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:

AIG1 and ADTRP are atypical integral membrane hydrolases that degrade bioactive FAHFAs

Abstract

Enzyme classes may contain outlier members that share mechanistic, but not sequence or structural, relatedness with more common representatives. The functional annotation of such exceptional proteins can be challenging. Here, we use activity-based profiling to discover that the poorly characterized multipass transmembrane proteins AIG1 and ADTRP are atypical hydrolytic enzymes that depend on conserved threonine and histidine residues for catalysis. Both AIG1 and ADTRP hydrolyze bioactive fatty acid esters of hydroxy fatty acids (FAHFAs) but not other major classes of lipids. We identify multiple cell-active, covalent inhibitors of AIG1 and show that these agents block FAHFA hydrolysis in mammalian cells. These results indicate that AIG1 and ADTRP are founding members of an evolutionarily conserved class of transmembrane threonine hydrolases involved in bioactive lipid metabolism. More generally, our findings demonstrate how chemical proteomics can excavate potential cases of convergent or parallel protein evolution that defy conventional sequence- and structure-based predictions.

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: Discovery and characterization of AIG1 and ADTRP as FP-reactive proteins in the human proteome.
Figure 2: Identification of Thr and His residues critical for FP labeling of AIG1 and ADTRP.
Figure 3: AIG1 and ADTRP are FAHFA hydrolases.
Figure 4: Discovery of inhibitors and structurally related inactive control compounds for AIG1.
Figure 5: AIG1 inhibitors block FAHFA hydrolysis in mammalian cells.

Similar content being viewed by others

References

  1. Willems, L.I., Overkleeft, H.S. & van Kasteren, S.I. Current developments in activity-based protein profiling. Bioconjug. Chem. 25, 1181–1191 (2014).

    Article  CAS  Google Scholar 

  2. Niphakis, M.J. & Cravatt, B.F. Enzyme inhibitor discovery by activity-based protein profiling. Annu. Rev. Biochem. 83, 341–377 (2014).

    Article  CAS  Google Scholar 

  3. Berger, A.B., Vitorino, P.M. & Bogyo, M. Activity-based protein profiling: applications to biomarker discovery, in vivo imaging and drug discovery. Am. J. Pharmacogenomics 4, 371–381 (2004).

    Article  CAS  Google Scholar 

  4. Liu, Y., Patricelli, M.P. & Cravatt, B.F. Activity-based protein profiling: the serine hydrolases. Proc. Natl. Acad. Sci. USA 96, 14694–14699 (1999).

    Article  CAS  Google Scholar 

  5. Simon, G.M. & Cravatt, B.F. Activity-based proteomics of enzyme superfamilies: serine hydrolases as a case study. J. Biol. Chem. 285, 11051–11055 (2010).

    Article  CAS  Google Scholar 

  6. Bachovchin, D.A. et al. Superfamily-wide portrait of serine hydrolase inhibition achieved by library-versus-library screening. Proc. Natl. Acad. Sci. USA 107, 20941–20946 (2010).

    Article  CAS  Google Scholar 

  7. Jessani, N. et al. A streamlined platform for high-content functional proteomics of primary human specimens. Nat. Methods 2, 691–697 (2005).

    Article  CAS  Google Scholar 

  8. Higa, H.H., Diaz, S. & Varki, A. Biochemical and genetic evidence for distinct membrane-bound and cytosolic sialic acid O-acetyl-esterases: serine-active-site enzymes. Biochem. Biophys. Res. Commun. 144, 1099–1108 (1987).

    Article  CAS  Google Scholar 

  9. Jessani, N. et al. Class assignment of sequence-unrelated members of enzyme superfamilies by activity-based protein profiling. Angew. Chem. Int. Edn. Engl. 44, 2400–2403 (2005).

    Article  CAS  Google Scholar 

  10. Long, J.Z. & Cravatt, B.F. The metabolic serine hydrolases and their functions in mammalian physiology and disease. Chem. Rev. 111, 6022–6063 (2011).

    Article  CAS  Google Scholar 

  11. Elias, M. & Tawfik, D.S. Divergence and convergence in enzyme evolution: parallel evolution of paraoxonases from quorum-quenching lactonases. J. Biol. Chem. 287, 11–20 (2012).

    Article  CAS  Google Scholar 

  12. Lone, A.M. et al. A substrate-free activity-based protein profiling screen for the discovery of selective PREPL inhibitors. J. Am. Chem. Soc. 133, 11665–11674 (2011).

    Article  CAS  Google Scholar 

  13. Kidd, D., Liu, Y. & Cravatt, B.F. Profiling serine hydrolase activities in complex proteomes. Biochemistry 40, 4005–4015 (2001).

    Article  CAS  Google Scholar 

  14. Adibekian, A. et al. Click-generated triazole ureas as ultrapotent in vivo–active serine hydrolase inhibitors. Nat. Chem. Biol. 7, 469–478 (2011).

    Article  CAS  Google Scholar 

  15. Seo, J., Kim, J. & Kim, M. Cloning of androgen-inducible gene 1 (AIG1) from human dermal papilla cells. Mol. Cells 11, 35–40 (2001).

    CAS  PubMed  Google Scholar 

  16. Wu, G., Sun, M., Zhang, W. & Huo, K. AIG1 is a novel Pirh2-interacting protein that activates the NFAT signaling pathway. Front. Biosci. 3, 834–842 (2011).

    Google Scholar 

  17. Lupu, C., Zhu, H., Popescu, N.I., Wren, J.D. & Lupu, F. Novel protein ADTRP regulates TFPI expression and function in human endothelial cells in normal conditions and in response to androgen. Blood 118, 4463–4471 (2011).

    Article  CAS  Google Scholar 

  18. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  CAS  Google Scholar 

  19. Dodson, G. & Wlodawer, A. Catalytic triads and their relatives. Trends Biochem. Sci. 23, 347–352 (1998).

    Article  CAS  Google Scholar 

  20. Ekici, O.D., Paetzel, M. & Dalbey, R.E. Unconventional serine proteases: variations on the catalytic Ser/His/Asp triad configuration. Protein Sci. 17, 2023–2037 (2008).

    Article  CAS  Google Scholar 

  21. Yore, M.M. et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell 159, 318–332 (2014).

    Article  CAS  Google Scholar 

  22. Cognetta, A.B. III et al. Selective N-hydroxyhydantoin carbamate inhibitors of mammalian serine hydrolases. Chem. Biol. 22, 928–937 (2015).

    Article  CAS  Google Scholar 

  23. Kamat, S.S. et al. Immunomodulatory lysophosphatidylserines are regulated by ABHD16A and ABHD12 interplay. Nat. Chem. Biol. 11, 164–171 (2015).

    Article  CAS  Google Scholar 

  24. Hoover, H.S., Blankman, J.L., Niessen, S. & Cravatt, B.F. Selectivity of inhibitors of endocannabinoid biosynthesis evaluated by activity-based protein profiling. Bioorg. Med. Chem. Lett. 18, 5838–5841 (2008).

    Article  CAS  Google Scholar 

  25. Nomura, D.K. et al. Monoacylglycerol lipase exerts dual control over endocannabinoid and fatty acid pathways to support prostate cancer. Chem. Biol. 18, 846–856 (2011).

    Article  CAS  Google Scholar 

  26. Brown, M.S., Ye, J., Rawson, R.B. & Goldstein, J.L. Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell 100, 391–398 (2000).

    Article  CAS  Google Scholar 

  27. Wolfe, M.S. Intramembrane-cleaving proteases. J. Biol. Chem. 284, 13969–13973 (2009).

    Article  CAS  Google Scholar 

  28. Urban, S., Lee, J.R. & Freeman, M. Drosophila rhomboid-1 defines a family of putative intramembrane serine proteases. Cell 107, 173–182 (2001).

    Article  CAS  Google Scholar 

  29. Strisovsky, K. Why cells need intramembrane proteases—a mechanistic perspective. FEBS J. 10.1111/febs.13638 (2015).

  30. Sherratt, A.R., Blais, D.R., Ghasriani, H., Pezacki, J.P. & Goto, N.K. Activity-based protein profiling of the Escherichia coli GlpG rhomboid protein delineates the catalytic core. Biochemistry 51, 7794–7803 (2012).

    Article  CAS  Google Scholar 

  31. Vosyka, O. et al. Activity-based probes for rhomboid proteases discovered in a mass spectrometry-based assay. Proc. Natl. Acad. Sci. USA 110, 2472–2477 (2013).

    Article  CAS  Google Scholar 

  32. Nguyen, M.T., Van Kersavond, T. & Verhelst, S.H. Chemical tools for the study of intramembrane proteases. ACS Chem. Biol. 10, 2423–2434 (2015).

    Article  CAS  Google Scholar 

  33. Rath, A., Glibowicka, M., Nadeau, V.G., Chen, G. & Deber, C.M. Detergent binding explains anomalous SDS-PAGE migration of membrane proteins. Proc. Natl. Acad. Sci. USA 106, 1760–1765 (2009).

    Article  CAS  Google Scholar 

  34. Gerlt, J.A. et al. The enzyme function initiative. Biochemistry 50, 9950–9962 (2011).

    Article  CAS  Google Scholar 

  35. Adibekian, A. et al. Confirming target engagement for reversible inhibitors in vivo by kinetically tuned activity-based probes. J. Am. Chem. Soc. 134, 10345–10348 (2012).

    Article  CAS  Google Scholar 

  36. Söding, J., Biegert, A. & Lupas, A.N. The HHpred interactive server for protein homology detection and structure prediction. Nucleic Acids Res. 33, W244–W248 (2005).

    Article  Google Scholar 

  37. Patricelli, M.P., Giang, D.K., Stamp, L.M. & Burbaum, J.J. Direct visualization of serine hydrolase activities in complex proteomes using fluorescent active site-directed probes. Proteomics 1, 1067–1071 (2001).

    Article  CAS  Google Scholar 

  38. Tully, S.E. & Cravatt, B.F. Activity-based probes that target functional subclasses of phospholipases in proteomes. J. Am. Chem. Soc. 132, 3264–3265 (2010).

    Article  CAS  Google Scholar 

  39. Hsu, K.L. et al. DAGLβ inhibition perturbs a lipid network involved in macrophage inflammatory responses. Nat. Chem. Biol. 8, 999–1007 (2012).

    Article  CAS  Google Scholar 

  40. Washburn, M.P., Wolters, D. & Yates, J.R. III. Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat. Biotechnol. 19, 242–247 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to K. Masuda and L. Bar-Peled for discussions on and assistance with cloning; M. Dix, J. Wu and K. Lum for discussions and technical expertise in designing and analyzing proteomics experiments; J. Teijaro for providing T cells; G. Simon for assistance with the HHpred analysis; and C. Walsh and M. Niphakis for numerous helpful discussions. This work was supported by the US National Institutes of Health (DA033760, DK909810), The Leona M. and Harry B. Helmsley Charitable Trust (grant no. 2012-PG-MED002 to A.S.), National Cancer Institute Cancer Center Support grant P30 (CA014195 MASS core, A.S.), Dr. Frederick Paulsen Chair/Ferring Pharmaceuticals (A.S.), a Hewitt Foundation for Medical Research Fellowship (to W.H.P.), a Chapman Charitable Trust Fellowship (to M.J.K.), UCSD Medical Scientist Training Program funding (T32 GM007198 to M.J.K.) and an Irving S. Sigal postdoctoral fellowship from the American Chemical Society (to S.S.K.).

Author information

Authors and Affiliations

Authors

Contributions

W.H.P., M.J.K., A.S. and B.F.C. conceived the project. W.H.P., M.J.K., S.S.K., A.S. and B.F.C. designed experiments. W.H.P. performed the molecular biology and proteomics experiments. W.H.P., A.B.C., J.J.H. and A.S. synthesized compounds. M.J.K., S.S.K. and W.H.P. performed substrate assays and biochemical experiments. W.H.P., M.J.K., S.S.K., E.S., B.B.K., A.S. and B.F.C. analyzed and interpreted the data. W.H.P. and B.F.C. wrote the paper. M.J.K., S.S.K. and A.S. edited the paper.

Corresponding authors

Correspondence to Alan Saghatelian or Benjamin F Cravatt.

Ethics declarations

Competing interests

B.F.C. is a founder and advisor to Abide Therapeutics, a biotechnology company interested in developing serine hydrolase inhibitors and therapeutics.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–18, Supplementary Table 1 and 2 captions, and Supplementary Tables 3 and 4. (PDF 3492 kb)

Supplementary Note

Synthetic Procedures. (PDF 270 kb)

Supplementary Table 1

Complete ABPP-SILAC data sets. (XLSX 5746 kb)

Supplementary Table 2

HHpred search results for AIG1/ADTRP alignment. (XLSX 20 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Parsons, W., Kolar, M., Kamat, S. et al. AIG1 and ADTRP are atypical integral membrane hydrolases that degrade bioactive FAHFAs. Nat Chem Biol 12, 367–372 (2016). https://doi.org/10.1038/nchembio.2051

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2051

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research