DOI QR코드

DOI QR Code

Comparative proteomic analysis of PK-15 cells infected with wild-type strain and its EP0 gene-deleted mutant strain of pseudorabies virus

  • Di Wang (School of Agroforestry and Medicine, The Open University of China) ;
  • Dongjie Chen (Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine) ;
  • Shengkui Xu (College of Animal Science and Technology, Beijing University of Agriculture) ;
  • Fang Wei (Institute of Animal Inspection and Quarantine, Chinese Academy of Inspection and Quarantine) ;
  • Hongyuan Zhao (School of Modern Agriculture & Biotechnology, Ankang University)
  • Received : 2024.02.20
  • Accepted : 2024.06.06
  • Published : 2024.07.31

Abstract

Importance: As one of the main etiologic agents of infectious diseases in pigs, pseudorabies virus (PRV) infections have caused enormous economic losses worldwide. EP0, one of the PRV early proteins (EP) plays a vital role in PRV infections, but the mechanisms are unclear. Objective: This study examined the function of EP0 to provide a direction for its in-depth analysis. Methods: In this study, the EP0-deleted PRV mutant was obtained, and Tandem Mass Tag-based proteomic analysis was used to screen the differentially expressed proteins (DEPs) quantitatively in EP0-deleted PRV- or wild-type PRV-infected porcine kidney 15 cells. Results: This study identified 7,391 DEPs, including 120 and 21 up-regulated and down-regulated DEPs, respectively. Western blot analysis confirmed the changes in the expression of the selected proteins, such as speckled protein 100. Comprehensive analysis revealed 141 DEPs involved in various biological processes and molecular functions, such as transcription regulator activity, biological regulation, and localization. Conclusions and Relevance: These results holistically outlined the functions of EP0 during a PRV infection and might provide a direction for more detailed function studies of EP0 and the stimulation of lytic PRV infections.

Keywords

Acknowledgement

This research was funded by China National Key R&D Program (2023YFF0614402) and the National Natural Science Foundation of China, grant number: 31902302.

References

  1. Pomeranz LE, Reynolds AE, Hengartner CJ. Molecular biology of pseudorabies virus: impact on neurovirology and veterinary medicine. Microbiol Mol Biol Rev. 2005;69(3):462-500.
  2. Tong W, Liu F, Zheng H, Liang C, Zhou YJ, Jiang YF, et al. Emergence of a Pseudorabies virus variant with increased virulence to piglets. Vet Microbiol. 2015;181(3-4):236-240.
  3. Yang H, Han H, Wang H, Cui Y, Liu H, Ding S. A case of human viral encephalitis caused by pseudorabies virus infection in China. Front Neurol. 2019;10:534.
  4. Wang Y, Nian H, Li Z, Wang W, Wang X, Cui Y. Human encephalitis complicated with bilateral acute retinal necrosis associated with pseudorabies virus infection: a case report. Int J Infect Dis. 2019;89:51-54.
  5. Liu Q, Wang X, Xie C, Ding S, Yang H, Guo S, et al. A novel human acute encephalitis caused by pseudorabies virus variant strain. Clin Infect Dis. 2021;73(11):e3690-e3700.
  6. Nauwynck H, Glorieux S, Favoreel H, Pensaert M. Cell biological and molecular characteristics of pseudorabies virus infections in cell cultures and in pigs with emphasis on the respiratory tract. Vet Res. 2007;38(2):229-241.
  7. Li ML, Cui W, Zhao ZY, Mo CC, Wang JL, Chen YL, et al. Molecular cloning and characterization of pseudorabies virus EP0 gene. Indian J Biochem Biophys. 2014;51(2):100-114. 
  8. Lanfranca MP, Mostafa HH, Davido DJ. HSV-1 ICP0: an E3 ubiquitin ligase that counteracts host intrinsic and innate immunity. Cells. 2014;3(2):438-454.
  9. Zhang L, Huang F, Liu J, Xu Y, Miao Y, Yuan Y, et al. HSV-1-encoded ICP0 degrades the host deubiquitinase BRCC36 to antagonize interferon antiviral response. Mol Immunol. 2021;135:28-35.
  10. Halford WP, Schaffer PA. ICP0 is required for efficient reactivation of herpes simplex virus type 1 from neuronal latency. J Virol. 2001;75(7):3240-3249.
  11. Rodriguez MC, Dybas JM, Hughes J, Weitzman MD, Boutell C. The HSV-1 ubiquitin ligase ICP0: modifying the cellular proteome to promote infection. Virus Res. 2020;285:198015.
  12. Guo H, Zhou R, Xi Y, Xiao S, Chen H. Transcriptional suppression of IE180 and TK promoters by the EP0 of pseudorabies virus strains Ea and Fa. Virus Genes. 2009;38(2):269-275.
  13. Wang M, Liu Y, Qin C, Lang Y, Xu A, Yu C, et al. Pseudorabies virus EP0 antagonizes the type i interferon response via inhibiting IRF9 transcription. J Virol. 2022;96(13):e0217121.
  14. Wang T, Tong W, Ye C, Yu Z, Chen J, Gao F, et al. Construction of an infectious bacterial artificial chromosome clone of a pseudorabies virus variant: reconstituted virus exhibited wild-type properties in vitro and in vivo. J Virol Methods. 2018;259:106-115.
  15. Xu S, Chen D, Chen D, Hu Q, Zhou L, Ge X, et al. Pseudorabies virus infection inhibits stress granules formation via dephosphorylating eIF2α. Vet Microbiol. 2020;247:108786.
  16. Ho TY, Wu SL, Chang TJ, Hsiang CH, Chang SH, Hsiang CY. Pseudorabies virus early protein 0 trans-activates the TATA-associated promoter by stimulating the transcription initiation. Virus Res. 1999;61(1):77-86.
  17. Yin Y, Romero N, Favoreel HW. Pseudorabies virus inhibits type I and type III interferon-induced signaling via proteasomal degradation of Janus kinases. J Virol. 2021;95(20):e0079321.
  18. Kovacs-Nagy R, Elek Z, Szekely A, Nanasi T, Sasvari-Szekely M, Ronai Z. Association of aggression with a novel microRNA binding site polymorphism in the wolframin gene. Am J Med Genet B Neuropsychiatr Genet. 2013;162B(4):404-412.
  19. Chen L, Ni M, Ahmed W, Xu Y, Bao X, Zhuang T, et al. Pseudorabies virus infection induces endoplasmic reticulum stress and unfolded protein response in suspension-cultured BHK-21 cells. J Gen Virol. 2022;103(12):10.
  20. Chelbi-Alix MK, Quignon F, Pelicano L, Koken MH, de The H. Resistance to virus infection conferred by the interferon-induced promyelocytic leukemia protein. J Virol. 1998;72(2):1043-1051.
  21. Lallemand-Breitenbach V, de The H. PML nuclear bodies: from architecture to function. Curr Opin Cell Biol. 2018;52:154-161.
  22. Jan Fada B, Reward E, Gu H. The role of ND10 nuclear bodies in herpesvirus infection: a frenemy for the virus? Viruses. 2021;13(2):239.
  23. Batty EC, Jensen K, Freemont PS. PML nuclear bodies and other TRIM-defined subcellular compartments. Adv Exp Med Biol. 2012;770:39-58.
  24. Maul GG, Negorev D, Bell P, Ishov AM. Review: properties and assembly mechanisms of ND10, PML bodies, or PODs. J Struct Biol. 2000;129(2-3):278-287.
  25. Suzich JB, Cuddy SR, Baidas H, Dochnal S, Ke E, Schinlever AR, et al. PML-NB-dependent type I interferon memory results in a restricted form of HSV latency. EMBO Rep. 2021;22(9):e52547.
  26. Szekely L, Pokrovskaja K, Jiang WQ, de The H, Ringertz N, Klein G. The Epstein-Barr virus-encoded nuclear antigen EBNA-5 accumulates in PML-containing bodies. J Virol. 1996;70(4):2562-2568.
  27. Bell P, Lieberman PM, Maul GG. Lytic but not latent replication of Epstein-Barr virus is associated with PML and induces sequential release of nuclear domain 10 proteins. J Virol. 2000;74(24):11800-11810.
  28. Fraschilla I, Jeffrey KL. The speckled protein (SP) family: immunity's chromatin readers. Trends Immunol. 2020;41(7):572-585.
  29. Sengupta I, Das D, Singh SP, Chakravarty R, Das C. Host transcription factor Speckled 110 kDa (Sp110), a nuclear body protein, is hijacked by hepatitis B virus protein X for viral persistence. J Biol Chem. 2017;292(50):20379-20393.
  30. Nicewonger J, Suck G, Bloch D, Swaminathan S. Epstein-Barr virus (EBV) SM protein induces and recruits cellular Sp110b to stabilize mRNAs and enhance EBV lytic gene expression. J Virol. 2004;78(17):9412-9422.
  31. Everett RD, Parada C, Gripon P, Sirma H, Orr A. Replication of ICP0-null mutant herpes simplex virus type 1 is restricted by both PML and Sp100. J Virol. 2008;82(6):2661-2672.
  32. Negorev DG, Vladimirova OV, Maul GG. Differential functions of interferon-upregulated Sp100 isoforms: herpes simplex virus type 1 promoter-based immediate-early gene suppression and PML protection from ICP0-mediated degradation. J Virol. 2009;83(10):5168-5180.
  33. Ma Y, Li J, Dong H, Yang Z, Zhou L, Xu P. PML body component Sp100A restricts wild-type herpes simplex virus 1 infection. J Virol. 2022;96(8):e0027922.
  34. Perusina Lanfranca M, Mostafa HH, Davido DJ. Two overlapping regions within the N-terminal half of the herpes simplex virus 1 E3 ubiquitin ligase ICP0 facilitate the degradation and dissociation of PML and dissociation of Sp100 from ND10. J Virol. 2013;87(24):13287-13296.
  35. Xu P, Mallon S, Roizman B. PML plays both inimical and beneficial roles in HSV-1 replication. Proc Natl Acad Sci USA. 2016;113(21):E3022-E3028.
  36. Gu H, Roizman B. The degradation of promyelocytic leukemia and Sp100 proteins by herpes simplex virus 1 is mediated by the ubiquitin-conjugating enzyme UbcH5a. Proc Natl Acad Sci USA. 2003;100(15):8963-8968.