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:

Comparative proteomics identifies Schlafen 5 (SLFN5) as a herpes simplex virus restriction factor that suppresses viral transcription

Abstract

Intrinsic antiviral host factors confer cellular defence by limiting virus replication and are often counteracted by viral countermeasures. We reasoned that host factors that inhibit viral gene expression could be identified by determining proteins bound to viral DNA (vDNA) in the absence of key viral antagonists. Herpes simplex virus 1 (HSV-1) expresses E3 ubiquitin-protein ligase ICP0 (ICP0), which functions as an E3 ubiquitin ligase required to promote infection. Cellular substrates of ICP0 have been discovered as host barriers to infection but the mechanisms for inhibition of viral gene expression are not fully understood. To identify restriction factors antagonized by ICP0, we compared proteomes associated with vDNA during HSV-1 infection with wild-type virus and a mutant lacking functional ICP0 (ΔICP0). We identified the cellular protein Schlafen family member 5 (SLFN5) as an ICP0 target that binds vDNA during HSV-1 ΔICP0 infection. We demonstrated that ICP0 mediates ubiquitination of SLFN5, which leads to its proteasomal degradation. In the absence of ICP0, SLFN5 binds vDNA to repress HSV-1 transcription by limiting accessibility of RNA polymerase II to viral promoters. These results highlight how comparative proteomics of proteins associated with viral genomes can identify host restriction factors and reveal that viral countermeasures can overcome SLFN antiviral activity.

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: ICP0 targets SLFN5 for degradation.
Fig. 2: ICP0 interacts with SLFN5.
Fig. 3: SLFN5 colocalizes with HSV-1 DNA.
Fig. 4: ICP0 counteracts SLFN5-mediated suppression of HSV-1 replication.
Fig. 5: SLFN5 associates with vDNA to suppress HSV-1 gene transcription.

Similar content being viewed by others

Data availability

The MS proteomics data have been deposited with the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the PRIDE75 partner repository with the dataset no. PXD018773. Source data are provided with this paper.

Code availability

The scripts used to analyse the iPOND proteomics data are available from the corresponding author upon reasonable request or can be accessed via GitHub (https://github.com/JosephDybas/HSV_iPOND).

References

  1. Malim, M. H. & Bieniasz, P. D. HIV restriction factors and mechanisms of evasion. Cold Spring Harb. Perspect. Med. 2, a006940 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Gu, H. & Zheng, Y. Role of ND10 nuclear bodies in the chromatin repression of HSV-1. Virol. J. 13, 62 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Tavalai, N. & Stamminger, T. Intrinsic cellular defense mechanisms targeting human cytomegalovirus. Virus Res. 157, 128–133 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Glass, M. & Everett, R. D. Components of promyelocytic leukemia nuclear bodies (ND10) act cooperatively to repress herpesvirus infection. J. Virol. 87, 2174–2185 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Crow, M. S., Lum, K. K., Sheng, X., Song, B. & Cristea, I. M. Diverse mechanisms evolved by DNA viruses to inhibit early host defenses. Crit. Rev. Biochem. Mol. Biol. 51, 452–481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rajsbaum, R. & García-Sastre, A. Viral evasion mechanisms of early antiviral responses involving regulation of ubiquitin pathways. Trends Microbiol. 21, 421–429 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Dybas, J. M., Herrmann, C. & Weitzman, M. D. Ubiquitination at the interface of tumor viruses and DNA damage responses. Curr. Opin. Virol. 32, 40–47 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Colomer-Lluch, M., Ruiz, A., Moris, A. & Prado, J. G. Restriction factors: from intrinsic viral restriction to shaping cellular immunity against HIV-1. Front. Immunol. 9, 2876 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Simon, V., Bloch, N. & Landau, N. R. Intrinsic host restrictions to HIV-1 and mechanisms of viral escape. Nat. Immunol. 16, 546–553 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Komatsu, T., Nagata, K. & Wodrich, H. The role of nuclear antiviral factors against invading DNA viruses: the immediate fate of incoming viral genomes. Viruses 8, 290 (2016).

    Article  PubMed Central  Google Scholar 

  11. Bieniasz, P. D. Restriction factors: a defense against retroviral infection. Trends Microbiol. 11, 286–291 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Harris, R. S., Hultquist, J. F. & Evans, D. T. The restriction factors of human immunodeficiency virus. J. Biol. Chem. 287, 40875–40883 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Boutell, C. & Everett, R. D. Regulation of alphaherpesvirus infections by the ICP0 family of proteins. J. Gen. Virol. 94, 465–481 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Knipe, D. M. & Cliffe, A. Chromatin control of herpes simplex virus lytic and latent infection. Nat. Rev. Microbiol. 6, 211–221 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Cai, W. & Schaffer, P. A. Herpes simplex virus type 1 ICP0 regulates expression of immediate-early, early, and late genes in productively infected cells. J. Virol. 66, 2904–2915 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rodríguez, M. C., Dybas, J. M., Hughes, J., Weitzman, M. D. & Boutell, C. The HSV-1 ubiquitin ligase ICP0: modifying the cellular proteome to promote infection. Virus Res. 285, 198015 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Boutell, C. & Everett, R. D. The herpes simplex virus type 1 (HSV-1) regulatory protein ICP0 interacts with and ubiquitinates p53. J. Biol. Chem. 278, 36596–36602 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Everett, R. D., Orr, A. & Preston, C. M. A viral activator of gene expression functions via the ubiquitin–proteasome pathway. EMBO J. 17, 7161–7169 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lium, E. K. & Silverstein, S. Mutational analysis of the herpes simplex virus type 1 ICP0 C3HC4 zinc ring finger reveals a requirement for ICP0 in the expression of the essential α27 gene. J. Virol. 71, 8602–8614 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Parkinson, J., Lees-Miller, S. P. & Everett, R. D. Herpes simplex virus type 1 immediate-early protein Vmw110 induces the proteasome-dependent degradation of the catalytic subunit of DNA-dependent protein kinase. J. Virol. 73, 650–657 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lees-Miller, S. P. et al. Attenuation of DNA-dependent protein kinase activity and its catalytic subunit by the herpes simplex virus type 1 transactivator ICP0. J. Virol. 70, 7471–7477 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chaurushiya, M. S. et al. Viral E3 ubiquitin ligase-mediated degradation of a cellular E3: viral mimicry of a cellular phosphorylation mark targets the RNF8 FHA domain. Mol. Cell 46, 79–90 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lilley, C. E. et al. A viral E3 ligase targets RNF8 and RNF168 to control histone ubiquitination and DNA damage responses. EMBO J. 29, 943–955 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chelbi-Alix, M. K. & de Thé, H. Herpes virus induced proteasome-dependent degradation of the nuclear bodies-associated PML and Sp100 proteins. Oncogene 18, 935–941 (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Everett, R. D. et al. The disruption of ND10 during herpes simplex virus infection correlates with the Vmw110- and proteasome-dependent loss of several PML isoforms. J. Virol. 72, 6581–6591 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Perusina Lanfranca, M., Mostafa, H. H. & Davido, D. J. Two overlapping regions within the N-terminal half of the herpes simplex virus 1 E3 ubiquitin ligase ICP0 facilitate the degradation and dissociation of PML and dissociation of Sp100 from ND10. J. Virol. 87, 13287–13296 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Lukashchuk, V. & Everett, R. D. Regulation of ICP0-null mutant herpes simplex virus type 1 infection by ND10 components ATRX and hDaxx. J. Virol. 84, 4026–4040 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Orzalli, M. H., DeLuca, N. A. & Knipe, D. M. Nuclear IFI16 induction of IRF-3 signaling during herpesviral infection and degradation of IFI16 by the viral ICP0 protein. Proc. Natl Acad. Sci. USA 109, E3008–E3017 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Everett, R. D., Earnshaw, W. C., Findlay, J. & Lomonte, P. Specific destruction of kinetochore protein CENP-C and disruption of cell division by herpes simplex virus immediate-early protein Vmw110. EMBO J. 18, 1526–1538 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lomonte, P., Sullivan, K. F. & Everett, R. D. Degradation of nucleosome-associated centromeric histone H3-like protein CENP-A induced by herpes simplex virus type 1 protein ICP0. J. Biol. Chem. 276, 5829–5835 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Lomonte, P. & Morency, E. Centromeric protein CENP-B proteasomal degradation induced by the viral protein ICP0. FEBS Lett. 581, 658–662 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Perusina Lanfranca, M., Mostafa, H. H. & Davido, D. J. HSV-1 ICP0: an E3 ubiquitin ligase that counteracts host intrinsic and innate immunity. Cells 3, 438–454 (2014).

    Article  Google Scholar 

  33. Alandijany, T. Host intrinsic and innate intracellular immunity during herpes simplex virus type 1 (HSV-1) infection. Front. Microbiol. 10, 2611 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Merkl, P. E., Orzalli, M. H. & Knipe, D. M. Mechanisms of host IFI16, PML, and Daxx protein restriction of herpes simplex virus 1 replication. J. Virol. 92, e00057-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Lilley, C. E., Chaurushiya, M. S., Boutell, C., Everett, R. D. & Weitzman, M. D. The intrinsic antiviral defense to incoming HSV-1 genomes includes specific DNA repair proteins and is counteracted by the viral protein ICP0. PLoS Pathog. 7, e1002084 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cuchet-Lourenço, D. et al. SUMO pathway dependent recruitment of cellular repressors to herpes simplex virus type 1 genomes. PLoS Pathog. 7, e1002123 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Sirbu, B. M. et al. Analysis of protein dynamics at active, stalled, and collapsed replication forks. Genes Dev. 25, 1320–1327 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dembowski, J. A. & DeLuca, N. A. Selective recruitment of nuclear factors to productively replicating herpes simplex virus genomes. PLoS Pathog. 11, e1004939 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Dembowski, J. A. & DeLuca, N. A. Temporal viral genome-protein interactions define distinct stages of productive herpesviral infection. mBio 9, e01182–18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Reyes, E. D. et al. Identifying host factors associated with DNA replicated during virus infection. Mol. Cell. Proteomics 16, 2079–2097 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Liu, F., Zhou, P., Wang, Q., Zhang, M. & Li, D. The Schlafen family: complex roles in different cell types and virus replication. Cell Biol. Int. 42, 2–8 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Mavrommatis, E., Fish, E. N. & Platanias, L. C. The Schlafen family of proteins and their regulation by interferons. J. Interferon Cytokine Res. 33, 206–210 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Katsoulidis, E. et al. Role of interferon α (IFNα)-inducible Schlafen-5 in regulation of Anchorage-independent growth and invasion of malignant melanoma cells. J. Biol. Chem. 285, 40333–40341 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sassano, A. et al. Human Schlafen 5 (SLFN5) is a regulator of motility and invasiveness of renal cell carcinoma cells. Mol. Cell. Biol. 35, 2684–2698 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Arslan, A. D. et al. Human SLFN5 is a transcriptional co-repressor of STAT1-mediated interferon responses and promotes the malignant phenotype in glioblastoma. Oncogene 36, 6006–6019 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, M. et al. Codon-usage-based inhibition of HIV protein synthesis by human schlafen 11. Nature 491, 125–128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Seong, R.-K. et al. Schlafen 14 (SLFN14) is a novel antiviral factor involved in the control of viral replication. Immunobiology 222, 979–988 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yang, J.-Y. et al. Structure of Schlafen13 reveals a new class of tRNA/rRNA-targeting RNase engaged in translational control. Nat. Commun. 9, 1165 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Valdez, F. et al. Schlafen 11 restricts flavivirus replication. J. Virol. 93, e00104-19 (2019).

  50. de la Casa-Esperón, E. From mammals to viruses: the Schlafen genes in developmental, proliferative and immune processes. Biomol. Concepts 2, 159–169 (2011).

    Article  PubMed  Google Scholar 

  51. Kulej, K. et al. Time-resolved global and chromatin proteomics during herpes simplex virus type 1 (HSV-1) infection. Mol. Cell. Proteomics 16, S92–S107 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Schreiner, S. et al. Proteasome-dependent degradation of Daxx by the viral E1B-55K protein in human adenovirus-infected cells. J. Virol. 84, 7029–7038 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dutrieux, J. et al. PML/TRIM19-dependent inhibition of retroviral reverse-transcription by Daxx. PLoS Pathog. 11, e1005280 (2015).

  54. Bernassola, F. et al. Ubiquitin-dependent degradation of p73 is inhibited by PML. J. Exp. Med. 199, 1545–1557 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Louria-Hayon, I. et al. The promyelocytic leukemia protein protects p53 from Mdm2-mediated inhibition and degradation. J. Biol. Chem. 278, 33134–33141 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Boutell, C. et al. A viral ubiquitin ligase has substrate preferential SUMO targeted ubiquitin ligase activity that counteracts intrinsic antiviral defence. PLoS Pathog. 7, e1002245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sloan, E. et al. Analysis of the SUMO2 proteome during HSV-1 Infection. PLoS Pathog. 11, e1005059 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Kumar, R., González-Prieto, R., Xiao, Z., Verlaan-de Vries, M. & Vertegaal, A. C. O. The STUbL RNF4 regulates protein group SUMOylation by targeting the SUMO conjugation machinery. Nat. Commun. 8, 1809 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Neumann, B., Zhao, L., Murphy, K. & Gonda, T. J. Subcellular localization of the Schlafen protein family. Biochem. Biophys. Res. Commun. 370, 62–66 (2008).

    Article  CAS  PubMed  Google Scholar 

  60. Taylor, K. E., Chew, M. V., Ashkar, A. A. & Mossman, K. L. Novel roles of cytoplasmic ICP0: proteasome-independent functions of the RING finger are required to block interferon-stimulated gene production but not to promote viral replication. J. Virol. 88, 8091–8101 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Lou, D. I. et al. An intrinsically disordered region of the DNA repair protein Nbs1 is a species-specific barrier to herpes simplex virus 1 in primates. Cell Host Microbe 20, 178–188 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Alandijany, T. et al. Distinct temporal roles for the promyelocytic leukaemia (PML) protein in the sequential regulation of intracellular host immunity to HSV-1 infection. PLoS Pathog. 14, e1006769 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Geserick, P., Kaiser, F., Klemm, U., Kaufmann, S. H. E. & Zerrahn, J. Modulation of T cell development and activation by novel members of the Schlafen (slfn) gene family harbouring an RNA helicase-like motif. Int. Immunol. 16, 1535–1548 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Stabell, A. C. et al. Non-human primate Schlafen11 inhibits production of both host and viral proteins. PLoS Pathog. 12, e1006066 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Stow, N. D. & Stow, E. C. Isolation and characterization of a herpes simplex virus type 1 mutant containing a deletion within the gene encoding the immediate early polypeptide Vmw110. J. Gen. Virol. 67, 2571–2585 (1986).

    Article  CAS  PubMed  Google Scholar 

  66. Everett, R. D. Construction and characterization of herpes simplex virus type 1 mutants with defined lesions in immediate early gene 1. J. Gen. Virol. 70, 1185–1202 (1989).

    Article  CAS  PubMed  Google Scholar 

  67. Sirbu, B. M., Couch, F. B. & Cortez, D. Monitoring the spatiotemporal dynamics of proteins at replication forks and in assembled chromatin using isolation of proteins on nascent DNA. Nat. Protoc. 7, 594–605 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Stacklies, W., Redestig, H., Scholz, M., Walther, D. & Selbig, J. pcaMethods: a Bioconductor package providing PCA methods for incomplete data. Bioinformatics 23, 1164–1167 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Pascovici, D., Handler, D. C. L., Wu, J. X. & Haynes, P. A. Multiple testing corrections in quantitative proteomics: a useful but blunt tool. Proteomics 16, 2448–2453 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Halford, W. P., Kemp, C. D., Isler, J. A., Davido, D. J. & Schaffer, P. A. ICP0, ICP4, or VP16 expressed from adenovirus vectors induces reactivation of latent herpes simplex virus type 1 in primary cultures of latently infected trigeminal ganglion cells. J. Virol. 75, 6143–6153 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Poling, B. C., Price, A. M., Luftig, M. A. & Cullen, B. R. The Epstein–Barr virus miR-BHRF1 microRNAs regulate viral gene expression in cis. Virology 512, 113–123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Russo, J., Heck, A. M., Wilusz, J. & Wilusz, C. J. Metabolic labeling and recovery of nascent RNA to accurately quantify mRNA stability. Methods 120, 39–48 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Price, A. M., Messinger, J. E. & Luftig, M. A. c-Myc represses transcription of Epstein–Barr virus latent membrane protein 1 early after primary B cell infection. J. Virol. 92, e01178–17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Dölken, L. et al. High-resolution gene expression profiling for simultaneous kinetic parameter analysis of RNA synthesis and decay. RNA 14, 1959–1972 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Weitzman laboratory for insightful discussions and input. We thank G. H. Cohen, R. D. Everett, D. M. Knipe, S. L. Sawyer and P. A. Schaffer for generous gifts of reagents. C.B. was supported by a Medical Research Council grant no. MC_UU_12014/5. Work in the Weitzman laboratory was supported in part by grants from the National Institutes of Health (grant nos. AI115104 and NS082240 to M.D.W.) and funds from the Children’s Hospital of Philadelphia. A.M.P. was supported by the National Cancer Institute T32 Training Grant in Tumor Virology no. T32-CA115299 and Individual National Research Service Award no. F32-AI138432. J.M.D. was supported by the Individual National Research Service Award no. F32-AI147587.

Author information

Authors and Affiliations

Authors

Contributions

E.T.K., J.M.D., E.D.R. and M.D.W. conceived and designed the study. E.D.R., K.K. and B.A.G. performed the iPOND MS and subsequent analysis. J.M.D. provided the computational analysis. E.T.K., E.D.R., A.M.P., A.O. and C.B. performed the cell imaging experiments. A.M.P. performed the RNA stability assays. E.T.K. and L.N.A. performed the virological, biochemical and molecular biology experiments. E.T.K. and M.D.W. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Matthew D. Weitzman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Comparative analysis of protein abundance between PCA-based clustering of HSV iPOND data and whole cell proteome data identifies potential substrates of ICP0.

a, Venn diagram of PML, ATRX, IFI16, and DNA-PKcs PCA-based clusters identifies proteins that are clustered with each known substrate. b, Volcano plots showing log2 fold change (x-axis) and associated statistical significance (y-axis) for proteins identified by iPOND proteomics, quantifying proteins associated with HSV genomes. Known ICP0 substrates (green) and clustered proteins (blue and red) are indicated. iPOND data comparing HSV WT and ΔICP0 infection shows known substrates and clustered proteins enriched on the ΔICP0 genome. P-values were calculated using the two-tailed unpaired Student’s t-test. n = 3 biologically independent experiments. c, Heatmap showing iPOND abundances as z-scores for mock, WT HSV-1, and ΔICP0 during infection. This list is proteins that are more than 2-fold enriched (dashed gray box in b) on the ΔICP0 virus genome compared to WT HSV-1 genome. These are clustered using a hierarchical clustering algorithm that analyzes the abundance for each condition. Therefore, the proteins with the same trends for mock-HSV-ΔICP0 are close together on the heatmap. d, Whole cell proteome abundance data over a time course of WT HSV-1 infection show decreases in known ICP0 substrates PML, IFI16, and DNA-PKcs, as well as SLFN5. Additional proteins (gray line) showing similar trends in the heat map (c) are not decreased during HSV infection.

Extended Data Fig. 2 Comparison of SLFN5 degradation in different cells.

a, HEK293T cells were transfected with plasmids encoding SLFN5-V5 and GFP-ICP0 (WT or ΔRING), as indicated. At 24 h after transfection, total cell lysates were prepared, and immunoblot analysis was carried out with indicated antibodies. GAPDH was used as a loading control. b, HEK293T cells were transfected with plasmids encoding GFP-SLFN5 and ICP0 (WT or ΔRING), and the cells were monitored for the GFP signals and the fluorescence intensity was measured by ImageJ software. Scale bar, 300 µm. c, HEK293 cells were infected with WT HSV-1 or ΔRING virus at an MOI of 3. Cell lysates were prepared at the indicated time points and subjected to immunoblot analysis. d, HeLa cells were infected with WT HSV-1 or ΔRING virus at an MOI of 3. Cell lysates were prepared at the indicated time points and subjected to immunoblot analysis. e, HeLa cells were transduced with recombinant adenoviral vectors encoding ICP0 together with Ad.C-rtTA, an adenovirus expressing the doxycycline regulated transactivator. At 24 h after transduction in the absence or presence of doxycycline (1 µg/ml), immunoblot analysis was performed with indicated antibodies. f, g, Similar experimental setup as in (d and e); U2OS cells were used. h, SLFN5 degradation is independent of SUMOylation, and is detected in cells depleted of the SUMO E2 conjugating enzyme UBC9. i, In vitro ubiquitination assay with bacterially purified SLFN5 Δ730-891 truncation mutant which lacks the ICP0 interaction region demonstrates it is not ubiquitinated. Immunoblot data shows representative data from n = 3 biologically independent experiments.

Source data

Extended Data Fig. 3 ICP0 interacts SLFN5 via the carboxy terminal region of SLFN5.

a, Subcellular localization of SLFN5 mutants was analyzed. HEK293T cells were transfected with plasmids encoding GFP-SLFN5 (full-length or various deletion mutants). At 24 hours after transfection, the cells monitored for the GFP signals. Scale bar, 50 µm. b, HEK293T cells were co-transfected with plasmids encoding various version of GFP-SLFN5 and ICP0 ΔRING or cytoplasmic ICP0 mutant (cICP0, also called D8) ΔRING, as indicated. The cells were subject to co-IP with anti-GFP Ab, followed by immunoblotting. Immunoblot and immunofluorescence images show representative data from n = 3 biologically independent experiments. c, SLFN5 interaction with ICP0 maps to a part of the intrinsicly disordered region of SLFN5. The SLFN5 and SLFN11 protein sequences were analyzed for disorder tendency using IUPred2A. The domain structure of SLFN is shown above. The green bar indicates an interaction region of ICP0 or a homolgous region of SLFN11 with SLFN5. Red lines indicate unique disorder region of SLFN5.

Source data

Extended Data Fig. 4 Colocalization of SLFN5 with replication compartments of HSV-1.

a, HFF cells were infected with WT HSV-1 at an MOI of 10 and immunofluorescence was examined at 2 hpi. ICP4 marks viral pre-replication foci from incoming viral genomes. b, HFF cells were infected with WT HSV or ΔICP0 mutant virus at an MOI of 3 and immunofluorescence was examined at 8 hpi. ICP8 marks viral replication compartments and DAPI marks cellular DNA. Scale bar, 10 µm. Fluorescence plot profile analyzed by ImageJ showed colocalization of SLFN5 and viral proteins (at dashed line). Immunofluorescence images show representative data from n = 3 biologically independent experiments.

Extended Data Fig. 5 SLFN5 suppresses HSV-1 replication.

a, Efficiency of SLFN5 knockdown in HFF and HeLa cells. Immunoblot analysis indicating SLFN5 and β-actin levels following transduction with lentiviruses encoding shRNAs that are nontargeting control (Ctrl) or specific for SLFN5 (#1 to #5). b, Viral DNA replication yields from shRNA-transduced HFF cells infected with ΔRING virus at an MOI of 0.1 and harvested at 24 h after infection. Quantification of viral DNA was carried out by qPCR. n = 5 biologically independent experiments. c, HeLa cells transduced with shRNA were mock infected or infected with HSV-1 (WT or ΔRING) at different MOIs (0.5 to 3) as indicated. Cell lysates were prepared at 9 h after infection and immunoblot nalysis was performed with antibodies to ICP0, VP21, and β-actin. d, Phase micrograph of cell morphology under subconfluent or confluent culture conditions. Scale bar, 100 µm. e, Growth of shRNA transduced cells over time. n = 3 biologically independent experiments. f, 2,000 cells were seeded in a 96-well plate, and cell proliferation was measured over 6 days by colorimetric analysis using the water-soluble tetrazolium salt, WST-8 (Cell Counting Kit-8, Dojindo) at the indicated time point. n = 3 biologically independent experiments. g, CRISPR/Cas9-mediated SLFN5 gene editing and permanent depletion from HeLa cells. Guide RNA transfected cells were selected and cloned for a month. Immunoblot analysis of SLFN5 in control cells or SLFN5 knockout cells. Viral DNA replication yields from cells infected with ΔRING at an MOI of 0.1 were analyzed by qPCR. HeLa cells were infected with ΔRING at an MOI of 0.1 for 24 h. Viral DNA was measured by qPCR. n = 3 biologically independent experiments. Data are the mean ± SD. Comparisons between groups were performed using the two-tailed unpaired Student’s t-test. **, p < 0.005, ***, p < 0.0005, n.s., not significant. Immunoblots show representative data from n = 3 biologically independent experiments.

Source data

Extended Data Fig. 6 SLFN5 Δ730-891 mutant does not inhibit HSV-1 replication.

a, HeLa cells were stably transduced with lentivirus containing tetracycline-inducible SLFN5-HA genes (WT and Δ730-891). SLFN5-HA was induced with doxycycline (0.5 μg/ml) for 48 h. SLFN5 expression was confirmed by immunoblot analysis. b, c, Cells were infected with HSV-1 (b) and ΔRING virus (c) at an MOI of 0.1. At 24 hpi, viral DNA was measured by qPCR. n = 3 biologically independent experiments. Data are the mean ± SD. Comparisons between groups were performed using the two-tailed unpaired Student’s t-test. **, p < 0.005. n.s., not significant.

Source data

Extended Data Fig. 7 Degradation of SLFN5 by other DNA viruses.

a, HFF cells were mock infected or infected with HSV-2 at an MOI of 3, and MG132 (5 µM) was added at 2 h after virus inoculation as indicated. Total cell lysates were prepared at indicated time points and examined by immunoblot analysis with indicated antibodies. ICP8 serves as an infection control. b, HFF cells were mock infected or infected with HCMV (TB40/E) at an MOI of 3 and total cell lysates were prepared at indicated time points and examined by immunoblot analysis with indicated antibodies. UL44 serves as an infection control. Circle marks cross-reacting viral protein. c, A549 cells were mock infected or infected with adenovirus (Ad5) at an MOI of 20 and MG132 (5 µM) was added at 2 h after virus inoculation, as indicated. Cell lysates were prepared at the indicated time points and examined by immunoblot analysis with indicated antibodies. RAD50 served as a degradation control and DBP as a control for infection. Immunoblots show representative data from n = 3 biologically independent experiments.

Source data

Extended Data Fig. 8 Restriction of other DNA viruses by SLFN5.

HeLa cells were mock infected or infected with other DNA viruses as indicated. a, Immunoblot analysis demonstrating increased HSV-2 protein levels in SLFN5-depleted cells. b, HCMV proteins are slightly increased in SLFN5-depleted cells at immediate-early time point (24 hpi), but decreased at late times (72 hpi) as compared to control cells. The ISG15 protein was increased in the absence of SLFN5 and further increased upon HCMV infection. c, SLFN5 does not affect adenovirus protein levels. d, SLFN5 knockdown results in higher level of ISG15 expression in HFF and HeLa cells. Immunoblots show representative data from n = 3 biologically independent experiments.

Source data

Extended Data Fig. 9 Effects of SLFN5 on HCMV infection.

a, b, HCMV mRNA (UL123, an immediate-early gene and UL94, a late gene) expression from shRNA-transduced HFF cells infected with HCMV at an MOI of 3 and harvested at 12 h (a) and 72 h (b) after infection. Quantification of viral transcripts was carried out by RT-qPCR. Viral DNA replication yields from shRNA-transduced HFF cells infected with ΔRING virus at an MOI of 0.1 and harvested at 24 h after infection. Quantification of viral DNA was carried out by qPCR. c, Progeny production of HCMV was monitored by infectious center assays using anti-IE1 antibody. n = 3 biologically independent experiments. Data are the mean ± SD. Comparisons between groups were performed using the two-tailed unpaired Student’s t-test. *, p < 0.05, **, p < 0.005.

Source data

Extended Data Fig. 10 Interaction of SLFN5 with HSV-1 DNA.

a. ChIP assay with anti-HA antibody was performed in SLFN5-HA-expressed HeLa cells. ΔRING virus was infected at an MOI of 3 for indicated time points in the absence or presence of PAA. ChIP DNA was assayed by qPCR using primers specific for US11 TSS. n = 3 biologically independent experiments. b. ΔRING virus was infected at an MOI of 3 for 3 h. ChIP DNA was assayed by qPCR using primers specific for the HSV-1 DNA or human DNA loci. n = 3 biologically independent experiments. Data are the mean ± SD. Comparisons between groups were performed using the two-tailed unpaired Student’s t-test. *, p < 0.05, **, p < 0.005.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables

iPOND proteomics quantification data and associated analysis.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, E.T., Dybas, J.M., Kulej, K. et al. Comparative proteomics identifies Schlafen 5 (SLFN5) as a herpes simplex virus restriction factor that suppresses viral transcription. Nat Microbiol 6, 234–245 (2021). https://doi.org/10.1038/s41564-020-00826-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-00826-3

This article is cited by

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