Publikationsserver der Universitätsbibliothek Marburg

Titel:Analysis of the Host Stress Response to Ebola Virus Infection and Generation of a Recombinant Marburg Virus Expressing EGFP to Study Viral Spread
Autor:Schmidt, Kristina Maria
Weitere Beteiligte: Renkawitz-Pohl, Renate (Prof. Dr.)
Veröffentlicht:2012
URI:https://archiv.ub.uni-marburg.de/diss/z2012/0492
DOI: https://doi.org/10.17192/z2012.0492
URN: urn:nbn:de:hebis:04-z2012-04922
DDC: Biowissenschaften, Biologie
Titel (trans.):Interaktion von Ebola-Virus mit der zellulären Stressantwort und Etablierung eines EGFP exprimierenden Marburg-Virus
Publikationsdatum:2012-07-11
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
Marburg-Virus-Krankheit, Processing body, Stressreaktion, VP35, Ebola virus, Recombinant Marburg virus expressing EGFP, Ebola-Virus, PACT, Stress granule, VP30, Fluoreszenz, NP, Protein-Serin-Threonin-Kinasen, PKR

Summary:
Ebola virus (EBOV) and Marburg virus (MARV) belong to the filovirus family and cause outbreaks with case fatality rates up to 90%. Currently there is no approved vaccine or antiviral treatment available. Therefore filoviruses are classified as priority A select agents, which can only be handled in high containment biosafety level 4 laboratories. Generation of an infectious Marburg virus clone expressing EGFP The generation of recombinant viruses expressing enhanced green fluorescent protein (EGFP) has significantly improved the study of their replication cycle and opened up the possibility for the rapid screening of antiviral drugs. The goal of this part of the work was to generate a recombinant MARV expressing EGFP from an additional transcription unit inserted between the second and third genes, encoding VP35 and VP40, respectively. The recombinant MARV containing the EGFP gene (rMARV-EGFP) was successfully rescued and used in live-cell imaging to follow viral spread in real time, revealing EGFP expression at 32 hours post infection (hpi), and infection of neighboring cells at 55 hpi. A slight growth restriction of rMARV-EGFP compared to the wt rMARV was observed which might be due to the additional gene insertion. During filovirus infection characteristic viral inclusions are formed in the cytoplasm of infected cells, induced by self-aggregation of the nucleoprotein (NP). Immunofluorescence analysis of rMARV-EGFP-infected cells revealed an accumulation of EGFP in these viral inclusions. This was reproduced by transient expression of both EGFP and other fluorescent proteins (FPs) along with filovirus nucleocapsid proteins, which further showed that NP-induced inclusion formation was sufficient for the recruitment. In contrast, ectopic GFP fusion proteins containing a localization signal were not relocated into inclusions formed by MARV NP and VP35. Taken together, the observed relocalization of ectopically expressed, untagged FPs suggests an unspecific recruitment to the viral inclusions based on a weak interaction. Interestingly, EGFP aggregates observed by autofluorescence were undetected by antibody-based immunofluorescence. This indicates that antibodies might not be able to penetrate viral inclusions. Cellular stress response to Ebola virus infection EBOV is known to antagonize various antiviral signaling pathways including the interferon response. Here, the interaction of EBOV with another host antiviral defense mechanism, the cellular stress response, was analyzed. During environmental stress a small range of kinases phosphorylate the eukaryotic initiation factor 2α (eIF2α), which plays a central role in the control of translational arrest. Cellular stress is also accompanied by the formation of cytoplasmic stress granules (SGs) and processing bodies (PBs) containing stalled messenger ribonucleoprotein bodies. Since viruses depend on the cellular translation apparatus for viral protein synthesis, many viruses have evolved strategies to antagonize cellular stress response mechanisms. Here we demonstrate that EBOV interferes with the cellular stress response. Since, SGs and PBs are highly dynamic structures, which cannot be isolated from cells, most of the studies presented here were performed by microscopic analysis. Formation of endogenous SGs was not observed in EBOV-infected cells, suggesting that filovirus infection per se does not induce a stress response. This raised the question of whether SG formation is actively inhibited in EBOV-infected cells. The oxidative stressor sodium arsenite (As) induces eIF2α phosphorylation and SG formation. After As- treatment, SGs formed in EBOV-infected cells, albeit in fewer cells compared to non-infected cells, indicating that EBOV is able to inhibit As-induced SG formation to a certain level. For better visualization SG formation was further analyzed in U2OS cells expressing an EGFP-tagged SG maker protein, ras-GAP SH3 domain binding protein 1 (G3BP). In EBOV-infected G3BP-EGFP-expressing cells, SG formation was induced by As and Hippuristanol (Hip). Hip leads to SG formation by inhibiting eIF4A-dependent translation initiation, a process, which does not involve phosphorylation of eIF2α. In some EBOV-infected cells treated with As, G3BP-EGFP was diffusely distributed and SG formation was impaired. This was not seen in Hip-treated cells, suggesting that EBOV inhibits SG formation to a certain level in response to phospho-eIF2α-mediated stress. This inhibition seemed to be dependent on the size of the viral inclusions, suggesting that the level of viral protein expression is important for the inhibition. Intriguingly, G3BP-EGFP SG-like aggregates were observed within the viral inclusions in unstressed and in stressed EBOV-infected cells. Transient expression of viral nucleocapsid proteins leading to inclusion formation was not sufficient for the aggregation of G3BP granules inside the inclusions, suggesting that other viral components, including the viral RNA, are needed to sequester SG components in the inclusions. Together this data suggest a mechanism of EBOV to interfere with SGs by sequestering SG components in the viral inclusions. To further understand how EBOV interacts with SGs, the EBOV RNA-binding proteins NP, VP30, and VP35 were individually examined for their ability to interact with SGs. While VP30 colocalized with SGs without affecting their structure, the double-strand (ds) RNA-binding protein VP35 inhibited SG formation induced by phospho-eIF2α-mediated stress when expressed at high levels. However, when VP30 and VP35 were coexpressed with NP, both proteins were relocated into NP-derived viral inclusions and colocalization with SGs was strongly reduced or absent, which was also observed in EBOV-infected G3BP-EGFP cells. This indicates that only free VP35 and VP30 are able to colocalize with SGs and that both proteins are preferentially relocated to viral inclusions. It is conceivable that the amount of free VP35 and VP30 in infected cells is too low to be detected by IFA. In As-treated cells the dsRNA-dependent protein kinase (PKR) is activated by the cellular protein, PKR activating protein (PACT). Activated PKR phosphorylates eIF2α, inducing SG formation. In EBOV-infected cells, activation of PKR by dsRNA has been shown to be efficiently inhibited by VP35. Our data revealed that EBOV was not able to block As-induced PKR activation. This indicates that PKR activation by PACT cannot be antagonized by VP35. Previous studies have reported that VP35 binds to PACT in unstressed cells (Fabozzi et al., 2011) Here we show that the VP35-PACT interaction was disrupted in stressed cells. This indicates that VP35 loses the ability to sequester PACT from binding to PKR, such that PKR can be activated. This further explains the presence of SGs in EBOV-infected cells but the questions remains, how eIF2α-induced SG formation is inhibited in EBOV-infected and VP35 expressing cells? Possible mechanisms include the inhibitory function of VP35 on SG formation or the sequestering of SG components. To analyze the role of PBs in EBOV infection, a U2OS cell line expressing the PB marker protein mRNA-decapping enzyme 1A (DCP1a) fused to mRFP was used. In EBOV-infected cells, As-induced PBs were observed to surround viral inclusions, suggesting a recruitment of PBs. In transient transfection experiments, PBs colocalized with VP35 but not with VP30 or NP. Expression of VP35 in U2OS cells containing both G3BP-EGFP and DCP1a-mRFP resulted in diffusely aggregated SGs that colocalized with VP35 and intermingled with PBs. This suggests that VP35 interacts and links constituents of PBs and SGs. The results indicate that EBOV exhibits antiviral control strategies at the level of the host stress response, where VP35 functions as a key player, since it directly interacts with cellular stress components of both SGs and PBs. Furthermore, the binding between VP35 and PACT seems to play an important role in the control of PKR.

Bibliographie / References

  1. Kedersha, N.L., Gupta, M., Li, W., Miller, I., Anderson, P., 1999. RNA-binding proteins TIA-1 and TIAR link the phosphorylation of eIF-2 alpha to the assembly of mammalian stress granules. J Cell Biol 147, 1431–1442.
  2. García-Mata, R., Bebök, Z., Sorscher, E.J., Sztul, E.S., 1999. Characterization and dynamics of aggresome formation by a cytosolic GFP-chimera. J Cell Biol 146, 1239–1254.
  3. Chu, C.-Y., Rana, T.M., 2006. Translation repression in human cells by microRNA-induced gene silencing requires RCK/p54. PLoS Biol 4, e210.
  4. Valmas, C., Grosch, M.N., Schümann, M., Olejnik, J., Martinez, O., Best, S.M., Krähling, V., Basler, C.F., Mühlberger, E., 2010. Marburg Virus Evades Interferon Responses by a Mechanism Distinct from Ebola Virus. PLoS Pathog. 6, e1000721.
  5. Montero, H., Rojas, M., Arias, C.F., Lopez, S., 2008. Rotavirus Infection Induces the Phosphorylation of eIF2 but Prevents the Formation of Stress Granules. J Virol 82, 1496– 1504.
  6. Slenczka, W.G., 1999. The Marburg virus outbreak of 1967 and subsequent episodes. Curr Top Microbiol Immunol 235, 49–75.
  7. Hartman, A.L., Towner, J.S., Nichol, S.T., 2004. A C-terminal basic amino acid motif of Zaire ebolavirus VP35 is essential for type I interferon antagonism and displays high identity with the RNA-binding domain of another interferon antagonist, the NS1 protein of influenza A virus. Virology 328, 177–184.
  8. REID, S., CARDENAS, W., BASLER, C., 2005. Homo-oligomerization facilitates the interferon- antagonist activity of the ebolavirus VP35 protein. Virology 341, 179–189.
  9. Hoenen, T., Jung, S., Herwig, A., Groseth, A., Becker, S., 2010. Both matrix proteins of Ebola virus contribute to the regulation of viral genome replication and transcription. Virology 403, 56–66.
  10. Kedersha, N., Anderson, P., 2002. Stress granules: sites of mRNA triage that regulate mRNA stability and translatability. Biochem. Soc. Trans. 30, 963–969.
  11. Bharat, T.A.M., Noda, T., Riches, J.D., Kraehling, V., Kolesnikova, L., Becker, S., Kawaoka, Y., Briggs, J.A.G., 2012. Structural dissection of Ebola virus and its assembly determinants using cryo-electron tomography. Proceedings of the National Academy of Sciences.
  12. Modrof, J., Mühlberger, E., Klenk, H.-D., Becker, S., 2002. Phosphorylation of VP30 impairs ebola virus transcription. J Biol Chem 277, 33099–33104.
  13. McCampbell, A., Taylor, J.P., Taye, A.A., Robitschek, J., Li, M., Walcott, J., Merry, D., Chai, Y., Paulson, H., Sobue, G., Fischbeck, K.H., 2000. CREB-binding protein sequestration by expanded polyglutamine. Human Molecular Genetics 9, 2197–2202.
  14. Schmidt, K.M., Schümann, M., Olejnik, J., Krähling, V., Mühlberger, E., 2011. Recombinant Marburg virus expressing EGFP allows rapid screening of virus growth and real-time visualization of virus spread. J Infect Dis 204 Suppl 3, S861–70.
  15. Mateo, M., Carbonnelle, C., Martinez, M.J., Reynard, O., Page, A., Volchkova, V.A., Volchkov, V.E., 2011. Knockdown of Ebola Virus VP24 Impairs Viral Nucleocapsid Assembly and Prevents Virus Replication. J Infect Dis 204, S892–S896.
  16. Martínez, M.J., Volchkova, V.A., Raoul, H., Alazard-Dany, N., Reynard, O., Volchkov, V.E., 2011. Role of VP30 phosphorylation in the Ebola virus replication cycle. J Infect Dis 204 Suppl 3, S934–40.
  17. Noda, T., Hagiwara, K., Sagara, H., Kawaoka, Y., 2010. Characterization of the Ebola virus nucleoprotein-RNA complex. Journal of General Virology 91, 1478–1483.
  18. Franks, T.M., Lykke-Andersen, J., 2007. TTP and BRF proteins nucleate processing body formation to silence mRNAs with AU-rich elements. Genes & Development 21, 719–735.
  19. Feng, Z., Cerveny, M., Yan, Z., He, B., 2006. The VP35 Protein of Ebola Virus Inhibits the Antiviral Effect Mediated by Double-Stranded RNA-Dependent Protein Kinase PKR. J Virol 81, 182– 192.
  20. Martínez, M.J., Biedenkopf, N., Volchkova, V., Hartlieb, B., Alazard-Dany, N., Reynard, O., Becker, S., Volchkov, V., 2008. Role of Ebola virus VP30 in transcription reinitiation. J Virol 82, 12569– 12573.
  21. Cardenas, W.B., Loo, Y.M., Gale, M., Hartman, A.L., Kimberlin, C.R., Martinez-Sobrido, L., Saphire, E.O., Basler, C.F., 2006. Ebola Virus VP35 Protein Binds Double-Stranded RNA and Inhibits Alpha/Beta Interferon Production Induced by RIG-I Signaling. J Virol 80, 5168–5178.
  22. Weik, M., Modrof, J., Klenk, H.-D., Becker, S., Mühlberger, E., 2002. Ebola virus VP30-mediated transcription is regulated by RNA secondary structure formation. J Virol 76, 8532–8539.
  23. Basler, C.F., Mikulasova, A., Martinez-Sobrido, L., Paragas, J., Mühlberger, E., Bray, M., Klenk, H.-D., Palese, P., García-Sastre, A., 2003. The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. J Virol 77, 7945–7956.
  24. Yang, Z., 2004. GW182 is critical for the stability of GW bodies expressed during the cell cycle and cell proliferation. J. Cell. Sci. 117, 5567–5578.
  25. Wilczynska, A., 2005. The translational regulator CPEB1 provides a link between dcp1 bodies and stress granules. J. Cell. Sci. 118, 981–992.
  26. Souquere, S., Mollet, S., Kress, M., Dautry, F., Pierron, G., Weil, D., 2009. Unravelling the ultrastructure of stress granules and associated P-bodies in human cells. J. Cell. Sci. 122, 3619–3626.
  27. Abrahamyan, L.G., Chatel-Chaix, L., Ajamian, L., Milev, M.P., Monette, A., Clément, J.-F., Song, R., Lehmann, M., DesGroseillers, L., Laughrea, M., Boccaccio, G., Mouland, A.J., 2010. Novel Staufen1 ribonucleoproteins prevent formation of stress granules but favour encapsidation of HIV-1 genomic RNA. J. Cell. Sci. 123, 369–383.
  28. Yang, W.H., 2006. RNA-associated protein 55 (RAP55) localizes to mRNA processing bodies and stress granules. RNA 12, 547–554.
  29. Andrei, M.A., 2005. A role for eIF4E and eIF4E-transporter in targeting mRNPs to mammalian processing bodies. RNA 11, 717–727.
  30. Yang, W.H., Bloch, D.B., 2007. Probing the mRNA processing body using protein macroarrays and " autoantigenomics. " RNA 13, 704–712.
  31. Bedard, K.M., Walter, B.L., Semler, B.L., 2004. Multimerization of poly(rC) binding protein 2 is required for translation initiation mediated by a viral IRES. RNA 10, 1266–1276.
  32. Teixeira, D., Sheth, U., Valencia-Sanchez, M.A., Brengues, M., Parker, R., 2005. Processing bodies require RNA for assembly and contain nontranslating mRNAs. RNA 11, 371–382.
  33. Fujimura, K., Kano, F., Murata, M., 2008. Identification of PCBP2, a facilitator of IRES-mediated translation, as a novel constituent of stress granules and processing bodies. RNA 14, 425– 431.
  34. Bharat, T.A.M., Riches, J.D., Kolesnikova, L., Welsch, S., Krähling, V., Davey, N., Parsy, M.-L., Becker, S., Briggs, J.A.G., 2011. Cryo-Electron Tomography of Marburg Virus Particles and Their Morphogenesis within Infected Cells. PLoS Biol 9, e1001196.
  35. Olejnik, J., Ryabchikova, E., Corley, R.B., Mühlberger, E., 2011. Intracellular events and cell fate in filovirus infection. Viruses 3, 1501–1531.
  36. Ramanan, P., Shabman, R.S., Brown, C.S., Amarasinghe, G.K., Basler, C.F., Leung, D.W., 2011. Filoviral Immune Evasion Mechanisms. Viruses 3, 1634–1649.
  37. Ryabchikova, E., Kolesnikova, L., Smolina, M., Tkachev, V., Pereboeva, L., Baranova, S., Grazhdantseva, A., Rassadkin, Y., 1996. Ebola virus infection in guinea pigs: presumable role of granulomatous inflammation in pathogenesis. Arch Virol 141, 909–921.
  38. Imataka, H., Gradi, A., Sonenberg, N., 1998. A newly identified N-terminal amino acid sequence of human eIF4G binds poly(A)-binding protein and functions in poly(A)-dependent translation. EMBO J. 17, 7480–7489.
  39. Chien, C.Y., Tejero, R., Huang, Y., Zimmerman, D.E., Rios, C.B., Krug, R.M., Montelione, G.T., 1997. A novel RNA-binding motif in influenza A virus non-structural protein 1. Nat Struct Biol 4, 891–895.
  40. Berlanga, J.J., Ventoso, I., Harding, H.P., Deng, J., Ron, D., Sonenberg, N., Carrasco, L., de Haro, C., 2006. Antiviral effect of the mammalian translation initiation factor 2alpha kinase GCN2 against RNA viruses. EMBO J. 25, 1730–1740.
  41. Parker, F., Maurier, F., Delumeau, I., Duchesne, M., Faucher, D., Debussche, L., Dugue, A., Schweighoffer, F., Tocque, B., 1996. A Ras-GTPase-activating protein SH3-domain-binding protein. Molecular and Cellular Biology 16, 2561–2569.
  42. Kolesnikova, L., Bohil, A.B., Cheney, R.E., Becker, S., 2007. Budding of Marburgvirus is associated with filopodia. Cell Microbiol 9, 939–951.
  43. Cougot, N., van Dijk, E., Babajko, S., Séraphin, B., 2004. " Cap-tabolism. " Trends in Biochemical Sciences 29, 436–444.
  44. Kedersha, N., Tisdale, S., Hickman, T., Anderson, P., 2008. Chapter 26 -Real-Time and Quantitative Imaging of Mammalian Stress Granules and Processing Bodies. RNA Turnover in Eukaryotes: Nucleases, Pathways and Anaylsis of mRNA Decay 448, 521–552.
  45. Berlanga, J.J., Santoyo, J., De Haro, C., 1999. Characterization of a mammalian homolog of the GCN2 eukaryotic initiation factor 2alpha kinase. Eur J Biochem 265, 754–762.
  46. Complementary Minus-Strand RNA and Facilitate Virus Replication. J Virol 76, 11989–12000.
  47. Wek, R.C., Jiang, H.-Y., Anthony, T.G., 2006. Coping with stress: eIF2 kinases and translational control. Biochem. Soc. Trans. 34, 7–11.
  48. Liu, J., Lynch, P.A., Chien, C.Y., Montelione, G.T., Krug, R.M., Berman, H.M., 1997. Crystal structure of the unique RNA-binding domain of the influenza virus NS1 protein. Nat Struct Biol 4, 896–899.
  49. Baize, S., Leroy, E.M., Georges-Courbot, M.C., Capron, M., Lansoud-Soukate, J., Debre, P., Fisher-Hoch, S.P., McCormick, J.B., Georges, A.J., 1999. Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nat Med 5, 423–426.
  50. Jakymiw, A., Lian, S., Eystathioy, T., Li, S., Satoh, M., Hamel, J.C., Fritzler, M.J., Chan, E.K.L., 2005. Disruption of GW bodies impairs mammalian RNA interference. Nat Cell Biol 7, 1167– 1174.
  51. Feldmann, H., Geisbert, T.W., 2011. Ebola haemorrhagic fever. The Lancet 377, 849–862.
  52. Watanabe, S., Noda, T., Halfmann, P., Jasenosky, L., Kawaoka, Y., 2007. Ebola virus (EBOV) VP24 inhibits transcription and replication of the EBOV genome. J Infect Dis 196 Suppl 2, S284–90.
  53. Prins, K.C., Cardenas, W.B., Basler, C.F., 2009. Ebola Virus Protein VP35 Impairs the Function of Interferon Regulatory Factor-Activating Kinases IKK and TBK-1. J Virol 83, 3069–3077.
  54. John, S.P., Wang, T., Steffen, S., Longhi, S., Schmaljohn, C.S., Jonsson, C.B., 2007. Ebola Virus References Page 102 VP30 Is an RNA Binding Protein. J Virol 81, 8967–8976.
  55. Modrof, J., Becker, S., Mühlberger, E., 2003. Ebola virus transcription activator VP30 is a zinc- binding protein. J Virol 77, 3334–3338.
  56. Rogers, G.W.J., Komar, A.A., Merrick, W.C., 2002. eIF4A: the godfather of the DEAD box helicases. Prog Nucleic Acid Res Mol Biol 72, 307–331.
  57. Kedersha, N., Chen, S., Gilks, N., Li, W., Miller, I.J., Stahl, J., Anderson, P., 2002. Evidence that ternary complex (eIF2-GTP-tRNA(i)(Met))-deficient preinitiation complexes are core constituents of mammalian stress granules. Mol. Biol. Cell 13, 195–210.
  58. Chudakov, D.M., Matz, M.V., Lukyanov, S., Lukyanov, K.A., 2010. Fluorescent proteins and their applications in imaging living cells and tissues. Physiol Rev 90, 1103–1163.
  59. Bordeleau, M.-E., Mori, A., Oberer, M., Lindqvist, L., Chard, L.S., Higa, T., Belsham, G.J., Wagner, G., Tanaka, J., Pelletier, J., 2006. Functional characterization of IRESes by an inhibitor of the RNA helicase eIF4A. Nat Chem Biol 2, 213–220.
  60. Towner, J.S., Paragas, J., Dover, J.E., Gupta, M., Goldsmith, C.S., Huggins, J.W., Nichol, S.T., 2005. Generation of eGFP expressing recombinant Zaire ebolavirus for analysis of early pathogenesis events and high-throughput antiviral drug screening. Virology 332, 20–27.
  61. Muhlberger, E., 2004. Genome organization replication and transcription of filoviruses, in: Klenk, H.-D., Feldmann, H. (Eds.), Ebola and Marburg Viruses: Molecular and Cellular Biology. Horizon Bioscience.
  62. Han, A.P., Yu, C., Lu, L., Fujiwara, Y., Browne, C., Chin, G., Fleming, M., Leboulch, P., Orkin, S.H., Chen, J.J., 2001. Heme-regulated eIF2alpha kinase (HRI) is required for translational regulation and survival of erythroid precursors in iron deficiency. EMBO J. 20, 6909–6918.
  63. McEwen, E., 2005. Heme-regulated Inhibitor Kinase-mediated Phosphorylation of Eukaryotic Translation Initiation Factor 2 Inhibits Translation, Induces Stress Granule Formation, and Mediates Survival upon Arsenite Exposure. Journal of Biological Chemistry 280, 16925– 16933.
  64. Kok, K.H., Ng, M.-H.J., Ching, Y.-P., Jin, D.-Y., 2007. Human TRBP and PACT directly interact with each other and associate with dicer to facilitate the production of small interfering RNA. J Biol Chem 282, 17649–17657.
  65. Shi, Y., Vattem, K.M., Sood, R., An, J., Liang, J., Stramm, L., Wek, R.C., 1998. Identification and characterization of pancreatic eukaryotic initiation factor 2 alpha-subunit kinase, PEK, involved in translational control. Molecular and Cellular Biology 18, 7499–7509.
  66. McInerney, G.M., Kedersha, N.L., Kaufman, R.J., Anderson, P., Liljeström, P., 2005. Importance of eIF2alpha phosphorylation and stress granule assembly in alphavirus translation regulation. Mol. Biol. Cell 16, 3753–3763.
  67. Shaner, N.C., Campbell, R.E., Steinbach, P.A., Giepmans, B.N.G., Palmer, A.E., Tsien, R.Y., 2004. Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22, 1567–1572.
  68. Gil, J., Esteban, M., 2000. Induction of apoptosis by the dsRNA-dependent protein kinase (PKR): mechanism of action. Apoptosis 5, 107–114.
  69. White, J.P., Cardenas, A.M., Marissen, W.E., Lloyd, R.E., 2007. Inhibition of Cytoplasmic mRNA Stress Granule Formation by a Viral Proteinase. Cell Host Microbe 2, 295–305.
  70. Mazroui, R., Sukarieh, R., Bordeleau, M.-E., Kaufman, R.J., Northcote, P., Tanaka, J., Gallouzi, I., Pelletier, J., 2006. Inhibition of ribosome recruitment induces stress granule formation independently of eukaryotic initiation factor 2alpha phosphorylation. Mol. Biol. Cell 17, 4212– 4219.
  71. Liu, S., 2006. Insertion and deletion analyses identify regions of non-structural protein 5A of Hepatitis C virus that are dispensable for viral genome replication. Journal of General Virology 87, 323–327.
  72. Laraki, G., Clerzius, G., Daher, A., Melendez-Peña, C., Daniels, S., Gatignol, A., 2008. Interactions between the double-stranded RNA-binding proteins TRBP and PACT define the Medipal domain that mediates protein-protein interactions. RNA Biol 5, 92–103.
  73. Becker, S., Rinne, C., Hofsäss, U., Klenk, H.D., Muhlberger, E., 1998. Interactions of Marburg virus nucleocapsid proteins. Virology 249, 406–417.
  74. Linero, F.N., Thomas, M.G., Boccaccio, G.L., Scolaro, L.A., 2011. Junin virus infection impairs stress-granule formation in Vero cells treated with arsenite via inhibition of eIF2 phosphorylation. Journal of General Virology 92, 2889–2899.
  75. Goodin, M., Yelton, S., Ghosh, D., Mathews, S., Lesnaw, J., 2005. Live-cell imaging of rhabdovirus-induced morphological changes in plant nuclear membranes. Mol. Plant Microbe Interact. 18, 703–709.
  76. Kahvejian, A., Svitkin, Y.V., Sukarieh, R., M'Boutchou, M.-N., Sonenberg, N., 2005. Mammalian poly(A)-binding protein is a eukaryotic translation initiation factor, which acts via multiple mechanisms. Genes & Development 19, 104–113.
  77. Martini, G.A., 1973. Marburg virus disease. Postgrad Med J 49, 542–546.
  78. Lu, L., Chen, J.J., 2002. Molecular cloning and characterization of the promoter of mouse heme- regulated eIF2alpha kinase. Biochim Biophys Acta 1574, 193–199.
  79. Brengues, M., Teixeira, D., Parker, R., 2005. Movement of eukaryotic mRNAs between polysomes and cytoplasmic processing bodies. Science 310, 486–489.
  80. Patel, C.V., 2000. PACT, a Stress-modulated Cellular Activator of Interferon-induced Double- stranded RNA-activated Protein Kinase, PKR. Journal of Biological Chemistry 275, 37993– 37998.
  81. Li, S., Sen, G.C., 2003. PACT-mediated enhancement of reporter gene expression at the translational level. J. Interferon Cytokine Res. 23, 689–697.
  82. Geisbert, T.W., Young, H.A., Jahrling, P.B., Davis, K.J., Larsen, T., Kagan, E., Hensley, L.E., 2003. Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. Am. J. Pathol. 163, 2371–2382.
  83. Lyakhov, D.L., He, B., Zhang, X., Studier, F.W., Dunn, J.J., McAllister, W.T., 1998. Pausing and termination by bacteriophage T7 RNA polymerase. Journal of Molecular Biology 280, 201–213.
  84. Eulalio, A., Behm-Ansmant, I., Izaurralde, E., 2007. P bodies: at the crossroads of post- transcriptional pathways. Nature Publishing Group 8, 9–22.
  85. Harding, H.P., Zhang, Y., Bertolotti, A., Zeng, H., Ron, D., 2000. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol. Cell 5, 897–904.
  86. Srivastava, S.P., Kumar, K.U., Kaufman, R.J., 1998. Phosphorylation of eukaryotic translation initiation factor 2 mediates apoptosis in response to activation of the double-stranded RNA- dependent protein kinase. J Biol Chem 273, 2416–2423.
  87. Peters, G.A., Li, S., Sen, G.C., 2006. Phosphorylation of specific serine residues in the PKR activation domain of PACT is essential for its ability to mediate apoptosis. J Biol Chem 281, 35129–35136.
  88. Williams, B.R., 1999. PKR; a sentinel kinase for cellular stress. Oncogene 18, 6112–6120.
  89. Harding, H.P., Zhang, Y., Ron, D., 1999. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 397, 271–274.
  90. References Page 101
  91. References Page 103
  92. References Page 105
  93. Harding, H.P., Novoa, I., Zhang, Y., Zeng, H., Wek, R., Schapira, M., Ron, D., 2000. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 6, 1099–1108.
  94. Sanchez, A., Kiley, M.P., Klenk, H.D., Feldmann, H., 1992. Sequence analysis of the Marburg virus nucleoprotein gene: comparison to Ebola virus and other non-segmented negative-strand RNA viruses. J. Gen. Virol. 73 ( Pt 2), 347–357.
  95. Williams, B.R., 2001. Signal integration via PKR. Sci. STKE 2001, re2.
  96. Singh, M., Castillo, D., Patel, C.V., Patel, R.C., 2011. Stress-induced phosphorylation of PACT reduces its interaction with TRBP and leads to PKR activation. Biochemistry 50, 4550–4560.
  97. Huang, Y., Xu, L., Sun, Y., Nabel, G.J., 2002. The assembly of Ebola virus nucleocapsid requires virion-associated proteins 35 and 24 and posttranslational modification of nucleoprotein. Mol. Cell 10, 307–316.
  98. Gupta, V., 2003. The carboxy-terminal, M3 motifs of PACT and TRBP have opposite effects on PKR activity. Virology 315, 283–291.
  99. Hovanessian, A.G., 1989. The double stranded RNA-activated protein kinase induced by interferon: dsRNA-PK. J Interferon Res 9, 641–647.
  100. Basler, C.F., Wang, X., Muhlberger, E., Volchkov, V., Paragas, J., Klenk, H.D., Garcia-Sastre, A., Palese, P., 2000. The Ebola virus VP35 protein functions as a type I IFN antagonist. Proc Natl Acad Sci USA 97, 12289–12294.
  101. Haasnoot, J., de Vries, W., Geutjes, E.-J., Prins, M., de Haan, P., Berkhout, B., 2007. The Ebola virus VP35 protein is a suppressor of RNA silencing. PLoS Pathog. 3, e86.
  102. van Hoof, A., Parker, R., 1999. The exosome: a proteasome for RNA? Cell 99, 347–350.
  103. Atlas, R., Behar, L., Elliott, E., Ginzburg, I., 2004. The insulin-like growth factor mRNA binding- protein IMP-1 and the Ras-regulatory protein G3BP associate with tau mRNA and HuD protein in differentiated P19 neuronal cells. J Neurochem 89, 613–626.
  104. Muhlberger, E., Sanchez, A., Randolf, A., Will, C., Kiley, M.P., Klenk, H.D., Feldmann, H., 1992. The nucleotide sequence of the L gene of Marburg virus, a filovirus: homologies with paramyxoviruses and rhabdoviruses. Virology 187, 534–547.
  105. Mellor, H., 2010. The role of formins in filopodia formation. Biochim Biophys Acta 1803, 191–200.
  106. Muhlberger, E., Lotfering, B., Klenk, H.D., Becker, S., 1998. Three of the four nucleocapsid proteins of Marburg virus, NP, VP35, and L, are sufficient to mediate replication and transcription of Marburg virus-specific monocistronic minigenomes. J Virol 72, 8756–8764.
  107. Duprex, P., Rima, B., 2011. Using Green Fluorescent Protein to Monitor Measels Virus cell-to-cell spread by time-lapse confocal microscopy 1–11.
  108. Hierholzer, J., Killington, R., 1996. Virus isolation and quantitation. Virology methods manual.
  109. Khaperskyy, D.A., Hatchette, T.F., McCormick, C., 2011. Influenza A virus inhibits cytoplasmic stress granule formation. The FASEB Journal.
  110. Zhou, C., Arslan, F., Wee, S., Krishnan, S., Ivanov, A.R., Oliva, A., Leatherwood, J., Wolf, D.A., 2005. PCI proteins eIF3e and eIF3m define distinct translation initiation factor 3 complexes. BMC Biol 3, 14.
  111. Bradfute, S.B., Warfield, K.L., Bavari, S., 2008. Functional CD8+ T cell responses in lethal Ebola virus infection. J Immunol 180, 4058–4066.
  112. Ohn, T., Kedersha, N., Hickman, T., Tisdale, S., Anderson, P., 2008. A functional RNAi screen links O-GlcNAc modification of ribosomal proteins to stress granule and processing body assembly. Nat Cell Biol 10, 1224–1231.
  113. Eystathioy, T., Chan, E.K.L., Tenenbaum, S.A., Keene, J.D., Griffith, K., Fritzler, M.J., 2002. A phosphorylated cytoplasmic autoantigen, GW182, associates with a unique population of human mRNAs within novel cytoplasmic speckles. Mol. Biol. Cell 13, 1338–1351.
  114. Gallouzi, I.E., Parker, F., Chebli, K., Maurier, F., Labourier, E., Barlat, I., Capony, J.P., Tocque, B., Tazi, J., 1998. A novel phosphorylation-dependent RNase activity of GAP-SH3 binding protein: a potential link between signal transduction and RNA stability. Molecular and Cellular Biology 18, 3956–3965.
  115. Kolesnikova, L., Muhlberger, E., Ryabchikova, E., Becker, S., 2000. Ultrastructural Organization of Recombinant Marburg Virus Nucleoprotein: Comparison with Marburg Virus Inclusions. J Virol 74, 3899–3904.
  116. Duprex, W.P., McQuaid, S., Hangartner, L., Billeter, M.A., Rima, B.K., 1999. Observation of measles virus cell-to-cell spread in astrocytoma cells by using a green fluorescent protein- expressing recombinant virus. J Virol 73, 9568–9575.
  117. Benkirane, M., Neuveut, C., Chun, R.F., Smith, S.M., Samuel, C.E., Gatignol, A., Jeang, K.T., 1997. Oncogenic potential of TAR RNA binding protein TRBP and its regulatory interaction with RNA-dependent protein kinase PKR. EMBO J. 16, 611–624.
  118. Patel, R.C., Sen, G.C., 1998. PACT, a protein activator of the interferon-induced protein kinase, PKR. EMBO J. 17, 4379–4390.
  119. Enterlein, S., Volchkov, V., Weik, M., Kolesnikova, L., Volchkova, V., Klenk, H.-D., Mühlberger, E., 2006. Rescue of recombinant Marburg virus from cDNA is dependent on nucleocapsid protein VP30. J Virol 80, 1038–1043.
  120. Peters, G.A., Khoo, D., Mohr, I., Sen, G.C., 2002. Inhibition of PACT-mediated activation of PKR by the herpes simplex virus type 1 Us11 protein. J Virol 76, 11054–11064.
  121. Noda, T., Ebihara, H., Muramoto, Y., Fujii, K., Takada, A., Sagara, H., Kim, J.H., Kida, H., Feldmann, H., Kawaoka, Y., 2006. Assembly and budding of Ebolavirus. PLoS Pathog. 2, 864–872.
  122. Leung, A.K.L., Calabrese, J.M., Sharp, P.A., 2006. Quantitative analysis of Argonaute protein reveals microRNA-dependent localization to stress granules. Proc Natl Acad Sci USA 103, 18125–18130.
  123. Sheth, U., Parker, R., 2006. Targeting of aberrant mRNAs to cytoplasmic processing bodies. Cell References Page 106 125, 1095–1109.
  124. Sheth, U., Parker, R., 2003. Decapping and decay of messenger RNA occur in cytoplasmic processing bodies. Science 300, 805–808.
  125. Emara, M.M., Brinton, M.A., 2007. Interaction of TIA-1/TIAR with West Nile and dengue virus products in infected cells interferes with stress granule formation and processing body assembly. Proc Natl Acad Sci USA 104, 9041–9046.
  126. Burkhardt, J.K., Echeverri, C.J., Nilsson, T., Vallee, R.B., 1997. Overexpression of the dynamitin (p50) subunit of the dynactin complex disrupts dynein-dependent maintenance of membrane organelle distribution. J Cell Biol 139, 469–484.
  127. Gradi, A., Svitkin, Y.V., Imataka, H., Sonenberg, N., 1998. Proteolysis of human eukaryotic translation initiation factor eIF4GII, but not eIF4GI, coincides with the shutoff of host protein synthesis after poliovirus infection. Proc Natl Acad Sci USA 95, 11089–11094.
  128. Kedersha, N., Stoecklin, G., Ayodele, M., Yacono, P., Lykke-Andersen, J., Fritzler, M.J., Scheuner, D., Kaufman, R.J., Golan, D.E., Anderson, P., 2005. Stress granules and processing bodies are dynamically linked sites of mRNP remodeling. J Cell Biol 871–884.
  129. Tourrière, H., 2003. The RasGAP-associated endoribonuclease G3BP assembles stress granules. J Cell Biol 160, 823–831.
  130. Halfmann, P., Kim, J.H., Ebihara, H., Noda, T., Neumann, G., Feldmann, H., Kawaoka, Y., 2008. Generation of biologically contained Ebola viruses. Proceedings of the National Academy of Sciences 105, 1129–1133.
  131. Habjan, M., Andersson, I., Klingström, J., Schümann, M., Martin, A., Zimmermann, P., Wagner, V., Pichlmair, A., Schneider, U., Mühlberger, E., Mirazimi, A., Weber, F., 2008. Processing of Genome 5% Termini as a Strategy of Negative-Strand RNA Viruses to Avoid RIG-I-Dependent Interferon Induction. PLoS ONE 3, e2032.
  132. Mollet, S., Cougot, N., Wilczynska, A., Dautry, F., Kress, M., Bertrand, E., Weil, D., 2008. Translationally repressed mRNA transiently cycles through stress granules during stress. Mol. Biol. Cell 19, 4469–4479.
  133. Daher, A., Laraki, G., Singh, M., Melendez-Peña, C.E., Bannwarth, S., Peters, A.H.F.M., Meurs, E.F., Braun, R.E., Patel, R.C., Gatignol, A., 2009. TRBP control of PACT-induced phosphorylation of protein kinase R is reversed by stress. Molecular and Cellular Biology 29, 254–265.
  134. Franks, T.M., Lykke-Andersen, J., 2008. The Control of mRNA Decapping and P-Body Formation. Mol. Cell 32, 605–615.
  135. Enterlein, S., Schmidt, K.M., Schümann, M., Conrad, D., Krähling, V., Olejnik, J., Mühlberger, E., 2009. The marburg virus 3' noncoding region structurally and functionally differs from that of ebola virus. J Virol 83, 4508–4519.
  136. Chang, T.-H., Kubota, T., Matsuoka, M., Jones, S., Bradfute, S.B., Bray, M., Ozato, K., 2009. Ebola Zaire Virus Blocks Type I Interferon Production by Exploiting the Host SUMO Modification Machinery. PLoS Pathog. 5, e1000493.
  137. Schumann, M., Gantke, T., Muhlberger, E., 2009. Ebola Virus VP35 Antagonizes PKR Activity through Its C-Terminal Interferon Inhibitory Domain. J Virol 83, 8993–8997.
  138. Qin, Q., Hastings, C., Miller, C.L., 2009. Mammalian Orthoreovirus Particles Induce and Are Recruited into Stress Granules at Early Times Postinfection. J Virol 83, 11090–11101.
  139. Hanley, L.L., McGivern, D.R., Teng, M.N., Djang, R., Collins, P.L., Fearns, R., 2010. Roles of the respiratory syncytial virus trailer region: Effects of mutations on genome production and stress granule formation. Virology 406, 241–252.
  140. Lindquist, M.E., Lifland, A.W., Utley, T.J., Santangelo, P.J., Crowe, J.E., 2010. Respiratory Syncytial Virus Induces Host RNA Stress Granules To Facilitate Viral Replication. J Virol 84, 12274–12284.
  141. Fabozzi, G., Nabel, C.S., Dolan, M.A., Sullivan, N.J., 2011. Ebolavirus Proteins Suppress the Effects of Small Interfering RNA by Direct Interaction with the Mammalian RNA Interference Pathway. J Virol 85, 2512–2523.
  142. Lindquist, M.E., Mainou, B.A., Dermody, T.S., Crowe, J.E., Jr, 2011. Activation of protein kinase R is required for induction of stress granules by respiratory syncytial virus but dispensable for viral replication. Virology 413, 103–110.
  143. Kuhn, J.H., Becker, S., Ebihara, H., Geisbert, T.W., Johnson, K.M., Kawaoka, Y., Lipkin, W.I., Negredo, A.I., Netesov, S.V., Nichol, S.T., Palacios, G., Peters, C.J., Tenorio, A., Volchkov, V.E., Jahrling, P.B., 2010. Proposal for a revised taxonomy of the family Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol 155, 2083–2103.
  144. Yi, Z., Pan, T., Wu, X., Song, W., Wang, S., Xu, Y., Rice, C.M., MacDonald, M.R., Yuan, Z., 2011. Hepatitis C Virus Co-Opts Ras-GTPase-Activating Protein-Binding Protein 1 for Its Genome Replication. J Virol 85, 6996–7004.
  145. Ariumi, Y., Kuroki, M., Kushima, Y., Osugi, K., Hijikata, M., Maki, M., Ikeda, M., Kato, N., 2011. Hepatitis C Virus Hijacks P-Body and Stress Granule Components around Lipid Droplets. J Virol 85, 6882–6892.
  146. Maggi, L.B.J., Heitmeier, M.R., Scheuner, D., Kaufman, R.J., Buller, R.M., Corbett, J.A., 2000. Potential role of PKR in double-stranded RNA-induced macrophage activation. EMBO J. 19, 3630–3638.
  147. Ward, A.M., Bidet, K., Yinglin, A., Ler, S.G., Hogue, K., Blackstock, W., Gunaratne, J., Garcia- Blanco, M.A., 2011. Quantitative mass spectrometry of DENV-2 RNA-interacting proteins reveals that the DEAD-box RNA helicase DDX6 binds the DB1 and DB2 3' UTR structures. References Page 107 RNA Biol 8, 1173–1186.
  148. Matthews, J.D., Frey, T.K., 2011. Analysis of subcellular G3BP redistribution during rubella virus infection. Journal of General Virology.
  149. Nover, L., Scharf, K.D., Neumann, D., 1989. Cytoplasmic heat shock granules are formed from precursor particles and are associated with a specific set of mRNAs. Molecular and Cellular Biology 9, 1298–1308.
  150. Nover, L., Scharf, K.D., Neumann, D., 1983. Formation of cytoplasmic heat shock granules in tomato cell cultures and leaves. Molecular and Cellular Biology 3, 1648–1655.
  151. Towner, J.S., Rollin, P.E., Bausch, D.G., Sanchez, A., Crary, S.M., Vincent, M., Lee, W.F., Spiropoulou, C.F., Ksiazek, T.G., Lukwiya, M., Kaducu, F., Downing, R., Nichol, S.T., 2004. Rapid diagnosis of Ebola hemorrhagic fever by reverse transcription-PCR in an outbreak setting and assessment of patient viral load as a predictor of outcome. J Virol 78, 4330–4341.
  152. Mühlberger, E., 2007. Filovirus replication and transcription. Future Virol 2, 205–215.
  153. Tarun, S.Z.J., Sachs, A.B., 1996. Association of the yeast poly(A) tail binding protein with translation initiation factor eIF-4G. EMBO J. 15, 7168–7177.
  154. Gilks, N., Kedersha, N., Ayodele, M., Shen, L., Stoecklin, G., Dember, L.M., Anderson, P., 2004. Stress granule assembly is mediated by prion-like aggregation of TIA-1. Mol. Biol. Cell 15, 5383–5398.
  155. Peters, G.A., Hartmann, R., Qin, J., Sen, G.C., 2001. Modular structure of PACT: distinct domains for binding and activating PKR. Molecular and Cellular Biology 21, 1908–1920.
  156. Tourrière, H., Gallouzi, I.E., Chebli, K., Capony, J.P., Mouaikel, J., van der Geer, P., Tazi, J., 2001. RasGAP-associated endoribonuclease G3Bp: selective RNA degradation and phosphorylation-dependent localization. Molecular and Cellular Biology 21, 7747–7760.
  157. Himeda, T., Hosomi, T., Asif, N., Shimizu, H., Okuwa, T., Muraki, Y., Ohara, Y., 2011. The preparation of an infectious full-length cDNA clone of Saffold virus. Virology Journal 8, 110.


* Das Dokument ist im Internet frei zugänglich - Hinweise zu den Nutzungsrechten