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 common mechanism of Sec61 translocon inhibition by small molecules

Abstract

The Sec61 complex forms a protein-conducting channel in the endoplasmic reticulum membrane that is required for secretion of soluble proteins and production of many membrane proteins. Several natural and synthetic small molecules specifically inhibit Sec61, generating cellular effects that are useful for therapeutic purposes, but their inhibitory mechanisms remain unclear. Here we present near-atomic-resolution structures of human Sec61 inhibited by a comprehensive panel of structurally distinct small molecules—cotransin, decatransin, apratoxin, ipomoeassin, mycolactone, cyclotriazadisulfonamide and eeyarestatin. All inhibitors bind to a common lipid-exposed pocket formed by the partially open lateral gate and plug domain of Sec61. Mutations conferring resistance to the inhibitors are clustered at this binding pocket. The structures indicate that Sec61 inhibitors stabilize the plug domain in a closed state, thereby preventing the protein-translocation pore from opening. Our study provides the atomic details of Sec61–inhibitor interactions and the structural framework for further pharmacological studies and drug design.

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: Cryo-EM structures of the human Sec61 complex inhibited by various small-molecule inhibitors.
Fig. 2: Structural plasticity of the inhibitor-binding pocket.
Fig. 3: Maps for interactions between Sec61 and inhibitors.
Fig. 4: Inhibitor-resistant mutations.
Fig. 5: Proposed model for Sec61 inhibition.

Similar content being viewed by others

Data availability

EM maps and models are available through EM Data Bank (EMDB) and Protein Data Bank (PDB) under the following accession codes: EMD-27581 and PDB-8DNV for the apo class 1 structure, EMD-27582 and PDB-8DNW for the apo class 2 structure, EMD-27583 and PDB-8DNX for the cotransin CP2-bound complex, EMD-27584 and PDB-8DNY for the decatransin-bound complex, EMD-27585 and PDB-8DNZ for the apratoxin F-bound complex, EMD-27586 and PDB-8DO0 for the mycolactone bound complex, EMD-27587 and PDB-8DO1 for ipomoeassin F-bound complex, EMD-27588 and PDB-8DO2 for the CADA-bound complex, and EMD-27589 and PDB-8DO3 for the eeyarestatin I-bound complex. Additional full Sec complex maps were also deposited to EMDB (for accession codes, see Supplementary Table 1). Source data are provided with this paper.

References

  1. Itskanov, S. et al. Mechanism of protein translocation by the Sec61 translocon complex. Cold Spring Harb. Perspect. Biol. 15, a041250 (2023).

    CAS  PubMed  Google Scholar 

  2. Van den Berg, B. et al. X-ray structure of a protein-conducting channel. Nature 427, 36–44 (2004).

    PubMed  Google Scholar 

  3. Deshaies, R. J. et al. Assembly of yeast Sec proteins involved in translocation into the endoplasmic reticulum into a membrane-bound multisubunit complex. Nature 349, 806–808 (1991).

    CAS  PubMed  Google Scholar 

  4. Panzner, S. et al. Posttranslational protein transport in yeast reconstituted with a purified complex of Sec proteins and Kar2p. Cell 81, 561–570 (1995).

    CAS  PubMed  Google Scholar 

  5. Egea, P. F. et al. Lateral opening of a translocon upon entry of protein suggests the mechanism of insertion into membranes. Proc. Natl Acad. Sci. USA 107, 17182–17187 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Park, E. et al. Structure of the SecY channel during initiation of protein translocation. Nature 506, 102–106 (2014).

    CAS  PubMed  Google Scholar 

  7. Voorhees, R. M. et al. Structure of the mammalian ribosome-Sec61 complex to 3.4 Å resolution. Cell 157, 1632–1643 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gogala, M. et al. Structures of the Sec61 complex engaged in nascent peptide translocation or membrane insertion. Nature 506, 107–110 (2014).

    CAS  PubMed  Google Scholar 

  9. Voorhees, R. M. et al. Structure of the Sec61 channel opened by a signal sequence. Science 351, 88–91 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, L. et al. Crystal structure of a substrate-engaged SecY protein-translocation channel. Nature 531, 395–399 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Itskanov, S. et al. Structure of the posttranslational Sec protein-translocation channel complex from yeast. Science 363, 84–87 (2019).

    CAS  PubMed  Google Scholar 

  12. Wu, X. et al. Structure of the post-translational protein translocation machinery of the ER membrane. Nature 566, 136–139 (2019).

    CAS  PubMed  Google Scholar 

  13. Itskanov, S. et al. Stepwise gating of the Sec61 protein-conducting channel by Sec63 and Sec62. Nat. Struct. Mol. Biol. 28, 162–172 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Weng, T. H. et al. Architecture of the active post-translational Sec translocon. EMBO J. 40, e105643 (2021).

    CAS  PubMed  Google Scholar 

  15. Pauwels, E. et al. Inhibitors of the Sec61 complex and novel high throughput screening strategies to target the protein translocation pathway. Int. J. Mol. Sci. 22, 12007 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Luesch, H. et al. Natural products as modulators of eukaryotic protein secretion. Nat. Prod. Rep. 37, 717–736 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Guenin-Mace, L. et al. Shaping mycolactone for therapeutic use against inflammatory disorders. Sci. Transl. Med. 7, 289ra285 (2015).

    Google Scholar 

  18. Heaton, N. S. et al. Targeting viral proteostasis limits influenza virus, HIV, and dengue virus infection. Immunity 44, 46–58 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Vermeire, K. et al. CADA inhibits human immunodeficiency virus and human herpesvirus 7 replication by down-modulation of the cellular CD4 receptor. Virology 302, 342–353 (2002).

    CAS  PubMed  Google Scholar 

  20. O’Keefe, S. et al. Ipomoeassin-F inhibits the in vitro biogenesis of the SARS-CoV-2 spike protein and its host cell membrane receptor. J. Cell Sci. 134, jcs257758 (2021).

    PubMed  PubMed Central  Google Scholar 

  21. Lowe, E. et al. Preclinical evaluation of KZR-261, a novel small molecule inhibitor of Sec61. J. Clin. Oncol. 38, 3582–3582 (2020).

    Google Scholar 

  22. Besemer, J. et al. Selective inhibition of cotranslational translocation of vascular cell adhesion molecule 1. Nature 436, 290–293 (2005).

    CAS  PubMed  Google Scholar 

  23. Garrison, J. L. et al. A substrate-specific inhibitor of protein translocation into the endoplasmic reticulum. Nature 436, 285–289 (2005).

    CAS  PubMed  Google Scholar 

  24. MacKinnon, A. L. et al. Photo-leucine incorporation reveals the target of a cyclodepsipeptide inhibitor of cotranslational translocation. J. Am. Chem. Soc. 129, 14560–14561 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Junne, T. et al. Decatransin, a new natural product inhibiting protein translocation at the Sec61/SecYEG translocon. J. Cell Sci. 128, 1217–1229 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Paatero, A. O. et al. Apratoxin kills cells by direct blockade of the Sec61 protein translocation channel. Cell Chem. Biol. 23, 561–566 (2016).

    CAS  PubMed  Google Scholar 

  27. McKenna, M. et al. Mechanistic insights into the inhibition of Sec61-dependent co- and post-translational translocation by mycolactone. J. Cell Sci. 129, 1404–1415 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Baron, L. et al. Mycolactone subverts immunity by selectively blocking the Sec61 translocon. J. Exp. Med. 213, 2885–2896 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Zong, G. et al. Ipomoeassin F binds Sec61alpha to inhibit protein translocation. J. Am. Chem. Soc. 141, 8450–8461 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Tranter, D. et al. Coibamide A targets Sec61 to prevent biogenesis of secretory and membrane proteins. ACS Chem. Biol. 15, 2125–2136 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Vermeire, K. et al. Signal peptide-binding drug as a selective inhibitor of co-translational protein translocation. PLoS Biol. 12, e1002011 (2014).

    PubMed  PubMed Central  Google Scholar 

  32. Cross, B. C. et al. Eeyarestatin I inhibits Sec61-mediated protein translocation at the endoplasmic reticulum. J. Cell Sci. 122, 4393–4400 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Pauwels, E. et al. A proteomic study on the membrane protein fraction of T cells confirms high substrate selectivity for the ER translocation inhibitor cyclotriazadisulfonamide. Mol. Cell Proteom. 20, 100144 (2021).

    CAS  Google Scholar 

  34. Mackinnon, A. L. et al. An allosteric Sec61 inhibitor traps nascent transmembrane helices at the lateral gate. eLife 3, e01483 (2014).

    PubMed  PubMed Central  Google Scholar 

  35. Gerard, S. F. et al. Structure of the inhibited state of the sec translocon. Mol. Cell 79, 406–415 e407 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Rehan, S. et al. Signal peptide mimicry primes Sec61 for client-selective inhibition. Preprint at bioRxiv https://doi.org/10.1101/2022.07.03.498529 (2022).

  37. Pauwels, E. et al. Structural insights into TRAP association with ribosome-Sec61 complex and translocon inhibition by a CADA derivative. Sci. Adv. 9, eadf0797 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Carlson, M. L. et al. The Peptidisc, a simple method for stabilizing membrane proteins in detergent-free solution. eLife 7, e34085 (2018).

    PubMed  PubMed Central  Google Scholar 

  39. Hommel, U. et al. The 3D-structure of a natural inhibitor of cell adhesion molecule expression. FEBS Lett. 379, 69–73 (1996).

    CAS  PubMed  Google Scholar 

  40. Luesch, H. et al. Total structure determination of apratoxin A, a potent novel cytotoxin from the marine cyanobacterium Lyngbya majuscula. J. Am. Chem. Soc. 123, 5418–5423 (2001).

    CAS  PubMed  Google Scholar 

  41. Trueman, S. F. et al. A gating motif in the translocation channel sets the hydrophobicity threshold for signal sequence function. J. Cell Biol. 199, 907–918 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Smith, M. A. et al. Modeling the effects of prl mutations on the Escherichia coli SecY complex. J. Bacteriol. 187, 6454–6465 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Junne, T. et al. Mutations in the Sec61p channel affecting signal sequence recognition and membrane protein topology. J. Biol. Chem. 282, 33201–33209 (2007).

    CAS  PubMed  Google Scholar 

  44. Klein, W. et al. Defining a conformational consensus motif in cotransin-sensitive signal sequences: a proteomic and site-directed mutagenesis study. PLoS ONE 10, e0120886 (2015).

    PubMed  PubMed Central  Google Scholar 

  45. Van Puyenbroeck, V. et al. Preprotein signature for full susceptibility to the co-translational translocation inhibitor cyclotriazadisulfonamide. Traffic 21, 250–264 (2020).

    PubMed  Google Scholar 

  46. Fessl, T. et al. Dynamic action of the Sec machinery during initiation, protein translocation and termination. eLife 7, e35112 (2018).

    PubMed  PubMed Central  Google Scholar 

  47. Mercier, E. et al. Lateral gate dynamics of the bacterial translocon during cotranslational membrane protein insertion. Proc. Natl Acad. Sci. USA 118, e2100474118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Bhadra, P. et al. Mycolactone enhances the Ca2+ leak from endoplasmic reticulum by trapping Sec61 translocons in a Ca2+ permeable state. Biochem. J. 478, 4005–4024 (2021).

    CAS  PubMed  Google Scholar 

  49. Gamayun, I. et al. Eeyarestatin compounds selectively enhance Sec61-mediated Ca2+ leakage from the endoplasmic reticulum. Cell Chem. Biol. 26, 571–583 e576 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Xiao, L. Synthetic Apratoxin F and Novel AnaloguesMolecules for Anticancer Mechanistic and Therapeutic Applications. Dissertation, The Ohio State University (2017).

  51. Zong, G. et al. Total synthesis and biological evaluation of ipomoeassin F and its unnatural 11R-epimer. J. Org. Chem. 80, 9279–9291 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Chany, A. C. et al. A diverted total synthesis of mycolactone analogues: an insight into Buruli ulcer toxins. Chemistry 17, 14413–14419 (2011).

    CAS  PubMed  Google Scholar 

  53. Lee, M. E. et al. A highly characterized yeast toolkit for modular, multipart assembly. ACS Synth. Biol. 4, 975–986 (2015).

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  55. Tegunov, D. et al. Real-time cryo-electron microscopy data preprocessing with Warp. Nat. Methods 16, 1146–1152 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Punjani, A. et al. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    CAS  PubMed  Google Scholar 

  57. Emsley, P. et al. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D 74, 531–544 (2018).

    CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  60. Pilon, M. et al. Sec61p mediates export of a misfolded secretory protein from the endoplasmic reticulum to the cytosol for degradation. EMBO J. 16, 4540–4548 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Hoepfner, D. et al. Selective and specific inhibition of the Plasmodium falciparum lysyl-tRNA synthetase by the fungal secondary metabolite cladosporin. Cell Host Microbe 11, 654–663 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Toso for support for electron microscope operation, G. Zong for ipomoeassin F synthesis, P. Mathys and R. Riedl for help acquiring the IC50 data. E.P. was supported by the Vallee Scholars Program, Pew Biomedical Scholars Program and Hellman Fellowship. S.I. and L.W. were supported by a National Institutes of Health training grant (5T32GM008295). M.S and T.J. were supported by the Swiss National Science Foundation (31003A-182519). N.B. was supported by Fondation Raoul Follereau and Fondation Pour Le Développement De La Chimie Des Substances Naturelles Et Ses Applications. W.Q.S. (synthesis of ipomoeassin F) was supported by an AREA grant from National Institutes of Health (GM116032). C.F. and L.X. were supported by the Ohio State University.

Author information

Authors and Affiliations

Authors

Contributions

E.P. conceived the project and supervised the cryo-EM study. L.W. and S.I. cloned the chimeric Sec construct and prepared protein samples. S.I., L.W. and E.P. collected and analyzed cryo-EM data and built atomic models. L.W. performed the human cell-based assays. R.S. helped purification of the human Sec complex and cloning of the chimeric Sec complex. T.J., M.S. and D.H. performed the yeast mutational study. D.H. provided cotransin CP2 and decatransin. C.F. and L.X. provided apratoxin F. W.Q.S. provided ipomoeassin F. N.B. provided mycolactone. All authored contributed to interpretation of results. E.P. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Eunyong Park.

Ethics declarations

Competing interests

During the revision of the manuscript, the Park lab (E.P. and L.W.) signed a sponsored research collaboration agreement with Kezar Life Sciences. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Richard Zimmermann and Karin Römisch for their contribution to the peer review of this work.

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 Cryo-EM analysis of the yeast and human Sec complexes.

a, A schematic of the single-particle cryo-EM analysis of the yeast Sec (ScSec) complex incubated with cotransin CP2. Note that the particles were sorted into two 3D classes, with and without Sec62, due to partial occupancy of Sec62. b, 3D reconstructions of the ScSec complex with and without ScSec62 (shown in yellow). No cotransin-like density was observed in either class. For this experiment, we used a pore ring mutant (PM; M90L/T185I/M294I/M450L) that stabilizes the plug towards a closed conformation. c, Purification of the human Sec (HsSec) complex. Shown is a Superose 6 size-exclusion chromatography elution profile with fractions analyzed on a Coomassie-stained SDS gel. Note that under the used purification condition, HsSec62 does not co-purify at a stoichiometric ratio or stably comigrate with the Sec61–Sec63 complex. The fractions indicated by gray shade were used for cryo-EM. MW standards: Tg, thyroglobulin; F, ferritin; Ald, aldolase. The experiment was repeated twice independently with similar results. d, A schematic of the single-particle analysis of HsSec complex incubated with cotransin CP2. Due to a poor refinement result from nonuniform refinement in cryoSPARC, the final reconstruction was obtained by the ab-initio refinement function of cryoSPARC (see f). e, Representative 2D classes of the HsSec complex. Diffuse cytosolic features of Sec63 (green arrowheads) suggest its flexibility or disorderedness. f, The 3D reconstruction of the HsSec complex. A putative cotransin CP2 feature (cyan) is visible at the lateral gate.

Source data

Extended Data Fig. 2 Cryo-EM analysis of the chimeric Sec complex in an apo form.

a, Purification of the chimeric Sec complex reconstituted in a peptidisc. Left, Superose 6 elution profile; right, Coomassie-stained SDS gel of the peak fraction. The fraction marked by gray shade was used for cryo-EM. Asterisks, putative species of glycosylated ScSec71. The experiment was repeated at least four times independently with similar results. b, A schematic of the cryo-EM analysis of the chimeric Sec complex in an apo state. c and d, Distributions of particle view orientations in the final reconstructions of Classes 1 (c) and 2 (d). e and f, Fourier shell correlation (FSC) curves and local resolution maps of the final reconstructions. g, Superimposition of the Class 1 and 2 atomic models (based on the cytosolic domains) shows a slight difference in relative positions between Sec63–Sec71–Sec72 and the Sec61 complex. h, Side views showing the contact between the engineered cytosolic loops of Sec61α and the FN3 domain of ScSec63. Note that in Apo Class 2, the contact is more poorly packed than Class 1.

Source data

Extended Data Fig. 3 Cryo-EM analysis of the chimeric Sec complex in an inhibitor (apratoxin F)-bound form.

a, Images of a representative micrograph and particles of the apratoxin F-bound chimeric Sec complex. Scale bar, 10 nm. b, A schematic of the cryo-EM analysis of the apratoxin F-bound chimeric Sec complex. c, Representative 2D classes of the apratoxin F-bound Sec complex. d, Distribution of particle view orientations in the final reconstruction. e, The FSC curve and local resolution map of the final reconstruction (full Sec complex map). f, As in e, but for the map from focused (local) refinement. g, Segmented density maps of the apratoxin F-bound Sec61α subunit. h, Segmented density features of bound natural inhibitors.

Extended Data Fig. 4 FSC curve and local resolution maps of inhibitor-bound Sec complexes.

As in Extended Data Fig. 3e and f, but for all other inhibitor-bound structures.

Extended Data Fig. 5 Comparison between the structures of cotransin CP2-bound human and chimeric Sec complexes.

The high-resolution structure of the cotransin CP2-bound chimeric Sec complex (ribbon representation for Sec61 and stick representation for cotransin CP2) is docked into the low-resolution cotransin CP2-bound human Sec complex structure (the semi-transparent gray density map; also see Extended Data Fig. 1f). The features of Sec61α and the bound cotransin CP2 are essentially superimposable between the two structures. Dashed lines indicate lateral gate helices (TM2b, TM3, and TM7).

Extended Data Fig. 6 Variation in the extent of lateral gate opening in inhibitor-bound structures.

As in Fig. 2 a and b, but showing other inhibitor-bound structures. In all panels showing a lateral gate comparison, cylindrical representations in red and pink are the cotransin CP2- and ipomoeassin F- bound structures, respectively, whereas the representation in green is the structure with the indicated inhibitor.

Extended Data Fig. 7 Conformational flexibility of the chimeric Sec complex allows ipomoeassin F binding.

Binding of ipomoeassin F causes a narrower opening of the Sec61 lateral gate compared to the apo complex structures (also see Fig. 2), and this is enabled by disengagement of the Sec61 channel from TM3 (Class 1; panel a) or FN3 domain (Class 2; panel b) of Sec63. For comparison, the structures of the apo complex are also shown.

Extended Data Fig. 8 3D maps for interactions between Sec61 and inhibitors.

Shown are stereo-views into the inhibitor-binding site. Inhibitors and adjacent protein side chains are shown in a stick representation together with Cα traces for TM2b, TM3, TM7, and the plug. The views are roughly similar between the different structures but adjusted for each structure for clearer representations. The following colors are used to differentiate parts: brown, pore ring residues; magenta, plug; lighter orange; N300, darker orange, Q127. All inhibitors are shown in cyan with certain atom-dependent coloring (nitrogen-blue, oxygen-red, sulfur-yellow, and chlorine-green).

Extended Data Fig. 9 Generation of HEK293 cell lines with expression of additional SEC61A1 and effects of CADA in CD4 expression.

a, Expression of indicated human Sec61A1 in stable HEK293 (T-Rex-293) cells was confirmed by western-blotting with anti-HA-tag and anti-Sec61A1 antibodies. b, Human CD4 with a C-terminal Strep-tag was expressed in the indicated HEK293 cell lines by transient transfection, and the CD4 expression level after treating cells with the indicated concentrations of CADA was measured by SDS-PAGE and western-blotting. Four replicates were performed, and the dose-response curves are shown in Fig. 4k.

Source data

Extended Data Fig. 10 Comparison with the mycolactone and CK147 structures by others.

a, Chemical structure of mycolactone A/B. b, Structure of mycolactone-bound Sec61 in the current study. c, Structure of mycolactone-bound Sec61 in Gérard et al. (ref. 35). Note that the position and orientation of mycolactone are markedly different between the two structures. The southern chain of mycolactone is buried into the cytosolic funnel of Sec61 in our study, whereas it is in the membrane in the study by Gérard et al. Another notable discrepancy is a one-residue-shifted helical register of the Sec61α TM7 in Gérard et al, which includes the N300 residue. d, Chemical structure of CADA. e, Chemical structure of CK147. f, Structure of CADA-bound Sec61 in the current study. In the lower panel, the surface was clipped along the dashed line shown in the upper panel. g, As in f, but shown is the structure of CK147-bound Sec61 in Pauwels et al. (ref. 36). Note that in this structure, CK147 is almost completely buried within Sec61α. Our CADA-Sec61 structure (f) does not show such a pocket as the space is occupied by the loops flanking the plug helix.

Supplementary information

Supplementary Information

Supplementary Tables 1–3 and Figs. 1 and 2.

Reporting Summary

Supplementary Table

Statistical source data for Supplementary Table 3.

Source data

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Extended Data Fig. 1

Unprocessed gel image of Extended Data Fig. 1c.

Source Data Extended Data Fig. 2

Unprocessed gel image of Extended Data Fig. 2a.

Source Data Extended Data Fig. 9

Unprocessed blot images of Extended Data Fig. 9.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Itskanov, S., Wang, L., Junne, T. et al. A common mechanism of Sec61 translocon inhibition by small molecules. Nat Chem Biol 19, 1063–1071 (2023). https://doi.org/10.1038/s41589-023-01337-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01337-y

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