Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Herpesviruses assimilate kinesin to produce motorized viral particles

Abstract

Neurotropic alphaherpesviruses initiate infection in exposed mucosal tissues and, unlike most viruses, spread rapidly to sensory and autonomic nerves where life-long latency is established1. Recurrent infections arise sporadically from the peripheral nervous system throughout the life of the host, and invasion of the central nervous system may occur, with severe outcomes2. These viruses directly recruit cellular motors for transport along microtubules in nerve axons, but how the motors are manipulated to deliver the virus to neuronal nuclei is not understood. Here, using herpes simplex virus type I and pseudorabies virus as model alphaherpesviruses, we show that a cellular kinesin motor is captured by virions in epithelial cells, carried between cells, and subsequently used in neurons to traffic to nuclei. Viruses assembled in the absence of kinesin are not neuroinvasive. The findings explain a critical component of the alphaherpesvirus neuroinvasive mechanism and demonstrate that these viruses assimilate a cellular protein as an essential proviral structural component. This principle of viral assimilation may prove relevant to other virus families and offers new strategies to combat infection.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Kinesin-1 supports initial infection of RPE cells by HSV-1.
Fig. 2: The presence of kinesin-1 during virus assembly promotes trafficking of capsids to nuclei during subsequent infection.
Fig. 3: Kinesin-1 is captured in viral particles and delivered to other cells.
Fig. 4: Virally captured kinesin transports capsids to nuclei.

Similar content being viewed by others

Data availability

Unfiltered mass spectrometry data, raw microscopy images and plasmid and virus construction details (including primer sequences) are available from the corresponding author upon request.

References

  1. Smith, G. Herpesvirus transport to the nervous system and back again. Annu. Rev. Microbiol. 66, 153–176 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Lafaille, F. G. et al. Deciphering human cell-autonomous anti-HSV-1 immunity in the central nervous system. Front. Immunol. 6, 208 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Zaichick, S. V. et al. The herpesvirus VP1/2 protein is an effector of dynein-mediated capsid transport and neuroinvasion. Cell Host Microbe 13, 193–203 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Smith, G. A., Pomeranz, L., Gross, S. P. & Enquist, L. W. Local modulation of plus-end transport targets herpesvirus entry and egress in sensory axons. Proc. Natl Acad. Sci. USA 101, 16034–16039 (2004).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Antinone, S. E. & Smith, G. A. Retrograde axon transport of herpes simplex virus and pseudorabies virus: a live-cell comparative analysis. J. Virol. 84, 1504–1512 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Pernigo, S., Lamprecht, A., Steiner, R. A. & Dodding, M. P. Structural basis for kinesin-1:cargo recognition. Science 340, 356–359 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Robert, A. et al. Kinesin-dependent transport of keratin filaments: a unified mechanism for intermediate filament transport. FASEB J. 33, 388–399 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Bish, S. E., Song, W. & Stein, D. C. Quantification of bacterial internalization by host cells using a β-lactamase reporter strain: Neisseria gonorrhoeae invasion into cervical epithelial cells requires bacterial viability. Microbes Infect. 10, 1182–1191 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Heine, J. W., Honess, R. W., Cassai, E. & Roizman, B. Proteins specified by herpes simplex virus. XII. The virion polypeptides of type 1 strains. J Virol 14, 640–651 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cavrois, M., De Noronha, C. & Greene, W. C. A sensitive and specific enzyme-based assay detecting HIV-1 virion fusion in primary T lymphocytes. Nat. Biotechnol. 20, 1151–1154 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Lyman, M. G., Feierbach, B., Curanovic, D., Bisher, M. & Enquist, L. W. PRV Us9 directs axonal sorting of viral capsids. J. Virol. 81, 11363–11371 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Scherer, J. et al. A Kinesin-3 recruitment complex facilitates axonal sorting of enveloped alpha herpesvirus capsids. PLoS Pathog. 16, e1007985 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Diwaker, D., Murray, J. W., Barnes, J., Wolkoff, A. W. & Wilson, D. W. Deletion of the pseudorabies virus gE/gI–US9p complex disrupts kinesin KIF1A and KIF5C recruitment during egress, and alters the properties of microtubule-dependent transport in vitro. PLoS Pathog. 16, e1008597 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. DuRaine, G., Wisner, T. W., Howard, P., Williams, M. & Johnson, D. C. Herpes simplex virus gE/gI and US9 promote both envelopment and sorting of virus particles in the cytoplasm of neurons, two processes that precede anterograde transport in axons. J. Virol. 91, e00050-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Engelke, M. F. et al. Engineered kinesin motor proteins amenable to small-molecule inhibition. Nat. Commun. 7, 11159 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Schipke, J. et al. The C terminus of the large tegument protein pUL36 contains multiple capsid binding sites that function differently during assembly and cell entry of herpes simplex virus. J. Virol. 86, 3682–3700 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dohner, K. et al. Function of dynein and dynactin in herpes simplex virus capsid transport. Mol. Biol. Cell. 13, 2795–2809 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. DuRaine, G., Wisner, T. W., Howard, P. & Johnson, D. C. Kinesin-1 proteins KIF5A, 5B and 5C promote anterograde transport of herpes simplex virus enveloped virions in axons. J. Virol. 92, e01269-18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Radtke, K. et al. Plus- and minus-end directed microtubule motors bind simultaneously to herpes simplex virus capsids using different inner tegument structures. PLoS Pathog. 6, e1000991 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Diefenbach, R. J. et al. The basic domain of herpes simplex virus 1 pUS9 recruits kinesin-1 to facilitate egress from neurons. J. Virol. 90, 2102–2111 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Loret, S., Guay, G. & Lippe, R. Comprehensive characterization of extracellular herpes simplex virus type 1 virions. J. Virol. 82, 8605–8618 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kramer, T., Greco, T. M., Enquist, L. W. & Cristea, I. M. Proteomic characterization of pseudorabies virus extracellular virions. J. Virol. 85, 6427–6441 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Miranda-Saksena, M. et al. Herpes simplex virus utilizes the large secretory vesicle pathway for anterograde transport of tegument and envelope proteins and for viral exocytosis from growth cones of human fetal axons. J. Virol. 83, 3187–3199 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Smith, C. L. Culturing Nerve Cells 2nd Edn (eds Banker, G. & Goslin, K.) 261–287 (MIT Press, 1998).

  25. Smith, G. A., Gross, S. P. & Enquist, L. W. Herpesviruses use bidirectional fast-axonal transport to spread in sensory neurons. Proc. Natl Acad. Sci. USA 98, 3466–3470 (2001).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tanaka, M., Kagawa, H., Yamanashi, Y., Sata, T. & Kawaguchi, Y. Construction of an excisable bacterial artificial chromosome containing a full-length infectious clone of herpes simplex virus type 1: viruses reconstituted from the clone exhibit wild-type properties in vitro and in vivo. J. Virol. 77, 1382–1391 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Smith, G. A. & Enquist, L. W. Construction and transposon mutagenesis in Escherichia coli of a full-length infectious clone of pseudorabies virus, an alphaherpesvirus. J. Virol. 73, 6405–6414 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kaufman, H. E., Ellison, E. D. & Waltman, S. R. Double-stranded RNA, an interferon inducer, in herpes simplex keratitis. Am. J. Ophthalmol. 68, 486–491 (1969).

    Article  CAS  PubMed  Google Scholar 

  29. Bohannon, K. P., Sollars, P. J., Pickard, G. E. & Smith, G. A. Fusion of a fluorescent protein to the pUL25 minor capsid protein of pseudorabies virus allows live-cell capsid imaging with negligible impact on infection. J. Gen. Virol. 93, 124–129 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huffmaster, N. J., Sollars, P. J., Richards, A. L., Pickard, G. E. & Smith, G. A. Dynamic ubiquitination drives herpesvirus neuroinvasion. Proc. Natl Acad. Sci. USA 112, 12818–12823 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Richards, A. L. et al. The pUL37 tegument protein guides alpha-herpesvirus retrograde axonal transport to promote neuroinvasion. PLoS Pathog. 13, e1006741 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Stults, A. M. & Smith, G. A. The herpes simplex virus 1 deamidase enhances propagation but is dispensable for retrograde axonal transport into the nervous system. J Virol 93, e01172-19 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tischer, B. K., Smith, G. A. & Osterrieder, N. En passant mutagenesis: a two step markerless red recombination system. Methods Mol. Biol. 634, 421–430 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Szpara, M. L. et al. A wide extent of inter-strain diversity in virulent and vaccine strains of alphaherpesviruses. PLoS Pathog. 7, e1002282 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Abramoff, M. D., Magelhaes, P. J. & Ram, S. J. Image processing with ImageJ. Biophotonics Int. 11, 36–42 (2004).

    Google Scholar 

  36. Luxton, G. W. et al. Targeting of herpesvirus capsid transport in axons is coupled to association with specific sets of tegument proteins. Proc. Natl Acad. Sci. USA 102, 5832–5837 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Leelawong, M., Lee, J. I. & Smith, G. A. Nuclear egress of pseudorabies virus capsids is enhanced by a subspecies of the large tegument protein that is lost upon cytoplasmic maturation. J. Virol. 86, 6303–6314 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lee, J. I., Luxton, G. W. & Smith, G. A. Identification of an essential domain in the herpesvirus VP1/2 tegument protein: the carboxy terminus directs incorporation into capsid assemblons. J. Virol. 80, 12086–12094 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dodding, M. P., Mitter, R., Humphries, A. C. & Way, M. A kinesin-1 binding motif in vaccinia virus that is widespread throughout the human genome. EMBO J. 30, 4523–4538 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tombacz, D., Toth, J. S., Petrovszki, P. & Boldogkoi, Z. Whole-genome analysis of pseudorabies virus gene expression by real-time quantitative RT-PCR assay. BMC Genomics 10, 491 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Lee, G. E., Murray, J. W., Wolkoff, A. W. & Wilson, D. W. Reconstitution of herpes simplex virus microtubule-dependent trafficking in vitro. J. Virol. 80, 4264–4275 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kharkwal, H., Smith, C. G. & Wilson, D. W. Blocking ESCRT-mediated envelopment inhibits microtubule-dependent trafficking of alphaherpesviruses in vitro. J. Virol. 88, 14467–14478 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Howard, J. & Hyman, A. A. Preparation of marked microtubules for the assay of the polarity of microtubule-based motors by fluorescence microscopy. Methods Cell. Biol. 39, 105–113 (1993).

    Article  CAS  PubMed  Google Scholar 

  44. Shanda, S. K. & Wilson, D. W. UL36p is required for efficient transport of membrane-associated herpes simplex virus type 1 along microtubules. J. Virol. 82, 7388–7394 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Homa, F. L. et al. Structure of the pseudorabies virus capsid: comparison with herpes simplex virus type 1 and differential binding of essential minor proteins. J. Mol. Biol. 425, 3415–3428 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Huet, A. et al. Extensive subunit contacts underpin herpesvirus capsid stability and interior-to-exterior allostery. Nat. Struct. Mol. Biol. 23, 531–539 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass. Spectrom. 5, 976–989 (1994).

    Article  CAS  PubMed  Google Scholar 

  48. Xu, T. et al. ProLuCID: An improved SEQUEST-like algorithm with enhanced sensitivity and specificity. J. Proteomics 129, 16–24 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cociorva, D., D, L. T. & Yates, J. R. Validation of tandem mass spectrometry database search results using DTASelect. Curr. Protoc. Bioinformatics Ch. 13, Unit 13 14 (2007).

    Google Scholar 

  50. Tabb, D. L., McDonald, W. H. & Yates, J. R., 3rd. DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics. J. Proteome Res. 1, 21–26 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. UniProt, C. UniProt: a hub for protein information. Nucleic Acids Res. 43, D204–D212 (2015).

    Article  Google Scholar 

  52. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Gelfand and K. Verhey for suggestions that helped inform the experimental design and for reagents, and A. Roberts, M. Englke, M. Way, S. Brady and G. Gunderson for reagents. D. Kirchenbuechler of the Northwestern University Cell Imaging Facility, supported by NCI CCSG P30 CA060553, provided invaluable support in the development of the automated image analysis pipeline used in this report. Some confocal imaging was performed on an Andor XDI Revolution microscope purchased through the support of NCRR 1S10 RR031680-01 at the Northwestern University Cell Imaging Facility. Sequencing services were performed at the Northwestern University Genomics Core Facility. In vitro microtubule-dependent motility assays and analyses were performed in the Analytical Imaging Facility of the Albert Einstein College of Medicine, with support from National Cancer Institute cancer center grant P30CA013330. This work was funded by NIH AI056346 (G.A.S., P.J.S. and G.E.P.), AI125244 (D.W.W.), AI141470 (D.W.) and AI148780 (J.N.S.). E.B.W. was supported by NIH NS106812. C.E.P. received support from National Science Foundation DGE-1324585 and NIH Cellular and Molecular Basis of Disease Training Grant T32GM08061.

Author information

Authors and Affiliations

Authors

Contributions

C.E.P., S.V.Z. and G.A.S. conceived the study; C.E.P. performed experiments concerning kinesin-1 assimilation and its discovery, and experiments concerning kinesin-1 involvement in centrosome-to-nucleus trafficking; S.V.Z. performed experiments concerning pUL36 involvement in centrosome-to-nucleus trafficking and kinesin-1 interaction; J.N.S. and E.B.W. performed mass spectrometry and data analysis; D.W. and V.J. performed kinesin siRNA experiments; D.K. and C.E.P. performed nitrocefin experiments; D.W.W. and H.K. performed in vitro motility experiments; P.J.S. and G.E.P. performed mouse infection studies; C.E.P., S.V.Z. and G.A.S. made the figures; C.E.P. and G.A.S. wrote and edited the manuscript; and all authors discussed results and commented on the manuscript.

Corresponding author

Correspondence to Gregory A. Smith.

Ethics declarations

Competing interests

P.J.S., G.E.P. and G.A.S. have disclosed a significant financial interest in Thyreos, Inc. In accordance with their Conflict of Interest policies, the University of Nebraska-Lincoln and Northwestern University Feinberg School of Medicine Conflict of Interest Review Committees have determined that this must be disclosed.

Additional information

Peer review information Nature thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Transiently-expressed PRV pUL36 co-localizes with microtubules and accumulates at the cell periphery.

(A) Schematic representation of seven regions of pUL36. Regions 2 and 6 are proline rich (pink). Position of the capsid binding domain (CBD) consisting of amino acids 3033-3095 is indicated. Amino acid positions are indicated above the schematic. (B) Transiently expressed GFP-pUL36∆R7, but not GFP-pUL36 (full length), formed small punctae that moved in curvilinear trajectories (also see Supplementary Video 1) and accumulated at the periphery of Vero cells (arrowheads). The 6.9 µm x 2.4 µm boxed region is expanded as a time-lapse montage to the right of the image (n=3 independent experiments). (C) GFP-pUL36∆R7 decorates microtubules. Vero cells were transiently co-transfected with GFP-pUL36∆R7 and mCherry-tubulin. The 7.4 µm x 7.4 µm boxed region is expanded below each image. All images were captured between 24-28 h post transfection (hpt) (n=3 independent experiments). Scale bars are 20 µm.

Extended Data Fig. 2 Transiently-expressed PRV pUL36 interacts with components of the Kif5 motor complex.

(A) pUL36 region 7 is dispensable for co-immunoprecipitation (co-IP) with Kif5 heavy chain (KHC) and Kif5 light chain (KLC). HEK293 cells were lysed 16-18 hpt and GFP-pUL36 was immunoprecipitated (IP) with anti-GFP antibody and detected by western blot as indicated. Inputs are 6% of crude lysates (n=3 independent experiments) . (B) Peripheral accumulations of transiently expressed GFP-pUL36ΔR7 co-localize with endogenous cellular Kif5 components (arrows). Vero cells were fixed and immunostained for KHC or KLC at 20 hpt (n=3 independent experiments). Scale bars are 20 µm. (C) GFP-pUL36ΔR7 co-immunoprecipitates with KLC1 and KLC2 isoforms. HEK293 cells transiently expressing GFP or GFP-pUL36ΔR7 were co-transfected with HA-KLC1 and HA-KLC2. Endogenous KHC served as a positive co-immunoprecipitation control (n=3 independent experiments). (D) Transient expression mCherry-KLC1 and mCherry-KLC2 in Vero cells. Both fusion proteins show varying degrees of juxtanuclear localization and an absence of peripheral accumulation (n=3 independent experiments). (E) mCherry-KLC1 and mCherry-KLC2 redistribute with GFP-pUL36ΔR7 to the cell periphery during transient expression. Vero cells were imaged 24-28 hpt (n=3 independent experiments). Scale bars are 20 µm.

Extended Data Fig. 3 Mapping the PRV pUL36 kinesin-binding region.

(A) Illustration of pUL36 constructs with amino- and carboxyl-terminal GFP fusions indicated (grey circles). Location of a cryptic nuclear localization signal (NLS) at the end of region 2 (vertical grey line in R2) that becomes active when region 6 is deleted is also indicated. Constructs are named for each intact region number with a dash representing deleted regions. In the case of region 6, a subregion consisting of the majority of the sequence was deleted (denoted “s” and consisting of amino acids 2087-2796). (B) pUL36 region 5 is required for interaction with the Kif5 complex (KHC, Kif5 heavy chain; KLC, Kif5 light chain; n = 3 independent experiments). (C) pUL36 region 6 is required for interaction with the Kif5 complex. Deletion of region 6 also resulted in nuclear localization of the protein due to the NLS in region 2. Mutation of the NLS (K285RRR > AAAA) restored protein localization to the cytoplasm (not shown) but did not restore Kif5 interaction (n = 3 independent experiments). (D–E) pUL36 regions 5-6, is sufficient for Kif5 binding. Interaction with a dynactin component, p150/glued (p150), was used as a positive co-immunoprecipitation control (n = 3 independent experiments each).

Extended Data Fig. 4 PRV pUL36 WD motifs are required for kinesin-1 binding.

(A) Schematic representation of two potential bipartite kinesin-binding motifs (WD1/WD2 and WD3/WD4) in region 5 of pUL36. Corresponding regions from ten different alphaherpesvirinae are aligned. Prospective WD motifs in PRV are highlighted in red. Conserved tryptophans and/or surrounding E, D, N and Q at positions ±1 and +2 in pUL36 orthologs are in blue. The number of amino acids between the WD3 and WD4 tryptophans are indicated only when no other tryptophan was present in between (W-W); otherwise, n.a. is indicated. Table on the right summarizes mutants used for transient expression (RKB, reduced kinesin binding mutant that was subsequently introduced into PRV – see Extended Data Fig. 5 & 6). (B–D) Co-immunoprecipitations of pUL36 proteins and endogenous cellular Kif5 demonstrate that WD3 and WD4 contribute to the pUL36-Kif5 interaction. Interaction with dynactin component, p150/glued (p150) was used as a positive co-immunoprecipitation control (n = 3 independent experiments each). (E) Quantitation of cells displaying pUL36 localization at the cell periphery or juxtanuclear between 24-28 hpt. Representative images of cells and scoring are provided at top. Percentile distributions of pUL36 accumulation are illustrated by Venn diagrams (average of n = 3 independent experiments >250 cells each). Scale bars are 20 µm.

Extended Data Fig. 5 The PRV[RKB] mutant is defective for nuclear trafficking and gene expression.

(A) Mutation of WD3 attenuated PRV spread, as measured by plaque size, whereas mutations of WD4 rendered PRV non-viable (n = 200 plaques over 3 independent experiments). (B) Mutation of the WD4 tryptophan alone also rendered PRV non-viable, whereas mutation of the surrounding acidic residues did not impair viral spread. PRV encoding both the ∆WD3 mutation and WD4(DE>AA) attenuated viral spread but could be propagated and further studied and was designated as the reduced-kinesin binding mutant (RKB; also see Figure S4). Plaque diameters were measured 43 hpt. Data are presented as scatter plots, with each dot representing a single plaque (n = 200 plaques over 3 independent experiments). Mean values ± SD are indicated. (P-values were determined by one-way analysis of variance with a post-hoc Tukey test). (C) Incoming capsids of PRV encoding the pUL36 RKB mutant accumulate juxtanuclear in explanted chick dorsal root ganglia (DRG). Representative images of neural soma (transmitted and fluorescent capsid image pairs with nuclei highlighted by dotted lines). Percentile distributions of juxtanuclear capsid accumulation are illustrated at right (n= 3 independent experiments > 500 infected cells each). DRG explants were imaged between 2.5-3 hpi. Scale bars are 20 µm. Mean values ± SD are indicated. (P values based on two-tailed unpaired t test). (D) PRV[RKB] has delayed viral gene expression. PK15 cells were infected at MOI 10 and total RNA was collected at 4 hpi. Relative IE180 mRNA levels were measured by RT-PCR first-strand DNA synthesis and qPCR amplification using primers specific for PRV IE180 and the swine ribosomal S28 rRNA (loading control). Data was normalized to S28 rRNA levels and depicted as a fold change with respect to IE180 mRNA levels during WT infection (n = independent experiments). Mean values ± SD are indicated. (P values based on two-tailed unpaired t test).

Extended Data Fig. 6 PRV[RKB] is impaired for kinesin-based transport.

Viral particles were harvested from the cytoplasm of infected RK13 cells and allowed to bind to fluorescent microtubules in vitro. (A) Numbers of bound fluorescent viral particles were counted and are represented as a percentage of those initially added to the imaging chamber. Four independent chambers containing a total of 1204 and 805 bound particles were counted for wild-type PRV (WT) and PRV encoding the reduced kinesin binding mutant pUL36 allele [RKB]. (B) Motility was measured after addition of ATP supplemented with: no inhibitor (light grey bars), 5 μM sodium orthovanadate (a dynein inhibitor; medium grey bars) or 1 mM AMP-PNP (a kinesin inhibitor; dark grey bars). Numbers of motile virions were determined from triplicate motility chambers, and motility plotted as a percentage of the number of motile particles seen in the absence of inhibitor. For WT and RKB respectively, the numbers of individual viral particles examined for each set of conditions were as follows: no inhibitor (658, 706), sodium orthovanadate (776, 714), AMP-PNP (776, 714). Mean values ± SD are indicated (n = 3 independent experiments). (P values based on two-tailed unpaired t test).

Extended Data Fig. 7 Kinesin-1 knockdown impairs HSV-1 infection in primary human fibroblasts.

(A) Normal human dermal fibroblasts (NHDF) cells were depleted of Kif5 isoforms by siRNA knockdown. Kif5 expression profile in SK-N-SH cells as a standard that expresses all three Kif5 isoforms (n = 1 independent experiment). (B) Control and Kif5B-depleted NHDF cells were infected with HSV-1 in the presence of 100 µM cycloheximide (n = 1 independent experiment). At 8 hpi, cells were fixed and immunofluorescence was performed. Scale bar is 20 µm.

Extended Data Fig. 8 Kif5B and Kif5C function interchangeably to rescue pUL36 peripheral accumulation in RPE∆Kif5B (KO) cells.

Quantitation of transiently-transfected cells displaying GFP-pUL36∆R7 localization at the cell periphery (n = 3 independent experiments > 150 each). Scoring methodology is provided in Extended Data Fig. 4. Mean values ± SD are indicated. (P values based on two-tailed unpaired t test).

Extended Data Fig. 9 Model of viral assimilation of kinesin-1.

Neuroinvasive herpesviruses capture Kif5 (conventional kinesin; kinesin-1) into the tegument of newly formed virions during infection of epithelial cells. The captured epithelial kinesin-1 is carried between cells as a structural virion component and is deposited into cells (epithelia and neurons) upon the subsequent round of infection (first blue panel). Upon entry, cytosolic capsids engage in retrograde axonal transport effected by the cytoplasmic dynein/dynactin microtubule motor. The assimilated kinesin is presumably carried in an inactive state during this step of infection (second blue panel). Dynein/dynactin-based transport directs the capsid ‘minus-ended’ along microtubules ending at the centrosome, where the virus uses assimilated kinesin to transport to nuclei (third blue panel). When viruses are attenuated for assimilated-kinesin binding (e.g., PRV[RKB]) or are produced in the absence of Kif5, capsids predominately accumulate at the centrosome and do not progress toward the nucleus despite the presence of endogenous neuronal kinesin-1 (pink panel). Nevertheless, endogenous kinesin also supports nuclear trafficking of capsids.

Extended Data Table 1 Description of recombinant viruses and mammalian expression plasmids used in this study

Supplementary information

Supplementary Information

This file contains Supplementary Data 1 and 2, legends for Supplementary Data 3 and 4, legends for Supplementary Videos 1 and 2 and Supplementary Fig. 1.

Reporting Summary

Supplementary Video 1

See Supplementary Information for description.

Supplementary Video 2

See Supplementary Information for description.

Supplementary Data 3

Data derived from automated analysis of capsid localization that support Fig. 1c. The spreadsheet includes all formula for post-analysis of automated data pipeline.

Supplementary Data 4

Data derived from automated analysis of capsid localization that support Fig. 2d. This spreadsheet includes all formulae for post-analysis of the automated data pipeline.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pegg, C.E., Zaichick, S.V., Bomba-Warczak, E. et al. Herpesviruses assimilate kinesin to produce motorized viral particles. Nature 599, 662–666 (2021). https://doi.org/10.1038/s41586-021-04106-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04106-w

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing