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.

  • Letter
  • Published:

Cryo-EM structures of a human ABCG2 mutant trapped in ATP-bound and substrate-bound states

Abstract

ABCG2 is a transporter protein of the ATP-binding-cassette (ABC) family that is expressed in the plasma membrane in cells of various tissues and tissue barriers, including the blood–brain, blood–testis and maternal–fetal barriers1,2,3,4. Powered by ATP, it translocates endogenous substrates, affects the pharmacokinetics of many drugs and protects against a wide array of xenobiotics, including anti-cancer drugs5,6,7,8,9,10,11,12. Previous studies have revealed the architecture of ABCG2 and the structural basis of its inhibition by small molecules and antibodies13,14. However, the mechanisms of substrate recognition and ATP-driven transport are unknown. Here we present high-resolution cryo-electron microscopy (cryo-EM) structures of human ABCG2 in a substrate-bound pre-translocation state and an ATP-bound post-translocation state. For both structures, we used a mutant containing a glutamine replacing the catalytic glutamate (ABCG2EQ), which resulted in reduced ATPase and transport rates and facilitated conformational trapping for structural studies. In the substrate-bound state, a single molecule of estrone-3-sulfate (E1S) is bound in a central, hydrophobic and cytoplasm-facing cavity about halfway across the membrane. Only one molecule of E1S can bind in the observed binding mode. In the ATP-bound state, the substrate-binding cavity has collapsed while an external cavity has opened to the extracellular side of the membrane. The ATP-induced conformational changes include rigid-body shifts of the transmembrane domains, pivoting of the nucleotide-binding domains (NBDs), and a change in the relative orientation of the NBD subdomains. Mutagenesis and in vitro characterization of transport and ATPase activities demonstrate the roles of specific residues in substrate recognition, including a leucine residue that forms a ‘plug’ between the two cavities. Our results show how ABCG2 harnesses the energy of ATP binding to extrude E1S and other substrates, and suggest that the size and binding affinity of compounds are important for distinguishing substrates from inhibitors.

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: Structures and transport cycle of ABCG2.
Fig. 2: Substrate-binding cavity and mutant analysis.
Fig. 3: ATP-induced conformational changes.
Fig. 4: Substrate-translocation pathway.

Similar content being viewed by others

Data availability

Atomic coordinates for ABCG2EQ–E1S (including only the variable domain of 5D3-Fab) and ABCG2EQ–ATP were deposited in the Protein Data Bank under accession codes 6HCO and 6HBU, respectively. Electron microscopy data for the two structures were deposited in the Electron Microscopy Data Bank under accession codes EMD-0196 (ABCG2EQ–E1S) and EMD-0190 (ABCG2EQ–ATP). Source Data for Fig. 2e, f and Extended Data Figs. 1e, 2b, d, f and 5 are available online. All other data are available from the corresponding author upon reasonable request. A Life Sciences Reporting Summary for this article is available.

References

  1. Diestra, J. E. et al. Frequent expression of the multi-drug resistance-associated protein BCRP/MXR/ABCP/ABCG2 in human tumours detected by the BXP-21 monoclonal antibody in paraffin-embedded material. J. Pathol. 198, 213–219 (2002).

    Article  CAS  Google Scholar 

  2. Fetsch, P. A. et al. Localization of the ABCG2 mitoxantrone resistance-associated protein in normal tissues. Cancer Lett. 235, 84–92 (2006).

    Article  CAS  Google Scholar 

  3. Robey, R. W. et al. ABCG2: a perspective. Adv. Drug Deliv. Rev. 61, 3–13 (2009).

    Article  CAS  Google Scholar 

  4. Vlaming, M. L., Lagas, J. S. & Schinkel, A. H. Physiological and pharmacological roles of ABCG2 (BCRP): recent findings in Abcg2 knockout mice. Adv. Drug Deliv. Rev. 61, 14–25 (2009).

    Article  CAS  Google Scholar 

  5. Gillet, J. P. & Gottesman, M. M. Advances in the molecular detection of ABC transporters involved in multidrug resistance in cancer. Curr. Pharm. Biotechnol. 12, 686–692 (2011).

    Article  CAS  Google Scholar 

  6. Gottesman, M. M., Fojo, T. & Bates, S. E. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat. Rev. Cancer 2, 48–58 (2002).

    Article  CAS  Google Scholar 

  7. Imai, Y. et al. Breast cancer resistance protein exports sulfated estrogens but not free estrogens. Mol. Pharmacol. 64, 610–618 (2003).

    Article  CAS  Google Scholar 

  8. Ishikawa, T., Aw, W. & Kaneko, K. Metabolic interactions of purine derivatives with human ABC transporter ABCG2: genetic testing to assess gout risk. Pharmaceuticals 6, 1347–1360 (2013).

    Article  Google Scholar 

  9. Mao, Q. & Unadkat, J. D. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport—an update. AAPS J. 17, 65–82 (2015).

    Article  CAS  Google Scholar 

  10. Mo, W. & Zhang, J. T. Human ABCG2: structure, function, and its role in multidrug resistance. Int. J. Biochem. Mol. Biol. 3, 1–27 (2012).

    CAS  PubMed  Google Scholar 

  11. Sarkadi, B., Homolya, L., Szakács, G. & Váradi, A. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiol. Rev. 86, 1179–1236 (2006).

    Article  CAS  Google Scholar 

  12. Sharom, F. J. The P-glycoprotein multidrug transporter. Essays Biochem. 50, 161–178 (2011).

    Article  CAS  Google Scholar 

  13. Taylor, N. M. I. et al. Structure of the human multidrug transporter ABCG2. Nature 546, 504–509 (2017).

    ADS  CAS  PubMed  Google Scholar 

  14. Jackson, S. M. et al. Structural basis of small-molecule inhibition of human multidrug transporter ABCG2. Nat. Struct. Mol. Biol. 25, 333–340 (2018).

    Article  CAS  Google Scholar 

  15. Zhou, S. et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat. Med. 7, 1028–1034 (2001).

    Article  CAS  Google Scholar 

  16. Suzuki, M., Suzuki, H., Sugimoto, Y. & Sugiyama, Y. ABCG2 transports sulfated conjugates of steroids and xenobiotics. J. Biol. Chem. 278, 22644–22649 (2003).

    Article  CAS  Google Scholar 

  17. Lee, J. Y. et al. Crystal structure of the human sterol transporter ABCG5/ABCG8. Nature 533, 561–564 (2016).

    Article  ADS  CAS  Google Scholar 

  18. Hanekop, N., Zaitseva, J., Jenewein, S., Holland, I. B. & Schmitt, L. Molecular insights into the mechanism of ATP-hydrolysis by the NBD of the ABC-transporter HlyB. FEBS Lett. 580, 1036–1041 (2006).

    Article  CAS  Google Scholar 

  19. Johnson, Z. L. & Chen, J. ATP binding enables substrate release from multidrug resistance protein 1. Cell 172, 81–89 (2018).

    Article  CAS  Google Scholar 

  20. Kim, Y. & Chen, J. Molecular structure of human P-glycoprotein in the ATP-bound, outward-facing conformation. Science 359, 915–919 (2018).

    Article  ADS  CAS  Google Scholar 

  21. Shintre, C. A. et al. Structures of ABCB10, a human ATP-binding cassette transporter in apo- and nucleotide-bound states. Proc. Natl Acad. Sci. USA 110, 9710–9715 (2013).

    Article  ADS  CAS  Google Scholar 

  22. Zhang, Z., Liu, F. & Chen, J. Conformational changes of CFTR upon phosphorylation and ATP binding. Cell 170, 483–491 (2017).

    Article  CAS  Google Scholar 

  23. Korkhov, V. M., Mireku, S. A. & Locher, K. P. Structure of AMP-PNP-bound vitamin B12 transporter BtuCD-F. Nature 490, 367–372 (2012).

    Article  ADS  CAS  Google Scholar 

  24. Zaitseva, J. et al. A structural analysis of asymmetry required for catalytic activity of an ABC-ATPase domain dimer. EMBO J. 25, 3432–3443 (2006).

    Article  CAS  Google Scholar 

  25. Aller, S. G. et al. Structure of P-glycoprotein reveals a molecular basis for poly-specific drug binding. Science 323, 1718–1722 (2009).

    Article  ADS  CAS  Google Scholar 

  26. Johnson, Z. L. & Chen, J. Structural basis of substrate recognition by the multidrug resistance protein MRP1. Cell 168, 1075–1085 (2017).

    Article  CAS  Google Scholar 

  27. Alam, A. et al. Structure of a zosuquidar and UIC2-bound human-mouse chimeric ABCB1. Proc. Natl Acad. Sci. USA 115, E1973–E1982 (2018).

    Article  CAS  Google Scholar 

  28. Choudhury, H. G. et al. Structure of an antibacterial peptide ATP-binding cassette transporter in a novel outward occluded state. Proc. Natl Acad. Sci. USA 111, 9145–9150 (2014).

    Article  ADS  CAS  Google Scholar 

  29. Hohl, M., Briand, C., Grütter, M. G. & Seeger, M. A. Crystal structure of a heterodimeric ABC transporter in its inward-facing conformation. Nat. Struct. Mol. Biol. 19, 395–402 (2012).

    Article  CAS  Google Scholar 

  30. Verhalen, B. et al. Energy transduction and alternating access of the mammalian ABC transporter P-glycoprotein. Nature 543, 738–741 (2017).

    Article  ADS  CAS  Google Scholar 

  31. Ritchie, T. K. et al. Chapter 11—reconstitution of membrane proteins in phospholipid bilayer nanodiscs. Methods Enzymol. 464, 211–231 (2009).

    Article  CAS  Google Scholar 

  32. Geertsma, E. R., Nik Mahmood, N. A., Schuurman-Wolters, G. K. & Poolman, B. Membrane reconstitution of ABC transporters and assays of translocator function. Nat. Protocols 3, 256–266 (2008).

    Article  CAS  Google Scholar 

  33. Schaffner, W. & Weissmann, C. A rapid, sensitive, and specific method for the determination of protein in dilute solution. Anal. Biochem. 56, 502–514 (1973).

    Article  CAS  Google Scholar 

  34. Chifflet, S., Torriglia, A., Chiesa, R. & Tolosa, S. A method for the determination of inorganic phosphate in the presence of labile organic phosphate and high concentrations of protein: application to lens ATPases. Anal. Biochem. 168, 1–4 (1988).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  36. Biyani, N. et al. Focus: the interface between data collection and data processing in cryo-EM. J. Struct. Biol. 198, 124–133 (2017).

    Article  CAS  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  Google Scholar 

  38. Rohou, A. & Grigorieff, N. CTFFIND4: Fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  Google Scholar 

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

    Article  ADS  CAS  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  Google Scholar 

  41. Kimanius, D., Forsberg, B. O., Scheres, S. H. & Lindahl, E. Accelerated cryo-EM structure determination with parallelisation using GPUs in RELION-2. eLife 5, e18722 (2016).

    Article  Google Scholar 

  42. Rosenthal, P. B. & Henderson, R. Optimal determination of particle orientation, absolute hand, and contrast loss in single-particle electron cryomicroscopy. J. Mol. Biol. 333, 721–745 (2003).

    Article  CAS  Google Scholar 

  43. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    Article  CAS  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  Google Scholar 

  45. 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 65, 1074–1080 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  47. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by the Swiss National Science Foundation through the National Centre of Competence in Research (NCCR) TransCure and by a Swiss Federal Institute of Technology Zurich (ETH Zurich) research grant (ETH-22-14-1). N.M.I.T. was also supported by the Research Fund Junior Researchers of the University of Basel. J.K. was also supported by the TransCure Young Investigator Award (2017). Cryo-EM data were collected at C-CINA, University of Basel; we thank K. Goldie, L. Kováčik and A. Fecteau-Lefebvre for technical support. We thank N. Tremp for help with cell culture and B. Sorrentino (St Jude Children’s Research Hospital) for providing the 5D3-producing hybridoma cell line.

Reviewer information

Nature thanks H. Mchaourab and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

I.M. expressed and purified wild-type ABCG2 and 5D3-Fab. I.M. and S.M.J. cloned, expressed and purified the ABCG2 mutants. S.M.J. reconstituted ABCG2 into liposomes and lipidic nanodiscs for cryo-EM and functional studies and carried out all functional experiments. J.K. prepared cryo-grids. N.M.I.T. collected cryo-EM data with the assistance of H.S. I.M. processed cryo-EM data of ATP-bound ABCG2 and determined the structure with the assistance of J.K. N.M.I.T. processed electron microscopy data and determined the structure of E1S-bound ABCG2. I.M and K.P.L. built, refined and validated the structures. K.P.L., I.M. and S.M.J. conceived the project, designed the experiments and wrote the manuscript. All authors contributed to revision of the manuscript.

Corresponding authors

Correspondence to Henning Stahlberg or Kaspar P. Locher.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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 Purification, activity and cryo-EM micrographs of ABCG2.

a, Preparative SEC profile (milli absorbance units (mAU) at 280 nm plotted against retention volume (ml)) of the nanodisc-reconstituted ABCG2EQ–E1S complex. The fraction used for cryo-EM grid preparation is indicated by a red asterisk. Inset: reducing (lane 1) and non-reducing (lane 2) SDS–PAGE of the complex, showing bands for ABCG2 (G2), 5D3-Fab (Fab) and nanodisc (ND). b, Preparative SEC profile of the nanodisc-reconstituted ABCG2EQ–ATP complex. The fraction used for cryo-EM grid preparation is indicated by a red asterisk. Inset: reducing (lane 1) and non-reducing (lane 2) SDS–PAGE of the complex, showing bands for ABCG2 (G2) and nanodisc (ND). c, An example micrograph (drift-corrected, dose-weighted and low-pass-filtered to 20 Å) of the nanodisc-reconstituted ABCG2EQ–E1S sample. White scale bar, 50 nm. d, An example micrograph (drift-corrected, dose-weighted and low-pass-filtered to 20 Å) of the nanodisc-reconstituted ABCG2EQ–ATP sample. White scale bar, 50 nm. e, ATPase activities of nanodisc-reconstituted and E1S-transport activities of liposome-reconstituted ABCG2. In both cases, data for wild-type and mutant (E211Q) ABCG2 are shown. The standard deviation from n technical replicates (same batch of nanodiscs or liposomes) is shown.

Source data

Extended Data Fig. 2 Effect of 5D3-Fab on ABCG2 function.

a, Analytical SEC profile of the nanodisc-reconstituted ABCG2EQ–E1S complex in the presence of 5 mM ATP and 5 mM MgCl2. ‘1’ denotes the peak collected. Inset: non-reducing SDS–PAGE of the complex, showing bands for ABCG2 (G2), 5D3-Fab (Fab) and nanodisc (ND). b, ATPase activity of liposome-reconstituted ABCG2, in the presence or absence of 5D3-Fab, and with 0–300 µM E1S. The basal ATPase activity has been normalized (norm) to 0. c, As for b, but with the maximal ATPase activity set to 100%. Each point represents the mean rate derived from technical replicates. For G2 n = 6, except in the case of 0 and 200 µM E1S, for which n = 9. For G2 + Fab, n = 3. d, ATPase activities of ABCG2 in the presence and absence of 5D3-Fab, and either 0 or 50 µM E1S. e, As for d, but with activities in the presence of E1S set to 100%. Bars show means and dots show the rates derived from each technical replicate (same batch of liposomes). Error bars show the standard deviation. f, The EC50 of E1S ATPase stimulation determined using the curves in b and c with the error of the fit (standard deviation) shown. PL, proteoliposome.

Source data

Extended Data Fig. 3 Cryo-EM map generation, data processing and atomic-model refinement of ABCG2EQ–E1S.

a, Twelve representative 2D class averages of the final round of 2D classification, sorted in decreasing order by the number of particles assigned to each class. b, FSC from the CryoSPARC auto-refine procedure of the unmasked half-maps (blue), the half-maps after masking (green), and the half-maps after masking and correction for the influence of the mask (pink). A horizontal dotted line (blue) is drawn for the FSC = 0.143 criterion. For both the unmasked and the corrected FSC curves, their intersection with the FSC = 0.143 and the FSC = 0.5 lines are marked by arrows, and the resolutions at these points are indicated. c, FSC curve of the final 3.58 Å refined model versus the map against it was refined (FSCfull; black line). The FSC curve of the final refined model with introduced shifts (mean value of 0.3 Å) versus the first of two independent half-maps (half-map 1, against which it was refined; FSCwork; green line) or the same model versus the second independent half-map (against which it was not refined; FSChalf2; red line) is also shown. d, Flow chart for cryo-EM data processing and structure determination of the ABCG2EQ–E1S complex. e, Full view of the final CryoSPARC B-factor-sharpened map of ABCG2EQ–E1S, coloured by local resolution in Å, as calculated by ResMap with the clipping plane in the middle of the molecule. f, Angular distribution plot for the final reconstruction.

Extended Data Fig. 4 Fit of the models to the densities.

a, Fit of the TM helices of the final model of the ABCG2EQ–E1S TMD to the post-processed and masked C2 map from CryoSPARC. A region of up to 2 Å around the atoms is shown. b, The fit of one E1S molecule (pink or turquoise sticks) in two possible orientations, flipped by 180°, docked into the C2-symmetrized substrate density of the final model of ABCG2EQ–E1S. The contour level has been reduced by comparison with Fig. 2a to show the strongest density at the core of the polycyclic rings. c, As for b, but showing the fit of one E1S into the electron microscopy density of the post-processed and masked C1 map from CryoSPARC. d, Fit of the TM helices of the final model of the ABCG2EQ–ATP TMD to the post-processed and masked C2 map from RELION. A region of up to 2 Å around the atoms is shown.

Extended Data Fig. 5 Purification and functional analysis of mutants.

a, Analytical SEC profiles of the detergent-purified wild-type and substrate-binding cavity mutants used to make proteoliposomes for functional assays. b, ATPase rates of the liposome-reconstituted wild-type and mutant proteins in the presence of 0–300 µM E1S. Each point represents the mean rate derived from technical replicates (same batch of liposomes) and error bars show the standard deviation. For G2, n = 6, except in the case of 0 and 200 µM E1S, for which n = 9. For the mutants, n = 3. c, Table showing the EC50 of E1S ATPase stimulation determined after normalizing the curves in b with the error of the fit (standard deviation) shown.

Source data

Extended Data Fig. 6 Cryo-EM map generation, data processing and atomic-model refinement of ABCG2EQ–ATP.

a, Twelve representative 2D class averages of the final round of 2D classification, sorted in decreasing order by the number of particles assigned to each class. b, FSC from the RELION auto-refine procedure of the unmasked half-maps (blue), the half-maps after masking (green), and the half-maps after masking and correction for the influence of the mask (pink). A horizontal dotted line (blue) is drawn for the FSC = 0.143 criterion. For both the unmasked and the corrected FSC curves, their intersection with the FSC = 0.143 and the FSC = 0.5 lines are marked by arrows, and the resolutions at these points are indicated. c, FSC curve of the final 3.09 Å refined model versus the map against which it was refined (FSCfull; black line). FSC curves of the final refined model with introduced shifts (mean value of 0.3 Å) versus the first of two independent half-maps (half-map 1, against which it was refined; FSCwork; green line) or the same model versus the second independent half-map (against which it was not refined; FSChalf2; red line) are also shown. d, Flow chart for cryo-EM data processing and structure determination of the ABCG2EQ–ATP complex. The map used for model building is indicated by a red square. e, Full view of the RELION local-resolution-filtered map of ABCG2EQ–ATP, coloured by local resolution in Å as calculated by ResMap, with the clipping plane in the middle of the molecule. f, Angular distribution plot for the final reconstruction.

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Manolaridis, I., Jackson, S.M., Taylor, N.M.I. et al. Cryo-EM structures of a human ABCG2 mutant trapped in ATP-bound and substrate-bound states. Nature 563, 426–430 (2018). https://doi.org/10.1038/s41586-018-0680-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0680-3

Keywords

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