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:

Activation of STING by targeting a pocket in the transmembrane domain

Abstract

Stimulator of interferon genes (STING) is an adaptor protein in innate immunity against DNA viruses or bacteria1,2,3,4,5. STING-mediated immunity could be exploited in the development of vaccines or cancer immunotherapies. STING is a transmembrane dimeric protein that is located in the endoplasmic reticulum or in the Golgi apparatus. STING is activated by the binding of its cytoplasmic ligand-binding domain to cyclic dinucleotides that are produced by the DNA sensor cyclic GMP-AMP (cGAMP) synthase or by invading bacteria1,6,7. Cyclic dinucleotides induce a conformational change in the STING ligand-binding domain, which leads to a high-order oligomerization of STING that is essential for triggering the downstream signalling pathways8,9. However, the cGAMP-induced STING oligomers tend to dissociate in solution and have not been resolved to high resolution, which limits our understanding of the activation mechanism. Here we show that a small-molecule agonist, compound 53 (C53)10, promotes the oligomerization and activation of human STING through a mechanism orthogonal to that of cGAMP. We determined a cryo-electron microscopy structure of STING bound to both C53 and cGAMP, revealing a stable oligomer that is formed by side-by-side packing and has a curled overall shape. Notably, C53 binds to a cryptic pocket in the STING transmembrane domain, between the two subunits of the STING dimer. This binding triggers outward shifts of transmembrane helices in the dimer, and induces inter-dimer interactions between these helices to mediate the formation of the high-order oligomer. Our functional analyses show that cGAMP and C53 together induce stronger activation of STING than either ligand alone.

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: Structure of the high-order oligomer of human STING bound to both cGAMP and C53.
Fig. 2: Interaction between STING and C53.
Fig. 3: Mutational analyses of the C53-binding site in human STING.
Fig. 4: TMD-mediated interactions contribute to STING oligomerization.

Similar content being viewed by others

Data availability

The atomic coordinates and the cryo-EM map of STING bound to both cGAMP and C53 have been deposited into the RCSB PDB (PDB ID: 7SII) and the Electron Microscopy Data Bank (EMD) (EMDB ID: EMD-25142). Source data are provided with this paper.

References

  1. Zhang, X., Bai, X. C. & Chen, Z. J. Structures and mechanisms in the cGAS–STING innate immunity pathway. Immunity 53, 43–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Ishikawa, H. & Barber, G. N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 455, 674–678 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhong, B. et al. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity 29, 538–550 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Jin, L. et al. MPYS, a novel membrane tetraspanner, is associated with major histocompatibility complex class II and mediates transduction of apoptotic signals. Mol. Cell. Biol. 28, 5014–5026 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sun, W. et al. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proc. Natl Acad. Sci. USA 106, 8653–8658 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wu, J. et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Shang, G., Zhang, C., Chen, Z. J., Bai, X. C. & Zhang, X. Cryo-EM structures of STING reveal its mechanism of activation by cyclic GMP-AMP. Nature 567, 389–393 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhang, C. et al. Structural basis of STING binding with and phosphorylation by TBK1. Nature 567, 394–398 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pryde, D. C. et al. The discovery of potent small molecule activators of human STING. Eur. J. Med. Chem. 209, 112869 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Burdette, D. L. et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature 478, 515–518 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dou, Z. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gluck, S. et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 19, 1061–1070 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mackenzie, K. J. et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yang, H., Wang, H., Ren, J., Chen, Q. & Chen, Z. J. cGAS is essential for cellular senescence. Proc. Natl Acad. Sci. USA 114, E4612–E4620 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Motedayen Aval, L., Pease, J. E., Sharma, R. & Pinato, D. J. Challenges and opportunities in the clinical development of STING agonists for cancer immunotherapy. J. Clin. Med. 9, 3323 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  17. Zhao, B. et al. A conserved PLPLRT/SD motif of STING mediates the recruitment and activation of TBK1. Nature 569, 718–722 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tanaka, Y. & Chen, Z. J. STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway. Sci. Signal. 5, ra20 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Liu, S. et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 347, aaa2630 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Ergun, S. L., Fernandez, D., Weiss, T. M. & Li, L. STING polymer structure reveals mechanisms for activation, hyperactivation, and inhibition. Cell 178, 290–301 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. 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  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  22. Saitoh, T. et al. Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response. Proc. Natl Acad. Sci. USA 106, 20842–20846 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Zhang, X. et al. Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol. Cell 51, 226–235 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Kranzusch, P. J. et al. Ancient origin of cGAS-STING reveals mechanism of universal 2',3' cGAMP signaling. Mol. Cell 59, 891–903 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shi, H., Wu, J., Chen, Z. J. & Chen, C. Molecular basis for the specific recognition of the metazoan cyclic GMP-AMP by the innate immune adaptor protein STING. Proc. Natl Acad. Sci. USA 112, 8947–8952 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao, P. et al. Structure–function analysis of STING activation by c[G(2',5')pA(3',5')p] and targeting by antiviral DMXAA. Cell 154, 748–762 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ernst, A. M. et al. S-palmitoylation sorts membrane cargo for anterograde transport in the Golgi. Dev. Cell 47, 479–493 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cong, X. et al. Crystal structures of porcine STING(CBD)–CDN complexes reveal the mechanism of ligand recognition and discrimination of STING proteins. J. Biol. Chem. 294, 11420–11432 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Conlon, J. et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J. Immunol. 190, 5216–5225 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Morales-Perez, C. L., Noviello, C. M. & Hibbs, R. E. Manipulation of subunit stoichiometry in heteromeric membrane proteins. Structure 24, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang, K. Gctf: real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bepler, T. et al. Positive-unlabeled convolutional neural networks for particle picking in cryo-electron micrographs. Nat. Methods 16, 1153–1160 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gao, P. et al. Cyclic [G(2',5')pA(3',5')p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153, 1094–1107 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ablasser, A. et al. cGAS produces a 2'-5'-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Robert, X. & Gouet, P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 42, W320–W324 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Laskowski, R. A. & Swindells, M. B. LigPlot+: multiple ligand-protein interaction diagrams for drug discovery. J. Chem. Inf. Model. 51, 2778–2786 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Woodward, J. J., Iavarone, A. T. & Portnoy, D. A. c-di-AMP secreted by intracellular Listeria monocytogenes activates a host type I interferon response. Science 328, 1703–1705 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Cryo-EM data were collected at the University of Texas Southwestern Medical Center (UTSW) Cryo-Electron Microscopy Facility, funded in part by the Cancer Prevention and Research Institute of Texas (CPRIT) Core Facility Support Award RP170644. We thank D. Stoddard and J. Martinez Diaz for facility access. We thank the Structural Biology Laboratory at UTSW for equipment use. This work is supported in part by grants from the National Institutes of Health (R35GM130289 to X.Z. and R01GM143158 to X.-c.B.), the Welch foundation (I-1702 to X.Z. and I-1944 to X.-c.B.) and CPRIT (RP160082 to X.-c.B.). X.-c.B. and X.Z. are Virginia Murchison Linthicum Scholars in Medical Research at UTSW.

Author information

Authors and Affiliations

Authors

Contributions

X.-c.B. and X.Z. conceived the project. D.L., G.S. and J.L. prepared the cryo-EM samples. X.-c.B. and X.Z. collected the cryo-EM data and solved the structure. Y.L. synthesized C53. D.L. did the biochemical and functional assays. All of the authors contributed to drafting the manuscript.

Corresponding authors

Correspondence to Xiao-chen Bai or Xuewu Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Osamu Nureki, Hao Wu and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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 Purification of human STING and its interaction with C53 and cGAMP.

a, Gel filtration profile of human STING on a Superdex S200 10/300 column. b, SDS–PAGE analyses of fractions indicated by the bracket in a. c, Analyses of STING oligomerization by native gel. C53 and cGAMP together induced robust high-order oligomerization of purified STING solubilized in detergent solution, whereas either one alone failed to do so under the same condition. The results shown are representative of three biological repeats.

Extended Data Fig. 2 Image processing procedure of the human STING tetramer bound to both cGAMP and C53.

a, Motion corrected micrograph. Red arrows highlight high-order oligomers of STING. The curved overall shape of the oligomers is evident from these examples. b, 2D class averages of high-order oligomers of human STING. Large oligomers were segmented into particles containing four dimers at the maximum. c, Final 3D reconstruction of the tetramer coloured based on local resolution. d, Gold-standard FSC curve of the final 3D reconstruction. e, FSC between the final map and the atomic model. f, Image processing procedure.

Extended Data Fig. 3 Sample density maps of various parts of the structure.

Various parts of the structure with the cryo-EM density overlayed are shown. Most of the protein sidechains are clearly visible. The high-quality of the densities for cGAMP and C53 allows unambiguous docking of the compounds.

Extended Data Fig. 4 C53-induced dilation of the binding pocket in the STING TMD.

a, Comparison of the TMD of human STING in the C53-bound and the apo states. It is evident that the TMD pocket in apo-STING is much smaller and cannot accommodate C53, and binding of C53 induces dilation of this pocket. b, Sequence alignment of the TMD of STING from human (h), mouse (m) and chicken (ch). Black circles highlight residues in the C53-binding pocket. Stars highlight residues in the TMD–TMD interface that contribute to the oligomerization of STING.

Extended Data Fig. 5 Effects of mutations in the C53-binding site on STING oligomerization in cells.

a, c, Representative images of cells expressing STING wild type or the mutants in the binding pocket and the TM3-TM4 loop, respectively. Hela cells were transfected with GFP-tagged human STING wild type or mutants. Cells were stimulated with cGAMP, C53 or both. Localization of STING-GFP in cells was monitored by the fluorescence signal of GFP. Nuclei were stained with DAPI. The experiments were repeated three times. The images are representative images from these experiments. As expected, STING showed a diffuse pattern in cells in the absence of an agonist. Both C53 and cGAMP induced puncta formation of STING wild type. C53-induced puncta formation was reduced or abolished by the mutations. b, d, Quantification of STING puncta formation in cells expressing the wild type or mutants. The bar graph shows the individual data points, mean and s.e.m. of the percentage of cells with STING forming large puncta from the three biological repeats. Statistical significance p-values between the wild type and mutants were calculated by two-tailed Welch’s t-test. Scale bar, 10 µm. Source data of puncta counting results are provided in the source data file.

Source data

Extended Data Fig. 6 Further characterization of the activation of STING in cells by C53 and cGAMP.

a, b, Activation of STING in cells by C53 and cGAMP at a series of concentrations. Notably, although cGAMP at 10 nM and C53 at 100 nM individually induced minimal phosphorylation of STING, the combination of the two led to robust activation of STING. c, d, Activation of STING by cGAMP or C53 with or without digitonin-mediated cell permeabilization. It is clear from these results that cGAMP-mediated activation of STING is dependent on digitonin-mediated permeabilization, whereas C53 can enter the cell and activate STING on its own. The experiments were carried out in a similar manner as in Fig. 3 and Fig. 4, and the results shown are representative of three biological repeats. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 7 Additional mutational analyses of the activation of STING by C53 and cGAMP.

a, C53 could induce phosphorylation of the R238A/Y240A mutant of STING in cells, which has its cGAMP-pocket disrupted and could not be activated by cGAMP. b, M120L, a mutation in the C53-binding pocket, reduced STING phosphorylation when cells were stimulated with cGAMP at a lower concentration (100 nM). c, L30A, a mutation in TMD–TMD oligomerization interface, reduced STING phosphorylation when cells were stimulated with cGAMP at a lower concentration (100 nM). These effects of M120L and L30A were suppressed when cells were treated with cGAMP at 1,000 nM. These results suggest that both the TMD pocket and the TMD–TMD interface are important for cGAMP-mediated activation of STING, although high concentrations of cGAMP can overcome the detrimental effects of some mutations in these regions. The experiments were carried out in a similar manner as in Figs. 3, 4, and the results shown are representative of three biological repeats. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 8 Effects of mutations in the TMD–TMD interface on STING oligomerization in cells.

a, Representative images of cells expressing STING wild type or the mutants in the TMD–TMD interface. Hela cells were transfected with GFP-tagged human STING wild type or mutants. Cells were stimulated with cGAMP, C53 or both. Localization of STING-GFP in cells was monitored by the fluorescence signal of GFP. Nuclei were stained with DAPI. The experiments were repeated three times. The images are representative images from these experiments. As expected, STING showed a diffuse pattern in cells in the absence of an agonist. Both C53 and cGAMP induced puncta formation of STING wild type, which was reduced or abolished by the mutants. b, Quantification of STING puncta formation in cells expressing the wild type or mutants. The bar graph shows the individual data points, mean and s.e.m. of the percentage of cells with STING forming large puncta from the three biological repeats. Statistical significance p-values between the wild type and mutants were calculated by two-tailed Welch’s t-test. Scale bar, 10 µm. Source data of puncta counting results are provided in the source data file.

Source data

Extended Data Table 1 Data collection and model refinement statistics

Supplementary information

Supplementary Information

The supplementary material contains two parts. The first part is the detailed description of the synthesis process of C53. The second part is Supplementary Figure 1, showing the uncropped western blots.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, D., Shang, G., Li, J. et al. Activation of STING by targeting a pocket in the transmembrane domain. Nature 604, 557–562 (2022). https://doi.org/10.1038/s41586-022-04559-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04559-7

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