Abstract
Ubiquitination is a central process affecting all facets of cellular signaling and function1. A critical step in ubiquitination is the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to a substrate or a growing ubiquitin chain, which is mediated by E3 ubiquitin ligases. RING-type E3 ligases typically facilitate the transfer of ubiquitin from the E2 directly to the substrate2,3. The RBR family of RING-type E3 ligases, however, breaks this paradigm by forming a covalent intermediate with ubiquitin similarly to HECT-type E3 ligases4–6. The RBR family includes Parkin4 and HOIP, the central catalytic factor of the linear ubiquitin chain assembly complex (LUBAC)7. While structural insights into the RBR E3 ligases Parkin and HHARI in their overall autoinhibited forms are available8–13, no structures exist of intact fully active RBR E3 ligases or any of their complexes. Thus, the RBR mechanism of action has remained largely enigmatic. Here we present the first structure of the fully active HOIP-RBR in its transfer complex with an E2~ubiquitin conjugate, which elucidates the intricate nature of RBR E3 ligases. The active HOIP-RBR adopts a conformation markedly different from that of autoinhibited RBRs. HOIP-RBR binds the E2~ubiquitin conjugate in an elongated fashion, with the E2 and E3 catalytic centers ideally aligned for ubiquitin transfer, which structurally both requires and enables a HECT-like mechanism. In addition, surprisingly, three distinct helix–IBR-fold motifs inherent to RBRs form ubiquitin-binding regions that engage the activated ubiquitin of the E2~Ub conjugate as well as an additional regulatory ubiquitin molecule. The features uncovered reveal critical states of the HOIP-RBR E3 ligase cycle, and comparison with Parkin and HHARI suggests a general mechanism for RBR E3 ligases.
RBR E3 ligases are characterized by an extended RING domain (RING1) followed by an ‘in-between RING’ (IBR) domain and the catalytic domain, which is structurally an IBR domain but is commonly designated RING2 (Extended Data Fig. 1a,b)8–11,14. HOIP, one of the most studied RBRs, is the key E3 ligase of the LUBAC. It is a prototypical RBR yet contains an extended RING2 domain that includes the linear ubiquitin chain determining domain (LDD), enabling the selective formation of linear ubiquitin linkages (Extended Data Fig. 1c)5–7,14, and is thus denoted RING2L. The HOIP-RBR is kept in an autoinhibited state by the HOIP-UBA domain, whose sequestration by the LUBAC constituent HOIL-1L activates HOIP to trigger, together with SHARPIN, NF-κB signaling and other cellular processes5–7,15–20. To obtain the first insight into an active RBR in a key catalytic complex, we generated a stable E2~ubiquitin conjugate (UbcH5B C85K~ubiquitin)21 and isolated its complex with HOIP-RBR. The subsequent addition of free ubiquitin proved necessary for crystal formation, allowing us to solve the HOIP-RBR/UbcH5B~ubiquitin transfer complex structure at 3.5 Å resolution (Fig. 1a; Extended Data Figs. 2, 3).
The asymmetric unit contains two HOIP-RBR molecules interacting with two UbcH5B~ubiquitin conjugates and an additional ubiquitin or E2~ubiquitin conjugate, arranged in a swapped dimer configuration (Extended Data Fig. 3a). While this arrangement could have functional relevance, analysis of interfaces and biophysical examination (Extended Data Fig. 3b–f) indicate a monomeric assembly of the HOIP/E2~ubiquitin loading complex (Fig. 1), represented in the crystal structure by the RING1–IBR module (residues 699–852) from one HOIP molecule and the RING2L (residues 853–1072) from the second HOIP molecule in the asymmetric unit. In this assembly, the RING1–IBR module forms an elongated arm-like unit (Fig. 2a) that together with the RING2L embraces the E2~ubiquitin conjugate in a clamp-like manner (Fig. 1a). This active HOIP-RBR conformation is markedly different from previous structures of autoinhibited RBRs (Extended Data Fig. 1d) and enables an astounding array of features inherent to the active RBR. Most notably, three distinct helix–IBR-fold motifs function as essential discrete ubiquitin-binding regions (UBR) (Fig. 1b).
The HOIP-RING1/E2 interaction is tailored towards a HECT-like mechanism, setting it apart from classic RING E3 ligases. While RING/E2 interactions of both classic RING and RBR E3 ligases utilize similar surfaces (Extended Data Fig. 4a)21–26, the position of the HOIP-RING1 domain relative to the E2 is shifted compared to classic RING/E2 complexes (Fig. 2b). Therefore, the RBR-RING1 and the E2 do not form a composite surface to bind the E2-conjugated activated ubiquitin (Ubact, Extended Data Fig. 4b,c,e), which is key to the mechanism of classic RING E3 ligases21,24,27. Instead, two extension helices (hE1, hE2) link the RBR-RING1 to the IBR domain (Figs. 1, 2a)8–11, and helix hE2 with the IBR forms an UBR (UBR1) that engages the activated ubiquitin (Fig. 2c; Extended Data Fig. 5a,b). UBR1 binds ubiquitin in a distinctive mode (mode 1) that utilizes a salt bridge system involving HOIP-hE2 residues K783 and E787 and ubiquitin residues K11 and E34, with further support from the HOIP-IBR (Fig. 2c; Extended Data Fig. 5b). Thus, in HOIP the entire RING1–IBR arm mediates bipartate binding of the E2~ubiquitin conjugate, with RING1 binding E2 and the hE2–IBR module binding activated ubiquitin (Fig. 2a). Sequence and structural comparisons with Parkin and HHARI suggest conservation of this mechanism among RBR E3 ligases (Extended Data Figs. 4c, 5c). Importantly, the bipartite binding mode results in an elongated conformation of the E2~ubiquitin conjugate with its thioester linkage not suited for direct attack by the amine function of a substrate (Fig. 2d; Extended Data Fig. 4e–f). The consequence is an entirely different catalytic arrangement compared to classic RING-supported catalysis, as emphasized by the observed lack of effect of mutations in UbcH5B L104 and S108, two residues crucial for classic RING/E2 catalysis21,25,26 (Extended Data Fig. 4d). Instead, the E2~ubiquitin thioester is ideally positioned for transfer of the activated ubiquitin onto HOIP-RING2L, thus both enabling and requiring a HECT-like mechanism. The significance of each interaction site is demonstrated in thioester transfer and polyubiquitination assays, where mutation of key RING1 and UbcH5B residues drastically impairs RBR activity (Fig. 2e–g). Single mutations in the hE2 salt bridge moderately diminish activity in thioester assays but dramatically impair polyubiquitination activity (Fig. 2e,f), indicating a cumulative effect due to a potential role of UBR1 in coordinating Ubact in steps subsequent to its initial transfer to HOIP. However, removal of the salt-bridge in HOIP/ubiquitin double mutants and mutation of the HOIP-IBR/Ubact interface cause the expected drastic reduction in thioester transfer activity (Extended Data Fig. 5d–f).
The other portion of the RBR/E2~ubiquitin embrace is centered around the catalytic HOIP-RING2L (Figs.1a; 3a). Here a helix–IBR-fold motif consisting of helix hL2 from the IBR–RING2L linker and RING2L form a second UBR (UBR2) binding the activated ubiquitin (Fig. 3a; Extended Data Fig. 6a–c). UBR2, which is conserved in Parkin and HHARI (Extended Data Fig. 6d), uses a hydrophobic pattern in helix hL2 and RING2 to interact with the canonical and a second hydrophobic patch28 of ubiquitin (Extended Data Fig. 6b–e). These interactions support the engagement of the ubiquitin R72/R74 di-Arg motif by polar residues, which is the key characteristic of UBR binding mode 2. This ultimately places the ubiquitin C-terminus onto RING2 (Extended Data Fig. 6b,d) and thus the ubiquitin/E2-thioester linkage onto the RBR active site. A previous structure14 of the isolated HOIP-RING2L with two ubiquitin molecules bound in a linear non-covalent arrangement mimics the final HOIP-RING2L donor/acceptor ubiquitin transfer complex (Fig. 3b; Extended Data Fig. 6f). Remarkably, in this structure the donor ubiquitin adopts a position identical to the activated ubiquitin in HOIP-UBR2 despite lacking the hL2 interaction (Extended Data Fig. 6f). This indicates that the binding mode of the activated ubiquitin observed in the HOIP/E2~ubiquitin complex persists from the E2~ubiquitin/E3 HOIP transfer complex to the HOIP~ubiquitin/acceptor ubiquitin transfer complex. The E2 portion of the E2~ubiquitin conjugate instead interacts with a region of RING2L that overlaps with that observed for the acceptor ubiquitin in the second transfer reaction (Fig. 3b; Extended Data Fig. 6f). Thus, the binding of E2~ubiquitin and of acceptor ubiquitin/substrate are mutually exclusive, requiring formation of a covalent HECT-like RBR~ubiquitin intermediate in the RBR E3 ligase cycle.
Importantly, the HOIP-RBR/E2~ubiquitin complex structure lacks the spatial gap between the E2 and E3 catalytic centers that is frequently observed in HECT/E2 complex structures and that was also predicted for RBR/E2~ubiquitin transfer complexes8–11,28,29. Thus, except for the ~3.5 Å spacer due to the C85 to lysine substitution in the E2~ubiquitin conjugate, the HOIP-RBR/E2~ubiquitin structure accurately depicts the immediate transfer complex. Here the catalytic centers of HOIP-RING2L and E2 come in close proximity via two contact conduits involving all three proteins (Fig. 3c; Extended Data Fig. 7a). The first conduit consists of ubiquitin R72, which interacts with D983 and Q974 in the β5/6-hairpin of HOIP-RING2L. Additionally, E976 in this hairpin mediates a salt bridge with ubiquitin R74 and UbcH5B R90, thus facilitating interactions among all three proteins.
The second conduit consists of catalytic residues of UbcH5B (N77, D117) and HOIP (H887, Q896)5,6,10,14,21–24,26. These residues appear permissive to close proximity between the reaction centers, yet not crucial for transesterification because, for example, a H887A mutation does not affect the thioester transfer reaction10,14 (Extended Data Fig. 7b). Surprisingly mutation to alanine of UbcH5B D117, a critical residue for classic RING-supported catalysis21,24, enhances transesterification (Extended Data Fig. 7b) further underlining the vastly different catalytic mechanism of RBR E3 ligases. This finding also points to a trade-off in the E2 active site to support both classic RING and HECT-type RBR E3 ligases. Interestingly UbcH7, which is specialized for HECT-like E3 catalysis4, features a histidine instead of D117 (Extended Data Fig. 7a). Mutational analysis demonstrates a crucial role for conduit 1 and also indicates that the close proximity between the ubiquitin thioester (and thus C85 of UbcH5B) and HOIP catalytic cysteine C885 is the driving factor for E2/RBR E3 ubiquitin transfer (Fig. 3c,d; Extended Data Fig. 7). Analysis of Parkin and HHARI shows conservation of the conduits (Extended Data Fig. 7a). However, while Parkin and HHARI lack the β5/6-hairpin indigenous to HOIP-RING2L, they instead possess a pair of conserved polar residues in the RING2 active site loop that are capable of binding the di-Arg motif in conduit 1.
Surprisingly, our structure reveals that an additional allosteric ubiquitin molecule (Uballo) interacts with a third HOIP-UBR. UBR3 is located in the RING1–IBR arm immediately across UBR1 and Ubact (Figs.1b,4a; Extended Data Fig. 8a,b). Uballo uses a binding mode similar to that of Ubact with UBR2 (mode 2), characterized by hydrophobic interactions and a di-arginine binding clamp (Extended Data Fig. 8a). Uballo interacts with helix hE2 of the extended RING1 and with the IBR, and makes additional interactions with helix hE1 (Fig. 4a, Extended Data Fig. 8c). Through this binding, Uballo induces a “straight” conformation of helix hE2 locking RING1 and IBR in their relative position, forming UBR1 to accommodate the activated ubiquitin (Fig. 4a; Extended Data Fig. 8b–d). Notably, UBR3 in the HOIP-RBR/UbcH5B~ubiquitin complex binds linear di-ubiquitin (Kd=7μM) better than mono-ubiquitin (Kd>50 μM) (Extended Data Fig. 9a). Importantly, pre-incubation of HOIP-RBR with linear di-ubiquitin leads to improved binding of UbcH5B~ubiquitin (Extended Data Fig. 9b), emphasizing the allosteric function of UBR3. Accordingly, HOIP-UBR3 I807A and E809A mutants show moderately decreased activity in thioester transfer assays but more pronounced effects in polyubiquitination assays, where linear di-ubiquitin/polyubiquitin are intrinsically produced (Fig. 4b; Extended Data Fig. 9c). Importantly, N- and C-terminally capped linear di-ubiquitin increases HOIP-RBR thioester transfer activity in a dose-dependent manner, but cannot activate HOIP-UBR3 mutants (Fig. 4b; Extended Data Fig. 9d). Moreover, the linear di-ubiquitin I44A mutant also fails to activate HOIP-RBR (Extended Data Fig. 9d).
Excitingly, the interaction of Uballo with UBR3 is structurally similar to that recently reported for phospho-ubiquitin in a tethered complex with ΔUBL-Pediculus humanus Parkin (Ph-Parkin)13 (Extended Data Fig. 8c–e). Binding of phospho-ubiquitin leads to a straight conformation of Parkin helix hE2 and an accompanying reorientation of RING1 and IBR, indicating a general role of UBR3 and ubiquitin in allosteric regulation of RBR proteins. Functionally, binding of phospho-ubiquitin activates Parking by counteracting the autoinhibitory function of the Parkin UBL domain (Extended Data Fig. 8c–e)13. In HOIP, the UBA domain exerts intramolecular autoinhibition5,6. While the structure of autoinhibited HOIP is unknown, the structure of autoinhibited HHARI shows binding of its UBA domain to a region analogous to UBR3 (Extended Data Fig. 8f–h)11. To determine if linear di-ubiquitin can overcome HOIP autoinhibition, we examined its effect on HOIP-UBA–RBR. As expected, HOIP-UBA–RBR alone exhibits low E3 activity but is activated by HOIL-1L (Fig. 4c). Strikingly, linear di-ubiquitin can also remove HOIP-UBA autoinhibition and at high concentrations allows the processive formation of polyubiquitin chains by HOIP-UBA–RBR (Fig. 4c). Importantly, in HEK293T cells expressing full-length HOIP, the HOIP-UBR3 I807A and E809A mutants fail to activate NF-κB regardless of HOIL-1L expression, demonstrating an essential physiological role of UBR3 (Fig. 4d). Thus, UBR3 likely serves as a critical sensor of ubiquitin chains that regulates LUBAC function. Whether this role is tailored to linear ubiquitin chains or ubiquitin chains in general (Extended Data Fig. 9b,e) needs further investigation in the context of other LUBAC constituents and binding partners.
The features revealed by the HOIP-RBR/E2~Ubact/Uballo complex structure provide the missing links in our understanding of these enigmatic multidomain E3 ligases8–11,14 and yield a mechanistic model for the RBR E3 ubiquitin ligase cycle, as summarized in Extended Data Fig. 10. Furthermore, the conservation of key mechanistic features in HOIP, HHARI, Parkin and other RBRs (Supplementary Data 2) underlines the general nature of the catalytic RBR cycle unraveled in this study.
Methods
No statistical methods were used to predetermine sample size.
Constructs
Human HOIP and HOIL-1L cDNA were purchased from Open Biosystems (cloneIDs 4653017 and 3877587, respectively). HOIP-RBR (residues 696–1072), HOIP-RING2L (residues 853–1072) and full-length HOIL-1L were cloned into the pET-NKI-6xHis-3C-LIC vector30 coding for an N-terminal 6x-His-tag with a 3C protease cleavage site. HOIP-UBA-RBR (residues 475–1072) was cloned into a pET-NKI-6xHis-eGFP-3C-LIC vector that codes for a 3C-cleavable His-tagged enhanced green-fluorescent protein (eGFP) followed by the HOIP sequence. Human UbcH5B and Cdc34 DNA were a gift of M. Petroski. Coding sequences for UbcH13 and Uev1a were extracted out of a human cDNA library (Agilent Megaman). For crystallization, UbcH5B (residues 2–147) with the mutations S22R (to prevent backside ubiquitin binding31) and C85K (to enable covalent ubiquitin linkage21) was cloned into the pET-NKI-6xHis-3C-LIC vector. UbcH5B without S22R and C85K mutations (used for enzymatic assays), Cdc34, UbcH13 and Uev1a were cloned into the same vector. Untagged mono-ubiquitin with native N- and C-termini, used for crystallization and linear ubiquitination assays, was cloned into the pET29 vector (Novagen) using NdeI/XhoI restriction sites. N-terminally blocked mono-ubiquitin used for thioester assays was cloned in the pET-NKI-6xHis-3C-LIC vector. Untagged linear di-ubiquitin was cloned with overlap extension PCR and ligated into the pET29 vector (Novagen) using NdeI/XhoI restriction sites. N- and C-terminally blocked di-ubiquitin with a N-terminal His-tag and a C-terminal Ala-Ser sequence was cloned into the pET-NKI-6xHis-3C-LIC vector. Human ubiquitin activating enzyme E1 (Ube1) was cloned into a pET28 vector resulting in an N-terminal His-tag. For NFκB assays full-length HOIP with an N-terminal FLAG-tag and HOIL-1L with an N-terminal myc-tag were cloned into pcDNA3.1(+) (Invitrogen) using EcoRI/NotI restriction sites. Mutations in UbcH5B, ubiquitin and HOIP were introduced using standard site-directed mutagenesis techniques.
Protein expression and purification
All proteins were expressed in BL21(DE3) E. coli after induction with 0.5 mM IPTG overnight at 20°C. For expression of HOIP and HOIL-1L constructs, 0.5 mM ZnCl2 was added to the cultures before induction. Bacteria were harvested by centrifugation, lysed by addition of lysozyme and sonication in the presence of protease inhibitors (PMSF and leupeptin) and DNase. Lysates were cleared by centrifugation and His-tagged proteins were initially purified using Ni-NTA agarose (Qiagen). For HOIP RBR used for crystallization, and UbcH5B, Cdc34, UbcH13, Uev1a, wild-type ubiquitin to generate K48-linked di-ubiquitin and HOIL-1L His tags were removed by addition of 3C protease overnight at 4 °C. HOIP-RBR and HOIL-1L were further purified using Superdex 200 10/300 GL or HiLoad 16/600 Superdex 200 pg size exclusion chromatography columns (GE Healthcare) equilibrated in protein buffer (10 mM HEPES pH 7.9, 100 mM NaCl). UbcH5B used for biochemical assays was further purified on a Superdex 75 10/300 GL size exclusion chromatography column (GE Healthcare) equilibrated in protein buffer. HOIP mutants for activity assays, and Cdc34, UbcH13 and Uev1a were desalted into protein buffer directly after Ni-NTA purification using PD MidiTrap G-25 desalting columns (GE Healthcare). Ube1 for biochemical assays was further purified using ion exchange chromatography (Source Q) in 10 mM HEPES pH 7.9, 10 mM NaCl and eluted with a gradient from 10 – 500 mM NaCl. N-terminally His-tagged (di)-ubiquitin was purified using Ni-NTA as described above followed by size exclusion chromatography using a Superdex 75 10/300 GL column (GE Healthcare) equilibrated in protein buffer or buffer exchange into protein buffer using PD MidiTrap G-25 desalting columns. To purify untagged mono- or di-ubiquitin, 0.5 mM EDTA and 100 mM sodium acetate pH 4.5 were added to the bacterial lysates and lysates were cleared by centrifugation, diluted 7-fold with 50 mM sodium acetate pH 4.5 and applied to a Source S 10/100 ion exchange column (GE Healthcare) equilibrated in 50 mM sodium acetate pH 4.5. Ubiquitin was eluted with a 0 – 500 mM NaCl gradient and further purified by size exclusion chromatography on a Superdex 75 10/300 GL column (GE Healthcare) equilibrated in protein buffer. His-eGFP-HOIP was purified using size exclusion chromatography as described for HOIP-RBR, followed by 3C cleavage and removal of His-eGFP via a second round of size exclusion chromatography. All proteins were generally flash frozen in liquid nitrogen in small aliquots and stored at −80°C.
UbcH5B~ubiquitin linkage
UbcH5B~ubiquitin linkage was performed based on published methods21. Briefly, Ube1, UbcH5B(S22R/C85K) and ubiquitin were mixed and buffer exchanged into 50 mM Tris pH 10, 150 mM NaCl using PD-10 desalting columns (GE Healthcare). 10 mM MgCl2, 5 mM ATP and 1 mM TCEP were added and the protein was incubated at 37°C for 16 h. The completeness of the reaction was monitored using SDS-PAGE and covalently linked UbcH5B~ubiquitin was purified from unreacted proteins and Ube1 using a Superdex 75 10/300 GL size exclusion chromatography column (GE Healthcare) equilibrated in protein buffer.
HOIP-RBR/UbcH5B~ubiquitin complex formation
HOIP-RBR was mixed with a 1.3-fold molar excess of UbcH5B~ubiquitin and applied to a Superdex 200 10/300 GL size exclusion chromatography column equilibrated in protein buffer. Complex formation and purity was confirmed using SDS-PAGE, and complex containing fractions were pooled and concentrated to ~12 mg/ml for crystallization.
HOIP/UbcH5B~ubiquitin/ubiquitin crystallization
Crystallization was performed using the vapour diffusion technique in sitting drop MRC 96-well plates (Molecular Dimensions). Initial crystals were obtained mixing HOIP/UbcH5B~ubiquitin complex solution with an equimolar amount of free ubiquitin in the Morpheus Screen (Molecular Dimensions). Subsequently, 2 μl of the protein complex were mixed with 0.6 μl reservoir solution (0.1 M Morpheus Buffer 3 pH 8.5 (Tris/Bicine), 0.12 M Morpheus Alcohols Mix (0.02 M each of 1,6-Hexanediol; 1-Butanol; 1,2-Propanediol (racemic); 2-Propanol; 1,4-Butanediol; 1,3-Propanediol), 30% Morpheus P550MME_P20K mix (20% PEG550MME, 10% PEG20K) and 8% glycerol) in MRC 48-well plates (Molecular Dimensions). Crystals appeared after about one week at 12°C and were cryo-cooled, and evaluated on a rotating anode X-ray generator (Rigaku FR-E superbright). Seeding and dehydration of the crystals was performed to improve crystal diffraction. For successful dehydration, reservoir was slowly added to the protein drop (3x 0.5 μl within ~2 h) and subsequently equilibrated overnight at 12°C against a reservoir solution with increased P550MME_P20K concentration by adding 11 μl 60% Morpheus P550MME_P20K stock solution to 50 μl reservoir solution. The new reservoir solution was then slowly added to the protein drop (3x 0.5 μl, followed by 2x 1 μl with removal of 1 μl each in the last steps). After further overnight equilibration, crystals were harvested from the drop and directly cryo-cooled in a cryogenic nitrogen stream at 100 K. Crystals diffracted in house to 4–6 Å. Complete diffraction data were measured at 100 K at beamline 23ID-D of the General Medical Sciences and Cancer Institutes Structural Biology Facility at the Advanced Photon Source (GM/CA @ APS), Argonne National Laboratory. Despite their size (common dimensions of ~200 × 140 × 100 μm3) crystals exhibited substantial inhomogeneity resulting in split and smeared diffraction spots. Using raster scans32, a suitable region for data collection could be identified at the edge of the crystal. Utilizing a small (20 μm diameter) beam, split spots could be separated to allow reliable indexing and integration. Utilization of a small beam necessitated higher flux to retain reliable diffraction. To mitigate the radiation damage, the total dose was distributed over a 100-μm stretch of the crystal by using the “helical” mode of “vector” data collection as implemented in JBluIce33. Data were measured at 1.282 Å wavelength with a Pilatus3 6m pixel array detector with a 1 mm-thick sensor (Dectris).
Data processing and structure solution
Data were collected from a single crystal and indexed, integrated and scaled in XDS/XSCALE34. Data were further processed using AIMLESS35 from the CCP4 suite36 with a resolution cut-off of 3.48 Å, resulting in an <I/σI> = 1.6 and CC1/2 = 0.648 in the highest resolution shell. Phasing was done in Phaser37 using the MR-SAD protocol as implemented in PHENIX38. For this, independent molecular replacement searches were initially performed for the RING2L domain of HOIP (from PDBid: 4LJP14), UbcH5B (from PDBid: 3A3339), and ubiquitin (from PDBid: 4LJP14) with the four C-terminal residues deleted. Various ambiguous solutions were identified that could not be separated, and Zn2+ sites could not be identified using MR-SAD due to incompleteness of resultant models. However, manual inspection revealed that some MR solutions contained ubiquitin oriented near identically to the symmetry-related donor ubiquitin observed in the HOIP-RING2L/ubiquitin-ubiquitin transfer complex (PDBid: 4LJP14). Based on this observation, a trimmed search model was created that contained a complex of the core of HOIP-RING2L (with residues 906–924 and 949–999 removed) and C-terminally truncated ubiquitin. An MR search using this model found a single solution for two copies of the complex. After successful iterative searches for two UbcH5B molecules and two further ubiquitin molecules, MR-SAD using Phaser identified 15 distinct Zn2+ sites including the known Zn2+ sites in the RING2L domain of HOIP. Further molecular replacement in Phaser using a single unit of the initial HOIP-RING2L/UbcH5B~ubiquitin complex (without the additional second ubiquitin), and the NMR structure of HOIP-IBR (zinc atoms removed, deposited in Protein Data Bank40 under PDBid: 2CT7, unpublished) correctly placed a single HOIP-IBR domain, which was then manually copied to the other NCS-related HOIP in the asymmetric unit. For molecular replacement of the HOIP-RING1, Sculptor41 was used to generate a search model based on the structure of the RING1 domain of HHARI (PDBid: 4KBL11). However, Phaser was not able to correctly place this domain, probably due to the low sequence conservation of only 27% identity. However, since mutational analysis of HOIP suggested that the RING/E2 interaction is preserved between RING-type E3 ligases and RBR-type E3 ligases5, we overlaid the E2 of the published RNF4-RING/UbcH5A~ubiquitin structure (PDBid: 4AP421) with the E2 in our structure and then used this overlay to add the RING1 model generated by Sculptor. This overlay placed the HOIP-RING1 Zn2+-coordinating residues near the last remaining free Zn2+-ions found earlier by Phaser MR-SAD, indicating correct placement of the RING1 domain. In the final round of molecular replacement, the two additional ubiquitin (Uballo) molecules were reinstated at the RING1-IBR interface. At this stage, Refmac42 was used for refinement using settings optimized for low-resolution refinement43 including “jelly body refinement” and Babinet scaling. ProSMART44 was used to generate external restraints against high-resolution structures (PDBid: 4LJO14 for HOIP-RING2L and ubiquitin, and PDBid: 2ESK45 for UbcH5B). After this, clear extra electron density became visible for the unmodelled helical linker regions of the RING1-IBR and IBR-RING2L transitions and for other regions omitted in the initial search models. Further model building and refinement was manually performed in Coot46 and Refmac. During refinement additional clear positive difference map electron density became visible and Phaser was used to place one additional UbcH5B molecule (UbcH5Badd) into this density. TLS restraints were generated using the TLSMD server47 and NCS restraints were used throughout refinement. One overall B-factor was refined in Refmac. In later rounds of refinement the PDB_REDO server48 was used for refinement optimization and MolProbity49 was used for structure validation. Data processing and refinement statistics are summarized in Extended Data Fig. 2b. Ramachandran statistics were calculated using MolProbity and 94.8% of all residues are in favoured regions, 4.9% in allowed regions and 0.3% are outliers. The final structure has a MolProbity score of 1.75 (100th percentile). In the final structure the two HOIP-RBR molecules (see also Extended Data Fig. 3) are defined by electron density from residues 699 to 707, 711 to 948, 969 to 991, and 996 to 1011 (chain A) and 699 to 754, 760 to 957, 967 to 1015, 1019 to 1035 and 1054 to 1066 (chain B). The catalytic UbcH5B~ubiquitin conjugates are defined from UbcH5B residues 3 to 147 and ubiquitin residues 1 to 76 (chains C and E), and UbcH5B residues 2 to 147 and ubiquitin residues 1 to 76 (chains D and F). The allosteric ubiquitin chains (chains G and H) are defined from residues 1 to 76 and the additional UbcH5B (chain I) is defined from residues 2 to 146. Phenix was used to calculate simulated annealing (SA) composite omit maps and feature enhanced maps (FEM). All molecular figures were prepared in PyMOL (Schroedinger, LLC).
K48-linked and K63-linked ubiquitin chain formation
K48-linked and K63-linked ubiquitin chains were formed through a linkage specific enzymatic reaction using Cdc34 and UbcH13/Uev1a E2 ubiquitin-conjugating enzymes, respectively, as described in the literature50. Ubiquitin chains were separated using ion exchange chromatography as described above for purification of mono-ubiquitin. Purified K48-linked di-ubiquitin was directly desalted into protein buffer using PD-10 desalting columns, whereas K63-linked di-ubiquitin was further purified on a Superdex 75 10/300 GL size exclusion chromatography column equilibrated in protein buffer. Native ubiquitin without additional residues was used to generate di-ubiquitin chains for ITC experiments, whereas N-terminally blocked ubiquitin was used to form K48-linked di-ubiquitin for testing allosteric activation of HOIP RBR.
Linear polyubiquitination assay
Linear ubiquitin formation assays were performed in 50mM HEPES pH7.9, 100 mM NaCl, 10 mM MgCl2 and 0.6 mM DTT using 200 nM E1, 1 μM UbcH5B, 1 μM HOIP-RBR or HOIP-RING2L and 40 μM untagged ubiquitin. Reactions were started by addition of 10 mM ATP and were incubated at 30°C for 2 h. Samples were taken at the indicated time points and treated with 50 mM sodium acetate pH4.5 as described previously6, mixed with SDS sample buffer and analysed by SDS-PAGE using 12% Bolt Bis-Tris gels (Life technologies). Proteins were visualized with Coomassie Brilliant blue dye. To test the activating effect of linear di-ubiquitin on autoinhibited HOIP-UBA-RBR, 5μM HOIP-UBA-RBR was pre-incubated with N- and C-terminally blocked linear di-ubiquitin or HOIL-1L at the indicated concentrations before addition of the remaining assay components. Samples were taken after 60 min and subsequently treated as described above.
UbcH5B~ubiquitin to HOIP-RBR ubiquitin transfer assay
To monitor HOIP~ubiquitin thioester ubiquitin transfer from UbcH5B to HOIP, Ube1 (100 nM), UbcH5B (4 μM) and N-terminally blocked ubiquitin (32 μM) were mixed in 50 mM HEPES pH 7.9, 100 mM NaCl, 10 mM MgCl2 and 5 mM ATP and incubated at 25°C for 5 min when 2 μM HOIP-RBR was added. Samples were taken 10 s after HOIP addition, quenched by addition of pre-heated SDS protein loading buffer without DTT, and run on a 12% SDS-PAGE gel (Life Technologies). The 10-second time point used was empirically determined with a time-course experiment (Extended Data Fig. 9g). Gels were stained with Coomassie Brilliant blue dye and scanned on a Li-COR Odyssey scanner using the 700 nm (red) channel. For the thioester transfer assay shown in Fig. 3d, 200 nM Ube1, 2 μM UbcH5B, 8 μM HOIP-RBR, 8 μM N-terminally blocked ubiquitin and 10 mM ATP were used and samples taken after 30 sec. Proteins were transferred to a PVDF membrane and ubiquitin was visualized on a LI-COR Odyssey scanner at 800 nm using an anti-ubiquitin antibody (P4D1, Santa Cruz, 1:200 dilution in TBST [50 mM Tris pH 7.4, 150 mM NaCl, 0.05% Tween-20]) followed by an IRDye 800CW secondary antibody (LI-COR, 1:10,000 dilution in TBST). All quantitative experiments shown in graphs were performed in triplicates and band intensities were quantified using the ImageStudio software (LI-COR). HOIP thioester transfer activity was calculated as the fraction of HOIP~Ub to total HOIP for each mutant and normalized against thioester transfer activity of WT HOIP. Data were analysed in GraphPad Prism using two-tailed unpaired Student’s t-test or one-way ANOVA followed by Tukey’s post-test.
Allosteric activation of HOIP-RBR by di-ubiquitin
To test the allosteric activation of HOIP-RBR by linear di-ubiquitin, a modified ubiquitin transfer assay was performed. HOIP-RBR was pre-incubated with N- and C-terminally blocked linear di-ubiquitin at the indicated final concentrations for 5 min at 25°C. At the same time, Ube1, UbcH5B, ubiquitin and ATP were premixed and incubated for 5 min at 25°C, resulting in fully loaded UbcH5B~Ub. Both mixtures were subsequently mixed together, resulting in final concentrations of 100 nM Ube1, 2 μM UbcH5B, 8 μM N-terminally blocked ubiquitin and 2 μM HOIP-RBR in the final buffer described for the standard ubiquitin transfer assay. Samples were taken after 30 s and further treated as described for the standard transfer assay. A 30 sec time point was determined to give the best results in this assay, in which lower E2 and mono-ubiquitin concentrations were used resulting in an overall slower reaction rate. The experiments comparing the effects of linear versus K48-linked di-ubiquitin (Extended Data Fig. 9e) were performed similarly, with the difference that all samples were incubated with apyrase (Sigma) for 5 min to deplete ATP before addition of HOIP/di-ubiquitin and prevent E2-loading of K48-linked di-ubiquitin, which features a free C-terminus on one of the ubiquitin units.
Analytical ultracentrifugation (AUC)
Sedimentation equilibrium experiments were performed in a ProteomeLab XL-I (Beckman Coulter) analytical ultracentrifuge. HOIP-RBR/UbcH5B~ubiquitin as used for crystallization was loaded into a 6-channel equilibrium cell at 5.0, 2.5 and 1.25 μM concentration and centrifuged at 10,000 rpm, 20°C in an An-50 Ti 8-place rotor until equilibrium was achieved. Data were analysed using HeteroAnalysis software (J.L. Cole and J.W. Lary, University of Connecticut; http://www.biotech.uconn.edu/auf/).
Isothermal titration calorimetry (ITC)
ITC experiments were performed on an ITC200 calorimeter (Microcal). Aliquots (2 μl each) of 500–650 μM UbcH5B~ubiquitin or di-ubiquitin solution were injected into the cell containing 40–50 μM HOIP-RBR or HOIP-RBR/di-ubiquitin complexes. The experiments were performed at 23°C in buffer containing 10 mM HEPES pH 7.9, 100 mM NaCl. For titrations of UbcH5B~ubiquitin into HOIP-RBR/di-ubiquitin complexes, HOIP-RBR was pre-incubated with an equimolar amount of di-ubiquitin before the ITC experiments. Data were analysed using the Origin software (Microcal).
NF-κB luciferase assay
Human embryonic kidney (HEK) 293T cells (ATCC) were co-transfected with NF-κB-luc reporter plasmid that contains an NF-κB response element upstream of the promoter driving the luciferase reporter gene, pGL4.74[hRluc/TK] control vector (Promega) and epitope tagged FLAG-HOIP or myc-HOIL-1L pcDNA3.1(+) plasmids in 6-well plates in triplicates using Lipofectamine 2000 transfection reagent. Since this assay could be carried out in a variety of cellular contexts, HEK293T cells were used because they are easy to transfect and suitable for the assay. The cells tested negative for mycoplasma contamination. Empty pcDNA3.1(+) vector was used as control. After 36 h, cells were lysed and 20 μl cell lysates were used to measure firefly luciferase and Renilla luciferase (transfection control) signals using the dual luciferase reporter assay system according to the manufacturer’s protocol (Promega). Data were analysed in GraphPad Prism and one-way ANOVA followed by Tukey post-tests were used for statistical analysis. Immunoblotting was performed with anti-FLAG (clone M2, Sigma-Aldrich) and anti-myc (clone 9E10, Sigma-Aldrich) antibodies, to confirm equivalent WT and mutant protein expression levels.
Extended Data
Supplementary Material
Acknowledgments
The authors thank A. Bobkov (SBP Protein Analysis Facility) for performing ITC and AUC experiments, M. Petroski (SBP) for providing UbcH5B and Cdc34 constructs, J. Badger (DeltaG technologies) for assistance in model evaluation and E. Pasquale (SBP) for help with manuscript writing. This work was supported by NIH grant R01AA017238 and institutional funding (S.J.R.), an EMBO Long-term Postdoctoral Fellowship (B.C.L.), a Rutherford Discovery Fellowship from the New Zealand government administered by the Royal Society of New Zealand (P.D.M.) and NCI Cancer Center Support Grant P30CA030199 (SBP Protein Analysis Core Facility). This research used resources of the Advanced Photon Source, a U.S. Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No. DE-AC02-06CH11357. GM/CA@APS has been funded in whole or in part with Federal funds from the National Cancer Institute (ACB-12002) and the National Institute of General Medical Sciences (AGM-12006).
Footnotes
Online Content Methods, along with any additional Extended Data display items and Source Data, are available in the online version of the paper; references unique to these sections appear only in the online paper.
Supplementary Information is available in the online version of the paper.
Author Contributions B.C.L. designed and carried out all experiments (except for the cell-based experiments), including crystallization, structure solution and refinement, and wrote the manuscript. M.K.D. expressed proteins and performed initial purification. P.D.M. participated in early stages of the study, structure solution and writing of the manuscript. R.S. collected and processed diffraction data. A.R. performed the HEK293T cell experiments under the supervision of C.F.W. S.J.R. oversaw and actively participated in all steps of the study and wrote the manuscript.
Coordinates and structure factors have been deposited in the Protein Data Bank under accession code 5EDV.
The authors declare no competing financial interests.
References
- 1.Komander D, Rape M. The ubiquitin code. Annu Rev Biochem. 2012;81:203–229. doi: 10.1146/annurev-biochem-060310-170328. [DOI] [PubMed] [Google Scholar]
- 2.Metzger MB, Pruneda JN, Klevit RE, Weissman AM. RING-type E3 ligases: master manipulators of E2 ubiquitin-conjugating enzymes and ubiquitination. Biochim Biophys Acta. 2014;1843:47–60. doi: 10.1016/j.bbamcr.2013.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Berndsen CE, Wolberger C. New insights into ubiquitin E3 ligase mechanism. Nat Struct Mol Biol. 2014;21:301–307. doi: 10.1038/nsmb.2780. [DOI] [PubMed] [Google Scholar]
- 4.Wenzel DM, Lissounov A, Brzovic PS, Klevit RE. UBCH7 reactivity profile reveals parkin and HHARI to be RING/HECT hybrids. Nature. 2011;474:105–108. doi: 10.1038/nature09966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Smit JJ, Monteferrario D, Noordermeer SM, van Dijk WJ, van der Reijden BA, Sixma TK. The E3 ligase HOIP specifies linear ubiquitin chain assembly through its RING-IBR-RING domain and the unique LDD extension. EMBO J. 2012;31:3833–3844. doi: 10.1038/emboj.2012.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Stieglitz B, Morris-Davies AC, Koliopoulos MG, Christodoulou E, Rittinger K. LUBAC synthesizes linear ubiquitin chains via a thioester intermediate. EMBO Rep. 2012;13:840–846. doi: 10.1038/embor.2012.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kirisako T, Kamei K, Murata S, Kato M, Fukumoto H, Kanie M, Sano S, Tokunaga F, Tanaka K, Iwai K. A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J. 2006;25:4877–4887. doi: 10.1038/sj.emboj.7601360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Riley BE, Lougheed JC, Callaway K, Velasquez M, Brecht E, Nguyen L, Shaler T, Walker D, Yang Y, Regnstrom K, Diep L, Zhang Z, Chiou S, Bova M, Artis DR, Yao N, Baker J, Yednock T, Johnston JA. Structure and function of Parkin E3 ubiquitin ligase reveals aspects of RING and HECT ligases. Nat Commun. 2013;4:1982. doi: 10.1038/ncomms2982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Trempe JF, Sauve V, Grenier K, Seirafi M, Tang MY, Menade M, Al-Abdul-Wahid S, Krett J, Wong K, Kozlov G, Nagar B, Fon EA, Gehring K. Structure of parkin reveals mechanisms for ubiquitin ligase activation. Science. 2013;340:1451–1455. doi: 10.1126/science.1237908. [DOI] [PubMed] [Google Scholar]
- 10.Wauer T, Komander D. Structure of the human Parkin ligase domain in an autoinhibited state. EMBO J. 2013;32:2099–2112. doi: 10.1038/emboj.2013.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Duda DM, Olszewski JL, Schuermann JP, Kurinov I, Miller DJ, Nourse A, Alpi AF, Schulman BA. Structure of HHARI, a RING-IBR-RING ubiquitin ligase: autoinhibition of an Ariadne-family E3 and insights into ligation mechanism. Structure. 2013;21:1030–1041. doi: 10.1016/j.str.2013.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kumar A, Aguirre JD, Condos TE, Martinez-Torres RJ, Chaugule VK, Toth R, Sundaramoorthy R, Mercier P, Knebel A, Spratt DE, Barber KR, Shaw GS, Walden H. Disruption of the autoinhibited state primes the E3 ligase parkin for activation and catalysis. EMBO J. 2015 doi: 10.15252/embj.201592337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wauer T, Simicek M, Schubert A, Komander D. Mechanism of phospho-ubiquitin-induced PARKIN activation. Nature. 2015;524:370–374. doi: 10.1038/nature14879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Stieglitz B, Rana RR, Koliopoulos MG, Morris-Davies AC, Schaeffer V, Christodoulou E, Howell S, Brown NR, Dikic I, Rittinger K. Structural basis for ligase-specific conjugation of linear ubiquitin chains by HOIP. Nature. 2013;503:422–426. doi: 10.1038/nature12638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gerlach B, Cordier SM, Schmukle AC, Emmerich CH, Rieser E, Haas TL, Webb AI, Rickard JA, Anderton H, Wong WW, Nachbur U, Gangoda L, Warnken U, Purcell AW, Silke J, Walczak H. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature. 2011;471:591–596. doi: 10.1038/nature09816. [DOI] [PubMed] [Google Scholar]
- 16.Ikeda F, Deribe YL, Skanland SS, Stieglitz B, Grabbe C, Franz-Wachtel M, van Wijk SJ, Goswami P, Nagy V, Terzic J, Tokunaga F, Androulidaki A, Nakagawa T, Pasparakis M, Iwai K, Sundberg JP, Schaefer L, Rittinger K, Macek B, Dikic I. SHARPIN forms a linear ubiquitin ligase complex regulating NF-kappaB activity and apoptosis. Nature. 2011;471:637–641. doi: 10.1038/nature09814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tokunaga F, Nakagawa T, Nakahara M, Saeki Y, Taniguchi M, Sakata S, Tanaka K, Nakano H, Iwai K. SHARPIN is a component of the NF-kappaB-activating linear ubiquitin chain assembly complex. Nature. 2011;471:633–636. doi: 10.1038/nature09815. [DOI] [PubMed] [Google Scholar]
- 18.Yagi H, Ishimoto K, Hiromoto T, Fujita H, Mizushima T, Uekusa Y, Yagi-Utsumi M, Kurimoto E, Noda M, Uchiyama S, Tokunaga F, Iwai K, Kato K. A non-canonical UBA-UBL interaction forms the linear-ubiquitin-chain assembly complex. EMBO Rep. 2012;13:462–468. doi: 10.1038/embor.2012.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sasaki K, Iwai K. Roles of linear ubiquitinylation, a crucial regulator of NF-kappaB and cell death, in the immune system. Immunol Rev. 2015;266:175–189. doi: 10.1111/imr.12308. [DOI] [PubMed] [Google Scholar]
- 20.Tokunaga F, Sakata S, Saeki Y, Satomi Y, Kirisako T, Kamei K, Nakagawa T, Kato M, Murata S, Yamaoka S, Yamamoto M, Akira S, Takao T, Tanaka K, Iwai K. Involvement of linear polyubiquitylation of NEMO in NF-kappaB activation. Nat Cell Biol. 2009;11:123–132. doi: 10.1038/ncb1821. [DOI] [PubMed] [Google Scholar]
- 21.Plechanovova A, Jaffray EG, Tatham MH, Naismith JH, Hay RT. Structure of a RING E3 ligase and ubiquitin-loaded E2 primed for catalysis. Nature. 2012;489:115–120. doi: 10.1038/nature11376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mace PD, Linke K, Feltham R, Schumacher FR, Smith CA, Vaux DL, Silke J, Day CL. Structures of the cIAP2 RING domain reveal conformational changes associated with ubiquitin-conjugating enzyme (E2) recruitment. J Biol Chem. 2008;283:31633–31640. doi: 10.1074/jbc.M804753200. [DOI] [PubMed] [Google Scholar]
- 23.Plechanovova A, Jaffray EG, McMahon SA, Johnson KA, Navratilova I, Naismith JH, Hay RT. Mechanism of ubiquitylation by dimeric RING ligase RNF4. Nat Struct Mol Biol. 2011;18:1052–1059. doi: 10.1038/nsmb.2108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Dou H, Buetow L, Sibbet GJ, Cameron K, Huang DT. BIRC7-E2 ubiquitin conjugate structure reveals the mechanism of ubiquitin transfer by a RING dimer. Nat Struct Mol Biol. 2012;19:876–883. doi: 10.1038/nsmb.2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zheng N, Wang P, Jeffrey PD, Pavletich NP. Structure of a c-Cbl-UbcH7 complex: RING domain function in ubiquitin-protein ligases. Cell. 2000;102:533–539. doi: 10.1016/s0092-8674(00)00057-x. [DOI] [PubMed] [Google Scholar]
- 26.Pruneda JN, Littlefield PJ, Soss SE, Nordquist KA, Chazin WJ, Brzovic PS, Klevit RE. Structure of an E3:E2~Ub complex reveals an allosteric mechanism shared among RING/U-box ligases. Mol Cell. 2012;47:933–942. doi: 10.1016/j.molcel.2012.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Branigan E, Plechanovova A, Jaffray EG, Naismith JH, Hay RT. Structural basis for the RING-catalyzed synthesis of K63-linked ubiquitin chains. Nat Struct Mol Biol. 2015 doi: 10.1038/nsmb.3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kamadurai HB, Souphron J, Scott DC, Duda DM, Miller DJ, Stringer D, Piper RC, Schulman BA. Insights into ubiquitin transfer cascades from a structure of a UbcH5B~ubiquitin-HECT(NEDD4L) complex. Mol Cell. 2009;36:1095–1102. doi: 10.1016/j.molcel.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Huang L, Kinnucan E, Wang G, Beaudenon S, Howley PM, Huibregtse JM, Pavletich NP. Structure of an E6AP-UbcH7 complex: insights into ubiquitination by the E2-E3 enzyme cascade. Science. 1999;286:1321–1326. doi: 10.1126/science.286.5443.1321. [DOI] [PubMed] [Google Scholar]
- 30.Luna-Vargas MP, Christodoulou E, Alfieri A, van Dijk WJ, Stadnik M, Hibbert RG, Sahtoe DD, Clerici M, Marco VD, Littler D, Celie PH, Sixma TK, Perrakis A. Enabling high-throughput ligation-independent cloning and protein expression for the family of ubiquitin specific proteases. J Struct Biol. 2011;175:113–119. doi: 10.1016/j.jsb.2011.03.017. [DOI] [PubMed] [Google Scholar]
- 31.Brzovic PS, Lissounov A, Christensen DE, Hoyt DW, Klevit RE. A UbcH5/ubiquitin noncovalent complex is required for processive BRCA1-directed ubiquitination. Mol Cell. 2006;21:873–880. doi: 10.1016/j.molcel.2006.02.008. [DOI] [PubMed] [Google Scholar]
- 32.Hilgart MC, Sanishvili R, Ogata CM, Becker M, Venugopalan N, Stepanov S, Makarov O, Smith JL, Fischetti RF. Automated sample-scanning methods for radiation damage mitigation and diffraction-based centering of macromolecular crystals. Journal of synchrotron radiation. 2011;18:717–722. doi: 10.1107/S0909049511029918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Stepanov S, Makarov O, Hilgart M, Pothineni SB, Urakhchin A, Devarapalli S, Yoder D, Becker M, Ogata C, Sanishvili R, Venugopalan N, Smith JL, Fischetti RF. JBluIce-EPICS control system for macromolecular crystallography. Acta Crystallogr D Biol Crystallogr. 2011;67:176–188. doi: 10.1107/S0907444910053916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kabsch W. Xds. Acta Crystallogr D Biol Crystallogr. 2010;66:125–132. doi: 10.1107/S0907444909047337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Evans PR, Murshudov GN. How good are my data and what is the resolution? Acta Crystallogr D Biol Crystallogr. 2013;69:1204–1214. doi: 10.1107/S0907444913000061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Winn MD, Ballard CC, Cowtan KD, Dodson EJ, Emsley P, Evans PR, Keegan RM, Krissinel EB, Leslie AG, McCoy A, McNicholas SJ, Murshudov GN, Pannu NS, Potterton EA, Powell HR, Read RJ, Vagin A, Wilson KS. Overview of the CCP4 suite and current developments. Acta Crystallogr D Biol Crystallogr. 2011;67:235–242. doi: 10.1107/S0907444910045749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.McCoy AJ, Grosse-Kunstleve RW, Adams PD, Winn MD, Storoni LC, Read RJ. Phaser crystallographic software. J Appl Crystallogr. 2007;40:658–674. doi: 10.1107/S0021889807021206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Adams PD, Afonine PV, Bunkoczi G, Chen VB, Davis IW, Echols N, Headd JJ, Hung LW, Kapral GJ, Grosse-Kunstleve RW, McCoy AJ, Moriarty NW, Oeffner R, Read RJ, Richardson DC, Richardson JS, Terwilliger TC, Zwart PH. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr. 2010;66:213–221. doi: 10.1107/S0907444909052925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sakata E, Satoh T, Yamamoto S, Yamaguchi Y, Yagi-Utsumi M, Kurimoto E, Tanaka K, Wakatsuki S, Kato K. Crystal structure of UbcH5b~ubiquitin intermediate: insight into the formation of the self-assembled E2~Ub conjugates. Structure. 2010;18:138–147. doi: 10.1016/j.str.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 40.Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN, Bourne PE. The Protein Data Bank. Nucleic Acids Res. 2000;28:235–242. doi: 10.1093/nar/28.1.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bunkoczi G, Read RJ. Improvement of molecular-replacement models with Sculptor. Acta Crystallogr D Biol Crystallogr. 2011;67:303–312. doi: 10.1107/S0907444910051218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Murshudov GN, Skubak P, Lebedev AA, Pannu NS, Steiner RA, Nicholls RA, Winn MD, Long F, Vagin AA. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr D Biol Crystallogr. 2011;67:355–367. doi: 10.1107/S0907444911001314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nicholls RA, Long F, Murshudov GN. Low-resolution refinement tools in REFMAC5. Acta Crystallogr D Biol Crystallogr. 2012;68:404–417. doi: 10.1107/S090744491105606X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nicholls RA, Fischer M, McNicholas S, Murshudov GN. Conformation-independent structural comparison of macromolecules with ProSMART. Acta Crystallogr D Biol Crystallogr. 2014;70:2487–2499. doi: 10.1107/S1399004714016241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ozkan E, Yu H, Deisenhofer J. Mechanistic insight into the allosteric activation of a ubiquitin-conjugating enzyme by RING-type ubiquitin ligases. Proc Natl Acad Sci U S A. 2005;102:18890–18895. doi: 10.1073/pnas.0509418102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of Coot. Acta Crystallogr D Biol Crystallogr. 2010;66:486–501. doi: 10.1107/S0907444910007493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Painter J, Merritt EA. Optimal description of a protein structure in terms of multiple groups undergoing TLS motion. Acta Crystallogr D Biol Crystallogr. 2006;62:439–450. doi: 10.1107/S0907444906005270. [DOI] [PubMed] [Google Scholar]
- 48.Joosten RP, Long F, Murshudov GN, Perrakis A. The PDB_REDO server for macromolecular structure model optimization. IUCrJ. 2014;1:213–220. doi: 10.1107/S2052252514009324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen VB, Arendall WB, 3rd, Headd JJ, Keedy DA, Immormino RM, Kapral GJ, Murray LW, Richardson JS, Richardson DC. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr D Biol Crystallogr. 2010;66:12–21. doi: 10.1107/S0907444909042073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dong KC, Helgason E, Yu C, Phu L, Arnott DP, Bosanac I, Compaan DM, Huang OW, Fedorova AV, Kirkpatrick DS, Hymowitz SG, Dueber EC. Preparation of distinct ubiquitin chain reagents of high purity and yield. Structure. 2011;19:1053–1063. doi: 10.1016/j.str.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 51.Elliott PR, Nielsen SV, Marco-Casanova P, Fiil BK, Keusekotten K, Mailand N, Freund SM, Gyrd-Hansen M, Komander D. Molecular basis and regulation of OTULIN-LUBAC interaction. Mol Cell. 2014;54:335–348. doi: 10.1016/j.molcel.2014.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Spratt DE, Martinez-Torres RJ, Noh YJ, Mercier P, Manczyk N, Barber KR, Aguirre JD, Burchell L, Purkiss A, Walden H, Shaw GS. A molecular explanation for the recessive nature of parkin-linked Parkinson’s disease. Nat Commun. 2013;4:1983. doi: 10.1038/ncomms2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Spratt DE, Mercier P, Shaw GS. Structure of the HHARI catalytic domain shows glimpses of a HECT E3 ligase. PLoS ONE. 2013;8:e74047. doi: 10.1371/journal.pone.0074047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Drozdetskiy A, Cole C, Procter J, Barton GJ. JPred4: a protein secondary structure prediction server. Nucleic Acids Res. 2015 doi: 10.1093/nar/gkv332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Grishin AM, Condos TE, Barber KR, Campbell-Valois FX, Parsot C, Shaw GS, Cygler M. Structural basis for the inhibition of host protein ubiquitination by Shigella effector kinase OspG. Structure. 2014;22:878–888. doi: 10.1016/j.str.2014.04.010. [DOI] [PubMed] [Google Scholar]
- 56.Lehninger AL, Nelson DL, Cox MM. Principles of Biochemistry. 2. Worth Publishers; 1993. [Google Scholar]
- 57.Afonine PV, Moriarty NW, Mustyakimov M, Sobolev OV, Terwilliger TC, Turk D, Urzhumtsev A, Adams PD. FEM: feature-enhanced map. Acta Crystallogr D Biol Crystallogr. 2015;71:646–666. doi: 10.1107/S1399004714028132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Emmerich CH, Ordureau A, Strickson S, Arthur JS, Pedrioli PG, Komander D, Cohen P. Activation of the canonical IKK complex by K63/M1-linked hybrid ubiquitin chains. Proc Natl Acad Sci U S A. 2013;110:15247–15252. doi: 10.1073/pnas.1314715110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sato Y, Fujita H, Yoshikawa A, Yamashita M, Yamagata A, Kaiser SE, Iwai K, Fukai S. Specific recognition of linear ubiquitin chains by the Npl4 zinc finger (NZF) domain of the HOIL-1L subunit of the linear ubiquitin chain assembly complex. Proc Natl Acad Sci U S A. 2011;108:20520–20525. doi: 10.1073/pnas.1109088108. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.