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:

The Toxoplasma effector TEEGR promotes parasite persistence by modulating NF-κB signalling via EZH2

Abstract

The protozoan parasite Toxoplasma gondii has co-evolved with its homeothermic hosts (humans included) strategies that drive its quasi-asymptomatic persistence in hosts, hence optimizing the chance of transmission to new hosts. Persistence, which starts with a small subset of parasites that escape host immune killing and colonize the so-called immune privileged tissues where they differentiate into a low replicating stage, is driven by the interleukin 12 (IL-12)–interferon-γ (IFN-γ) axis. Recent characterization of a family of Toxoplasma effectors that are delivered into the host cell, in which they rewire the host cell gene expression, has allowed the identification of regulators of the IL-12–IFN-γ axis, including repressors. We now report on the dense granule-resident effector, called TEEGR (Toxoplasma E2F4-associated EZH2-inducing gene regulator) that counteracts the nuclear factor-κB (NF-κB) signalling pathway. Once exported into the host cell, TEEGR ends up in the nucleus where it not only complexes with the E2F3 and E2F4 host transcription factors to induce gene expression, but also promotes shaping of a non-permissive chromatin through its capacity to switch on EZH2. Remarkably, EZH2 fosters the epigenetic silencing of a subset of NF-κB-regulated cytokines, thereby strongly contributing to the host immune equilibrium that influences the host immune response and promotes parasite persistence in mice.

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

Fig. 1: The export of TEEGR in the host cell nucleus.
Fig. 2: TEEGR activates gene expression in human cells in a E2F3 and E2F4-dependent manner.
Fig. 3: TEEGR forms a partnership with the E2F–DP host transcription factors.
Fig. 4: E2F3 and E2F4 DNA association with the EZH2 promoter is enhanced by T. gondii infection in a TEEGR-dependent manner.
Fig. 5: TEEGR-dependent repression of NF-κB-regulated genes is mediated by EZH2.
Fig. 6: In vivo control of teegr-deficient T. gondii tachyzoite population is likely to be mediated by NF-κB-regulated pro-inflammatory cytokines.

Similar content being viewed by others

Data availability

Correspondence and requests for materials should be addressed to M.-A.H. The microarray data have been deposited to the GEO datasets under the accession numbers GSE113618 and GSE113626.

References

  1. Jeffers, V., Tampaki, Z., Kim, K. & Sullivan, W. J. A latent ability to persist: differentiation in Toxoplasma gondii. Cell. Mol. Life Sci. 75, 2355–2373 (2018).

  2. Melo, M. B., Jensen, K. D. C. & Saeij, J. P. J. Toxoplasma gondii effectors are master regulators of the inflammatory response. Trends Parasitol. 27, 487–495 (2011).

    Article  CAS  Google Scholar 

  3. Hunter, C. A. & Sibley, L. D. Modulation of innate immunity by Toxoplasma gondii virulence effectors. Nat. Rev. Microbiol. 10, 766–778 (2012).

    Article  CAS  Google Scholar 

  4. Bougdour, A. et al. Host cell subversion by Toxoplasma GRA16, an exported dense granule protein that targets the host cell nucleus and alters gene expression. Cell Host Microbe 13, 489–500 (2013).

    Article  CAS  Google Scholar 

  5. Braun, L. et al. A Toxoplasma dense granule protein, GRA24, modulates the early immune response to infection by promoting a direct and sustained host p38 MAPK activation. J. Exp. Med. 210, 2071–2086 (2013).

    Article  CAS  Google Scholar 

  6. Gay, G. et al. Toxoplasma gondii TgIST co-opts host chromatin repressors dampening STAT1-dependent gene regulation and IFN-γ-mediated host defenses. J. Exp. Med. 213, 1779–1798 (2016).

    Article  CAS  Google Scholar 

  7. Olias, P., Etheridge, R. D., Zhang, Y., Holtzman, M. J. & Sibley, L. D. Toxoplasma effector recruits the Mi-2/NuRD complex to repress STAT1 transcription and block IFN-γ-dependent gene expression. Cell Host Microbe 20, 72–82 (2016).

    Article  CAS  Google Scholar 

  8. Hakimi, M.-A., Olias, P. & Sibley, L. D. Toxoplasma effectors targeting host signaling and transcription. Clin. Microbiol. Rev. 30, 615–645 (2017).

    Article  Google Scholar 

  9. Fischer, M., Grossmann, P., Padi, M. & DeCaprio, J. A. Integration of TP53, DREAM, MMB-FOXM1 and RB-E2F target gene analyses identifies cell cycle gene regulatory networks. Nucleic Acids Res. 44, 6070–6086 (2016).

    Article  CAS  Google Scholar 

  10. Marson, A. et al. Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature 445, 931–935 (2007).

    Article  CAS  Google Scholar 

  11. Julian, L. M. et al. Tissue-specific targeting of cell fate regulatory genes by E2f factors. Cell Death Differ. 23, 565–575 (2016).

    Article  CAS  Google Scholar 

  12. Zheng, N., Fraenkel, E., Pabo, C. O. & Pavletich, N. P. Structural basis of DNA recognition by the heterodimeric cell cycle transcription factor E2F–DP. Gene Dev. 13, 666–764 (1999).

    Article  CAS  Google Scholar 

  13. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  Google Scholar 

  14. Bracken, A. P. et al. EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplied in cancer. EMBO J. 22, 5323–5335 (2003).

    Article  CAS  Google Scholar 

  15. Garber, M. et al. A high-throughput chromatin immunoprecipitation approach reveals principles of dynamic gene regulation in mammals. Mol. Cell 47, 810–822 (2012).

    Article  CAS  Google Scholar 

  16. Xing, Y., Zhou, F. & Wang, J. Subset of genes targeted by transcription factor NF-κB in TNFα-stimulated human HeLa cells. Funct. Integr. Genomics 13, 143–154 (2013).

    Article  CAS  Google Scholar 

  17. Luo, X., Chae, M., Krishnakumar, R., Danko, C. G. & Kraus, W. Dynamic reorganization of the AC16 cardiomyocyte transcriptome in response to TNFα signaling revealed by integrated genomic analyses. BMC Genom. 15, 155 (2014).

    Article  Google Scholar 

  18. Afonina, I. S., Zhong, Z., Karin, M. & Beyaert, R. Limiting inflammation—the negative regulation of NF-κB and the NLRP3 inflammasome. Nat. Immunol. 18, 861–869 (2017).

    Article  CAS  Google Scholar 

  19. Sun, F. et al. Combinatorial pharmacologic approaches target EZH2-mediated gene repression in breast cancer cells. Mol. Cancer Ther. 8, 3191–3202 (2009).

    Article  CAS  Google Scholar 

  20. Lee, S. T. et al. Context-specific regulation of NF-κB target gene expression by EZH2 in breast cancers. Mol. Cell 43, 798–810 (2011).

    Article  CAS  Google Scholar 

  21. Liu, Y. et al. Epithelial EZH2 serves as an epigenetic determinant in experimental colitis by inhibiting TNFα-mediated inflammation and apoptosis. Proc. Natl Acad. Sci. USA 114, E3796–E3805 (2017).

    Article  CAS  Google Scholar 

  22. McCabe, M. T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108–112 (2012).

    Article  CAS  Google Scholar 

  23. Howard, J. C., Hunn, J. P. & Steinfeldt, T. The IRG protein-based resistance mechanism in mice and its relation to virulence in Toxoplasma gondii. Curr. Opin. Microbiol. 14, 414–421 (2011).

    Article  CAS  Google Scholar 

  24. Virreira Winter, S. et al. Determinants of GBP recruitment to Toxoplasma gondii vacuoles and the parasitic factors that control it. PLoS ONE 6, e24434 (2011).

    Article  Google Scholar 

  25. Robben, P. M., LaRegina, M., Kuziel, W. A. & Sibley, L. D. Recruitment of Gr-1+ monocytes is essential for control of acute toxoplasmosis. J. Exp. Med. 201, 1761–1769 (2005).

    Article  CAS  Google Scholar 

  26. Dunay, I. R. et al. Gr1(+) inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity 29, 306–317 (2008).

    Article  CAS  Google Scholar 

  27. Mordue, D. G., Monroy, F., La Regina, M., Dinarello, C. A. & Sibley, L. D. Acute toxoplasmosis leads to lethal overproduction of Th1 cytokines. J. Immunol. 167, 4574–4584 (2001).

    Article  CAS  Google Scholar 

  28. Chang, H. R., Grau, G. E. & Pechère, J. C. Role of TNF and IL-1 in infections with Toxoplasma gondii. Immunology 69, 33–37 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hunter, C. A., Chizzonite, R. & Remington, J. S. IL-1 beta is required for IL-12 to induce production of IFN-gamma by NK cells. A role for IL-1 beta in the T cell-independent mechanism of resistance against intracellular pathogens. J. Immunol. 155, 4347–4354 (1995).

    CAS  PubMed  Google Scholar 

  30. Rosowski, E. E. et al. Strain-specific activation of the NF-κB pathway by GRA15, a novel Toxoplasma gondii dense granule protein. J. Exp. Med. 208, 195–212 (2011).

    Article  CAS  Google Scholar 

  31. Gorfu, G. et al. Dual role for inflammasome sensors NLRP1 and NLRP3 in murine resistance to Toxoplasma gondii. mBio 5, e01117-13 (2014).

  32. Mason, N. J., Liou, H.-C. & Hunter, C. A. T cell-intrinsic expression of c-Rel regulates Th1 cell responses essential for resistance to Toxoplasma gondii. J. Immunol. 172, 3704–3711 (2004).

    Article  CAS  Google Scholar 

  33. Bhatt, D. & Ghosh, S. Regulation of the NF-κB-mediated transcription of inflammatory genes. Front. Immunol. 5, 71 (2014).

    Article  Google Scholar 

  34. Lima, T. S., Gov, L. & Lodoen, M. B. Evasion of human neutrophil-mediated host defense during Toxoplasma gondii infection. mBio 9, e02027-17 (2018).

    Article  Google Scholar 

  35. Berger, S. L., Kouzarides, T., Shiekhattar, R. & Shilatifard, A. An operational definition of epigenetics. Genes Dev. 23, 781–783 (2009).

    Article  CAS  Google Scholar 

  36. Lee, B. K., Bhinge, A. A. & Iyer, V. R. Wide-ranging functions of E2F4 in transcriptional activation and repression revealed by genome-wide analysis. Nucleic Acids Res. 39, 3558–3573 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Laboratoire d’Excellence (LabEx) ParaFrap (ANR-11-LABX-0024), the Agence Nationale pour la Recherche (Project HostQuest, ANR-18-CE15-0023) and the European Research Council (ERC Consolidator grant no. 614880 Hosting TOXO to M.-A.H.). The proteomic experiments were partly supported by the Agence Nationale pour la Recherche (Investissement d’Avenir Infrastructures, ProFi project ANR-10-INBS-08-01).

Author information

Authors and Affiliations

Authors

Contributions

M.-A.H., L.B. and A.B. conceived the project. L.B., M.-P.B.-P., P.-M.H., D.C., S.K.-J., J.V., V.J., B.T., Y.C., I.T. and A.B. designed, performed and interpreted the experimental work. M.-A.H. supervised the research. M.-A.H. wrote the paper with editorial support from I.T., L.B. and A.B.

Corresponding authors

Correspondence to Alexandre Bougdour or Mohamed-Ali Hakimi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figures 1–10 and Supplementary Table legends.

Reporting Summary

Supplementary Table 1

Differentially expressed genes between human cells infected with Pruku80 and Pruku80teegr.

Supplementary Table 2

Differentially expressed genes between murine BMDMs infected with Pruku80 and Pruku80teegr.

Supplementary Table 3

Strains, vectors and primers.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Braun, L., Brenier-Pinchart, MP., Hammoudi, PM. et al. The Toxoplasma effector TEEGR promotes parasite persistence by modulating NF-κB signalling via EZH2. Nat Microbiol 4, 1208–1220 (2019). https://doi.org/10.1038/s41564-019-0431-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0431-8

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