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:

Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs

An Erratum to this article was published on 18 August 2015

A Corrigendum to this article was published on 17 February 2015

This article has been updated

Abstract

Agonists of mouse STING (TMEM173) shrink and even cure solid tumors by activating innate immunity; human STING (hSTING) agonists are needed to test this therapeutic hypothesis in humans. The endogenous STING agonist is 2′3′-cGAMP, a second messenger that signals the presence of cytosolic double-stranded DNA. We report activity-guided partial purification and identification of ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP1) to be the dominant 2′3′-cGAMP hydrolyzing activity in cultured cells. The hydrolysis activity of ENPP1 was confirmed using recombinant protein and was depleted in tissue extracts and plasma from Enpp1−/− mice. We synthesized a hydrolysis-resistant bisphosphothioate analog of 2′3′-cGAMP (2′3′-cGsAsMP) that has similar affinity for hSTING in vitro and is ten times more potent at inducing IFN-β secretion from human THP1 monocytes. Studies in mouse Enpp1−/− lung fibroblasts indicate that resistance to hydrolysis contributes substantially to its higher potency. 2′3′-cGsAsMP is therefore improved over natural 2′3′-cGAMP as a model agonist and has potential as a vaccine adjuvant and cancer therapeutic.

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: There is 2′-5′ phosphodiesterase activity in the plasma membrane or heavy organelles and it is not due to PDE12.
Figure 2: ENPP1 is an efficient hydrolase for 2′3′-cGAMP.
Figure 3: ENPP1 is the dominant hydrolase activity for 2′3′-cGAMP.
Figure 4: Development of hydrolysis-resistant hSTING agonists.
Figure 5: ENPP1 dampens 2′3′-cGAMP signaling.

Similar content being viewed by others

Change history

  • 29 January 2015

    In the version of this article initially published, the chemical structures of the two cyclic dinucleotides in Figure 4a were incorrect at the triphosphate group, with each of the phosphates shown as containing two extra hydrogen atoms. The error has been corrected in the HTML and PDF versions of the article.

  • 15 July 2015

    In the version of this article published on 29 January 2015, in Figure 4 the hydroxyl group was depicted in the 3′ position on the deoxyribose ring of the cyclic dinucleotide precursor to 3′3′-cGAMP rather than in the 2′ position and the R group was in the 2′ position rather than in the 3′ position. This error has been corrected in the HTML and PDF versions of the article.

References

  1. Duthie, M.S., Windish, H.P., Fox, C.B. & Reed, S.G. Use of defined TLR ligands as adjuvants within human vaccines. Immunol. Rev. 239, 178–196 (2011).

    Article  CAS  Google Scholar 

  2. Coffman, R.L., Sher, A. & Seder, R.A. Vaccine adjuvants: putting innate immunity to work. Immunity 33, 492–503 (2010).

    Article  CAS  Google Scholar 

  3. Hemmi, H. et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat. Immunol. 3, 196–200 (2002).

    Article  CAS  Google Scholar 

  4. 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 

  5. 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 

  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  CAS  Google Scholar 

  7. Kim, S. et al. Anticancer flavonoids are mouse-selective STING agonists. ACS Chem. Biol. 8, 1396–1401 (2013).

    Article  CAS  Google Scholar 

  8. 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  Google Scholar 

  9. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  12. Davies, B.W., Bogard, R.W., Young, T.S. & Mekalanos, J.J. Coordinated regulation of accessory genetic elements produces cyclic di-nucleotides for V. cholerae virulence. Cell 149, 358–370 (2012).

    Article  CAS  Google Scholar 

  13. 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 

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

    Article  CAS  Google Scholar 

  15. Diner, E.J. et al. The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell Rep. 3, 1355–1361 (2013).

    Article  CAS  Google Scholar 

  16. Li, X.D. et al. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341, 1390–1394 (2013).

    Article  CAS  Google Scholar 

  17. Bender, A.T. & Beavo, J.A. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol. Rev. 58, 488–520 (2006).

    Article  CAS  Google Scholar 

  18. Maurice, D.H. et al. Advances in targeting cyclic nucleotide phosphodiesterases. Nat. Rev. Drug Discov. 13, 290–314 (2014).

    Article  CAS  Google Scholar 

  19. Wielinga, P.R. et al. Characterization of the MRP4- and MRP5-mediated transport of cyclic nucleotides from intact cells. J. Biol. Chem. 278, 17664–17671 (2003).

    Article  CAS  Google Scholar 

  20. Jedlitschky, G., Burchell, B. & Keppler, D. The multidrug resistance protein 5 functions as an ATP-dependent export pump for cyclic nucleotides. J. Biol. Chem. 275, 30069–30074 (2000).

    Article  CAS  Google Scholar 

  21. Guo, Y. et al. MRP8, ATP-binding cassette C11 (ABCC11), is a cyclic nucleotide efflux pump and a resistance factor for fluoropyrimidines 2′,3′-dideoxycytidine and 9′-(2′-phosphonylmethoxyethyl)adenine. J. Biol. Chem. 278, 29509–29514 (2003).

    Article  CAS  Google Scholar 

  22. 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  Google Scholar 

  23. 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  CAS  Google Scholar 

  24. Kubota, K. et al. Identification of 2′-phosphodiesterase, which plays a role in the 2–5A system regulated by interferon. J. Biol. Chem. 279, 37832–37841 (2004).

    Article  CAS  Google Scholar 

  25. Poulsen, J.B. et al. Human 2′-phosphodiesterase localizes to the mitochondrial matrix with a putative function in mitochondrial RNA turnover. Nucleic Acids Res. 39, 3754–3770 (2011).

    Article  CAS  Google Scholar 

  26. Goding, J.W. et al. Ecto-phosphodiesterase/pyrophosphatase of lymphocytes and non-lymphoid cells: structure and function of the PC-1 family. Immunol. Rev. 161, 11–26 (1998).

    Article  CAS  Google Scholar 

  27. Bollen, M., Gijsbers, R., Ceulemans, H., Stalmans, W. & Stefan, C. Nucleotide pyrophosphatases/phosphodiesterases on the move. Crit. Rev. Biochem. Mol. Biol. 35, 393–432 (2000).

    Article  CAS  Google Scholar 

  28. Bischoff, E., Tran-Thi, T.A. & Decker, K.F. Nucleotide pyrophosphatase of rat liver. A comparative study on the enzymes solubilized and purified from plasma membrane and endoplasmic reticulum. Eur. J. Biochem. 51, 353–361 (1975).

    Article  CAS  Google Scholar 

  29. Kato, K. et al. Crystal structure of Enpp1, an extracellular glycoprotein involved in bone mineralization and insulin signaling. Proc. Natl. Acad. Sci. USA 109, 16876–16881 (2012).

    Article  CAS  Google Scholar 

  30. Belli, S.I., van Driel, I.R. & Goding, J.W. Identification and characterization of a soluble form of the plasma cell membrane glycoprotein PC-1 (5′-nucleotide phosphodiesterase). Eur. J. Biochem. 217, 421–428 (1993).

    Article  CAS  Google Scholar 

  31. Rutsch, F. et al. PC-1 nucleoside triphosphate pyrophosphohydrolase deficiency in idiopathic infantile arterial calcification. Am. J. Pathol. 158, 543–554 (2001).

    Article  CAS  Google Scholar 

  32. Hessle, L. et al. Tissue-nonspecific alkaline phosphatase and plasma cell membrane glycoprotein-1 are central antagonistic regulators of bone mineralization. Proc. Natl. Acad. Sci. USA 99, 9445–9449 (2002).

    Article  CAS  Google Scholar 

  33. Lau, W.M. et al. Enpp1: A potential facilitator of breast cancer bone metastasis. PLoS ONE 8, e66752 (2013).

    Article  CAS  Google Scholar 

  34. Umar, A. et al. Identification of a putative protein profile associated with tamoxifen therapy resistance in breast cancer. Mol. Cell. Proteomics 8, 1278–1294 (2009).

    Article  CAS  Google Scholar 

  35. Meyre, D. et al. Variants of ENPP1 are associated with childhood and adult obesity and increase the risk of glucose intolerance and type 2 diabetes. Nat. Genet. 37, 863–867 (2005).

    Article  CAS  Google Scholar 

  36. Rey, D. et al. Amerindians show no association of PC-1 gene Gln121 allele and obesity: a thrifty gene population genetics. Mol. Biol. Rep. 39, 7687–7693 (2012).

    Article  CAS  Google Scholar 

  37. Maddux, B.A. et al. Membrane glycoprotein PC-1 and insulin resistance in non-insulin-dependent diabetes mellitus. Nature 373, 448–451 (1995).

    Article  CAS  Google Scholar 

  38. Chin, C.N. et al. Evidence that inhibition of insulin receptor signaling activity by PC-1/ENPP1 is dependent on its enzyme activity. Eur. J. Pharmacol. 606, 17–24 (2009).

    Article  CAS  Google Scholar 

  39. Konno, H., Konno, K. & Barber, G.N. Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. Cell 155, 688–698 (2013).

    Article  CAS  Google Scholar 

  40. Liang, Q. et al. Crosstalk between the cGAS DNA sensor and Beclin-1 autophagy protein shapes innate antimicrobial immune responses. Cell Host Microbe 15, 228–238 (2014).

    Article  CAS  Google Scholar 

  41. Ablasser, A. et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013).

    Article  CAS  Google Scholar 

  42. Sali,, A.F.J.M., Terkeltaub, R. & Goding, J.W. in Ecto-ATPases and Related Ectonucleotidases (eds. Vanduffel, L. & Lemmens, R.) 267–282 (Shaker Publishing BV, Maastricht, the Netherlands, 1999).

  43. Narisawa, S. et al. Novel inhibitors of alkaline phosphatase suppress vascular smooth muscle cell calcification. J. Bone Miner. Res. 22, 1700–1710 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Koch, P. Choi, K. Krukenberg, A. Groen, M. Loose, S. Kim and S. Gruver for helpful discussions. We thank R. Jiang for help with data analysis. We thank K. Chu for providing mouse livers. We thank S. Walker for sharing the MicroBeta plate reader and C. Fan for technical assistance. We thank J.J. Mekalanos for providing the DncV expression plasmid and T. Bernhard (both from Harvard Medical School) for providing the pTB146 plasmid. We thank G. Heffron and C. Sheahan for assistance with the NMR data collection and analysis. We thank R. Ward for help with manuscript preparation. This research was supported by the National Cancer Institute (CA139980, AR53102, AI050872 and 1K99AI108793-01). L. Li thanks the Jane Coffin Childs Fund for her postdoctoral fellowship.

Author information

Authors and Affiliations

Authors

Contributions

L.L. and T.J.M. developed the hypothesis and designed the study. L.L., Q.Y., P.K. and Z.M. conducted the experiments. All authors interpreted and discussed the results. J.L.M. advised P.K., and H.W. advised Q.Y. Both J.L.M. and H.W. funded part of the research. L.L. and T.J.M. wrote the manuscript.

Corresponding author

Correspondence to Lingyin Li.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results and Supplementary Figures 1–15. (PDF 29368 kb)

Supplementary Data Set 1

Mass spectrometry analysis of fraction 26. (XLSX 77 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Yin, Q., Kuss, P. et al. Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat Chem Biol 10, 1043–1048 (2014). https://doi.org/10.1038/nchembio.1661

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1661

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer