1_Nluc vs pA/BV_Nluc | No | NS | >0.9999 |
| BVΔE2_Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔE2_Nluc vs pA/BV_Nluc | No | NS | >0.9999 |
| BVΔC-E2_Nluc vs pA/BV_Nluc |
No | NS | >0.9999 |
| 24 h | |||
| BVΔC_Nluc vs BVΔERNS_156__Nluc | No | NS | 0.833 |
| BVΔC_Nluc vs BVΔE1_Nluc | Yes | **** | <0.0001 |
| BVΔC_Nluc vs BVΔE2_Nluc | No | NS | 0.0904 |
| BVΔC_Nluc vs BVΔC-E2_Nluc | No | NS | 0.1087 |
| BVΔC_Nluc vs pA/BV_Nluc | No | NS | 0.1383 |
| BVΔERNS_156__Nluc vs BVΔE1_Nluc | Yes | ** | 0.0077 |
| BVΔERNS_156__Nluc vs BVΔE2_Nluc | No | NS | 0.7003 |
| BVΔERNS_156__Nluc vs BVΔC-E2_Nluc | No | NS | 0.7465 |
| BVΔERNS_156__Nluc vs pA/BV_Nluc | Yes | ** | 0.0041 |
| BVΔE1_Nluc vs BVΔE2_Nluc | No | NS | 0.3186 |
| BVΔE1_Nluc vs BVΔC-E2_Nluc | No | NS | 0.2785 |
| BVΔE1_Nluc vs BV_Nluc | Yes | **** | <0.0001 |
| BVΔE2_Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔE2_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔC-E2_Nluc vs pA/BV_Nluc |
Yes | **** | <0.0001 |
| 48 h | |||
| BVΔC_Nluc vs BVΔERNS_156__Nluc | No | NS | 0.9853 |
| BVΔC_Nluc vs BVΔE1_Nluc | Yes | *** | 0.0004 |
| BVΔC_Nluc vs BVΔE2_Nluc | No | NS | 0.1805 |
| BVΔC_Nluc vs BVΔC-E2_Nluc | No | NS | 0.2317 |
| BVΔC_Nluc vs pA/BV_Nluc | Yes | *** | 0.0003 |
| BVΔERNS_156__Nluc vs BVΔE1_Nluc | Yes | ** | 0.0054 |
| BVΔERNS_156__Nluc vs BVΔE2_Nluc | No | NS | 0.5467 |
| BVΔERNS_156__Nluc vs BVΔC-E2_Nluc | No | NS | 0.6283 |
| BVΔERNS_156__Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔE1_Nluc vs BVΔE2_Nluc | No | NS | 0.3958 |
| BVΔE1_Nluc vs BVΔC-E2_Nluc | No | NS | 0.3242 |
| BVΔE1_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔE2_Nluc vs BVΔC-E2_Nluc | No | NS | >0.9999 |
| BVΔE2_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔC-E2_Nluc vs pA/BV_Nluc |
Yes | **** | <0.0001 |
| 72 h | |||
| BVΔC_Nluc vs BVΔERNS_156__Nluc | No | NS | 0.7169 |
| BVΔC_Nluc vs BVΔE1_Nluc | Yes | * | 0.0186 |
| BVΔC_Nluc vs BVΔE2_Nluc | No | NS | 0.6407 |
| BVΔC_Nluc vs BVΔC-E2_Nluc | No | NS | 0.7918 |
| BVΔC_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔERNS_156__Nluc vs BVΔE1_Nluc | Yes | **** | <0.0001 |
| BVΔERNS_156__Nluc vs BVΔE2_Nluc | Yes | * | 0.0406 |
| BVΔERNS_156__Nluc vs BVΔC-E2_Nluc | No | NS | 0.0792 |
| BVΔERNS_156__Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔE1_Nluc vs BVΔE2_Nluc | No | NS | 0.5450 |
| BVΔE1_Nluc vs BVΔC-E2_Nluc | No | NS | 0.3856 |
| BVΔE1_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔE2_Nluc vs BVΔC-E2_Nluc | No | NS | 0.9999 |
| BVΔE2_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
| BVΔC-E2_Nluc vs pA/BV_Nluc | Yes | **** | <0.0001 |
One-way ANOVA followed by Tukey`s multiple-comparison test was performed using GraphPad Prism version 8.0.1 for Windows (GraphPad Software, San Diego, CA, USA).
*, <0.05; **, <0.01; ***, <0.001; ****, <0.0001; NS, not significant.
Characterization of BuPV replicons with ERNS deletions.
We next investigated whether deletions within functional regions of BuPV ERNS or the size of the ERNS deletion had an impact on the generation of infectious progeny virus. BuPV replicons with deletions of 55 to 112 amino acids within ERNS (BVΔERNS_55, BVΔERNS_88, and BVΔERNS_112) or a complete ERNS deletion of 222 amino acids (BVΔERNS) were constructed (Fig. 4A). In the construct BVΔERNS_55, the deletion of 55 aa between aa 84 and aa 139 in the middle of ERNS does not affect any of the functional domains. In the construct BVΔERNS_88, the deletion from aa 51 to aa 139 covers the second RNase domain with its catalytic center H80 (BuPV-ERNS aa numbering [57–59]). In deletion mutant BVΔERNS_112, in which aa 51 to aa 163 have been deleted, the first cluster of basic amino acids (140KKGH143), which has been shown to be important for double-stranded RNA (dsRNA) binding and regulation of interferon (IFN) induction in pestivirus-infected cells (32, 58), is also deleted. In replicon BVΔERNS_156, amino acids between positions 51 and 207 were deleted, while 51 N-terminal amino acids, including the first RNase domain with its active center H31 and the 15 C-terminal amino acids harboring a part of the amphipathic helix with a second cluster of basic amino acids (209KIENTKKK216) responsible for membrane binding (34, 35) were retained.
FIG 4.
(A) Schematic representation of the BuPV genome and the generated replicons with deletion of various functional domains within the ERNS. Horizontal dotted lines represent the deletions; RNaDI, RNase domain I; RNaDII, RNase domain II; dsRNaD, double-stranded RNA binding domain; horizontal black lines represent conserved cysteines. Numbers above vertical lines on the bar indicate amino acid positions. (B) IF analysis of SK-6 cells electroporated with the respective replicons (BVΔERNS_156, BVΔERNS_112, BVΔERNS_88, BVΔERNS_55, BVΔERNS). Supernatants were harvested, and the cells were immunostained at 72 hpt using the pan-pestivirus NS3-specific MAb or the BuPV ERNS-specific MAb. Supernatants of the transfected cells were used for inoculation of fresh SK-6 cells. The supernatants contained infectious recombinant viruses that could be passaged for up to two further rounds, as demonstrated by IF staining with NS3 (1st passage, 2nd passage, 3rd passage). Bar, 100 μm. (C) Neutralization test of BVΔERNS replicons with BuPV-positive serum. Supernatants from electroporated SK-6 cells were neutralized, and IF staining was used for analysis with BuPV E2-specific MAb. (D) IF analysis of the generated SK-6 cell line expressing the BuPV ERNS protein (SK-6_BV-ERNS). IF staining with the BuPV ERNS-specific MAb led to positive IF signals, while IF staining using the NS3-specific MAb and the BuPV E2-specific MAb remained negative. (E and F) Passage history of trans-complemented VRPs with ERNS deletions in SK-6_BuPV-ERNS cells (E) and alternating in SK-6 cells and SK-6_BuPV-ERNS cells (F). SK-6_BuPV-ERNS cells were electroporated with the respective replicons. At 72 hpi, supernatants were harvested (TF, transfection supernatant) and the generated complemented viruses (BVΔERNS_156_comp, BVΔERNS_112_comp, BVΔERNS_88_comp, BVΔERNS_55_comp, BVΔERNS_comp) were serially passaged using complementing SK-6_BV-ERNS cells with increasing virus titers. In contrast, alternating passages in complementing (boxed passage numbers) and noncomplementing cells always resulted in a dramatic drop in virus titers, demonstrating the limited replication of the packaged replicons in noncomplementing cells and the absence of recombination events. Endpoint titers are given in log10 TCID50/ml. After electroporation of SK-6 cells with BVΔERNS_156, supernatants were harvested and passaged using SK-6 or SK-6-BV-ERNS cells. Viral titers (H) and Cq values (G) were determined after every passage.
To investigate the influence of the deletions on RNA replication, the constructs were transfected into SK-6 cells by electroporation, and expression of NS3, a marker for autonomous pestivirus RNA replication, could be detected. Furthermore, ERNS staining of the cells was negative. The intensity of NS3 immunostaining and the number of NS3-positive cells differed only marginally among the replicons (Fig. 4B).
Transfection of all different replicons with an ERNS deletion resulted in the generation of infectious progeny virus, and a large number of small foci of infected cells could be observed. However, all ERNS-deleted recombinant viruses showed a significant growth defect and could only be passaged up to two further rounds in SK-6 cells (Fig. 4B).
The BVDV-1 plasmid NCP7ΔERNS (51) and a BVDV-2 plasmid, pA/BVDV-2ΔERNS (I. Reimann, unpublished data), were used as controls; however, infectious virus could never be rescued (data not shown).
For further characterization of the generated particles, a neutralization test was performed using serum of an animal inoculated experimentally with the wild-type virus. All BVΔERNS virus particles could be neutralized, indicating that these are actually infectious and not artifacts of the electroporation process (Fig. 4C). Transfection supernatants incubated with a negative serum, which was used as a control, were not neutralized.
Establishment of a BuPV ERNS-expressing SK-6 cell line and trans-complementation studies.
For a further characterization of the different ERNS deletion mutants, we established an SK-6 cell line (SK-6_BV-ERNS) that constitutively expresses BuPV ERNS (Fig. 4D). SK-6 cells were electroporated with plasmid pCAGGS_BuPV-ERNS, which contains the complete region encoding the BuPV structural protein ERNS under the control of the chicken beta-actin (CAG) promoter, and a puromycin resistance gene. IF staining of puromycin-resistant cell clones using the BuPV ERNS-specific monoclonal antibody (MAb) revealed BuPV ERNS expression in more than 90% of the cells. IF staining of the cells using pan-pestivirus NS3-specific and BuPV E2-specific MAbs served as negative controls (Fig. 4D). After continuous passaging of the SK-6_BV-ERNS cell lines for 6 months in the presence of puromycin, the portion of ERNS-expressing cells remained stable (data not shown).
To investigate whether BuPV ERNS can be complemented in trans, in vitro-transcribed RNA of the constructs BVΔERNS_55, BVΔERNS_88, BVΔERNS_112, BVΔERNS_156, and BVΔERNS was electroporated into the SK-6_BV-ERNS cell line. At 72 hpt, supernatants of the electroporated cells were collected, and secreted trans-complemented virion replicon particles (VRPs) (BVΔERNS_55_comp, BVΔERNS_88_comp, BVΔERNS_112_comp, BVΔERNS_156_comp, and BVΔERNS_comp) with virus titers of 103.25 to 104.0 TCID50/ml could be detected in all transfection supernatants (Fig. 4E). Subsequent serial passages in the SK-6_BV-ERNS helper cell line resulted in virus titers of about 107 TCID50/ml after nine passages for all mutants, independent of the size of the ERNS deletion.
In contrast, in alternating passaging experiments using the parental SK-6 cells and SK-6_BV-ERNS cells, the titers of the trans-complemented viruses decreased clearly in all cases after two passages in SK-6 cells but increased immediately after one passage in complementing SK-6_BV-ERNS cells, before dropping down again to virus titers just below 101 TCID50/ml (Fig. 4F). Finally, after three consecutive passages in SK-6 cells, again no progeny virus could be detected.
These results could be confirmed by electroporation of BVΔERNS _156 in SK-6 cells and passaging the supernatants of electroporated cells in both SK-6 and SK-6_BV-ERNS cells. Supernatants of the first to fifth passages in SK-6 cells were again passaged once in SK-6_BV-ERNS cells, and virus titers as well as quantification cycle (Cq) values of BuPV RNA were determined (Fig. 4G and H). In passage one, virus titers of 100.75 TCID50/ml in SK-6 cells and 106.5 TCID50/ml in SK-6_BV-ERNS cells could be determined (Fig. 4H). In this experiment, in passage two, no virus could be detected in SK-6 cells by IF staining; however, in SK-6_BV-ERNS cells, trans-complemented virus was detectable and high virus titers of 105.75 TCID50/ml could be obtained. Only after three passages in SK-6 cells, no infectious virus could be rescued after passaging these supernatants in SK-6_BV-ERNS cells (= passage P4) (Fig. 4H). The real-time RT-PCR results confirmed these results, as the viral genome of BVΔERNS_156 was detectable in SK-6 cells up to passage two, with Cq values of 29.8 in SK-6 cells and 19.93 in SK-6_BV-ERNS cells, and in passage four, no viral RNA was detectable in either cell line (Fig. 4G).
From these results and from sequence analyses of the generated VRPs (passage 9 in SK-6_BV-ERNS cells, alternating passages 7 and 8), we conclude that neither genetic changes such as adaptive mutations nor recombination with the ERNS gene of the cell line occurred and that the VRPs are stable in SK-6_BV-ERNS cells.
The growth defect of BVΔERNS is not related to replication efficiency but is a consequence of virion assembly and/or egress.
In order to investigate whether the growth defect of the ERNS-defective VRPs is caused by reduced RNA replication efficiency, we analyzed the replication capacity of BVΔERNS_156 replicon RNA and the full-length BuPV RNA after electroporation into SK-6 cells earlier. The results show that the full-length construct replicates only at a slightly higher level at different time points (Fig. 3C).
In order to have a closer look at these processes, SK-6 cells and the helper cell line SK-6_BV-ERNS were infected with recombinant BuPV (rBuPV) or packaged BVΔERNS_156_comp VRPs, as an example for the ERNS-deleted mutants, at a multiplicity of infection (MOI) of 0.1 TCID50/ml. After virus adsorption and several washing steps, to ensure that the starting material did not affect subsequent investigations, supernatants and cells of the same well were separated and RNA was isolated from both fractions. BVΔE1 was used as a negative control. The RNA amounts were quantified 2 and 48 hpi by real-time RT-PCR using BuPV-specific primers and an external standard. At 2 hpi, very low copy numbers and no apparent differences were estimated between the replicon and full-length RNA levels in SK-6 and SK-6_BV-ERNS cells and supernatants. This indicates for both cell lines that the infection with VRPs as well as rBuPV were under equal conditions and the same amounts of input RNA (Fig. 5).
FIG 5.
Quantitative RT-PCR analysis of SK-6 cells and SK-6_BV-ERNS cells infected with BVΔERNS_156_comp and full-length BuPV. RNA copy numbers in SK-6 and SK-6_BV-ERNS cells infected with the packaged replicon BVΔERNS_156_comp and rBuPV at an MOI of 0.1 were estimated per well at 2 and 48 hpi. SK-6 and SK-6_BV-ERNS cells inoculated with supernatants of SK-6_BV-ERNS cells electroporated with BVΔE1 were used as a negative control. Experiments were done in biological duplicates. Significance analysis was performed by ANOVA using GraphPad Prism software. Asterisks indicate significant differences between groups.
As expected, no BuPV-specific RNA could be detected in cells inoculated with supernatants of BVΔE1 electroporated SK-6_BV-ERNS cells. However, compared to the 2-h RNA values, at 48 hpi, a significant increase in full-length BuPV RNA could be detected for both cell lines in the cells as well as in the supernatants (SK-6_BV-ERNS cells, P = 0.0001, supernatant, P = 0.0202; SK-6 cells, full-length BuPV, P = 0.043, supernatant, P = 0.0028). For BVΔERNS_156, significantly increased RNA copy numbers could be likewise detected in the complementing cell line after 48 hpi compared to the 2-h values (cells, P = 0.0155; supernatant, P = 0.0187). In the SK-6 cells, however, no statistically significant difference was observed (Table 2). Comparable increases in RNA copy numbers by about 3 log steps were measured in the cell fraction for BVΔERNS_156 in both cell lines, whereas the copy number in the supernatant did not change significantly 48 hpi in the noncomplementing cell line. In contrast, RNA copy numbers of around 107 in cells and 109 in the supernatant could be detected in the complementing cell line infected with the BVΔERNS_156 replicon particles or rBuPV at 48 hpi. Significantly higher RNA copy numbers of the full-length BuPV RNA than of BVΔERNS_156 replicon RNA could be observed in SK-6 cells, with P values of 0.0468 in cells and 0.0028 in the supernatant at that time point. This indicates a defect of BVΔERNS in the assembly or release of RNA-carrying virions.
TABLE 2.
Significance analysis of RNA copy numbers from pA/BV-, BVΔERNS_comp-, and BVΔE1-infected SK-6 or SK-6-BV-ERNS cellsa
| Cell type or supernatant and comparison | Significant? | Summaryb | Adjusted P value |
|---|---|---|---|
| SK-6-BV-ERNS supernatant | |||
| 2h BVΔERNS vs 2h pA/BV | No | NS | >0.9999 |
| 2h BVΔERNS vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h BVΔERNS | Yes | * | 0.0187 |
| 2h BVΔERNS vs 48h pA/BV | Yes | * | 0.0202 |
| 2h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 48h BVΔERNS | Yes | * | 0.0187 |
| 2h pA/BV vs 48h pA/BV | Yes | * | 0.0202 |
| 2h pA/BV vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h BVΔERNS | Yes | * | 0.0187 |
| 2h BVΔE1 vs 48h pA/BV | Yes | * | 0.0202 |
| 2h BVΔE1 vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h BVΔERNS vs 48h pA/BV | No | NS | 0.9611 |
| 48h BVΔERNS vs 48h BVΔE1 | Yes | * | 0.0187 |
| 48h pA/BV vs 48h BVΔE1 |
Yes | * | 0.0202 |
| SK-6-BV-ERNS cells | |||
| 2h BVΔERNS vs 2h pA/BV | No | NS | >0.9999 |
| 2h BVΔERNS vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h BVΔERNS | Yes | * | 0.0155 |
| 2h BVΔERNS vs 48h pA/BV | Yes | *** | 0.0001 |
| 2h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 48h BVΔERNS | Yes | * | 0.0155 |
| 2h pA/BV vs 48h pA/BV | Yes | *** | 0.0001 |
| 2h pA/BV vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h BVΔERNS | Yes | * | 0.0155 |
| 2h BVΔE1 vs 48h pA/BV | Yes | *** | 0.0001 |
| 2h BVΔE1 vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h BVΔERNS vs 48h pA/BV | No | NS | 0.0594 |
| 48h BVΔERNS vs 48h BVΔE1 | Yes | * | 0.0155 |
| 48h pA/BV vs 48h BVΔE1 |
Yes | *** | 0.0001 |
| SK-6 supernatant | |||
| 2h BVΔERNS vs 2h pA/BV | No | NS | >0.9999 |
| 2h BVΔERNS vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h pA/BV | Yes | ** | 0.0028 |
| 2h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h pA/BV vs 48h pA/BV | Yes | ** | 0.0028 |
| 2h pA/BV vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h pA/BV | Yes | ** | 0.0065 |
| 2h BVΔE1 vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h BVΔERNS vs 48h pA/BV | Yes | ** | 0.0028 |
| 48h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h pA/BV vs 48h BVΔE1 |
Yes | ** | 0.0028 |
| SK-6 cells | |||
| 2h BVΔERNS vs 2h pA/BV | No | NS | >0.9999 |
| 2h BVΔERNS vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h BVΔERNS vs 48h pA/BV | Yes | * | 0.043 |
| 2h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 2h BVΔE1 | No | NS | >0.9999 |
| 2h pA/BV vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h pA/BV vs 48h pA/BV | Yes | * | 0.043 |
| 2h pA/BV vs 48h BVΔE1 | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h BVΔERNS | No | NS | >0.9999 |
| 2h BVΔE1 vs 48h pA/BV | Yes | * | 0.043 |
| 2h BVΔE1 vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h BVΔERNS vs 48h pA/BV | Yes | * | 0.0468 |
| 48h BVΔERNS vs 48h BVΔE1 | No | NS | >0.9999 |
| 48h pA/BV vs 48h BVΔE1 | Yes | * | 0.043 |
One-way ANOVA followed by Tukey`s multiple-comparison test was performed using GraphPad Prism version 8.0.1 for Windows (GraphPad Software, San Diego, CA, USA).
*, <0.05; **, <0.01; ***, <0.001; NS, not significant.
BuPV replicons with ERNS deletions are suitable for the stable expression of heterologous viral genes and selected immunogenic domains.
To investigate whether the generated BVΔERNS replicons are a suitable system for expressing foreign genes, the CSFV E2 protein, the N-terminal domain of the SBV glycoprotein Gc (GcN), and the receptor-binding domain (RBD) of the MERS-CoV spike protein served as model immunogens (60–62). Based on BVΔERNS_156, we generated replicons expressing GcN by insertion of two GcN domains connected by a 16-aa-long linker sequence (BVΔERNS_2GcN) or the RBD of MERS-CoV spike protein (BVΔERNS_RBD). For the correct processing of both the BuPV C and E1 proteins by cellular signalases, in both replicons the foreign genes were flanked by 51 N-terminal amino acids and 15 C-terminal amino acids of BuPV ERNS. The CSFV E2-expressing replicon BVΔERNS_CSFV_E2 was constructed on the basis of BVΔERNS with a complete ERNS deletion and was inserted between C and E1 of the replicon (Fig. 6A). Since both BuPV ERNS and CSFV E2 are pestiviral glycoproteins with a C-terminal region, which allows cleavage from the polyprotein by cellular signalases, the complete substitution of CSFV E2 for BuPV ERNS should not negatively affect correct processing of BVΔERNS_CSFV-E2.
FIG 6.
Analysis of chimeric replicons and chimeric VRPs. (A) Schematic depiction of the constructed chimeric replicons with deletions within the ERNS coding region. Structural proteins are shown in dark gray, nonstructural proteins are shown in light gray, and foreign genes are shown in hatched boxes. The blowups show the insertion and the surrounding genomic region. For construction of BVΔERNS_2GcN, a double construct of the N-terminal SBV Gc region connected by a linker was inserted into the ERNS_156 deletion. The same deletion mutant was used for the insertion of the receptor binding domain of MERS-CoV (BVΔERNS_RBD). CSFV E2 was inserted into the complete ERNS deletion mutant BVΔERNS, resulting in BVΔERNS-CSFV-E2. (B) IF analysis after electroporation of in vitro-transcribed RNA into SK-6 cells at 72 hpt by using the NS3-specific MAb WB112, a MERS-CoV RBD-specific MAb, the SBV GcN-specific MAb 5E1_A3, and the CSFV E2-specific MAb Bio275. Nontransfected cells were used a negative control. SK-6 cells were inoculated with collected supernatants and investigated by IF staining at 72 hpi using pan-pestivirus NS3-specific MAb WB112. (C) Trans-complementation of the indicated constructs by RNA transfection of SK-6_BV-ERNS cells. The chimeric VRPs were passaged 6 times in SK-6_BV-ERNS cells 72 hpi and analyzed by IF staining of the cells using the pan-pestivirus NS3-specific MAb (WB112) and the insert-specific MAbs 5E1_A3 (SBV-GcN), MERS-CoV-RBD, and Bio275 (CSFV-E2). Nontransfected cells were used as a negative control. (D) SK-6 cells were infected with complemented replicons (BVΔERNS_2GcN_comp, BVΔERNS_RBD_comp, BVΔERNS_CSFV_E2_comp, BVΔERNS_156_comp), and expression of the foreign genes was analyzed at 48 hpi by Western blotting using MAbs 5E1_A3 (SBV GcN), Bio275 (CSFV E2), and PAb (MERS-CoV S1) (spike protein S1). Bar, 100 μm.
The successful expression of the different foreign genes could be demonstrated by immunostaining of SK-6 cells electroporated with in vitro-transcribed RNA of the replicon constructs BVΔERNS_2GcN, BVΔERNS_RBD, and BVΔERNS_CSFV_E2, respectively, at 72 hpt, using SBV GcN-specific, MERS-CoV RBD-specific, or CSFV E2-specific MAbs. In contrast, full-length BuPV electroporated cells, which have been used as a control, reacted negatively in IF staining with those MAbs.
After electroporation, all replicon RNA-transfected cells were positive by NS3 staining, while the mock-transfected cells reacted negatively and pA/BV, which was used as positive control, reacted correctly positive. Transfection supernatants were used for inoculation of SK-6 cells, and at 72 hpi, small foci of IF-positive cells could be detected. However, the number of IF-positive foci was lower than that observed after the first passage of BVΔERNS and BVΔERNS_156 virions without insertions of foreign genes, and no progeny virus could be further passaged in SK-6 cells (Fig. 6B).
However, the expression of foreign genes had no negative effect on the packaging of the replicon RNA in SK-6_BV-ERNS helper cells. After electroporation of the constructs BVΔERNS_2GcN, BVΔERNS_RBD, and BVΔERNS_CSFV_E2 into complementing SK-6_BV-ERNS cells, RNA replication could be detected by NS3-specific immunostaining and expression of the SBV GcN domain, MERS-CoV RBD, and CSFV E2 was confirmed at 72 hpt (Fig. 6C). Virus titers between 103.875 and 105.625 TCID50/ml could be determined at 72 hpt (Fig. 6C). The complemented viruses could be further passaged in SK-6_BV-ERNS cells; the titers are given in Fig. 5.
Stable expression of foreign genes.
In order to investigate the stability of protein expression after six passages of the complemented viruses in SK-6_BV-ERNS cells, virus titers were also determined by IF staining using the respective foreign-gene-specific MAbs. For all complemented viruses, the titers determined by NS3 immunostaining as well as by SBV GcN, MERS-CoV RBD, and CSFV E2 immunostaining did not differ significantly and indicated a stable expression of the foreign genes (Fig. 6C). Recombination events could be excluded, since the complemented viruses could not be continuously passaged in noncomplementing SK-6 cells.
The expression of the SBV GcN domain, the receptor-binding domain of MERS-CoV S1 spike protein, and CSFV E2 protein could also be shown by Western blotting. SK-6 cells were inoculated with the complemented viruses BVΔERNS_RBD_comp, BVΔERNS_2GcN_comp, BVΔERNS_CSFV_E2_comp, and BVΔERNS_156_comp or remained noninfected (negative control). In lysates of cells infected with BVΔERNS_2GcN, the double GcN domain could be detected as a fusion protein with an estimated size of approximately 62 kDa, similar to the calculated size. In lysates of cells infected with BVΔERNS_RBD_comp, MERS-CoV RBD, which was also expressed as a fusion protein, could be detected at 38 kDa, slightly higher than the calculated molecular weight of 34 kDa. In lysates of cells infected with BVΔERNS_CSFV_E2_comp, expression of CSFV E2 with a calculated size of about 55 kDa could be demonstrated by Western blotting as well (Fig. 6D).
Expression of foreign genes does not alter the cell tropism of VRPs.
In order to investigate whether the chimeric BuPV VRPs were able to infect cells originating from the most relevant target species of SBV (sheep and cattle) and MERS-CoV (Bactrian camel, human), cells of these species were inoculated with the chimeric VRPs or VRPs without insertions (negative control). At 72 hpi, RNA replication and expression of GcN, RBD, and CSFV-E2 could be detected in sheep (SFT-R), human (HEK-293), Bactrian camel (TT-R), and cattle (KOP-R) cells. In addition, the RBD domain was successfully expressed in llama (LGL-2-R) and alpaca (LPK-1-R, LPL-2-R) cells (data not shown). IF analysis of the infected cells demonstrated that the broad cell tropism of BuPV also applies to the chimeric VRPs (Fig. 7A and B). In order to determine the infection rate of the cell lines with the respective VRPs and to compare the chimeric VRPs with the BuPV VRPs without foreign genes, fluorescence-activated cell sorting (FACS) analysis was performed. For this comparison, the measured values of the construct BVΔERNS were set as 100% for each of the tested cell lines, and it could be demonstrated that all replicons showed a high infection rate in SK-6 cells (>86%) and a slightly lower infectivity in SFT cells (45% to 73%), HEK cells (52% to 75%), and KOP-R cells (49% to 73%) than that of the parental clone BVΔERNS. An improved infection rate due to the insertion could therefore not be observed (Fig. 7C).
FIG 7.
Infection of cells originating from the target species of SBV, CSFV, and MERS-CoV with packaged BVΔERNS replicons. (A) Sheep cells (SFT-R), cattle cells (KOP-R), cells originating from Bactrian camel (TT-R), and (B) cells of human origin (HEK-293) were infected with packaged BVΔERNS_2GcN_comp, BVΔERNS_RBD_comp, and/or BVΔERNS_CSFV_E2_comp. Infected cells were analyzed by IF staining 72 hpi by using the NS3-specific MAb WB112, a MERS-CoV RBD-specific MAb, a CSFV E2-specific MAb, and the SBV GcN-specific MAb 5E1_A3, nontransfected cells were used as a negative control, and full-length pA/BV was used as a positive control. Bar, 100 μm. (C) To determine whether the insertion influences the infection rate of the replicons, flow cytometry analysis (FACS) was performed for BVΔERNS_CSFV_E2, BVΔERNS_2GcN, and BVΔERNS_RBD. The replicon BVΔERNS and uninfected cells were used as controls. The infection rate of BVΔERNS was defined as 100%.
DISCUSSION
Autonomously replicating RNAs are important tools to study the role of single proteins, e.g., concerning their role in the viral replication cycle. Pestivirus replicons with deletions within the structural proteins C, ERNS, E1, and E2 have therefore been constructed previously and have been used for the development of safe and efficient vaccine candidates that are defective in the second cycle against BVDV and CSFV infections.
In all cases, those pestiviral replicons with deletions in the structural protein-encoding genes were not able to form infectious virus progeny, with the exception of a CSFV construct with a nearly complete deletion of the C protein. A mutation within the NS3 protein seems to compensate for the functionality of the C protein. This construct has the ability to produce infectious viruses in vitro (45). Furthermore, replicons could be passaged on trans-complementing cell lines and packaged into viral replicon particles (VRPs). For BVDV replicons, passaging and packaging were often very inefficient or even not possible due to severe interference effects (43, 51, 63). In contrast, CSFV replicons lacking the ERNS protein could be efficiently passaged on complementing helper cells (56). However, the host range of those packaged pestivirus replicons was very restricted, mainly to the natural hosts, i.e., pigs and ruminants.
Here, for first time, we studied replicons of the atypical Bungowannah pestivirus. Since BuPV has only been found on one farm until now, a vaccine against this virus itself seems to be unnecessary, but since BuPV can infect a very broad range of cells of, e.g., mouse, monkey, bat, camelid, porcine, ruminant, and human origin—in sharp contrast to all other known pestiviruses (9)—and replicate efficiently in these cells, BuPV replicons might have the advantage as a universal expression vector. Therefore, a first BuPV replicon set with the deletion of genes encoding the structural proteins was constructed and characterized. We also tested their suitability for use as expression vectors for immunogens of important human and veterinary viruses and designed a novel packaging cell line expressing ERNS of BuPV.
Electroporation of the in vitro-transcribed RNAs of all replicon constructs resulted in efficient RNA replication, and expression of BuPV NS3 was readily detected. By also using Nluc-expressing replicon constructs, the replication efficiency of the replicon RNAs could be further investigated. Interestingly, all replicon RNAs (BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE1, BVΔE2_Nluc, BVΔC-E2_Nluc) replicated on comparable levels, indicating that the deletions did not result in altered RNA secondary structures or any other circumstances which could influence RNA replication (49, 64, 65). In accordance with previous investigations using BVDV or the majority of CSFV replicons, no infectious virus could be recovered from cells electroporated with the replicons BVΔC, BVΔE1, BVΔE2, or BVΔC-E2 (42–44, 51). Unexpectedly and in clear contrast to all previous studies, BVΔERNS RNA electroporation resulted in the production of ERNS replicon-based virus particles, which could be passaged for at least two additional rounds in susceptible SK-6 cells. Although these particles were not capable of continued propagation, this is to our knowledge the first description of a viable pestivirus ERNS deletion mutant. However, Wang et al. (66) demonstrated by using pseudovirions that in addition to those containing the three viral glycoproteins E1, E1, and ERNS, pseudovirions were also infectious when ERNS was missing. Thus, it could be shown that ERNS is obviously not essential for CSFV to enter the cell. This observation is in accordance to our data, and these pseudovirions showed a lower infectivity than those containing ERNS (66).
Our study therefore proves that BuPV glycoprotein ERNS is not essential for the rescue of infectious virions. The resulting virions could infect and replicate in SK-6 cells. Neutralization tests demonstrated that the particles did not differ from wild-type BuPV and could be completely neutralized by BuPV-specific sera; therefore, we assume that they were not artifacts from the electroporation procedure. Nevertheless, the produced particles showed a distinct growth defect, and only small foci of infected cells could be detected. This could not be observed for any other analyzed pestivirus used as controls. Previous studies with both CSFV and BVDV replicons also clearly demonstrated that ERNS is strictly essential for virion production, and supernatants of transfected cells did not contain any infectious virus. In all experiments, IF staining of susceptible cells inoculated with transfection supernatants was always negative (51, 56, 67).
Our further experiments also demonstrated that virus rescue from BuPV replicons was independent of the size or the functional domains of the ERNS deletion (BVΔERNS_55, BVΔERNS_88, BVΔERNS_112, BVΔERNS_156) and could also be observed for a mutant with a complete ERNS deletion (BVΔERNS). In contrast, for CSFV, it was shown that deletions within the amphipathic helix strongly influenced the functionality and properties of the ERNS protein and virus production. For instance, mutations in the C-terminal region resulted in reduced activity in blocking dsRNA-induced interferon synthesis (34, 68). We consider this not an irregular nature of the virus particles, since the BuPV particles could be neutralized efficiently by an anti-BuPV serum. However, it remains unclear why Bungowannah virus particles without ERNS protein can be generated. BuPV ERNS has a distinctly lower molecular weight, about 38 kDa, than that of other pestiviruses (48 kDa) and exhibits a lower N-glycosylation. It can be therefore speculated that BuPV ERNS is connected to the viral envelope in a different manner than the ERNS of classical pestiviruses. Although it had been demonstrated that the integrity of the ERNS membrane anchor is important for the recovery of infectious BVDV and CSFV (34, 35), BuPV ERNS shows a deletion of 7 amino acids within this essential membrane anchor region. Furthermore, the formation of an ERNS-E1 protein, which is important in the life cycle of classical pestiviruses, seems to be not essential for the BuPV life cycle (29). Most likely, these different features, alone or in combination, are the basis of the different behavior of BVΔERNS replicons.
Unfortunately, most likely because of the low number of generated ERNS-deleted BuPV particles, all attempts to visualize the defective viruses by electron microscopy were unsuccessful (data not shown).
We also investigated whether the observed growth defect of the ERNS-deleted BuPV replicons is caused by reduced RNA replication compared to that of full-length BuPV RNA, disturbed virus assembly, or reduced virus egress. Our results demonstrated that the replicon BVΔERNS_156 replicated on nearly the same level as full-length BuPV RNA. By comparing infection of SK-6 cells and SK-6_BV-ERNS helper cells with that of packaged BVΔERNS_156_comp VRPs, we could show that the newly generated ERNS-defective virus particles were assembled and could be released from infected SK-6 cells. Nevertheless, virus assembly and/or egress seems to be inefficient in noncomplementing cells but could be fully compensated by the helper cell line providing BuPV-ERNS in trans.
The novel characteristics of BVΔERNS replicons, like the release of infectious particles with the restriction to a few further replication cycles, the highly efficient trans-complementation and passaging on helper cell lines, and the broad range of susceptible cell lines, render these replicons an ideal system for the long-term expression of foreign genes and the development of efficient and safe packaged replicon vaccines. We therefore investigated the trans-complementation of BuPV replicons with the different ERNS deletions and demonstrated that all of these replicons could be efficiently complemented. For BVDV VRPs with E2 or E2 and p7 deletions, it has been reported that they could not be further propagated in cells constitutively expressing the missing proteins, probably due to virus interference (42, 43). In contrast, the packaged BuPV VRPs could be efficiently passaged in the complementing cell line, and virus titers increased to about 107.0 TCID50/ml. Slightly lower maximum virus titers of 105.0 to 105.8 TCID50/ml have been described for packaged CSFV replicons with ERNS deletions (67). In order to point out a pronounced difference in packaging efficiency, it would be interesting to compare the CSFV and BuPV VRP systems directly.
After serial passages of the VRPs in SK-6_BV-ERNS cells, neither mutations within the replicon genome nor recombination of the replicon genome and the ERNS gene of the complementing cells could be seen. Since the BuPV replicons with ERNS deletions are viable for at least two rounds in SK-6 cells, the VRPs could also be detected after two passages in SK-6 cells. However, the virus titer was clearly reduced, which has been also demonstrated by real-time RT-PCR, as no viral RNA could be detected after three to four passages. The generation of an autonomously growing variant after passaging could also be excluded after alternating passages of the BuPV VRPs in complementing SK-6_BV-ERNS cells and SK-6 cells. Neither recombination nor reversion to the wild-type BuPV occurred; however, this cannot be ruled out at higher passage numbers.
Replicons are useful tools for the expression of heterologous genes. In this context, the advantage of noncytopathogenic replicons may be that they continuously replicate at a low level in a broad range of cell lines. In contrast, cytopathogenic replicons seem to be more suitable for high-level expression in the cytoplasm (69–71). Replicon RNA of ssRNA viruses like alphaviruses (e.g., Sindbis virus, Semliki Forest virus, and Venezuelan equine encephalitis virus), flaviviruses (e.g., Kunjin virus, West Nile virus, and yellow fewer virus), measles virus, and rhabdoviruses (rabies virus, vesicular stomatitis virus) have been used as self-replicating expression systems and for vaccine development (72). These viruses are known for their high-level RNA replication in the cytoplasm, which is a prerequisite for a transient expression of heterologous genes. For the production of recombinant particles, replicons can be packaged by cotransfection of a helper vector (73) or packaging cell lines (74), of which the latter approach has been selected in the present study.
Replicon RNA vectors allow the induction of a strong protective immune response in various laboratory animals and have also been tested in clinical trials (72). In previous studies, mainly surface antigens capable of inducing neutralizing antibodies against pathogenic infectious diseases have been selected as heterologous genes to be inserted into the replicon vectors (75–77). Accordingly, the major immunogens of CSFV, SBV, and MERS-CoV were selected to be expressed in the context of the BuPV replicon system.
We constructed chimeric BuPV replicons with insertions of three model antigens, CSFV E2 (BVΔERNS_CSFV_E2), the N-terminal domain GcN of the SBV glycoprotein Gc (BVΔERNS_2GcN), and the receptor-binding domain (RBD) of the MERS-CoV spike protein (BVΔERNS_RBD). All proteins could be expressed, and trans-complementation and propagation of the expression replicons using the SK-6_BV-ERNS cell line was highly efficient. Titers of the expressing VRPs were comparable with those of the VRPs without insertions, and neither recombination nor loss of expression could be observed. Furthermore, the broad cell tropism of BuPV allowed, e.g., the infection of alpaca, llama, Bactrian camel, and human cells as well as cells of ovine and bovine origin by the chimeric BuPV replicons. Future studies will now focus on immunization/challenge experiments with the different constructs in the respective target animals.
In summary, the BuPV ERNS deletion mutants and VRPs described in this study represent the first pestivirus replicons that could be passaged in noncomplementing cells despite the deletion of a complete envelope protein. However, the ERNS-deleted BuPV replicons and VRPs are still growth defective and abortive in further growth. Nevertheless, their unique properties make BuPV ΔERNS replicons and VRPs a promising novel replicon-based expression and vaccine platform.
MATERIALS AND METHODS
Cells.
SK-6 (swine kidney cells; RIE262), HEK-293 (human kidney cells; RIE197), TT-R (Bactrian camel umbilical cord cells; RIE1006), SFT-R (sheep thymus cells; RIE43), KOP-R (cattle esophagus/pharynx cells; RIE244) (Collection of Cell Lines in Veterinary Medicine, Friedrich-Loeffler-Institut, Insel Riems, Germany [CCLV]) were grown in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal calf serum (FCS) at 37°C and 5% CO2.
Plasmid constructs.
Plasmids were amplified in Escherichia coli DH10B cells (Invitrogen, USA). Plasmid DNA was purified by use of Qiagen plasmid mini or midi kits (Qiagen, Germany). All cDNA constructs were generated by fusion PCR, a modified restriction-free cloning method (78, 79). Pairs of forward and reverse PCR primers used for the respective constructs are given in Table 3. The identity of the constructs was confirmed by DNA sequencing.
TABLE 3.
Constructs and PCR primers used for construction
| Construct | Primer | Sequence (5′ → 3′)a |
|---|---|---|
| BVΔC | Bungo_dC_1012F | GACAAAGCCTAAAGAGACTGAACCAGAAGCGTCCAGGAAGAAG |
| Bungo_1499R | GATTGATCCACGGCTCTATATTG | |
| BVΔE2 | Bungo_2060F | CTGAGCTTAATTGCAATGTC |
| Bungo_dE2_R | GAGCCACTATGTTCTCGTCGGCTTGTGCTCCCCCTATG | |
| BVΔERNS_156 | Bungo_dErns_F | CATCTAGCAGCAGACTATGAAAGTAAGATTGAAAACACCAAGA |
| Bungo_2164R | CATCACGAAGTCCCTGTTGTC | |
| BVΔC-E2 | Ph_dCE2_Bungo _F | GACAAAGCCTAAAGAGACTGAATGGTTTGATCTCACAGCAAAG |
| Bungo_4021R | GAGACTTATTACAGCCACAAG | |
| BVΔERNS_55 | Ph_Bungo_dErns_55F | CAAGCATGGGTGGTGCAACAAGAAAGGGCACAACTTCTC |
| Bungo_2164R | CATCACGAAGTCCCTGTTGTC | |
| BVΔERNS_88 | Ph_Bungo_dErns_88F | CATCTAGCAGCAGACTATGAAAAGAAAGGGCACAACTTCTC |
| Bungo_2164R | CATCACGAAGTCCCTGTTGTC | |
| BVΔERNS_112 | Ph_Bungo_dErns_112F | CATCTAGCAGCAGACTATGAAGACTTGCTCAGGATCATGG |
| Bungo_2164R | CATCACGAAGTCCCTGTTGTC | |
| BVΔERNS | BV_dErns_kompF | GCCTATTGGTCGTGCCGGTAGGGTCCAGTCCTTACTGCCCAGTGGC |
| Bungo_2164R | CATCACGAAGTCCCTGTTGTC | |
| BVΔE1 | BVdE1A_F | CAGTCCTTACTGCCCAGTGGCTTTGTGGACCGAAGCAACAGC |
| Bungo_2874R | CTAGGGCATTCAAGTTCAAAGC | |
| BV_GAA_Nluc | Bungo_GAA_F | GATACATGTGTGTGGAGCTGCTGGTTTCTTGATAACTGAG |
| Bungo_11835R | CTGAGCCAATTTAGAGAAC | |
| pA/BV_Nluc | Ph_CNlucF | CTGACGACAAAGGAGCAAAAATGGTCTTCACACTCGAAGATTTC |
| Ph_NlucR | GAAGTTCGTGGCTCCGGAACCCGCCAGAATGCGTTCGCACAG | |
| pCAGGS_sigERNS_BV | CAGGS_sigErns_F | GGCAAAGAATTCGCCACCATGGAGGCCAGCCGGAAGAAGCTG |
| CAGGS_Erns_R | GAAAATAACATATGCGGCCGCTAGCTCAGGCCTCGGCGCCGAAG | |
| BVdERNS_2GcN | 2Gc_ F | CATCTAGCAGCAGACTATGAAAGTATTAACTGCAAAAACATT |
| Erns_GC_amino_R | CTTGGTGTTTTCAATCTTACTGATCAGAGATAAGGTAGTGAG | |
| BVdERNS_RBD | BV_dErns_RBD_F | CTAGCAGCAGACTATGAATCTTTCGAAGCAAAACCTTCTGG |
| BV_dErns_RBD_R | GGTGTTTTCAATCTTACTATATTCCACGCAATTGCCTAATTGAG | |
| BVdERNS_CSFV_E2 | BV_CSFV_E2_F | GGTCGTGCCGGTAGGGTCCCAGCTAGCCTGCAAGGAAG |
| BV_CSFV_E2_R | GCCACTGGGCAGTAAGGACTACCAGCGGCGAGTTGTTCT |
Restriction enzyme sites are underlined.
The infectious full-length cDNA clone pA/BV (9) was generated on the basis of published BuPV sequences (NCBI GenBank accession numbers NC_023176.1 and DQ901402.1). The completely synthetic construct was constituted from six plasmids harboring synthetic peptides (Geneart AG, Regensburg, Germany), which were assembled into plasmid vector pA (40) by fusion PCR cloning. In this plasmid, the full-length BuPV cDNA is flanked by the bacteriophage T7 promoter sequence at the 5′ end and a SmaI restriction site at the 3′ end. The construct pA/BV_Nluc was generated on the basis of plasmid pA/BV_GFP (I. Reimann, unpublished data) by replacing the eGFP gene with that of the Nluc gene (Fig. 1C). The eGFP gene has been located between the genes encoding NPRO and the capsid protein C and is flanked upstream by 21 nt of the capsid gene and downstream by a linker and the teschovirus 2Apro gene.
In brief, the Nluc gene was amplified from plasmid DNA pNL1.1 (Promega, Germany) using primers Ph_CNlucF and Ph_NlucR (biomers.net GmbH, Germany) by PCR using Phusion high-fidelity PCR master mix (New England Biolabs, USA). Subsequently, the purified amplicon was used as a megaprimer in a fusion PCR with plasmid pA/BV_GFP as the template to generate full-length pA/BV_Nluc.
The subgenomic replicons (BVΔC, BVΔERNS, BVΔERNS_156, BVΔERNS_55, BVΔERNS_88, BVΔERNS_112, BVΔE2, BVΔE1, BVΔC-E2) were generated on the basis of pA/BV by complete or partial deletion of the genomic regions encoding the respective structural proteins (Fig. 1A) by fusion PCR using the appropriate primers. Nluc-expressing replicons (BVΔC_Nluc, BVΔERNS_156_ Nluc, BVΔE2_Nluc, BVΔE1_Nluc, BVΔC-E2_Nluc, BV_GAA_Nluc) were constructed in the same way, using plasmid pA/BV_Nluc as the template.
Bungowannah virus replicons containing foreign genes were constructed based on replicons with deletions within the ERNS-encoding region. For the construction of the replicon expressing the antigenic domain GcN of SBV, a megaprimer was amplified by using primers 2Gc_F and Erns_Gc_amino_R and plasmid syn_2xGc_amino_linker_pMK_RQ (Geneart AG, Germany) as the template DNA. Subsequently, the megaprimer was used for fusion PCR with the DNA template BVΔERNS_156 to generate the construct BVΔERNS_2GcN. The construct BVΔERNS_RBD, expressing the receptor binding domain (RBD) of the MERS-CoV spike protein, was also generated on the basis of plasmid BVΔERNS_156 but using a megaprimer, which was generated by PCR with primers BV_dErns_RBD_F and BV_dErns_RBD_R and plasmid S1_RBD_pMA-T (Geneart AG, Germany) as the DNA template. The CSFV E2-expressing plasmid BVΔERNS_CSFV_E2 was based on BVΔERNS with a complete ERNS deletion. A DNA fragment was amplified by PCR by using primers BV_CSFV_E2_F and BV_CSFV_E2_R and plasmid pA/CP7_E2alf (80) as the template DNA, which was used as a megaprimer in a fusion PCR with the DNA template BVΔERNS.
For construction of the expression plasmid pCAGGS_BuPV-ERNS, the genomic region encoding the C-terminal parts of the BuPV C and BuPV ERNS proteins was amplified from the chemically synthesized and sequence-optimized ORF syn_sigERNS_BV_opt_pMA-T (Geneart AG, Germany). The amplicon was inserted into the plasmid vector pCAGGS_T7POLiresPuro (kindly provided by Stefan Finke, Friedrich-Loeffler-Institut) by fusion PCR using primers CAGGS_sigErns_F and CAGGS_Erns_R.
Further details for the generation of the plasmid constructs are available upon request.
Establishment of BuPV ERNS-expressing SK6 cells (SK-6_BV-ERNS).
Subconfluent monolayers of SK-6 cells were transfected with plasmid pCAGGS_BuPV-ERNS using Lipofectamine 3000 (Invitrogen, USA) according to the manufacturer’s protocol with a 1.5-to-1 (vol/wt) Lipofectamine 3000-to-DNA ratio. In short, plasmid DNA and transfection reagent were added to Opti-MEM I (1×) plus GlutaMAX-I reduced serum medium (Gibco; Life Technologies, USA) separately and incubated for 5 min at room temperature. After 20 min of incubation at room temperature, the transfection mixture was added directly to the cell monolayer. After 48 h, the cell culture medium was replaced with fresh DMEM with 10% FCS supplemented with 2 μg/ml puromycin (Gibco, Life Technologies, USA). Single colonies were picked, replaced, and propagated under selection with 2 μg/ml puromycin and stained for ERNS expression by using the BuPV ERNS-specific MAb 43C3.
In vitro transcription, electroporation, and virus rescue.
For transfection experiments, cDNA plasmids were linearized with SmaI (New England Biolabs, USA) and in vitro transcribed using a T7 RiboMax large-scale RNA production system (Promega, Germany) according to the manufacturer’s instructions. The in vitro-synthesized RNA was transfected (1 to 5 μg) into SK-6 cells by electroporation using a Gene Pulser Xcell electroporation system (Bio-Rad, USA). After two pulses (850 V, 25 μF, 50 Ω), the cells were seeded in cell culture plates and incubated for 48 to 72 h at 37°C and 5% CO2. For recovery of infectious virus, cell culture supernatants of the electroporated cells were harvested at 72 h posttransfection (hpt), and after centrifugation (3,000 rpm, 5 min) and filtration with a 0.22-μm Millex filter unit (Merck, Germany), the supernatants were passaged on fresh cells. To further exclude any carryover events, the supernatant of BVΔERNS_156 RNA-electroporated cells was additionally passaged onto fresh SK-6 cells, the medium was removed after 2 h, the cells were washed several times with phosphate-buffered saline (PBS), and, thereafter, fresh DMEM was added.
Virus stocks were prepared by passaging the viruses on SK-6 cells. Virus titers were calculated and expressed as 50% tissue culture infective dose (TCID50) per ml. The identity of the recombinant viruses was confirmed by RT-PCR and sequencing using the appropriate primers.
IF assays.
For immunofluorescence (IF) analysis, electroporated or infected cells were fixed and permeabilized with 80% acetone. Subsequently, the cells were incubated with the working dilution of the monoclonal antibody of interest for 30 to 45 min. These included the pan-pestivirus NS3-specific MAb WB112 (1:500; CVL, UK), the BuPV E2-specific MAb 682/45F12 or the BuPV ERNS-specific MAb 682/43C3 (1:20; M. Dauber, Friedrich-Loeffler-Institut, Insel Riems, Germany) as well as the SBV GcN-specific MAb 5E1_A3 (1:50; A. Aebischer, Friedrich-Loeffler-Institut, Insel Riems, Germany), CSFV E2-specific MAb Bio275 (1:100; Bio-X Diagnostics, Belgium), and a MERS-CoV RBD-specific MAb (1:10,000; kindly provided by B. J. Bosch, Leiden University Medical Center, The Netherlands; unpublished). After two washing steps with PBS, cell cultures were incubated with a goat anti-mouse Ig Alexa488 conjugate or, for MERS-CoV RBD, a goat anti-human Ig Alexa488 (1:1,000; Thermo Fisher Scientific, Inc., USA) for 30 min and finally washed with PBS. Images were captured with a fluorescence microscope (Nikon Eclipse) with 4×, 10×, 20×, and 40× lens objectives.
Virus neutralization assay.
One hundred microliters of supernatant from SK-6 cells electroporated with in vitro-transcribed RNA of BVΔERNS deletion mutants was incubated in a 24-well cell culture plate for 2 h at 37°C with 50 μl undiluted BuPV antibody-positive serum generated by experimental infection with the first Australian cell culture isolate (permission number 7221.3-1.1-064/11). Thereafter, an SK-6 cell suspension (2 × 104 cells/ml) was added and the plates were incubated for 72 h at 37°C. Neutralizations were detected by IF staining using the BuPV E2-specific MAb 682/45F12. As a negative control, SK-6 cells electroporated with in vitro-transcribed RNA of BVΔERNS deletion mutants were incubated as described above with a BuPV-negative serum.
Nluc activity detection.
For nanoluciferase (Nluc) assays, 250 ng of SmaI-digested DNA of the Nluc constructs (BVΔC_Nluc, BVΔERNS_156_Nluc, BVΔE2_Nluc, BVΔC-E2_Nluc, BVΔE1_Nluc, and BV_GAA_Nluc) was in vitro transcribed. A total of 1 × 107 SK-6 cells were transfected with 1 μg synthesized RNA by electroporation. The cells were diluted with 15 ml medium, and 100 μl cell suspension per cavity was added to a 96-well plate. After 4, 24, 48, and 72 h of incubation at 37°C and 5% CO2, the expression of the Nluc reporter gene was detected by using a Nano-Glo luciferase assay system (Promega, USA) according to the manufacturer’s protocol. Briefly, at the different time points, the supernatant was removed and the electroporated SK-6 cells were detached with fresh medium. Nano-Glo luciferase assay buffer and Nano-Glo luciferase assay substrate (Furimazine) were mixed (50:1) and added to the cells (1:1). Luciferase activity was measured as relative light units (RLU) by using a luminometer (Tecan Infinite F200 PRO; Switzerland). Three independent experiments were performed, and samples were measured in triplicate. The measured values were normalized to the values of the replication-deficient GAA mutant at the different time points. Significance analyses were performed using one-way analysis of variance (ANOVA), whereby the Tukey test was used for pairwise comparison as implemented in GraphPad Prism software 8.0.1 for Windows (GraphPad Software, USA).
Complementation studies.
For trans-complementation experiments, in vitro-transcribed RNA of the BuPV replicons (BVΔERNS_156, BVΔERNS_112, BVΔERNS_88, BVΔERNS_55, BVΔERNS, BVΔERNS_2GcN, BVΔERNS_RBD, BVΔERNS_CSFV_E2) was electroporated into SK-6_BV-ERNS cells. Cell culture supernatants were collected 72 hpt and filtered using a 0.22-μm Millex filter unit (Merck, Germany). For detection of trans-complemented VRPs, SK-6 cells grown in six-well plates were inoculated with 1 ml of collected supernatant and further incubated at 37°C and 5% CO2. At 72 h postinfection (hpi), cells were investigated by IF staining using anti-NS3-specific MAb WB 112. VRPs were serially passaged either in SK-6 or SK-6_BV-ERNS cells or alternating in SK-6 and SK-6_BV-ERNS cells and titrated using SK-6 cells. VRPs expressing SBV GcN (BVΔERNS_2GcN_comp) were also passaged in KOP-R and SFT-R cells, and VPRs expressing MERS-CoV RBD (BVΔERNS_RBD_comp) were passaged also in HEK-293T and TT-R cells. Virus titers were determined as TCID50/ml by IF staining using the pan-pestivirus NS3-specific MAb WB112 or one of the insert-specific MAbs, 5E1_A3 (anti-GcN), Bio275 (anti-CSFV E2), and MERS-CoV RBD-specific MAb (anti-RBD).
PCR and sequencing.
PCR was carried out using a C1000 thermal cycler (Bio-Rad, USA). DNA-based amplification was done using GoTaq polymerase (Promega, Germany) according to the supplier’s protocols. For RT-PCR, total RNA of virus-infected cells was extracted using a QIAamp viral RNA minikit (Qiagen, Germany) according to the manufacturer’s instruction and cDNA was amplified using a OneStep RT-PCR kit (Qiagen, Germany). Sequencing was carried out using a BigDye Terminator v1.1 cycle sequencing kit (Applied Biosystems, USA). Nucleotide sequences were read with an automatic sequencer (3130 Genetic Analyzer; Applied Biosystems, USA) and analyzed using Geneious software (version 10.2.3; Biomatters Ltd., New Zealand).
Real-time RT-PCR.
For quantification of viral RNA, SK-6 cells and SK-6_BV-ERNS cells were infected with full-length BuPV or BVΔERNS _156_comp at a multiplicity of infection (81) of 0.1 TCID50/ml. SK-6 cells and SK-6_BV-ERNS cells were also inoculated with supernatants of SK-6_BV-ERNS cells electroporated with BVΔE1 RNA (negative control). At 2 and 48 hpi, cells and supernatant of the respective wells were separated. Total RNA was extracted using TRIzol reagent (cells) or TRIzol/LS reagent (supernatant; Thermo Fisher Scientific, Inc., USA) according to the manufacturer’s instruction. The real-time PCR (RT-qPCR) was performed using an AgPath-ID one-step RT-PCR kit (Thermo Fisher Scientific, Inc., USA), the BuPV capsid protein targeting primers BV-1100-F (5′-CGAAACCCAAGACTCA AGACG-3′) and BV-1203-R (5′-GCAGGCTAATATTGCCCATGC-3′), and a BuPV-specific FAM-labeled probe (5′-FAM-CACAACAAGAACAAACCAGAAGCGTCC-3′). Real-time RT-PCR was carried out using a CFX 96 real-time detection system (Bio-Rad, USA) with the following thermal profile: a reverse transcription step at 45°C for 10 min, a PCR initial activation step at 95°C for 10 min, and then 42 cycles of three-step cycling consisting of denaturation at 95°C for 15 s, annealing at 57°C for 20 s, and extension at 72°C for 30 s. All samples were tested in triplicate, and to determine the genome load in the samples, an external standard was included in every real-time RT-PCR run. Two biological and three technical replicates were performed. Significance analysis was performed using one-way analysis of variance (ANOVA), whereby the Tukey test was used for pairwise comparison as implemented in GraphPad Prism software 8.4.0 for Windows (GraphPad Software, USA).
Western blot analysis.
For Western blot analysis, SK-6 cells were infected with BVΔERNS_CSFV_E2_comp, BVΔERNS_RBD_comp, BVΔERNS_2GcN_comp, and BVΔERNS_156_comp, respectively, at an MOI of 0.1 TCID50/ml. Total cell lysates were prepared at 48 hpi. The proteins were separated by 10% SDS-PAGE under reducing (BVΔERNS_CSFV_E2_comp, BVΔERNS_RBD_comp) or nonreducing (BVΔERNS_2GcN_comp) conditions and transferred onto a nitrocellulose membrane (Bio-Rad, USA). After overnight blocking at 4°C with 1× ROTIBlock solution (Carl Roth, Germany), the membrane was incubated with the anti-GcN MAb (5E1_A3, 1:20), anti-E2 CSFV MAb (1:300; Bio-X Diagnostics, Belgium), or MERS-CoV spike protein S1 rabbit polyclonal antibody (PAb) (1:2,000; Sino Biological Inc., USA) for 2 h. As a secondary antibody, a horseradish peroxidase-conjugated anti-mouse or anti-rabbit antibody (1:20,000; Dianova, Germany) was used. After washing, the membrane was incubated with SuperSignal-West Pico chemiluminescent detection reagent (Thermo Fisher Scientific, Inc., USA) and visualized using a ChemoCam Western blot imaging system (Intas Science Imaging Instruments GmbH, Germany). BVΔERNS_156_comp was used as a negative control. The protein sizes were calculated by Geneious software (version 10.2.3; Biomatters Ltd., Auckland, New Zealand).
FACS analysis.
For the detection of VRP infection rates in different cell lines and to investigate a possible influence of the foreign genes, FACS analysis was performed. The cells were infected with the packaged replicons BVΔERNS_CSFV_E2_comp, BVΔERNS_RBD_comp, BVΔERNS_2GcN_comp, and BVΔERNS_156_comp (MOI of 1). At 24 hpi, cells were fixed (4% paraformaldehyde), permeabilized (digitonin; Fluka Chemie GmbH, Switzerland), and incubated for 45 to 60 min with the pan-pestivirus NS3-specific MAb WB112 on ice. After a washing step with PBS, the cells were incubated with a goat anti-mouse Ig Alexa488 conjugate (Thermo Fisher Scientific, Inc., USA) for 45 to 60 min on ice, washed, and analyzed by using a FACSCalibur flow cytometer (Becton, Dickinson, USA). Two biological and technical replicates were performed. The FACS data obtained were analyzed with CellQuest Pro software (Becton, Dickinson, USA).
Data availability.
All raw data are available upon request.
ACKNOWLEDGMENTS
We thank Doreen Schulz, Gabriela Adam, Bianka Hillmann, and Aileen Stoll for excellent technical assistance.
This work was financially supported by the Zoonoses Anticipation and Preparedness Initiative (ZAPI; grant agreement no. 115760) within the Innovative Medicines Initiative (IMI Call 11–IMI-JU-11-2013-04).
REFERENCES
- 1.Simmonds P, Becher P, Bukh J, Gould EA, Meyers G, Monath T, Muerhoff S, Pletnev A, Rico-Hesse R, Smith DB, Stapleton JT, ICTV Report Consortium. 2017. ICTV virus taxonomy profile: Flaviviridae. J Gen Virol 98:2–3. doi: 10.1099/jgv.0.000672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Finlaison DS, King KR, Frost MJ, Kirkland PD. 2009. Field and laboratory evidence that Bungowannah virus, a recently recognised pestivirus, is the causative agent of the porcine myocarditis syndrome (PMC). Vet Microbiol 136:259–265. doi: 10.1016/j.vetmic.2008.11.026. [DOI] [PubMed] [Google Scholar]
- 3.Kirkland PD, Frost MJ, Finlaison DS, King KR, Ridpath JF, Gu X. 2007. Identification of a novel virus in pigs—Bungowannah virus: a possible new species of pestivirus. Virus Res 129:26–34. doi: 10.1016/j.virusres.2007.05.002. [DOI] [PubMed] [Google Scholar]
- 4.Kirkland PD, Read AJ, Frost MJ, Finlaison DS. 2015. Bungowannah virus—a probable new species of pestivirus—what have we found in the last 10 years? Anim Health Res Rev 16:60–63. doi: 10.1017/S1466252315000031. [DOI] [PubMed] [Google Scholar]
- 5.Abrahante JE, Zhang JW, Rossow K, Zimmerman JJ, Murtaugh MP. 2014. Surveillance of Bungowannah pestivirus in the upper Midwestern USA. Transbound Emerg Dis 61:375–377. doi: 10.1111/tbed.12035. [DOI] [PubMed] [Google Scholar]
- 6.Lamp B, Schwarz L, Hogler S, Riedel C, Sinn L, Rebel-Bauder B, Weissenbock H, Ladinig A, Rümenapf T. 2017. Novel pestivirus species in pigs, Austria, 2015. Emerg Infect Dis 23:1176–1179. doi: 10.3201/eid2307.170163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Jo WK, van Elk C, van de Bildt M, van Run P, Petry M, Jesse ST, Jung K, Ludlow M, Kuiken T, Osterhaus A. 2019. An evolutionary divergent pestivirus lacking the NPRO gene systemically infects a whale species. Emerg Microbes Infect 8:1383–1392. doi: 10.1080/22221751.2019.1664940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fernelius AL, Lambert G, Hemness GJ. 1969. Bovine viral diarrhea virus-host cell interactions: adaptation and growth of virus in cell lines. Am J Vet Res 30:1561–1572. [PubMed] [Google Scholar]
- 9.Richter M, Reimann I, Schirrmeier H, Kirkland PD, Beer M. 2014. The viral envelope is not sufficient to transfer the unique broad cell tropism of Bungowannah virus to a related pestivirus. J Gen Virol 95:2216–2222. doi: 10.1099/vir.0.065995-0. [DOI] [PubMed] [Google Scholar]
- 10.Collett MS, Larson R, Belzer SK, Retzel E. 1988. Proteins encoded by bovine viral diarrhea virus: the genomic organization of a pestivirus. Virology 165:200–208. doi: 10.1016/0042-6822(88)90673-3. [DOI] [PubMed] [Google Scholar]
- 11.Collett MS, Anderson DK, Retzel E. 1988. Comparisons of the pestivirus bovine viral diarrhoea virus with members of the flaviviridae. J Gen Virol 69:2637–2643. doi: 10.1099/0022-1317-69-10-2637. [DOI] [PubMed] [Google Scholar]
- 12.Donis RO. 1995. Molecular biology of bovine viral diarrhea virus and its interactions with the host. Vet Clin North Am Food Anim Pract 11:393–423. doi: 10.1016/s0749-0720(15)30459-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Becher P, Orlich M, Thiel HJ. 1998. Complete genomic sequence of border disease virus, a pestivirus from sheep. J Virol 72:5165–5173. doi: 10.1128/JVI.72.6.5165-5173.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hause BM, Collin EA, Peddireddi L, Yuan F, Chen Z, Hesse RA, Gauger PC, Clement T, Fang Y, Anderson G. 2015. Discovery of a novel putative atypical porcine pestivirus in pigs in the USA. J Gen Virol 96:2994–2998. doi: 10.1099/jgv.0.000251. [DOI] [PubMed] [Google Scholar]
- 15.Meyers G, Rümenapf T, Thiel HJ. 1989. Molecular cloning and nucleotide sequence of the genome of hog cholera virus. Virology 171:555–567. doi: 10.1016/0042-6822(89)90625-9. [DOI] [PubMed] [Google Scholar]
- 16.Rümenapf T, Thiel H-J. 2008. Molecular biology of pestiviruses In Mettenleiter TC, Sobrino F (ed), Animal viruses: molecular biology. Caister Academic Press, Norfolk, United Kingdom. [Google Scholar]
- 17.Yu H, Grassmann CW, Behrens SE. 1999. Sequence and structural elements at the 3′ terminus of bovine viral diarrhea virus genomic RNA: functional role during RNA replication. J Virol 73:3638–3648. doi: 10.1128/JVI.73.5.3638-3648.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yu H, Isken O, Grassmann CW, Behrens SE. 2000. A stem-loop motif formed by the immediate 5′ terminus of the bovine viral diarrhea virus genome modulates translation as well as replication of the viral RNA. J Virol 74:5825–5835. doi: 10.1128/jvi.74.13.5825-5835.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fletcher SP, Jackson RJ. 2002. Pestivirus internal ribosome entry site (IRES) structure and function: elements in the 5′ untranslated region important for IRES function. J Virol 76:5024–5033. doi: 10.1128/jvi.76.10.5024-5033.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pestova TV, Hellen CU. 1999. Internal initiation of translation of bovine viral diarrhea virus RNA. Virology 258:249–256. doi: 10.1006/viro.1999.9741. [DOI] [PubMed] [Google Scholar]
- 21.Rümenapf T, Unger G, Strauss JH, Thiel HJ. 1993. Processing of the envelope glycoproteins of pestiviruses. J Virol 67:3288–3294. doi: 10.1128/JVI.67.6.3288-3294.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lindenbach BD, Rice CM. 2003. Molecular biology of flaviviruses. Adv Virus Res 59:23–61. doi: 10.1016/s0065-3527(03)59002-9. [DOI] [PubMed] [Google Scholar]
- 23.Thiel HJ, Stark R, Weiland E, Rümenapf T, Meyers G. 1991. Hog cholera virus: molecular composition of virions from a pestivirus. J Virol 65:4705–4712. doi: 10.1128/JVI.65.9.4705-4712.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gong Y, Trowbridge R, Macnaughton TB, Westaway EG, Shannon AD, Gowans EJ. 1996. Characterization of RNA synthesis during a one-step growth curve and of the replication mechanism of bovine viral diarrhoea virus. J Gen Virol 77:2729–2736. doi: 10.1099/0022-1317-77-11-2729. [DOI] [PubMed] [Google Scholar]
- 25.Behrens SE, Grassmann CW, Thiel HJ, Meyers G, Tautz N. 1998. Characterization of an autonomous subgenomic pestivirus RNA replicon. J Virol 72:2364–2372. doi: 10.1128/JVI.72.3.2364-2372.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Riedel C, Lamp B, Heimann M, Rümenapf T. 2010. Characterization of essential domains and plasticity of the classical swine fever virus core protein. J Virol 84:11523–11531. doi: 10.1128/JVI.00699-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chu HJ, Zee YC. 1984. Morphology of bovine viral diarrhea virus. Am J Vet Res 45:845–850. [PubMed] [Google Scholar]
- 28.Weiland F, Weiland E, Unger G, Saalmüller A, Thiel HJ. 1999. Localization of pestiviral envelope proteins Erns and E2 at the cell surface and on isolated particles. J Gen Virol 80:1157–1165. doi: 10.1099/0022-1317-80-5-1157. [DOI] [PubMed] [Google Scholar]
- 29.Wegelt A, Reimann I, Zemke J, Beer M. 2009. New insights into processing of bovine viral diarrhea virus glycoproteins Erns and E1. J Gen Virol 90:2462–2467. doi: 10.1099/vir.0.012559-0. [DOI] [PubMed] [Google Scholar]
- 30.Mayer D, Hofmann MA, Tratschin JD. 2004. Attenuation of classical swine fever virus by deletion of the viral NPRO gene. Vaccine 22:317–328. doi: 10.1016/j.vaccine.2003.08.006. [DOI] [PubMed] [Google Scholar]
- 31.Tratschin JD, Moser C, Ruggli N, Hofmann MA. 1998. Classical swine fever virus leader proteinase NPRO is not required for viral replication in cell culture. J Virol 72:7681–7684. doi: 10.1128/JVI.72.9.7681-7684.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Iqbal M, Flick-Smith H, McCauley JW. 2000. Interactions of bovine viral diarrhoea virus glycoprotein Erns with cell surface glycosaminoglycans. J Gen Virol 81:451–459. doi: 10.1099/0022-1317-81-2-451. [DOI] [PubMed] [Google Scholar]
- 33.Zürcher C, Sauter KS, Mathys V, Wyss F, Schweizer M. 2014. Prolonged activity of the pestiviral RNase Erns as an interferon antagonist after uptake by clathrin-mediated endocytosis. J Virol 88:7235–7243. doi: 10.1128/JVI.00672-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fetzer C, Tews BA, Meyers G. 2005. The carboxy-terminal sequence of the pestivirus glycoprotein Erns represents an unusual type of membrane anchor. J Virol 79:11901–11913. doi: 10.1128/JVI.79.18.11901-11913.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tews BA, Meyers G. 2007. The pestivirus glycoprotein Erns is anchored in plane in the membrane via an amphipathic helix. J Biol Chem 282:32730–32741. doi: 10.1074/jbc.M706803200. [DOI] [PubMed] [Google Scholar]
- 36.Hausmann Y, Roman-Sosa G, Thiel HJ, Rümenapf T. 2004. Classical swine fever virus glycoprotein Erns is an endoribonuclease with an unusual base specificity. J Virol 78:5507–5512. doi: 10.1128/jvi.78.10.5507-5512.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Schneider R, Unger G, Stark R, Schneider-Scherzer E, Thiel HJ. 1993. Identification of a structural glycoprotein of an RNA virus as a ribonuclease. Science 261:1169–1171. doi: 10.1126/science.8356450. [DOI] [PubMed] [Google Scholar]
- 38.Windisch JM, Schneider R, Stark R, Weiland E, Meyers G, Thiel HJ. 1996. RNase of classical swine fever virus: biochemical characterization and inhibition by virus-neutralizing monoclonal antibodies. J Virol 70:352–358. doi: 10.1128/JVI.70.1.352-358.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Meyers G, Saalmüller A, Büttner M, 1999. Mutations abrogating the RNase activity in glycoprotein Erns of the pestivirus classical swine fever virus lead to virus attenuation. J Virol 73:10224–10235. doi: 10.1128/JVI.73.12.10224-10235.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Meyers G, Tautz N, Becher P, Thiel HJ, Kümmerer BM. 1996. Recovery of cytopathogenic and noncytopathogenic bovine viral diarrhea viruses from cDNA constructs. J Virol 70:8606–8613. doi: 10.1128/JVI.70.12.8606-8613.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tautz N, Harada T, Kaiser A, Rinck G, Behrens S, Thiel HJ. 1999. Establishment and characterization of cytopathogenic and noncytopathogenic pestivirus replicons. J Virol 73:9422–9432. doi: 10.1128/JVI.73.11.9422-9432.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Harada T, Tautz N, Thiel HJ. 2000. E2-p7 region of the bovine viral diarrhea virus polyprotein: processing and functional studies. J Virol 74:9498–9506. doi: 10.1128/jvi.74.20.9498-9506.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Reimann I, Meyers G, Beer M. 2003. Trans-complementation of autonomously replicating bovine viral diarrhea virus replicons with deletions in the E2 coding region. Virology 307:213–227. doi: 10.1016/s0042-6822(02)00129-0. [DOI] [PubMed] [Google Scholar]
- 44.Moser C, Stettler P, Tratschin JD, Hofmann MA. 1999. Cytopathogenic and noncytopathogenic RNA replicons of classical swine fever virus. J Virol 73:7787–7794. doi: 10.1128/JVI.73.9.7787-7794.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Riedel C, Lamp B, Heimann M, König M, Blome S, Moennig V, Schüttler C, Thiel HJ, Rümenapf T. 2012. The core protein of classical swine fever virus is dispensable for virus propagation in vitro. PLoS Pathog 8:e1002598. doi: 10.1371/journal.ppat.1002598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lo MK, Tilgner M, Shi PY. 2003. Potential high-throughput assay for screening inhibitors of West Nile virus replication. J Virol 77:12901–12906. doi: 10.1128/jvi.77.23.12901-12906.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rossi SL, Zhao Q, O'Donnell VK, Mason PW. 2005. Adaptation of West Nile virus replicons to cells in culture and use of replicon-bearing cells to probe antiviral action. Virology 331:457–470. doi: 10.1016/j.virol.2004.10.046. [DOI] [PubMed] [Google Scholar]
- 48.Ng CY, Gu F, Phong WY, Chen YL, Lim SP, Davidson A, Vasudevan SG. 2007. Construction and characterization of a stable subgenomic dengue virus type 2 replicon system for antiviral compound and siRNA testing. Antiviral Res 76:222–231. doi: 10.1016/j.antiviral.2007.06.007. [DOI] [PubMed] [Google Scholar]
- 49.Leardkamolkarn V, Sirigulpanit W, Chotiwan N, Kumkate S, Huang CY. 2012. Development of dengue type-2 virus replicons expressing GFP reporter gene in study of viral RNA replication. Virus Res 163:552–562. doi: 10.1016/j.virusres.2011.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Xie X, Zou J, Shan C, Yang Y, Kum DB, Dallmeier K, Neyts J, Shi PY. 2016. Zika virus replicons for drug discovery. EBioMedicine 12:156–160. doi: 10.1016/j.ebiom.2016.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Reimann I, Semmler I, Beer M. 2007. Packaged replicons of bovine viral diarrhea virus are capable of inducing a protective immune response. Virology 366:377–386. doi: 10.1016/j.virol.2007.05.006. [DOI] [PubMed] [Google Scholar]
- 52.Maurer R, Stettler P, Ruggli N, Hofmann MA, Tratschin JD. 2005. Oronasal vaccination with classical swine fever virus (CSFV) replicon particles with either partial or complete deletion of the E2 gene induces partial protection against lethal challenge with highly virulent CSFV. Vaccine 23:3318–3328. doi: 10.1016/j.vaccine.2005.01.076. [DOI] [PubMed] [Google Scholar]
- 53.Pijlman GP, Suhrbier A, Khromykh AA. 2006. Kunjin virus replicons: an RNA-based, non-cytopathic viral vector system for protein production, vaccine and gene therapy applications. Expert Opin Biol Ther 6:135–145. doi: 10.1517/14712598.6.2.135. [DOI] [PubMed] [Google Scholar]
- 54.Khromykh AA. 2000. Replicon-based vectors of positive strand RNA viruses. Curr Opin Mol Ther 2:555–569. [PubMed] [Google Scholar]
- 55.Yang Z, Wu R, Li RW, Li L, Xiong Z, Zhao H, Guo D, Pan Z. 2012. Chimeric classical swine fever (CSF)-Japanese encephalitis (JE) viral replicon as a non-transmissible vaccine candidate against CSF and JE infections. Virus Res 165:61–70. doi: 10.1016/j.virusres.2012.01.007. [DOI] [PubMed] [Google Scholar]
- 56.Frey CF, Bauhofer O, Ruggli N, Summerfield A, Hofmann MA, Tratschin JD. 2006. Classical swine fever virus replicon particles lacking the Erns gene: a potential marker vaccine for intradermal application. Vet Res 37:655–670. doi: 10.1051/vetres:2006028. [DOI] [PubMed] [Google Scholar]
- 57.Hulst MM, Himes G, Newbigin E, Moormann RJ. 1994. Glycoprotein E2 of classical swine fever virus: expression in insect cells and identification as a ribonuclease. Virology 200:558–565. doi: 10.1006/viro.1994.1218. [DOI] [PubMed] [Google Scholar]
- 58.Iqbal M, Poole E, Goodbourn S, McCauley JW. 2004. Role for bovine viral diarrhea virus Erns glycoprotein in the control of activation of beta interferon by double-stranded RNA. J Virol 78:136–145. doi: 10.1128/jvi.78.1.136-145.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mätzener P, Magkouras I, Rümenapf T, Peterhans E, Schweizer M. 2009. The viral RNase Erns prevents IFN type-I triggering by pestiviral single- and double-stranded RNAs. Virus Res 140:15–23. doi: 10.1016/j.virusres.2008.10.015. [DOI] [PubMed] [Google Scholar]
- 60.Roman-Sosa G, Brocchi E, Schirrmeier H, Wernike K, Schelp C, Beer M. 2016. Analysis of the humoral immune response against the envelope glycoprotein Gc of Schmallenberg virus reveals a domain located at the amino terminus targeted by mAbs with neutralizing activity. J Gen Virol 97:571–580. doi: 10.1099/jgv.0.000377. [DOI] [PubMed] [Google Scholar]
- 61.Wensvoort G. 1989. Topographical and functional mapping of epitopes on hog cholera virus with monoclonal antibodies. J Gen Virol 70:2865–2876. doi: 10.1099/0022-1317-70-11-2865. [DOI] [PubMed] [Google Scholar]
- 62.Du L, Yang Y, Zhou Y, Lu L, Li F, Jiang S. 2017. MERS-CoV spike protein: a key target for antivirals. Expert Opin Ther Targets 21:131–143. doi: 10.1080/14728222.2017.1271415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lee YM, Tscherne DM, Yun SI, Frolov I, Rice CM. 2005. Dual mechanisms of pestiviral superinfection exclusion at entry and RNA replication. J Virol 79:3231–3242. doi: 10.1128/JVI.79.6.3231-3242.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang CC, Tsai MH, Hu HS, Pu SY, Wu RH, Wu SH, Lin HM, Song JS, Chao YS, Yueh A. 2013. Characterization of an efficient dengue virus replicon for development of assays of discovery of small molecules against dengue virus. Antiviral Res 98:228–241. doi: 10.1016/j.antiviral.2013.03.001. [DOI] [PubMed] [Google Scholar]
- 65.Risager PC, Fahnøe U, Gullberg M, Rasmussen TB, Belsham GJ. 2013. Analysis of classical swine fever virus RNA replication determinants using replicons. J Gen Virol 94:1739–1748. doi: 10.1099/vir.0.052688-0. [DOI] [PubMed] [Google Scholar]
- 66.Wang Z, Nie Y, Wang P, Ding M, Deng H. 2004. Characterization of classical swine fever virus entry by using pseudotyped viruses: E1 and E2 are sufficient to mediate viral entry. Virology 330:332–341. doi: 10.1016/j.virol.2004.09.023. [DOI] [PubMed] [Google Scholar]
- 67.Widjojoatmodjo MN, van Gennip HG, Bouma A, van Rijn PA, Moormann RJ. 2000. Classical swine fever virus Erns deletion mutants: trans-complementation and potential use as nontransmissible, modified, live-attenuated marker vaccines. J Virol 74:2973–2980. doi: 10.1128/jvi.74.7.2973-2980.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Aberle D, Muhle-Goll C, Bürck J, Wolf M, Reißer S, Luy B, Wenzel W, Ulrich AS, Meyers G. 2014. Structure of the membrane anchor of pestivirus glycoprotein Erns, a long tilted amphipathic helix. PLoS Pathog 10:e1003973. doi: 10.1371/journal.ppat.1003973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Agapov EV, Frolov I, Lindenbach BD, Pragai BM, Schlesinger S, Rice CM. 1998. Noncytopathic Sindbis virus RNA vectors for heterologous gene expression. Proc Natl Acad Sci U S A 95:12989–12994. doi: 10.1073/pnas.95.22.12989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Varnavski AN, Khromykh AA. 1999. Noncytopathic flavivirus replicon RNA-based system for expression and delivery of heterologous genes. Virology 255:366–375. doi: 10.1006/viro.1998.9564. [DOI] [PubMed] [Google Scholar]
- 71.Johanning FW, Conry RM, LoBuglio AF, Wright M, Sumerel LA, Pike MJ, Curiel DT. 1995. A Sindbis virus mRNA polynucleotide vector achieves prolonged and high level heterologous gene expression in vivo. Nucleic Acids Res 23:1495–1501. doi: 10.1093/nar/23.9.1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lundstrom K. 2016. Replicon RNA viral vectors as vaccines. Vaccines 4:39. doi: 10.3390/vaccines4040039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Xiong C, Levis R, Shen P, Schlesinger S, Rice CM, Huang HV. 1989. Sindbis virus: an efficient, broad host range vector for gene expression in animal cells. Science 243:1188–1191. doi: 10.1126/science.2922607. [DOI] [PubMed] [Google Scholar]
- 74.Khromykh AA, Varnavski AN, Westaway EG. 1998. Encapsidation of the flavivirus Kunjin replicon RNA by using a complementation system providing Kunjin virus structural proteins in trans. J Virol 72:5967–5977. doi: 10.1128/JVI.72.7.5967-5977.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ryder AB, Nachbagauer R, Buonocore L, Palese P, Krammer F, Rose JK. 2015. Vaccination with vesicular stomatitis virus-vectored chimeric hemagglutinins protects mice against divergent influenza virus challenge strains. J Virol 90:2544–2550. doi: 10.1128/JVI.02598-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Anraku I, Mokhonov VV, Rattanasena P, Mokhonova EI, Leung J, Pijlman G, Cara A, Schroder WA, Khromykh AA, Suhrbier A. 2008. Kunjin replicon-based simian immunodeficiency virus gag vaccines. Vaccine 26:3268–3276. doi: 10.1016/j.vaccine.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Agnihothram S, Menachery VD, Yount BL Jr, Lindesmith LC, Scobey T, Whitmore A, Schafer A, Heise MT, Baric RS. 2018. Development of a broadly accessible Venezuelan equine encephalitis virus replicon particle vaccine platform. J Virol 92:e00027-18. doi: 10.1128/JVI.00027-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Geiser M, Cebe R, Drewello D, Schmitz R. 2001. Integration of PCR fragments at any specific site within cloning vectors without the use of restriction enzymes and DNA ligase. Biotechniques 31:88–92. doi: 10.2144/01311st05. [DOI] [PubMed] [Google Scholar]
- 79.Richter M, Reimann I, Wegelt A, Kirkland PD, Beer M. 2011. Complementation studies with the novel “Bungowannah” virus provide new insights in the compatibility of pestivirus proteins. Virology 418:113–122. doi: 10.1016/j.virol.2011.07.014. [DOI] [PubMed] [Google Scholar]
- 80.Reimann I, Depner K, Trapp S, Beer M. 2004. An avirulent chimeric pestivirus with altered cell tropism protects pigs against lethal infection with classical swine fever virus. Virology 322:143–157. doi: 10.1016/j.virol.2004.01.028. [DOI] [PubMed] [Google Scholar]
- 81.Yamanaka A, Moi ML, Takasaki T, Kurane I, Matsuda M, Suzuki R, Konishi E. 2017. Utility of Japanese encephalitis virus subgenomic replicon-based single-round infectious particles as antigens in neutralization tests for Zika virus and three other flaviviruses. J Virol Methods 243:164–171. doi: 10.1016/j.jviromet.2017.02.011. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All raw data are available upon request.







