DOI QR코드

DOI QR Code

Establishment of inflammatory model induced by Pseudorabies virus infection in mice

  • Ren, Chun-Zhi (College of Animal Science and Technology, Guangxi University) ;
  • Hu, Wen-Yue (School of Life Sciences & Biotechnology, Shanghai Jiao Tong University) ;
  • Zhang, Jin-Wu (College of Animal Science and Technology, Guangxi University) ;
  • Wei, Ying-Yi (College of Animal Science and Technology, Guangxi University) ;
  • Yu, Mei-Ling (College of Animal Science and Technology, Guangxi University) ;
  • Hu, Ting-Jun (College of Animal Science and Technology, Guangxi University)
  • Received : 2020.10.05
  • Accepted : 2021.01.07
  • Published : 2021.03.31

Abstract

Background: Pseudorabies virus (PRV) infection leads to high mortality in swine. Despite extensive efforts, effective treatments against PRV infection are limited. Furthermore, the inflammatory response induced by PRV strain GXLB-2013 is unclear. Objectives: Our study aimed to investigate the inflammatory response induced by PRV strain GXLB-2013, establish an inflammation model to elucidate the pathogenesis of PRV infection further, and develop effective drugs against PRV infection. Methods: Kunming mice were infected intramuscularly with medium, LPS, and different doses of PRV-GXLB-2013. Viral spread and histopathological damage to brain, spleen, and lung were determined at 7 days post-infection (dpi). Immune organ indices, levels of reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines, as well as levels of activity of COX-2 and iNOS were determined at 4, 7, and 14 dpi. Results: At 105-106 TCID50 PRV produced obviously neurological symptoms and 100% mortality in mice. Viral antigens were detectable in kidney, heart, lung, liver, spleen, and brain. In addition, inflammatory injuries were apparent in brain, spleen, and lung of PRV-infected mice. Moreover, PRV induced increases in immune organ indices, ROS and NO levels, activity of COX-2 and iNOS, and the content of key pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, tumor necrosis factor-α, interferon-γ and MCP-1. Among the tested doses, 102 TCID50 of PRV produced a significant inflammatory mediator increase. Conclusions: An inflammatory model induced by PRV infection was established in mice, and 102 TCID50 PRV was considered as the best concentration for the establishment of the model.

Keywords

Acknowledgement

We thank Dr. Huang Weijian in the Laboratory of Animal Infectious Disease in College of Animal Science and Technology at Guangxi University for his kindness in providing the PRV.

References

  1. Muller T, Hahn EC, Tottewitz F, Kramer M, Klupp BG, Mettenleiter TC, et al. Pseudorabies virus in wild swine: a global perspective. Arch Virol. 2011;156(10):1691-1705. https://doi.org/10.1007/s00705-011-1080-2
  2. Laval K, Enquist LW. The neuropathic itch caused by pseudorabies virus. Pathogens. 2020;9(4):E254.
  3. Ai JW, Weng SS, Cheng Q, Cui P, Li YJ, Wu HL, et al. Human endophthalmitis caused by pseudorabies virus infection, China, 2017. Emerg Infect Dis. 2018;24(6):1087-1090. https://doi.org/10.3201/eid2406.171612
  4. Hu F, Wang J, Peng XY. Bilateral necrotizing retinitis following encephalitis caused by the pseudorabies virus confirmed by next-generation sequencing. Ocul Immunol Inflamm. 2020:1-4.
  5. He W, Auclert LZ, Zhai X, Wong G, Zhang C, Zhu H, et al. Interspecies transmission, genetic diversity, and evolutionary dynamics of pseudorabies virus. J Infect Dis. 2019;219(11):1705-1715. https://doi.org/10.1093/infdis/jiy731
  6. Wang Y, Xia SL, Lei JL, Cong X, Xiang GT, Luo Y, et al. Dose-dependent pathogenicity of a pseudorabies virus variant in pigs inoculated via intranasal route. Vet Immunol Immunopathol. 2015;168(3-4):147-152. https://doi.org/10.1016/j.vetimm.2015.10.011
  7. Takahashi H, Yoshikawa Y, Kai C, Yamanouchi K. Mechanism of pruritus and peracute death in mice induced by pseudorabies virus (PRV) infection. J Vet Med Sci. 1993;55(6):913-920. https://doi.org/10.1292/jvms.55.913
  8. Sehl J, Teifke JP. Comparative pathology of pseudorabies in different naturally and experimentally infected species-a review. Pathogens. 2020;9(8):E633.
  9. Colomer MA, Margalida A, Fraile L. Vaccination is a suitable tool in the control of Aujeszky's disease outbreaks in pigs using a population dynamics P systems model. Animals (Basel). 2020;10(5):E909.
  10. Lin W, Shao Y, Tan C, Shen Y, Zhang X, Xiao J, et al. Commercial vaccine against pseudorabies virus: a hidden health risk for dogs. Vet Microbiol. 2019;233:102-112. https://doi.org/10.1016/j.vetmic.2019.04.031
  11. Hahn EC, Fadl-Alla B, Lichtensteiger CA. Variation of Aujeszky's disease viruses in wild swine in USA. Vet Microbiol. 2010;143(1):45-51. https://doi.org/10.1016/j.vetmic.2010.02.013
  12. Yu ZQ, Tong W, Zheng H, Li LW, Li GX, Gao F, et al. Variations in glycoprotein B contribute to immunogenic difference between PRV variant JS-2012 and Bartha-K61. Vet Microbiol. 2017;208:97-105. https://doi.org/10.1016/j.vetmic.2017.07.019
  13. 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. https://doi.org/10.1016/j.jviromet.2018.06.004
  14. Liu J, Chen C, Li X. Novel Chinese pseudorabies virus variants undergo extensive recombination and rapid interspecies transmission. Transbound Emerg Dis. 2020;67(6):2274-2276. https://doi.org/10.1111/tbed.13784
  15. Ye C, Guo JC, Gao JC, Wang TY, Zhao K, Chang XB, et al. Genomic analyses reveal that partial sequence of an earlier pseudorabies virus in China is originated from a Bartha-vaccine-like strain. Virology. 2016;491:56-63. https://doi.org/10.1016/j.virol.2016.01.016
  16. 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. 2020;ciaa987.
  17. Zhao X, Tong W, Song X, Jia R, Li L, Zou Y, et al. Antiviral effect of resveratrol in piglets infected with virulent pseudorabies virus. Viruses. 2018;10(9):E457.
  18. Kim JS, Enquist LW, Card JP. Circuit-specific coinfection of neurons in the rat central nervous system with two pseudorabies virus recombinants. J Virol. 1999;73(11):9521-9531. https://doi.org/10.1128/jvi.73.11.9521-9531.1999
  19. Brittle EE, Reynolds AE, Enquist LW. Two modes of pseudorabies virus neuroinvasion and lethality in mice. J Virol. 2004;78(23):12951-12963. https://doi.org/10.1128/JVI.78.23.12951-12963.2004
  20. Li X, Zhang W, Liu Y, Xie J, Hu C, Wang X. Role of p53 in pseudorabies virus replication, pathogenicity, and host immune responses. Vet Res (Faisalabad). 2019;50(1):9. https://doi.org/10.1186/s13567-019-0627-1
  21. Chen HL, Yang J, Fu YF, Meng XN, Zhao WD, Hu TJ. Effect of total flavonoids of Spatholobus suberectus Dunn on PCV2 induced oxidative stress in RAW264.7 cells. BMC Complement Altern Med. 2017;17(1):244. https://doi.org/10.1186/s12906-017-1764-6
  22. Qin SY. Complete genome analysis and biological characteristics of PRV GXLB-2015 and GXGG-2016 strain [Master's dissertation]. Nanning: Guangxi University; 2017.
  23. 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. https://doi.org/10.1128/MMBR.69.3.462-500.2005
  24. Laval K, Vernejoul JB, Van Cleemput J, Koyuncu OO, Enquist LW. Virulent pseudorabies virus infection induces a specific and lethal systemic inflammatory response in mice. J Virol. 2018;92(24):e01614-18.
  25. Poeck H, Ruland J. From virus to inflammation: mechanisms of RIG-I-induced IL-1β production. Eur J Cell Biol. 2012;91(1):59-64. https://doi.org/10.1016/j.ejcb.2011.01.013
  26. Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze MG. Into the eye of the cytokine storm. Microbiol Mol Biol Rev. 2012;76(1):16-32. https://doi.org/10.1128/MMBR.05015-11
  27. Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta. 2014;1843(11):2563-2582. https://doi.org/10.1016/j.bbamcr.2014.05.014
  28. Ohashi W, Hattori K, Hattori Y. Control of macrophage dynamics as a potential therapeutic approach for clinical disorders involving chronic inflammation. J Pharmacol Exp Ther. 2015;354(3):240-250. https://doi.org/10.1124/jpet.115.225540
  29. Miller LC, Zanella EL, Waters WR, Lager KM. Cytokine protein expression levels in tracheobronchial lymph node homogenates of pigs infected with pseudorabies virus. Clin Vaccine Immunol. 2010;17(5):728-734. https://doi.org/10.1128/CVI.00485-09
  30. Rahman I. Oxidative stress, transcription factors and chromatin remodelling in lung inflammation. Biochem Pharmacol. 2002;64(5-6):935-942. https://doi.org/10.1016/S0006-2952(02)01153-X
  31. Hensley K, Robinson KA, Gabbita SP, Salsman S, Floyd RA. Reactive oxygen species, cell signaling, and cell injury. Free Radic Biol Med. 2000;28(10):1456-1462. https://doi.org/10.1016/S0891-5849(00)00252-5
  32. Guzik TJ, Korbut R, Adamek-Guzik T. Nitric oxide and superoxide in inflammation and immune regulation. J Physiol Pharmacol 2003;54(4):469-487.
  33. Chen L, Kang YH. Antioxidant and enzyme inhibitory activities of Plebeian herba (Salvia plebeia R. Br.) under different cultivation conditions. J Agric Food Chem. 2014;62(10):2190-2197. https://doi.org/10.1021/jf404570s
  34. Gopez JJ, Yue H, Vasudevan R, Malik AS, Fogelsanger LN, Lewis S, et al. Cyclooxygenase-2-specific inhibitor improves functional outcomes, provides neuroprotection, and reduces inflammation in a rat model of traumatic brain injury. Neurosurgery. 2005;56(3):590-604. https://doi.org/10.1227/01.NEU.0000154060.14900.8F
  35. Jang SI, Kim BH, Lee WY, An SJ, Choi HG, Jeon BH, et al. Stylopine from Chelidonium majus inhibits LPS-induced inflammatory mediators in RAW 264.7 cells. Arch Pharm Res. 2004;27(9):923-929. https://doi.org/10.1007/BF02975845
  36. Talar-Wojnarowska R, Gasiorowska A, Smolarz B, Romanowicz-Makowska H, Kulig A, Malecka-Panas E. Clinical significance of interleukin-6 (IL-6) gene polymorphism and IL-6 serum level in pancreatic adenocarcinoma and chronic pancreatitis. Dig Dis Sci. 2009;54(3):683-689. https://doi.org/10.1007/s10620-008-0390-z
  37. Brandt J, Haibel H, Cornely D, Golder W, Gonzalez J, Reddig J, et al. Successful treatment of active ankylosing spondylitis with the anti-tumor necrosis factor alpha monoclonal antibody infliximab. Arthritis Rheum. 2000;43(6):1346-1352. https://doi.org/10.1002/1529-0131(200006)43:6<1346::AID-ANR18>3.0.CO;2-E
  38. McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5(1):45. https://doi.org/10.1186/1742-2094-5-45
  39. Wilson MR, Choudhury S, Takata M. Pulmonary inflammation induced by high-stretch ventilation is mediated by tumor necrosis factor signaling in mice. Am J Physiol Lung Cell Mol Physiol. 2005;288(4):L599-L607. https://doi.org/10.1152/ajplung.00304.2004
  40. Ogando DG, Paz D, Cella M, Franchi AM. The fundamental role of increased production of nitric oxide in lipopolysaccharide-induced embryonic resorption in mice. Reproduction. 2003;125(1):95-110. https://doi.org/10.1530/reprod/125.1.95
  41. O'Neill E, Griffin EW, O'Sullivan R, Murray C, Ryan L, Yssel J, et al. Acute neuroinflammation, sickness behavior and working memory in response to acute systemic challenge with LPS following noradrenergic lesion in mice. Brain Behav Immun 2020;8:S0889-1591(20)32445-4.
  42. Fuijkschot WW, Morrison MC, Zethof IP, Krijnen PA, Kleemann R, Niessen HW, et al. LPS-induced systemic inflammation does not alter atherosclerotic plaque area or inflammation in APOE3*LEIDEN mice in the early phase up to 15 days. Shock. 2018;50(3):360-365. https://doi.org/10.1097/SHK.0000000000001026