Tula orthohantavirus nucleocapsid protein is cleaved in infected cells and may sequester activated caspase-3 during persistent infection to suppress apoptosis Free

Abstract

The family mostly comprises rodent-borne segmented negative-sense RNA viruses, many of which are capable of causing devastating disease in humans. In contrast, hantavirus infection of rodent hosts results in a persistent and inapparent infection through their ability to evade immune detection and inhibit apoptosis. In this study, we used Tula hantavirus (TULV) to investigate the interplay between viral and host apoptotic responses during early, peak and persistent phases of virus infection in cell culture. Examination of early-phase TULV infection revealed that infected cells were refractory to apoptosis, as evidenced by the complete lack of cleaved caspase-3 (casp-3C) staining, whereas in non-infected bystander cells casp-3C was highly abundant. Interestingly, at later time points, casp-3C was abundant in infected cells, but the cells remained viable and able to continue shedding infectious virus, and together these observations were suggestive of a TULV-associated apoptotic block. To investigate this block, we viewed TULV-infected cells using laser scanning confocal and wide-field deconvolution microscopy, which revealed that TULV nucleocapsid protein (NP) colocalized with, and sequestered, casp-3C within cytoplasmic ultrastructures. Consistent with casp-3C colocalization, we showed for the first time that TULV NP was cleaved in cells and that TULV NP and casp-3C could be co-immunoprecipitated, suggesting that this interaction was stable and thus unlikely to be solely confined to NP binding as a substrate to the casp-3C active site. To account for these findings, we propose a novel mechanism by which TULV NP inhibits apoptosis by spatially sequestering casp-3C from its downstream apoptotic targets within the cytosol.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001291
2019-08-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/100/8/1208.html?itemId=/content/journal/jgv/10.1099/jgv.0.001291&mimeType=html&fmt=ahah

References

  1. Schmaljohn CS, Dalrymple JM. Analysis of Hantaan virus RNA: evidence for a new genus of Bunyaviridae. Virology 1983; 131:482–491 [View Article]
    [Google Scholar]
  2. Schmaljohn CS, Hasty SE, Harrison SA, Dalrymple JM. Characterization of hantaan virions, the prototype virus of hemorrhagic fever with renal syndrome. J Infect Dis 1983; 148:1005–1012 [View Article]
    [Google Scholar]
  3. Plyusnin A. Genetics of hantaviruses: implications to taxonomy. Arch Virol 2002; 147:665–682 [View Article]
    [Google Scholar]
  4. Virtanen JO, Jääskeläinen KM, Djupsjöbacka J, Vaheri A, Plyusnin A et al. Tula hantavirus NSs protein accumulates in the perinuclear area in infected and transfected cells. Arch Virol 2010; 155:117–121 [View Article]
    [Google Scholar]
  5. Matthys V, Mackow ER. Hantavirus regulation of type I interferon responses. Adv Virol 2012; 2012:1–9 [View Article]
    [Google Scholar]
  6. Briese T, Alkhovsky SV, Beer M, Calisher CH, Charrel RN et al. Bunyavirales proposal. Ictv 20161–45
    [Google Scholar]
  7. Guo WP, Lin XD, Wang W, Tian JH, Cong ML et al. Phylogeny and origins of hantaviruses harbored by bats, insectivores, and rodents. PLoS Pathog 2013; 9:e1003159 [View Article]
    [Google Scholar]
  8. Shi M, Lin XD, Chen X, Tian JH, Chen LJ et al. The evolutionary history of vertebrate RNA viruses. Nature 2018; 556:197–202 [View Article]
    [Google Scholar]
  9. Plyusnin A, Morzunov SP. Virus Evolution and Genetic Diversity of Hantaviruses and Their Rodent Hosts Berlin, Heidelberg: Springer; pp 47–75
    [Google Scholar]
  10. Rowe JE, Wagoner K, Jentes ES, Root JJ, Beaty BJ et al. Epizootiology of Sin Nombre and El Moro canyon hantavirus, southeastern Colorado, 1995-2000. J Wildl Dis 2013; 41:1–11
    [Google Scholar]
  11. Kallio ER, Voutilainen L, Vapalahti O, Vaheri A, Henttonen H et al. Endemic hantavirus infection impairs the winter survival of its rodent host. Ecology 2007; 88:1911–1916 [View Article]
    [Google Scholar]
  12. Kallio ER, Klingström J, Gustafsson E, Manni T, Vaheri A et al. Prolonged survival of Puumala hantavirus outside the host: evidence for indirect transmission via the environment. J Gen Virol 2006; 87:2127–2134 [View Article]
    [Google Scholar]
  13. Botten J, Mirowsky K, Kusewitt D, Ye C, Gottlieb K et al. Persistent Sin Nombre virus infection in the deer mouse (Peromyscus maniculatus) model: sites of replication and strand-specific expression. J Virol 2003; 77:1540–1550 [View Article]
    [Google Scholar]
  14. Miedema K, Schountz T, McGuire A, Fauver J, Rico A et al. Maporal hantavirus causes mild pathology in deer mice (Peromyscus maniculatus). Viruses 2016; 8:286
    [Google Scholar]
  15. Martinez-Valdebenito C, Calvo M, Vial C, Mansilla R, Marco C et al. Person-to-person household and nosocomial transmission of Andes hantavirus, southern Chile, 2011. Emerg Infect Dis 2014; 20:1637–1644 [View Article]
    [Google Scholar]
  16. Brummer-Korvenkontio M, Vaheri A, Hovi T, von Bonsdorff CH, Vuorimies J et al. Nephropathia epidemica: detection of antigen in bank voles and serologic diagnosis of human infection. J Infect Dis 1980; 141:131–134 [View Article]
    [Google Scholar]
  17. Feldmann H, Sanchez A, Morzunov S, Spiropoulou CF, Rollin PE et al. Utilization of autopsy RNA for the synthesis of the nucleocapsid antigen of a newly recognized virus associated with hantavirus pulmonary syndrome. Virus Res 1993; 30:351–367 [View Article]
    [Google Scholar]
  18. Cosgriff TM. Mechanisms of disease in hantavirus infection: pathophysiology of hemorrhagic fever with renal syndrome. Clin Infect Dis 1991; 13:97–107 [View Article]
    [Google Scholar]
  19. Meyer BJ, Schmaljohn CS. Persistent hantavirus infections: characteristics and mechanisms. Trends Microbiol 2000; 8:61–67 [View Article]
    [Google Scholar]
  20. Vapalahti O, Lundkvist A, Kukkonen SK, Cheng Y, Gilljam M et al. Isolation and characterization of Tula virus, a distinct serotype in the genus hantavirus, family Bunyaviridae. J Gen Virol 1996; 77:3063–3067 [View Article]
    [Google Scholar]
  21. Jääskeläinen KM, Plyusnina A, Lundkvist A, Vaheri A, Plyusnin A. Tula hantavirus isolate with the full-length ORF for nonstructural protein NSS survives for more consequent passages in interferon-competent cells than the isolate having truncated NSS ORF. Virol J 2008; 5:3 [View Article]
    [Google Scholar]
  22. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35:495–516 [View Article]
    [Google Scholar]
  23. Li X-D, Mäkelä TP, Guo D, Soliymani R, Koistinen V et al. Hantavirus nucleocapsid protein interacts with the Fas-mediated apoptosis enhancer Daxx. J Gen Virol 2002; 83:759–766 [View Article]
    [Google Scholar]
  24. Park SW, Han MG, Park C, Ju YR, Ahn BY et al. Hantaan virus nucleocapsid protein stimulates MDM2-dependent p53 degradation. J Gen Virol 2013; 94:2424–2428 [View Article]
    [Google Scholar]
  25. Solà-Riera C, Gupta S, Ljunggren H-G, Klingström J. Orthohantaviruses belonging to three phylogroups all inhibit apoptosis in infected target cells. Sci Rep 2019; 9:834 [View Article]
    [Google Scholar]
  26. Gupta S, Braun M, Tischler ND, Stoltz M, Sundström KB et al. Hantavirus-infection confers resistance to cytotoxic lymphocyte-mediated apoptosis. PLoS Pathog 2013; 9:e1003272 [View Article]
    [Google Scholar]
  27. Kramski M, Meisel H, Klempa B, Krüger DH, Pauli G et al. Detection and typing of human pathogenic hantaviruses by real-time reverse transcription-PCR and pyrosequencing. Clin Chem 2007; 53:1899–1905 [View Article]
    [Google Scholar]
  28. McElroy AK, Smith JM, Hooper JW, Schmaljohn CS. Andes virus M genome segment is not sufficient to confer the virulence associated with Andes virus in Syrian hamsters. Virology 2004; 326:130–139 [View Article]
    [Google Scholar]
  29. Ontiveros SJ, Li Q, Jonsson CB. Modulation of apoptosis and immune signaling pathways by the Hantaan virus nucleocapsid protein. Virology 2010; 401:165–178 [View Article]
    [Google Scholar]
  30. Li X-D, Lankinen H, Putkuri N, Vapalahti O, Vaheri A. Tula hantavirus triggers pro-apoptotic signals of ER stress in Vero E6 cells. Virology 2005; 333:180–189 [View Article]
    [Google Scholar]
  31. Markotic A, Hensley L, Geisbert T, Spik K, Schmaljohn C. Hantaviruses induce cytopathic effects and apoptosis in continuous human embryonic kidney cells. J Gen Virol 2003; 84:2197–2202 [View Article]
    [Google Scholar]
  32. Li X-D, Kukkonen S, Vapalahti O, Plyusnin A, Lankinen H et al. Tula hantavirus infection of Vero E6 cells induces apoptosis involving caspase 8 activation. J Gen Virol 2004; 85:3261–3268 [View Article]
    [Google Scholar]
  33. Garg H, Mohl J, Joshi A. HIV-1 induced bystander apoptosis. Viruses 2012; 4:3020–3043 [View Article]
    [Google Scholar]
  34. Karjoo Z, Chen X, Hatefi A. Progress and problems with the use of suicide genes for targeted cancer therapy. Adv Drug Deliv Rev 2016; 99:113–128 [View Article]
    [Google Scholar]
  35. Wolff S, Becker S, Groseth A. Cleavage of the Junin virus nucleoprotein serves a decoy function to inhibit the induction of apoptosis during infection. J Virol 2013; 87:224–233 [View Article]
    [Google Scholar]
  36. Chang YJ, Linh NH, Shih YH, Yu HM, Li MS et al. Alzheimer's amyloid-β sequesters caspase-3 in vitro via its C-terminal tail. ACS Chem Neurosci 2016; 7:1097–1106 [View Article]
    [Google Scholar]
  37. Concannon CG, Orrenius S, Samali A. Hsp27 inhibits cytochrome c-mediated caspase activation by sequestering both pro-caspase-3 and cytochrome c . Gene Expr 2001; 9:195–201 [View Article]
    [Google Scholar]
  38. Hengartner MO, Ellis RE, Horvitz HR. Caenorhabditis elegans gene CED-9 protects cells from programmed cell death. Nature 1992; 356:494–499 [View Article]
    [Google Scholar]
  39. Yan N, Chai J, Lee ES, Gu L, Liu Q et al. Structure of the CED-4-CED-9 complex provides insights into programmed cell death in caenorhabditis elegans. Nature 2005; 437:831–837 [View Article]
    [Google Scholar]
  40. Tan FJ, Fire AZ, Hill RB. Regulation of apoptosis by C. elegans CED-9 in the absence of the C-terminal transmembrane domain. Cell Death Differ 2007; 14:1925–1935 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001291
Loading
/content/journal/jgv/10.1099/jgv.0.001291
Loading

Data & Media loading...

Most cited Most Cited RSS feed