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.

  • Letter
  • Published:

Inhibition of the histone demethylase LSD1 blocks α-herpesvirus lytic replication and reactivation from latency

Abstract

Reversible methylation of histone tails serves as either a positive signal recognized by transcriptional assemblies or a negative signal that result in repression1,2,3,4. Invading viral pathogens that depend upon the host cell's transcriptional apparatus are also subject to the regulatory impact of chromatin assembly and modifications5,6,7,8. Here we show that infection by the α-herpesviruses, herpes simplex virus (HSV) and varicella zoster virus (VZV), results in the rapid accumulation of chromatin bearing repressive histone H3 Lys9 methylation. To enable expression of viral immediate early (IE) genes, both viruses use the cellular transcriptional coactivator host cell factor-1 (HCF-1) to recruit the lysine-specific demethylase-1 (LSD1) to the viral immediate early promoters. Depletion of LSD1 or inhibition of its activity with monoamine oxidase inhibitors (MAOIs) results in the accumulation of repressive chromatin and a block to viral gene expression. As HCF-1 is a component of the Set1 and MLL1 histone H3 Lys4 methyltransferase complexes9,10, it thus coordinates modulation of repressive H3 Lys9 methylation levels with addition of activating H3 Lys4 trimethylation marks. Strikingly, MAOIs also block the reactivation of HSV from latency in sensory neurons, indicating that the HCF-1 complex is a crucial component of the reactivation mechanism. The results support pharmaceutical control of histone modifying enzymes as a strategy for controlling herpesvirus infections.

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

Access options

Buy this article

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

Figure 1: LSD1 is crucial for viral activator–mediated transcription of VZV IE and HSV IE model promoters.
Figure 2: An HCF-1–LSD1 complex is essential for α-herpesvirus IE gene transcription.
Figure 3: Inhibition of LSD1 with MAOIs blocks α-herpesviral lytic gene expression.
Figure 4: Inhibition of LSD1 with MAOIs blocks HSV-1 reactivation from latency.

Similar content being viewed by others

References

  1. Klose, R.J. & Zhang, Y. Regulation of histone methylation by demethylimination and demethylation. Nat. Rev. Mol. Cell Biol. 8, 307–318 (2007).

    Article  CAS  Google Scholar 

  2. Ruthenburg, A.J., Allis, C.D. & Wysocka, J. Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol. Cell 25, 15–30 (2007).

    Article  CAS  Google Scholar 

  3. Shi, Y. & Whetstine, J.R. Dynamic regulation of histone lysine methylation by demethylases. Mol. Cell 25, 1–14 (2007).

    Article  CAS  Google Scholar 

  4. Kouzarides, T. & Berger, S.L. Chromatin modifications and their mechanism of action. in Epigenetics (eds. Allis, C.D., Jenuwein, T. & Reinberg, D.) 191–209 (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2007).

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

    Article  CAS  Google Scholar 

  6. Lieberman, P.M. Chromatin organization and virus gene expression. J. Cell. Physiol. 216, 295–302 (2008).

    Article  CAS  Google Scholar 

  7. Mok, H.P. & Lever, A.M. Chromatin, gene silencing and HIV latency. Genome Biol. 8, 228 (2007).

    Article  Google Scholar 

  8. Sinclair, J. Chromatin structure regulates human cytomegalovirus gene expression during latency, reactivation and lytic infection. Biochim. Biophys. Acta published online, doi:10.1016/j.bbagrm.2009.08.001 (12 August 2009).

  9. Wysocka, J., Myers, M.P., Laherty, C.D., Eisenman, R.N. & Herr, W. Human Sin3 deacetylase and trithorax-related Set1/Ash2 histone H3–K4 methyltransferase are tethered together selectively by the cell-proliferation factor HCF-1. Genes Dev. 17, 896–911 (2003).

    Article  CAS  Google Scholar 

  10. Yokoyama, A. et al. Leukemia proto-oncoprotein MLL forms a SET1-like histone methyltransferase complex with menin to regulate Hox gene expression. Mol. Cell. Biol. 24, 5639–5649 (2004).

    Article  CAS  Google Scholar 

  11. Kristie, T.M. Early events pre-initiation of alphaherpesvirus viral gene expression. in Human Herpesviruses Biology, Therapy, and Immunoprophylaxis (eds. Arvin, A. et al.) 112–127 (Cambridge University Press, New York, 2007).

  12. Narayanan, A., Ruyechan, W.T. & Kristie, T.M. The coactivator host cell factor-1 mediates Set1 and MLL1 H3K4 trimethylation at herpesvirus immediate early promoters for initiation of infection. Proc. Natl. Acad. Sci. USA 104, 10835–10840 (2007).

    Article  CAS  Google Scholar 

  13. Huang, J. et al. Trimethylation of histone H3 lysine 4 by Set1 in the lytic infection of human herpes simplex virus 1. J. Virol. 80, 5740–5746 (2006).

    Article  CAS  Google Scholar 

  14. Garcia-Bassets, I. et al. Histone methylation–dependent mechanisms impose ligand dependency for gene activation by nuclear receptors. Cell 128, 505–518 (2007).

    Article  CAS  Google Scholar 

  15. Metzger, E. et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 437, 436–439 (2005).

    Article  CAS  Google Scholar 

  16. Perillo, B. et al. DNA oxidation as triggered by H3K9me2 demethylation drives estrogen-induced gene expression. Science 319, 202–206 (2008).

    Article  CAS  Google Scholar 

  17. Wang, J. et al. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 446, 882–887 (2007).

    Article  CAS  Google Scholar 

  18. Dou, Y. et al. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat. Struct. Mol. Biol. 13, 713–719 (2006).

    Article  CAS  Google Scholar 

  19. Lee, M.G., Wynder, C., Cooch, N. & Shiekhattar, R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 437, 432–435 (2005).

    Article  CAS  Google Scholar 

  20. Shi, Y.J. et al. Regulation of LSD1 histone demethylase activity by its associated factors. Mol. Cell 19, 857–864 (2005).

    Article  CAS  Google Scholar 

  21. Forneris, F., Binda, C., Vanoni, M.A., Mattevi, A. & Battaglioli, E. Histone demethylation catalysed by LSD1 is a flavin-dependent oxidative process. FEBS Lett. 579, 2203–2207 (2005).

    Article  CAS  Google Scholar 

  22. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    Article  CAS  Google Scholar 

  23. Lee, M.G., Wynder, C., Schmidt, D.M., McCafferty, D.G. & Shiekhattar, R. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chem. Biol. 13, 563–567 (2006).

    Article  CAS  Google Scholar 

  24. Schmidt, D.M. & McCafferty, D.G. trans-2-phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry 46, 4408–4416 (2007).

    Article  CAS  Google Scholar 

  25. Yang, M. et al. Structural basis for the inhibition of the LSD1 histone demethylase by the antidepressant trans-2-phenylcyclopropylamine. Biochemistry 46, 8058–8065 (2007).

    Article  CAS  Google Scholar 

  26. Shin, S. & Janknecht, R. Activation of androgen receptor by histone demethylases JMJD2A and JMJD2D. Biochem. Biophys. Res. Commun. 359, 742–746 (2007).

    Article  CAS  Google Scholar 

  27. Wissmann, M. et al. Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor–dependent gene expression. Nat. Cell Biol. 9, 347–353 (2007).

    Article  CAS  Google Scholar 

  28. Amelio, A.L., Giordani, N.V., Kubat, N.J., O'Neil, J.E. & Bloom, D.C. Deacetylation of the herpes simplex virus type 1 latency-associated transcript (LAT) enhancer and a decrease in LAT abundance precede an increase in ICP0 transcriptional permissiveness at early times postexplant. J. Virol. 80, 2063–2068 (2006).

    Article  CAS  Google Scholar 

  29. Kubat, N.J., Tran, R.K., McAnany, P. & Bloom, D.C. Specific histone tail modification and not DNA methylation is a determinant of herpes simplex virus type 1 latent gene expression. J. Virol. 78, 1139–1149 (2004).

    Article  CAS  Google Scholar 

  30. Neumann, D.M., Bhattacharjee, P.S., Giordani, N.V., Bloom, D.C. & Hill, J.M. In vivo changes in the patterns of chromatin structure associated with the latent herpes simplex virus type 1 genome in mouse trigeminal ganglia can be detected at early times after butyrate treatment. J. Virol. 81, 13248–13253 (2007).

    Article  CAS  Google Scholar 

  31. Wang, Q.Y. et al. Herpesviral latency–associated transcript gene promotes assembly of heterochromatin on viral lytic-gene promoters in latent infection. Proc. Natl. Acad. Sci. USA 102, 16055–16059 (2005).

    Article  CAS  Google Scholar 

  32. Kolb, G. & Kristie, T.M. Association of the cellular coactivator HCF-1 with the Golgi apparatus in sensory neurons. J. Virol. 82, 9555–9563 (2008).

    Article  CAS  Google Scholar 

  33. Kristie, T.M., Vogel, J.L. & Sears, A.E. Nuclear localization of the C1 factor (host cell factor) in sensory neurons correlates with reactivation of herpes simplex virus from latency. Proc. Natl. Acad. Sci. USA 96, 1229–1233 (1999).

    Article  CAS  Google Scholar 

  34. Whitlow, Z. & Kristie, T.M. Recruitment of the transcriptional coactivator HCF-1 to viral immediate-early promoters during initiation of reactivation from latency of herpes simplex virus type 1. J. Virol. 83, 9591–9595 (2009).

    Article  CAS  Google Scholar 

  35. Pesola, J.M., Zhu, J., Knipe, D.M. & Coen, D.M. Herpes simplex virus 1 immediate-early and early gene expression during reactivation from latency under conditions that prevent infectious virus production. J. Virol. 79, 14516–14525 (2005).

    Article  CAS  Google Scholar 

  36. Sawtell, N.M., Thompson, R.L. & Haas, R.L. Herpes simplex virus DNA synthesis is not a decisive regulatory event in the initiation of lytic viral protein expression in neurons in vivo during primary infection or reactivation from latency. J. Virol. 80, 38–50 (2006).

    Article  CAS  Google Scholar 

  37. Oh, J. & Fraser, N.W. Temporal association of the herpes simplex virus genome with histone proteins during a lytic infection. J. Virol. 82, 3530–3537 (2008).

    Article  CAS  Google Scholar 

  38. Placek, B.J. et al. The histone variant H3.3 regulates gene expression during lytic infection with herpes simplex virus type 1. J. Virol. 83, 1416–1421 (2009).

    Article  CAS  Google Scholar 

  39. Huang, J. et al. p53 is regulated by the lysine demethylase LSD1. Nature 449, 105–108 (2007).

    Article  CAS  Google Scholar 

  40. Wang, J. et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet. 41, 125–129 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Skinner for expert statistical analysis of the experimental data; B. Moss, J. Yewdell, T. Pierson, J. Bennink and A. McBride for critical discussions and comments on this manuscript; members of the Molecular Genetics Section of the Laboratory of Viral Diseases for discussion, advice and technical assistance; the US National Institute of Allergy and Infectious Diseases Bld33 Animal Care Facility staff; N. Fraser (University of Pennsylvania School of Medicine) for HSV-1 strain 17, W. Ruyechan (University at Buffalo, State University of New York) for ICP8–specific sera and X. Chen (University of California at Davis) for MCF7 inducible LSD shRNA cells. These studies were supported by the Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Y.L. performed ChIP, quantitative PCR, immunofluorescence and animal reactivation studies; A.N. and H.P. performed ChIP and quantitative PCR analyses; J.L.V. performed reporter and immunoprecipitation assays; T.M.K. designed the study, performed animal reactivation studies and wrote the paper. J.L.V. and A.N. contributed equally to this study. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Thomas M Kristie.

Ethics declarations

Competing interests

The US National Institutes of Health Department of Health and Human Services has filed US patent application No. 61/083,304 and international patent application No. PCT/US2009/051557 for methods of preventing infection by herpes viruses or treating reactivation after latency in a host by inhibitors of the Lsd1 protein.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–4 and Supplementary Methods (PDF 609 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Liang, Y., Vogel, J., Narayanan, A. et al. Inhibition of the histone demethylase LSD1 blocks α-herpesvirus lytic replication and reactivation from latency. Nat Med 15, 1312–1317 (2009). https://doi.org/10.1038/nm.2051

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2051

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