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:

Structural basis of inhibition of the human SGLT2–MAP17 glucose transporter

Abstract

Human sodium–glucose cotransporter 2 (hSGLT2) mediates the reabsorption of the majority of filtrated glucose in the kidney1. Pharmacological inhibition of hSGLT2 by oral small-molecule inhibitors, such as empagliflozin, leads to enhanced excretion of glucose and is widely used in the clinic to manage blood glucose levels for the treatment of type 2 diabetes1. Here we determined the cryogenic electron microscopy structure of the hSGLT2–MAP17 complex in the empagliflozin-bound state to an overall resolution of 2.95 Å. Our structure shows eukaryotic SGLT-specific structural features. MAP17 interacts with transmembrane helix 13 of hSGLT2. Empagliflozin occupies both the sugar-substrate-binding site and the external vestibule to lock hSGLT2 in an outward-open conformation, thus inhibiting the transport cycle. Our work provides a framework for understanding the mechanism of SLC5A family glucose transporters and also develops a foundation for the future rational design and optimization of new inhibitors targeting these transporters.

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: Architecture of the hSGLT2–MAP17 complex.
Fig. 2: The eukaryotic-specific features of hSGLT2.
Fig. 3: The empagliflozin-binding site of hSGLT2.
Fig. 4: Empagliflozin locks hSGLT2 in the outward-facing conformation.

Similar content being viewed by others

Data availability

Cryo-EM maps and atomic coordinates of the hSGLT2–MAP17 complex have been deposited in the Electron Microscopy Data Bank and PDB under the ID codes EMD-31558 and 7FEN, respectively. Source data are provided with this paper.

References

  1. Ferrannini, E. Sodium-glucose co-transporters and their inhibition: clinical physiology. Cell Metab. 26, 27–38 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Wright, E. M., Loo, D. D. & Hirayama, B. A. Biology of human sodium glucose transporters. Physiol. Rev. 91, 733–794 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Coady, M. J. et al. MAP17 is a necessary activator of renal Na+/glucose cotransporter SGLT2. J. Am. Soc. Nephrol. 28, 85–93 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Santer, R. & Calado, J. Familial renal glucosuria and SGLT2: from a mendelian trait to a therapeutic target. Clin. J. Am. Soc. Nephrol. 5, 133–141 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Faillie, J. L. Pharmacological aspects of the safety of gliflozins. Pharmacol. Res. 118, 71–81 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Choi, C. I. Sodium-glucose cotransporter 2 (SGLT2) inhibitors from natural products: discovery of next-generation antihyperglycemic agents. Molecules 21, 1136 (2016).

    Article  PubMed Central  Google Scholar 

  7. Frampton, J. E. Empagliflozin: a review in type 2 diabetes. Drugs 78, 1037–1048 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Ghezzi, C. et al. SGLT2 inhibitors act from the extracellular surface of the cell membrane. Physiol. Rep. 2, e12058 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Faham, S. et al. The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321, 810–814 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Watanabe, A. et al. The mechanism of sodium and substrate release from the binding pocket of vSGLT. Nature 468, 988–991 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kawate, T. & Gouaux, E. Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673–681 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Kirchhofer, A. et al. Modulation of protein properties in living cells using nanobodies. Nat. Struct. Mol. Biol. 17, 133–138 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Chang, H. C. et al. Development of a novel non-radioactive cell-based method for the screening of SGLT1 and SGLT2 inhibitors using 1-NBDG. Mol. Biosyst. 9, 2010–2020 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Patel, A., Toso, D., Litvak, A. & Nogales, E. Efficient graphene oxide coating improves cryo-EM sample preparation and data collection from tilted grids. Preprint at https://doi.org/10.1101/2021.03.08.434344 (2021).

  15. Wang, N. et al. Structural basis of human monocarboxylate transporter 1 inhibition by anti-cancer drug candidates. Cell 184, 370–383.e13 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Yamashita, A., Singh, S. K., Kawate, T., Jin, Y. & Gouaux, E. Crystal structure of a bacterial homologue of Na+/Cl-dependent neurotransmitter transporters. Nature 437, 215–223 (2005).

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Jungnickel, K. E. J., Parker, J. L. & Newstead, S. Structural basis for amino acid transport by the CAT family of SLC7 transporters. Nat. Commun. 9, 550 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  18. Yan, R., Zhao, X., Lei, J. & Zhou, Q. Structure of the human LAT1-4F2hc heteromeric amino acid transporter complex. Nature 568, 127–130 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Lee, Y. et al. Cryo-EM structure of the human l-type amino acid transporter 1 in complex with glycoprotein CD98hc. Nat. Struct. Mol. Biol. 26, 510–517 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Gagnon, D. G., Bissonnette, P. & Lapointe, J. Y. Identification of a disulfide bridge linking the fourth and the seventh extracellular loops of the Na+/glucose cotransporter. J. Gen. Physiol. 127, 145–158 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wright, E. M., Turk, E. & Martin, M. G. Molecular basis for glucose-galactose malabsorption. Cell Biochem. Biophys. 36, 115–121 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Santer, R. et al. Molecular analysis of the SGLT2 gene in patients with renal glucosuria. J. Am. Soc. Nephrol. 14, 2873–2882 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Calado, J. et al. Twenty-one additional cases of familial renal glucosuria: absence of genetic heterogeneity, high prevalence of private mutations and further evidence of volume depletion. Nephrol. Dial. Transplant. 23, 3874–3879 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Bhattacharya, S. et al. An exhaustive perspective on structural insights of SGLT2 inhibitors: a novel class of antidiabetic agent. Eur. J. Med. Chem. 204, 112523 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Turner, R. J. & Silverman, M. Sugar uptake into brush border vesicles from normal human kidney. Proc. Natl Acad. Sci. USA 74, 2825–2829 (1977).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Coady, M. J., Wallendorff, B. & Lapointe, J. Y. Characterization of the transport activity of SGLT2/MAP17, the renal low-affinity Na+-glucose cotransporter. Am. J. Physiol. Renal Physiol. 313, F467–F474 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Abramson, J. & Wright, E. M. Structure and function of Na+-symporters with inverted repeats. Curr. Opin. Struct. Biol. 19, 425–432 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Penmatsa, A. & Gouaux, E. How LeuT shapes our understanding of the mechanisms of sodium-coupled neurotransmitter transporters. J. Physiol. 592, 863–869 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Forrest, L. R. & Rudnick, G. The rocking bundle: a mechanism for ion-coupled solute flux by symmetrical transporters. Physiology 24, 377–386 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Penmatsa, A., Wang, K. H. & Gouaux, E. X-ray structure of dopamine transporter elucidates antidepressant mechanism. Nature 503, 85–90 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Coleman, J. A., Green, E. M. & Gouaux, E. X-ray structures and mechanism of the human serotonin transporter. Nature 532, 334–339 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li, N. et al. Structure of a pancreatic ATP-sensitive potassium channel. Cell 168, 101–110.e10 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Scheich, C., Kummel, D., Soumailakakis, D., Heinemann, U. & Bussow, K. Vectors for co-expression of an unrestricted number of proteins. Nucleic Acids Res. 35, e43 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Goehring, A. et al. Screening and large-scale expression of membrane proteins in mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nasr, M. L. et al. Covalently circularized nanodiscs for studying membrane proteins and viral entry. Nat. Methods 14, 49–52 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Hummel, C. S. et al. Glucose transport by human renal Na+/d-glucose cotransporters SGLT1 and SGLT2. Am. J. Physiol. Cell Physiol. 300, C14–C21 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. 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 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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 

  40. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, S. et al. High-resolution noise substitution to measure overfitting and validate resolution in 3D structure determination by single particle electron cryomicroscopy. Ultramicroscopy 135, 24–35 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Biasini, M. et al. SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Res. 42, W252–W258 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Ye and W. Zhang from Boehringer Ingelheim International GmbH for assistance; L.-C. Hsu from the School of Pharmacy, National Taiwan University for providing the 1-NBD-glucose sample for our initial pilot experiment; and S. Huang for assistance with the radioactive experiments. Cryo-EM data collection was supported by the Electron Microscopy Laboratory and Cryo-EM Platform of Peking University with the assistance of X. Li, Z. Guo, B. Shao, X. Pei and G. Wang. Part of the structural computation was also performed on the Computing Platform of the Center for Life Science and High-performance Computing Platform of Peking University. We thank the National Center for Protein Sciences at Peking University in Beijing, China, for assistance with negative-stain EM. The work is supported by grants from the National Natural Science Foundation of China (91957201 and 31870833 to L.C., 31821091 to L.C. and Z.C., and 31971375 to Z.C.) and Beijing Municipal Science & Technology Commission Project (Z201100005320017 to Z.C.). C.G. was partially supported by a postdoctoral fellowship sponsored by Boehringer Ingelheim International GmbH.

Author information

Authors and Affiliations

Authors

Contributions

L.C. initiated the project and wrote the draft of the manuscript. C.G. screened the SGLT homologues. Y.N. purified protein, prepared the cryo-EM sample, processed data and carried out functional assays. Y.N. and R.L. collected the cryo-EM data. L.C. built and refined the model. Y.Z. and Z.C. synthesized 1-NBD-glucose. S.H. and H.N. provided empagliflozin and [3H]-empagliflozin. All authors contributed to the preparation of the manuscript.

Corresponding author

Correspondence to Lei Chen.

Ethics declarations

Competing interests

S.H. and H.N. are employees of Boehringer Ingelheim Pharma, GmbH & Co KG.

Additional information

Peer review information Nature thanks David Drew and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Biochemical characterization of the human SGLT2-MAP17 heterodimer.

a, FSEC traces of GFP-tagged hSGLT2 (red) alone, in complex with non-tagged MAP17 (green) and in complex with C-terminal MBP-mScarlet tagged MAP17 (blue). The fluorescence signals were monitored in the GFP channel. b, HPLC chromatograph of synthesized 1-NBD-glucose, showing high purity. ESI-MS: m/z calculated for C12H14N4O8 [M+H]+ 343.09, found 343.18. c, [3H]-empagliflozin binding to the purified hSGLT2-MAP17 protein. Unlabeled empagliflozin was used for competition. CPM, counts per minute. (Data are shown as means ± standard deviations, n = 3 biologically independent samples). d, Size-exclusion chromatography profile of the hSGLT2-MAP17 complex in nanodisc in the presence of empagliflozin. Fractions between dashes were used for cryo-EM sample preparation. e, SDS-PAGE gel of purified hSGLT2-MAP17 protein in the nanodisc. Asterisk indicates the fraction for cryo-EM sample preparation. The purifications were repeated 3 times with similar results. (For gel source data, see Supplementary Fig. 1.).

Source data

Extended Data Fig. 2 Cryo-EM image analysis of the hSGLT2-MAP17 complex.

a, Representative raw micrograph of hSGLT2-MAP17 complex in nanodisc. The other 7899 micrographs are similar to this one. b, 2D class averages of hSGLT2-MAP17 complex output from cryoSPARC. c, Data processing flow chart. d, The locations of GFP (green) and nanobody (brown) shown in 4 Å low-pass filtered cryo-EM map. e, Gold-standard Fourier shell correlation curves of the final reconstruction. f, Angular distributions of the final reconstruction. g, Local resolution distribution of the hSGLT2-MAP17 complex. h, The cut-open view of local resolution distribution of the hSGLT2-MAP17 complex.

Source data

Extended Data Fig. 3 Representative cryo-EM density maps of the hSGLT2-MAP17 complex.

The densities of transmembrane helices, empagliflozin, and putative palmitoylated Cys residues are contoured at 1.3 σ, 1.0 σ and 1.0 σ, respectively.

Extended Data Fig. 4 Sequence alignment of SGLT.

a, The sequences of the Homo sapiens SGLT2 (hSGLT2), Mus Musculus SGLT2 (mSGLT2), Homo sapiens SGLT1 (hSGLT1), and Vibrio parahaemolyticus SGLT (vSGLT) are aligned using MEGA-X. α helices are shown as cylinders. Unmodeled residues are shown as dashed lines. Conserved residues are colored from cyan to blue. Residues that form disulfide bonds are highlighted in yellow. FRG mutations on extracellular protrusion are indicated by asterisks. The residues that are mutated in Fig. 3g are boxed in red. The colors of cylinders are the same as in Fig. 1d. b, The sequences of transmembrane helices of Homo sapiens MAP17, Homo sapiens SMIM24 are aligned using MEGA-X. Conserved residues are colored from cyan to blue. Residues that interact with hSGLT2 are boxed in red. c, The superposition of hSGLT2 (colored) with vSGLT (gray). The scaffold domain of hSGLT2 is colored in green. The bundle helices of TM1, TM2, TM6 and TM7 of hSGLT2 are colored in brown, blue, pink and red respectively. d, A 90° rotated view of c. e, Interactions between hydroxyl groups of empagliflozin and hSGLT2. The helices of hSGLT2 are colored the same as in c. Empagliflozin and its interacting residues are shown as sticks. Putative hydrogen bonds are depicted as black dashed lines. f, Interactions of D-galactose with vSGLT. The helices of vSGLT are numbered according to LeuT nomenclature for comparison. D-galactose and its interacting residues are shown as sticks. Hydrogen bonds are depicted as black dashed lines. g, Top view of the cross-section of the transmembrane domain of the hSGLT2-MAP17 complex at the approximate level indicated by the dashed lines in Fig. 1c, colored the same as in Fig. 1c. The numbers of transmembrane helices are labeled above. h, Top view of the cross-section of the transmembrane domain of the GkApcT-MgtS complex. GkApcT is colored in grey and MgtS is colored in yellow. i, Top view of the cross-section of the transmembrane domain of the LAT1-4F2hc complex. LAT1 is colored in grey and the transmembrane helix of 4F2hc is colored in yellow.

Extended Data Fig. 5 Distributions of human disease mutations.

a, hSGLT1 mutations found in glucose galactose malabsorption human patients are mapped onto the homology structural model of hSGLT1. Cα positions of mutations are shown as purple spheres. hSGLT1 is shown in blue ribbons. b, A 180° rotated view of a. c, hSGLT2 mutations found in familial renal glycosuria patients are mapped onto the structure of hSGLT2. Cα positions of mutations are shown as purple spheres. hSGLT2 is shown in brown ribbons. d, A 180° rotated view of c.

Extended Data Fig. 6 Chemical structures of related compounds.

a, Chemical structures of glucose and its various analogues. Carbons of D-glucose are numbered in blue. The C2-OH and C3-OH groups of glucose that are important for hSGLT2 binding are shown in red dashed circles. b, Chemical structures of representative SGLT2i.

Extended Data Fig. 7 Electron density map of hSGLT2 at the putative sodium-binding site.

a, The electron density map of hSGLT2 is contoured at 1.8 σ. b, a 180° rotated view of a.

Extended Data Fig. 8 The model of hSGLT2 inhibition by SGLT2i.

a, hSGLT2 is shown in blue. MAP17 is shown in red. SGLT2i is shown in green. Glucose is shown in yellow. Sodium ions are shown as purple spheres. b, The cut-open view of hSGLT2 shown with the surface colored in pink. Empagliflozin is shown as green spheres. c, The cut-open view of SERT (PDB ID: 5I73) with the surface colored in brown. (S)-citaloprams are shown as green spheres. d, The cut-open view of DAT (PDB ID: 4M48), with the surface colored in red. Nortriptyline is shown as green spheres.

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics
Extended Data Table 2 The potencies of empagliflozin on various hSGLT2 constructs

Supplementary information

Supplementary Fig. 1

The uncropped gel of Extended Data Fig. 1e with size marker indicated.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Niu, Y., Liu, R., Guan, C. et al. Structural basis of inhibition of the human SGLT2–MAP17 glucose transporter. Nature 601, 280–284 (2022). https://doi.org/10.1038/s41586-021-04212-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04212-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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