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:

HUWE1 employs a giant substrate-binding ring to feed and regulate its HECT E3 domain

Abstract

HUWE1 is a universal quality-control E3 ligase that marks diverse client proteins for proteasomal degradation. Although the giant HECT enzyme is an essential component of the ubiquitin–proteasome system closely linked with severe human diseases, its molecular mechanism is little understood. Here, we present the crystal structure of Nematocida HUWE1, revealing how a single E3 enzyme has specificity for a multitude of unrelated substrates. The protein adopts a remarkable snake-like structure, where the C-terminal HECT domain heads an extended alpha-solenoid body that coils in on itself and houses various protein–protein interaction modules. Our integrative structural analysis shows that this ring structure is highly dynamic, enabling the flexible HECT domain to reach protein targets presented by the various acceptor sites. Together, our data demonstrate how HUWE1 is regulated by its unique structure, adapting a promiscuous E3 ligase to selectively target unassembled orphan proteins.

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: HUWE1N targets orphan proteins for degradation.
Fig. 2: Overall architecture of the giant E3 ligase HUWE1N.
Fig. 3: Functional insertions within the HUWE1N ring.
Fig. 4: Dynamic interfaces stabilize the HUWE1N ring.
Fig. 5: Behavior of HUWE1N in solution.
Fig. 6: Regulation of the HUWE1N HECT domain.

Similar content being viewed by others

Data availability

Coordinates of the HUWE1N crystal structure have been deposited at the Protein Data Bank (PDB) under accession code 7BII. Cryo-EM maps and atomic coordinates have been deposited in the Electron Microscopy Data Bank (EMDB) with accession codes EMD-12318 and EMD-12319, and in the PDB under 7NH1 and 7NH3. Source data are provided with this paper.

References

  1. Ciechanover, A., Orian, A. & Schwartz, A. L. Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays 22, 442–451 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Hochstrasser, M. Lingering mysteries of ubiquitin-chain assembly. Cell 124, 27–34 (2006).

    Article  PubMed  Google Scholar 

  3. Cardozo, T. & Pagano, M. The SCF ubiquitin ligase: insights into a molecular machine. Nat. Rev. Mol. Cell Biol. 5, 739–751 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Alfieri, C., Zhang, S. & Barford, D. Visualizing the complex functions and mechanisms of the anaphase promoting complex/cyclosome (APC/C). Open Biol. 7, 1700204 (2017).

    Article  CAS  Google Scholar 

  5. Kamadurai, H. B. et al. Mechanism of ubiquitin ligation and lysine prioritization by a HECT E3. eLife 2, e00828 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Chen, Z. et al. A tunable brake for HECT ubiquitin ligases. Mol. Cell 66, 345–357 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lorenz, S. Structural mechanisms of HECT-type ubiquitin ligases. Biol. Chem. 399, 127–145 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Kao, S. H., Wu, H. T. & Wu, K. J. Ubiquitination by HUWE1 in tumorigenesis and beyond. J. Biomed. Sci. 25, 67 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Zhong, Q., Gao, W., Du, F. & Wang, X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 121, 1085–1095 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Inoue, S. et al. Mule/Huwe1/Arf-BP1 suppresses Ras-driven tumorigenesis by preventing c-Myc/Miz1-mediated down-regulation of p21 and p15. Genes Dev. 27, 1101–1114 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, D. et al. ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 121, 1071–1083 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Adhikary, S. et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell 123, 409–421 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Urbán, N. et al. Return to quiescence of mouse neural stem cells by degradation of a proactivation protein. Science 353, 292–295 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Hao, Z. et al. K48-linked KLF4 ubiquitination by E3 ligase Mule controls T-cell proliferation and cell cycle progression. Nat. Commun. 8, 14003 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. DeGroot, R. E. A. et al. Huwe1-mediated ubiquitylation of dishevelled defines a negative feedback loop in the wnt signaling pathway. Sci. Signal. 7, ra26 (2014).

    Article  CAS  Google Scholar 

  16. Hall, J. R. et al. Cdc6 stability is regulated by the Huwe1 ubiquitin ligase after DNA damage. Mol. Biol. Cell 18, 3340–3350 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Parsons, J. L. et al. Ubiquitin ligase ARF-BP1/Mule modulates base excision repair. EMBO J. 28, 3207–3215 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang, X. et al. HUWE1 interacts with BRCA1 and promotes its degradation in the ubiquitin-proteasome pathway. Biochem. Biophys. Res. Commun. 444, 549–554 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Xu, Y., Anderson, D. E. & Ye, Y. The HECT domain ubiquitin ligase HUWE1 targets unassembled soluble proteins for degradation. Cell Discov. 2, 16040 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sung, M. K. et al. A conserved quality-control pathway that mediates degradation of unassembled ribosomal proteins. eLife 5, e19105 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Liu, Z., Oughtred, R. & Wing, S. S. Characterization of E3Histone, a novel testis ubiquitin protein ligase which ubiquitinates histones. Mol. Cell. Biol. 25, 2819–2831 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Singh, R. K., Kabbaj, M. H., Paik, J. & Gunjan, A. Histone levels are regulated by phosphorylation and ubiquitylation-dependent proteolysis. Nat. Cell Biol. 11, 925–933 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Deshaies, R. J. Proteotoxic crisis, the ubiquitin-proteasome system, and cancer therapy. BMC Biol. 12, 94 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Oromendia, A. B. & Amon, A. Aneuploidy: implications for protein homeostasis and disease. Dis. Model Mech. 7, 15–20 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Myant, K. B. et al. HUWE1 is a critical colonic tumour suppressor gene that prevents MYC signalling, DNA damage accumulation and tumour initiation. EMBO Mol. Med. 9, 181–197 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Peter, S. et al. Tumor cell-specific inhibition of MYC function using small molecule inhibitors of the HUWE1 ubiquitin ligase. EMBO Mol. Med. 6, 1525–1541 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Froyen, G. et al. Submicroscopic duplications of the hydroxysteroid dehydrogenase HSD17B10 and the E3 ubiquitin ligase HUWE1 are associated with mental retardation. Am. J. Hum. Genet. 82, 432–443 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jäckl, M. et al. β-Sheet augmentation is a conserved mechanism of priming HECT E3 ligases for ubiquitin ligation. J. Mol. Biol. 430, 3218–3233 (2018).

    Article  PubMed  CAS  Google Scholar 

  29. Michel, M. A., Swatek, K. N., Hospenthal, M. K. & Komander, D. Ubiquitin linkage-specific affimers reveal insights into K6-linked ubiquitin signaling. Mol. Cell 68, 233–246 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. White, A. E., Hieb, A. R. & Luger, K. A quantitative investigation of linker histone interactions with nucleosomes and chromatin. Sci. Rep. 6, 19122 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, S., Sun, S., Li, Z., Zhang, R. & Xu, J. Accurate de novo prediction of protein contact map by ultra-deep learning model. PLoS Comput. Biol. 13, e1005324 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. Hunkeler, M. et al. Modular HUWE1 architecture serves as hub for degradation of cell-fate decision factors. Preprint at https://www.biorxiv.org/content/10.1101/2020.08.19.257352v1 (2020).

  34. Conti, E., Uy, M., Leighton, L., Blobel, G. & Kuriyan, J. Crystallographic analysis of the recognition of a nuclear localization signal by the nuclear import factor karyopherin alpha. Cell 94, 193–204 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Huber, A. H. & Weis, W. I. The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell 105, 391–402 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Kamadurai, H. B. et al. Insights into ubiquitin transfer cascades from a structure of a UbcH5B approximately ubiquitin-HECT(NEDD4L) complex. Mol. Cell 36, 1095–1102 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sander, B., Xu, W., Eilers, M., Popov, N. & Lorenz, S. A conformational switch regulates the ubiquitin ligase HUWE1. eLife 6, e21036 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Pandya, R. K., Partridge, J. R., Love, K. R., Schwartz, T. U. & Ploegh, H. L. A structural element within the HUWE1 HECT domain modulates self-ubiquitination and substrate ubiquitination activities. J. Biol. Chem. 285, 5664–5673 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Forget, A. et al. Shh signaling protects Atoh1 from degradation mediated by the E3 ubiquitin ligase Huwe1 in neural precursors. Dev. Cell 29, 649–661 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Baek, K. et al. NEDD8 nucleates a multivalent cullin-RING-UBE2D ubiquitin ligation assembly. Nature 578, 461–466 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Neuhold, J. et al. GoldenBac: a simple, highly efficient, and widely applicable system for construction of multi-gene expression vectors for use with the baculovirus expression vector system. BMC Biotechnol. 20, 26 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Vonrhein, C. et al. Data processing and analysis with the autoPROC toolbox. Acta Crystallogr. D Biol. Crystallogr. 67, 293–302 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tickle, I. J. et al. STARANISO. https://staraniso.globalphasing.org/cgi-bin/staraniso.cgi (2018).

  45. Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69, 1204–1214 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Terwilliger, T. C. et al. Decision-making in structure solution using Bayesian estimates of map quality: the PHENIX AutoSol wizard. Acta Crystallogr. D Biol. Crystallogr. 65, 582–601 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Smart, O. S. et al. Exploiting structure similarity in refinement: automated NCS and target-structure restraints in BUSTER. Acta Crystallogr. D Biol. Crystallogr. 68, 368–380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  51. 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 

  52. 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 

  53. Wagner, T. et al. SPHIRE-crYOLO is a fast and accurate fully automated particle picker for cryo-EM. Commun. Biol. 2, 218 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Zhong, E. D., Bepler, T., Berger, B. & Davis, J. H. CryoDRGN: reconstruction of heterogeneous cryo-EM structures using neural networks. Nat. Methods 18, 176–185 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Dorfer, V. et al. MS Amanda, a universal identification algorithm optimized for high accuracy tandem mass spectra. J. Proteome Res. 13, 3679–3684 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Doblmann, J. et al. apQuant: accurate label-free quantification by quality filtering. J. Proteome Res. 18, 535–541 (2019).

    CAS  PubMed  Google Scholar 

  59. Graham, M., Combe, C., Kolbowski, L. & Rappsilber, J. xiView: a common platform for the downstream analysis of crosslinking mass spectrometry data. Preprint at https://www.biorxiv.org/content/10.1101/561829v1 (2019).

Download references

Acknowledgements

We thank all members of the Clausen group for remarks on the manuscript and discussions, and the Mass Spectrometry and ProTech services from the Vienna BioCenter Core Facilities for their support. We thank the scientific staff at beamline X10SA, Paul Scherrer Institute (Villigen, Switzerland), as well as those at beamline P11, DESY (Hamburg, Germany) for support in crystallographic data collection. We thank the following cryo-EM facilities for their access and support: CEITEC MU of CIISB, Instruct-CZ Centrer (proposal no. LM2018127); the UK national Electron Bio-imaging Center (Diamond Lightsource, proposal no. EM BI25222); the EM facility at the Institute of Science and Technology (IST) Austria; and the EM facility at the Vienna BioCenter Core Facilities. We thank members of the Protein Science laboratory at Boehringer Ingelheim for support in connection with the expression and purification of HUWE1N, and V.-V. Hodirnau for help with cryo-EM analysis. This project received funding from the European Research Council under the European Union’s Horizon 2020 research and innovation program (no. AdG 694978), a Marie Skłodowska-Curie grant (agreement no. 847548), an FFG Headquarter grant (no. 852936) and the Austrian Science Fund (no. FWF, SFB F 79). P.M. and O.A.P are members of the Boehringer Ingelheim Discovery Research global postdoctorate program. IMP is supported by Boehringer Ingelheim.

Author information

Authors and Affiliations

Authors

Contributions

D.B.G. and T.C. designed experiments. L.D., O.A.P., D.B.G., R.K., P.M., R.G., V.F., D. Kordic, A.L. and J.N. prepared expression constructs, purified HUWE1 and performed biochemical assays. O.A.P., I.G., J.A. and D.H. performed cryo-EM analysis. A.V. and R.I. performed XL–MS analysis. A.S. performed bioinformatic analysis. D.B.G., T.C., A.M., G.B., P.S.-B., J.B., B.W., G.F. and D. Kessler performed crystallographic analysis. D.B.G. built the molecular models. D.B.G. and T.C. coordinated the research project and prepared the manuscript, with input from all authors.

Corresponding authors

Correspondence to Daniel B. Grabarczyk or Tim Clausen.

Ethics declarations

Competing interests

G.B., J.B., P.S.-B., G.F., B.W. and D. Kessler are currently employees of Boehringer Ingelheim.

Additional information

Peer review information Nature Chemical Biology thanks Ronald Hay 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

Extended Data Fig. 1 Model and electron density quality of the HUWE1N crystal structure.

a, The anomalous difference Fourier map at 2.5 σ is shown over representative ARM repeats from the structure of SeMet HUWE1N. Selenomethionines are colored in cyan. b, The asymmetric unit is shown with the two copies of HUWE1N colored in green and cyan, in cartoon representation. The 2FO-FC map at 1 σ is displayed. c, Representative 2FO-FC map at 1 σ of the ARM repeats.

Extended Data Fig. 2 Details of the HUWE1N solenoid and comparison with human HUWE1.

a, The primary and tertiary structure of HUWE1N is shown as in Fig. 2a, but with every ARM repeat labelled. b, The predicted positions of the previously annotated human HUWE1 domains in the context of the HUWE1N structure. The D/E-rich region is highly conserved in character and corresponds to HUWE1N IDR1. c, The predicted positions of X-linked intellectual disability mutations on the structure of HUWE1N. Mutated residues are shown in sphere representation, with HECT domain mutations in orange and all other mutations in blue.

Extended Data Fig. 3 Molecular details of the HUWE1N crystal structure.

a, Comparison of the HUWE1N ARM repeats with peptide-binding ARM repeat proteins. HUWE1N is shown in gray, bound peptides in red, and in the upper panel p120 catenin (PDB 3L6X) in light blue, and in the lower panel, a designed ARM repeat protein (PDB 6S9O) in dark blue. b, The surface conservation of the ring-closing interface is shown as a spectrum with magenta indicating strong conservation, and cyan low conservation. The score was calculated using AACon1 from an alignment of 49 HUWE1 orthologs. The protein was separated to show the surface of the interface on the side of the neck (upper panel) and tail (lower panel). c, Comparison of the two chains in the asymmetric unit superimposed by their first 300 amino acids.

Extended Data Fig. 4 Three-dimensional representation of the XL-MS data.

The cross-links shown in Fig. 5a are displayed as dashed tubes on the structure of HUWE1N, represented as in Fig. 2a.

Extended Data Fig. 5 Models and sharpened maps for the two refined CryoEM classes.

a, Overall fit between the model and map for Class 1. b, Overall fit between the model and map for Class 2. c,d, Density for ARM repeat helices in regions of the structure with high local resolution.

Extended Data Fig. 6 Structural features of HUWE1N.

a, Relief of potential clashes during catalysis. The structure of the Rsp5 HECT domain (in red), charged with Ub (in blue) and in complex with a peptide substrate (PDB 4LCD), is superimposed on the HUWE1N HECT domain from the crystal structure in the upper panel, and CryoEM Class 2 in the lower panel. b, ARM34 as a molecular hinge. The H1, H2, H3 helices of ARM34 and the N-terminal HECT helix are shown in the leftmost panel together with an overview and cartoon presentation. The two copies present in the crystal structure of an extended HUWE1 HECT domain (PDB 5LP8) are overlaid on our structure and shown in cyan. Structural rearrangements of ARM34 helices H2 and H3 are indicated. c, The position of F2105. d, Autoubiquitination activity of full-length CeHUWE1 compared to the isolated HECT domain (residues 3793-4180) followed using Ub-DyLight800.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2 and Fig. 1.

Reporting Summary

Supplementary Video 1

Electron density quality of the HUWE1N crystal structure. The 2FO-FC map at 1 σ is shown with the structure colored as in Fig. 2.

Supplementary Video 2

Heterogeneity of the Cryo-EM data analyzed using CryoDRGN.

Supplementary Video 3

Heterogeneity of the Cryo-EM data analyzed using CryoDRGN, second viewpoint.

Source data

Source Data Fig. 1

Unprocessed and Coomassie-stained gels, and mass spectrometry datasets.

Source Data Fig. 3

Unprocessed and Coomassie-stained gels.

Source Data Fig. 5

Mass spectrometry data used for Fig. 5a and Extended Data Fig. 4.

Source Data Fig. 6

Unprocessed and Coomassie-stained gels.

Source Data Extended Data Fig. 6

Unprocessed and Coomassie-stained gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grabarczyk, D.B., Petrova, O.A., Deszcz, L. et al. HUWE1 employs a giant substrate-binding ring to feed and regulate its HECT E3 domain. Nat Chem Biol 17, 1084–1092 (2021). https://doi.org/10.1038/s41589-021-00831-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00831-5

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