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:

Structure of the bile acid transporter and HBV receptor NTCP

Abstract

Chronic infection with hepatitis B virus (HBV) affects more than 290 million people worldwide, is a major cause of cirrhosis and hepatocellular carcinoma, and results in an estimated 820,000 deaths annually1,2. For HBV infection to be established, a molecular interaction is required between the large glycoproteins of the virus envelope (known as LHBs) and the host entry receptor sodium taurocholate co-transporting polypeptide (NTCP), a sodium-dependent bile acid transporter from the blood to hepatocytes3. However, the molecular basis for the virus–transporter interaction is poorly understood. Here we report the cryo-electron microscopy structures of human, bovine and rat NTCPs in the apo state, which reveal the presence of a tunnel across the membrane and a possible transport route for the substrate. Moreover, the cryo-electron microscopy structure of human NTCP in the presence of the myristoylated preS1 domain of LHBs, together with mutation and transport assays, suggest a binding mode in which preS1 and the substrate compete for the extracellular opening of the tunnel in NTCP. Our preS1 domain interaction analysis enables a mechanistic interpretation of naturally occurring HBV-insusceptible mutations in human NTCP. Together, our findings provide a structural framework for HBV recognition and a mechanistic understanding of sodium-dependent bile acid translocation by mammalian NTCPs.

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: Structure of mammalian NTCP.
Fig. 2: Substrate transport pathway of NTCP.
Fig. 3: Interactions between NTCP and preS1.
Fig. 4: Model of the mode of preS1 binding.

Similar content being viewed by others

Data availability

Cryo-EM maps and structure coordinates have been deposited in the Electron Microscopy Data Bank and the PDB, respectively, under the accession codes EMD-31837 and 7VAD (human NTCP(Q261A)–Fab); EMD-31838 and 7VAE (bovine NTCP(Q261A)–Fab); EMD-31839 and 7VAF (rat NTCP(Q261A)–Fab); EMD-31840 and 7VAG (human NTCP–Fab–myr-preS1); and EMD-32759 and 7WSI (human NTCP(WT)–Fab).

References

  1. The Polaris Observatory Collaborators. Global prevalence, treatment, and prevention of hepatitis B virus infection in 2016: a modelling study. Lancet Gastroenterol. Hepatol. 3, 383–403 (2018).

    Article  Google Scholar 

  2. Global Progress Report on HIV, Viral Hepatitis and Sexually Transmitted Infections (World Health Organization, 2021).

  3. Yan, H. et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. eLife 1, e00049 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Winer, B. Y. & Ploss, A. Determinants of hepatitis B and delta virus host tropism. Curr. Opin. Virol. 13, 109–116 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Watashi, K. & Wakita, T. Hepatitis B virus and Hepatitis D virus entry, species specificity, and tissue tropism. Cold Spring Harb. Perspect. Med. 5, a021378 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Urban, S., Bartenschlager, R., Kubitz, R. & Zoulim, F. Strategies to inhibit entry of HBV and HDV into hepatocytes. Gastroenterology 147, 48–64 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Leistner, C. M., Gruen-Bernhard, S. & Glebe, D. Role of glycosaminoglycans for binding and infection of hepatitis B virus. Cell. Microbiol. 10, 122–133 (2008).

    CAS  PubMed  Google Scholar 

  8. Schulze, A., Gripon, P. & Urban, S. Hepatitis B virus infection initiates with a large surface protein-dependent binding to heparan sulfate proteoglycans. Hepatology 46, 1759–1768 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Ni, Y. et al. Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes. Gastroenterology 146, 1070–1083 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Appelman, M. D., Wettengel, J. M., Protzer, U., Oude Elferink, R. P. J. & van de Graaf, S. F. J. Molecular regulation of the hepatic bile acid uptake transporter and HBV entry receptor NTCP. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 1866, 158960 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Li, M. et al. Apical sodium-dependent bile acid transporter, drug target for bile acid related diseases and delivery target for prodrugs: current and future challenges. Pharmacol. Ther. 212, 107539 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Anwer, M. S. & Stieger, B. Sodium-dependent bile salt transporters of the SLC10A transporter family: more than solute transporters. Pflugers Arch. 466, 77–89 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Li, W. The hepatitis B virus receptor. Annu. Rev. Cell Dev. Biol. 31, 125–147 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Bogomolov, P. et al. Treatment of chronic hepatitis D with the entry inhibitor myrcludex B: first results of a phase Ib/IIa study. J. Hepatol. 65, 490–498 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Passioura, T. et al. De novo macrocyclic peptide inhibitors of hepatitis B virus cellular entry. Cell Chem. Biol. 25, 906–915 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Yan, H. et al. Molecular determinants of hepatitis B and D virus entry restriction in mouse sodium taurocholate cotransporting polypeptide. J. Virol. 87, 7977–7991 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Takeuchi, J. S. et al. A Single adaptive mutation in sodium taurocholate cotransporting polypeptide induced by hepadnaviruses determines virus species specificity. J. Virol. 93, e01432-18 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Jacquet, S. et al. Evolution of hepatitis B virus receptor NTCP reveals differential pathogenicities and species specificities of hepadnaviruses in primates, rodents, and bats. J. Virol. 93, e01738-18 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Muller, S. F., Konig, A., Doring, B., Glebe, D. & Geyer, J. Characterisation of the hepatitis B virus cross-species transmission pattern via Na+/taurocholate co-transporting polypeptides from 11 New World and Old World primate species. PLoS ONE 13, e0199200 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Wang, P. et al. Genetic variations of NTCP are associated with susceptibility to HBV infection and related hepatocellular carcinoma. Oncotarget 8, 105407–105424 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fu, L., Hu, H., Liu, Y., Jing, Z. & Li, W. Woodchuck sodium taurocholate cotransporting polypeptide supports low-level hepatitis B and D virus entry. Virology 505, 1–11 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Saito, S. et al. Catalog of 238 variations among six human genes encoding solute carriers (hSLCs) in the Japanese population. J. Hum. Genet. 47, 576–584 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Zhou, X. et al. Structural basis of the alternating-access mechanism in a bile acid transporter. Nature 505, 569–573 (2014).

    Article  CAS  PubMed  ADS  Google Scholar 

  24. Hu, N. J., Iwata, S., Cameron, A. D. & Drew, D. Crystal structure of a bacterial homologue of the bile acid sodium symporter ASBT. Nature 478, 408–411 (2011).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  25. Lee, C. et al. A two-domain elevator mechanism for sodium/proton antiport. Nature 501, 573–577 (2013).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  26. Fang, S. et al. Molecular mechanism underlying transport and allosteric inhibition of bicarbonate transporter SbtA. Proc. Natl Acad. Sci. USA 118, e2101632118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bruss, V., Hagelsten, J., Gerhardt, E. & Galle, P. R. Myristylation of the large surface protein is required for hepatitis B virus in vitro infectivity. Virology 218, 396–399 (1996).

    Article  CAS  PubMed  Google Scholar 

  28. Gripon, P., Leseyec, J., Rumin, S. & Guguenguillouzo, C. Myristylation of the hepatitis-B virus large surface protein is essential for viral infectivity. Virology 213, 292–299 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Schulze, A., Schieck, A., Ni, Y., Mier, W. & Urban, S. Fine mapping of pre-S sequence requirements for hepatitis B virus large envelope protein-mediated receptor interaction. J. Virol. 84, 1989–2000 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Kirstgen, M. et al. Selective hepatitis B and D virus entry inhibitors from the group of pentacyclic lupane-type betulin-derived triterpenoids. Sci. Rep. 10, 21772 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  31. Grosser, G., Muller, S. F., Kirstgen, M., Doring, B. & Geyer, J. Substrate specificities and Inhibition pattern of the solute carrier family 10 members NTCP, ASBT and SOAT. Front. Mol. Biosci. 8, 689757 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yan, H. et al. Viral entry of hepatitis B and D viruses and bile salts transportation share common molecular determinants on sodium taurocholate cotransporting polypeptide. J. Virol. 88, 3273–3284 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Konig, A. et al. Kinetics of the bile acid transporter and hepatitis B virus receptor Na+/taurocholate cotransporting polypeptide (NTCP) in hepatocytes. J. Hepatol. 61, 867–875 (2014).

    Article  PubMed  CAS  Google Scholar 

  34. Hu, H. H. et al. The rs2296651 (S267F) variant on NTCP (SLC10A1) is inversely associated with chronic hepatitis B and progression to cirrhosis and hepatocellular carcinoma in patients with chronic hepatitis B. Gut 65, 1514–1521 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Peng, L. et al. The p.Ser267Phe variant in SLC10A1 is associated with resistance to chronic hepatitis B. Hepatology 61, 1251–1260 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Zou, T. T., Zhu, Y., Wan, C. M. & Liao, Q. Clinical features of sodium-taurocholate cotransporting polypeptide deficiency in pediatric patients: case series and literature review. Transl. Pediatr. 10, 1045–1054 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Dong, C. et al. Clinical and histopathologic features of sodium taurocholate cotransporting polypeptide deficiency in pediatric patients. Medicine 98, e17305 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Drew, D. & Boudker, O. Shared molecular mechanisms of membrane transporters. Annu. Rev. Biochem. 85, 543–572 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Lu, P. H. et al. Dissecting the conformational dynamics of the bile acid transporter homologue ASBTNM. J. Mol. Biol. 433, 166764 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Noppes, S. et al. Homo- and heterodimerization is a common feature of the solute carrier family SLC10 members. Biol. Chem. 400, 1371–1384 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Bijsmans, I. T., Bouwmeester, R. A., Geyer, J., Faber, K. N. & van de Graaf, S. F. Homo- and hetero-dimeric architecture of the human liver Na+-dependent taurocholate co-transporting protein. Biochem. J. 441, 1007–1015 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Fukano, K. et al. Troglitazone impedes the oligomerization of sodium taurocholate cotransporting polypeptide and entry of hepatitis B virus Into hepatocytes. Front. Microbiol. 9, 3257 (2018).

    Article  PubMed  Google Scholar 

  43. Iwamoto, M. et al. Epidermal growth factor receptor is a host-entry cofactor triggering hepatitis B virus internalization. Proc. Natl Acad. Sci. USA 116, 8487–8492 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jaenecke, F. et al. Generation of conformation-specific antibody fragments for crystallization of the multidrug resistance transporter MdfA. Methods Mol. Biol. 1700, 97–109 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  46. 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 

  47. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

  49. 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  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  51. 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 

  52. 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 

  53. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Tsutsumi, H. Yanagisawa, Y. Kise, Y. Sakamaki, M. Kikkawa, Y. Sugita and D. Im for their help in cryo-EM data collection, T. Tomita for allowing us to use the ultracentrifuge, and S.-Y. Park for discussions. Preliminary cryo-EM screening for antibody as a fiducial marker was performed using a Glacios at the RIKEN SPring-8 Center. This work was supported by a Grant-in-Aid from the Japanese Ministry of Education, Culture, Sports, Science and Technology (grant numbers 19H03164 (U.O.), 19H00976 (T.S.), 18K05334 (N.N.) and 19H00923 (S.I. and N.N.)). This work is partially supported by the Knut and Alice Wallenberg foundation (D.D.), the Platform Project for Supporting Drug Discovery and Life Science Research (Basis for Supporting Innovative Drug Discovery and Life Science Research (BINDS)) from Japan Agency for Medical Research and Development (AMED) under grant numbers JP19am0101115 (support no. 1570, 1846, 1848) and JP21am0101079 (S.I. and N.N.), an AMED Research Program on Emerging and Re-emerging Infectious Disease (20fk0108270h0001) (T.N.), the JST Core Research for Evolutional Science and Technology (JPMJCR20HA) (T.N.), JSPS Core-to-Core Program A (T.N.), the Joint Research Project of the Institute of Medical Science, the University of Tokyo (T.N.) and the Joint Usage/Research Center Program of Institute for Frontier Life and Medical Sciences, Kyoto University (N.N. and T.N.)

Author information

Authors and Affiliations

Authors

Contributions

J.A., N.N. and U.O. designed the experiments. J.A., H.I., Y.N., Y.S., M.O., D.D., N.N. and U.O. prepared recombinant proteins. K.L., T.U., S.I. and N.N. generated antibodies. J.A., K.T.K., Y.F.-F., Z.Z., M.Y., T.N., H.S., N.N. and U.O. performed cryo-EM analyses. J.A. and H.I performed the in vitro preS1 binding assay. K.T.K. performed the transport assay. J.A., D.D., N.N. and U.O. wrote the paper with assistance from all of the authors. S.I., T.S., N.N. and U.O. supervised the project.

Corresponding authors

Correspondence to Norimichi Nomura or Umeharu Ohto.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Christoph Seeger and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 figures and tables

Extended Data Fig. 1 Sequence alignment of NTCP and ASBT.

Sequence alignment of human, bovine, and rat NTCPs and human ASBT, as well as bacterial homologues from Yersinia frederiksenii (ASBTYf) and Neisseria meningitidis (ASBTNM), were calculated using Clustal Omega. Coloured bars indicate locations of the transmembrane helices in the human NTCP. Residues involved in myr-preS1 binding are indicated by red boxes. Residues involved in Na binding (Na1 and Na2 sites) are highlighted in cyan and orange, respectively. Residues near the unmodelled lipid densities in the human NTCP–Fab structure and near the TCA molecule in the ASBTNM structure are highlighted in pink and yellow, respectively. Residues forming TM1 in bASBTs are boxed with black lines.

Extended Data Fig. 2 Sample preparation of mammalian NTCP.

a–c, Representative SEC of human (a), bovine (b), and rat (c) NTCP(Q261A). Absorbance profiles (top) and SDS–PAGE analysis of the peak fractions stained with CBB (bottom) are shown. Pooled fractions are shown as bars. Absorbances at 280 nm and 260 nm are shown as solid and dashed lines, respectively. Purifications were performed four, one, and four times for human, bovine, and rat NTCP, respectively. Uncropped blots are shown in Supplementary Fig. 1.

Extended Data Fig. 3 Cryo-EM analysis of mammalian NTCP–Fab complexes.

a–d, Data processing workflow of cryo-EM analysis of human (a), bovine (b), and rat (c) NTCP–YN69202Fab complexes and human NTCP–YN69202Fab complex with myr-preS1 (d). Representative motion-corrected micrograph (out of 5,434 (a), 2,712 (b), 9,932 (c), and 7,496 (d) total micrographs), 2D class averages, 3D class averages, gold-standard FSC curves of the final 3D reconstruction (resolution cut-off at FSC = 0.143), and the final 3D map (coloured according to the local resolution) are shown. 2D class averages were calculated using refined particles that were used for the final reconstruction. 3D classes selected for the following analyses are indicated with red dotted boxes.

Extended Data Fig. 4 Cryo-EM density maps of mammalian NTCPs in complex with Fab.

a, Overall structures of the human (left), bovine (middle), and rat (right) NTCP–YN69202Fab complexes. Cryo-EM maps (top) and ribbon models (bottom) are shown. NTCP and heavy and light chains of Fab are shown in green, cyan, and blue, respectively. b, Cryo-EM density maps of the rat NTCPYN69202Fab complex around each TM helix of NTCP are shown.

Extended Data Fig. 5 Structural comparison between mammalian NTCP and bASBTs.

a, Structural comparison of human NTCP(Q261A), ASBTYf(E254A) (PDB ID: 4N7X, outward-open), wild-type ASBTYf (PDB ID: 4N7W, inward-open), and wild-type ASBTNM (PDB ID: 3ZUX, inward-open). Each structure was viewed from the extracellular side (top) and parallel to the membrane (bottom). RMSD values of each ASBT structure against the human NTCP structure are indicated (top). b, Structural comparison of the core (left) and panel (right) domains of NTCP and ASBT. Structures of human NTCP(Q261A) (light green), ASBTYf(E254A) (orange), wild-type ASBTYf (cyan), and wild-type ASBTNM (blue) are superposed for each domain. RMSD values of the ASBT structures against the human NTCP structure are indicated (top). c, Close-up view of sodium-binding sites. Na1 (top) and Na2 (bottom) binding sites in the structure of ASBTNM (left) and the corresponding sites of human NTCP (right) are shown. Side chains of residues involved in Na+ binding are labelled and shown as stick representations.

Extended Data Fig. 6 Sodium-dependent transport of NTCP constructs used in this study.

Michaelis–Menten transport kinetics of human NTCP wild type (circle)- and Q261A mutant (triangle)-mediated [3H]-TCA uptake. The specific uptake was calculated by subtracting the sodium-independent and endogenous TCA transport activities from the total uptake, as detailed in Methods. Data represent mean ± standard deviation of three independent measurements. The observed Michaelis constant, Km, for [3H]-TCA is ~9.5 µM (wild type). Estimation of accurate Km value of the Q261A mutant for [3H]-TCA is technically impossible owing to the low transport activity. The Vmax value of the Q261A-mediated uptake was 349.2 ± 103.3 pmol min−1 mg−1, which is fivefold lower than the corresponding value for wild type (1791 ± 154.7 pmol min−1 mg−1).

Source data

Extended Data Fig. 7 Structural comparison between wild-type and Q261A-mutant human NTCP.

a, Representative SEC of human NTCP (WT). Absorbance profiles (top) and SDS–PAGE analysis of the peak fractions stained with CBB (bottom) are shown. Pooled fractions are shown as bars. Absorbances at 280 nm and 260 nm are shown as solid and dashed lines, respectively. Purifications were performed nine times with similar results. The uncropped blot is shown in Supplementary Fig. 1. b, Data processing workflow of cryo-EM analysis of human NTCP (wild type)-YN69202Fab complex. Representative motion-corrected micrograph, 2D class averages, 3D class averages, gold-standard FSC curves of the final 3D reconstruction (resolution cut-off at FSC = 0.143), and the final 3D map (coloured according to the local resolution) are shown. 2D class averages were calculated using refined particles that were used for the final reconstruction. 3D classes selected for the following analyses are indicated with red dotted boxes. c, Structural comparison of wild-type and Q261A-mutant human NTCP. Structures of human NTCP (wild type) (grey) and human NTCP(Q261A) (light green) are superposed (upper left). Na1 (upper right) and Na2 (bottom right) binding sites in the structure of human NTCP (wild type) and human NTCP(Q261A) are shown. Side chains of residues involved in Na+ binding are labelled and shown as stick representations. Close-up view of human NTCP (wild type) marked with a black rectangle is shown (bottom left). The two observed densities are indicated in orange. Side chains of residues near the densities are shown as stick representations. Crossover regions of TM3 and TM8 is indicated by a black dotted circle.

Extended Data Fig. 8 NTCP–preS1 binding interface.

a, Topology diagrams of HBV envelope glycoproteins LHBs. b, Sequences of preS1 mutants used in Fig. 3c. Sequence of wild-type preS1 (248) and the residue numbers are indicated (top). Dots in the sequence indicate the same residues as the wild type. c, Structural comparison of predicted preS1-binding sites, patch 1 (left), and patch 2 (right), among mammalian NTCPs. Superposition of human (light green), bovine (cyan), and rat (magenta) NTCPs with patch 1 (in TM23 loop) and patch 2 (in TM5), indicated with rectangles (top). A detailed view and sequence of each site are shown (bottom).

Extended Data Fig. 9 Cryo-EM densities in the tunnel in NTCP.

Cryo-EM density maps for the human NTCPYN69202Fab complex in the presence (green) and absence (orange) of myr-preS1. Only the densities in the channel are displayed.

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

Asami, J., Kimura, K.T., Fujita-Fujiharu, Y. et al. Structure of the bile acid transporter and HBV receptor NTCP. Nature 606, 1021–1026 (2022). https://doi.org/10.1038/s41586-022-04845-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04845-4

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