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:

SF2312 is a natural phosphonate inhibitor of enolase

Abstract

Despite being crucial for energy generation in most forms of life, few if any microbial antibiotics specifically inhibit glycolysis. To develop a specific inhibitor of the glycolytic enzyme enolase 2 (ENO2) for the treatment of cancers with deletion of ENO1 (encoding enolase 1), we modeled the synthetic tool compound inhibitor phosphonoacetohydroxamate (PhAH) into the active site of human ENO2. A ring-stabilized analog of PhAH, in which the hydroxamic nitrogen is linked to Cα by an ethylene bridge, was predicted to increase binding affinity by stabilizing the inhibitor in a bound conformation. Unexpectedly, a structure-based search revealed that our hypothesized backbone-stabilized PhAH bears strong similarity to SF2312, a phosphonate antibiotic of unknown mode of action produced by the actinomycete Micromonospora, which is active under anaerobic conditions. Here, we present multiple lines of evidence, including a novel X-ray structure, that SF2312 is a highly potent, low-nanomolar inhibitor of enolase.

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: SF2312 is a potent inhibitor of enolase with mixed competitive and noncompetitive kinetics.
Figure 2: SF2312 stabilizes ENO2 against thermal denaturation.
Figure 3: PhAH and SF2312 interact with ENO2 through a complex network of electrostatic, metal coordination and hydrogen bond interactions.
Figure 4: SF2312 inhibits enolase activity and is selectively toxic to D423 cells.
Figure 5: SF2312 selectively blocks glycolysis in ENO1-deleted glioma cells.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Fothergill-Gilmore, L.A. & Michels, P.A. Evolution of glycolysis. Prog. Biophys. Mol. Biol. 59, 105–235 (1993).

    Article  CAS  Google Scholar 

  2. Deutscher, D., Meilijson, I., Kupiec, M. & Ruppin, E. Multiple knockout analysis of genetic robustness in the yeast metabolic network. Nat. Genet. 38, 993–998 (2006).

    Article  CAS  Google Scholar 

  3. Baba, T. et al. Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol. Syst. Biol. 2, 2006.0008 (2006).

    Article  Google Scholar 

  4. Vander Heiden, M.G., Cantley, L.C. & Thompson, C.B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  5. Muller, F.L. et al. Passenger deletions generate therapeutic vulnerabilities in cancer. Nature 488, 337–342 (2012).

    Article  CAS  Google Scholar 

  6. Poyner, R.R. & Reed, G.H. Structure of the bis divalent cation complex with phosphonoacetohydroxamate at the active site of enolase. Biochemistry 31, 7166–7173 (1992).

    Article  CAS  Google Scholar 

  7. Anderson, V.E., Weiss, P.M. & Cleland, W.W. Reaction intermediate analogues for enolase. Biochemistry 23, 2779–2786 (1984).

    Article  CAS  Google Scholar 

  8. Qin, J., Chai, G., Brewer, J.M., Lovelace, L.L. & Lebioda, L. Structures of asymmetric complexes of human neuron specific enolase with resolved substrate and product and an analogous complex with two inhibitors indicate subunit interaction and inhibitor cooperativity. J. Inorg. Biochem. 111, 187–194 (2012).

    Article  CAS  Google Scholar 

  9. de AS Navarro, M.V. et al. Structural flexibility in Trypanosoma brucei enolase revealed by X-ray crystallography and molecular dynamics. FEBS J. 274, 5077–5089 (2007).

    Article  Google Scholar 

  10. Wedekind, J.E., Poyner, R.R., Reed, G.H. & Rayment, I. Chelation of serine 39 to Mg2+ latches a gate at the active site of enolase: structure of the bis(Mg2+) complex of yeast enolase and the intermediate analog phosphonoacetohydroxamate at 2.1-Å resolution. Biochemistry 33, 9333–9342 (1994).

    Article  CAS  Google Scholar 

  11. Zhang, E., Hatada, M., Brewer, J.M. & Lebioda, L. Catalytic metal ion binding in enolase: the crystal structure of an enolase-Mn2+-phosphonoacetohydroxamate complex at 2.4-Å resolution. Biochemistry 33, 6295–6300 (1994).

    Article  CAS  Google Scholar 

  12. Watanabe, H., Yoshida, J., Tanaka, E., Ito, M., Miyadoh, S. & Shomura, T. Studies on a new phosphonic acid antibiotic, SF-2312. Scientific Reports of Meiji Seika Kaisha 25, 12–17 (1986).

    Google Scholar 

  13. Hanaya, T. & Itoh, C. An efficient synthesis of antibiotic SF-2312 (3-dihydroxyphosphoryl-1,5-dihydroxy-2-pyrrolidone). Heterocycles 82, 1675–1683 (2011).

    Article  CAS  Google Scholar 

  14. Liu, Y., Jacobs, H.K. & Gopalan, A.S. A new approach to cyclic hydroxamic acids: intramolecular cyclization of N-benzyloxy carbamates with carbon nucleophiles. Tetrahedron 67, 2206–2214 (2011).

    Article  CAS  Google Scholar 

  15. Martinez Molina, D. et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 341, 84–87 (2013).

    Article  Google Scholar 

  16. Brewer, J.M. & Wampler, J.E. A differential scanning calorimetric study of the effects of metal ions, substrate/product, substrate analogues and chaotropic anions on the thermal denaturation of yeast enolase 1. Int. J. Biol. Macromol. 28, 213–218 (2001).

    Article  CAS  Google Scholar 

  17. Zhang, E., Brewer, J.M., Minor, W., Carreira, L.A. & Lebioda, L. Mechanism of enolase: the crystal structure of asymmetric dimer enolase-2-phospho-D-glycerate/enolase-phosphoenolpyruvate at 2.0 Å resolution. Biochemistry 36, 12526–12534 (1997).

    Article  CAS  Google Scholar 

  18. Marangos, P.J., Parma, A.M. & Goodwin, F.K. Functional properties of neuronal and glial isoenzymes of brain enolase. J. Neurochem. 31, 727–732 (1978).

    Article  CAS  Google Scholar 

  19. Muller, F.L., Fletcher-Sananikone, E., Colla, S., Aquilanti, E. & DePinho, R. Collateral gene inactivation biomarkers and targets for cancer therapy. US patent WO2013090732A2 (2013).

  20. Dette, G.A., Knothe, H., Schönenbach, B. & Plage, G. Comparative study of fosfomycin activity in Mueller–Hinton media and in tissues. J. Antimicrob. Chemother. 11, 517–524 (1983).

    Article  CAS  Google Scholar 

  21. Horii, S. et al. Structure of alahopcin (nourseimycin), a new dipeptide antibiotic. J. Antibiot. (Tokyo) 38, 302–311 (1985).

    Article  CAS  Google Scholar 

  22. Higashide, E., Kanamaru, T., Fukase, H. & Horii, S. Isolation of dealanylalahopcin, a new amino acid, and its biological activity. J. Antibiot. (Tokyo) 38, 296–301 (1985).

    Article  CAS  Google Scholar 

  23. Gerlt, J.A., Babbitt, P.C. & Rayment, I. Divergent evolution in the enolase superfamily: the interplay of mechanism and specificity. Arch. Biochem. Biophys. 433, 59–70 (2005).

    Article  CAS  Google Scholar 

  24. Brewer, J.M. Yeast enolase: mechanism of activation by metal ions. CRC Crit. Rev. Biochem. 11, 209–254 (1981).

    Article  CAS  Google Scholar 

  25. Lee, J. & Kim, S.H. High-throughput T7 LIC vector for introducing C-terminal poly-histidine tags with variable lengths without extra sequences. Protein Expr. Purif. 63, 58–61 (2009).

    Article  CAS  Google Scholar 

  26. Studier, F.W. Protein production by auto-induction in high-density shaking cultures. Protein Exp. Purif. 41, 207–234 (2005).

    Article  CAS  Google Scholar 

  27. Battye, T.G., Kontogiannis, L., Johnson, O., Powell, H.R. & Leslie, A.G. iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM. Acta Crystallogr. D Biol. Crystallogr. 67, 271–281 (2011).

    Article  CAS  Google Scholar 

  28. Evans, P.R. & Murshudov, G.N. How good are my data and what is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69, 1204–1214 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Chai, G. et al. Expression, purification and the 1.8 angstroms resolution crystal structure of human neuron specific enolase. J. Mol. Biol. 341, 1015–1021 (2004).

    Article  CAS  Google Scholar 

  31. 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 

  32. Duncan, C.G. et al. Integrated genomic analyses identify ERRFI1 and TACC3 as glioblastoma-targeted genes. Oncotarget 1, 265–277 (2010).

    Article  Google Scholar 

  33. Stommel, J.M. et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318, 287–290 (2007).

    Article  CAS  Google Scholar 

  34. Bady, P. et al. DNA fingerprinting of glioma cell lines and considerations on similarity measurements. Neuro-oncol. 14, 701–711 (2012).

    Article  CAS  Google Scholar 

  35. Torsvik, A. et al. U-251 revisited: genetic drift and phenotypic consequences of long-term cultures of glioblastoma cells. Cancer Med. 3, 812–824 (2014).

    Article  CAS  Google Scholar 

  36. Yuan, M., Breitkopf, S.B., Yang, X. & Asara, J.M. A positive/negative ion-switching, targeted mass spectrometry-based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue. Nat. Protoc. 7, 872–881 (2012).

    Article  CAS  Google Scholar 

  37. Ying, H. et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 149, 656–670 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Holton and G. Meigs for their assistance with X-ray diffraction data collection at the Advanced Light Source, beamline 8.3.1. We thank K. Kalhurachi and J. McMurray for assistance with 13C-NMR measurements. We thank S. Millward, J. Marszalek and Y. Sun for critical comments and suggestions. We thank M. Yuan and S. Breitkopf for help with MS experiments. We thank M. Protopopova for assistance with Operetta cell imaging. We thank L. Lebioda, G. Reed and R. Poyner for useful discussions about the catalytic mechanism of enolase. We thank R. Zielinski for assistance with hypoxia experiments. The cell line D423-MG (D423) was kindly shared by D. Bigner (Duke University Medical Center) while the Gli56 cell line was shared by D. Louis (Massachusetts General Hospital) and TS neurospheres were shared by C. Brennan (Memorial Sloan Kettering Cancer Center). Inhibitors were provided by the Pharmaceutical Chemistry Facility at MD Anderson, supported by the US National Institutes of Health (NIH) National Cancer Institute under award number P30CA016672. Financial support was provided by NIH-NCI grant 7P01CA095616-10 (R.A.D.); Cancer Prevention Research Institute of Texas (CPRIT) grant RP140612 (R.A.D.); NCI grants 5P01CA120964 (J.M.A.) and 5P30CA006516 (J.M.A.) and NIH Career Development Project through the Specialized Programs of Research Excellence (SPORE) P50CA127001-07 (F.L.M.). F.L.M. was supported by Research Scholar Grant RSG-15-145-01-CDD from the American Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

D.M. performed structural modeling and docking; F.L.M. and D.M. conceived the cyclized inhibitors, which were synthesized by Z.P., D.S., B.A.B.P. and W.B.; P.G.L., G.R.L. and T.M.L. performed isolation of recombinant protein and X-ray crystallography; M.E.D.F., F.P., F.L.M. and B.C. repeated chemical syntheses and wrote synthetic procedures with characterizations. F.P. and F.L.M. performed chiral chromatography. F.L.M. and N.S. performed in vitro enzymatic activity experiments. N.S., N.H. and Y.-H.L. performed cell culture, western blots, 13C-NMR tracing and biochemical profiling experiments. N.S. performed thermal shift assays. J.M.A. performed MS small-molecule metabolite analysis. R.A.D., Y.A.W., F.L.M., N.S. and P.G.L. oversaw overall experimental design and data analysis and wrote the manuscript.

Corresponding author

Correspondence to Florian L Muller.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Tables 1–2 and Supplementary Figures 1–26. (PDF 5726 kb)

Supplementary Note 1

Synthetic procedures. (PDF 115 kb)

Supplementary Note 2 (PDF 544 kb)

Source Data

Source data for Figures 1, 2, 4, and 5. (ZIP 142 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leonard, P., Satani, N., Maxwell, D. et al. SF2312 is a natural phosphonate inhibitor of enolase. Nat Chem Biol 12, 1053–1058 (2016). https://doi.org/10.1038/nchembio.2195

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2195

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