Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2002;8(5):381–391. doi: 10.1080/13550280260422686

Enhanced green fluorescent protein expression may be used to monitor murine coronavirus spread in vitro and in the mouse central nervous system

Jayasri Das Sarma 1, Esther Scheen 2, Su-hun Seo 2, Michael Koval 1, Susan R Weiss 2,
PMCID: PMC7095158  PMID: 12402164

Abstract

Targeted recombination was used to select mouse hepatitis virus isolates with stable and efficient expression of the gene encoding the enhanced green fluorescent protein (EGFP). The EGFP gene was inserted into the murine coronavirus genome in place of the nonessential gene 4. These viruses expressed the EGFP gene from an mRNA of slightly slower electrophoretic mobility than mRNA 4. EGFP protein was detected on a Western blot of infected cell lysates and EGFP activity (fluorescence) was visualized by microscopy in infected cells and in viral plaques. Expression of EGFP remained stable through at least six passages in tissue culture and during acute infection in the mouse central nervous system. These viruses replicated with similar kinetics and to similar final extents as wild-type virus both in tissue culture and in the mouse central nervous system (CNS). They caused encephalitis and demyelination in animals as wild-type virus; however, they were somewhat attenuated in virulence. Isogenic EGFP-expressing viruses that differ only in the spike gene and express either the spike gene of the highly neurovirulent MHV-4 strain or the more weakly neurovirulent MHV-A59 strain were compared; the difference in virulence and patterns of spread of viral antigen reflected the differences between parental viruses expressing each of these spike genes. Thus, EGFP-expressing viruses will be useful in the studies of murine coronavirus pathogenesis in mice.

Keywords: murine coronavirus, viral genetics, viral pathogenesis

Footnotes

This work was supported by NIH grants AI-17418, NS-30606, and NS-21954, and a National Multiple Sclerosis Society grant RG-2585 to SRW, and an American Heart Association Grant-in-Aid and NIH GM61012 to MK. Jayasri Das Sarma was supported in part by an advanced postdoctoral fellowship, FG1431-A-1, from the National Multiple Sclerosis Society.

References

  1. Bond CW, Leibowitz JL, Robb JA. Pathogenic murine coronaviruses. II. Characterization of virus-specific proteins of murine coronaviruses JHMV and A59V. Virology. 1979;94:371–384. doi: 10.1016/0042-6822(79)90468-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Cormack BP, Valdivia RH, Falkow S. FACS-optimized mutants of the green fluorescent protein (GFP) Gene. 1996;173:33–38. doi: 10.1016/0378-1119(95)00685-0. [DOI] [PubMed] [Google Scholar]
  3. Das Sarma J, Fu L, Tsai JC, Weiss SR, Lavi E. Demyelination determinants map to the spike glycoprotein gene of coronavirus mouse hepatitis virus. J Virol. 2000;74:9206–9213. doi: 10.1128/JVI.74.19.9206-9213.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Fischer F, Stegen CF, Koetzner CA, Masters PS. Analysis of a recombinant mouse hepatitis virus expressing a foreign gene reveals a novel aspect of coronavirus transcription. J Virol. 1997;71:5148–5160. doi: 10.1128/jvi.71.7.5148-5160.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Gombold JL, Hingley ST, Weiss SR. Fusion-defective mutants of mouse hepatitis virus A59 contain a mutation in the spike protein cleavage signal. J Virol. 1993;67:4504–4512. doi: 10.1128/jvi.67.8.4504-4512.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Gombold JL, Weiss SR. Mouse hepatitis virus A59 increases steady state levels of MHC mRNAs in primary glial cell cultures and in the murine central nervous system. Microb Pathogen. 1992;13:493–505. doi: 10.1016/0882-4010(92)90015-G. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Haas J, Park EC, Seed B. Codon usage limitation in the expression of HIV-1 envelope glycoprotein. Curr Biol. 1996;6:315–324. doi: 10.1016/S0960-9822(02)00482-7. [DOI] [PubMed] [Google Scholar]
  8. Houtman JJ, Fleming JO. Dissociation of demyelination and viral clearance in congenitally immunodeficient mice infected with murine coronavirus JHM. J NeuroVirol. 1996;2:101–110. doi: 10.3109/13550289609146543. [DOI] [PubMed] [Google Scholar]
  9. Kuo L, Godeke GJ, Raamsman MJ, Masters PS, Rottier PJ. Retargeting of coronavirus by substitution of the spike glycoprotein ectodomain: Crossing the host cell species barrier. J Virol. 2000;74:1393–1406. doi: 10.1128/JVI.74.3.1393-1406.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Lavi E, Gilden DH, Wroblewska Z, Rorke LB, Weiss SR. Experimental demyelination produced by the A59 strain of mouse hepatitis virus. Neurology. 1984;34:597–603. doi: 10.1212/wnl.34.5.597. [DOI] [PubMed] [Google Scholar]
  11. Lavi E, Murray EM, Makino S, Stohlman SA, Lai MMC, Weiss SR. Determinants of coronavirus MHV pathogenesis are localized to 3′ portions of the genome as determined by ribonucleic acid-ribonucleic acid recombination. Lab Invest. 1990;62:570–578. [PubMed] [Google Scholar]
  12. Leparc-Goffart I, Hingley ST, Chua MM, Jiang X, Lavi E, Weiss SR. Altered pathogenesis of a mutant of the murine coronavirus MHV-A59 is associated with a Q159L amino acid substitution in the spike protein. Virology. 1997;239:1–10. doi: 10.1006/viro.1997.8877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Leparc-Goffart I, Hingley ST, Chua MM, Phillips J, Lavi E, Weiss SR. Targeted recombination within the spike gene of murine coronavirus mouse hepatitis virus-A59: Q159 is a determinant of hepatotropism. J Virol. 1998;72:9628–9636. doi: 10.1128/jvi.72.12.9628-9636.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Luytjes W, Bredenbeek PJ, Noten AF, Horzinek MC, Spaan WJM. Sequence of mouse hepatitis virus A59 mRNA 2: indications for RNA recombination between coronaviruses and influenza C virus. Virology. 1988;166:415–422. doi: 10.1016/0042-6822(88)90512-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Matthews AE, Weiss SR, Shlomchik MJ, Hannum LG, Gombold JL, Paterson Y. Antibody is required for clearance of infectious murine hepatitis virus A59 from the central nervous system, but not the liver. J Immunol. 2001;167:5254–5263. doi: 10.4049/jimmunol.167.9.5254. [DOI] [PubMed] [Google Scholar]
  16. Navas S, Seo SH, Chua MM, Sarma JD, Lavi E, Hingley ST, Weiss SR. Murine coronavirus spike protein determines the ability of the virus to replicate in the liver and cause hepatitis. J Virol. 2001;75:2452–2457. doi: 10.1128/JVI.75.5.2452-2457.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Ontiveros E, Kuo L, Masters PS, Perlman S. Inactivation of expression of gene 4 of mouse hepatitis virus strain JHM does not affect virulence in the murine CNS. Virology. 2001;289:230–238. doi: 10.1006/viro.2001.1167. [DOI] [PubMed] [Google Scholar]
  18. Phillips JJ, Chua MM, Lavi E, Weiss SR. Pathogenesis of chimeric MHV4/MHV-A59 recombinant viruses: The murine coronavirus spike protein is a major determinant of neurovirulence. J Virol. 1999;73:7752–7760. doi: 10.1128/jvi.73.9.7752-7760.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Phillips JJ, Chua M, Seo SH, Weiss SR. Multiple regions of the murine coronavirus spike glycoprotein influence neurovirulence. J NeuroVirol. 2001;7:421–431. doi: 10.1080/135502801753170273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Sanger F, Nicklen S, Coulson AP. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci USA. 1977;74:5463–5467. doi: 10.1073/pnas.74.12.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Schwarz B, Routledge E, Siddell SG. Murine coronavirus nonstructural protein ns2 is not essential for virus replication in transformed cells. J Virol. 1990;64:4784–4791. doi: 10.1128/jvi.64.10.4784-4791.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Slifka MK, Pagarigan R, Mena I, Feuer R, Whitton JL. Using recombinant coxsackievirus B3 to evaluate the induction and protective efficacy of CD8+ T cells during picornavirus infection. J Virol. 2001;75:2377–2387. doi: 10.1128/JVI.75.5.2377-2387.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Smith BN, Banfield BW, Smeraski CA, Wilcox CL, Dudek FE, Enquist LW, Pickard GE. Pseudorabies virus expressing enhanced green fluorescent protein: a tool for in vitro electrophysiological analysis of transsynaptically labeled neurons in identified central nervous system circuits. Proc Natl Acad Sci USA. 2000;97:9264–9269. doi: 10.1073/pnas.97.16.9264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Sutherland RM, Chua MM, Lavi E, Weiss SR, Paterson Y. CD4+ and CD8+ T cells are not major effectors of mouse hepatitis virus A59-induced demyelinating disease. J NeuroVirol. 1997;3:225–228. doi: 10.3109/13550289709018297. [DOI] [PubMed] [Google Scholar]
  25. Yokomori K, Lai MMC. Mouse hepatitis virus S sequence reveals that nonstructural proteins ns4 and ns5a are not essential for murine coronavirus replication. J Virol. 1991;65:5605–5608. doi: 10.1128/jvi.65.10.5605-5608.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Neurovirology are provided here courtesy of Nature Publishing Group

RESOURCES