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 amylase inhibitor montbretin A reveals a new glycosidase inhibition motif

Abstract

The complex plant flavonol glycoside montbretin A is a potent (Ki = 8 nM) and specific inhibitor of human pancreatic α-amylase with potential as a therapeutic for diabetes and obesity. Controlled degradation studies on montbretin A, coupled with inhibition analyses, identified an essential high-affinity core structure comprising the myricetin and caffeic acid moieties linked via a disaccharide. X-ray structural analyses of the montbretin A–human α-amylase complex confirmed the importance of this core structure and revealed a novel mode of glycosidase inhibition wherein internal π-stacking interactions between the myricetin and caffeic acid organize their ring hydroxyls for optimal hydrogen bonding to the α-amylase catalytic residues D197 and E233. This novel inhibitory motif can be reproduced in a greatly simplified analog, offering potential for new strategies for glycosidase inhibition and therapeutic development.

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: Characterization of MbA functionalities.
Figure 2: The binding mode of MbA.
Figure 3: Pre-formed characteristics of MbA structure in solution.
Figure 4: Chemical structure of mini-MbA analog inhibitor 10.

Similar content being viewed by others

References

  1. World Health Organization. WHO Fact Sheet 312: Diabetes (WHO, 2013) http://www.who.int/mediacentre/factsheets/fs312/en/.

  2. Chiasson, J.L. et al. The efficacy of acarbose in the treatment of patients with non-insulin-dependent diabetes mellitus—a multicenter controlled clinical trial. Ann. Intern. Med. 121, 928–935 (1994).

    Article  CAS  Google Scholar 

  3. Mooradian, A.D. & Thurman, J.E. Drug therapy of postprandial hyperglycaemia. Drugs 57, 19–29 (1999).

    Article  CAS  Google Scholar 

  4. Scott, L.J. & Spencer, C.M. Miglitol—A review of its therapeutic potential in type 2 diabetes mellitus. Drugs 59, 521–549 (2000).

    Article  CAS  Google Scholar 

  5. Brayer, G.D., Luo, Y.G. & Withers, S.G. The structure of human pancreatic alpha-amylase at 1.8 Angstrom resolution and comparisons with related enzymes. Protein Sci. 4, 1730–1742 (1995).

    Article  CAS  Google Scholar 

  6. Li, C.M. et al. Acarbose rearrangement mechanism implied by the kinetic and structural analysis of human pancreatic alpha-amylase in complex with analogues and their elongated counterparts. Biochemistry 44, 3347–3357 (2005).

    Article  CAS  Google Scholar 

  7. Tarling, C.A. et al. The search for novel human pancreatic alpha-amylase inhibitors: high-throughput screening of terrestrial and marine natural product extracts. ChemBioChem 9, 433–438 (2008).

    Article  CAS  Google Scholar 

  8. Asada, Y., Hirayama, Y. & Furuya, T. Acylated flavonols from Crocosmia crocosmiiflora. Phytochemistry 27, 1497–1501 (1988).

    Article  CAS  Google Scholar 

  9. Lo Piparo, E. et al. Flavonoids for controlling starch digestion: structural requirements for inhibiting human alpha-amylase. J. Med. Chem. 51, 3555–3561 (2008).

    Article  CAS  Google Scholar 

  10. Ong, K.C. & Khoo, H.E. Effects of myricetin on glycemia and glycogen metabolism in diabetic rats. Life Sci. 67, 1695–1705 (2000).

    Article  CAS  Google Scholar 

  11. Yoshida, K., Hishida, A., Iida, O., Hosokawa, K. & Kawabata, J. Flavonol caffeoylglycosides as alpha-glucosidase inhibitors from Spiraea cantoniensis flower. J. Agric. Food Chem. 56, 4367–4371 (2008).

    Article  CAS  Google Scholar 

  12. Manaharan, T., Appleton, D., Cheng, H.M. & Palanisamy, U.D. Flavonoids isolated from Syzygium aqueum leaf extract as potential antihyperglycaemic agents. Food Chem. 132, 1802–1807 (2012).

    Article  CAS  Google Scholar 

  13. Williams, L.K., Li, C.M., Withers, S.G. & Brayer, G.D. Order and disorder: differential structural impacts of myricetin and ethyl caffeate on human amylase, an antidiabetic target. J. Med. Chem. 55, 10177–10186 (2012).

    Article  CAS  Google Scholar 

  14. Brayer, G.D. et al. Subsite mapping of the human pancreatic alpha-amylase active site through structural, kinetic, and mutagenesis techniques. Biochemistry 39, 4778–4791 (2000).

    Article  CAS  Google Scholar 

  15. Sim, L., Quezada-Calvillo, R., Sterchi, E.E., Nichols, B.L. & Rose, D.R. Human intestinal maltase-glucoamylase: crystal structure of the N-terminal catalytic subunit and basis of inhibition and substrate specificity. J. Mol. Biol. 375, 782–792 (2008).

    Article  CAS  Google Scholar 

  16. Sim, L. et al. Structural basis for substrate selectivity in human maltase-glucoamylase and sucrase-isomaltase N-terminal domains. J. Biol. Chem. 285, 17763–17770 (2010).

    Article  CAS  Google Scholar 

  17. McCarter, J.D. & Withers, S.G. Mechanisms of enzymatic glycoside hydrolysis. Curr. Opin. Struct. Biol. 4, 885–892 (1994).

    Article  CAS  Google Scholar 

  18. Mhlongo, N.N., Skelton, A.A., Kruger, G., Soliman, M.E.S. & Williams, I.H. A critical survey of average distances between catalytic carboxyl groups in glycoside hydrolases. Proteins 82, 1747–1755 (2014).

    Article  CAS  Google Scholar 

  19. Wang, Q.P., Graham, R.W., Trimbur, D., Warren, R.A.J. & Withers, S.G. Changing enzymatic reaction mechanisms by mutagenesis- conversion of a retaining glucosidase to an inverting enzyme. J. Am. Chem. Soc. 116, 11594–11595 (1994).

    Article  CAS  Google Scholar 

  20. Rydberg, E.H. et al. Mechanistic analyses of catalysis in human pancreatic alpha-amylase: detailed kinetic and structural studies of mutants of three conserved carboxylic acids. Biochemistry 41, 4492–4502 (2002).

    Article  CAS  Google Scholar 

  21. The PyMOL Molecular Graphics System, version 1.3r1 (Schrodinger LLC, San Diego, California, USA, 2010).

  22. Rydberg, E.H. et al. Cloning, mutagenesis, and structural analysis of human pancreatic alpha-amylase expressed in Pichia pastoris. Protein Sci. 8, 635–643 (1999).

    Article  CAS  Google Scholar 

  23. Sambrook, J., Fritsch, E.F. & Maniatis, T. Molecular Cloning: A Laboratory Manual edn. 2 (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, USA, 1989).

  24. Numao, S. et al. Probing the role of the chloride ion in the mechanism of human pancreatic alpha-amylase. Biochemistry 41, 215–225 (2002).

    Article  CAS  Google Scholar 

  25. Woods, K.B. Bioactive natural products. PhD Thesis, University of British Columbia (2010).

  26. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  Google Scholar 

  27. Karplus, P.A. & Diederichs, K. Linking crystallographic model and data quality. Science 336, 1030–1033 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  Google Scholar 

  31. Schüttelkopf, A.W. & van Aalten, D.M.F. PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60, 1355–1363 (2004).

    Article  Google Scholar 

  32. Moriarty, N.W., Grosse-Kunstleve, R.W. & Adams, P.D. electronic Ligand Builder and Optimization Workbench (eLBOW): a tool for ligand coordinate and restraint generation. Acta Crystallogr. D Biol. Crystallogr. 65, 1074–1080 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank R. Maurus for helpful discussions regarding structural analyses and E. Goddard-Borger for advice on synthesis. We thank E. Kwan for preparation of HPA, T. Hill for isolation of human gut bacterial amylases, C. Hii, P. Lincez and K. Liu for additional technical assistance, H. Flint for providing clones of the human gut bacterial amylases and D.R. Rose (University of Waterloo) for supplies of ntMGAM, ctMGAM and ctSI. This work was supported by Operating and Proof of Principle grants from the Canadian Institutes of Health Research (CIHR; ref. nos. (FRN) 111082, 200704PPP (to G.D.B. and S.G.W.)), by an operating grant from the Natural Sciences and Engineering Research Council (NSERC) of Canada to R.J.A. and through the CDRD-Pfizer Innovation Fund. L.K.W. was supported by an Alexander Graham Bell Canada Graduate Scholarship (NSERC). S.G.W. is supported by a Tier 1 Canada Research Chair. Portions of this research were carried out at the Stanford Synchrotron Radiation Lightsource (SSRL), SLAC National Accelerator Laboratory, which is supported by the US Department of Energy (DOE), Office of Science, Office of Basic Energy Sciences under contract no. DE-AC02-76SF00515. The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research and by the National Institutes of Health (NIH), National Institute of General Medical Sciences (NIGMS) (including P41GM103393). The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of NIGMS or NIH. The graphics program PyMOL (ref. 21) was used in part to generate figures.

Author information

Authors and Affiliations

Authors

Contributions

N.T.N. crystallized the MbA–HPA complex, and L.K.W. and S.C. solved and analyzed its structure; X.Z. performed the degradation studies of MbA and kinetic studies thereon; C.T. synthesized inhibitor 10 and performed kinetic studies thereon; J.W. performed kinetic studies on MbA variants; D.E.W. performed the ROESY NMR studies on MbA; J.C., V.Y., J.M., R.J.A., S.G.W. and G.D.B. designed and supervised the project. Writing was primarily done by V.Y., G.D.B., L.K.W. and S.G.W. with contributions from all.

Corresponding author

Correspondence to Stephen G Withers.

Ethics declarations

Competing interests

The University of British Columbia holds a patent on the use of MbA as a blood glucose controlling agent with R.J.A., G.D.B. and S.G.W. as inventors.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Tables 1–3, Supplementary Figures 1–5 and Supplementary Notes 1 and 2. (PDF 1922 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Williams, L., Zhang, X., Caner, S. et al. The amylase inhibitor montbretin A reveals a new glycosidase inhibition motif. Nat Chem Biol 11, 691–696 (2015). https://doi.org/10.1038/nchembio.1865

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1865

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