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:

Bidirectional modulation of HIF-2 activity through chemical ligands

Abstract

Hypoxia-inducible factor-2 (HIF-2) is a heterodimeric transcription factor formed through dimerization between an oxygen-sensitive HIF-2α subunit and its obligate partner subunit ARNT. Enhanced HIF-2 activity drives some cancers, whereas reduced activity causes anemia in chronic kidney disease. Therefore, modulation of HIF-2 activity via direct-binding ligands could provide many new therapeutic benefits. Here, we explored HIF-2α chemical ligands using combined crystallographic, biophysical, and cell-based functional studies. We found chemically unrelated antagonists to employ the same mechanism of action. Their binding displaced residue M252 from inside the HIF-2α PAS-B pocket toward the ARNT subunit to weaken heterodimerization. We also identified first-in-class HIF-2α agonists and found that they significantly displaced pocket residue Y281. Its dramatic side chain movement increases heterodimerization stability and transcriptional activity. Our findings show that despite binding to the same HIF-2α PAS-B pocket, ligands can manifest as inhibitors versus activators by mobilizing different pocket residues to allosterically alter HIF-2α–ARNT heterodimerization.

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: Inhibition of HIF-2 by PT2385.
Fig. 2: Allosteric mechanism of PT2385 revealed by the crystal structure and H/D-ex MS.
Fig. 3: PT2385 disrupts subunit–subunit interactions in HIF-2 heterodimers.
Fig. 4: Newly identified antagonist T1001 points to a common inhibitory mechanism.
Fig. 5: The allosteric mechanism of agonist M1001.
Fig. 6: The agonistic effects of M1002.

Similar content being viewed by others

Data availability

The three sets of structural data from HIF-2α–ARNT crystals in complex with PT2385, T1001 and M1001 are available from the PDB under accession codes 6E3S, 6E3T and 6E3U, respectively. Other data shown in the article are available from the correspond authors upon reasonable request.

References

  1. Wu, D. & Rastinejad, F. Structural characterization of mammalian bHLH-PAS transcription factors. Curr. Opin. Struct. Biol. 43, 1–9 (2017).

    Article  Google Scholar 

  2. Möglich, A., Ayers, R. A. & Moffat, K. Structure and signaling mechanism of Per-ARNT-Sim domains. Structure 17, 1282–1294 (2009).

    Article  Google Scholar 

  3. Wu, D., Su, X., Potluri, N., Kim, Y. & Rastinejad, F. NPAS1-ARNT and NPAS3-ARNT crystal structures implicate the bHLH-PAS family as multi-ligand binding transcription factors. eLife 5, e18790 (2016).

    Article  Google Scholar 

  4. McIntosh, B. E., Hogenesch, J. B. & Bradfield, C. A. Mammalian Per-Arnt-Sim proteins in environmental adaptation. Annu. Rev. Physiol. 72, 625–645 (2010).

    Article  CAS  Google Scholar 

  5. Schito, L. & Semenza, G. L. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer 2, 758–770 (2016).

    Article  Google Scholar 

  6. Keith, B., Johnson, R. S. & Simon, M. C. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 12, 9–22 (2011).

    Article  Google Scholar 

  7. Ravenna, L., Salvatori, L. & Russo, M. A. HIF3α: the little we know. FEBS. J. 283, 993–1003 (2016).

    Article  CAS  Google Scholar 

  8. Wu, D., Potluri, N., Lu, J., Kim, Y. & Rastinejad, F. Structural integration in hypoxia-inducible factors. Nature 524, 303–308 (2015).

    Article  CAS  Google Scholar 

  9. Ivan, M. et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292, 464–468 (2001).

    Article  CAS  Google Scholar 

  10. Jaakkola, P. et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292, 468–472 (2001).

    Article  CAS  Google Scholar 

  11. Yu, F., WhiteS. B., Zhao, Q. & Lee, F. S. HIF-1α binding to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc. Natl Acad. Sci. USA 98, 9630–9635 (2001).

    Article  CAS  Google Scholar 

  12. Lando, D., Peet, D. J., Whelan, D. A., Gorman, J. J. & Whitelaw, M. L. Asparagine hydroxylation of the HIF transactivation domain a hypoxic switch. Science 295, 858–861 (2002).

    Article  CAS  Google Scholar 

  13. Lando, D. et al. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. 16, 1466–1471 (2002).

    Article  CAS  Google Scholar 

  14. Huang, P., Chandra, V. & Rastinejad, F. Structural overview of the nuclear receptor superfamily: insights into physiology and therapeutics. Annu. Rev. Physiol. 72, 247–272 (2010).

    Article  CAS  Google Scholar 

  15. Denison, M. S. & Nagy, S. R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 43, 309–334 (2003).

    Article  CAS  Google Scholar 

  16. Denison, M. S., Soshilov, A. A., He, G., DeGroot, D. E. & Zhao, B. Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol. Sci. 124, 1–22 (2011).

    Article  CAS  Google Scholar 

  17. Scheuermann, T. H. et al. Artificial ligand binding within the HIF2α PAS-B domain of the HIF2 transcription factor. Proc. Natl Acad. Sci. USA 106, 450–455 (2009).

    Article  CAS  Google Scholar 

  18. Key, J., Scheuermann, T. H., Anderson, P. C., Daggett, V. & Gardner, K. H. Principles of ligand binding within a completely buried cavity in HIF2αa PAS-B. J. Am. Chem. Soc. 131, 17647–17654 (2009).

    Article  CAS  Google Scholar 

  19. Cardoso, R. et al. Identification of Cys255 in HIF-1α as a novel site for development of covalent inhibitors of HIF-1α/ARNT PasB domain protein-protein interaction. Protein Sci. 21, 1885–1896 (2012).

    Article  CAS  Google Scholar 

  20. Guo, Y. et al. Regulating the ARNT/TACC3 axis: multiple approaches to manipulating protein/protein interactions with small molecules. ACS. Chem. Biol. 8, 626–635 (2013).

    Article  CAS  Google Scholar 

  21. Fala, A. M. et al. Unsaturated fatty acids as high-affinity ligands of the C-terminal Per-ARNT-Sim domain from the hypoxia-inducible factor 3α. Sci. Rep. 5, 12698 (2015).

    Article  CAS  Google Scholar 

  22. Hewitson, K. S. & Schofield, C. J. The HIF pathway as a therapeutic target. Drug Discov. Today 9, 704–711 (2004).

    Article  CAS  Google Scholar 

  23. Wallace, E. M. et al. A small-molecule antagonist of HIF2α is efficacious in preclinical models of renal cell carcinoma. Cancer Res. 76, 5491–5500 (2016).

    Article  CAS  Google Scholar 

  24. Chen, W. et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature 539, 112–117 (2016).

    Article  CAS  Google Scholar 

  25. Cho, H. et al. On-target efficacy of a HIF-2α antagonist in preclinical kidney cancer models. Nature 539, 107–111 (2016).

    Article  CAS  Google Scholar 

  26. Maxwell, P. H. & Eckardt, K. U. HIF prolyl hydroxylase inhibitors for the treatment of renal anaemia and beyond. Nat. Rev. Nephrol. 12, 157–168 (2016).

    Article  CAS  Google Scholar 

  27. Yousaf, F. & Spinowitz, B. Hypoxia-inducible factor stabilizers: a new avenue for reducing BP while helping hemoglobin? Curr. Hypertens. Rep. 18, 23 (2016).

    Article  Google Scholar 

  28. Gupta, N. & Wish, J. B. Hypoxia-inducible factor prolyl hydroxylase inhibitors: a potential new treatment for anemia in patients With CKD. Am. J. Kidney. Dis. 69, 815–826 (2017).

    Article  CAS  Google Scholar 

  29. Yeh, T. L. et al. Molecular and cellular mechanisms of HIF prolyl hydroxylase inhibitors in clinical trials. Chem. Sci. 8, 7651–7668 (2017).

    Article  CAS  Google Scholar 

  30. Seidel, S. A. et al. Microscale thermophoresis quantifies biomolecular interactions under previously challenging conditions. Methods 59, 301–315 (2013).

    Article  CAS  Google Scholar 

  31. Ciulli, A. & Abell, C. Fragment-based approaches to enzyme inhibition. Curr. Opin. Biotechnol. 18, 489–496 (2007).

    Article  CAS  Google Scholar 

  32. Scheuermann, T. H. et al. Allosteric inhibition of hypoxia inducible factor-2 with small molecules. Nat. Chem. Biol. 9, 271–276 (2013).

    Article  CAS  Google Scholar 

  33. Forbes, S. A. et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 43, D805–D811 (2015).

    Article  CAS  Google Scholar 

  34. Annis, D. A., Nickbarg, E., Yang, X., Ziebell, M. R. & Whitehurst, C. E. Affinity selection-mass spectrometry screening techniques for small molecule drug discovery. Curr. Opin. Chem. Biol. 11, 518–526 (2007).

    Article  CAS  Google Scholar 

  35. Bonomini, M., Del Vecchio, L., Sirolli, V. & Locatelli, F. New treatment approaches for the anemia of CKD. Am. J. Kidney. Dis. 67, 133–142 (2016).

    Article  Google Scholar 

  36. Besarab, A. et al. Roxadustat (FG-4592): correction of anemia in incident dialysis patients. J. Am. Soc. Nephrol. 27, 1225–1233 (2016).

    Article  CAS  Google Scholar 

  37. Brigandi, R. A. et al. A novel hypoxia-inducible factor-prolyl hydroxylase inhibitor (GSK1278863) for anemia in CKD: a 28-day, phase 2A randomized trial. Am. J. Kidney. Dis. 67, 861–871 (2016).

    Article  CAS  Google Scholar 

  38. Pergola, P. E., Spinowitz, B. S., Hartman, C. S., Maroni, B. J. & Haase, V. H. Vadadustat, a novel oral HIF stabilizer, provides effective anemia treatment in nondialysis-dependent chronic kidney disease. Kidney Int. 90, 1115–1122 (2016).

    Article  CAS  Google Scholar 

  39. Beck, H. et al. Discovery of molidustat (BAY 85-3934): a small-molecule oral HIF-prolyl hydroxylase (HIF-PH) inhibitor for the treatment of renal anemia. ChemMedChem 13, 988–1003 (2018).

    Article  CAS  Google Scholar 

  40. Rogers, J. L. et al. Development of inhibitors of the PAS-B domain of the HIF-2α transcription factor. J. Med. Chem. 56, 1739–1747 (2013).

    Article  CAS  Google Scholar 

  41. Scheuermann, T. H. et al. Isoform-selective and stereoselective inhibition of hypoxia inducible factor-2. J. Med. Chem. 58, 5930–5941 (2015).

    Article  CAS  Google Scholar 

  42. Wehn, P. M. et al. Design and activity of specific hypoxia-inducible factor-2α (HIF-2α) inhibitors for the treatment of clear cell renal cell carcinoma: discovery of clinical candidate (S)-3-((2,2-difluoro-1-hydroxy-7-(methylsulfonyl)-2,3-dihydro-1H-inden-4-yl)oxy)-5-fluorobenzonitrile (PT2385). J. Med. Chem. 61, 9691–9721 (2018).

    Article  CAS  Google Scholar 

  43. Minor, W., Cymborowski, M., Otwinowski, Z. & Chruszcz, M. HKL-3000: the integration of data reduction and structure solution--from diffraction images to an initial model in minutes. Acta Crystallogr. D. Biol. Crystallogr. 62, 859–866 (2006).

    Article  Google Scholar 

  44. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Google Scholar 

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

    Article  CAS  Google Scholar 

  48. Marsh, J. J. et al. Structural insights into fibrinogen dynamics using amide hydrogen/deuterium exchange mass spectrometry. Biochemistry 52, 5491–5502 (2013).

    Article  CAS  Google Scholar 

  49. Woods, V. L. Jr. & Hamuro, Y. High resolution, high-throughput amide deuterium exchange-mass spectrometry (DXMS) determination of protein binding site structure and dynamics: utility in pharmaceutical design. J. Cell. Biochem. Suppl. 84, 89–98 (2001).

    Article  Google Scholar 

  50. Walters, B. T., Ricciuti, A., Mayne, L. & Englander, S. W. Minimizing back exchange in the hydrogen exchange-mass spectrometry experiment. J. Am. Soc. Mass. Spectrom. 23, 2132–2139 (2012).

    Article  CAS  Google Scholar 

  51. Li, S. et al. Mechanism of intracellular cAMP sensor Epac2 activation: cAMP-induced conformational changes identified by amide hydrogen/deuterium exchange mass spectrometry (DXMS). J. Biol. Chem. 286, 17889–17897 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Structural Biology Center at Argonne National Laboratory for their help with data collection at the 19-ID beamline, D. Liu and I. Pass at Sanford Burnham Prebys for kindly providing materials, and G.N. Murshudov at University of Cambridge for the help with ligand building at the 2016 CCP4/APS summer school. This work was supported by the Wellcome Trust and by grants from the National Institutes of Health (R01GM117013, R01DK118297) and US ARMY Medical Research (W81XWH-16-1-0322) to F.R., as well as grants from Shandong University (Qilu Young Scholar 86963072), National Natural Science Foundation of China (31700114), Natural Science Foundation of Jiangsu Province (BK20170399), and the 111 Project (B16030) to D.W.

Author information

Authors and Affiliations

Authors

Contributions

D.W. purified proteins, carried out crystallization and solved the structures. X.S. conducted cell-based experiments. J.L. purified proteins and performed biochemical assays. S.L. executed the H/D-ex MS analysis. B.L.H. and S.V. performed the TR-FRET binding assays and thermal-shift-based screening experiments. N.P. produced the expression and mutation constructs. X.D. contributed to biochemical assays and structure refinement. Y.K. collected and processed synchrotron diffraction data. S.K. provided critical instruments and training for biochemical studies. All authors analyzed results. D.W. and F.R. conceived this study, designed experiments and wrote the manuscript.

Corresponding authors

Correspondence to Dalei Wu or Fraydoon Rastinejad.

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.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1 and 2, Supplementary Figures 1–14

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, D., Su, X., Lu, J. et al. Bidirectional modulation of HIF-2 activity through chemical ligands. Nat Chem Biol 15, 367–376 (2019). https://doi.org/10.1038/s41589-019-0234-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0234-5

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