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:

Synergistic activation of the insulin receptor via two distinct sites

Abstract

Insulin receptor (IR) signaling controls multiple facets of animal physiology. Maximally four insulins bind to IR at two distinct sites, termed site-1 and site-2. However, the precise functional roles of each binding event during IR activation remain unresolved. Here, we showed that IR incompletely saturated with insulin predominantly forms an asymmetric conformation and exhibits partial activation. IR with one insulin bound adopts a Γ-shaped conformation. IR with two insulins bound assumes a Ƭ-shaped conformation. One insulin binds at site-1 and another simultaneously contacts both site-1 and site-2 in the Ƭ-shaped IR dimer. We further show that concurrent binding of four insulins to sites-1 and -2 prevents the formation of asymmetric IR and promotes the T-shaped symmetric, fully active state. Collectively, our results demonstrate how the synergistic binding of multiple insulins promotes optimal IR activation.

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: Binding to IR by insulin site-1 and site-2 mutants.
Fig. 2: Structure of IR with insulin bound only to site-2.
Fig. 3: Structures of IR with insulin LeuA13R bound only to site-1.
Fig. 4: Structures of IR with insulin LeuB17R bound only to site-1 and functional importance of site-2 insulin binding.
Fig. 5: Mixture of insulin site-1 and site-2 mutants promotes the formation of T-shaped IR dimer and activates IR signaling.
Fig. 6: Structures of IR/insulin WT complex at subsaturated insulin concentrations.
Fig. 7: Working models for insulin-induced IR activation.

Similar content being viewed by others

Data availability

All reagents generated in this study are available with a completed Materials Transfer Agreement. All cryo-EM maps and models reported in this work have been deposited into EMDB/PDB database, under the entry ID: EMD-25188/PDB 7SL1, EMD-25189/PDB 7SL2, EMD-25190/PDB 7SL3, EMD-25191/PDB 7SL4, EMD-25192/PDB 7SL6, EMD-25193/PDB 7SL7, EMD-25428/PDB 7STH, EMD-25249/PDB 7STI, EMD-25430/7STJ and EMD-25431/7STK. Source data are provided with this paper.

References

  1. Haeusler, R. A., McGraw, T. E. & Accili, D. Biochemical and cellular properties of insulin receptor signalling. Nat. Rev. Mol. Cell Biol. 19, 31–44 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Ullrich, A. et al. Human insulin receptor and its relationship to the tyrosine kinase family of oncogenes. Nature 313, 756–761 (1985).

    Article  CAS  PubMed  Google Scholar 

  3. White, M. F. Insulin signaling in health and disease. Science 302, 1710–1711 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Boucher, J., Kleinridders, A. & Kahn, C. R. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb. Perspect. Biol. 6, a009191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Petersen, M. C. & Shulman, G. I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 98, 2133–2223 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Olefsky, J. M. The insulin receptor: its role in insulin resistance of obesity and diabetes. Diabetes 25, 1154–1162 (1976).

    Article  CAS  PubMed  Google Scholar 

  7. Lawrence, M. C. Understanding insulin and its receptor from their three-dimensional structures. Mol. Metab. 52, 101255 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed  Google Scholar 

  9. Ferguson, K. M., Hu, C. & Lemmon, M. A. Insulin and epidermal growth factor receptor family members share parallel activation mechanisms. Protein Sci. 29, 1331–1344 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. 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 

  12. Gutmann, T., Kim, K. H., Grzybek, M., Walz, T. & Coskun, U. Visualization of ligand-induced transmembrane signaling in the full-length human insulin receptor. J. Cell Biol. 217, 1643–1649 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  14. 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 

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

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. 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  PubMed  Google Scholar 

  19. Bai, X. C., Rajendra, E., Yang, G., Shi, Y. & Scheres, S. H. Sampling the conformational space of the catalytic subunit of human gamma-secretase. eLife 4, e11182 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Li, J. et al. Cryo-EM analyses reveal the common mechanism and diversification in the activation of RET by different ligands. eLife 8, e47650 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ciszak, E. & Smith, G. D. Crystallographic evidence for dual coordination around zinc in the T3R3 human insulin hexamer. Biochemistry 33, 1512–1517 (1994).

    Article  CAS  PubMed  Google Scholar 

  22. Kavran, J. M. et al. How IGF-1 activates its receptor. eLife 3, e03772 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Xu, Y. et al. How ligand binds to the type 1 insulin-like growth factor receptor. Nat. Commun. 9, 821 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Tofteng, A. P., Jensen, K. J., Schaffer, L. & Hoeg-Jensen, T. Total synthesis of desB30 insulin analogues by biomimetic folding of single-chain precursors. ChemBioChem 9, 2989–2996 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Zaykov, A. N., Mayer, J. P., Gelfanov, V. M. & DiMarchi, R. D. Chemical synthesis of insulin analogs through a novel precursor. ACS Chem. Biol. 9, 683–691 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Ball, K. A. et al. An isothermal shift assay for proteome scale drug-target identification. Commun. Biol. 3, 75 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Uchikawa, E., Chen, Z., Xiao, G. Y., Zhang, X. & Bai, X. C. Structural basis of the activation of c-MET receptor. Nat. Commun. 12, 4074 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  29. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang, K. Gctf: Real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  32. 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  PubMed  PubMed Central  Google Scholar 

  33. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D. Struct. Biol. 75, 861–877 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Choi, E., Zhang, X., Xing, C. & Yu, H. Mitotic checkpoint regulators control insulin signaling and metabolic homeostasis. Cell 166, 567–581 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Choi, E. et al. Mitotic regulators and the SHP2-MAPK pathway promote IR endocytosis and feedback regulation of insulin signaling. Nat. Commun. 10, 1473 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Li, J., Choi, E., Yu, H. & Bai, X. C. Structural basis of the activation of type 1 insulin-like growth factor receptor. Nat. Commun. 10, 4567 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Keller, S. et al. High-precision isothermal titration calorimetry with automated peak-shape analysis. Anal. Chem. 84, 5066–5073 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brautigam, C. A., Zhao, H., Vargas, C., Keller, S. & Schuck, P. Integration and global analysis of isothermal titration calorimetry data for studying macromolecular interactions. Nat. Protoc. 11, 882–894 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Brautigam, C. A. Calculations and publication-quality illustrations for analytical ultracentrifugation data. Methods Enzymol. 562, 109–133 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Cryo-EM data were collected at the University of Texas Southwestern Medical Center (UTSW) Cryo-Electron Microscopy Facility, funded in part by the Cancer Prevention and Research Institute of Texas (CPRIT) Core Facility Support Award RP170644. We thank D. Stoddard for facility access. This work is supported in part by grants from the National Institutes Health (R01GM136976 to X.-C.B., R35GM142937, P30DK063608 and UL1TR001873 to E.C., AG061829 to M.H.B.S and R35GM130289 to X.Z), the Welch foundation (I-1944 to X.-C.B. and I-1702 to X.Z.), CPRIT (RP160082 to X.-C.B.), the MCDB Neurodegenerative Disease Fund (to M.H.B.S.), the T. Curtius Peptide Facility (to M.H.B.S.) and the Alice Bohmfalk Charitable (to E.C.). X.-C.B. and X.Z. are Virginia Murchison Linthicum Scholars in Medical Research at UTSW. This paper is dedicated to the memory of Dr. Misha Plam.

Author information

Authors and Affiliations

Authors

Contributions

M.H.B.S., E.C. and X.-C.B. designed and supervised the research. All the authors performed the research. J.P.M., X.Z., M.H.B.S., E.C. and X.-C.B. analyzed the data. J.P.M., M.H.B.S., E.C. and X.-C.B. wrote the paper with input from the other authors.

Corresponding authors

Correspondence to Michael H. B. Stowell, Eunhee Choi or Xiao-chen Bai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Stevan Hubbard, John Rubinstein and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Florian Ullrich, in collaboration with the Nature Structural & Molecular Biology team. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Domains of insulin receptor (IR) and activities of insulin analogs in primary mouse hepatocytes.

a. Domains and disulfide connectivity of IR. L1 and L2, leucine rich domains 1 and 2; CR, cysteine rich domain; F1, F2 and F3, fibronectin III (FnIII) domains; ID, insert in FnIII-2 domain; TM, transmembrane domain; TK, tyrosine kinase domain. b. HPLC traces for each of the insulins synthesized and utilized for both functional and structural studies. c. MS1 spectra of the purified insulins in B analyzed in the Orbitrap mass analyzer. d. Binding of insulin WT and site1 mutant (IleA2A;ValA3A) labeled with Alexa Fluor 488 to purified IR WT in the indicated conditions (Mean ± SD, WT, n = 9 independent experiments; IleA2A;ValA3A, n = 3). Significance calculated using two-tailed student t-test; **p < 0.01 and ***p < 0.001 (The exact p values are provided in the source data). e. Insulin competition-binding assay for isolated FnIII-1 domain and insulin WT and mutants (ValA3E and LeuB17R) (Mean ± SD, n = 3). f. Insulin-induced IR autophosphorylation in 293FT cells expressing IR wild-type (WT). Cells were treated with the indicated insulin WT or site-2 mutants for 10 min. The IR autophosphorylation levels were assessed by quantitative western blotting with a phospho-tyrosine (pY) IRβ antibody. Expression levels of IRβ were monitored by anti-Myc blotting against the C-terminal Myc-tag. g. Quantification of the western blot data shown in e (Mean ± SD). Each experiment was repeated four times. Significance calculated using two-tailed student t-test; *p < 0.05; **p < 0.01, ***p < 0.001, and ****p < 0.0001 (The exact p values are provided in the source data). Uncropped images for all blots and gels are available as source data.

Source data

Extended Data Fig. 2 Purification of the full-length mouse insulin receptor (IR).

a. A representative size-exclusion chromatography of IR. b. The peak fractions were combined and visualized on SDS-PAGE by Coomassie staining, in the absence or presence of dithiothreitol (DTT). Most of IR was processed into α-chain (IRα) and β-chain (IRβ). This experiment was repeated for 10 times independently with similar results.

Extended Data Fig. 3 Cryo-EM analysis of the IR-insulin site-1 mutant (ValA3E) complex.

a. Representative electron micrograph and 2D class averages of the IR-insulin site-1 mutant (ValA3E) complex. Scale bar: 200 Å. This experiment was repeated for 2,879 times independently with similar results. b. Unsharpened cryo-EM map colored by local resolution. c. The gold-standard Fourier Shell Correlation (FSC) curve for the cryo-EM map shown in Fig. 2a. d. Flowchart of cryo-EM data processing.

Extended Data Fig. 4 Cryo-EM analysis of the IR-insulin site-2 mutant (LeuA13R) complex.

a. Representative electron micrograph and 2D class averages of the IR-insulin site-2 mutant (LeuA13R) complex. Scale bar: 200 Å. This experiment was repeated for 3,783 times independently with similar results. b. Unsharpened cryo-EM map colored by local resolution. c. The gold-standard Fourier Shell Correlation (FSC) curve for the cryo-EM map shown in Fig. 3a. d. Flowchart of cryo-EM data processing.

Extended Data Fig. 5 Close-up view of asymmetric IR/insulin LeuA13R complex.

a. Overall view of asymmetric IR/insulin LeuA13R complex in two orthogonal views. b. The close-up view of the contact of site-1 bound insulin LeuA13R at FnIII-1 domain in the asymmetric IR/insulin LeuA13R complex. The location of this interaction in the asymmetric dimer is indicated by a blue box in a. c. The close-up view of the binding of a dimeric insulin at asymmetric IR/insulin LeuA13R complex. The location of this interaction in the asymmetric dimer is indicated by a red box in a.

Extended Data Fig. 6 Cryo-EM analysis of the IR-insulin site-2 mutant (LeuB17R) complex.

a. Representative electron micrograph and 2D class averages of the IR-insulin site-2 mutant (LeuB17R) complex. Scale bar: 200 Å. This experiment was repeated for 3,995 times independently with similar results. b. Unsharpened cryo-EM map colored by local resolution. c. The gold-standard Fourier Shell Correlation (FSC) curve for the cryo-EM map shown in Fig. 4a. d. Flowchart of cryo-EM data processing.

Extended Data Fig. 7 Insulin binding to site-2 of IR facilitates IR activation and signaling.

a. IR signaling in 293FT cells expressing IR wild-type (WT). Cells were treated with the 10 nM insulin WT and site-2 mutants for the indicated times. Cell lysates were blotted with the indicated antibodies. b. Quantification of the western blot data shown in a (Mean ± SEM, For pY/IR, n = 7 independent experiments; pAKT/AKT, n = 4, pERK/ERK, n = 6). Significance calculated using two-tailed student t-test; *p < 0.05; **p < 0.01, ***p < 0.001, and ****p < 0.0001 (The exact p values are provided in the source data). c. IR signaling in primary mouse hepatocytes treated with the indicated concentrations of insulin for 10 min. Cell lysates were blotted with the indicated antibodies. Quantification of the western blot data shown in Fig. 4e. d. IR signaling in primary mouse hepatocytes treated with 10 nM insulin for the indicated times. Cell lysates were blotted with the indicated antibodies. Quantification of the western blot data shown in Fig. 4g. Uncropped images for all blots and gels are available as source data.

Source data

Extended Data Fig. 8 Cryo-EM analysis of the IR-insulin site-1 (ValA3E) and site-2 (LeuA13R) mutants complex.

a. Representative electron micrograph and 2D class averages of the IR-insulin site-1 (ValA3E) and site-2 (LeuA13R) mutants complex. Scale bar: 200 Å. This experiment was repeated for 4,895 times independently with similar results. b. Unsharpened cryo-EM map colored by local resolution. c. The gold-standard Fourier Shell Correlation (FSC) curve for the cryo-EM map shown in Fig. 5a, b. d. Flowchart of cryo-EM data processing.

Extended Data Fig. 9 Cryo-EM analysis of the IR-insulin WT complex at subsaturated insulin concentrations.

a. Representative electron micrograph and 2D class averages of the IR-insulin WT complex at subsaturated insulin concentrations. Scale bar: 200 Å. This experiment was repeated for 3,635 times independently with similar results. b. Unsharpened cryo-EM maps colored by local resolution. c. The gold-standard Fourier Shell Correlation (FSC) curve for the cryo-EM map shown in Fig. 6a-d. d. Flowchart of cryo-EM data processing.

Extended Data Fig. 10 Close-up view of insulin, α-CT in asymmetric IR dimer and membrane proximal domains in asymmetric and symmetric IR dimer.

a. The close-up view of the contact of site-2 bound insulin at α-CT in the asymmetric IR/insulin complex. b. Close-up view of α-CT in 1 and 2 insulins bound asymmetric IR dimer. c. Superposition of the hybrid sites between the Ƭ-shaped asymmetric conformations 1 and 2, showing the rotation of insulin around the α-CT and two different insulin binding modes. d. Close-up view of the membrane proximal domains in asymmetric and symmetric IR dimer. e. Superposition between the membrane proximal domains in asymmetric and symmetric IR dimer.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, J., Park, J., Mayer, J.P. et al. Synergistic activation of the insulin receptor via two distinct sites. Nat Struct Mol Biol 29, 357–368 (2022). https://doi.org/10.1038/s41594-022-00750-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00750-6

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