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 Ca2+ sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum

Abstract

Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the type I interferon response to DNA pathogens. Aberrant activation of STING is linked to the pathology of autoimmune and autoinflammatory diseases. The rate-limiting step for the activation of STING is its translocation from the ER to the ER-Golgi intermediate compartment. Here, we found that deficiency in the Ca2+ sensor stromal interaction molecule 1 (STIM1) caused spontaneous activation of STING and enhanced expression of type I interferons under resting conditions in mice and a patient with combined immunodeficiency. Mechanistically, STIM1 associated with STING to retain it in the ER membrane, and coexpression of full-length STIM1 or a STING-interacting fragment of STIM1 suppressed the function of dominant STING mutants that cause autoinflammatory diseases. Furthermore, deficiency in STIM1 strongly enhanced the expression of type I interferons after viral infection and prevented the lethality of infection with a DNA virus in vivo. This work delineates a STIM1-STING circuit that maintains the resting state of the STING pathway.

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: STIM1 deficiency spontaneously induces type I IFN response in murine and human cells.
Fig. 2: STING-TBK1-IRF3 pathway links loss of STIM1 expression to Ifnb1 transcription.
Fig. 3: STIM1 deficiency causes enhanced type I IFN response in patient cells.
Fig. 4: STIM1 interacts with STING for its retention in the ER.
Fig. 5: STIM1 inhibits STING trafficking to the ERGIC.
Fig. 6: Ablation of STIM1 enhances host defense towards DNA viruses and HIV by priming type I IFN responses.
Fig. 7: STIM1 deficiency enhances host defense against HSV-1 infection in vivo.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon request. The manuscript describing the clinical phenotype of the STIM1 patient is available from OSR Preprints (https://doi.org/10.31219/osf.io/4duxt).

References

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. 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  CAS  Google Scholar 

  4. Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15, 760–770 (2015).

    Article  CAS  Google Scholar 

  5. Chen, Q., Sun, L. & Chen, Z. J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 17, 1142–1149 (2016).

    Article  CAS  Google Scholar 

  6. Li, T. & Chen, Z. J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 17, 1142–1149 (2018).

    Google Scholar 

  7. Li, Y., Wilson, H. L. & Kiss-Toth, E. Regulating STING in health and disease. J. Inflamm. (Lond.) 14, 11 (2017).

    Article  Google Scholar 

  8. Yan, N., Regalado-Magdos, A. D., Stiggelbout, B., Lee-Kirsch, M. A. & Lieberman, J. The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1. Nat. Immunol. 11, 1005–1013 (2010).

    Article  CAS  Google Scholar 

  9. Ahn, J., Gutman, D., Saijo, S. & Barber, G. N. STING manifests self DNA-dependent inflammatory disease. Proc. Natl Acad. Sci. USA 109, 19386–19391 (2012).

    Article  CAS  Google Scholar 

  10. Crow, Y. J. & Manel, N. Aicardi–Goutieres syndrome and the type I interferonopathies. Nat. Rev. Immunol. 15, 429–440 (2015).

    Article  CAS  Google Scholar 

  11. Liu, Y. et al. Activated STING in a vascular and pulmonary syndrome. N. Engl. J. Med. 371, 507–518 (2014).

    Article  CAS  Google Scholar 

  12. Melki, I. et al. Disease-associated mutations identify a novel region in human STING necessary for the control of type I interferon signaling. J. Allergy Clin. Immunol. 140, 543–552.e545 (2017).

    Article  CAS  Google Scholar 

  13. Jeremiah, N. et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. J. Clin. Invest. 124, 5516–5520 (2014).

    Article  Google Scholar 

  14. 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 

  15. 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  Google Scholar 

  16. Prakriya, M. & Lewis, R. S. Store-operated calcium channels. Physiol. Rev. 95, 1383–1436 (2015).

    Article  CAS  Google Scholar 

  17. Feske, S., Skolnik, E. Y. & Prakriya, M. Ion channels and transporters in lymphocyte function and immunity. Nat. Rev. Immunol. 12, 532–547 (2012).

    Article  CAS  Google Scholar 

  18. Baba, Y. & Kurosaki, T. Role of calcium signaling in B cell activation and biology. Curr. Top. Microbiol. Immunol. 393, 143–174 (2016).

    CAS  PubMed  Google Scholar 

  19. Srikanth, S., Woo, J. S., Sun, Z. & Gwack, Y. Immunological disorders: regulation of Ca2+ signaling in T lymphocytes. Adv. Exp. Med. Biol. 993, 397–424 (2017).

  20. Lacruz, R. S. & Feske, S. Diseases caused by mutations in ORAI1 and STIM1. Ann. N. Y. Acad. Sci. 1356, 45–79 (2015).

    Article  CAS  Google Scholar 

  21. Notarangelo, L. D., Kim, M. S., Walter, J. E. & Lee, Y. N. Human RAG mutations: biochemistry and clinical implications. Nat. Rev. Immunol. 16, 234–246 (2016).

    Article  CAS  Google Scholar 

  22. Brandman, O., Liou, J., Park, W. S. & Meyer, T. STIM2 is a feedback regulator that stabilizes basal cytosolic and endoplasmic reticulum Ca2+ levels. Cell 131, 1327–1339 (2007).

    Article  CAS  Google Scholar 

  23. Soboloff, J., Rothberg, B. S., Madesh, M. & Gill, D. L. STIM proteins: dynamic calcium signal transducers. Nat. Rev. Mol. Cell Biol. 13, 549–565 (2012).

    Article  CAS  Google Scholar 

  24. Picard, C. et al. STIM1 mutation associated with a syndrome of immunodeficiency and autoimmunity. N. Engl. J. Med. 360, 1971–1980 (2009).

    Article  CAS  Google Scholar 

  25. Rice, A. et al. A report of novel STIM1 deficiency and 6 year follow up of two previous cases associated with mild immunological phenotype. Preprint at https://doi.org/10.31219/osf.io/4duxt (2018).

  26. Guo, H. et al. NLRX1 sequesters STING to negatively regulate the interferon response, thereby facilitating the replication of HIV-1 and DNA viruses. Cell Host Microbe 19, 515–528 (2016).

    Article  CAS  Google Scholar 

  27. 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  CAS  Google Scholar 

  28. Lau, L., Gray, E. E., Brunette, R. L. & Stetson, D. B. DNA tumor virus oncogenes antagonize the cGAS-STING DNA-sensing pathway. Science 350, 568–571 (2015).

    Article  CAS  Google Scholar 

  29. Yuan, J. P. et al. SOAR and the polybasic STIM1 domains gate and regulate Orai channels. Nat. Cell Biol. 11, 337–343 (2009).

    Article  CAS  Google Scholar 

  30. Ishikawa, H., Ma, Z. & Barber, G. N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 461, 788–792 (2009).

    Article  CAS  Google Scholar 

  31. Mukai, K. et al. Activation of STING requires palmitoylation at the Golgi. Nat. Commun. 7, 11932 (2016).

    Article  CAS  Google Scholar 

  32. Ouyang, S. et al. Structural analysis of the STING adaptor protein reveals a hydrophobic dimer interface and mode of cyclic di-GMP binding. Immunity 36, 1073–1086 (2012).

    Article  CAS  Google Scholar 

  33. Gao, D. et al. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903–906 (2013).

    Article  CAS  Google Scholar 

  34. Cheshenko, N. et al. Herpes simplex virus triggers activation of calcium-signaling pathways. J. Cell Biol. 163, 283–293 (2003).

    Article  CAS  Google Scholar 

  35. 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 

  36. Wang, Y. et al. TRIM30alpha 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 

  37. Srikanth, S. et al. A novel EF-hand protein, CRACR2A, is a cytosolic Ca2+ sensor that stabilizes CRAC channels in T cells. Nat. Cell Biol. 12, 436–446 (2010).

    Article  CAS  Google Scholar 

  38. Gwack, Y. et al. Hair loss and defective T- and B-cell function in mice lacking ORAI1. Mol. Cell. Biol. 28, 5209–5222 (2008).

    Article  CAS  Google Scholar 

  39. Stetson, D. B. & Medzhitov, R. Recognition of cytosolic DNA activates an IRF3-dependent innate immune response. Immunity 24, 93–103 (2006).

    Article  CAS  Google Scholar 

  40. Srikanth, S., Jung, H. J., Ribalet, B. & Gwack, Y. The intracellular loop of Orai1 plays a central role in fast inactivation of Ca2+ release-activated Ca2+ channels. J. Biol. Chem. 285, 5066–5075 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Bensinger (UCLA) for sharing THP1 cells, and X. Liu and S. Smale (UCLA) for BMDM differentiation protocols and reagents. We thank N.-H. Park, K.-H. Shin, M.K. Kang, R. Kim, Y. Wang, and T.M. Vondriska for sharing their confocal imaging facilities. We thank J.-L. Casanova (Rockefeller University) for providing STIM1 patient B cell line. This work was supported by the National Institutes of Health grants AI083432 and DE028432 (Y.G.) and AI130653 (S. Srikanth). This work was also supported in part by CA180779, CA200422, AI073099, AI116585, AI129496, AI140705, DE023926, DE027888, the Fletcher Jones Foundation, and the Whittier Foundation (J.U.J.).

Author information

Authors and Affiliations

Authors

Contributions

Y.G. and S.S. designed the research. S.S. performed all of the in vitro experiments using MEFs, THP1, and BMDMs with technical help from J.L. J.S.W. performed biochemical experiments of interaction between STIM1 and STING, SAVI mutant analyses, and in vivo HSV infection experiments with help from B.W. Y.M.E.-S., L.R., and S. Savic collected and analyzed patient samples together with S.S. and J.S.W. K.C. and D.S.A. performed the HIV infection experiments. G.J.S. and J.U.J. provided reagents and protocols for in vitro HSV infections. G.C. helped with statistical analysis. C.R. and E.C. provided reagents and protocols for in vivo HSV infections. T.T.W. and R.S. provided reagents and protocols for MHV-68 infections. S.S. and Y.G. wrote the manuscript with input from all authors and supervised the project.

Corresponding authors

Correspondence to Sonal Srikanth or Yousang Gwack.

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.

Integrated supplementary information

Supplementary Figure 1 Sequencing data for STIM1−/− THP1 clones generated using CRISPR–Cas9 system.

Top: alignment of sequences within exons 2 and 3 of human STIM1 gene with the ones from individual clones (sg#2 and sg#3). The sgRNA sequences are highlighted in red. Below: sequencing results of individual clones. Boxed areas mark the indel sites. Clone sg#2 shows insertion of three nucleotides and deletion of two nucleotides, whereas sg#3 shows deletion of five nucleotides

Supplementary Figure 2 Lack of spontaneous induction of the type I IFN response in STIM2−/− THP1 cells.

Left, immunoblot showing expression of STIM2 in control or STIM2−/− THP1 cells generated using two independent sgRNAs (sg#1 and sg#2). Loss of STIM2 does not alter expression of STIM1 or STING in these cells. Right: qPCR analysis of IFNB1 transcripts in unstimulated control and STIM2−/− THP1 cells. Data show representative triplicates from two independent experiments

Supplementary Figure 3 STIM1 deficiency induces dimerization of STING.

a, Immunoblots from two representative experiments (Exp. no. 1 and no. 2) showing endogenous STING in control, Stim1−/−, and Orai1−/− MEFs under non-reducing SDS–PAGE conditions. *The position of monomeric and dimeric STING. b, Lysates from untreated or DSP-treated (0.125, 0.25, 0.5, 1.0, and 2.0 mM, 1 h on ice) control or STIM1−/− HEK293T cells expressing STING-FLAG were immunoblotted for detection of STING under non-reducing conditions (left). Bar graph shows averaged ratio of STING (dimer/monomer) from three independent experiments (right). *P <0.05 (unpaired/two-tailed t-test)

Supplementary Figure 4 Overexpression or deletion of STING affects SOCE and STIM1 translocation kinetics.

a, Representative traces of averaged SOCE from HEK293T cells expressing empty vector (24 cells) or that encoding STING (27 cells) after passive depletion of intracellular Ca2+ stores with 1 μM TG in the presence of external solution containing 2 mM Ca2+, as indicated. b, Representative traces of averaged SOCE from Jurkat T cells expressing empty vector (54 cells) or that encoding STING (49 cells). c, Representative traces of averaged SOCE from Jurkat T cells expressing empty vector (49 cells) or that encoding STING (53 cells) after stimulation with 10 µg ml−1 α-CD3 antibody or 0.5 µM ionomycin in the presence of external solution containing 2 mM Ca2+, as indicated. d, Representative epifluorescence (left) and TIRF (right) images of WT or Sting−/− MEFs stably expressing STIM1-YFP under resting conditions or 10 min after depletion of intracellular Ca2+ stores using 1 μM TG. Scale bar, 10 µm. Line graph on the right shows mean normalized fluorescence intensity ± s.e.m. of STIM1-YFP in WT (6 cells) and Tmem173−/− MEFs (9 cells) from 2 independent experiments. e, Left, immunoblot showing expression of indicated proteins in control and TMEM173−/− Jurkat T cells. Right panels show representative traces of averaged SOCE from control (54 cells) and TMEM173−/− (58 cells) Jurkat T cells. f, Representative traces of averaged SOCE from control (66 cells) and TMEM173−/− (60 cells) Jurkat T cells after stimulation with anti-CD3 antibody (2 μg ml−1) in the presence of external solution containing 2 mM Ca2+, as indicated. Bar graphs in panels ac, e, and f show averaged baseline subtracted peak SOCE (±s.e.m.) from three independent experiments. *P <0.05, **P <0.005, ***P <0.0005 (unpaired/two-tailed t-test)

Supplementary Figure 5 Functional interaction of constitutively active STING mutants with STIM1.

a, FLAG immunoprecipitates from lysates of HEK293T cells stably expressing FLAG-tagged WT or indicated mutants of STING were immunoblotted for detection of endogenous STIM1 proteins (left). Bar graph (right) shows densitometry analysis of normalized (relative to WT STING) fold change in STIM1 band intensity from seven independent experiments. b, Reporter assay for Ifnb1 promoter activity in HEK293T cells transfected with WT or mutant STING proteins and full-length STIM1 or its N-terminal fragment (a.a. 1–249) for 24 h (right). Data show representative triplicates from two independent experiments. c, Representative confocal microscopy images of MEFs stably expressing WT STING-GFP or indicated SAVI mutants without or with STIM1. Cells were either stained for endogenous p58 (ERGIC marker, left three panels) or STIM1 (+STIM1, right three panels). Scale bar, 10 µm. Images are representative of 25–30 cells for each condition. *P <0.05, **P <0.005, and ***P <0.0005 (unpaired/two-tailed t-test, a; one-way ANOVA, b)

Supplementary Figure 6 STIM1 plays a Ca2+-independent role in the antiviral immune response.

a, qPCR analysis of GFP (left) and Ifnb1 (right) transcripts in WT, Orai1−/−, or Stim1−/− MEFs uninfected or HSV-1-GFP-infected with indicated MOI. b, qPCR analysis of GFP (left) transcripts in HSV-GFP-infected (MOI 0.5, 24 h) WT or Stim1−/− MEFs in the presence or absence of indicated amounts of tofacitinib. Cells were preincubated with the inhibitor for 30 min before infection. Data show representative triplicates from two independent experiments. Data are shown as mean ± s.e.m. *P <0.05, **P <0.005, and ***P <0.0005 (two-way ANOVA, a; unpaired/two-tailed t-test, b)

Supplementary Figure 7 Generation of mice with conditionally targeted Orai1 deficiency.

a, Schematic showing the architecture of mouse Orai1 gene. Exon 2 of the Orai1 gene was targeted. By homologous recombination, Orai1+/fl mice with loxP sites (red arrowheads) flanking exon 2 were generated. Germ-line transmission of flox alleles was validated by genotyping PCR (lower panel). The location of the primer pair is indicated as green (5′ and 3′) arrows. A 378-base pair (bp) band is derived from the flox allele while a 200-bp band denotes the WT allele. b, qPCR analysis of Orai1 transcripts in control (Orai1fl/fl) or Orai1−/− BMDMs 7 d after differentiation. Data show representative triplicates (mean ± s.e.m., unpaired/two-tailed t-test) from two independent experiments c, Representative traces of averaged SOCE from Orai1fl/fl (36 cells) and Orai1−/− (41 cells) BMDMs 7 d after differentiation and after passive depletion of intracellular Ca2+ stores with 1 μM TG in the presence of external solution containing 2 mM Ca2+, as indicated. Data are representative of three independent experiments

Supplementary information

Supplementary Figures 1–7

Supplementary Figures 1–7, Supplementary Table 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Srikanth, S., Woo, J.S., Wu, B. et al. The Ca2+ sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum. Nat Immunol 20, 152–162 (2019). https://doi.org/10.1038/s41590-018-0287-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0287-8

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