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:

A structurally minimized yet fully active insulin based on cone-snail venom insulin principles

An Author Correction to this article was published on 12 June 2020

This article has been updated

Abstract

Human insulin and its current therapeutic analogs all show propensity, albeit varyingly, to self-associate into dimers and hexamers, which delays their onset of action and makes blood glucose management difficult for people with diabetes. Recently, we described a monomeric, insulin-like peptide in cone-snail venom with moderate human insulin-like bioactivity. Here, with insights from structural biology studies, we report the development of mini-Ins—a human des-octapeptide insulin analog—as a structurally minimal, full-potency insulin. Mini-Ins is monomeric and, despite the lack of the canonical B-chain C-terminal octapeptide, has similar receptor binding affinity to human insulin. Four mutations compensate for the lack of contacts normally made by the octapeptide. Mini-Ins also has similar in vitro insulin signaling and in vivo bioactivities to human insulin. The full bioactivity of mini-Ins demonstrates the dispensability of the PheB24–PheB25–TyrB26 aromatic triplet and opens a new direction for therapeutic insulin development.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of crystal structures of Con-Ins–G1-bound Fv83-7-bound μIR and of human insulin-bound Fv83-7-bound μIR.
Fig. 2: Structural biology of Con-Ins–G1 and human insulin within their respective μIR cocomplexes.
Fig. 3: Side-chain substitution of human insulin PheB24 by Con-Ins–G1 TyrB15.
Fig. 4: Structure–activity studies on Con-Ins–G1 and DOI.
Fig. 5: Structure–activity studies on DOI.
Fig. 6: Biochemical, in vitro and in vivo characterization of mini-Ins.
Fig. 7: Probing the interaction of mini-Ins with hIR.

Similar content being viewed by others

Data availability

Coordinates and structures factors for the structures presented here have been deposited in the Protein Data Bank as follows: Cons-Ins–G1-bound Fv83-7-bound μIR, PDB 6VEQ; human insulin-bound Fv83-7-bound μIR, PDB 6VEP and mini-Ins, PDB 6VET. Supplementary Methods and Source Data for Figs. 4a,b,d, 5b–d, 6 and 7a,b and Extended Data Figs. 2 and 3 are available with the paper online.

Change history

References

  1. Safavi-Hemami, H. et al. Specialized insulin is used for chemical warfare by fish-hunting cone snails. Proc. Natl Acad. Sci. USA 112, 1743–1748 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Menting, J. G. et al. A minimized human insulin-receptor-binding motif revealed in a Conus geographus venom insulin. Nat. Struct. Mol. Biol. 23, 916–920 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Carpenter, F. H. Relationship of structure to biological activity of insulin as revealed by degradative studies. Am. J. Med. 40, 750–758 (1966).

    Article  CAS  PubMed  Google Scholar 

  4. Gradel, A. K. J. et al. Factors affecting the absorption of subcutaneously administered insulin: effect on variability. J. Diabetes Res. 2018, 1205121 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Maikawa, C. L. et al. Stable monomeric insulin formulations enabled by supramolecular pegylation of insulin analogues. Adv. Ther. 3, 1900094 (2020).

    Article  Google Scholar 

  6. Kang, S., Brange, J., Burch, A., Vølund, A. & Owens, D. R. Subcutaneous insulin absorption explained by insulin’s physicochemical properties. Evidence from absorption studies of soluble human insulin and insulin analogues in humans. Diabetes Care 14, 942–948 (1991).

    Article  CAS  PubMed  Google Scholar 

  7. Kristensen, C., Andersen, A. S., Østergaard, S., Hansen, P. H. & Brandt, J. Functional reconstitution of insulin receptor binding site from non-binding receptor fragments. J. Biol. Chem. 277, 18340–18345 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Uchikawa, E., Choi, E., Shang, G., Yu, H. & Bai, X.-C. Activation mechanism of the insulin receptor revealed by cryo-EM structure of the fully liganded receptor-ligand complex. Elife 8, e48630 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Scapin, G. et al. Structure of the insulin receptor–insulin complex by single-particle cryo-EM analysis. Nature 556, 122–125 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Weis, F. et al. The signalling conformation of the insulin receptor ectodomain. Nat. Commun. 9, 4420 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Gutmann, T. et al. Cryo-EM structure of the complete and ligand-saturated insulin receptor ectodomain. J. Cell Biol. 219, e201907210 (2020).

    Article  PubMed  CAS  Google Scholar 

  12. Menting, J. G. et al. Protective hinge in insulin opens to enable its receptor engagement. Proc. Natl Acad. Sci. USA 111, E3395–E3404 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Menting, J. G. et al. How insulin engages its primary binding site on the insulin receptor. Nature 493, 241–245 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. De Meyts, P. & Whittaker, J. Structural biology of insulin and IGF1 receptors: implications for drug design. Nat. Rev. Drug Discov. 1, 769–783 (2002).

    Article  CAS  PubMed  Google Scholar 

  15. Kiselyov, V. V., Versteyhe, S., Gauguin, L. & De Meyts, P. Harmonic oscillator model of the insulin and IGF1 receptors’ allosteric binding and activation. Mol. Sys. Biol. 5, 243 (2009).

    Article  CAS  Google Scholar 

  16. Lawrence, C. F. et al. Insulin mimetic peptide disrupts the primary binding site of the insulin receptor. J. Biol. Chem. 291, 15473–15481 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nakagawa, S. H. & Tager, H. S. Role of the phenylalanine B25 side chain in directing insulin interaction with its receptor. Steric and conformational effects. J. Biol. Chem. 261, 7332–7341 (1986).

    Article  CAS  PubMed  Google Scholar 

  18. Pandyarajan, V. et al. Aromatic anchor at an invariant hormone-receptor interface: function of insulin residue B24 with application to protein design. J. Biol. Chem. 289, 34709–34727 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Kikuchi, K. et al. Studies on the biological activity of degraded insulins and insulin fragments. J. Biol. Chem. 255, 9281–9288 (1980).

    Article  CAS  PubMed  Google Scholar 

  20. Schwartz, G. P., Burke, G. T. & Katsoyannis, P. G. A superactive insulin: [B10-aspartic acid]insulin(human). Proc. Natl Acad. Sci. USA 84, 6408–6411 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kaarsholm, N. C. et al. Engineering stability of the insulin monomer fold with application to structure-activity relationships. Biochemistry 32, 10773–10778 (1993).

    Article  CAS  PubMed  Google Scholar 

  22. Kristensen, C. et al. Alanine scanning mutagenesis of insulin. J. Biol. Chem. 272, 12978–12983 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Thayer, W. P., Kraft, J. R., Tompkins, S. M., Moore, J. C. T. & Jensen, P. E. Assessment of the role of determinant selection in genetic control of the immune response to insulin in H-2b mice. J. Immunol. 163, 2549–2554 (1999).

    CAS  PubMed  Google Scholar 

  24. Menting, J. G., Ward, C. W., Margetts, M. B. & Lawrence, M. C. A thermodynamic study of ligand binding to the first three domains of the human insulin receptor: relationship between the receptor α-chain C-terminal peptide and the Site 1 insulin mimetic peptides. Biochemistry 48, 5492–5500 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Schäffer, L. A model for insulin binding to the insulin receptor. Eur. J. Biochem. 221, 1127–1132 (1994).

    Article  PubMed  Google Scholar 

  26. Smith, G. D., Pangborn, W. A. & Blessing, R. H. The structure of T6 human insulin at 1.0 Å resolution. Acta Crystallogr. D. 59, 474–482 (2003).

    Article  PubMed  CAS  Google Scholar 

  27. Ahorukomeye, P. et al. Fish-hunting cone snail venoms are a rich source of minimized ligands of the vertebrate insulin receptor. Elife 8, e41574 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  28. De Meyts, P. Insulin/receptor binding: the last piece of the puzzle? What recent progress on the structure of the insulin/receptor complex tells us (or not) about negative cooperativity and activation. Bioessays 37, 389–397 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Glendorf, T. et al. Systematic evaluation of the metabolic to mitogenic potency ratio for B10-substituted insulin analogues. PLoS ONE 7, e29198 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cara, J. F., Mirmira, R. G., Nakagawa, S. H. & Tager, H. S. An insulin-like growth factor I/insulin hybrid exhibiting high potency for interaction with the type I insulin-like growth factor and insulin receptors of placental plasma membranes. J. Biol. Chem. 265, 17820–17825 (1990).

    Article  CAS  PubMed  Google Scholar 

  31. Kurtzhals, P. et al. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes 49, 999–1005 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Laskowski, R. A. SURFNET: a program for visualizing molecular surfaces, cavities, and intermolecular interactions. J. Mol. Graph. 13, 323–330 (1995).

    Article  CAS  PubMed  Google Scholar 

  33. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Aragão, D. et al. MX2: a high-flux undulator microfocus beamline serving both the chemical and macromolecular crystallography communities at the Australian Synchrotron. J. Synchrotron Radiat. 25, 885–891 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Kabsch, W. Integration, scaling, space-group assignment and post-refinement. Acta Crystallogr. D Biol. Crystallogr. 66, 133–144 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  39. Winn, M. D., Isupov, M. N. & Murshudov, G. N. Use of TLS parameters to model anisotropic displacements in macromolecular refinement. Acta Crystallogr. D Biol. Crystallogr. 57, 122–133 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Bricogne, G. et al. BUSTER v.2.10 (Global Phasing Ltd, 2011).

  41. McKern, N. M. et al. Structure of the insulin receptor ectodomain reveals a folded-over conformation. Nature 443, 218–221 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Berry, M. B. et al. Structure of an anti-HIV monoclonal Fab antibody fragment specific to a gp120 C-4 region peptide. Proteins 45, 281–282 (2001).

    Article  CAS  PubMed  Google Scholar 

  43. Teplyakov, A. et al. On the domain pairing in chimeric antibodies. Mol. Immunol. 47, 2422–2426 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Celikel, R., Ruggeri, Z. M. & Varughese, K. I. von Willebrand factor conformation and adhesive function is modulated by an internalized water molecule. Nat. Struct. Biol. 7, 881–884 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. 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  PubMed  PubMed Central  Google Scholar 

  46. Webb, B. & Sali, A. Comparative protein structure modeling using MODELLER. Curr. Protoc. Bioinformatics 47, 5.6.1–5.6.32 (2014).

    Article  Google Scholar 

  47. Sparrow, L. G. et al. N-linked glycans of the human insulin receptor and their distribution over the crystal structure. Proteins: Struct. Funct. Bioinform. 71, 426–439 (2008).

    Article  CAS  Google Scholar 

  48. Abraham, M. J. et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 1–2, 19–25 (2015).

    Article  Google Scholar 

  49. Guvench, O. et al. CHARMM additive all-atom force field for carbohydrate derivatives and its utility in polysaccharide and carbohydrate-protein modeling. J. Chem. Theory Comput. 7, 3162–3180 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Best, R. B. et al. Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone ϕ, ψ and side-chain χ1 and χ2 dihedral angles. J. Chem. Theory Comput. 8, 3257–3273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hess, B. P-LINCS: a parallel linear constraint solver for molecular simulation. J. Chem. Theory Comput. 4, 116–122 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Denley, A. et al. Structural determinants for high-affinity binding of insulin-like growth factor II to insulin receptor (IR)-A, the exon 11 minus isoform of the IR. Mol. Endocrinol. 18, 2502–2512 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Rajapaksha, H. & Forbes, B. E. Ligand-binding affinity at the insulin receptor isoform-A and subsequent IR-A tyrosine phosphorylation kinetics are important determinants of mitogenic biological outcomes. Front. Endocrinol. 6, 107 (2015).

    Article  Google Scholar 

Download references

Acknowledgements

X.X. is a Juvenile Diabetes Research Foundation Postdoctoral Fellow. N.A.S. acknowledges receipt of an Australian Research Training Scholarship. R.S.N acknowledges fellowship support from the Australian National Health and Medical Research Council. Part of this work was undertaken using resources from the National Computational Infrastructure, which is supported by the Australian Government and provided through Intersect Australia Ltd, and through the HPC-GPGPU Facility, which was established with the assistance of a Linkage Infrastructure, Equipment and Facilities grant (LE170100200). We thank the CSIRO Protein Production Facility for the production under contract of cIR485, the precursor of IR310.T. Crystallization screening was undertaken at the CSIRO Collaborative Crystallisation Centre (www.csiro.au/C3), Melbourne, Australia. This research was undertaken in part using the MX2 beamline at the Australian Synchrotron, part of the Australian Nuclear Science and Technology Organisation, and made use of the ACRF detector. We thank M. Margetts for production of the heavy and light chain fragments of Fv83-7. This work is supported by NIDDK (DK120430 to D.H.C.), NIGMS (GM125001 to D.H.C.), the Juvenile Diabetes Research Foundation (5-CDA-2018-572-A-N to D.H.C. and 1-INO-2017-441-A-N to H.S.H.), the Australian National Health and Medical Research Council (NHMRC) Project grant nos. APP1143546 (to M.C.L., R.S.N., B.J.S., B.E.F. and D.H.C.) and APP1099595 (to M.C.L.). M.C.L.’s research is also made possible at The Walter and Eliza Hall Institute of Medical Research through Victorian State Government Operational Infrastructure Support and the Australian NHMRC Independent Research Institutes Infrastructure Support Scheme.

Author information

Authors and Affiliations

Authors

Contributions

X.X., J.G.M., R.S.N., H.S.-H., B.O., M.C.L. and D.H.-C.C. designed the study. M.C.L. and D.H.-C.C. wrote the manuscript with input from all authors. M.M.D. and J.G. generated Con-Ins–G1 analogs and M.M.D. performed related in vitro assays with assistance from G.G., and N.A.S. and B.J.S. performed modeling studies. C.D. and B.E.F. performed receptor binding and signaling studies. R.A. and S.J.F. performed in vivo bioactivity assays. X.W. and X.H. performed antibody response assays. C.A.M. and R.S.N. performed analytical ultracentrifugation studies. J.G.M and M.C.L performed crystallographic studies, and J.G.M. produced protein.

Corresponding authors

Correspondence to Michael C. Lawrence or Danny Hung-Chieh Chou.

Ethics declarations

Competing interests

M.C.L.’s laboratory has a funded Agreement with Eli Lilly and Company (USA) to conduct research not connected to this publication. Patents associated with part of this work were licensed to Monolog LLC, which aims to develop new fast-acting insulin analogs.

Additional information

Peer review information Katarzyna Marcinkiewicz was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Stereo views of sample (2mFobs-DFcalc) electron density for the structures presented in the manuscript.

(a) L1 domain residues 32-35 within monomer 1 of the Con-Ins-G1-bound μIR + Fv 83-7 crystal structure. (b) L1 domain residues 32-35 within monomer 1 of the human-insulin- bound μIR + Fv 83-7 crystal structure. (c) B-chain residues 14-16 within monomer 1 of the mini-Ins crystal structure. All maps are contoured at the 1.0 σ level.

Extended Data Fig. 2 Sedimentation equilibrium analysis of mini-Ins.

Sedimentation equilibrium analysis of mini-Ins was performed at 35,000 rpm with the best fit (curves) to a single species of apparent mass 5080 ± 45 Da. The molecular weight of mini-Ins is 5067. Detailed procedure can be found in ref. 2. Source data are available with the paper online.

Source data

Extended Data Fig. 3 Antibody response of 21-day immunization of bovine insulin, human insulin and mini-Ins.

Data are the average of 4 independent animals. Error bar represents S.E.M. Source data are available with the paper online.

Source data

Extended Data Fig. 4 Isothermal titration calorimetry.

Representative ITC thermograms for the titration against IR485 + IR-A704–719 αCT peptide of (a) mini-Ins; (b) hIns; (c) Con-Ins G1 and (d) human DOI.

Extended Data Fig. 5 Separation of insulin and IR amino acid pairs at the secondary binding site during 1 ns MD simulation.

(a) Distance between GluB10 carboxylate carbon (Cδ) and Arg539 guanyl carbon (Cζ). This salt pair remain closely associated (~4 Å) throughout the simulation. (b) Distance between HisA8 imidazole Nε2 nitrogen and Asp574 carboxylate carbon. (c) Distance between ArgA9 guanyl carbon and Glu575 carboxylate carbon (Cγ): this salt bridge forms (separation ~4 Å) following ~6ns MD. The salt bridge is observed to dissociate and reform several times throughout the simulation. Dissociation of this interaction correlates with increase in separation of the HisA8-Asp574 pair, reflecting mobility in the Phe572-to-Tyr579 loop of the FnIII-1.

Supplementary information

Supplementary Information

Supplementary Tables 1–4 and methods.

Reporting Summary

Source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Unmodified gels

Source Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 3

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiong, X., Menting, J.G., Disotuar, M.M. et al. A structurally minimized yet fully active insulin based on cone-snail venom insulin principles. Nat Struct Mol Biol 27, 615–624 (2020). https://doi.org/10.1038/s41594-020-0430-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-020-0430-8

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