Next Article in Journal
From Field Tests to Molecular Tools—Evaluating Diagnostic Tests to Improve Rabies Surveillance in Namibia
Next Article in Special Issue
Establishment of an In Vitro Model of Pseudorabies Virus Latency and Reactivation and Identification of Key Viral Latency-Associated Genes
Previous Article in Journal
Modulation of the Aryl Hydrocarbon Receptor Signaling Pathway Impacts on Junín Virus Replication
Previous Article in Special Issue
Cytopathic and Genomic Characteristics of a Human-Originated Pseudorabies Virus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recombinant Pseudorabies Virus Usage in Vaccine Development against Swine Infectious Disease

1
Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China
2
Viral Oncogenesis Group, The Pirbright Institute, Ash Road Pirbright, Woking, Surrey GU24 0NF, UK
*
Authors to whom correspondence should be addressed.
Viruses 2023, 15(2), 370; https://doi.org/10.3390/v15020370
Submission received: 30 December 2022 / Revised: 19 January 2023 / Accepted: 20 January 2023 / Published: 28 January 2023
(This article belongs to the Special Issue Pseudorabies Virus, Volume II)

Abstract

:
Pseudorabies virus (PRV) is the pathogen of pseudorabies (PR), which belongs to the alpha herpesvirus subfamily with a double stranded DNA genome encoding approximately 70 proteins. PRV has many non-essential regions for replication, has a strong capacity to accommodate foreign genes, and more areas for genetic modification. PRV is an ideal vaccine vector, and multivalent live virus-vectored vaccines can be developed using the gene-deleted PRV. The immune system continues to be stimulated by the gene-deleted PRVs and maintain a long immunity lasting more than 4 months. Here, we provide a brief overview of the biology of PRV, recombinant PRV construction methodology, the technology platform for efficiently constructing recombinant PRV, and the applications of recombinant PRV in vaccine development. This review summarizes the latest information on PRV usage in vaccine development against swine infectious diseases, and it offers novel perspectives for advancing preventive medicine through vaccinology.

1. Introduction

Pseudorabies virus (PRV) is the etiological agent of pseudorabies (PR), belonging to the Herpesviridae family, primarily affects pigs and can occasionally be transmitted to cattle, goats, sheep, cats, and dogs [1,2,3,4,5]. The PRV virus is a double-stranded DNA virus approximately 143 kb in size, consisting of unique long (UL), internal repeat short (IRS), unique short (US), and a terminal repeat short (TRS) [6,7,8]. There is a minimum of 70 open reading frames (ORFs) in the genome encoding 70–100 viral proteins, including virulence-related proteins and replicase, as well as many proteins that are not essential for PRV replication [9,10]. The large PRV genome allows the insertion of several kilobases (kb) foreign DNA sequences. The expression of foreign genes does not influence the virus replication [11]. Therefore, multivalent vaccines were developed using PRV as a vector, in which the major antigen of different swine pathogens was expressed [12].
Live vector vaccine can stimulate humoral immunity and solid cellular immunity, which is the main goal for developing a swine infectious disease vaccine. Scientists have generated PRV vaccine strains with few side effects and good immunity by inserting foreign genes or knocking out self-virulence genes. This review summarizes the biological characteristics of PRV, the methodological principle and technology platform for constructing characteristics of PRV, the methodological principle and technology platform for constructing PRV recombinants efficiently, and the applications of PRV recombinants in vaccine development. This review is intended to serve as a reference for PRV live vector vaccine research and development.

2. The Biological Characteristics of PRV

There are 10 glycoproteins encoded by PRV, which are divided into two groups (essential or non-essential glycoproteins) according to the degree to which viral replication depends on them [13,14]. The glycoprotein B (gB) plays a crucial role in membrane fusion during infection and in spreading viruses from cell to cell [15,16,17]. A major glycoprotein encoded by the glycoprotein C gene mediates the attachment of PRV to target cells through heparin-binding domains [11,18,19,20]. In susceptible animals, the protective immune responses were induced by gB, gC, and glycoprotein D (gD) [21,22,23]. In the viral envelope of infected cells, a heterodimer complex formed by glycoprotein I (gI) and glycoprotein E (gE) plays a crucial role in PR infection and transmission [24,25,26,27].
Bartha-K61, the original PRV vaccine strain, contains the deletion of US2, gE, gI, and US9 genes. This vaccine strain has been widely used to control PR worldwide. Since the 1990s, few swine PR cases have been reported due to PRV Bartha-K61 [4,28,29,30,31]. However, the PR outbreak occurred at the farm where pigs were vaccinated with Bartha-K61, which was caused by PRV variants and resulted in severe economic losses from 2011 [32,33,34,35,36,37,38]. Vaccines remain the most effective method to control PRV infection. Consequently, the PRV variant has been used to generate several gene mutant vaccines with gE, gI, and TK deletions. When challenged with PRV variants, these mutants provided adequate protection [39,40,41,42,43,44,45,46,47,48]. By introducing foreign genes derived from other porcine pathogens into the PRV variants and subsequent vaccination, it is possible to establish immune protection against these swine infectious diseases.

3. PRV Recombinants Construction

There are some non-essential regions in the large genome of PRV. As a result of this character, several kb of foreign DNA can be accommodated by PRV without compromising its stability. Reverse genetics plays a crucial role in PRV gene modification, and with the development of molecular biology techniques, many reverse genetic techniques have been used to construct the recombinant PRV.

3.1. Homologous Recombination

In the past, PRV recombinants were generated by homologous recombination (HR) in permissive cells. The construction of recombinant viruses based on the principle of HR can be divided into two steps: construction of a transfer plasmid and screening of the recombinant virus. Usually, the virus genome is used as a template to amplify the two gene fragments used as recombinant homologous arms by PCR technology, and then the foreign gene expression cassette is inserted between the homologous arms to construct the transfer vector containing the foreign gene expression cassette and the left and right homologous arms. Gene deletion or replacement is achieved by homologous recombination of the transfer vector and parent strain in eukaryotic susceptible cells. At present, there are two methods used for transfection. One method is co-transfection of the transfer vector and the parent virus genome, and another method is transfection of the transfer vector first and then inoculation of the virus. The reporter gene carried on the transfer vector is used to screen the plaque of the recombinant virus. Generally, the recombinant virus with infective activity can be obtained after 5–10 generations of screening. By using this method, Tong et al. and Wang et al., constructed a recombinant PRV strain expressing the E2 protein of CSFV in a gE-deleted PRV variant strain [49,50]; Yan et al. constructed the recombinant virus with additional expression of the gC gene [51]; Zheng et al. produced recombinant PRV that expressed VP2 protein of porcine parvovirus and porcine IL-6 [52]. Because of the low efficiency of HR, it is not commonly used in the construction of PRV recombinants.

3.2. Bacterial Artificial Chromosome

In comparison with homologous recombination, bacterial artificial chromosomes (BAC) are more efficient. Red/ET recombination technology can be used to modify BAC in Escherichia coli, which can be divided into two steps [53]. The first step is to construct an expression cassette with marker genes and both ends containing homologous arm sequences and transform them into competent cells containing viral BAC by electroporation. After resistance screening, correct recombinants were obtained. The second step was constructing an expression cassette with foreign genes and both ends containing homologous arm sequences. The recombinants with deleted marker genes were obtained by counter-resistance screening. Zhang et al. construct a TK/gE-deleted AH02LA BAC using the virulent PRV AH02LA strain and inserted the pig epidemic diarrhea virus (PEDV) variant spike (S) expression cassette in four different noncoding regions using the En Passant mutagenesis method [54]. Furthermore, to develop the recombinant vaccine against the PRV variants, Zhang et al. constructed a BAC clone of Bartha-K61. Using Bartha-K61 BAC as the backbone, the gD and gC of Bartha were replaced with that of PRV variants (AH02LA) via the En Passant method. When compared to the parental Bartha-K61, there was no difference in growth properties in the gD/gC-substituted Bartha-K61 ST cells [22].

3.3. Fosmid Library

The traditional HR method is not efficient in obtaining recombinant viruses. Creating recombinant BAC constructs is a labor-intensive and time-consuming process. When the whole genome PRV is cloned to the BAC plasmid, fragment deletion often occurs during the electroporation process, affecting recombination efficiency. In contrast, fosmid library generation is more efficient. The huge genome of PRV is randomly sheared into various fragments, then each segment is ligated with a fosmid vector to screen out the various combinations covering the whole genome of PRV, and the fosmid combination was co-transfected into virus-susceptible eukaryotic cells to rescue the virus [55,56]. Construction of recombinant virus only needs to modify the corresponding fosmid in E coli. Since the fosmid only contains part of the PRV genome, the efficiency of obtaining virus mutants is greatly improved. Modification of fosmid in E. coli can also utilize Red/ET recombination technology. A fosmid library based on the PRV-TJ strain was constructed by Zhou et al., and recombinant PRV was rescued by transfecting a group of fosmid directly into Vero cells. Furthermore, the green fluorescent protein-expressing reporter virus was rescued successfully by the Red/ET system [55]. Abid et al. constructed a PRV co-expressing E2 of classical swine fever virus (CSFV) and Cap of porcine circovirus type 2 (PCV2) by using this fosmid library platform [57]. In addition, Qi et al. constructed a fosmid library for the PRV-SC strain. The EGFP gene was inserted into the N-terminal of the UL 36 gene of PRV-SC via the Red/ET recombination [56].

3.4. CRISPR/Cas9

Molecular biotechnology has dramatically contributed to studying viruses’ replication and vaccines. The CRISPR /Cas9-mediated genome editing system has been widely applied as a powerful tool to construct PRV mutants. The researchers used CRISPR/Cas9 technology to construct PRV gE, gI, or TK gene-deleted recombinants [42,43,46]. Furthermore, CRISPR/Cas9 can be used in combination with HR and BAC technology to improve the efficiency of recombination [53,58]. Fu et al., used CRISPR/Cas9 gene-editing technology to construct a double reporter virus with stable expression of EGFP and firefly luciferase [59]. Feng et al. used this technology to insert the African swine fever virus (ASFV) CD2v gene into the TK, gE, and gI-deleted PRV Fa strain. The ASFV CD2v was stability expressed in the recombinant virus [12]. Wu et al. inserted the tandem repeats of the ORF2 gene into PRV gE and gG sites through CRISPR/Cas9 mediated HR and constructed a bivalent vaccine based on the PRV-HNX strain [60]. To optimize the method used for generating infectious recombinant PRV expressing predicted or known ASFV immunogenic proteins, Fuchs et al. developed an efficient strategy for the insertion of foreign genes in PRV vector based on marker-enforced recombination and CRISPR/Cas9. They co-transfected the defective PRV Bartha-K61 BAC DNA (possesses a deletion of glycoprotein G and an additional deletion encompassing the initiation codon and promoter of glycoprotein D, which plays an essential role in binding to receptors), the rescue transfer plasmid (A PRV-encoded transgene flanked by homologous recombination sequences and an intact gD promoter are included in this construct), and the CRISPR/Cas9 plasmid to the cell. Transgene substitutions were observed in 99% of the obtained progeny viruses [61]. Additionally, CRISPR/Cas9 and Cre/Lox greatly improved multi-gene editing efficiency and reduced vaccine development time. Yao et al. combined the Cre/Lox and CRISPR/Cas9-mediated gene insertion system, deleted the TK and gE gene, and inserted the immunoregulatory factor gene simultaneously [62].
Homologous recombination is the traditional method to generate the recombinant PRV. BAC was used later and allowed manipulation of the whole genome in Escherichia coli. The homologous recombination method is labor-intensive due to several rounds of plaque purification. The BAC system was more efficient than homologous recombination, but the generation of recombinant BAC construct is time-consuming, and the huge PRV genome is easily broken. The fosmid library and CRISPR/Cas9-based genetic manipulation platform for PRV offer several advantages over the conventional technology, but some disadvantages still appeared during the construction of the recombinants (Table 1). Therefore, we need to consider the advantages of various approaches and develop a more efficient way to construct the recombinant PRV.

4. Principle behind Recombinant PRV

The expression of foreign genes is an important index to evaluate the efficacy of the live recombinant vaccine, which is affected by the promoter and the insertion site of the foreign gene.

4.1. Promoter Affects Foreign Gene Expression

The initial transcription of foreign genes is the critical step of gene expression, and the rate of transcription initiation is the rate-limiting step of gene expression. The promoters and related regulatory sequences are the premises of constructing a good expression system. The regulation elements of exogenous gene expression include a promoter, enhancer, Kozak sequence, and stop codon. The expression level of the foreign gene in PRV mainly depends on the strength of the upstream promoter, and a generally strong promoter is selected. The gG and gE promoters were always chosen to construct recombinant PRV, which have unique structures and vigorous activity [57,60]. A more potent heterologous enhancer or promoter, such as the HCMV or MCMV promoter, was also evaluated instead of the gG promoter [50,54]. Different promoters were tested to express the ASFV gene in PRV, such as HCMV, MCMV, and a chimeric promoter (CAG) consisting of the HCMV enhancer and the chicken beta-actin promoter [61]. Compared with cytomegalovirus immediate-early promoters, the CAG promoter enhanced the expression of foreign genes in PRV.

4.2. Insertion Site Is the Main Factor, Affects the Expression of Foreign Genes

For a live vector vaccine strain, genetic stability is crucial. The PRV genome contains numerous sites for inserting and expressing heterologous genes, but almost all are located near the gI/gE, TK, or gG genes. In a study by Tong et al., the gD and gG genes were inserted with E2 expression cassettes, and insertion of the cassettes did not significantly affect in vitro replication of the recombinant virus. And the piglets challenged with virulent PRV are protected by recombinant viruses [50]. Wu et al. also inserted the tandem repeats of the PCV2 Cap protein gene into PRV gG and gE sites. A high level of PCV2 Cap protein expression was detected in this recombinant PRV [60]. Intergenic regions between gG and gD proved to be excellent sites for expressing foreign genes, according to these results. To identify suitable areas to insert foreign genes into PRV genomes, Zhang et al. inserted an S protein expression cassette of a PEDV variant in different noncoding regions of the PRV genome with deletion of TK, gE, and gI, such as US2-1, UL11-10, UL46-27, and UL35-36. The US2-1 area is critical for PRV replication, as the insertion of a foreign gene failed to rescue the virus, another three noncoding regions are suitable sites for the insertion of the S gene; S gene mRNA expression was higher when it was inserted in the location of UL11-10 [50,54]. Therefore, the UL11-10 is a new suitable insertion site for S gene expression.
So far, when selecting the insertion sites of foreign genes, researchers have considered the non-essential regions of virus replication first. Although the genome sequence of PRV contains a large number of non-essential regions for replication, the basic theories of the relationship between these regions and virus virulence, immune escape mechanism, or host range need to be further studied. At the same time, the expression of exogenous genes in the vector is affected by the size of the exogenous fragment, the promoter of the expression vector, and the insertion site effect caused by the insertion of exogenous genes. Therefore, a certain size of a foreign gene, stable insertion sites of a foreign gene, and safe and efficient expression elements are the basis for the application of recombinant live vector vaccines.

5. Applications of Recombinant PRV

The large genome of PRV contains numerous insertion sites that allow heterologous genes to be integrated and expressed. Consequently, PRV has developed into a powerful vector system for expressing foreign proteins.

5.1. PRV Bartha K61 Strain as a Vector for Expressing Exogenous Antigens

In pigs, PRV Bartha K61 replicates reliably and elicits a wide range of humoral and cellular immune responses. Additionally, in order to establish clinical protection against different pathogens, foreign antigens can be introduced into the Bartha K61 backbone and vaccinated. By introducing the GP5 gene from the porcine reproductive and respiratory syndrome virus (PRRSV) into Bartha K61, and subsequently vaccinating, significant clinical protection was achieved, and pathological lesions were reduced after the PRRSV challenge in piglets [63]. Further, mice were protected against virulent challenges with swine influenza H3N2 by vaccination with a Bartha vector that expressed the hemagglutinin of swine influenza H3N2 [64]. Furthermore, pigs vaccinated with Bartha vectors expressing the pandemic H1N1 swine-origin influenza virus hemagglutinin or neuraminidase also demonstrated significant inhibition of virus replication after a challenge with the H1N1 virus [65]. Due to the limitations of Bartha K61 in protecting against PRV variants, Zhang et al. constructed the gD/gC-substituted Bartha K61, in which the gD/gC of Bartha K61 was substituted with the gD/gC of PRV variants (AH02LA). The gD/gC-substituted Bartha K61 was safe, and it effectively protected against virulent PRV variants [22]. Therefore, Bartha K61 was a safe and effective backbone for multivalent vaccine development.

5.2. Attenuated PRV Variant as a Vector for Expressing Exogenous Antigens

Since 2011, farms immunized with Bartha-K61 have experienced PR outbreaks caused by PRV variants. Therefore, the researchers try to generate several gene mutant vaccines involving gE/TK, gI/gE, and gI/gE/TK deletion based on different PRV variants. These vaccines provided adequate protection against the PRV variant challenge. These gene mutant vaccines also are used as a vector system for foreign proteins to develop multivalent live virus-vectored vaccines against several swine infectious diseases. Tong et al. and Wang et al. constructed a recombinant expressing the CSFV E2 protein and evaluated its efficacy against both CSFV and PRV variant strains [49,50]. The recombinant PRV appears to be a promising recombinant vaccine candidate for the control and eradication of PRV variants and CSFV. Through the fosmid library platform, Abid et al. constructed a recombinant co-expressing E2 of CSFV and Cap of PCV2 with gE/gI/TK gene deletions [57]. The recombinant strain was safe for pigs and rabbits, and anti-PRV antibodies could be detected in the serum of immunized rabbits and pigs. But anti- E2 and PCV2 antibodies could not be detected, which was presumed to be because the expression level of E2 and Cap proteins are too low to induce antibody production. In addition, a tandem repeat of PCV2 Cap protein gene ORF2 that links with a protein quantitation rationing linker was constructed by Wu et al., and endogenous PRV promoters were used to drive PCV2 Cap2 expression in PRV [60]. High levels of neutralizing antibodies were detected 14 days after immunization of the recombinant bivalent vaccine candidate, which was maintained at days 21, 42, and 60. In wild boars and domestic pigs, the African swine fever virus (ASFV) causes a fatal disease, and there is no effective vaccine against the ASFV. In order to develop the live vector vaccine against ASFV. Feng et al. insert the CD2v into the PRV variant Fa stain (TK, gE, and gI deleted) and obtained a recombinant strain expressing CD2v gene [12]. The recombinant virus stimulated the production of anti-CD2v antibodies and specific cellular immune responses. Furthermore, the recombinant virus could protect mice against the virulent strain (PRV-Fa) infection.

5.3. Attenuated PRV Variant as a Vector for Expressing Immunoregulatory Factors

Lymphokine-activated killer cells recognize infected cells associated with PRV gC during cell-mediated immunity, and PRV gC plays a role in cytotoxic T-lymphocytes recognition of infected cells [22,66,67]. To enhance gC expression, Yan et al. inserted an additional expression cassette of the gC gene into the gI and gE deletion regions [51]. Additional gC gene expression enhanced the efficacy of the PRV gI/gE-deleted vaccine in pigs. A hematopoietic growth factor, Fms-related tyrosine kinase 3 ligand (Flt3L), plays an important role in hematopoietic cell differentiation. In the periphery, FLT3L and its receptor regulate homeostatic DC division [68,69]. An FLT3L-dependent pathway mediates vaccine-induced protective immunity and enhances the activation of T cells [70]. Based on the immunomodulatory functions of Flt3L, a TK/gE gene deleted and Flt3L co-expressed recombinant PRV was constructed by Yao et al. [62]. The recombinant gene showed potential function in DC activation and protective immune response enhancement. These results indicated that t Flt3L is an ideal adjuvant, and the PRV can load the immunoregulatory factors and achieve simultaneous immunization with the adjuvant and vaccine.

5.4. Attenuated PRV Variant as Vector for Expressing Reporter Genes

Up to now, the mechanism of PRV variant virulence enhancement is unknown. A reporter virus is a valuable tool for basic virology studies. Therefore, Fu et al. constructed a double reporter virus with stable expression of EGFP and firefly luciferase based on the PRV variant [59]. The recombinant virus showed similar biological characteristics to the parental strain, including a stable viral titer and luciferase activity throughout 20 passages. Furthermore, Zhou and Qi constructed the reporter virus based on the PRV variant and classical stain, respectively [55,56]. The EGFP was fused into the amino-terminal of UL36 in these recombinants. The single viral particles with green fluorescence were used to monitor retrograde and anterograde moving virions in the axon, these reported viruses will accelerate the understanding of the biology of the PRV variant.

6. Conclusions and Prospects

The genetically engineered live vector vaccine can effectively avoid the traditional vaccines’ defects, simplify the breeding industry’s immunization procedures, and improve the efficiency of epidemic prevention and control. Attenuated PRV-based vector vaccines do not induce any clinical signs of PR, but provide complete protection against PRV and other swine pathogens, and it has been widely used in the study of a recombinant multivalent vaccine against swine infectious disease. The BAC technology, CRISPR/Cas9 technology, and fosmid library were used to construct PRV recombinant, significantly improving the efficiency of exogenous gene expression (Table 2). But we still need to consider how to ensure the genetic stability of the immune genes, and how to ensure the safety of recombinant viruses during live vector vaccine development. The significant virulence genes were deleted during the development of the recombinant PRV live vector vaccine, significantly reducing its virulence to animals, However, the safety of the recombinants still needs to be analyzed in detail. There are extensive replication non-essential regions in the PRV genome, which provide a basis for optimizing recombinant sites. The selection of better insertion sites and safer and more efficient expression elements, especially an efficient promoter, is the basis of recombinant live vector vaccine application. Further research and utilization of the advantages of recombinant PRV live vector vaccine will play an essential role in the prevention and control of animal diseases. The recombinant herpes virus live vector vaccine will play an important role in animal disease prevention and control in the future.

Author Contributions

M.Z. and S.C. are the major contributors to the review, M.Z., S.C. and S.Z. designed the concept and prepare the original draft of the review; S.Z. and M.A. conceived and revised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a grant from the key project of Jiangsu Province’s Key Research and Development Plan (modern Agriculture) (BE2020407), the project of Jiangsu Agri-animal Husbandry Vocational College (NSF2022CB04, NSF2022CB25), the Natural Science Research Project of Higher Education of Jiangsu Province (2020220375), and the Qing Lan Project of Jiangsu Province.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the insert article.

Acknowledgments

The authors would like to thank members of the Division of Swine Infectious disease prevention and control of Jiangsu Agri-animal Husbandry Vocational College for valuable discussion.

Conflicts of Interest

The authors declare that this work was conducted in the absence of any commercial or financial relationships that could be as a potential conflict of interest.

References

  1. Hu, R.M.; Zhou, Q.; Song, W.B.; Sun, E.C.; Zhang, M.M.; He, Q.G.; Chen, H.C.; Wu, B.; Liu, Z.F. Novel pseudorabies virus variant with defects in TK, gE and gI protects growing pigs against lethal challenge. Vaccine 2015, 33, 5733–5740. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, X.; Tong, W.; Song, X.; Jia, R.; Li, L.; Zou, Y.; He, C.; Liang, X.; Lv, C.; Jing, B.; et al. Antiviral Effect of Resveratrol in Piglets Infected with Virulent Pseudorabies Virus. Viruses 2018, 10, 457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yao, J.; Li, J.; Gao, L.; He, Y.; Xie, J.; Zhu, P.; Zhang, Y.; Zhang, X.; Duan, L.; Yang, S.; et al. Epidemiological Investigation and Genetic Analysis of Pseudorabies Virus in Yunnan Province of China from 2017 to 2021. Viruses 2022, 14, 895. [Google Scholar] [CrossRef] [PubMed]
  4. Zheng, H.H.; Fu, P.F.; Chen, H.Y.; Wang, Z.Y. Pseudorabies Virus: From Pathogenesis to Prevention Strategies. Viruses 2022, 14, 1638. [Google Scholar] [CrossRef] [PubMed]
  5. Ye, C.; Wu, J.; Tong, W.; Shan, T.; Cheng, X.; Xu, J.; Liang, C.; Zheng, H.; Li, G.; Tong, G. Comparative genomic analyses of a virulent pseudorabies virus and a series of its in vitro passaged strains. Virol. J. 2018, 15, 195. [Google Scholar] [CrossRef] [PubMed]
  6. Pomeranz, L.E.; Ekstrand, M.I.; Latcha, K.N.; Smith, G.A.; Enquist, L.W.; Friedman, J.M. Gene Expression Profiling with Cre-Conditional Pseudorabies Virus Reveals a Subset of Midbrain Neurons That Participate in Reward Circuitry. J. Neurosci. 2017, 37, 4128–4144. [Google Scholar] [CrossRef] [Green Version]
  7. Yin, Y.; Romero, N.; Favoreel, H.W. Pseudorabies Virus Inhibits Type I and Type III Interferon-Induced Signaling via Proteasomal Degradation of Janus Kinases. J. Virol. 2021, 95, e0079321. [Google Scholar] [CrossRef]
  8. Yin, Y.; Ma, J.; Van Waesberghe, C.; Devriendt, B.; Favoreel, H.W. Pseudorabies virus-induced expression and antiviral activity of type I or type III interferon depend on the type of infected epithelial cell. Front. Immunol. 2022, 13, 1016982. [Google Scholar] [CrossRef]
  9. Wang, Y.P.; Huang, L.P.; Du, W.J.; Wei, Y.W.; Xia, D.L.; Wu, H.L.; Feng, L.; Liu, C.M. The pseudorabies virus DNA polymerase processivity factor UL42 exists as a monomer in vitro and in vivo. Arch. Virol. 2016, 161, 1027–1031. [Google Scholar] [CrossRef]
  10. Xu, C.; Wang, M.; Song, Z.; Wang, Z.; Liu, Q.; Jiang, P.; Bai, J.; Li, Y.; Wang, X. Pseudorabies virus induces autophagy to enhance viral replication in mouse neuro-2a cells in vitro. Virus. Res. 2018, 248, 44–52. [Google Scholar] [CrossRef]
  11. Thomsen, D.R.; Marotti, K.R.; Palermo, D.P.; Post, L.E. Pseudorabies virus as a live virus vector for expression of foreign genes. Gene 1987, 57, 261–265. [Google Scholar] [CrossRef] [PubMed]
  12. Feng, Z.; Chen, J.; Liang, W.; Chen, W.; Li, Z.; Chen, Q.; Cai, S. The recombinant pseudorabies virus expressing African swine fever virus CD2v protein is safe and effective in mice. Virol. J. 2020, 17, 180. [Google Scholar] [CrossRef] [PubMed]
  13. Zhao, W.L.; Wu, Y.H.; Li, H.F.; Li, S.Y.; Fan, S.Y.; Wu, H.L.; Li, Y.J.; Lü, Y.L.; Han, J.; Zhang, W.C.; et al. Clinical experience and next-generation sequencing analysis of encephalitis caused by pseudorabies virus. Zhonghua Yi Xue Za Zhi 2018, 98, 1152–1157. [Google Scholar]
  14. Zhao, N.; Wang, F.; Kong, Z.; Shang, Y. Pseudorabies Virus Tegument Protein UL13 Suppresses RLR-Mediated Antiviral Innate Immunity through Regulating Receptor Transcription. Viruses 2022, 14, 1465. [Google Scholar] [CrossRef] [PubMed]
  15. Dory, D.; Rémond, M.; Béven, V.; Cariolet, R.; Backovic, M.; Zientara, S.; Jestin, A. Pseudorabies virus glycoprotein B can be used to carry foot and mouth disease antigens in DNA vaccination of pigs. Antivir. Res. 2009, 81, 217–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Li, X.; Yang, F.; Hu, X.; Tan, F.; Qi, J.; Peng, R.; Wang, M.; Chai, Y.; Hao, L.; Deng, J.; et al. Two classes of protective antibodies against Pseudorabies virus variant glycoprotein B: Implications for vaccine design. PLoS Pathog. 2017, 13, e1006777. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, M.; Wang, M.H.; Shen, X.G.; Liu, H.; Zhang, Y.Y.; Peng, J.M.; Meng, F.; Wang, T.Y.; Bai, Y.Z.; Sun, M.X.; et al. Neuropilin-1 Facilitates Pseudorabies Virus Replication and Viral Glycoprotein B Promotes Its Degradation in a Furin-Dependent Manner. J. Virol. 2022, 96, e0131822. [Google Scholar] [CrossRef]
  18. Trybala, E.; Bergström, T.; Spillmann, D.; Svennerholm, B.; Flynn, S.J.; Ryan, P. Interaction between pseudorabies virus and heparin/heparan sulfate. Pseudorabies virus mutants differ in their interaction with heparin/heparan sulfate when altered for specific glycoprotein C heparin-binding domain. J. Biol. Chem. 1998, 273, 5047–5052. [Google Scholar] [CrossRef] [Green Version]
  19. Tong, T.; Fan, H.; Tan, Y.; Xiao, S.; Ling, J.; Chen, H.; Guo, A. C3d enhanced DNA vaccination induced humoral immune response to glycoprotein C of pseudorabies virus. Biochem. Biophys. Res. Commun. 2006, 347, 845–851. [Google Scholar] [CrossRef]
  20. Fan, H.; Liu, Z.; Tong, T.; Liu, X.; Guo, A. C3d-M28 enhanced DNA vaccination induced humoral immune response to glycoprotein C of pseudorabies virus. Sheng Wu Gong Cheng Xue Bao 2009, 25, 987–992. [Google Scholar]
  21. Li, A.; Lu, G.; Qi, J.; Wu, L.; Tian, K.; Luo, T.; Shi, Y.; Yan, J.; Gao, G.F. Structural basis of nectin-1 recognition by pseudorabies virus glycoprotein D. PLoS Pathog. 2017, 13, e1006314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zhang, C.; Liu, Y.; Chen, S.; Qiao, Y.; Guo, M.; Zheng, Y.; Xu, M.; Wang, Z.; Hou, J.; Wang, J. A gD&gC-substituted pseudorabies virus vaccine strain provides complete clinical protection and is helpful to prevent virus shedding against challenge by a Chinese pseudorabies variant. BMC Vet. Res. 2019, 15, 2. [Google Scholar]
  23. Zhang, T.; Liu, Y.; Chen, Y.; Wang, A.; Feng, H.; Wei, Q.; Zhou, E.; Zhang, G. A single dose glycoprotein D-based subunit vaccine against pseudorabies virus infection. Vaccine 2020, 38, 6153–6161. [Google Scholar] [CrossRef]
  24. Tyborowska, J.; Reszka, N.; Kochan, G.; Szewczyk, B. Formation of Pseudorabies virus glycoprotein E/I complex in baculovirus recombinant system. Acta Virol. 2006, 50, 169–174. [Google Scholar] [PubMed]
  25. Lu, S.; Xiang, M.; Dan, H.; Wu, B.; Gao, Q.; Huang, H.; He, Q.; Chen, H. Immune-tolerizing procedure for preparation of monoclonal antibodies against glycoprotein E of Pseudorabies virus. Monoclon. Antib. Immunodiagn. Immunother. 2013, 32, 21–25. [Google Scholar] [CrossRef] [Green Version]
  26. Wu, C.Y.; Liao, C.M.; Chi, J.N.; Chien, M.S.; Huang, C. Growth properties and vaccine efficacy of recombinant pseudorabies virus defective in glycoprotein E and thymidine kinase genes. J. Biotechnol. 2016, 229, 58–64. [Google Scholar] [CrossRef] [PubMed]
  27. Xu, J.J.; Wu, J.Q.; Cheng, X.F.; Tong, W.; Zheng, H.; Zhu, H.J.; Liu, Y.T.; Jiang, Y.F.; Gao, F.; Yu, H.; et al. Identification of two novel epitopes targeting glycoprotein E of pseudorabies virus using monoclonal antibodies. Biochem. Biophys. Res. Commun. 2019, 519, 330–336. [Google Scholar] [CrossRef] [PubMed]
  28. Freuling, C.M.; Müller, T.F.; Mettenleiter, T.C. Vaccines against pseudorabies virus (PrV). Vet. Microbiol. 2017, 206, 3–9. [Google Scholar] [CrossRef]
  29. Delva, J.L.; Nauwynck, H.J.; Mettenleiter, T.C.; Favoreel, H.W. The Attenuated Pseudorabies Virus Vaccine Strain Bartha K61: A Brief Review on the Knowledge Gathered During 60 Years of Research. Pathogens 2020, 9, 897. [Google Scholar] [CrossRef]
  30. Mettenleiter, T.C. Aujeszky Disease and the Development of the Marker/DIVA Vaccination Concept. Pathogens 2020, 9, 563. [Google Scholar] [CrossRef]
  31. Zhou, H.; Pan, Y.; Liu, M.; Han, Z. Prevalence of Porcine Pseudorabies Virus and Its Coinfection Rate in Heilongjiang Province in China from 2013 to 2018. Viral Immunol. 2020, 33, 550–554. [Google Scholar] [CrossRef] [PubMed]
  32. Zheng, H.H.; Bai, Y.L.; Xu, T.; Zheng, L.L.; Li, X.S.; Chen, H.Y.; Wang, Z.Y. Isolation and Phylogenetic Analysis of Reemerging Pseudorabies Virus Within Pig Populations in Central China During 2012 to 2019. Front. Vet. Sci. 2021, 8, 764982. [Google Scholar] [CrossRef] [PubMed]
  33. Zheng, H.H.; Jin, Y.; Hou, C.Y.; Li, X.S.; Zhao, L.; Wang, Z.Y.; Chen, H.Y. Seroprevalence investigation and genetic analysis of pseudorabies virus within pig populations in Henan province of China during 2018–2019. Infect. Genet. Evol. 2021, 92, 104835. [Google Scholar] [CrossRef] [PubMed]
  34. Fan, J.; Zeng, X.; Zhang, G.; Wu, Q.; Niu, J.; Sun, B.; Xie, Q.; Ma, J. Molecular characterization and phylogenetic analysis of pseudorabies virus variants isolated from Guangdong province of southern China during 2013–2014. J. Vet. Sci. 2016, 17, 369–375. [Google Scholar] [CrossRef]
  35. Wang, X.; Wu, C.X.; Song, X.R.; Chen, H.C.; Liu, Z.F. Comparison of pseudorabies virus China reference strain with emerging variants reveals independent virus evolution within specific geographic regions. Virology 2017, 506, 92–98. [Google Scholar] [CrossRef]
  36. Zhai, X.; Zhao, W.; Li, K.; Zhang, C.; Wang, C.; Su, S.; Zhou, J.; Lei, J.; Xing, G.; Sun, H.; et al. Genome Characteristics and Evolution of Pseudorabies Virus Strains in Eastern China from 2017 to 2019. Virol. Sin. 2019, 34, 601–609. [Google Scholar] [CrossRef]
  37. Liu, J.; Chen, C.; Li, X. Novel Chinese pseudorabies virus variants undergo extensive recombination and rapid interspecies transmission. Transbound. Emerg. Dis. 2020, 67, 2274–2276. [Google Scholar] [CrossRef]
  38. Bo, Z.; Li, X. A Review of Pseudorabies Virus Variants: Genomics, Vaccination, Transmission, and Zoonotic Potential. Viruses 2022, 14, 1003. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, T.; Xiao, Y.; Yang, Q.; Wang, Y.; Sun, Z.; Zhang, C.; Yan, S.; Wang, J.; Guo, L.; Yan, H.; et al. Construction of a gE-Deleted Pseudorabies Virus and Its Efficacy to the New-Emerging Variant PRV Challenge in the Form of Killed Vaccine. Biomed. Res. Int. 2015, 2015, 684945. [Google Scholar]
  40. Wang, J.; Guo, R.; Qiao, Y.; Xu, M.; Wang, Z.; Liu, Y.; Gu, Y.; Liu, C.; Hou, J. An inactivated gE-deleted pseudorabies vaccine provides complete clinical protection and reduces virus shedding against challenge by a Chinese pseudorabies variant. BMC Vet. Res. 2016, 12, 277. [Google Scholar] [CrossRef] [Green Version]
  41. Yin, Y.; Xu, Z.; Liu, X.; Li, P.; Yang, F.; Zhao, J.; Fan, Y.; Sun, X.; Zhu, L. A live gI/gE-deleted pseudorabies virus (PRV) protects weaned piglets against lethal variant PRV challenge. Virus. Genes. 2017, 53, 565–572. [Google Scholar] [CrossRef] [PubMed]
  42. Li, J.; Fang, K.; Rong, Z.; Li, X.; Ren, X.; Ma, H.; Chen, H.; Li, X.; Qian, P. Comparison of gE/gI- and TK/gE/gI-Gene-Deleted Pseudorabies Virus Vaccines Mediated by CRISPR/Cas9 and Cre/Lox Systems. Viruses 2020, 12, 369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Lin, J.; Li, Z.; Feng, Z.; Fang, Z.; Chen, J.; Chen, W.; Liang, W.; Chen, Q. Pseudorabies virus (PRV) strain with defects in gE, gC, and TK genes protects piglets against an emerging PRV variant. J. Vet. Med. Sci. 2020, 82, 846–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yin, H.; Li, Z.; Zhang, J.; Huang, J.; Kang, H.; Tian, J.; Qu, L. Construction of a US7/US8/UL23/US3-deleted recombinant pseudorabies virus and evaluation of its pathogenicity in dogs. Vet. Microbiol. 2020, 240, 108543. [Google Scholar] [CrossRef]
  45. Zhao, Y.; Wang, L.Q.; Zheng, H.H.; Yang, Y.R.; Liu, F.; Zheng, L.L.; Jin, Y.; Chen, H.Y. Construction and immunogenicity of a gE/gI/TK-deleted PRV based on porcine pseudorabies virus variant. Mol. Cell Probes. 2020, 53, 101605. [Google Scholar] [CrossRef]
  46. Lv, L.; Liu, X.; Jiang, C.; Wang, X.; Cao, M.; Bai, J.; Jiang, P. Pathogenicity and immunogenicity of a gI/gE/TK/UL13-gene-deleted variant pseudorabies virus strain in swine. Vet. Microbiol. 2021, 258, 109104. [Google Scholar] [CrossRef]
  47. Xu, L.; Wei, J.F.; Zhao, J.; Xu, S.Y.; Lee, F.Q.; Nie, M.C.; Xu, Z.W.; Zhou, Y.C.; Zhu, L. The Immunity Protection of Central Nervous System Induced by Pseudorabies Virus DelgI/gE/TK in Mice. Front. Microbiol. 2022, 13, 862907. [Google Scholar] [CrossRef]
  48. Zhao, J.; Zhu, L.; Xu, L.; Li, F.; Deng, H.; Huang, Y.; Gu, S.; Sun, X.; Zhou, Y.; Xu, Z. The Construction and Immunogenicity Analyses of Recombinant Pseudorabies Virus with NADC30-Like Porcine Reproductive and Respiratory Syndrome Virus-Like Particles Co-expression. Front. Microbiol. 2022, 13, 846079. [Google Scholar] [CrossRef]
  49. Wang, Y.; Yuan, J.; Cong, X.; Qin, H.Y.; Wang, C.H.; Li, Y.; Li, S.; Luo, Y.; Sun, Y.; Qiu, H.J. Generation and Efficacy Evaluation of a Recombinant Pseudorabies Virus Variant Expressing the E2 Protein of Classical Swine Fever Virus in Pigs. Clin. Vaccine Immunol. 2015, 22, 1121–1129. [Google Scholar] [CrossRef] [Green Version]
  50. Tong, W.; Zheng, H.; Li, G.X.; Gao, F.; Shan, T.L.; Zhou, Y.J.; Yu, H.; Jiang, Y.F.; Yu, L.X.; Li, L.W.; et al. Recombinant pseudorabies virus expressing E2 of classical swine fever virus (CSFV) protects against both virulent pseudorabies virus and CSFV. Antiviral Res. 2020, 173, 104652. [Google Scholar] [CrossRef]
  51. Yan, Z.; Chen, M.; Tang, D.; Wu, X.; Ren, X.; Pan, H.; Li, Y.; Ji, Q.; Luo, Y.; Fan, H.; et al. Better immune efficacy triggered by the inactivated gI/gE-deleted pseudorabies virus with the additional insertion of gC gene in mice and weaned pigs. Virus. Res. 2021, 296, 198353. [Google Scholar] [CrossRef] [PubMed]
  52. Zheng, H.H.; Wang, L.Q.; Fu, P.F.; Zheng, L.L.; Chen, H.Y.; Liu, F. Characterization of a recombinant pseudorabies virus expressing porcine parvovirus VP2 protein and porcine IL-6. Virol. J. 2020, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  53. Zheng, K.; Jiang, F.F.; Su, L.; Wang, X.; Chen, Y.X.; Chen, H.C.; Liu, Z.F. Highly Efficient Base Editing in Viral Genome Based on Bacterial Artificial Chromosome Using a Cas9-Cytidine Deaminase Fused Protein. Virol. Sin. 2020, 35, 191–199. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, C.; Guo, S.; Guo, R.; Chen, S.; Zheng, Y.; Xu, M.; Wang, Z.; Liu, Y.; Wang, J. Identification of four insertion sites for foreign genes in a pseudorabies virus vector. BMC Vet. Res. 2021, 17, 190. [Google Scholar] [CrossRef]
  55. Zhou, M.; Abid, M.; Yin, H.; Wu, H.; Teklue, T.; Qiu, H.J.; Sun, Y. Establishment of an Efficient and Flexible Genetic Manipulation Platform Based on a Fosmid Library for Rapid Generation of Recombinant Pseudorabies Virus. Front. Microbiol. 2018, 9, 2132. [Google Scholar] [CrossRef]
  56. Qi, H.; Wu, H.; Abid, M.; Qiu, H.J.; Sun, Y. Establishment of a Fosmid Library for Pseudorabies Virus SC Strain and Application in Viral Neuronal Tracing. Front. Microbiol. 2020, 11, 1168. [Google Scholar] [CrossRef]
  57. Abid, M.; Teklue, T.; Li, Y.; Wu, H.; Wang, T.; Qiu, H.J.; Sun, Y. Generation and Immunogenicity of a Recombinant Pseudorabies Virus Co-Expressing Classical Swine Fever Virus E2 Protein and Porcine Circovirus Type 2 Capsid Protein Based on Fosmid Library Platform. Pathogens 2019, 8, 279. [Google Scholar] [CrossRef] [Green Version]
  58. Guo, J.C.; Tang, Y.D.; Zhao, K.; Wang, T.Y.; Liu, J.T.; Gao, J.C.; Chang, X.B.; Cui, H.Y.; Tian, Z.J.; Cai, X.H.; et al. Highly Efficient CRISPR/Cas9-Mediated Homologous Recombination Promotes the Rapid Generation of Bacterial Artificial Chromosomes of Pseudorabies Virus. Front. Microbiol. 2016, 7, 2110. [Google Scholar] [CrossRef] [Green Version]
  59. Fu, P.F.; Cheng, X.; Su, B.Q.; Duan, L.F.; Wang, C.R.; Niu, X.R.; Wang, J.; Yang, G.Y.; Chu, B.B. CRISPR/Cas9-based generation of a recombinant double-reporter pseudorabies virus and its characterization in vitro and in vivo. Vet. Res. 2021, 52, 95. [Google Scholar] [CrossRef]
  60. Wu, X.; Wu, H.; Wang, H.; Luo, L.; Wang, J.; Wu, B.; He, Q.; Cao, G.; Lei, Y.; Chen, X.; et al. A new strategy to develop pseudorabies virus-based bivalent vaccine with high immunogenicity of porcine circovirus type 2. Vet. Microbiol. 2021, 255, 109022. [Google Scholar] [CrossRef]
  61. Hübner, A.; Keil, G.M.; Kabuuka, T.; Mettenleiter, T.C.; Fuchs, W. Efficient transgene insertion in a pseudorabies virus vector by CRISPR/Cas9 and marker rescue-enforced recombination. J. Virol. Methods 2018, 262, 38–47. [Google Scholar] [CrossRef] [PubMed]
  62. Yao, L.; Hu, Q.; Chen, S.; Zhou, T.; Yu, X.; Ma, H.; H Ghonaim, A.; Wu, H.; Sun, Q.; Fan, S.; et al. Recombinant Pseudorabies Virus with TK/gE Gene Deletion and Flt3L Co-Expression Enhances the Innate and Adaptive Immune Response via Activating Dendritic Cells. Viruses 2021, 13, 691. [Google Scholar] [CrossRef] [PubMed]
  63. Qiu, H.J.; Tian, Z.J.; Tong, G.Z.; Zhou, Y.J.; Ni, J.Q.; Luo, Y.Z.; Cai, X.H. Protective immunity induced by a recombinant pseudorabies virus expressing the GP5 of porcine reproductive and respiratory syndrome virus in piglets. Vet. Immunol. Immunopathol. 2005, 106, 309–319. [Google Scholar] [CrossRef] [PubMed]
  64. Tian, Z.J.; Zhou, G.H.; Zheng, B.L.; Qiu, H.J.; Ni, J.Q.; Yang, H.L.; Yin, X.N.; Hu, S.P.; Tong, G.Z. A recombinant pseudorabies virus encoding the HA gene from H3N2 subtype swine influenza virus protects mice from virulent challenge. Vet. Immunol. Immunopathol. 2006, 111, 211–218. [Google Scholar] [CrossRef] [PubMed]
  65. Klingbeil, K.; Lange, E.; Teifke, J.P.; Mettenleiter, T.C.; Fuchs, W. Immunization of pigs with an attenuated pseudorabies virus recombinant expressing the haemagglutinin of pandemic swine origin H1N1 influenza A virus. J. Gen. Virol. 2014, 95, 948–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Flynn, S.J.; Ryan, P. The receptor-binding domain of pseudorabies virus glycoprotein gC is composed of multiple discrete units that are functionally redundant. J. Virol. 1996, 70, 1355–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zhang, P.; Lv, L.; Sun, H.; Li, S.; Fan, H.; Wang, X.; Bai, J.; Jiang, P. Identification of linear B cell epitope on gB, gC, and gE proteins of porcine pseudorabies virus using monoclonal antibodies. Vet. Microbiol. 2019, 234, 83–91. [Google Scholar] [CrossRef]
  68. Waskow, C.; Liu, K.; Darrasse-Jèze, G.; Guermonprez, P.; Ginhoux, F.; Merad, M.; Shengelia, T.; Yao, K.; Nussenzweig, M. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat. Immunol. 2008, 9, 676–683. [Google Scholar] [CrossRef]
  69. Yuan, X.; Qin, X.; Wang, D.; Zhang, Z.; Tang, X.; Gao, X.; Chen, W.; Sun, L. Mesenchymal stem cell therapy induces FLT3L and CD1c (+) dendritic cells in systemic lupus erythematosus patients. Nat. Commun. 2019, 10, 2498. [Google Scholar] [CrossRef] [Green Version]
  70. Wen, Y.; Wang, H.; Wu, H.; Yang, F.; Tripp, R.A.; Hogan, R.J.; Fu, Z.F. Rabies virus expressing dendritic cell-activating molecules enhances the innate and adaptive immune response to vaccination. J. Virol. 2011, 85, 1634–1644. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, K.; Huang, J.; Wang, Q.; He, Y.; Xu, Z.; Xiang, M.; Wu, B.; Chen, H. Recombinant pseudorabies virus expressing P12A and 3C of FMDV can partially protect piglets against FMDV challenge. Res. Vet. Sci. 2011, 91, 90–94. [Google Scholar] [CrossRef] [PubMed]
Table 1. Advantages and disadvantages of recombinant PRV construction approaches.
Table 1. Advantages and disadvantages of recombinant PRV construction approaches.
ApproachesAdvantagesDisadvantages
Homologous recombination (HR)The procedure is relatively easy; Site-specific insertion of foreign genes.Construction of transfer vector is time-consuming and labor-intensive; The efficiency of homologous recombination is low; The purification of viruses is time-consuming.
Bacterial Artificial Chromosome (BAC)More efficient than homologous recombination; Cloning the entire genome into a plasmid facilitates genome modification.Generation of recombinant BAC construct is time-consuming; The PRV genome is easily broken, and once the genome is broken, infectivity is lost and a recombinant virus cannot be obtained.
Fosmid libraryGeneration of the fosmid library is more efficient; High structural stability.The fracted genome was inserted into 5-6 fosmids; 5-6 fosmids need to be co-transfected.
CRISPR/Cas9 systemHighly efficient; Simultaneous targeting of multiple sites.It is necessary to construct homologous arms; Off-target effects usually occurred; This method also requires the purification of the virus.
Table 2. The characteristic features of recombinant PRV.
Table 2. The characteristic features of recombinant PRV.
VectorExogenous GenesInsertion SitesPromotersModification of PRVImmunization Dose/RouteAnimal ModelEfficacyReference
Bartha-K61respiratory syndrome virus (PRRSV) GP5UL23 (TK) siteCMV promoterhomologous recombination (HR)10 7.0 pla-
que-forming unit (PFU)/ intranasally (i.n.) and
intramuscularly (i.m.)
4-week-old pigletconfer
significant protection against clinical disease and reduce pathogenic lesions induced by PRRSV challenge in vaccinated pigs
[63]
Bartha-K61hemagglutinin (HA) gene of swine influenza virus (SIV)not mentionedSV40 promoterHR10 5.0 PFU/i.n.8-week-old miceprotect mice from heterologous virulent challenge[64]
Bartha-K61HA of swine-origin H1N1 virus.gG gene locusMCMV promoterbacterial artificial chromosome (BAC) technology-mediated HR2×10 7.0 PFU/i.n.7-week-old
pigs
protected pig from clinical signs after challenge with a related swine-origin H1N1
influenza A virus
[65]
Bartha-K61gD and gC genes of the AH02LA straingD and gC gene locus BAC technology and HR 10 6.0 TCID 50 / i.m4-week-old pigletPRV
B-gD&gC S is safe for piglets, and provides complete
clinical protection against a pseudorabies variant
(AH02LA) challenge
[22]
Bartha-K61open reading frames E199L, CP204L (p30) and KP177R (p22) of African swine fever virus.gG gene locusCAG promotersBAC technology and CRISPR/Cas9 [61]
PRV variant (HN1201strain)enhanced green fluorescent protein (EGFP) and firefly luciferasebetween gE partial and gI partialEGFP expression was under the control of the CAG promoter (a synthetic promoter composed of a CMV enhancer and chicken β-actin promoter); the firefly luciferase expression cassette was under the control of the SV40 promoterCRISPR/Cas9 [59]
PRV variant
(AH strain)
glycoprotein Cbetween gD and US9CMV promoterHR10 7.0 TCID 50 / i.m (piglet); 10 5.0 TCID 50 / i.m (mice)4-week-old piglet; 4-week- old SPF Kunming miceadditional
insertion of gC gene could enhance the protective efficacy in PRV gI/gE-deleted vaccine in pigs
[51]
PRV variant
(HNX strain)
cytokine Fms-related tyrosine kinase 3 ligand (Flt3L)after gDgD promoterCRISPR/Cas9 and Cre/Lox systems10 5.0 TCID 50 / i.mSix-week-old female BALB/c miceFlt3L can activate DCs and enhance protective immune
responses of recombinant pseudorabies virus with TK/gE gene deletion
[62]
PRV variant
(TJ strain)
classical swine fever virus (CSFV) E2 glycoprotein and capsid (Cap) protein of porcine circovirus type 2 (PCV2)E2 expression cassette was inserted after US9; Cap was fused with gG and co-expressed with gGE2 expression was under the control of CMV promoter; Cap was under the control of the gG promoterfosmid library platform and Red/ET systems10 7, 10 6, and 10 5 TCID 50 / i.m (rabit); 10 6.0 TCID 50 / i.m (pig)6-week-old rabbits; 6-week-old pigsrPRVTJ-delgE/gI/TK-E2-Cap elicited detectable anti-PRV antibodies, but not
anti-PCV2 or anti-CSFV antibodies
[57]
PRV variant (AH02LA strain)S gene of a Porcine epidemic diarrhea virus (PEDV) variantUL11-10, UL35-36, UL46-27MCMV promoter BAC technology and En Passant method [54]
PRV variant
(JS-2012)
CSFV E2 glycoproteinbetween the gG and gD genesnot mentionedHR10 5.0 TCID 50 / i.m3-week-old pigletsinduce the production of Abs to the
gE protein of PRV or to the CSFV proteins other than E2
[50]
PRV variant
(TJ strain)
CSFV E2 glycoproteinbetween gE partial and gI partialCMV promoterHR10 4, 10 5, and 106 TCID 50 / i.m6-week-old piglets provided complete protection against the lethal
challenge with either the PRV TJ strain or the CSFV Shimen strain
[49]
PRV variant
(HNX strain)
tandem repeats of porcine circovirus type 2 (PCV2) Capprotein gene ORF2gE and gG sites CRISPR/Cas9 and HR10 5.8 TCID 50/ i.m4-week-old female BALB/c micehigh titer of specific antibodies for PRV and neutralized antibodies for PCV2 were detected[60]
PRV (Fa strain)CD2v of African swine fever virusPRV UL23 (TK) siteCMV promoterCRISPR/Cas9 and HR10 5.0 TCID 50 / i.m5-week-old SPF mice (ICR)PRV-ΔgE/ΔgI/ΔTK-(CD2v) recombinant strain has strong immunogenicity[12]
PRVP12A and 3C of Foot-and-mouth disease virus (FMDV)between gE partial and gI partialCMV promoterHR10 6.0 TCID 50 / i.m6-week-old large white pigletsPRV-P12A3C induced a high level of neutralizing antibody and FMDV-specific lymphocytes.[71]
PRV variant
(TJ strain)
EGFPfused with UL35UL35 promoterfosmid library platform and Red/ET [55]
PRV
(SC strain)
EGFPfused with UL36UL36 promoterfosmid library platform and Red/ET [56]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhou, M.; Abid, M.; Cao, S.; Zhu, S. Recombinant Pseudorabies Virus Usage in Vaccine Development against Swine Infectious Disease. Viruses 2023, 15, 370. https://doi.org/10.3390/v15020370

AMA Style

Zhou M, Abid M, Cao S, Zhu S. Recombinant Pseudorabies Virus Usage in Vaccine Development against Swine Infectious Disease. Viruses. 2023; 15(2):370. https://doi.org/10.3390/v15020370

Chicago/Turabian Style

Zhou, Mo, Muhammad Abid, Shinuo Cao, and Shanyuan Zhu. 2023. "Recombinant Pseudorabies Virus Usage in Vaccine Development against Swine Infectious Disease" Viruses 15, no. 2: 370. https://doi.org/10.3390/v15020370

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop