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:

Linkage-specific ubiquitin chain formation depends on a lysine hydrocarbon ruler

Abstract

Virtually all aspects of cell biology are regulated by a ubiquitin code where distinct ubiquitin chain architectures guide the binding events and itineraries of modified substrates. Various combinations of E2 and E3 enzymes accomplish chain formation by forging isopeptide bonds between the C terminus of their transiently linked donor ubiquitin and a specific nucleophilic amino acid on the acceptor ubiquitin, yet it is unknown whether the fundamental feature of most acceptors—the lysine side chain—affects catalysis. Here, use of synthetic ubiquitins with non-natural acceptor site replacements reveals that the aliphatic side chain specifying reactive amine geometry is a determinant of the ubiquitin code, through unanticipated and complex reliance of many distinct ubiquitin-carrying enzymes on a canonical acceptor lysine.

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: UBE2N~UB/UBE2V1/RNF4 RING E3 complex reacts preferentially with free amino acids harboring amine acceptors and various side-chain hydrocarbon linkers.
Fig. 2: K48 and K63 chain-forming E2s show strong preferences for a native lysine acceptor on UB.
Fig. 3: K63 chain-forming HECT E3 ligases show strong preferences for a native lysine acceptor on UB.
Fig. 4: The location of lysine analogs on acceptor UB impacts the distribution of di-UB chain linkage types generated by the E2 enzyme UBE2D3.
Fig. 5: Molecular dynamics (MD) simulations reveal pleiotropic structural effects on UBs harboring lysine analogs.

Similar content being viewed by others

Data availability

All raw gels are included in source data files. The MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD021286. Source data are provided with this paper.

Code availability

ROSETTA software can be downloaded from www.rosettacommons.org and is available free to academic users.

References

  1. Dikic, I., Wakatsuki, S. & Walters, K. J. Ubiquitin-binding domains—from structures to functions. Nat. Rev. Mol. Cell Biol. 10, 659–671 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Yau, R. & Rape, M. The increasing complexity of the ubiquitin code. Nat. Cell Biol. 18, 579–586 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Kwon, Y. T. & Ciechanover, A. The ubiquitin code in the ubiquitin-proteasome system and autophagy. Trends Biochem. Sci. 42, 873–886 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Mulder, M. P. C., Witting, K. F. & Ovaa, H. Cracking the ubiquitin code: the ubiquitin toolbox. Curr. Issues Mol. Biol. 37, 1–20 (2020).

    Article  PubMed  Google Scholar 

  6. Singh, S. K. et al. Synthetic uncleavable ubiquitinated proteins dissect proteasome deubiquitination and degradation, and highlight distinctive fate of tetraubiquitin. J. Am. Chem. Soc. 138, 16004–16015 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Sun, H. et al. Diverse fate of ubiquitin chain moieties: the proximal is degraded with the target, and the distal protects the proximal from removal and recycles. Proc. Natl Acad. Sci. USA 116, 7805–7812 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang, X. et al. An interaction landscape of ubiquitin signaling. Mol. Cell 65, 941–955 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Buetow, L. & Huang, D. T. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 17, 626–642 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zheng, N. & Shabek, N. Ubiquitin ligases: structure, function and regulation. Annu. Rev. Biochem. 86, 129–157 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Mattiroli, F. & Sixma, T. K. Lysine-targeting specificity in ubiquitin and ubiquitin-like modification pathways. Nat. Struct. Mol. Biol. 21, 308–316 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Bernier-Villamor, V., Sampson, D. A., Matunis, M. J. & Lima, C. D. Structural basis for E2-mediated SUMO conjugation revealed by a complex between ubiquitin-conjugating enzyme Ubc9 and RanGAP1. Cell 108, 345–356 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Eddins, M. J., Carlile, C. M., Gomez, K. M., Pickart, C. M. & Wolberger, C. Mms2–Ubc13 covalently bound to ubiquitin reveals the structural basis of linkage-specific polyubiquitin chain formation. Nat. Struct. Mol. Biol. 13, 915–920 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Smit, J. J. et al. The E3 ligase HOIP specifies linear ubiquitin chain assembly through its RING–IBR–RING domain and the unique LDD extension. EMBO J. 31, 3833–3844 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Berndsen, C. E., Wiener, R., Yu, I. W., Ringel, A. E. & Wolberger, C. A conserved asparagine has a structural role in ubiquitin-conjugating enzymes. Nat. Chem. Biol. 9, 154–156 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Stieglitz, B. et al. Structural basis for ligase-specific conjugation of linear ubiquitin chains by HOIP. Nature 503, 422–426 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wenzel, D. M., Lissounov, A., Brzovic, P. S. & Klevit, R. E. UBCH7 reactivity profile reveals parkin and HHARI to be RING/HECT hybrids. Nature 474, 105–108 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pao, K. C. et al. Activity-based E3 ligase profiling uncovers an E3 ligase with esterification activity. Nature 556, 381–385 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Virdee, S., Macmillan, D. & Waksman, G. Semisynthetic Src SH2 domains demonstrate altered phosphopeptide specificity induced by incorporation of unnatural lysine derivatives. Chem. Biol. 17, 274–284 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Temimi, A. H. K. A. et al. Lysine possesses the optimal chain length for histone lysine methyltransferase catalysis. Sci. Rep. 7, 16148 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. McKenna, S. et al. Noncovalent interaction between ubiquitin and the human DNA repair protein Mms2 is required for Ubc13-mediated polyubiquitination. J. Biol. Chem. 276, 40120–40126 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Branigan, E., Plechanovová, A., Jaffray, E. G., Naismith, J. H. & Hay, R. T. Structural basis for the RING-catalyzed synthesis of K63-linked ubiquitin chains. Nat. Struct. Mol. Biol. 22, 597–602 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Branigan, E., Carlos Penedo, J. & Hay, R. T. Ubiquitin transfer by a RING E3 ligase occurs from a closed E2~ubiquitin conformation. Nat. Commun. 11, 2846 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Petroski, M. D. & Deshaies, R. J. Mechanism of lysine 48-linked ubiquitin-chain synthesis by the cullin–RING ubiquitin–ligase complex SCF–Cdc34. Cell 123, 1107–1120 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Choi, Y.-S. et al. Differential ubiquitin binding by the acidic loops of Ube2g1 and Ube2r1 enzymes distinguishes their Lys-48-ubiquitylation activities. J. Biol. Chem. 290, 2251–2263 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Hill, S., Harrison, J. S., Lewis, S. M., Kuhlman, B. & Kleiger, G. Mechanism of Lysine 48 selectivity during polyubiquitin chain formation by the Ube2R1/2 ubiquitin-conjugating enzyme. Mol. Cell. Biol. 36, 1720–1732 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lu, G. et al. UBE2G1 governs the destruction of cereblon neomorphic substrates. eLife 7, e40958 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Hill, S. et al. Robust cullin–RING ligase function is established by a multiplicity of poly-ubiquitylation pathways. eLife 8, e51163 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Skaar, J. R., Pagan, J. K. & Pagano, M. Mechanisms and function of substrate recruitment by F-box proteins. Nat. Rev. Mol. Cell Biol. 14, 369–381 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Kronke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    Article  PubMed  CAS  Google Scholar 

  32. Lu, G. et al. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Maspero, E. et al. Structure of a ubiquitin-loaded HECT ligase reveals the molecular basis for catalytic priming. Nat. Struct. Mol. Biol. 20, 696–701 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

  35. Kim, H. C. & Huibregtse, J. M. Polyubiquitination by HECT E3s and the determinants of chain type specificity. Mol. Cell. Biol. 29, 3307–3318 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gupta, R. et al. Ubiquitination screen using protein microarrays for comprehensive identification of Rsp5 substrates in yeast. Mol. Syst. Biol. 3, 116 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. 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 

  38. Brown, N. G. et al. Dual RING E3 architectures regulate multiubiquitination and ubiquitin chain elongation by APC/C. Cell 165, 1440–1453 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brzovic, P. S. & Klevit, R. E. Ubiquitin transfer from the E2 perspective: why is UbcH5 so promiscuous? Cell Cycle 5, 2867–2873 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Swatek, K. N. et al. Insights into ubiquitin chain architecture using Ub-clipping. Nature 572, 533–537 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lee, B. L., Singh, A., Mark Glover, J. N., Hendzel, M. J. & Spyracopoulos, L. Molecular basis for K63-linked ubiquitination processes in double-strand DNA break repair: a focus on kinetics and dynamics. J. Mol. Biol. 429, 3409–3429 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Garg, P. et al. Structural and functional analysis of ubiquitin-based inhibitors that target the backsides of E2 enzymes. J. Mol. Biol. 432, 952–966 (2020).

    Article  CAS  PubMed  Google Scholar 

  43. Rout, M. K. et al. Stochastic gate dynamics regulate the catalytic activity of ubiquitination enzymes. J. Am. Chem. Soc. 136, 17446–17458 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Yunus, A. A. & Lima, C. D. Lysine activation and functional analysis of E2-mediated conjugation in the SUMO pathway. Nat. Struct. Mol. Biol. 13, 491–499 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Vittal, V. et al. Intrinsic disorder drives N-terminal ubiquitination by Ube2w. Nat. Chem. Biol. 11, 83–89 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Jones, W. M., Davis, A. G., Wilson, R. H., Elliott, K. L. & Sumner, I. A conserved asparagine in a ubiquitin-conjugating enzyme positions the substrate for nucleophilic attack. J. Comput. Chem. 40, 1969–1977 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Wang, M., Cheng, D., Peng, J. & Pickart, C. M. Molecular determinants of polyubiquitin linkage selection by an HECT ubiquitin ligase. EMBO J. 25, 1710–1719 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Scott, D. C. et al. Structure of a RING E3 trapped in action reveals ligation mechanism for the ubiquitin-like protein NEDD8. Cell 157, 1671–1684 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Burslem, G. M. & Crews, C. M. Proteolysis-targeting chimeras as therapeutics and tools for biological discovery. Cell 181, 102–114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Verma, R., Mohl, D. & Deshaies, R. J. Harnessing the power of proteolysis for targeted protein inactivation. Mol. Cell 77, 446–460 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Scott, D. C. et al. Two distinct types of E3 ligases work in unison to regulate substrate ubiquitylation. Cell 166, 1198–1214 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

  53. Brown, N. G. et al. Mechanism of polyubiquitination by human anaphase-promoting complex: RING repurposing for ubiquitin chain assembly. Mol. Cell 56, 246–260 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Plechanovova, A. et al. Mechanism of ubiquitylation by dimeric RING ligase RNF4. Nat. Struct. Mol. Biol. 18, 1052–1059 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Sievers, Q. L. et al. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 362, eaat0572 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Weissmann, F. et al. biGBac enables rapid gene assembly for the expression of large multisubunit protein complexes. Proc. Natl Acad. Sci. USA 113, E2564–E2569 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Duda, D. M. et al. Structural insights into NEDD8 activation of cullin–RING ligases: conformational control of conjugation. Cell 134, 995–1006 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ziemba, A. et al. Multimodal mechanism of action for the Cdc34 acidic loop: a case study for why ubiquitin-conjugating enzymes have loops and tails. J. Biol. Chem. 288, 34882–34896 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Kulak, N. A., Pichler, G., Paron, I., Nagaraj, N. & Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 11, 319–324 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study is dedicated the memory of our inspiring mentor, colleague and beloved friend, Huib Ovaa, whom we miss dearly. We thank J.R. Prabu, J. Kellermann, S. von Gronau, D. Scott, S. Uebel, S. Pettera, V. Sanchez, K. Baek, D. Horn-Ghetko and S. Kostrhon for assistance, reagents and helpful discussions. We also thank C. Talavera-Ormeño and P. Hekking for assistance with peptide synthesis. B.A.S. has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant no. 789016-NEDD8Activate), and from the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation—SCHU 3196/1-1). B.A.S. and M.M. are supported by the Max Planck Society. Also, N.P., D.H., N.B. and G.K. were supported by a grant from the National Institutes of Health (R15GM117555-02). G.J.v.d.H.v.N was supported by grants from NWO (VIDI and Off-Road). H.O. was supported by a VICI grant from the Netherlands Foundation for Scientific Research (NWO). Work by M.S. and M.J.B. was performed within the framework of SFB 1035 (German Research Foundation DFG, Sonderforschungsbereich 1035, no. 201302640, project Z01).

Author information

Authors and Affiliations

Authors

Contributions

Syntheses of UB analogs were designed and executed by G.J.v.d.H.v.N. J.L. performed all biochemical assays. Kinetics experiments were carried out by G.K., N.P., D.H. and N.B. MS experiments were designed and conducted by M.M., F.M.H. and O.K. MD simulations were performed by V.H.T. and J.S.H. NMR was carried out by M.J.B. and M.S. The manuscript was prepared by J.L., D.T.K., G.J.v.d.H.v.N., G.K., H.O. and B.A.S., with input from all authors. The project was supervised by B.A.S., G.K., H.O. and D.T.K.

Corresponding authors

Correspondence to Gary Kleiger, Huib Ovaa or Brenda A. Schulman.

Ethics declarations

Competing interests

H.O. was a shareholder of UbiqBio. All other authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 UBE2N~UB/UBE2V1/RNF4 RING E3 complex reacts preferentially with free amino acids harboring amine acceptors and various side-chain.

a, Fluorescence scan of SDS-PAGE gels demonstrating the discharge of labeled UB (UB*) to L-lysine compared with the absence of amino acid acceptor using wild-type UBE2N. Electrophoresis was performed under both reducing and non-reducing conditions to differentiate thioester bonded complexes from isopeptide bonded E2-donor UB ones. b, Time-courses of fluorescent UB discharge from UBE2N K92R~UB/UBE2V1/RNF4 RING E3 to the indicated amino acids, normalized to starting signal of fluorescent UB thioester-bonded to UBE2N. For all, N=2 independent experiments. For samples derived from the same experiment, gels were processed in parallel.

Source data

Extended Data Fig. 2 K48 and K63 chain-forming E2s equally discharge to K63UBC1-C5 and K48UBC1-C5 acceptors respectively.

a, Di-UB formed by K48 UB chain-forming E2 UBE2G1 with K63UBC1-C5 acceptors in the absence (left) or presence (right) of neddylated CRL4 (N8CRL4). b, Di-UB formed by the K63 UB chain-forming E2 UBE2N/UBE2V1 complex with the K48UBC1-C5 acceptors in the absence (left) or presence (right) of the E3 RNF4 RING domain. For all plots graphs, di-UB levels (μMol) represent the final time-points from the reactions (Source Data Extended Data Fig. 2), N=2 independent experiments. For samples derived from the same experiment, gels were processed in parallel.

Source data

Extended Data Fig. 3 1D and 2D proton NMR spectra for synthetic UBC4, recombinant UB, and K48UBC5 are highly superimposabe.

a, 2D Nuclear Overhauser effect spectroscopy (NOESY) recorded at 298 K and 1D spectra (b) for UBC4 (blue), recombinant UB (C4 Bio; pink) and K48UBC5 (purple). The 2D NOESY spectra show NOE interactions between amide protons (x-axis) and amino acid side-chain protons (y-axis), whereas the 1D spectra show signals from methyl protons (-0.5–1.0 ppm), Cα-protons (3.5 – 6 ppm) and amide protons (6 – 10 ppm). The signal from water is at 4.7 ppm1. The observed dispersion of signals demonstrates that all three UBs are well folded, while the comparable overlays indicate that the UBs share a highly similar fold. c, same as (a), except data were recorded at 310 K. d, same as (b), except at 310 K.

Extended Data Fig. 4 Lack of preference for a native lysine on acceptor UBs for the K11 chain-forming E2 UBE2S.

a, Cartoon of experimental scheme monitoring the reactivity of E2s with K11UBC1-C5 acceptors (UBA). Plot of the discharge of labeled UB (UB*) from UBE2S_IsoT to K11RUB, UBC4 Bio or K11UBC1-5 acceptors (left) and representative fluorescence scans of SDS-PAGE gels representing the primary data (right). b, same as (a), except in the presence of the E3 APC/C. c, same as (a), except in the presence of K11UBC2 or UBC4 Bio acceptors or the same harboring an E34D mutation. d, same as (b), except in the presence of K48RUB, UBC4 Bio or K48UBC1-5 acceptors. e, same as (a), except with UBE2N/UBE2V1. f, same as (b), except with UBE2N/UBE2V1 and the RING domain from the E3 RNF4. For all plots graphs, di-UB levels (μMol) represent the final time-points from the reactions (N=2 independent experiments). For samples derived from the same experiment, gels were processed in parallel.

Source data

Extended Data Fig. 5 The location of lysine analogues on acceptor UB impacts the distribution of di-UB chain linkage types generated by the E2 enzyme UBE2D3.

a, Plots showing the relative changes in UBE2D3 generated di-UB chain linkages in the presence of the RING domain from the E3 RNF4, comparing products containing K11UBC5 or UBC4 acceptors (N=3 technical replicates). ND, not determined. b, Same as (a), except with K63UBC5 acceptor.

Extended Data Fig. 6 Molecular Dynamics simulations reveal pleiotropic structural effects on UBs harboring lysine analogs.

a, Plot showing the degree of various side-chain rotamer interconversions for K11UBC5, K48UBC5, or K63UBC5 versus UBC4 acceptor UBs. Bins are divided by 120° intervals. b, Distribution of the distances between lysine acceptor amine and Cα atoms for UBC4 versus UBC5 during 25 ns MD simulations (N=3 independent experiments) for the UBE2N~UB/UBE2V1/acceptor UB multi-subunit complex. Bins are divided by 10° intervals. c, Plot showing the dynamics of φ and ψ main-chain torsion angles for UBC4 or UBC5 acceptors in the UBE2N~UB/UBE2V1/acceptor UB multi-subunit complex. Bins are divided by 10° intervals. d, Plot showing the dynamics of the side-chain rotamers for UBC4 or UBC5 acceptors in the UBE2N~UB/UBE2V1/acceptor UB multi-subunit complex. Bins are divided by 120° intervals. e, Rose plot showing the distance and angle of the amine acceptor of UBC4 relative to the active-site during 25 ns MD simulations of the UBE2N~UB/UBE2V1/acceptor UB multi-subunit complex (N=3 independent experiments). Golden star indicated starting position. f, same as (e), but with K63UBC5 (g) RMSD of gate loop during the trajectory for UBC4. h, same as (g), except with K63UBC5.

Extended Data Fig. 7 Di-UB chain formation reactions with UBC4 or UBC5 acceptors produce distinct results depending on the identity of the ubiquitin carrying enzyme.

a, Graph of the reactions velocities as a function of pH. performed in the presence of wild-type UBE2N/UBE2V1, radiolabeled K63R donor UB and either K63UBC4 or K63UBC5 acceptor UBs (b) same as (a), except with K92R UBE2N/UBE2V1. c, Graph of the reaction velocities as a function of the acceptor UB concentration for UBE2N/UBE2V1. The inset shows the fit of the data to the model for reactions containing K63UBC5 acceptor UB since the magnitude of the velocities is far less than reactions containing UBC4. d, same as (c) except in the presence of the RING domain of RNF4. e, Graph of the reaction velocities performed as a function of pH, in the presence of UBE2R2 and radiolabeled K48R donor UB and either UBC4 or K48UBC5 acceptors. f, Graph of the reaction velocities as a function of the acceptor UB concentration and their fit to the Michaelis-Menten model for UBE2R2. g, same as (f), except in the presence of the yeast HECT E3 Rsp5p. N=2 independent experiments. Samples derive from the same experiment, gels were processed parallelly.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–5 and Notes 1–3.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liwocha, J., Krist, D.T., van der Heden van Noort, G.J. et al. Linkage-specific ubiquitin chain formation depends on a lysine hydrocarbon ruler. Nat Chem Biol 17, 272–279 (2021). https://doi.org/10.1038/s41589-020-00696-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-00696-0

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