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:

Homeostatic regulation of STING protein at the resting state by stabilizer TOLLIP

Abstract

STING (stimulator of interferon genes) is an important innate immune protein, but its homeostatic regulation at the resting state is unknown. Here, we identified TOLLIP as a stabilizer of STING through direct interaction to prevent its degradation. Tollip deficiency results in reduced STING protein in nonhematopoietic cells and tissues, and renders STING protein unstable in immune cells, leading to severely dampened STING signaling capacity. The competing degradation mechanism of resting-state STING requires IRE1α and lysosomes. TOLLIP mediates clearance of Huntington’s disease-linked polyQ protein aggregates. Ectopically expressed polyQ proteins in vitro or endogenous polyQ proteins in Huntington’s disease mouse striatum sequester TOLLIP away from STING, leading to reduced STING protein and dampened immune signaling. Tollip–/– also ameliorates STING-mediated autoimmune disease in Trex1–/– mice. Together, our findings reveal that resting-state STING protein level is strictly regulated by a constant tug-of-war between ‘stabilizer’ TOLLIP and ‘degrader’ IRE1α-lysosome that together maintain tissue immune homeostasis.

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: Tollip deficiency impairs STING-mediated IFN signaling.
Fig. 2: TOLLIP prevents lysosomal-mediated degradation of STING at the resting state.
Fig. 3: TOLLIP interacts with STING.
Fig. 4: PolyQ protein disrupts TOLLIP–STING interaction and results in STING protein turnover in vitro and in vivo.
Fig. 5: TOLLIP stabilizes STING protein in cells.
Fig. 6: Tollip deficiency selectively activates IRE1α, which facilitates resting-state STING protein turnover.
Fig. 7: Tollip–/– ameliorates autoinflammatory disease of Trex1–/– mice.
Fig. 8: STING protein level is reduced in Tollip–/– mouse heart tissue.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Tan, X., Sun, L., Chen, J. & Chen, Z. J. Detection of microbial infections through innate immune sensing of nucleic acids. Annu. Rev. Microbiol. 72, 447–478 (2018).

    Article  CAS  Google Scholar 

  2. Dobbs, N. et al. STING activation by translocation from the ER is associated with infection and autoinflammatory disease. Cell Host Microbe 18, 157–168 (2015).

    Article  CAS  Google Scholar 

  3. Yan, N. Immune diseases associated with TREX1 and STING dysfunction. J. Interferon Cytokine Res. 37, 198–206 (2017).

    Article  CAS  Google Scholar 

  4. Gao, D. et al. Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. Proc. Natl Acad. Sci. USA 112, E5699–E5705 (2015).

    Article  CAS  Google Scholar 

  5. Gray, E. E., Treuting, P. M., Woodward, J. J. & Stetson, D. B. Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi–Goutières syndrome. J. Immunol. 195, 1939–1943 (2015).

    Article  CAS  Google Scholar 

  6. Gall, A. et al. Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity 36, 120–131 (2012).

    Article  CAS  Google Scholar 

  7. Morita, M. et al. Gene-targeted mice lacking the Trex1 (DNase III) 3′→5′ DNA exonuclease develop inflammatory myocarditis. Mol. Cell Biol. 24, 6719–6727 (2004).

    Article  CAS  Google Scholar 

  8. Luo, W.-W. et al. iRhom2 is essential for innate immunity to DNA viruses by mediating trafficking and stability of the adaptor STING. Nat. Immunol. 17, 1057–1066 (2016).

    Article  CAS  Google Scholar 

  9. Zhang, M. et al. USP18 recruits USP20 to promote innate antiviral response through deubiquitinating STING/MITA. Cell Res. 26, 1302–1319 (2016).

    Article  CAS  Google Scholar 

  10. Zhong, B. et al. The ubiquitin ligase RNF5 regulates antiviral responses by mediating degradation of the adaptor protein MITA. Immunity 30, 397–407 (2009).

    Article  CAS  Google Scholar 

  11. Xing, J. et al. TRIM29 promotes DNA virus infections by inhibiting innate immune response. Nat. Commun. 8, 945 (2017).

    Article  Google Scholar 

  12. Wang, Y. et al. TRIM30α is a negative-feedback regulator of the intracellular DNA and DNA virus-triggered response by targeting STING. PLoS Pathog. 11, e1005012 (2015).

    Article  Google Scholar 

  13. Burns, K. et al. Tollip, a new component of the IL-1RI pathway, links IRAK to the IL-1 receptor. Nat. Cell Biol. 2, 346–351 (2000).

    Article  CAS  Google Scholar 

  14. Gonugunta, V. K. et al. Trafficking-mediated STING degradation requires sorting to acidified endolysosomes and can be targeted to enhance anti-tumor response. Cell Rep. 21, 3234–3242 (2017).

    Article  CAS  Google Scholar 

  15. Warner, J. D. et al. STING-associated vasculopathy develops independently of IRF3 in mice. J. Exp. Med. 214, 3279–3292 (2017).

    Article  CAS  Google Scholar 

  16. Yang, K., Huang, R., Fujihira, H., Suzuki, T. & Yan, N. N-glycanase NGLY1 regulates mitochondrial homeostasis and inflammation through NRF1. J. Exp. Med. 215, 2600–2616 (2018).

    Article  CAS  Google Scholar 

  17. Lu, K., Psakhye, I. & Jentsch, S. Autophagic clearance of polyQ proteins mediated by ubiquitin-Atg8 adaptors of the conserved CUET protein family. Cell 158, 549–563 (2014).

    Article  CAS  Google Scholar 

  18. Menalled, L. B., Sison, J. D., Dragatsis, I., Zeitlin, S. & Chesselet, M.-F. Time course of early motor and neuropathological anomalies in a knock-in mouse model of Huntington’s disease with 140 CAG repeats. J. Comp. Neurol. 465, 11–26 (2003).

    Article  CAS  Google Scholar 

  19. Wu, J. et al. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. J. Exp. Med. 216, 867–883 (2019).

    Article  CAS  Google Scholar 

  20. Fink, S. L. et al. IRE1α promotes viral infection by conferring resistance to apoptosis. Sci. Signal 10, eaai7814 (2017).

    Article  Google Scholar 

  21. Hur, K. Y. et al. IRE1α activation protects mice against acetaminophen-induced hepatotoxicity. J. Exp. Med. 209, 307–318 (2012).

    Article  CAS  Google Scholar 

  22. Aden, K. et al. ATG16L1 orchestrates interleukin-22 signaling in the intestinal epithelium via cGAS-STING. J. Exp. Med. 215, 2868–2886 (2018).

    Article  CAS  Google Scholar 

  23. Duennwald, M. L. & Lindquist, S. Impaired ERAD and ER stress are early and specific events in polyglutamine toxicity. Genes Dev. 22, 3308–3319 (2008).

    Article  CAS  Google Scholar 

  24. Gui, X. et al. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 567, 262–266 (2019).

    Article  CAS  Google Scholar 

  25. Sliter, D. A. et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018).

    Article  CAS  Google Scholar 

  26. Gulen, M. F. et al. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 8, 427 (2017).

    Article  Google Scholar 

  27. Larkin, B. et al. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J. Immunol. 199, 397–402 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Maillard (Centre hospitalier universitaire vaudois) for Tollip–/– mice; M. Lehrman (University of Texas Southwestern Medical Center) for Ern1+/+, Ern1–/–, Xbp1+/+ and Xbp1–/– MEFs; U. Deshmukh (Oklahoma Medical Research Foundation) for the initial IHC protocol; and members of the Yan laboratory for helpful discussion. This work was supported by the National Institutes of Health (grant nos. AR067135 and AI134877 to N.Y.; grant nos. NS0870778 and NS052325 to R.G.K.), the Cancer Prevention and Research Institute of Texas (CPRIT, RP180288 to N.Y.) and the Burroughs Wellcome Fund (N.Y.).

Author information

Authors and Affiliations

Authors

Contributions

V.P. initiated the project and performed most of the experiments. K.Y. and J.W. helped with some experiments. X.T. and N.D. helped with mouse IHC staining. R.G.K. performed the zQ175 mouse experiment. N.Y. and V.P. wrote the paper with input from all co-authors.

Corresponding author

Correspondence to Nan Yan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pokatayev, V., Yang, K., Tu, X. et al. Homeostatic regulation of STING protein at the resting state by stabilizer TOLLIP. Nat Immunol 21, 158–167 (2020). https://doi.org/10.1038/s41590-019-0569-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0569-9

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