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:

Molecular basis of Lys11-polyubiquitin specificity in the deubiquitinase Cezanne

Abstract

The post-translational modification of proteins with polyubiquitin regulates virtually all aspects of cell biology. Eight distinct chain linkage types co-exist in polyubiquitin and are independently regulated in cells. This ‘ubiquitin code’ determines the fate of the modified protein1. Deubiquitinating enzymes of the ovarian tumour (OTU) family regulate cellular signalling by targeting distinct linkage types within polyubiquitin2, and understanding their mechanisms of linkage specificity gives fundamental insights into the ubiquitin system. Here we reveal how the deubiquitinase Cezanne (also known as OTUD7B) specifically targets Lys11-linked polyubiquitin. Crystal structures of Cezanne alone and in complex with monoubiquitin and Lys11-linked diubiquitin, in combination with hydrogen–deuterium exchange mass spectrometry, enable us to reconstruct the enzymatic cycle in great detail. An intricate mechanism of ubiquitin-assisted conformational changes activates the enzyme, and while all chain types interact with the enzymatic S1 site, only Lys11-linked chains can bind productively across the active site and stimulate catalytic turnover. Our work highlights the plasticity of deubiquitinases and indicates that new conformational states can occur when a true substrate, such as diubiquitin, is bound at the active site.

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

Figure 1: Cezanne biochemistry and structures.
Figure 2: Conformational changes in the Cezanne catalytic cycle.
Figure 3: Mutational analysis of Cezanne dynamics.
Figure 4: Basis of Lys11 specificity and model of Cezanne mechanism.

Similar content being viewed by others

Accession codes

Data deposits

Coordinates and structure factors have been deposited with the Protein Data Bank under accession codes 5LRU (Cez apo), 5LRV (Cez–Lys11 diUb), 5LRW (Cez–Ub) and 5LRX (A20–Ub).

References

  1. Komander, D. & Rape, M. The ubiquitin code. Annu. Rev. Biochem. 81, 203–229 (2012)

    CAS  PubMed  Google Scholar 

  2. Mevissen, T. E. T. et al. OTU deubiquitinases reveal mechanisms of linkage specificity and enable ubiquitin chain restriction analysis. Cell 154, 169–184 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Keusekotten, K. et al. OTULIN antagonizes LUBAC signaling by specifically hydrolyzing Met1-linked polyubiquitin. Cell 153, 1312–1326 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wiener, R., Zhang, X., Wang, T. & Wolberger, C. The mechanism of OTUB1-mediated inhibition of ubiquitination. Nature 483, 618–622 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Juang, Y.-C. et al. OTUB1 co-opts Lys48-linked ubiquitin recognition to suppress E2 enzyme function. Mol. Cell 45, 384–397 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hymowitz, S. G. & Wertz, I. E. A20: from ubiquitin editing to tumour suppression. Nat. Rev. Cancer 10, 332–341 (2010)

    Article  CAS  PubMed  Google Scholar 

  7. Wertz, I. E. et al. Phosphorylation and linear ubiquitin direct A20 inhibition of inflammation. Nature 528, 370–375 (2015)

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Tran, H., Hamada, F., Schwarz-Romond, T. & Bienz, M. Trabid, a new positive regulator of Wnt-induced transcription with preference for binding and cleaving K63-linked ubiquitin chains. Genes Dev. 22, 528–542 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jin, J. et al. Epigenetic regulation of the expression of Il12 and Il23 and autoimmune inflammation by the deubiquitinase Trabid. Nat. Immunol. 17, 259–268 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Licchesi, J. D. F. et al. An ankyrin-repeat ubiquitin-binding domain determines TRABID’s specificity for atypical ubiquitin chains. Nat. Struct. Mol. Biol. 19, 62–71 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

  11. Christianson, J. C. & Ye, Y. Cleaning up in the endoplasmic reticulum: ubiquitin in charge. Nat. Struct. Mol. Biol. 21, 325–335 (2014)

    Article  CAS  PubMed  Google Scholar 

  12. Hu, H. et al. OTUD7B controls non-canonical NF-κB activation through deubiquitination of TRAF3. Nature 494, 371–374 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Enesa, K. et al. NF-κB suppression by the deubiquitinating enzyme Cezanne: a novel negative feedback loop in pro-inflammatory signaling. J. Biol. Chem. 283, 7036–7045 (2008)

    Article  CAS  PubMed  Google Scholar 

  14. Luong, A. et al. Cezanne regulates inflammatory responses to hypoxia in endothelial cells by targeting TRAF6 for deubiquitination. Circ. Res. 112, 1583–1591 (2013)

    Article  ADS  CAS  PubMed Central  Google Scholar 

  15. Hu, H. et al. Otud7b facilitates T cell activation and inflammatory responses by regulating Zap70 ubiquitination. J. Exp. Med. 213, 399–414 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pareja, F. et al. Deubiquitination of EGFR by Cezanne-1 contributes to cancer progression. Oncogene 31, 4599–4608 (2012)

    Article  CAS  PubMed  Google Scholar 

  17. Bremm, A., Moniz, S., Mader, J., Rocha, S. & Komander, D. Cezanne (OTUD7B) regulates HIF-1α homeostasis in a proteasome-independent manner. EMBO Rep. 15, 1268–1277 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Moniz, S. et al. Cezanne regulates E2F1-dependent HIF2α expression. J. Cell Sci. 128, 3082–3093 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bremm, A., Freund, S. M. V. & Komander, D. Lys11-linked ubiquitin chains adopt compact conformations and are preferentially hydrolyzed by the deubiquitinase Cezanne. Nat. Struct. Mol. Biol. 17, 939–947 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Geurink, P. P. et al. Development of diubiquitin-based FRET probes to quantify ubiquitin linkage specificity of deubiquitinating enzymes. ChemBioChem 17, 816–820 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mulder, M. P. C., El Oualid, F., ter Beek, J. & Ovaa, H. A native chemical ligation handle that enables the synthesis of advanced activity-based probes: diubiquitin as a case study. ChemBioChem 15, 946–949 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Komander, D. & Barford, D. Structure of the A20 OTU domain and mechanistic insights into deubiquitination. Biochem. J. 409, 77–85 (2008)

    Article  CAS  PubMed  Google Scholar 

  23. Lin, S.-C. et al. Molecular basis for the unique deubiquitinating activity of the NF-κB inhibitor A20. J. Mol. Biol. 376, 526–540 (2008)

    Article  CAS  PubMed  Google Scholar 

  24. Geurink, P. P., El Oualid, F., Jonker, A., Hameed, D. S. & Ovaa, H. A general chemical ligation approach towards isopeptide-linked ubiquitin and ubiquitin-like assay reagents. ChemBioChem 13, 293–297 (2012)

    Article  CAS  PubMed  Google Scholar 

  25. Wickliffe, K. E., Lorenz, S., Wemmer, D. E., Kuriyan, J. & Rape, M. The mechanism of linkage-specific ubiquitin chain elongation by a single-subunit E2. Cell 144, 769–781 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Berrow, N. S. et al. A versatile ligation-independent cloning method suitable for high-throughput expression screening applications. Nucleic Acids Res. 35, e45 (2007)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  27. Hospenthal, M. K., Mevissen, T. E. T. & Komander, D. Deubiquitinase-based analysis of ubiquitin chain architecture using Ubiquitin Chain Restriction (UbiCRest). Nat. Protocols 10, 349–361 (2015)

    Article  PubMed  Google Scholar 

  28. Faggiano, S., Alfano, C. & Pastore, A. The missing links to link ubiquitin: Methods for the enzymatic production of polyubiquitin chains. Anal. Biochem. 492, 82–90 (2016)

    Article  CAS  PubMed  Google Scholar 

  29. Borodovsky, A. et al. Chemistry-based functional proteomics reveals novel members of the deubiquitinating enzyme family. Chem. Biol. 9, 1149–1159 (2002)

    Article  CAS  PubMed  Google Scholar 

  30. Ekkebus, R. et al. On terminal alkynes that can react with active-site cysteine nucleophiles in proteases. J. Am. Chem. Soc. 135, 2867–2870 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Battye, T. G. G., Kontogiannis, L., Johnson, O., Powell, H. R. & Leslie, A. G. W. iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM. Acta Crystallogr. D Biol. Crystallogr. 67, 271–281 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Evans, P. Scaling and assessment of data quality. Acta Crystallogr. D Biol. Crystallogr. 62, 72–82 (2006)

    Article  PubMed  Google Scholar 

  34. 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 

  35. Bricogne, G., Vonrhein, C., Flensburg, C., Schiltz, M. & Paciorek, W. Generation, representation and flow of phase information in structure determination: recent developments in and around SHARP 2.0. Acta Crystallogr. D Biol. Crystallogr. 59, 2023–2030 (2003)

    Article  CAS  PubMed  Google Scholar 

  36. Vonrhein, C., Blanc, E., Roversi, P. & Bricogne, G. Automated structure solution with autoSHARP. Methods Mol. Biol. 364, 215–230 (2007)

    CAS  PubMed  Google Scholar 

  37. 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 

  38. Adams, P. D. et al. The Phenix software for automated determination of macromolecular structures. Methods 55, 94–106 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Vijay-Kumar, S., Bugg, C. E. & Cook, W. J. Structure of ubiquitin refined at 1.8 A resolution. J. Mol. Biol. 194, 531–544 (1987)

    Article  CAS  PubMed  Google Scholar 

  41. Pham, G. H. et al. Comparison of native and non-native ubiquitin oligomers reveals analogous structures and reactivities. Protein Sci. 25, 456–471 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Messick, T. E. et al. Structural basis for ubiquitin recognition by the Otu1 ovarian tumor domain protein. J. Biol. Chem. 283, 11038–11049 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wiener, R. et al. E2 ubiquitin-conjugating enzymes regulate the deubiquitinating activity of OTUB1. Nat. Struct. Mol. Biol. 20, 1033–1039 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Wakatsuki and N. Matsugaki for access to KEK PF BL-1A, and beam-line scientists at ESRF ID23-1, ID29 and Diamond I02 and I03; R. Williams and M. Skehel for help and discussion on HDX-MS; and C. Johnson and S. McLaughlin for help with biophysics. Access to DLS was supported in part by the EU FP7 infrastructure grant BIOSTRUCT-X (contract no. 283570). This work was supported by the Medical Research Council (U105192732 (D.K.)), the European Research Council (309756 (D.K.); 281699 (H.O.)], the Lister Institute for Preventive Medicine (D.K.), the British Heart Foundation (PG11/109/29247 (J.E.B.)), and a Netherlands Organization for Scientific Research VICI grant (724.013.002 (H.O.)). T.E.T.M. was supported by the Marie Curie ITN UPStream and Y.K. by Marie Curie and EMBO Long Term Fellowships.

Author information

Authors and Affiliations

Authors

Contributions

D.K. directed the research. T.E.T.M. performed all biochemical experiments, crystallized and determined the structure of the Cez–Lys11 diUb complex, and refined and analysed all structures. Y.K. crystallized and determined the structure of Cez apo, Cez–Ub and A20–Ub and performed preliminary biochemistry. M.P.C.M. and F.E.O. designed and generated diubiquitin ABPs. P.P.G. designed and generated (with B.D.M.v.T.) diubiquitin-based FRET probes. S.L.M. performed HDX-MS. J.E.B. performed preliminary HDX-MS. M.G. performed TSA assays. P.R.E., M.A. and M.K. helped with data collection and structure determination. S.M.V.F. performed NMR analysis. H.O. guided chemical biology efforts. T.E.T.M. and D.K. analysed the data and wrote the manuscript with help from all authors.

Corresponding author

Correspondence to David Komander.

Ethics declarations

Competing interests

D.K. and H.O. are part of the DUB Alliance, which includes Cancer Research Technology and FORMA Therapeutics. H.O. and F.E.O. are co-founders and shareholders of UbiQ Bio BV.

Extended data figures and tables

Extended Data Figure 1 Analysis of branched triUb substrates and FRET-based diUb cleavage kinetics.

a, Branched triUb molecules with different topologies were generated as shown in the schematic (bottom). Lys11 diUb, Lys63 diUb and branched Lys11/63 triUb (left panel) were treated with wild-type Cezanne (Cez WT; top) and OTUD1 (residues 287–481, bottom), a Lys63-specific enzyme2. Both DUBs cleaved their preferred diUb substrate as well as one linkage of the branched triUb molecule. Lys11 diUb, Lys48 diUb and branched Lys11/48 triUb (right panel) were incubated with wild-type Cezanne (top) and OTUB1 (full-length, bottom), a Lys48-specific OTU DUB2. Again, both enzymes showed similar activities towards their preferred linkage type in a diUb substrate and a branched triUb molecule. This shows that Cezanne can cleave Lys11 linkages in the context of Lys11/Lys63- and Lys11/Lys48-branched chains. For gel source images, see Supplementary Fig. 1. b, Schematic of FRET-based diUb cleavage assays to derive DUB kinetics. Distal and proximal Ub moieties were modified with a donor (D) and acceptor (A) fluorophore, respectively. Upon DUB treatment, the native isopeptide bond was cleaved and the FRET signal was lost. The increase in donor intensity was measured to follow the reaction. c, Kinetic parameters for all independently performed experiments of Lys11, Lys63 and Lys48 diUb cleavage by wild-type Cezanne. Values are in good agreement with previously published parameters derived from gel-based studies41. d, Summary of kinetic parameters for Lys11-, Lys63- and Lys48-linked diUb cleavage by Cezanne E157K. The determined KM values for Lys48 diUb lie above the highest tested substrate concentration, so kinetic parameters marked by an asterisk were calculated from experiments where substrate saturation could not be achieved owing to technical limitations. Catalytic efficiencies (kcat/KM) for this substrate were also derived from a linear fit of the lower concentration range (0–20 μM, linear part of the graph). These values are marked by a cross. The similarity of catalytic efficiencies calculated in two different ways indicate that the kinetic parameters marked by asterisks are good estimates. See Supplementary Fig. 2 for all corresponding graphs of initial rates.

Extended Data Figure 2 Crystal structures determined in this study and comparison of A20-like OTU apo structures.

a–e, Active site regions Cez apo (a), Cez–Ub-A (b), Cez–Ub-B (c), Cez–Lys11 diUb (d), and A20–Ub (e). 2|FO| – |FC| electron density maps contoured at 1σ (blue) cover catalytic residues, the Cys-loop and chemical linkers in the complex structures. Hydrogen bonds between the oxyanion hole and the Lys11 diUb ABP linker carbonyl are indicated in d, and the sp3-hybridized carbon atom that is linked to the oxyanion in a native first tetrahedral intermediate is highlighted (green arrowhead). f, g, Cezanne OTU (as in Fig. 1d) and A20 OTU (PDB 2VFJ22) apo structures with labelled secondary structure elements. Catalytic residues are shown in ball-and-stick representation. Three loops surrounding the active site are coloured (Cys-loop, orange; V-loop, green; His-loop, purple). h, Superposition of f and g showing structural similarities and differences between Cezanne and A20. i, j, Topology diagrams of f and g. The catalytic centre is indicated (red stars) and Ub-binding sites are highlighted. A20 contains two additional N-terminal and one additional C-terminal helices compared to Cezanne. The β1–β10 sheet in Cezanne corresponds to the A20 β7–β8 sheet. This explains why sequence-based alignments are challenging. k, Superposition of Cez apo (f) and TRABID AnkUBD (pink) and OTU (brown) domains (residues 245–697, PDB 3ZRH10).

Extended Data Figure 3 Comparison of Ub and diUb complexes within the OTU family.

Ub moieties are shown in cartoon representation under transparent surfaces in shades of yellow. Secondary structure elements involved in Ub binding are labelled, and active site loops are coloured as in Extended Data Fig. 2f. a, Cez–Ub-A complex as in Fig. 1d. b, A20–Ub complex as in Fig. 1g. c, Superposition of Ub complexes reveals a conserved S1 Ub-binding mode in A20-like OTU DUBs. d, Superposition of A20 apo (Extended Data Fig. 2g) and A20–Ub (b). No large conformational changes occur upon Ub binding. However, two unstructured loops in A20 apo are stabilized by Ub, forming helix α6′ and the β2′–β2′′ sheet (compare with Extended Data Fig. 2j). e, The structure of the yeast Otu1–Ub complex (PDB 3BY442) is representative of the OTUD subfamily of OTU DUBs. The Ub moiety in the S1 site is mainly bound by the short helix α3. f, The superposition of Cez–Ub (a) and Otu1 (e) reveals substantially different S1 sites between A20-like and OTUD subfamilies. Rotations around the roll axis of Ub (~80°) and the active site (~70°) would be required to align both Ub moieties. gi, Structures of OTU domains in identical orientation bound to their respective diUb substrate. The binding modes of proximal and distal Ub differ markedly between the here determined Cez–Lys11 diUb complex (g, as shown in Fig. 1d), the h/ceOTUB1–Ub UbcH5b–Ub structure (PDB 4LDT43; UbcH5b molecule is not shown), which resembles an OTUB1–Lys48 diUb complex (h), and OTULIN bound to Met1-linked diUb (PDB 3ZNZ3) (i).

Extended Data Figure 4 Conformational changes in the catalytic centre.

Cezanne structures (as in Fig. 2a) are shown in the corners, and transitions I–IV are overlays of neighbouring structures. Side chains of catalytic residues and other selected residues are highlighted. Loops are coloured as in Extended Data Fig. 2f. Cez apo shows a catalytically incompetent state. His358 and Glu157 are in flipped-out conformations. Transition I features structural rearrangements of the Cys-loop (orange arrowhead), helices α1 and α2 (red arrowheads) and the S1′-loop (black arrowhead). Cez–Lys11 diUb also features an inactive state; His358 remains flipped-out, which is caused by the Cys-loop residue Thr188 that is pushed into the active site by the proximal Ub. In transition II, another S1′-loop movement relocates S1′ site residues (black arrowhead). A similar inactive state is present in Cez–Ub-A, and Thr188 still resides in the active site. The absence of the proximal Ub allows the Cys-loop and Thr188 to move in transition III (orange arrow), allowing a ~100° rotation of His358. Hence, Cez–Ub-B contains an aligned catalytic centre. Hydrogen bonds are indicated. In transition IV, large conformational changes in various parts of the OTU domain regenerate the autoinhibited apoenzyme.

Extended Data Figure 5 Ub binding to Cezanne and mutational analysis of residues involved in catalysis and conformational dynamics.

a, NMR analysis of Ub binding to wild-type Cezanne and the covalent Cez–Ub complex. 1H-15N BEST-TROSY spectra of 50 μM 15N-labelled Ub alone (black) and in the presence of 130 μM unlabelled wild-type Cezanne (red, left) or unlabelled Cez–Ub (red, right). Strong chemical shift perturbations upon addition of wild-type Cezanne indicate binding to Ub. In contrast, no chemical shifts were detected with Cez–Ub, suggesting that all changes with wild-type Cezanne can be attributed to the S1 site (this site is occupied by unlabelled Ub in Cez–Ub). More importantly, this also indicates that a functional S1′ site is not present in the Cez–Ub complex. b, Fluorescence polarization experiment assessing the binding of FlAsH-tagged Ub to catalytically inactive Cezanne (C194A), wild-type Cezanne, an S1 site mutant (E295K, see below) and the Cez–Ub complex. c, Lys11 diUb cleavage assays of catalytic Cys194 and His358 mutants. d, e, Ub-KG* cleavage by catalytic Cys194 and His358 mutants (d), as well as Asn193 and helix α2 mutants that modulate the overall dynamics of Cezanne (e). This assay follows fluorescent dye release in the reaction; the fact that Cezanne H358A is inactive indicates an important role in the deprotonation of the catalytic Cys at the start of the reaction (that is, the catalytic centre transiently adopts an active state) and/or a role in resolving the first tetrahedral intermediate. If His358 was not required for either, we would expect a single turnover of the reaction, which would stop at the thioester intermediate. The release of KG-TAMRA would still occur, but this was not detected even at an enzyme concentration of 150 nM (the substrate concentration in all assays was 150 nM). The fluorescence polarization signal also did not increase, suggesting that no covalent first tetrahedral intermediate was formed due to impaired dye release. Hence, the data suggest a role for His358 at least in the initial Cys deprotonation in addition to the last reaction step. f, Fluorescence polarization binding assay of Asn193 and helix α2 mutants compared to constructs used in b. g, h, Hydrolysis of Lys11-linked diUb (g) and Ub-KG* (h) by Cezanne H197A and D210A. i, DUB assay with Cezanne variants (extended incubation at room temperature, RT). j, Lys11 diUb cleavage assay with His197 variants. k, Fluorescence polarization binding assay as in b testing His197 variants. l, Mutation of corresponding residues in A20 (A20 His256 corresponds to Cezanne His358, and A20 His106 to Cezanne His197) have similar effects on Lys48 diUb hydrolysis. All DUB assays are representative of at least two independent experiments for every construct. Ub-KG* cleavage experiments and fluorescence polarization binding assays were replicated at least twice for each variant with consistent results. Fluorescence polarization measurements were performed in triplicate. Error bars represent s.d. from the mean. mP, millipolarization unit. For gel source images, see Supplementary Fig. 1.

Extended Data Figure 6 HDX-MS analysis of the Cez apo state.

a, HDX-MS experiment showing the conformational dynamics of wild-type Cezanne. The relative fractional deuterium uptake is shown for four time points (0.3–300 s). Protein sequence and secondary structure elements of Cez apo (dark grey) and Cez–diUb (light grey) are aligned. Residues of the catalytic centre are indicated by stars. b, Cez apo structure coloured based upon the relative fractional deuterium uptake of wild-type Cezanne at 0.3 s, 3 s, 30 s and 300 s. The region spanning helices α1 and α2 shows a particularly high deuterium uptake, suggesting conformational flexibility in this region in solution. c, The H–D exchange of the α2-helix destabilizing mutant Cezanne L155G/I156G compared to wild-type Cezanne. Cez apo structure coloured based upon the difference in deuterium uptake (L155G/I156G-WT) at 0.3 s, 3 s, 30 s and 300 s (heat maps are shown in Supplementary Fig. 3). The data suggest that helix α2 is destabilized, as regions structurally adjacent to the mutation site (black arrowhead) show increased deuterium uptake as compared to wild-type Cezanne. Peptides containing the mutations could not be analysed owing to the different sequences, and are therefore coloured grey. Notably, most differences are stronger at shorter time points, indicating increased dynamics within this time frame (0.3–30 s). At the last time point (300 s), differences are not as pronounced, suggesting that wild-type Cezanne undergoes similar structural rearrangements at a slower speed. Importantly, the data also confirm that overall folding of the mutant was not affected by the two Gly residues introduced into helix α2.

Extended Data Figure 7 HDX-MS analysis of transitions I, II and IV.

a, HDX-MS experiments were performed with Cez apo, Cez–Lys11 diUb and Cez–Ub. Heat maps show differences in deuterium uptake between two states in each case: transition I (diUb–apo), transition II (Ub–diUb) and transition IV (apo–Ub). Hence, Cezanne regions that are stabilized or more protected upon Lys11 diUb binding (transition I), or more flexible or exposed upon the stepwise release of the proximal Ub (transition II) and the distal Ub (transition IV) are highlighted. The S1 site predominantly consists of helices α5 and α6 (that is, helical content with very low deuterium uptake in any state), and is not as easily detected as the S1′ site, which features various loops and the dynamic helix α2. Cezanne sequence and secondary structure schematics are shown as in Extended Data Fig. 6a. b, Cez–Lys11 diUb structure (shown without Lys11 diUb) coloured based upon transition I deuterium uptake at 30 s. c, Transition II deuterium uptake at 30 s plotted onto Cez–Ub-B (shown without Ub). d, Cez apo coloured based upon transition IV deuterium uptake at 30 s.

Extended Data Figure 8 Mutational analysis of the S1 Ub-binding site.

a, Thermal shift assay of wild-type and C194A Cezanne. In the presence of Ub, the melting temperature (Tm) of Cezanne increases. Data were recorded in triplicate and in two independent experiments. b, Ub-KG* hydrolysis by S1 site mutants. c, d, Fluorescence polarization-based affinity measurement using N-terminally FlAsH-tagged Ub. Dissociation constants (KD) for wild-type (c), C194A and C194S Cezanne (d) are shown. Data are representative of at least two independent experiments per construct. e, Pull-down assay with His-tagged Cezanne constructs (catalytically inactive C194A, S1 site mutant C194A/E295K or wild-type) and different Ub and diUb variants. MonoUb requires an intact C terminus to bind to Cezanne C194A. To prevent unspecific binding of differently linked diUb molecules with their proximal Ub to the S1 site, the C terminus was removed (∆LRGG). Variants marked by an asterisk were assembled using K11R, S20C and K63R mutations in the distal Ub, as well as K63R (only for Lys11 diUb) and ∆LRGG in the proximal Ub moiety. Pull-down and input samples were analysed by SDS–PAGE and silver staining. The pull-down assay was performed in two independent experiments. For gel source images, see Supplementary Fig. 1.

Extended Data Figure 9 Biochemical analysis of S1′ site mutations.

a, The interface between Cezanne and the proximal Ub in the Cez–Lys11 diUb complex. An unusual surface of Ub comprising Glu16, Asp32, Lys33 and Glu34 is contacted by the S1′ site (Leu155, Glu157, Met203, Phe206 and His207). b, c, Lys11 diUb cleavage (b) and Lys11 diUb ABP reactivity (c) assays with S1′ site mutants. d, DUB assays with wild-type Cezanne and Ub variants. Lys11 diUb substrates were assembled to specifically mutate the proximal Ub by using K11R, K63R mutations in the distal, and K63R, ∆LRGG in the proximal Ub moiety. No further mutations were introduced in WT*, while K33A* and K33E* variants additionally contained respective mutations in their proximal Ub only. e, Lys11 diUb cleavage assay with Glu157 variants. f, g, Gel-based specificity analysis of Cezanne E157K. The mutant shows a reduced activity towards Lys11-linked diUb and therefore specificity compared to wild-type Cezanne (compare Fig. 1b). Assays with each variant were performed at least twice with consistent results. For gel source images, see Supplementary Fig. 1.

Extended Data Table 1 Data collection and refinement statistics

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-3. (PDF 1929 kb)

Conformational changes in the Cezanne catalytic cycle

The four determined crystal structures delineate a catalytic cycle for Lys11 diUb cleavage. The video shows morphed states between individual crystal structures annotated as Transitions I-IV in the main text, which were generated using Chimera (UCSF - Yang, Z. et al. UCSF Chimera, MODELLER, and IMP: an integrated modeling system. J Struct Biol 179, 269–278 (2012)) and PyMol (www.pymol.org). The Cezanne OTU domain (blue) is shown in cartoon representation and Ub molecules (shades of yellow) as transparent surfaces. The same Ub moiety is shown for both Cez~Ub complexes. Side chains of catalytic centre residues as well as residues important for Cys-loop conformational changes and proximal Ub binding are shown in stick representation. The Cys-loop and the S1’ site region are coloured in orange and purple, respectively. Key features of the reaction cycle are highlighted. (MOV 12942 kb)

Cezanne active site and S1’ site in the catalytic cycle

The same hypothetical transitions as in Supplementary Video 1 are shown with a focus on the catalytic centre and the S1’ site region. (MOV 13429 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mevissen, T., Kulathu, Y., Mulder, M. et al. Molecular basis of Lys11-polyubiquitin specificity in the deubiquitinase Cezanne. Nature 538, 402–405 (2016). https://doi.org/10.1038/nature19836

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature19836

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