Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-06T21:47:52.331Z Has data issue: false hasContentIssue false

A Dual Laser Scanning Confocal and Transmission Electron Microscopy Analysis of the Intracellular Localization, Aggregation and Particle Formation of African Horse Sickness Virus Major Core Protein VP7

Published online by Cambridge University Press:  23 January 2017

Gayle V. Wall
Affiliation:
Department of Genetics, University of Pretoria, Pretoria, 0002, South Africa
Daria A. Rutkowska
Affiliation:
Department of Genetics, University of Pretoria, Pretoria, 0002, South Africa
Eshchar Mizrachi
Affiliation:
Department of Genetics, University of Pretoria, Pretoria, 0002, South Africa
Henk Huismans
Affiliation:
Department of Genetics, University of Pretoria, Pretoria, 0002, South Africa
Vida van Staden*
Affiliation:
Department of Genetics, University of Pretoria, Pretoria, 0002, South Africa
*
*Corresponding author. vida.vanstaden@up.ac.za
Get access

Abstract

The bulk of the major core protein VP7 in African horse sickness virus (AHSV) self-assembles into flat, hexagonal crystalline particles in a process appearing unrelated to viral replication. Why this unique characteristic of AHSV VP7 is genetically conserved, and whether VP7 aggregation and particle formation have an effect on cellular biology or the viral life cycle, is unknown. Here we investigated how different small peptide and enhanced green fluorescent protein (eGFP) insertions into the VP7 top domain affected VP7 localization, aggregation, and particle formation. This was done using a dual laser scanning confocal and transmission electron microscopy approach in conjunction with analyses of the solubility, aggregation, and fluorescence profiles of the proteins. VP7 top domain modifications did not prevent trimerization, or intracellular trafficking, to one or two discrete sites in the cell. However, modifications that resulted in a misfolded and insoluble VP7-eGFP component blocked trafficking, and precluded protein accumulation at a single cellular site, perhaps by interfering with normal trimer–trimer interactions. Furthermore, the modifications disrupted the stable layering of the trimers into characteristic AHSV VP7 crystalline particles. It was concluded that VP7 trafficking is driven by a balance between VP7 solubility, trimer forming ability, and trimer–trimer interactions.

Type
Biological Applications
Copyright
© Microscopy Society of America 2017 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

Current address: Council for Scientific and Industrial Research (CSIR), Biosciences, Pretoria, 0001, South Africa.

References

Amin, S., Barnett, G.V., Pathak, J.A., Roberts, C.J. & Sarangapani, P.S. (2014). Protein aggregation, particle formation, characterization & rheology. Curr Opin Colloid Interface Sci 19, 438449.CrossRefGoogle Scholar
Basak, A.K., Gouet, P., Grimes, J.M., Roy, P. & Stuart, D. (1996). Crystal structure of the top domain of African horse sickness virus VP7: Comparisons with bluetongue virus VP7. J Virol 70, 37973806.CrossRefGoogle ScholarPubMed
Bekker, S., Huismans, H. & van Staden, V. (2014). Factors that affect the intracellular localization and trafficking of African horse sickness virus core protein, VP7. Virology 456–457, 279291.CrossRefGoogle ScholarPubMed
Bolte, S. & Cordelieres, F.P. (2006). A guided tour into subcellular colocalisation analysis in light microscopy. J Microsc 224, 213232.CrossRefGoogle Scholar
Bremer, C.W. (1976). A gel electrophoretic study of the protein and nucleic acid components of African horsesickness virus. Onderstepoort J Vet Res 43, 193199.Google ScholarPubMed
Bucciantini, M., Giannoni, E., Chiti, F., Baroni, F., Formigli, L., Zurdo, J., Taddei, N., Ramponi, G., Dobson, C.M. & Stefani, M. (2002). Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases. Nature 416, 507511.CrossRefGoogle ScholarPubMed
Burroughs, J.N., O’Hara, R.S., Smale, C.J., Hamblin, C., Walton, A., Armstrong, R. & Mertens, P.P. (1994). Purification and properties of virus particles, infectious subviral particles, cores and VP7 crystals of African horsesickness virus serotype 9. J Gen Virol 75(Pt 8), 18491857.CrossRefGoogle ScholarPubMed
Chuma, T., Le Blois, H., Sanchez-Vizcaino, J.M., Diaz-Laviada, M. & Roy, P. (1992). Expression of the major core antigen VP7 of African horsesickness virus by a recombinant baculovirus and its use as a group-specific diagnostic reagent. J Gen Virol 73(Pt 4), 925931.CrossRefGoogle ScholarPubMed
Dobson, C.M. (2004). Principles of protein folding, misfolding and aggregation. Semin Cell Dev Biol 15, 316.CrossRefGoogle ScholarPubMed
Grimes, J.M., Basak, A.K., Roy, P. & Stuart, D. (1995). The crystal structure of bluetongue virus VP7. Nature 373, 167170.CrossRefGoogle ScholarPubMed
Grimes, J.M., Burroughs, J.N., Gouet, P., Diprose, J.M., Malby, R., Zientara, S., Mertens, P.P. & Stuart, D.I. (1998). The atomic structure of the bluetongue virus core. Nature 395, 470478.CrossRefGoogle ScholarPubMed
Hewat, E.A., Booth, T.F. & Roy, P. (1992). Structure of bluetongue virus particles by cryoelectron microscopy. J Struct Biol 109, 6169.CrossRefGoogle ScholarPubMed
Heymann, M.C., Rabe, S., Ruß, S., Kapplusch, F., Schulze, F., Stein, R., Winkler, S., Hedrich, C.M., Rösen-Wolff, A. & Hofmann, S.R. (2015). Fluorescent tags influence the enzymatic activity and subcellular localization of procaspase-1. Clin Immunol 160, 172179.CrossRefGoogle ScholarPubMed
Kar, A.K., Bhattacharya, B. & Roy, P. (2007). Bluetongue virus RNA binding protein NS2 is a modulator of viral replication and assembly. BMC Mol Biol 8, 4.CrossRefGoogle ScholarPubMed
Lorenzo, A. & Yankner, B.A. (1994). Beta-amyloid neurotoxicity requires fibril formation and is inhibited by congo red. Proc Natl Acad Sci U S A 91, 1224312247.CrossRefGoogle ScholarPubMed
Maree, S., Durbach, S. & Huismans, H. (1998). Intracellular production of African horsesickness virus core-like particles by expression of the two major core proteins, VP3 and VP7, in insect cells. J Gen Virol 79(Pt 2), 333337.CrossRefGoogle ScholarPubMed
Maree, S., Maree, F.F., Putterill, J.F., de Beer, T.A.P., Huismans, H. & Theron, J. (2016). Synthesis of empty African horse sickness virus particles. Virus Res 213, 184194.CrossRefGoogle ScholarPubMed
Mellor, P.S. & Hamblin, C. (2004). African horse sickness. Vet Res 35, 445466.CrossRefGoogle ScholarPubMed
Monastyrskaya, K., Staeuber, N., Sutton, G. & Roy, P. (1997). Effects of domain-switching and site-directed mutagenesis on the properties and functions of the VP7 proteins of two orbiviruses. Virology 237, 217227.CrossRefGoogle ScholarPubMed
Oellermann, R.A., Els, H.J. & Erasmus, B.J. (1970). Characterization of African horsesickness virus. Arch Gesamte Virusforsch 29, 163174.CrossRefGoogle ScholarPubMed
Oldfield, S., Adachi, A., Urakawa, T., Hirasawa, T. & Roy, P. (1990). Purification and characterization of the major group-specific core antigen VP7 of bluetongue virus synthesized by a recombinant baculovirus. J Gen Virol 71(Pt 11), 26492656.CrossRefGoogle ScholarPubMed
Reynolds, E.S. (1963). The use of lead citrate at high pH as an electron-opaque stain in electron microscopy. J Cell Biol 17, 208212.CrossRefGoogle ScholarPubMed
Roberts, C.J. (2014). Protein aggregation and its impact on product quality. Curr Opin Biotechnol 30, 211217.CrossRefGoogle ScholarPubMed
Roy, P. (1996). Orbivirus structure and assembly. Virology 216, 111.CrossRefGoogle ScholarPubMed
Roy, P., Hirasawa, T., Fernandez, M., Blinov, V.M. & Sanchez-Vixcain Rodrique, J.M. (1991). The complete sequence of the group-specific antigen, VP7, of African horsesickness disease virus serotype 4 reveals a close relationship to bluetongue virus. J Gen Virol 72(Pt 6), 12371241.CrossRefGoogle ScholarPubMed
Rutkowska, D.A., Meyer, Q.C., Maree, F., Vosloo, W., Fick, W. & Huismans, H. (2011). The use of soluble African horse sickness viral protein 7 as an antigen delivery and presentation system. Virus Res 156, 3548.CrossRefGoogle ScholarPubMed
Stefani, M. (2004). Protein misfolding and aggregation: New examples in medicine and biology of the dark side of the protein world. Biochim Biophys Acta 1739, 525.CrossRefGoogle Scholar
Studer, D., Graber, W., Al-Amoudi, A. & Eggli, P. (2001). A new approach for cryofixation by high-pressure freezing. J Microsc 203, 285294.CrossRefGoogle ScholarPubMed
Tsumoto, K., Ejima, D., Kita, Y. & Arakawa, T. (2005). Review: Why is arginine effective in suppressing aggregation? Protein Pept Lett 12, 613619.CrossRefGoogle ScholarPubMed
Venter, E., van der Merwe, C.F., Buys, A.V., Huismans, H. & van Staden, V. (2014). Comparative ultrastructural characterisation of African horse sickness virus-infected mammalian and insect cells reveals novel potential virus release mechanism from insect cells. J Gen Virol 95, 642651.CrossRefGoogle ScholarPubMed
Verwoerd, D., Els, H., De Villiers, E. & Huismans, H. (1972). Structure of the bluetongue virus capsid. J Virol 10, 783794.CrossRefGoogle ScholarPubMed
Wu, C. & Shea, J.-E. (2011). Coarse-grained models for protein aggregation. Curr Opin Struct Biol 21, 209220.CrossRefGoogle ScholarPubMed
Supplementary material: Image

Wall supplementary material

Figure

Download Wall supplementary material(Image)
Image 5.1 MB