Analyses of cell death mechanisms related to amino acid substitution at position 95 in the rabies virus matrix protein No Access

Abstract

We previously reported that the avirulent fixed rabies virus strain Ni-CE induces a clear cytopathic effect in mouse neuroblastoma cells, whereas its virulent progenitor, the Nishigahara strain, does not. Infection with Nishigahara and Ni-CE mutants containing a single amino acid substitution in the matrix protein (M) demonstrated that the amino acid at position 95 of M (M95) is a cytopathic determinant. The characteristics of cell death induced by Ni-CE infection resemble those of apoptosis (rounded and shrunken cells, DNA fragmentation), but the intracellular signalling pathway for this process has not been fully investigated. In this study, we aimed to elucidate the mechanism by which M95 affects cell death induced by human neuroblastoma cell infection with the Nishigahara, Ni-CE and M95-mutated strains. We demonstrated that the Ni-CE strain induced DNA fragmentation, cell membrane disruption, exposure of phosphatidylserine (PS), activation of caspase-3/7 and anti-poly (ADP-ribose) polymerase 1 (PARP-1) cleavage, an early apoptosis indicator, whereas the Nishigahara strain did not induce DNA fragmentation, caspase-3/7 activation, cell membrane disruption, or PARP-1 cleavage, but did induce PS exposure. We also demonstrated that these characteristics were associated with M95 using M95-mutated strains. However, we found that Ni-CE induced cell death despite the presence of a caspase inhibitor, Z-VAD-FMK. In conclusion, our data suggest that M95 mutation-related cell death is caused by both the caspase-dependent and -independent pathways.

Funding
This study was supported by the:
  • GlaxoSmithKline foundation
    • Principle Award Recipient: TatsunoriMasatani
  • Japan Society for the Promotion of Science (Award 17K08083)
    • Principle Award Recipient: TatsunoriMasatani
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001594
2021-04-23
2024-03-29
Loading full text...

Full text loading...

References

  1. Hampson K, Coudeville L, Lembo T, Sambo M, Kieffer A. Estimating the global burden of endemic canine rabies. PLoS Negl Trop Dis 2015; 9:1–20
    [Google Scholar]
  2. Chenik M, Chebli K, Gaudin Y, Blondel D. In vivo interaction of rabies virus phosphoprotein (P) and nucleoprotein (N): existence of two N-binding sites on P protein. J Gen Virol 1994; 75:2889–2896 [View Article][PubMed]
    [Google Scholar]
  3. Finke S, Conzelmann KK. Dissociation of rabies virus matrix protein functions in regulation of viral RNA synthesis and virus assembly. J Virol 2003; 77:12074–12082 [View Article][PubMed]
    [Google Scholar]
  4. Wirblich C, Tan GS, Papaneri A, Godlewski PJ, Orenstein JM et al. PPEY motif within the rabies virus (RV) matrix protein is essential for efficient virion release and RV pathogenicity. J Virol 2008; 82:9730–9738 [View Article][PubMed]
    [Google Scholar]
  5. Dietzschold B, Li J, Faber M, Schnell M. Concepts in the pathogenesis of rabies. Future Virol 2008; 3:481–490 [View Article][PubMed]
    [Google Scholar]
  6. Finke S, Conzelmann KK. Replication strategies of rabies virus. Virus Res 2005; 111:120–131 [View Article][PubMed]
    [Google Scholar]
  7. Shimizu K, Ito N, Mita T, Yamada K, Hosokawa-Muto J et al. Involvement of nucleoprotein, phosphoprotein, and matrix protein genes of rabies virus in virulence for adult mice. Virus Res 2007; 123:154–160 [View Article][PubMed]
    [Google Scholar]
  8. Ito N, Mita T, Shimizu K, Ito Y, Masatani T et al. Amino acid substitution at position 95 in rabies virus matrix protein affects viral pathogenicity. J Vet Med Sci 2011; 73:1363–1366 [View Article][PubMed]
    [Google Scholar]
  9. Mita T, Shimizu K, Ito N, Yamada K, Ito Y et al. Amino acid at position 95 of the matrix protein is a cytopathic determinant of rabies virus. Virus Res 2008; 137:33–39 [View Article][PubMed]
    [Google Scholar]
  10. Jackson AC, Rasalingam P, Weli SC. Comparative pathogenesis of recombinant rabies vaccine strain SAD-L16 and SAD-D29 with replacement of Arg333 in the glycoprotein after peripheral inoculation of neonatal mice: less neurovirulent strain is a stronger inducer of neuronal apoptosis. Acta Neuropathol 2006; 111:372–378 [View Article][PubMed]
    [Google Scholar]
  11. Morimoto K, Hooper DC, Spitsin S, Koprowski H, Dietzschold B. Pathogenicity of different rabies virus variants inversely correlates with apoptosis and rabies virus glycoprotein expression in infected primary neuron cultures. J Virol 1999; 73:510–518 [View Article][PubMed]
    [Google Scholar]
  12. Suja MS, Mahadevan A, Madhusudana SN, Shankar SK. Role of apoptosis in rabies viral encephalitis: a comparative study in mice, canine, and human brain with a review of literature. Patholog Res Int 2011; 2011:1–13 [View Article][PubMed]
    [Google Scholar]
  13. Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science 1998; 281:1305–1308 [View Article][PubMed]
    [Google Scholar]
  14. Julien O, Wells JA. Caspases and their substrates. Cell Death Differ 2017; 24:1380–1389 [View Article][PubMed]
    [Google Scholar]
  15. Larsen BD, Sørensen CS. The caspase-activated DNase: apoptosis and beyond. Febs J 2017; 284:1160–1170 [View Article][PubMed]
    [Google Scholar]
  16. Suzuki J, Imanishi E, Nagata S. Xkr8 phospholipid scrambling complex in apoptotic phosphatidylserine exposure. Proc Natl Acad Sci U S A 2016; 113:9509–9514 [View Article][PubMed]
    [Google Scholar]
  17. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D et al. Molecular mechanisms of cell death: recommendations of the nomenclature Committee on cell death 2018. Cell Death Differ 2018; 25:486–541 [View Article][PubMed]
    [Google Scholar]
  18. Yamada K, Ito N, Takayama-Ito M, Sugiyama M, Minamoto N. Multigenic relation to the attenuation of rabies virus. Microbiol Immunol 2006; 50:25–32 [View Article][PubMed]
    [Google Scholar]
  19. Minamoto N, Tanaka H, Hishida M, Goto H, Ito H et al. Linear and conformation-dependent antigenic sites on the nucleoprotein of rabies virus. Microbiol Immunol 1994; 38:449–455 [View Article][PubMed]
    [Google Scholar]
  20. Thoulouze M, Lafage M, Montano-Hirose JA, Lafon M. Rabies virus infects mouse and human lymphocytes and induces apoptosis. J Virol 1997; 71:207372–7380 [View Article][PubMed]
    [Google Scholar]
  21. Zan J, Liu J, Zhou JW, Wang HL, Mo K-K et al. Rabies virus matrix protein induces apoptosis by targeting mitochondria. Exp Cell Res 2016; 347:83–94 [View Article][PubMed]
    [Google Scholar]
  22. Chaitanya GV, Steven AJ, Babu PP. Parp-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun Signal 2010; 8:31 [View Article][PubMed]
    [Google Scholar]
  23. Yan X, Prosniak M, Curtis MT, Weiss ML, Faber M et al. Silver-haired bat rabies virus variant does not induce apoptosis in the brain of experimentally infected mice. J Neurovirol 2001; 7:518–527 [View Article][PubMed]
    [Google Scholar]
  24. Sarmento L, Tseggai T, Dhingra V, Fu ZF. Rabies virus-induced apoptosis involves caspase-dependent and caspase-independent pathways. Virus Res 2006; 121:144–151 [View Article][PubMed]
    [Google Scholar]
  25. Silva MT, do Vale A, dos Santos NMN. Secondary necrosis in multicellular animals: an outcome of apoptosis with pathogenic implications. Apoptosis 2008; 13:463–482 [View Article][PubMed]
    [Google Scholar]
  26. Segawa K, Kurata S, Yanagihashi Y, Brummelkamp TR, Matsuda F et al. Caspase-Mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 2014; 344:1164–1168 [View Article][PubMed]
    [Google Scholar]
  27. Suzuki J, Umeda M, Sims PJ, Nagata S. Calcium-Dependent phospholipid scrambling by TMEM16F. Nature 2010; 468:834–838 [View Article][PubMed]
    [Google Scholar]
  28. Suzuki J, Denning DP, Imanishi E, Horvitz HR, Nagata S. Xk-Related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science 2013a; 341:403–406 [View Article][PubMed]
    [Google Scholar]
  29. Suzuki J, Fujii T, Imao T, Ishihara K, Kuba H et al. Calcium-Dependent phospholipid scramblase activity of TMEM16 protein family members. J Biol Chem 2013b; 288:13305–13316 [View Article][PubMed]
    [Google Scholar]
  30. Stowell SR, Karmakar S, Arthur CM, Ju T, Rodrigues LC et al. Galectin-1 induces reversible phosphatidylserine exposure at the plasma membrane. Mol Biol Cell 2009; 20:1408–1418 [View Article][PubMed]
    [Google Scholar]
  31. Ivana Scovassi A, Diederich M. Modulation of poly(ADP-ribosylation) in apoptotic cells. Biochem Pharmacol 2004; 68:1041–1047 [View Article]
    [Google Scholar]
  32. Salvesen GS, Dixit VM. Caspases: intracellular signaling by proteolysis. Cell 1997; 91:443–446 [View Article][PubMed]
    [Google Scholar]
  33. Boucher D, Blais V, Denault JB. Caspase-7 uses an exosite to promote poly(ADP ribose) polymerase 1 proteolysis. Proc Natl Acad Sci U S A 2012; 109:5669–5674 [View Article][PubMed]
    [Google Scholar]
  34. Masdehors P, Glaisner S, Maciorowski Z, Magdelénat H, Delic J. Ubiquitin-dependent protein processing controls radiation-induced apoptosis through the N-end rule pathway. Exp Cell Res 2000; 257:48–57 [View Article][PubMed]
    [Google Scholar]
  35. Yang Y, Zhao S, Song J. Caspase-dependent apoptosis and -independent poly(ADP-ribose) polymerase cleavage induced by transforming growth factor beta1. Int J Biochem Cell Biol 2004; 36:223–234 [View Article][PubMed]
    [Google Scholar]
  36. Upton JW, Shubina M, Balachandran S. RIPK3-driven cell death during virus infections. Immunol Rev 2017; 277:90–101 [View Article][PubMed]
    [Google Scholar]
  37. Douzandegan Y, Tahamtan A, Gray Z, Nikoo HR, Tabarraei A et al. Cell death mechanisms in esophageal squamous cell carcinoma induced by vesicular stomatitis virus matrix protein. Osong Public Heal Res Perspect 2019; 10:246–252 [View Article][PubMed]
    [Google Scholar]
  38. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019; 29:347–364 [View Article][PubMed]
    [Google Scholar]
  39. Gadaleta P, Perfetti X, Mersich S, Coulombié F. Early activation of the mitochondrial apoptotic pathway in vesicular stomatitis virus-infected cells. Virus Res 2005; 109:65–69 [View Article][PubMed]
    [Google Scholar]
  40. Wang Y, Kim NS, Haince JF, Kang HC, David KK et al. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 2011; 4:ra20 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001594
Loading
/content/journal/jgv/10.1099/jgv.0.001594
Loading

Data & Media loading...

Most cited Most Cited RSS feed