Discovery of a Novel DCAF1 Ligand Using a Drug–Target Interaction Prediction Model: Generalizing Machine Learning to New Drug TargetsClick to copy article linkArticle link copied!
- Serah W. KimaniSerah W. KimaniStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaPrincess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 2C4, CanadaMore by Serah W. Kimani
- Julie Owen
- Stuart R. GreenStuart R. GreenStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Stuart R. Green
- Fengling LiFengling LiStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Fengling Li
- Yanjun LiYanjun LiStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Yanjun Li
- Aiping DongAiping DongStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Aiping Dong
- Peter J. BrownPeter J. BrownStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Peter J. Brown
- Suzanne AcklooSuzanne AcklooStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaMore by Suzanne Ackloo
- David Kuter
- Cindy Yang
- Miranda MacAskillMiranda MacAskillRecursion Pharmaceuticals Inc., Toronto, Ontario M5V 2A2, CanadaMore by Miranda MacAskill
- Stephen Scott MacKinnonStephen Scott MacKinnonRecursion Pharmaceuticals Inc., Toronto, Ontario M5V 2A2, CanadaMore by Stephen Scott MacKinnon
- Cheryl H. ArrowsmithCheryl H. ArrowsmithStructural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaPrincess Margaret Cancer Center, University Health Network, Toronto, Ontario M5G 2C4, CanadaDepartment of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A1, CanadaMore by Cheryl H. Arrowsmith
- Matthieu Schapira*Matthieu Schapira*Email: [email protected]. Tel.: +1 416-978-3092.Structural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaDepartment of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A1, CanadaMore by Matthieu Schapira
- Vijay Shahani*Vijay Shahani*Email: [email protected]. Tel.: +1 647-444-6226.Recursion Pharmaceuticals Inc., Toronto, Ontario M5V 2A2, CanadaMore by Vijay Shahani
- Levon Halabelian*Levon Halabelian*Email: [email protected]. Tel.: +1 416-946-3876.Structural Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, CanadaDepartment of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A1, CanadaMore by Levon Halabelian
Abstract
DCAF1 functions as a substrate recruitment subunit for the RING-type CRL4DCAF1 and the HECT family EDVPDCAF1 E3 ubiquitin ligases. The WDR domain of DCAF1 serves as a binding platform for substrate proteins and is also targeted by HIV and SIV lentiviral adaptors to induce the ubiquitination and proteasomal degradation of antiviral host factors. It is therefore attractive both as a potential therapeutic target for the development of chemical inhibitors and as an E3 ligase that could be recruited by novel PROTACs for targeted protein degradation. In this study, we used a proteome-scale drug–target interaction prediction model, MatchMaker, combined with cheminformatics filtering and docking to identify ligands for the DCAF1 WDR domain. Biophysical screening and X-ray crystallographic studies of the predicted binders confirmed a selective ligand occupying the central cavity of the WDR domain. This study shows that artificial intelligence-enabled virtual screening methods can successfully be applied in the absence of previously known ligands.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1
Figure 1. Conceptual diagram highlighting differences between ligand-based drug discovery (LBDD) models and drug–target interaction (DTI) models. LBDD models treat each protein as its own machine learning model, thereby limiting inference (prediction) to targets that already have sufficient data to train models. DTI models train a global model to predict binding drug–target pairs, such that protein targets learn from the bioactivities of similar proteins.
Experimental Methods
Computational Screening
Compound Library and Procurement
Human Proteome and DCAF1 Pockets
Figure 2
Figure 2. Pocket selection. (A) Visualization of P2Rank predicted pockets. (B,C) Cartoon representation of Vpx from the ternary complex of DCAF1-SAMHD1-Vpx (PDB code 4CC9) showing overlay of pocket prediction with protein contacts. (D) Inside pocket. (E) Top pocket. (F) Side pocket.
Modeling Drug–Target Interactions
DTI Evaluations
Candidate Selection
Biophysical Characterization of Computational Hits
Protein Expression and Purification
Surface Plasmon Resonance (SPR) Binding Studies
Protein Crystallography
DCAF1 WDR Domain Gene Cloning, Protein Expression, and Purification
Protein Crystallization
Diffraction Data Collection, Structure Determination, and Refinement
Results
Candidate Selection and Experimental Confirmation of the Hit Compound CYCA-117-70
Figure 3
Figure 3. Binding of CYCA-117-70 to DCAF1 and WDR5 by SPR. (A) Binding of CYCA-117-70 to DCAF1 WDR. (B) Binding to WDR5. SPR binding data (representative plot of N = 2) represented in the steady-state response (black circles) with the steady state 1:1 binding model fitting (red dashed line) and the sensorgram (solid green) with the kinetic fit (black dots). CYCA-117-70 showed binding to DCAF1 with an estimated KD of 70 μM (since the binding curve does not fully reach saturation) and no significant binding to WDR5 (KD not determined).
Crystal Structure of the Human DCAF1 WDR Domain Bound to CYCA-117-70
DCAF1-CYCA-117-70 | |
---|---|
PDB ID | 7SSE |
Wavelength (nm) | 0.9791 |
Resolution range (Å) | 50–1.62 (1.65–1.62) |
Space group | P 1 21 1 |
Unit cell (Å) | 48.956, 87.919, 73.878 |
Total reflections | 269,845 |
Unique reflections | 76,489 |
Multiplicity | 3.5 (2.1) |
Completeness (%) | 97.4 (78.4) |
Mean I/sigma (I) | 30.92 (1.45) |
R-merge | 0.041 (0.516) |
R-meas | 0.048 (0.651) |
R-pim | 0.024 (0.390) |
CC1/2 | 0.998 (0.666) |
CC* | 0.999 (0.894) |
Reflections used in refinement | 72,666 |
Reflections used for R-free | 3750 |
R-work | 0.206 |
R-free | 0.234 |
CC (work) | 0.960 |
CC (free) | 0.947 |
Number of nonhydrogen atoms | |
Macromolecules | 4608 |
Ligands | 26 |
Solvent | 224 |
Protein residues | |
RMS (bonds) | 0.005 |
RMS (angles) | 1.307 |
Ramachandran favored (%) | 96.88 |
Ramachandran allowed (%) | 96.81 |
Ramachandran outliers (%) | 0.0 |
Poor rotamer (%) | 0.8 |
Clash score | 2.32 |
Average B-factor | |
Macromolecules | 29.153 |
Ligands | 35.953 |
Solvent | 33.077 |
Statistics for the highest-resolution shell are shown in parentheses.
Figure 4
Figure 4. Cocrystal structure of the DCAF1 WDR domain in complex with CYCA-117-70. (A) Top and side views of the DCAF1 WDR domain shown as a cartoon representation in slate blue, bound to CYCA-117-70 shown as yellow sticks. The compound binds close to the surface of the WDR ring central channel. (B) Zoomed-in view of the CYCA-117-70 binding site in chain A of the DCAF1-CYCA-117-70 cocrystal structure. CYCA-117-70 is shown as yellow sticks, water molecules are shown as red spheres, and the putative hydrogen bond is shown as black dashes. (C) Overlay of the DCAF1 monomer (slate blue surface) bound to CYCA-117-70 (yellow sticks) on to lentiviral Vpx (green) (PDB ID 4CC9, data from ref (22), revealing a steric overlap between the two ligands.
Discussion
Conclusions
Data Availability
Atomic coordinates and structure factors for the reported crystal structure have been deposited in the Protein Data Bank under the accession code 7SSE. MatchMaker is a commercial software developed by Cyclica, Inc. and now owned by Recursion Pharmaceuticals Inc.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00082.
Figure showing the domain architecture of human DCAF1 and the WDR fold, figure of the electron density of CYCA-117-70 and its binding site, figure depicting the comparison of CYCA-117-70 with other recently reported DCAF1 ligands (PDF)
SMILES string of the 101 compounds computationally selected for experimental testing (XLSX)
SPR raw data for CYCA-117-70 (XLSX)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We would like to thank Peter Loppnau, Almagul Seitova, Ashley Hutchinson, Pegah Ghiabi, and Taraneh Hajian for protein expression and purification. This work is based upon research conducted at the Northeastern Collaborative Access Team beamlines, which are funded by the National Institute of General Medical Sciences from the National Institutes of Health (P30 GM124165). 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. Structural Genomics Consortium is a registered charity (no. 1097737) that receives funds from Bayer AG, Boehringer Ingelheim, Bristol Myers Squibb, Genentech, Genome Canada through Ontario Genomics Institute [OGI-196], EU/EFPIA/OICR/McGill/KTH/Diamond Innovative Medicines Initiative 2 Joint Undertaking [EUbOPEN grant 875510], Janssen, Merck KGaA (aka EMD in Canada and US), Pfizer, and Takeda. M.S. gratefully acknowledges financial support from NSERC [Grant RGPIN-2019-04416].
AI | artificial intelligence |
CLR4 | cullin-4 RING ubiquitin ligase |
DCAF1 | DDB1 and CUL4 associated factor 1 |
DDB1 | DNA damage-binding protein 1 |
DTI | drug–target interaction |
EDVP | EDD, DDB1, and VPRBP E3 ligase complex |
HECT | homologous to the E6-AP carboxyl terminus |
HIV | human immunodeficiency virus |
LBDD | ligand-based drug design |
RING | really interesting new gene |
PROTACS | proteolysis targeting chimeras |
SBDD | structure-based drug design |
SIV | simian immunodeficiency virus |
Vpr/Vpx | accessory proteins r or x |
WDR | WD40 repeat |
References
This article references 37 other publications.
- 1Ehrlich, P. The Mutual Relations between Toxin and Antitoxin. Boston Med. Surg. J. 1904, 150, 443– 445, DOI: 10.1056/NEJM190404281501701Google ScholarThere is no corresponding record for this reference.
- 2Langley, J. N. On the Reaction of Cells and of Nerve-Endings to Certain Poisons, Chiefly as Regards the Reaction of Striated Muscle to Nicotine and to Curari. J. Physiol. 1905, 33, 374– 413, DOI: 10.1113/jphysiol.1905.sp001128Google Scholar2Over the reaction of the cells and the nerve ending to certain poisons, in particular in consideration of the reaction of the transverse striped muscle to nicotine and to curare. [machine translation]Langley, J. N.Journal of Physiology (Oxford, United Kingdom) (1905), 33 (), 374-413CODEN: JPHYA7 ISSN:.[Machine Translation of Descriptors]. Nicotine causes extended contraction in certain muscles of the birds. Also, after cutting through the muscle supplying nerves as after paralysis, one obtains muscle contraction the same by nicotine or curare. The contraction caused by nicotine is decreased by the injection of a sufficient quantity of curare; the two poisons are in their effect on the muscle antagonist. Degeneration of the nerves belonging to the muscles does not change anything in their effect. From it, experiments results that nicotine and curare not on the nerve ending, but on the muscle affects, not directly on the contractile substance, but other materials in the muscle, which calls author "receptive substance" (receipt substance). As author of the closer implements, similar conditions are present not only with the transverse striped muscle fibers, but also with other cell kinds, and probably the larger number of the materials practices their effect not, as accepted, on the nerve ending, select on the "receptive substance".
- 3MacKinnon, S. S.; Madani Tonekaboni, S. A.; Windemuth, A. Proteome-Scale Drug-Target Interaction Predictions: Approaches and Applications. Curr. Protoc. 2021, 1, e302 DOI: 10.1002/cpz1.302Google ScholarThere is no corresponding record for this reference.
- 4Schapira, M.; Tyers, M.; Torrent, M.; Arrowsmith, C. H. WD40 Repeat Domain Proteins: A Novel Target Class?. Nat. Rev. Drug Discov. 2017, 16, 773– 786, DOI: 10.1038/nrd.2017.179Google Scholar4WD40 repeat domain proteins: a novel target class?Schapira, Matthieu; Tyers, Mike; Torrent, Maricel; Arrowsmith, Cheryl H.Nature Reviews Drug Discovery (2017), 16 (11), 773-786CODEN: NRDDAG; ISSN:1474-1776. (Nature Research)Antagonism of protein-protein interactions (PPIs) with small mols. is becoming more feasible as a therapeutic approach. Successful PPI inhibitors tend to target proteins contg. deep peptide-binding grooves or pockets rather than the more common large, flat protein interaction surfaces. Here, we review one of the most abundant PPI domains in the human proteome, the WD40 repeat (WDR) domain, which has a central peptide-binding pocket and is a member of the β-propeller domain-contg. protein family. Recently, two WDR domain-contg. proteins, WDR5 and EED, as well as other β-propeller domains have been successfully targeted by potent, specific, cell-active, drug-like chem. probes. Could WDR domains be a novel target class for drug discovery. Although the research is at an early stage and therefore not clin. validated, cautious optimism is justified, as WDR domain-contg. proteins are involved in multiple disease-assocd. pathways. The druggability and structural diversity of WDR domain binding pockets suggest that understanding how to target this prevalent domain class will open up areas of disease biol. that have so far resisted drug discovery efforts.
- 5Higa, L. A.; Wu, M.; Ye, T.; Kobayashi, R.; Sun, H.; Zhang, H. CUL4-DDB1 Ubiquitin Ligase Interacts with Multiple WD40-Repeat Proteins and Regulates Histone Methylation. Nat. Cell Biol. 2006, 8, 1277– 1283, DOI: 10.1038/ncb1490Google Scholar5CUL4-DDB1 ubiquitin ligase interacts with multiple WD40-repeat proteins and regulates histone methylationHiga, Leigh Ann; Wu, Min; Ye, Tao; Kobayashi, Ryuji; Sun, Hong; Zhang, HuiNature Cell Biology (2006), 8 (11), 1277-1283CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)The CUL4-DDB1-ROC1 ubiquitin E3 ligase regulates cell-cycle progression, replication and DNA damage response. However, the substrate-specific adaptors of this ligase remain uncharacterized. Here, we show that CUL4-DDB1 complexes interact with multiple WD40-repeat proteins (WDRs) including TLE1-3, WDR5, L2DTL (also known as CDT2) and the Polycomb-group protein EED (also known as ESC). WDR5 and EED are core components of histone methylation complexes that are essential for histone H3 methylation and epigenetic control at K4 or K9 and K27, resp., whereas L2DTL regulates CDT1 proteolysis after DNA damage through CUL4-DDB1 (ref. 8). We found that CUL4A-DDB1 interacts with H3 methylated mononucleosomes and peptides. Inactivation of either CUL4 or DDB1 impairs these histone modifications. However, loss of WDR5 specifically affects histone H3 methylation at K4 but not CDT1 degrdn., whereas inactivation of L2DTL prevents CDT1 degrdn. but not histone methylation. Our studies suggest that CUL4-DDB1 ligases use WDR proteins as mol. adaptors for substrate recognition, and modulate multiple biol. processes through ubiquitin-dependent proteolysis.
- 6Sharma, P.; Nag, A. CUL4A Ubiquitin Ligase: A Promising Drug Target for Cancer and Other Human Diseases. Open Biol. 2014, 4, 130217 DOI: 10.1098/rsob.130217Google Scholar6CUL4A ubiquitin ligase: a promising drug target for cancer and other human diseasesSharma, Puneet; Nag, AloOpen Biology (2014), 4 (2), 130217/1-130217/15, 15CODEN: OBPICQ; ISSN:2046-2441. (Royal Society)A review. The ability of cullin 4A (CUL4A), a scaffold protein, to recruit a repertoire of substrate adaptors allows it to assemble into distinct E3 ligase complexes to mediate turnover of key regulatory proteins. In the past decade, a considerable wealth of information has been generated regarding its biol., regulation, assembly, mol. architecture and novel functions. Importantly, unravelling of its assocn. with multiple tumors and modulation by viral proteins establishes it as one of the key proteins that may play an important role in cellular transformation. Considering the role of its substrate in regulating the cell cycle and maintenance of genomic stability, understanding the detailed aspects of these processes will have significant consequences for the treatment of cancer and related diseases. This review is an effort to provide a broad overview of this multifaceted ubiquitin ligase and addresses its crit. role in regulation of important biol. processes. More importantly, its tremendous potential to be exploited for therapeutic purposes has been discussed.
- 7Maddika, S.; Chen, J. Protein Kinase DYRK2 Is a Scaffold That Facilitates Assembly of an E3 Ligase. Nat. Cell Biol. 2009, 11, 409– 419, DOI: 10.1038/ncb1848Google Scholar7Protein kinase DYRK2 is a scaffold that facilitates assembly of an E3 ligaseMaddika, Subbareddy; Chen, JunjieNature Cell Biology (2009), 11 (4), 409-419CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Protein kinases have central functions in various cellular signal transduction pathways through their substrate phosphorylation. Here we show that a protein kinase, DYRK2, has unexpected role as a scaffold for an E3 ubiquitin ligase complex. DYRK2 assocs. with an E3 ligase complex contg. EDD, DDB1, and VPRBP proteins (EDVP complex). Strikingly, DYRK2 serves as a scaffold for the EDVP complex, because small-interfering-RNA-mediated depletion of DYRK2 disrupts the formation of the EDD-DDB1-VPRBP complex. Although the kinase activity of DYRK2 is dispensable for its ability to mediate EDVP complex formation, it is required for the phosphorylation and subsequent degrdn. of its downstream substrate, katanin p60. Collectively, our results reveal a new type of E3-ubiquitin ligase complex in humans that depends on a protein kinase for complex formation as well as for the subsequent phosphorylation, ubiquitylation, and degrdn. of their substrates.
- 8Angers, S.; Li, T.; Yi, X.; MacCoss, M. J.; Moon, R. T.; Zheng, N. Molecular Architecture and Assembly of the DDB1-CUL4A Ubiquitin Ligase Machinery. Nature 2006, 443, 590– 593, DOI: 10.1038/nature05175Google Scholar8Molecular architecture and assembly of the DDB1-CUL4A ubiquitin ligase machineryAngers, Stephane; Li, Ti; Yi, Xianhua; MacCoss, Michael J.; Moon, Randall T.; Zheng, NingNature (London, United Kingdom) (2006), 443 (7111), 590-593CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Protein ubiquitination is a common form of post-translational modification that regulates a broad spectrum of protein substrates in diverse cellular pathways. Through a three-enzyme (E1-E2-E3) cascade, the attachment of ubiquitin to proteins is catalyzed by the E3 ubiquitin ligase, which is best represented by the superfamily of cullin-RING complexes. Conserved from yeast to human, the DDB1-CUL4-ROC1 complex is a recently identified cullin-RING ubiquitin ligase, which regulates DNA repair, DNA replication and transcription, and can also be subverted by pathogenic viruses to benefit viral infection. Lacking a canonical SKP1-like cullin adaptor and a defined substrate recruitment module, how the DDB1-CUL4-ROC1 E3 app. is assembled for ubiquitinating various substrates remains unclear. Here we present crystallog. analyses of the virally hijacked form of the human DDB1-CUL4A-ROC1 machinery, which show that DDB1 uses one β-propeller domain for cullin scaffold binding and a variably attached sep. double-β-propeller fold for substrate presentation. Through tandem-affinity purifn. of human DDB1 and CUL4A complexes followed by mass spectrometry anal., we also identify a novel family of WD40-repeat proteins, which directly bind to the double-propeller fold of DDB1 and serve as the substrate-recruiting module of the E3. Together, our structural and proteomic results reveal the structural mechanisms and mol. logic underlying the assembly and versatility of a new family of cullin-RING E3 complexes.
- 9Fischer, E. S.; Scrima, A.; Böhm, K.; Matsumoto, S.; Lingaraju, G. M.; Faty, M.; Yasuda, T.; Cavadini, S.; Wakasugi, M.; Hanaoka, F.; Iwai, S.; Gut, H.; Sugasawa, K.; Thomä, N. H. The Molecular Basis of CRL4DDB2/CSA Ubiquitin Ligase Architecture, Targeting, and Activation. Cell 2011, 147, 1024– 1039, DOI: 10.1016/j.cell.2011.10.035Google Scholar9The Molecular Basis of CRL4DDB2/CSA Ubiquitin Ligase Architecture, Targeting, and ActivationFischer, Eric S.; Scrima, Andrea; Boehm, Kerstin; Matsumoto, Syota; Lingaraju, Gondichatnahalli M.; Faty, Mahamadou; Yasuda, Takeshi; Cavadini, Simone; Wakasugi, Mitsuo; Hanaoka, Fumio; Iwai, Shigenori; Gut, Heinz; Sugasawa, Kaoru; Thomae, Nicolas H.Cell (Cambridge, MA, United States) (2011), 147 (5), 1024-1039CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The DDB1-CUL4-RBX1 (CRL4) ubiquitin ligase family regulates a diverse set of cellular pathways through dedicated substrate receptors (DCAFs). The DCAF DDB2 detects UV-induced pyrimidine dimers in the genome and facilitates nucleotide excision repair. We provide the mol. basis for DDB2 receptor-mediated cyclobutane pyrimidine dimer recognition in chromatin. The structures of the fully assembled DDB1-DDB2-CUL4A/B-RBX1 (CRL4DDB2) ligases reveal that the mobility of the ligase arm creates a defined ubiquitination zone around the damage, which precludes direct ligase activation by DNA lesions. Instead, the COP9 signalosome (CSN) mediates the CRL4DDB2 inhibition in a CSN5 independent, nonenzymic, fashion. In turn, CSN inhibition is relieved upon DNA damage binding to the DDB2 module within CSN-CRL4DDB2. The Cockayne syndrome A DCAF complex crystal structure shows that CRL4DCAF(WD40) ligases share common architectural features. Our data support a general mechanism of ligase activation, which is induced by CSN displacement from CRL4DCAF on substrate binding to the DCAF. Binding pyrimidine dimers on chromatin displaces an inhibitory factor from the CRL4-DDB2 ubiquitin ligase complex and activates it. Other CLR4 complexes share similar architectural features, suggesting a general mechanism of ligase activation.
- 10Nakagawa, T.; Mondal, K.; Swanson, P. C. VprBP (DCAF1): A Promiscuous Substrate Recognition Subunit That Incorporates into Both RING-Family CRL4 and HECT-Family EDD/UBR5 E3 Ubiquitin Ligases. BMC Mol. Biol. 2013, 14, 22, DOI: 10.1186/1471-2199-14-22Google Scholar10VprBP (DCAF1): a promiscuous substrate recognition subunit that incorporates into both RING-family CRL4 and HECT-family EDD/UBR5 E3 ubiquitin ligasesNakagawa, Tadashi; Mondal, Koushik; Swanson, Patrick C.BMC Molecular Biology (2013), 14 (), 22CODEN: BMBMC4; ISSN:1471-2199. (BioMed Central Ltd.)A review. The terminal step in the ubiquitin modification system relies on an E3 ubiquitin ligase to facilitate transfer of ubiquitin to a protein substrate. The substrate recognition and ubiquitin transfer activities of the E3 ligase may be mediated by a single polypeptide or may rely on sep. subunits. The latter organization is particularly prevalent among members of largest class of E3 ligases, the RING family, although examples of this type of arrangement have also been reported among members of the smaller HECT family of E3 ligases. This review describes recent discoveries that reveal the surprising and distinctive ability of VprBP (DCAF1) to serve as a substrate recognition subunit for a member of both major classes of E3 ligase, the RING-type CRL4 ligase and the HECT-type EDD/UBR5 ligase. The cellular processes normally regulated by VprBP-assocd. E3 ligases, and their targeting and subversion by viral accessory proteins are also discussed. Taken together, these studies provide important insights and raise interesting new questions regarding the mechanisms that regulate or subvert VprBP function in the context of both the CRL4 and EDD/UBR5 E3 ligases.
- 11Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric Molecules That Target Proteins to the Skp1-Cullin-F Box Complex for Ubiquitination and Degradation. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 8554– 8559, DOI: 10.1073/pnas.141230798Google Scholar11Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradationSakamoto, Kathleen M.; Kim, Kyung B.; Kumagai, Akiko; Mercurio, Frank; Crews, Craig M.; Deshaies, Raymond J.Proceedings of the National Academy of Sciences of the United States of America (2001), 98 (15), 8554-8559CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex contg. Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degrdn. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compd., protein-targeting chimeric mol. 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the IκBα phosphopeptide that is recognized by the F-box protein β-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCFβ-TRCP, ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.
- 12Zhang, S.; Feng, Y.; Narayan, O.; Zhao, L. J. Cytoplasmic Retention of HIV-1 Regulatory Protein Vpr by Protein-Protein Interaction with a Novel Human Cytoplasmic Protein VprBP. Gene 2001, 263, 131– 140, DOI: 10.1016/s0378-1119(00)00583-7Google Scholar12Cytoplasmic retention of HIV-1 regulatory protein Vpr by protein-protein interaction with a novel human cytoplasmic protein VprBPZhang, S.; Feng, Y.; Narayan, O.; Zhao, L.-J.Gene (2001), 263 (1-2), 131-140CODEN: GENED6; ISSN:0378-1119. (Elsevier Science B.V.)Vpr is an HIV-1 auxiliary regulatory protein packaged in the virion. It has been shown to enhance the nuclear transport of the HIV-1 pre-integration complex, activate transcription of cellular and viral promoters, and arrest the cell cycle at the G2/M check-point. We previously identified a cellular protein of 180 kDa (RIP) that interacted with HIV-1 Vpr specifically. We now rename this cellular protein as Vpr-binding protein, or VprBP. In this report, we describe the cloning of the VprBP cDNA that encodes 1507 aa residues and is identical to the previously cloned cDNA KIAA0800. We demonstrate that Vpr specifically interacts with recombinantly expressed VprBP in vitro as well as in vivo. Furthermore, Vpr interacts with the cellular endogenous VprBP in the context of the HIV-1 life cycle. Mutational anal. of VprBP suggests that the Vpr binding domain is located within the C-terminal half of VprBP, which has a Pro-rich domain and several Phe-x-x-Phe repeats. Subcellular fractionation studies show that both the endogenous VprBP and the adenovirus-expressed VprBP are distributed predominantly in the cytoplasmic fraction. Consistent with previous reports, the adenovirus-expressed Vpr is distributed in both the cytoplasmic and the nuclear fractions. However, when VprBP and Vpr are expressed together, Vpr is found almost exclusively in the cytoplasm. Expression of VprBP does not affect the nuclear transport of the adenoviral nuclear protein, pTP. VprBP expressed in insect cells also blocks the nuclear transport of a Vpr-GFP fusion protein, and VprBP mutants incapable of interacting with Vpr fail to block Vpr-GFP nuclear transport. We hypothesize that Vpr interaction with VprBP may cause changes in the host cell cytoplasm that affect HIV-1 pathogenesis as well as HIV-1 replication.
- 13Hossain, D.; Ferreira Barbosa, J. A.; Cohen, É. A.; Tsang, W. Y. HIV-1 Vpr Hijacks EDD-DYRK2-DDB1DCAF1 to Disrupt Centrosome Homeostasis. J. Biol. Chem. 2018, 293, 9448– 9460, DOI: 10.1074/jbc.RA117.001444Google Scholar13HIV-1 Vpr hijacks EDD-DYRK2-DDB1DCAF1 to disrupt centrosome homeostasisHossain, Delowar; Ferreira Barbosa, Jeremy A.; Cohen, Eric A.; Tsang, William Y.Journal of Biological Chemistry (2018), 293 (24), 9448-9460CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Viruses exploit the host cell machinery for their own profit. To evade innate immune sensing and promote viral replication, HIV type 1 (HIV-1) subverts DNA repair regulatory proteins and induces G2/M arrest. The preintegration complex of HIV-1 is known to traffic along microtubules and accumulate near the microtubule-organizing center. The centrosome is the major microtubule-organizing center in most eukaryotic cells, but precisely how HIV-1 impinges on centrosome biol. remains poorly understood. We report here that the HIV-1 accessory protein viral protein R (Vpr) localized to the centrosome through binding to DCAF1, forming a complex with the ubiquitin ligase EDD-DYRK2-DDB1DCAF1 and Cep78, a resident centrosomal protein previously shown to inhibit EDD-DYRK2-DDB1DCAF1. Vpr did not affect ubiquitination of Cep78. Rather, it enhanced ubiquitination of an EDD-DYRK2-DDB1DCAF1 substrate, CP110, leading to its degrdn., an effect that could be overcome by Cep78 expression. The down-regulation of CP110 and elongation of centrioles provoked by Vpr were independent of G2/M arrest. Infection of T lymphocytes with HIV-1, but not with HIV-1 lacking Vpr, promoted CP110 degrdn. and centriole elongation. Elongated centrioles recruited more γ-tubulin to the centrosome, resulting in increased microtubule nucleation. Our results suggest that Vpr is targeted to the centrosome where it hijacks a ubiquitin ligase, disrupting organelle homeostasis, which may contribute to HIV-1 pathogenesis.
- 14Schabla, N. M.; Mondal, K.; Swanson, P. C. DCAF1 (VprBP): Emerging Physiological Roles for a Unique Dual-Service E3 Ubiquitin Ligase Substrate Receptor. J. Mol. Cell Biol. 2019, 11, 725– 735, DOI: 10.1093/jmcb/mjy085Google Scholar14DCAF1 (VprBP): emerging physiological roles for a unique dual-service E3 ubiquitin ligase substrate receptorSchabla, N. Max; Mondal, Koushik; Swanson, Patrick C.Journal of Molecular Cell Biology (Oxford, United Kingdom) (2019), 11 (9), 725-735CODEN: JMCBCU; ISSN:1759-4685. (Oxford University Press)A review. Cullin-RING ligases (CRLs) comprise a large group of modular eukaryotic E3 ubiquitin ligases. Within this family, the CRL4 ligase (consisting of the Cullin4 [CUL4] scaffold protein, the Rbx1 RING finger domain protein, the DNA damage-binding protein 1 [DDB1], and one of many DDB1-assocd. substrate receptor proteins) has been intensively studied in recent years due to its involvement in regulating various cellular processes, its role in cancer development and progression, and its subversion by viral accessory proteins. Initially discovered as a target for hijacking by the human immunodeficiency virus accessory protein r, the normal targets and function of the CRL4 substrate receptor protein DDB1-Cul4-assocd. factor 1 (DCAF1; also known as VprBP) had remained elusive, but newer studies have begun to shed light on these questions. Here, we review recent progress in understanding the diverse physiol. roles of this DCAF1 in supporting various general and cell type-specific cellular processes in its context with the CRL4 E3 ligase, as well as another HECT-type E3 ligase with which DCAF1 also assocs., called EDD/UBR5. We also discuss emerging questions and areas of future study to uncover the dynamic roles of DCAF1 in normal physiol.
- 15Hrecka, K.; Gierszewska, M.; Srivastava, S.; Kozaczkiewicz, L.; Swanson, S. K.; Florens, L.; Washburn, M. P.; Skowronski, J. Lentiviral Vpr Usurps Cul4-DDB1[VprBP] E3 Ubiquitin Ligase to Modulate Cell Cycle. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 11778– 11783, DOI: 10.1073/pnas.0702102104Google Scholar15Lentiviral Vpr usurps Cul4-DDB1[VprBP] E3 ubiquitin ligase to modulate cell cycleHrecka, Kasia; Gierszewska, Magdalena; Srivastava, Smita; Kozackiewicz, Lukasz; Swanson, Selene K.; Florens, Laurence; Washburn, Michael P.; Skowronski, JacekProceedings of the National Academy of Sciences of the United States of America (2007), 104 (28), 11778-11783, S11778/1-S11778/6CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The replication of viruses depends on the cell cycle status of the infected cells. Viruses have evolved functions that alleviate restrictions imposed on their replication by the host. Vpr, an accessory factor of primate lentiviruses, arrests cells at the DNA damage checkpoint in G2 phase of the cell cycle, but the mechanism underlying this effect has remained elusive. Here we report that Vpr proteins of both the human (HIV-1) and the distantly related simian (SIVmac) immunodeficiency viruses specifically assoc. with a protein complex comprising subunits of E3 ubiquitin ligase assembled on Cullin-4 scaffold (Cul4-DDB1[VprBP]). We show that Vpr binding to Cul4-DDB1[VprBP] leads to increased neddylation and elevated intrinsic ubiquitin ligase activity of this E3. This effect is mediated through the VprBP subunit of the complex, which recently has been suggested to function as a substrate receptor for Cu14. We also demonstrate that VprBP regulates G, phase and is essential for the completion of DNA replication in S phase. Furthermore, the ability of Vpr to arrest cells in G2 phase correlates with its ability to interact with Cu14-DDB1[VprBP] E3 complex. Our studies identify the Cu14-DDB1[VprBP] E3 ubiquitin ligase complex as the downstream effector of lentiviral Vpr for the induction of cell cycle arrest in G2 phase and suggest that Vpr may use this complex to perturb other aspects of the cell cycle and DNA metab. in infected cells.
- 16Guo, Z.; Kong, Q.; Liu, C.; Zhang, S.; Zou, L.; Yan, F.; Whitmire, J. K.; Xiong, Y.; Chen, X.; Wan, Y. Y. DCAF1 Controls T-Cell Function via P53-Dependent and -Independent Mechanisms. Nat. Commun. 2016, 7, 10307, DOI: 10.1038/ncomms10307Google Scholar16DCAF1 controls T-cell function via p53-dependent and -independent mechanismsGuo, Zengli; Kong, Qing; Liu, Cui; Zhang, Song; Zou, Liyun; Yan, Feng; Whitmire, Jason K.; Xiong, Yue; Chen, Xian; Wan, Yisong Y.Nature Communications (2016), 7 (), 10307CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)On activation, naive T cells grow in size and enter cell cycle to mount immune response. How the fundamental processes of T-cell growth and cell cycle entry are regulated is poorly understood. Here we report that DCAF1 (Ddb1-cullin4-assocd.-factor 1) is essential for these processes. The deletion of DCAF1 in T cells impairs their peripheral homeostasis. DCAF1 is upregulated on T-cell receptor activation and crit. for activation-induced T-cell growth, cell cycle entry and proliferation. In addn., DCAF1 is required for T-cell expansion and function during anti-viral and autoimmune responses in vivo. DCAF1 deletion leads to a drastic stabilization of p53 protein, which can be attributed to a requirement of DCAF1 for MDM2-mediated p53 poly-ubiquitination. Importantly, p53 deletion rescues the cell cycle entry defect but not the growth defect of DCAF1-deficient cells. Therefore, DCAF1 is vital for T-cell function through p53-dependent and -independent mechanisms.
- 17Tao, Y.; Remillard, D.; Vinogradova, E. V.; Yokoyama, M.; Banchenko, S.; Schwefel, D.; Melillo, B.; Schreiber, S. L.; Zhang, X.; Cravatt, B. F. Targeted Protein Degradation by Electrophilic PROTACs That Stereoselectively and Site-Specifically Engage DCAF1. J. Am. Chem. Soc. 2022, 144, 18688– 18699, DOI: 10.1021/jacs.2c08964Google Scholar17Targeted Protein Degradation by Electrophilic PROTACs that Stereoselectively and Site-Specifically Engage DCAF1Tao, Yongfeng; Remillard, David; Vinogradova, Ekaterina V.; Yokoyama, Minoru; Banchenko, Sofia; Schwefel, David; Melillo, Bruno; Schreiber, Stuart L.; Zhang, Xiaoyu; Cravatt, Benjamin F.Journal of the American Chemical Society (2022), 144 (40), 18688-18699CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Targeted protein degrdn. induced by heterobifunctional compds. and mol. glues presents an exciting avenue for chem. probe and drug discovery. To date, small-mol. ligands have been discovered for only a limited no. of E3 ligases, which is an important limiting factor for realizing the full potential of targeted protein degrdn. We report herein the discovery by chem. proteomics of azetidine acrylamides that stereoselectively and site-specifically react with a cysteine (C1113) in the E3 ligase substrate receptor DCAF1. We demonstrate that the azetidine acrylamide ligands for DCAF1 can be developed into electrophilic proteolysis-targeting chimeras (PROTACs) that mediated targeted protein degrdn. in human cells. We show that this process is stereoselective and does not occur in cells expressing a C1113A mutant of DCAF1. Mechanistic studies indicate that only low fractional engagement of DCAF1 is required to support protein degrdn. by electrophilic PROTACs. These findings, taken together, demonstrate how the chem. proteomic anal. of stereochem. defined electrophilic compd. sets can uncover ligandable sites on E3 ligases that support targeted protein degrdn.
- 18Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F. T.; de Beer, T. A. P.; Rempfer, C.; Bordoli, L.; Lepore, R.; Schwede, T. SWISS-MODEL: Homology Modelling of Protein Structures and Complexes. Nucleic Acids Res. 2018, 46, W296– W303, DOI: 10.1093/nar/gky427Google Scholar18SWISS-MODEL: homology modelling of protein structures and complexesWaterhouse, Andrew; Bertoni, Martino; Bienert, Stefan; Studer, Gabriel; Tauriello, Gerardo; Gumienny, Rafal; Heer, Florian T.; de Beer, Tjaart A. P.; Rempfer, Christine; Bordoli, Lorenza; Lepore, Rosalba; Schwede, TorstenNucleic Acids Research (2018), 46 (W1), W296-W303CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Homol. modeling has matured into an important technique in structural biol., significantly contributing to narrowing the gap between known protein sequences and exptl. detd. structures. Fully automated workflows and servers simplify and streamline the homol. modeling process, also allowing users without a specific computational expertise to generate reliable protein models and have easy access to modeling results, their visualization and interpretation. Here, we present an update to the SWISS-MODEL server, which pioneered the field of automated modeling 25 years ago and been continuously further developed. Recently, its functionality has been extended to the modeling of homo- and heteromeric complexes. Starting from the amino acid sequences of the interacting proteins, both the stoichiometry and the overall structure of the complex are inferred by homol. modeling. Other major improvements include the implementation of a new modeling engine, ProMod3 and the introduction a new local model quality estn. method, QMEANDisCo.
- 19Sugiyama, M. G.; Cui, H.; Redka, D. S.; Karimzadeh, M.; Rujas, E.; Maan, H.; Hayat, S.; Cheung, K.; Misra, R.; McPhee, J. B.; Viirre, R. D.; Haller, A.; Botelho, R. J.; Karshafian, R.; Sabatinos, S. A.; Fairn, G. D.; Madani Tonekaboni, S. A.; Windemuth, A.; Julien, J.-P.; Shahani, V.; MacKinnon, S. S.; Wang, B.; Antonescu, C. N. Multiscale Interactome Analysis Coupled with Off-Target Drug Predictions Reveals Drug Repurposing Candidates for Human Coronavirus Disease. Sci. Rep. 2021, 11, 23315, DOI: 10.1038/s41598-021-02432-7Google Scholar19Multiscale interactome analysis coupled with off-target drug predictions reveals drug repurposing candidates for human coronavirus diseaseSugiyama, Michael G.; Cui, Haotian; Redka, Dar'ya S.; Karimzadeh, Mehran; Rujas, Edurne; Maan, Hassaan; Hayat, Sikander; Cheung, Kyle; Misra, Rahul; McPhee, Joseph B.; Viirre, Russell D.; Haller, Andrew; Botelho, Roberto J.; Karshafian, Raffi; Sabatinos, Sarah A.; Fairn, Gregory D.; Madani Tonekaboni, Seyed Ali; Windemuth, Andreas; Julien, Jean-Philippe; Shahani, Vijay; MacKinnon, Stephen S.; Wang, Bo; Antonescu, Costin N.Scientific Reports (2021), 11 (1), 23315CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)Abstr.: The COVID-19 pandemic has highlighted the urgent need for the identification of new antiviral drug therapies for a variety of diseases. COVID-19 is caused by infection with the human coronavirus SARS-CoV-2, while other related human coronaviruses cause diseases ranging from severe respiratory infections to the common cold. We developed a computational approach to identify new antiviral drug targets and repurpose clin.-relevant drug compds. for the treatment of a range of human coronavirus diseases. Our approach is based on graph convolutional networks (GCN) and involves multiscale host-virus interactome anal. coupled to off-target drug predictions. Cell-based exptl. assessment reveals several clin.-relevant drug repurposing candidates predicted by the in silico analyses to have antiviral activity against human coronavirus infection. In particular, we identify the MET inhibitor capmatinib as having potent and broad antiviral activity against several coronaviruses in a MET-independent manner, as well as novel roles for host cell proteins such as IRAK1/4 in supporting human coronavirus infection, which can inform further drug discovery studies.
- 20He, Y.; Yang, C.; Wang, Y.; Sacher, J. R.; Sims, M. M.; Pfeffer, L. M.; Miller, D. D. Novel Structural-Related Analogs of PFI-3 (SRAPs) That Target the BRG1 Catalytic Subunit of the SWI/SNF Complex Increase the Activity of Temozolomide in Glioblastoma Cells. Bioorg. Med. Chem. 2022, 53, 116533 DOI: 10.1016/j.bmc.2021.116533Google Scholar20Novel structural-related analogs of PFI-3 (SRAPs) that target the BRG1 catalytic subunit of the SWI/SNF complex increase the activity of temozolomide in glioblastoma cellsHe, Yali; Yang, Chuanhe; Wang, Yinan; Sacher, Joshua R.; Sims, Michelle M.; Pfeffer, Lawrence M.; Miller, Duane D.Bioorganic & Medicinal Chemistry (2022), 53 (), 116533CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Glioblastoma (GBM) is the most aggressive and treatment-refractory malignant adult brain cancer. After std. of care therapy, the overall median survival for GBM is only ∼6 mo with a 5-yr survival <10%. Although some patients initially respond to the DNA alkylating agent temozolomide (TMZ), unfortunately most patients become resistant to therapy and brain tumors eventually recur. We previously found that knockout of BRG1 or treatment with PFI-3, a small mol. inhibitor of the BRG1 bromodomain, enhances sensitivity of GBM cells to temozolomide in vitro and in vivo GBM animal models. Those results demonstrated that the BRG1 catalytic subunit of the SWI/SNF chromatin remodeling complex appears to play a crit. role in regulating TMZ-sensitivity. In the present study we designed and synthesized Structurally Related Analogs of PFI-3 (SRAPs) and tested their bioactivity in vitro. Among of the SRAPs, 9f and 11d show better efficacy than PFI-3 in sensitizing GBM cells to the antiproliferative and cell death inducing effects of temozolomide in vitro, as well as enhancing the inhibitor effect of temozolomide on the growth of s.c. GBM tumors.
- 21Krivák, R.; Hoksza, D. P2Rank: Machine Learning Based Tool for Rapid and Accurate Prediction of Ligand Binding Sites from Protein Structure. Aust. J. Chem. 2018, 10, 39, DOI: 10.1186/s13321-018-0285-8Google ScholarThere is no corresponding record for this reference.
- 22Schwefel, D.; Groom, H. C. T.; Boucherit, V. C.; Christodoulou, E.; Walker, P. A.; Stoye, J. P.; Bishop, K. N.; Taylor, I. A. Structural Basis of Lentiviral Subversion of a Cellular Protein Degradation Pathway. Nature 2014, 505, 234– 238, DOI: 10.1038/nature12815Google Scholar22Structural basis of lentiviral subversion of a cellular protein degradation pathwaySchwefel, David; Groom, Harriet C. T.; Boucherit, Virginie C.; Christodoulou, Evangelos; Walker, Philip A.; Stoye, Jonathan P.; Bishop, Kate N.; Taylor, Ian A.Nature (London, United Kingdom) (2014), 505 (7482), 234-238CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Lentiviruses contain accessory genes that have evolved to counteract the effects of host cellular defense proteins that inhibit productive infection. One such restriction factor, SAMHD1, inhibits human immunodeficiency virus (HIV)-1 infection of myeloid-lineage cells as well as resting CD4+ T cells by reducing the cellular deoxynucleoside 5'-triphosphate (dNTP) concn. to a level at which the viral reverse transcriptase cannot function. In other lentiviruses, including HIV-2 and related simian immunodeficiency viruses (SIVs), SAMHD1 restriction is overcome by the action of viral accessory protein x (Vpx) or the related viral protein r (Vpr) that target and recruit SAMHD1 for proteasomal degrdn. The mol. mechanism by which these viral proteins are able to usurp the host cell's ubiquitination machinery to destroy the cell's protection against these viruses has not been defined. Here we present the crystal structure of a ternary complex of Vpx with the human E3 ligase substrate adaptor DCAF1 and the carboxy-terminal region of human SAMHD1. Vpx is made up of a three-helical bundle stabilized by a zinc finger motif, and wraps tightly around the disk-shaped DCAF1 mol. to present a new mol. surface. This adapted surface is then able to recruit SAMHD1 via its C terminus, making it a competent substrate for the E3 ligase to mark for proteasomal degrdn. The structure reported here provides a mol. description of how a lentiviral accessory protein is able to subvert the cell's normal protein degrdn. pathway to inactivate the cellular viral defense system.
- 23The UniProt Consortium UniProt: The Universal Protein Knowledgebase in 2023. Nucleic Acids Res. 2023, 51, D523– D531, DOI: 10.1093/nar/gkac1052Google ScholarThere is no corresponding record for this reference.
- 24Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235– 242, DOI: 10.1093/nar/28.1.235Google Scholar24The Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 25Hutchinson, A.; Seitova, A. Production of Recombinant PRMT Proteins Using the Baculovirus Expression Vector System. J. Vis. Exp. JoVE 2021, No. 173. DOI: 10.3791/62510 .Google ScholarThere is no corresponding record for this reference.
- 26Minor, W.; Cymborowski, M.; Otwinowski, Z.; Chruszcz, M. HKL-3000: The Integration of Data Reduction and Structure Solution--from Diffraction Images to an Initial Model in Minutes. Acta Crystallogr. D Biol. Crystallogr. 2006, 62, 859– 866, DOI: 10.1107/S0907444906019949Google Scholar26HKL-3000: the integration of data reduction and structure solution - from diffraction images to an initial model in minutesMinor, Wladek; Cymborowski, Marcin; Otwinowski, Zbyszek; Chruszcz, MaksymilianActa Crystallographica, Section D: Biological Crystallography (2006), D62 (8), 859-866CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)A new approach that integrates data collection, data redn., phasing and model building significantly accelerates the process of structure detn. and on av. minimizes the no. of data sets and synchrotron time required for structure soln. Initial testing of the HKL-3000 system (the beta version was named HKL-2000_ph) with more than 140 novel structure detns. has proven its high value for MAD/SAD expts. The heuristics for choosing the best computational strategy at different data resoln. limits of phasing signal and crystal diffraction are being optimized. The typical end result is an interpretable electron-d. map with a partially built structure and, in some cases, an almost complete refined model. The current development is oriented towards very fast structure soln. in order to provide feedback during the diffraction expt. Work is also proceeding towards improving the quality of phasing calcn. and model building.
- 27McCoy, A. J.; Grosse-Kunstleve, R. W.; Adams, P. D.; Winn, M. D.; Storoni, L. C.; Read, R. J. Phaser Crystallographic Software. J. Appl. Crystallogr. 2007, 40, 658– 674, DOI: 10.1107/S0021889807021206Google Scholar27Phaser crystallographic softwareMcCoy, Airlie J.; Grosse-Kunstleve, Ralf W.; Adams, Paul D.; Winn, Martyn D.; Storoni, Laurent C.; Read, Randy J.Journal of Applied Crystallography (2007), 40 (4), 658-674CODEN: JACGAR; ISSN:0021-8898. (International Union of Crystallography)Phaser is a program for phasing macromol. crystal structures by both mol. replacement and exptl. phasing methods. The novel phasing algorithms implemented in Phaser have been developed using max. likelihood and multivariate statistics. For mol. replacement, the new algorithms have proved to be significantly better than traditional methods in discriminating correct solns. from noise, and for single-wavelength anomalous dispersion exptl. phasing, the new algorithms, which account for correlations between F+ and F-, give better phases (lower mean phase error with respect to the phases given by the refined structure) than those that use mean F and anomalous differences ΔF. One of the design concepts of Phaser was that it be capable of a high degree of automation. To this end, Phaser (written in C++) can be called directly from Python, although it can also be called using traditional CCP4 keyword-style input. Phaser is a platform for future development of improved phasing methods and their release, including source code, to the crystallog. community.
- 28Emsley, P.; Cowtan, K. Coot: Model-Building Tools for Molecular Graphics. Acta Crystallogr. D Biol. Crystallogr. 2004, 60, 2126– 2132, DOI: 10.1107/S0907444904019158Google Scholar28Coot: model-building tools for molecular graphicsEmsley, Paul; Cowtan, KevinActa Crystallographica, Section D: Biological Crystallography (2004), D60 (12, Pt. 1), 2126-2132CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)CCP4mg is a project that aims to provide a general-purpose tool for structural biologists, providing tools for x-ray structure soln., structure comparison and anal., and publication-quality graphics. The map-fitting tools are available as a stand-alone package, distributed as 'Coot'.
- 29Murshudov, G. N.; Vagin, A. A.; Dodson, E. J. Refinement of Macromolecular Structures by the Maximum-Likelihood Method. Acta Crystallogr. D Biol. Crystallogr. 1997, 53, 240– 255, DOI: 10.1107/S0907444996012255Google Scholar29Refinement of macromolecular structures by the maximum-likelihood methodMurshudov, Garib N.; Vagin, Alexei A.; Dodson, Eleanor J.Acta Crystallographica, Section D: Biological Crystallography (1997), D53 (3), 240-255CODEN: ABCRE6; ISSN:0907-4449. (Munksgaard)A review with many refs. on the math. basis of max. likelihood. The likelihood function for macromol. structures is extended to include prior phase information and exptl. std. uncertainties. The assumption that different parts of a structure might have different errors is considered. A method for estg. σA using "free" reflections is described and its effects analyzed. The derived equations have been implemented in the program REFMAC. This has been tested on several proteins at different stages of refinement (bacterial α-amylase, cytochrome c', cross-linked insulin and oligopeptide binding protein). The results derived using the max.-likelihood residual are consistently better than those obtained from least-squares refinement.
- 30Winn, M. D.; Ballard, C. C.; Cowtan, K. D.; Dodson, E. J.; Emsley, P.; Evans, P. R.; Keegan, R. M.; Krissinel, E. B.; Leslie, A. G. W.; McCoy, A.; McNicholas, S. J.; Murshudov, G. N.; Pannu, N. S.; Potterton, E. A.; Powell, H. R.; Read, R. J.; Vagin, A.; Wilson, K. S. Overview of the CCP4 Suite and Current Developments. Acta Crystallogr. D Biol. Crystallogr. 2011, 67, 235– 242, DOI: 10.1107/S0907444910045749Google Scholar30Overview of the CCP4 suite and current developmentsWinn, Martyn D.; Ballard, Charles C.; Cowtan, Kevin D.; Dodson, Eleanor J.; Emsley, Paul; Evans, Phil R.; Keegan, Ronan M.; Krissinel, Eugene B.; Leslie, Andrew G. W.; McCoy, Airlie; McNicholas, Stuart J.; Murshudov, Garib N.; Pannu, Navraj S.; Potterton, Elizabeth A.; Powell, Harold R.; Read, Randy J.; Vagin, Alexei; Wilson, Keith S.Acta Crystallographica, Section D: Biological Crystallography (2011), 67 (4), 235-242CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)A review. The CCP4 (Collaborative Computational Project, No. 4) software suite is a collection of programs and assocd. data and software libraries which can be used for macromol. structure detn. by X-ray crystallog. The suite is designed to be flexible, allowing users a no. of methods of achieving their aims. The programs are from a wide variety of sources but are connected by a common infrastructure provided by std. file formats, data objects and graphical interfaces. Structure soln. by macromol. crystallog. is becoming increasingly automated and the CCP4 suite includes several automation pipelines. After giving a brief description of the evolution of CCP4 over the last 30 years, an overview of the current suite is given. While detailed descriptions are given in the accompanying articles, here it is shown how the individual programs contribute to a complete software package.
- 31Chen, V. B.; Arendall, W. B.; Headd, J. J.; Keedy, D. A.; Immormino, R. M.; Kapral, G. J.; Murray, L. W.; Richardson, J. S.; Richardson, D. C. MolProbity: All-Atom Structure Validation for Macromolecular Crystallography. Acta Crystallogr. D Biol. Crystallogr. 2010, 66, 12– 21, DOI: 10.1107/S0907444909042073Google Scholar31MolProbity: all-atom structure validation for macromolecular crystallographyChen, Vincent B.; Arendall, W. Bryan, III; Headd, Jeffrey J.; Keedy, Daniel A.; Immormino, Robert M.; Kapral, Gary J.; Murray, Laura W.; Richardson, Jane S.; Richardson, David C.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 12-21CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MolProbity is a structure-validation web service that provides broad-spectrum solidly based evaluation of model quality at both the global and local levels for both proteins and nucleic acids. It relies heavily on the power and sensitivity provided by optimized hydrogen placement and all-atom contact anal., complemented by updated versions of covalent-geometry and torsion-angle criteria. Some of the local corrections can be performed automatically in MolProbity and all of the diagnostics are presented in chart and graphical forms that help guide manual rebuilding. X-ray crystallog. provides a wealth of biol. important mol. data in the form of at. three-dimensional structures of proteins, nucleic acids and increasingly large complexes in multiple forms and states. Advances in automation, in everything from crystn. to data collection to phasing to model building to refinement, have made solving a structure using crystallog. easier than ever. However, despite these improvements, local errors that can affect biol. interpretation are widespread at low resoln. and even high-resoln. structures nearly all contain at least a few local errors such as Ramachandran outliers, flipped branched protein side chains and incorrect sugar puckers. It is crit. both for the crystallographer and for the end user that there are easy and reliable methods to diagnose and correct these sorts of errors in structures. MolProbity is the authors' contribution to helping solve this problem and this article reviews its general capabilities, reports on recent enhancements and usage, and presents evidence that the resulting improvements are now beneficially affecting the global database.
- 32Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF Chimera--a Visualization System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25, 1605– 1612, DOI: 10.1002/jcc.20084Google Scholar32UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 33DeLano, W.; Schrödinger, L. PyMOL. Retrieved Httpwwwpymolorgpymol.Google ScholarThere is no corresponding record for this reference.
- 34Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand Efficiency: A Useful Metric for Lead Selection. Drug Discovery Today 2004, 9, 430– 431, DOI: 10.1016/S1359-6446(04)03069-7Google Scholar34Ligand efficiency: a useful metric for lead selectionHopkins Andrew L; Groom Colin R; Alex AlexanderDrug discovery today (2004), 9 (10), 430-1 ISSN:1359-6446.There is no expanded citation for this reference.
- 35Li, A. S. M.; Kimani, S.; Wilson, B.; Noureldin, M.; González-Álvarez, H.; Mamai, A.; Hoffer, L.; Guilinger, J. P.; Zhang, Y.; von Rechenberg, M.; Disch, J. S.; Mulhern, C. J.; Slakman, B. L.; Cuozzo, J. W.; Dong, A.; Poda, G.; Mohammed, M.; Saraon, P.; Mittal, M.; Modh, P.; Rathod, V.; Patel, B.; Ackloo, S.; Santhakumar, V.; Szewczyk, M. M.; Barsyte-Lovejoy, D.; Arrowsmith, C. H.; Marcellus, R.; Guié, M.-A.; Keefe, A. D.; Brown, P. J.; Halabelian, L.; Al-Awar, R.; Vedadi, M. Discovery of Nanomolar DCAF1 Small Molecule Ligands. J. Med. Chem. 2023, 66, 5041– 5060, DOI: 10.1021/acs.jmedchem.2c02132Google Scholar35Discovery of Nanomolar DCAF1 Small Molecule LigandsLi, Alice Shi Ming; Kimani, Serah; Wilson, Brian; Noureldin, Mahmoud; Gonzalez-Alvarez, Hector; Mamai, Ahmed; Hoffer, Laurent; Guilinger, John P.; Zhang, Ying; von Rechenberg, Moritz; Disch, Jeremy S.; Mulhern, Christopher J.; Slakman, Belinda L.; Cuozzo, John W.; Dong, Aiping; Poda, Gennady; Mohammed, Mohammed; Saraon, Punit; Mittal, Manish; Modh, Pratik; Rathod, Vaibhavi; Patel, Bhashant; Ackloo, Suzanne; Santhakumar, Vijayaratnam; Szewczyk, Magdalena M.; Barsyte-Lovejoy, Dalia; Arrowsmith, Cheryl H.; Marcellus, Richard; Guie, Marie-Aude; Keefe, Anthony D.; Brown, Peter J.; Halabelian, Levon; Al-awar, Rima; Vedadi, MasoudJournal of Medicinal Chemistry (2023), 66 (7), 5041-5060CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)DCAF1 is a substrate receptor of two distinct E3 ligases (CRL4DCAF1 and EDVP), plays a crit. physiol. role in protein degrdn., and is considered a drug target for various cancers. Antagonists of DCAF1 could be used toward the development of therapeutics for cancers and viral treatments. We used the WDR domain of DCAF1 to screen a 114-billion-compd. DNA encoded library (DEL) and identified candidate compds. using similarity search and machine learning. This led to the discovery of a compd. (Z1391232269) with an SPR KD of 11μM. Structure-guided hit optimization led to the discovery of OICR-8268 (26e) with an SPR KD of 38 nM and cellular target engagement with EC50 of 10μM as measured by cellular thermal shift assay (CETSA). OICR-8268 is an excellent tool compd. to enable the development of next-generation DCAF1 ligands toward cancer therapeutics, further investigation of DCAF1 functions in cells, and the development of DCAF1-based PROTACs.
- 36Schröder, M.; Renatus, M.; Liang, X.; Meili, F.; Zoller, T.; Ferrand, S.; Gauter, F.; Li, X.; Sigoillot, F.; Gleim, S.; Stachyra, M.-T.; Thomas, J.; Begue, D.; Lefeuvre, P.; Andraos-Rey, R.; Chung, B. Y.; Ma, R.; Carbonneau, S.; Pinch, B.; Hofmann, A.; Schirle, M.; Schmiedberg, N.; Imbach, P.; Gorses, D.; Calkins, K.; Bauer-Probst, B.; Maschlej, M.; Niederst, M.; Maher, R.; Henault, M.; Alford, J.; Ahrne, E.; Hollingworth, G.; Thomä, N. H.; Vulpetti, A.; Radimerski, T.; Holzer, P.; Thoma, C. R. Reinstating Targeted Protein Degradation with DCAF1 PROTACs in CRBN PROTAC Resistant Settings. bioRxiv 2023, DOI: 10.1101/2023.04.09.536153Google ScholarThere is no corresponding record for this reference.
- 37Gironda-Martínez, A.; Donckele, E. J.; Samain, F.; Neri, D. DNA-Encoded Chemical Libraries: A Comprehensive Review with Succesful Stories and Future Challenges. ACS Pharmacol. Transl. Sci. 2021, 4, 1265– 1279, DOI: 10.1021/acsptsci.1c00118Google Scholar37DNA-encoded chemical libraries: A comprehensive review with successful stories and future challengesGironda-Martinez, Adrian; Donckele, Etienne J.; Samain, Florent; Neri, DarioACS Pharmacology & Translational Science (2021), 4 (4), 1265-1279CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)A review. DNA-encoded chem. libraries (DELs) represent a versatile and powerful technol. platform for the discovery of small-mol. ligands to protein targets of biol. and pharmaceutical interest. DELs are collections of mols., individually coupled to distinctive DNA tags serving as amplifiable identification barcodes. Thanks to advances in DNA-compatible reactions, selection methodologies, next-generation sequencing, and data anal., DEL technol. allows the construction and screening of libraries of unprecedented size, which has led to the discovery of highly potent ligands, some of which have progressed to clin. trials. In this Review, we present an overview of diverse approaches for the generation and screening of DEL mol. repertoires. Recent success stories are described, detailing how novel ligands were isolated from DEL screening campaigns and were further optimized by medicinal chem. The goal of the Review is to capture some of the most recent developments in the field, while also elaborating on future challenges to further improve DEL technol. as a therapeutic discovery platform.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 15 publications.
- Suzanne Ackloo, Fengling Li, Magda Szewczyk, Almagul Seitova, Peter Loppnau, Hong Zeng, Jin Xu, Shabbir Ahmad, Yelena A Arnautova, A. J. Baghaie, Serap Beldar, Albina Bolotokova, Paolo A. Centrella, Irene Chau, Matthew A. Clark, John W. Cuozzo, Saba Dehghani-Tafti, Jeremy S. Disch, Aiping Dong, Antoine Dumas, Jianwen A. Feng, Pegah Ghiabi, Elisa Gibson, Justin Gilmer, Brian Goldman, Stuart R Green, Marie-Aude Guié, John P. Guilinger, Nathan Harms, Oleksandra Herasymenko, Scott Houliston, Ashley Hutchinson, Steven Kearnes, Anthony D. Keefe, Serah W. Kimani, Trevor Kramer, Maria Kutera, Haejin A. Kwak, Cristina Lento, Yanjun Li, Jenny Liu, Joachim Loup, Raquel A. C. Machado, Christopher J. Mulhern, Sumera Perveen, Germanna L. Righetto, Patrick Riley, Suman Shrestha, Eric A. Sigel, Madhushika Silva, Michael D. Sintchak, Belinda L. Slakman, Rhys D. Taylor, James Thompson, Wen Torng, Carl Underkoffler, Moritz von Rechenberg, Ryan T. Walsh, Ian Watson, Derek J. Wilson, Esther Wolf, Manisha Yadav, Aliakbar K. Yazdi, Junyi Zhang, Ying Zhang, Vijayaratnam Santhakumar, Aled M. Edwards, Dalia Barsyte-Lovejoy, Matthieu Schapira, Peter J. Brown, Levon Halabelian, Cheryl H. Arrowsmith. A Target Class Ligandability Evaluation of WD40 Repeat-Containing Proteins. Journal of Medicinal Chemistry 2025, 68
(2)
, 1092-1112. https://doi.org/10.1021/acs.jmedchem.4c02010
- Shabbir Ahmad, Jin Xu, Jianwen A. Feng, Ashley Hutchinson, Hong Zeng, Pegah Ghiabi, Aiping Dong, Paolo A. Centrella, Matthew A. Clark, Marie-Aude Guié, John P. Guilinger, Anthony D. Keefe, Ying Zhang, Thomas Cerruti, John W. Cuozzo, Moritz von Rechenberg, Albina Bolotokova, Yanjun Li, Peter Loppnau, Alma Seitova, Yen-Yen Li, Vijayaratnam Santhakumar, Peter J. Brown, Suzanne Ackloo, Levon Halabelian. Discovery of a First-in-Class Small-Molecule Ligand for WDR91 Using DNA-Encoded Chemical Library Selection Followed by Machine Learning. Journal of Medicinal Chemistry 2023, 66
(23)
, 16051-16061. https://doi.org/10.1021/acs.jmedchem.3c01471
- Qi Miao, Vilas D. Kadam, Ayan Mukherjee, Zhi Tan, Mingxing Teng. Unlocking DCAFs To Catalyze Degrader Development: An Arena for Innovative Approaches. Journal of Medicinal Chemistry 2023, 66
(19)
, 13369-13383. https://doi.org/10.1021/acs.jmedchem.3c01209
- Hamad H. Alanazi. Role of artificial intelligence in advancing immunology. Immunologic Research 2025, 73
(1)
https://doi.org/10.1007/s12026-025-09632-7
- Wenxing Lv, Xiaojuan Jia, Bowen Tang, Chao Ma, Xiaopeng Fang, Xurui Jin, Zhangming Niu, Xin Han. In silico modeling of targeted protein degradation. European Journal of Medicinal Chemistry 2025, 289 , 117432. https://doi.org/10.1016/j.ejmech.2025.117432
- Bing Liao, Jing Chen, Qing Ye, Gang Cheng, Lu‐Ping Qin. Identification of Potential DCAF1 Inhibitors Through Virtual Screening Based on Molecular Docking and Molecular Dynamics Simulations. ChemistrySelect 2025, 10
(15)
https://doi.org/10.1002/slct.202501078
- Matthew T.J. Halma, Sowmiyaa Kumar, Jan van Eck, Sanne Abeln, Alexander Gates, Gijs J.L. Wuite. FAIR data for optical tweezers experiments. Biophysical Journal 2025, 124
(8)
, 1255-1272. https://doi.org/10.1016/j.bpj.2025.03.005
- Shuoyan Tan, Zhenglu Chen, Ruiqiang Lu, Huanxiang Liu, Xiaojun Yao. Rational Proteolysis Targeting Chimera Design Driven by Molecular Modeling and Machine Learning. WIREs Computational Molecular Science 2025, 15
(2)
https://doi.org/10.1002/wcms.70013
- Junhyeong Yim, Solbi Kim, Hyung Ho Lee, Jin Soo Chung, Jongmin Park. Fragment-based approaches to discover ligands for tumor-specific E3 ligases. Expert Opinion on Drug Discovery 2024, 19
(12)
, 1471-1484. https://doi.org/10.1080/17460441.2024.2415310
- Spoorthi R. Kulkarni, Vidya Niranjan, Shradha Anand Mulimani, Deeksha Umesh Rao. Designing Pharmaceuticals Through Ligand-Centric Approaches. 2024, 217-246. https://doi.org/10.4018/979-8-3693-5598-5.ch008
- Baiyun Wang, Shiyun Cao, Ning Zheng. Emerging strategies for prospective discovery of molecular glue degraders. Current Opinion in Structural Biology 2024, 86 , 102811. https://doi.org/10.1016/j.sbi.2024.102811
- Miaomiao Li, Wenjing Huang, Yuan Zhang, Yue Du, Shan Zhao, Longhao Wang, Yaxin Sun, Beibei Sha, Jie Yan, Yangcheng Ma, Jinlu Tang, Jianxiang Shi, Pei Li, Lijun Jia, Tao Hu, Ping Chen. Glucose deprivation triggers DCAF1-mediated inactivation of Rheb-mTORC1 and promotes cancer cell survival. Cell Death & Disease 2024, 15
(6)
https://doi.org/10.1038/s41419-024-06808-1
- Rahman Shah Zaib Saleem, Martin P. Schwalm, Stefan Knapp. Expanding the ligand spaces for E3 ligases for the design of protein degraders. Bioorganic & Medicinal Chemistry 2024, 105 , 117718. https://doi.org/10.1016/j.bmc.2024.117718
- Yanbei Li, Zhehuan Fan, Jingxin Rao, Zhiyi Chen, Qinyu Chu, Mingyue Zheng, Xutong Li. An overview of recent advances and challenges in predicting compound-protein interaction (CPI). Medical Review 2023, 3
(6)
, 465-486. https://doi.org/10.1515/mr-2023-0030
- Carrie Arnold. AlphaFold touted as next big thing for drug discovery — but is it?. Nature 2023, 622
(7981)
, 15-17. https://doi.org/10.1038/d41586-023-02984-w
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Conceptual diagram highlighting differences between ligand-based drug discovery (LBDD) models and drug–target interaction (DTI) models. LBDD models treat each protein as its own machine learning model, thereby limiting inference (prediction) to targets that already have sufficient data to train models. DTI models train a global model to predict binding drug–target pairs, such that protein targets learn from the bioactivities of similar proteins.
Figure 2
Figure 2. Pocket selection. (A) Visualization of P2Rank predicted pockets. (B,C) Cartoon representation of Vpx from the ternary complex of DCAF1-SAMHD1-Vpx (PDB code 4CC9) showing overlay of pocket prediction with protein contacts. (D) Inside pocket. (E) Top pocket. (F) Side pocket.
Figure 3
Figure 3. Binding of CYCA-117-70 to DCAF1 and WDR5 by SPR. (A) Binding of CYCA-117-70 to DCAF1 WDR. (B) Binding to WDR5. SPR binding data (representative plot of N = 2) represented in the steady-state response (black circles) with the steady state 1:1 binding model fitting (red dashed line) and the sensorgram (solid green) with the kinetic fit (black dots). CYCA-117-70 showed binding to DCAF1 with an estimated KD of 70 μM (since the binding curve does not fully reach saturation) and no significant binding to WDR5 (KD not determined).
Figure 4
Figure 4. Cocrystal structure of the DCAF1 WDR domain in complex with CYCA-117-70. (A) Top and side views of the DCAF1 WDR domain shown as a cartoon representation in slate blue, bound to CYCA-117-70 shown as yellow sticks. The compound binds close to the surface of the WDR ring central channel. (B) Zoomed-in view of the CYCA-117-70 binding site in chain A of the DCAF1-CYCA-117-70 cocrystal structure. CYCA-117-70 is shown as yellow sticks, water molecules are shown as red spheres, and the putative hydrogen bond is shown as black dashes. (C) Overlay of the DCAF1 monomer (slate blue surface) bound to CYCA-117-70 (yellow sticks) on to lentiviral Vpx (green) (PDB ID 4CC9, data from ref (22), revealing a steric overlap between the two ligands.
References
This article references 37 other publications.
- 1Ehrlich, P. The Mutual Relations between Toxin and Antitoxin. Boston Med. Surg. J. 1904, 150, 443– 445, DOI: 10.1056/NEJM190404281501701There is no corresponding record for this reference.
- 2Langley, J. N. On the Reaction of Cells and of Nerve-Endings to Certain Poisons, Chiefly as Regards the Reaction of Striated Muscle to Nicotine and to Curari. J. Physiol. 1905, 33, 374– 413, DOI: 10.1113/jphysiol.1905.sp0011282Over the reaction of the cells and the nerve ending to certain poisons, in particular in consideration of the reaction of the transverse striped muscle to nicotine and to curare. [machine translation]Langley, J. N.Journal of Physiology (Oxford, United Kingdom) (1905), 33 (), 374-413CODEN: JPHYA7 ISSN:.[Machine Translation of Descriptors]. Nicotine causes extended contraction in certain muscles of the birds. Also, after cutting through the muscle supplying nerves as after paralysis, one obtains muscle contraction the same by nicotine or curare. The contraction caused by nicotine is decreased by the injection of a sufficient quantity of curare; the two poisons are in their effect on the muscle antagonist. Degeneration of the nerves belonging to the muscles does not change anything in their effect. From it, experiments results that nicotine and curare not on the nerve ending, but on the muscle affects, not directly on the contractile substance, but other materials in the muscle, which calls author "receptive substance" (receipt substance). As author of the closer implements, similar conditions are present not only with the transverse striped muscle fibers, but also with other cell kinds, and probably the larger number of the materials practices their effect not, as accepted, on the nerve ending, select on the "receptive substance".
- 3MacKinnon, S. S.; Madani Tonekaboni, S. A.; Windemuth, A. Proteome-Scale Drug-Target Interaction Predictions: Approaches and Applications. Curr. Protoc. 2021, 1, e302 DOI: 10.1002/cpz1.302There is no corresponding record for this reference.
- 4Schapira, M.; Tyers, M.; Torrent, M.; Arrowsmith, C. H. WD40 Repeat Domain Proteins: A Novel Target Class?. Nat. Rev. Drug Discov. 2017, 16, 773– 786, DOI: 10.1038/nrd.2017.1794WD40 repeat domain proteins: a novel target class?Schapira, Matthieu; Tyers, Mike; Torrent, Maricel; Arrowsmith, Cheryl H.Nature Reviews Drug Discovery (2017), 16 (11), 773-786CODEN: NRDDAG; ISSN:1474-1776. (Nature Research)Antagonism of protein-protein interactions (PPIs) with small mols. is becoming more feasible as a therapeutic approach. Successful PPI inhibitors tend to target proteins contg. deep peptide-binding grooves or pockets rather than the more common large, flat protein interaction surfaces. Here, we review one of the most abundant PPI domains in the human proteome, the WD40 repeat (WDR) domain, which has a central peptide-binding pocket and is a member of the β-propeller domain-contg. protein family. Recently, two WDR domain-contg. proteins, WDR5 and EED, as well as other β-propeller domains have been successfully targeted by potent, specific, cell-active, drug-like chem. probes. Could WDR domains be a novel target class for drug discovery. Although the research is at an early stage and therefore not clin. validated, cautious optimism is justified, as WDR domain-contg. proteins are involved in multiple disease-assocd. pathways. The druggability and structural diversity of WDR domain binding pockets suggest that understanding how to target this prevalent domain class will open up areas of disease biol. that have so far resisted drug discovery efforts.
- 5Higa, L. A.; Wu, M.; Ye, T.; Kobayashi, R.; Sun, H.; Zhang, H. CUL4-DDB1 Ubiquitin Ligase Interacts with Multiple WD40-Repeat Proteins and Regulates Histone Methylation. Nat. Cell Biol. 2006, 8, 1277– 1283, DOI: 10.1038/ncb14905CUL4-DDB1 ubiquitin ligase interacts with multiple WD40-repeat proteins and regulates histone methylationHiga, Leigh Ann; Wu, Min; Ye, Tao; Kobayashi, Ryuji; Sun, Hong; Zhang, HuiNature Cell Biology (2006), 8 (11), 1277-1283CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)The CUL4-DDB1-ROC1 ubiquitin E3 ligase regulates cell-cycle progression, replication and DNA damage response. However, the substrate-specific adaptors of this ligase remain uncharacterized. Here, we show that CUL4-DDB1 complexes interact with multiple WD40-repeat proteins (WDRs) including TLE1-3, WDR5, L2DTL (also known as CDT2) and the Polycomb-group protein EED (also known as ESC). WDR5 and EED are core components of histone methylation complexes that are essential for histone H3 methylation and epigenetic control at K4 or K9 and K27, resp., whereas L2DTL regulates CDT1 proteolysis after DNA damage through CUL4-DDB1 (ref. 8). We found that CUL4A-DDB1 interacts with H3 methylated mononucleosomes and peptides. Inactivation of either CUL4 or DDB1 impairs these histone modifications. However, loss of WDR5 specifically affects histone H3 methylation at K4 but not CDT1 degrdn., whereas inactivation of L2DTL prevents CDT1 degrdn. but not histone methylation. Our studies suggest that CUL4-DDB1 ligases use WDR proteins as mol. adaptors for substrate recognition, and modulate multiple biol. processes through ubiquitin-dependent proteolysis.
- 6Sharma, P.; Nag, A. CUL4A Ubiquitin Ligase: A Promising Drug Target for Cancer and Other Human Diseases. Open Biol. 2014, 4, 130217 DOI: 10.1098/rsob.1302176CUL4A ubiquitin ligase: a promising drug target for cancer and other human diseasesSharma, Puneet; Nag, AloOpen Biology (2014), 4 (2), 130217/1-130217/15, 15CODEN: OBPICQ; ISSN:2046-2441. (Royal Society)A review. The ability of cullin 4A (CUL4A), a scaffold protein, to recruit a repertoire of substrate adaptors allows it to assemble into distinct E3 ligase complexes to mediate turnover of key regulatory proteins. In the past decade, a considerable wealth of information has been generated regarding its biol., regulation, assembly, mol. architecture and novel functions. Importantly, unravelling of its assocn. with multiple tumors and modulation by viral proteins establishes it as one of the key proteins that may play an important role in cellular transformation. Considering the role of its substrate in regulating the cell cycle and maintenance of genomic stability, understanding the detailed aspects of these processes will have significant consequences for the treatment of cancer and related diseases. This review is an effort to provide a broad overview of this multifaceted ubiquitin ligase and addresses its crit. role in regulation of important biol. processes. More importantly, its tremendous potential to be exploited for therapeutic purposes has been discussed.
- 7Maddika, S.; Chen, J. Protein Kinase DYRK2 Is a Scaffold That Facilitates Assembly of an E3 Ligase. Nat. Cell Biol. 2009, 11, 409– 419, DOI: 10.1038/ncb18487Protein kinase DYRK2 is a scaffold that facilitates assembly of an E3 ligaseMaddika, Subbareddy; Chen, JunjieNature Cell Biology (2009), 11 (4), 409-419CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)Protein kinases have central functions in various cellular signal transduction pathways through their substrate phosphorylation. Here we show that a protein kinase, DYRK2, has unexpected role as a scaffold for an E3 ubiquitin ligase complex. DYRK2 assocs. with an E3 ligase complex contg. EDD, DDB1, and VPRBP proteins (EDVP complex). Strikingly, DYRK2 serves as a scaffold for the EDVP complex, because small-interfering-RNA-mediated depletion of DYRK2 disrupts the formation of the EDD-DDB1-VPRBP complex. Although the kinase activity of DYRK2 is dispensable for its ability to mediate EDVP complex formation, it is required for the phosphorylation and subsequent degrdn. of its downstream substrate, katanin p60. Collectively, our results reveal a new type of E3-ubiquitin ligase complex in humans that depends on a protein kinase for complex formation as well as for the subsequent phosphorylation, ubiquitylation, and degrdn. of their substrates.
- 8Angers, S.; Li, T.; Yi, X.; MacCoss, M. J.; Moon, R. T.; Zheng, N. Molecular Architecture and Assembly of the DDB1-CUL4A Ubiquitin Ligase Machinery. Nature 2006, 443, 590– 593, DOI: 10.1038/nature051758Molecular architecture and assembly of the DDB1-CUL4A ubiquitin ligase machineryAngers, Stephane; Li, Ti; Yi, Xianhua; MacCoss, Michael J.; Moon, Randall T.; Zheng, NingNature (London, United Kingdom) (2006), 443 (7111), 590-593CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Protein ubiquitination is a common form of post-translational modification that regulates a broad spectrum of protein substrates in diverse cellular pathways. Through a three-enzyme (E1-E2-E3) cascade, the attachment of ubiquitin to proteins is catalyzed by the E3 ubiquitin ligase, which is best represented by the superfamily of cullin-RING complexes. Conserved from yeast to human, the DDB1-CUL4-ROC1 complex is a recently identified cullin-RING ubiquitin ligase, which regulates DNA repair, DNA replication and transcription, and can also be subverted by pathogenic viruses to benefit viral infection. Lacking a canonical SKP1-like cullin adaptor and a defined substrate recruitment module, how the DDB1-CUL4-ROC1 E3 app. is assembled for ubiquitinating various substrates remains unclear. Here we present crystallog. analyses of the virally hijacked form of the human DDB1-CUL4A-ROC1 machinery, which show that DDB1 uses one β-propeller domain for cullin scaffold binding and a variably attached sep. double-β-propeller fold for substrate presentation. Through tandem-affinity purifn. of human DDB1 and CUL4A complexes followed by mass spectrometry anal., we also identify a novel family of WD40-repeat proteins, which directly bind to the double-propeller fold of DDB1 and serve as the substrate-recruiting module of the E3. Together, our structural and proteomic results reveal the structural mechanisms and mol. logic underlying the assembly and versatility of a new family of cullin-RING E3 complexes.
- 9Fischer, E. S.; Scrima, A.; Böhm, K.; Matsumoto, S.; Lingaraju, G. M.; Faty, M.; Yasuda, T.; Cavadini, S.; Wakasugi, M.; Hanaoka, F.; Iwai, S.; Gut, H.; Sugasawa, K.; Thomä, N. H. The Molecular Basis of CRL4DDB2/CSA Ubiquitin Ligase Architecture, Targeting, and Activation. Cell 2011, 147, 1024– 1039, DOI: 10.1016/j.cell.2011.10.0359The Molecular Basis of CRL4DDB2/CSA Ubiquitin Ligase Architecture, Targeting, and ActivationFischer, Eric S.; Scrima, Andrea; Boehm, Kerstin; Matsumoto, Syota; Lingaraju, Gondichatnahalli M.; Faty, Mahamadou; Yasuda, Takeshi; Cavadini, Simone; Wakasugi, Mitsuo; Hanaoka, Fumio; Iwai, Shigenori; Gut, Heinz; Sugasawa, Kaoru; Thomae, Nicolas H.Cell (Cambridge, MA, United States) (2011), 147 (5), 1024-1039CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The DDB1-CUL4-RBX1 (CRL4) ubiquitin ligase family regulates a diverse set of cellular pathways through dedicated substrate receptors (DCAFs). The DCAF DDB2 detects UV-induced pyrimidine dimers in the genome and facilitates nucleotide excision repair. We provide the mol. basis for DDB2 receptor-mediated cyclobutane pyrimidine dimer recognition in chromatin. The structures of the fully assembled DDB1-DDB2-CUL4A/B-RBX1 (CRL4DDB2) ligases reveal that the mobility of the ligase arm creates a defined ubiquitination zone around the damage, which precludes direct ligase activation by DNA lesions. Instead, the COP9 signalosome (CSN) mediates the CRL4DDB2 inhibition in a CSN5 independent, nonenzymic, fashion. In turn, CSN inhibition is relieved upon DNA damage binding to the DDB2 module within CSN-CRL4DDB2. The Cockayne syndrome A DCAF complex crystal structure shows that CRL4DCAF(WD40) ligases share common architectural features. Our data support a general mechanism of ligase activation, which is induced by CSN displacement from CRL4DCAF on substrate binding to the DCAF. Binding pyrimidine dimers on chromatin displaces an inhibitory factor from the CRL4-DDB2 ubiquitin ligase complex and activates it. Other CLR4 complexes share similar architectural features, suggesting a general mechanism of ligase activation.
- 10Nakagawa, T.; Mondal, K.; Swanson, P. C. VprBP (DCAF1): A Promiscuous Substrate Recognition Subunit That Incorporates into Both RING-Family CRL4 and HECT-Family EDD/UBR5 E3 Ubiquitin Ligases. BMC Mol. Biol. 2013, 14, 22, DOI: 10.1186/1471-2199-14-2210VprBP (DCAF1): a promiscuous substrate recognition subunit that incorporates into both RING-family CRL4 and HECT-family EDD/UBR5 E3 ubiquitin ligasesNakagawa, Tadashi; Mondal, Koushik; Swanson, Patrick C.BMC Molecular Biology (2013), 14 (), 22CODEN: BMBMC4; ISSN:1471-2199. (BioMed Central Ltd.)A review. The terminal step in the ubiquitin modification system relies on an E3 ubiquitin ligase to facilitate transfer of ubiquitin to a protein substrate. The substrate recognition and ubiquitin transfer activities of the E3 ligase may be mediated by a single polypeptide or may rely on sep. subunits. The latter organization is particularly prevalent among members of largest class of E3 ligases, the RING family, although examples of this type of arrangement have also been reported among members of the smaller HECT family of E3 ligases. This review describes recent discoveries that reveal the surprising and distinctive ability of VprBP (DCAF1) to serve as a substrate recognition subunit for a member of both major classes of E3 ligase, the RING-type CRL4 ligase and the HECT-type EDD/UBR5 ligase. The cellular processes normally regulated by VprBP-assocd. E3 ligases, and their targeting and subversion by viral accessory proteins are also discussed. Taken together, these studies provide important insights and raise interesting new questions regarding the mechanisms that regulate or subvert VprBP function in the context of both the CRL4 and EDD/UBR5 E3 ligases.
- 11Sakamoto, K. M.; Kim, K. B.; Kumagai, A.; Mercurio, F.; Crews, C. M.; Deshaies, R. J. Protacs: Chimeric Molecules That Target Proteins to the Skp1-Cullin-F Box Complex for Ubiquitination and Degradation. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 8554– 8559, DOI: 10.1073/pnas.14123079811Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradationSakamoto, Kathleen M.; Kim, Kyung B.; Kumagai, Akiko; Mercurio, Frank; Crews, Craig M.; Deshaies, Raymond J.Proceedings of the National Academy of Sciences of the United States of America (2001), 98 (15), 8554-8559CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex contg. Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degrdn. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compd., protein-targeting chimeric mol. 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the IκBα phosphopeptide that is recognized by the F-box protein β-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCFβ-TRCP, ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.
- 12Zhang, S.; Feng, Y.; Narayan, O.; Zhao, L. J. Cytoplasmic Retention of HIV-1 Regulatory Protein Vpr by Protein-Protein Interaction with a Novel Human Cytoplasmic Protein VprBP. Gene 2001, 263, 131– 140, DOI: 10.1016/s0378-1119(00)00583-712Cytoplasmic retention of HIV-1 regulatory protein Vpr by protein-protein interaction with a novel human cytoplasmic protein VprBPZhang, S.; Feng, Y.; Narayan, O.; Zhao, L.-J.Gene (2001), 263 (1-2), 131-140CODEN: GENED6; ISSN:0378-1119. (Elsevier Science B.V.)Vpr is an HIV-1 auxiliary regulatory protein packaged in the virion. It has been shown to enhance the nuclear transport of the HIV-1 pre-integration complex, activate transcription of cellular and viral promoters, and arrest the cell cycle at the G2/M check-point. We previously identified a cellular protein of 180 kDa (RIP) that interacted with HIV-1 Vpr specifically. We now rename this cellular protein as Vpr-binding protein, or VprBP. In this report, we describe the cloning of the VprBP cDNA that encodes 1507 aa residues and is identical to the previously cloned cDNA KIAA0800. We demonstrate that Vpr specifically interacts with recombinantly expressed VprBP in vitro as well as in vivo. Furthermore, Vpr interacts with the cellular endogenous VprBP in the context of the HIV-1 life cycle. Mutational anal. of VprBP suggests that the Vpr binding domain is located within the C-terminal half of VprBP, which has a Pro-rich domain and several Phe-x-x-Phe repeats. Subcellular fractionation studies show that both the endogenous VprBP and the adenovirus-expressed VprBP are distributed predominantly in the cytoplasmic fraction. Consistent with previous reports, the adenovirus-expressed Vpr is distributed in both the cytoplasmic and the nuclear fractions. However, when VprBP and Vpr are expressed together, Vpr is found almost exclusively in the cytoplasm. Expression of VprBP does not affect the nuclear transport of the adenoviral nuclear protein, pTP. VprBP expressed in insect cells also blocks the nuclear transport of a Vpr-GFP fusion protein, and VprBP mutants incapable of interacting with Vpr fail to block Vpr-GFP nuclear transport. We hypothesize that Vpr interaction with VprBP may cause changes in the host cell cytoplasm that affect HIV-1 pathogenesis as well as HIV-1 replication.
- 13Hossain, D.; Ferreira Barbosa, J. A.; Cohen, É. A.; Tsang, W. Y. HIV-1 Vpr Hijacks EDD-DYRK2-DDB1DCAF1 to Disrupt Centrosome Homeostasis. J. Biol. Chem. 2018, 293, 9448– 9460, DOI: 10.1074/jbc.RA117.00144413HIV-1 Vpr hijacks EDD-DYRK2-DDB1DCAF1 to disrupt centrosome homeostasisHossain, Delowar; Ferreira Barbosa, Jeremy A.; Cohen, Eric A.; Tsang, William Y.Journal of Biological Chemistry (2018), 293 (24), 9448-9460CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Viruses exploit the host cell machinery for their own profit. To evade innate immune sensing and promote viral replication, HIV type 1 (HIV-1) subverts DNA repair regulatory proteins and induces G2/M arrest. The preintegration complex of HIV-1 is known to traffic along microtubules and accumulate near the microtubule-organizing center. The centrosome is the major microtubule-organizing center in most eukaryotic cells, but precisely how HIV-1 impinges on centrosome biol. remains poorly understood. We report here that the HIV-1 accessory protein viral protein R (Vpr) localized to the centrosome through binding to DCAF1, forming a complex with the ubiquitin ligase EDD-DYRK2-DDB1DCAF1 and Cep78, a resident centrosomal protein previously shown to inhibit EDD-DYRK2-DDB1DCAF1. Vpr did not affect ubiquitination of Cep78. Rather, it enhanced ubiquitination of an EDD-DYRK2-DDB1DCAF1 substrate, CP110, leading to its degrdn., an effect that could be overcome by Cep78 expression. The down-regulation of CP110 and elongation of centrioles provoked by Vpr were independent of G2/M arrest. Infection of T lymphocytes with HIV-1, but not with HIV-1 lacking Vpr, promoted CP110 degrdn. and centriole elongation. Elongated centrioles recruited more γ-tubulin to the centrosome, resulting in increased microtubule nucleation. Our results suggest that Vpr is targeted to the centrosome where it hijacks a ubiquitin ligase, disrupting organelle homeostasis, which may contribute to HIV-1 pathogenesis.
- 14Schabla, N. M.; Mondal, K.; Swanson, P. C. DCAF1 (VprBP): Emerging Physiological Roles for a Unique Dual-Service E3 Ubiquitin Ligase Substrate Receptor. J. Mol. Cell Biol. 2019, 11, 725– 735, DOI: 10.1093/jmcb/mjy08514DCAF1 (VprBP): emerging physiological roles for a unique dual-service E3 ubiquitin ligase substrate receptorSchabla, N. Max; Mondal, Koushik; Swanson, Patrick C.Journal of Molecular Cell Biology (Oxford, United Kingdom) (2019), 11 (9), 725-735CODEN: JMCBCU; ISSN:1759-4685. (Oxford University Press)A review. Cullin-RING ligases (CRLs) comprise a large group of modular eukaryotic E3 ubiquitin ligases. Within this family, the CRL4 ligase (consisting of the Cullin4 [CUL4] scaffold protein, the Rbx1 RING finger domain protein, the DNA damage-binding protein 1 [DDB1], and one of many DDB1-assocd. substrate receptor proteins) has been intensively studied in recent years due to its involvement in regulating various cellular processes, its role in cancer development and progression, and its subversion by viral accessory proteins. Initially discovered as a target for hijacking by the human immunodeficiency virus accessory protein r, the normal targets and function of the CRL4 substrate receptor protein DDB1-Cul4-assocd. factor 1 (DCAF1; also known as VprBP) had remained elusive, but newer studies have begun to shed light on these questions. Here, we review recent progress in understanding the diverse physiol. roles of this DCAF1 in supporting various general and cell type-specific cellular processes in its context with the CRL4 E3 ligase, as well as another HECT-type E3 ligase with which DCAF1 also assocs., called EDD/UBR5. We also discuss emerging questions and areas of future study to uncover the dynamic roles of DCAF1 in normal physiol.
- 15Hrecka, K.; Gierszewska, M.; Srivastava, S.; Kozaczkiewicz, L.; Swanson, S. K.; Florens, L.; Washburn, M. P.; Skowronski, J. Lentiviral Vpr Usurps Cul4-DDB1[VprBP] E3 Ubiquitin Ligase to Modulate Cell Cycle. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 11778– 11783, DOI: 10.1073/pnas.070210210415Lentiviral Vpr usurps Cul4-DDB1[VprBP] E3 ubiquitin ligase to modulate cell cycleHrecka, Kasia; Gierszewska, Magdalena; Srivastava, Smita; Kozackiewicz, Lukasz; Swanson, Selene K.; Florens, Laurence; Washburn, Michael P.; Skowronski, JacekProceedings of the National Academy of Sciences of the United States of America (2007), 104 (28), 11778-11783, S11778/1-S11778/6CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The replication of viruses depends on the cell cycle status of the infected cells. Viruses have evolved functions that alleviate restrictions imposed on their replication by the host. Vpr, an accessory factor of primate lentiviruses, arrests cells at the DNA damage checkpoint in G2 phase of the cell cycle, but the mechanism underlying this effect has remained elusive. Here we report that Vpr proteins of both the human (HIV-1) and the distantly related simian (SIVmac) immunodeficiency viruses specifically assoc. with a protein complex comprising subunits of E3 ubiquitin ligase assembled on Cullin-4 scaffold (Cul4-DDB1[VprBP]). We show that Vpr binding to Cul4-DDB1[VprBP] leads to increased neddylation and elevated intrinsic ubiquitin ligase activity of this E3. This effect is mediated through the VprBP subunit of the complex, which recently has been suggested to function as a substrate receptor for Cu14. We also demonstrate that VprBP regulates G, phase and is essential for the completion of DNA replication in S phase. Furthermore, the ability of Vpr to arrest cells in G2 phase correlates with its ability to interact with Cu14-DDB1[VprBP] E3 complex. Our studies identify the Cu14-DDB1[VprBP] E3 ubiquitin ligase complex as the downstream effector of lentiviral Vpr for the induction of cell cycle arrest in G2 phase and suggest that Vpr may use this complex to perturb other aspects of the cell cycle and DNA metab. in infected cells.
- 16Guo, Z.; Kong, Q.; Liu, C.; Zhang, S.; Zou, L.; Yan, F.; Whitmire, J. K.; Xiong, Y.; Chen, X.; Wan, Y. Y. DCAF1 Controls T-Cell Function via P53-Dependent and -Independent Mechanisms. Nat. Commun. 2016, 7, 10307, DOI: 10.1038/ncomms1030716DCAF1 controls T-cell function via p53-dependent and -independent mechanismsGuo, Zengli; Kong, Qing; Liu, Cui; Zhang, Song; Zou, Liyun; Yan, Feng; Whitmire, Jason K.; Xiong, Yue; Chen, Xian; Wan, Yisong Y.Nature Communications (2016), 7 (), 10307CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)On activation, naive T cells grow in size and enter cell cycle to mount immune response. How the fundamental processes of T-cell growth and cell cycle entry are regulated is poorly understood. Here we report that DCAF1 (Ddb1-cullin4-assocd.-factor 1) is essential for these processes. The deletion of DCAF1 in T cells impairs their peripheral homeostasis. DCAF1 is upregulated on T-cell receptor activation and crit. for activation-induced T-cell growth, cell cycle entry and proliferation. In addn., DCAF1 is required for T-cell expansion and function during anti-viral and autoimmune responses in vivo. DCAF1 deletion leads to a drastic stabilization of p53 protein, which can be attributed to a requirement of DCAF1 for MDM2-mediated p53 poly-ubiquitination. Importantly, p53 deletion rescues the cell cycle entry defect but not the growth defect of DCAF1-deficient cells. Therefore, DCAF1 is vital for T-cell function through p53-dependent and -independent mechanisms.
- 17Tao, Y.; Remillard, D.; Vinogradova, E. V.; Yokoyama, M.; Banchenko, S.; Schwefel, D.; Melillo, B.; Schreiber, S. L.; Zhang, X.; Cravatt, B. F. Targeted Protein Degradation by Electrophilic PROTACs That Stereoselectively and Site-Specifically Engage DCAF1. J. Am. Chem. Soc. 2022, 144, 18688– 18699, DOI: 10.1021/jacs.2c0896417Targeted Protein Degradation by Electrophilic PROTACs that Stereoselectively and Site-Specifically Engage DCAF1Tao, Yongfeng; Remillard, David; Vinogradova, Ekaterina V.; Yokoyama, Minoru; Banchenko, Sofia; Schwefel, David; Melillo, Bruno; Schreiber, Stuart L.; Zhang, Xiaoyu; Cravatt, Benjamin F.Journal of the American Chemical Society (2022), 144 (40), 18688-18699CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Targeted protein degrdn. induced by heterobifunctional compds. and mol. glues presents an exciting avenue for chem. probe and drug discovery. To date, small-mol. ligands have been discovered for only a limited no. of E3 ligases, which is an important limiting factor for realizing the full potential of targeted protein degrdn. We report herein the discovery by chem. proteomics of azetidine acrylamides that stereoselectively and site-specifically react with a cysteine (C1113) in the E3 ligase substrate receptor DCAF1. We demonstrate that the azetidine acrylamide ligands for DCAF1 can be developed into electrophilic proteolysis-targeting chimeras (PROTACs) that mediated targeted protein degrdn. in human cells. We show that this process is stereoselective and does not occur in cells expressing a C1113A mutant of DCAF1. Mechanistic studies indicate that only low fractional engagement of DCAF1 is required to support protein degrdn. by electrophilic PROTACs. These findings, taken together, demonstrate how the chem. proteomic anal. of stereochem. defined electrophilic compd. sets can uncover ligandable sites on E3 ligases that support targeted protein degrdn.
- 18Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F. T.; de Beer, T. A. P.; Rempfer, C.; Bordoli, L.; Lepore, R.; Schwede, T. SWISS-MODEL: Homology Modelling of Protein Structures and Complexes. Nucleic Acids Res. 2018, 46, W296– W303, DOI: 10.1093/nar/gky42718SWISS-MODEL: homology modelling of protein structures and complexesWaterhouse, Andrew; Bertoni, Martino; Bienert, Stefan; Studer, Gabriel; Tauriello, Gerardo; Gumienny, Rafal; Heer, Florian T.; de Beer, Tjaart A. P.; Rempfer, Christine; Bordoli, Lorenza; Lepore, Rosalba; Schwede, TorstenNucleic Acids Research (2018), 46 (W1), W296-W303CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Homol. modeling has matured into an important technique in structural biol., significantly contributing to narrowing the gap between known protein sequences and exptl. detd. structures. Fully automated workflows and servers simplify and streamline the homol. modeling process, also allowing users without a specific computational expertise to generate reliable protein models and have easy access to modeling results, their visualization and interpretation. Here, we present an update to the SWISS-MODEL server, which pioneered the field of automated modeling 25 years ago and been continuously further developed. Recently, its functionality has been extended to the modeling of homo- and heteromeric complexes. Starting from the amino acid sequences of the interacting proteins, both the stoichiometry and the overall structure of the complex are inferred by homol. modeling. Other major improvements include the implementation of a new modeling engine, ProMod3 and the introduction a new local model quality estn. method, QMEANDisCo.
- 19Sugiyama, M. G.; Cui, H.; Redka, D. S.; Karimzadeh, M.; Rujas, E.; Maan, H.; Hayat, S.; Cheung, K.; Misra, R.; McPhee, J. B.; Viirre, R. D.; Haller, A.; Botelho, R. J.; Karshafian, R.; Sabatinos, S. A.; Fairn, G. D.; Madani Tonekaboni, S. A.; Windemuth, A.; Julien, J.-P.; Shahani, V.; MacKinnon, S. S.; Wang, B.; Antonescu, C. N. Multiscale Interactome Analysis Coupled with Off-Target Drug Predictions Reveals Drug Repurposing Candidates for Human Coronavirus Disease. Sci. Rep. 2021, 11, 23315, DOI: 10.1038/s41598-021-02432-719Multiscale interactome analysis coupled with off-target drug predictions reveals drug repurposing candidates for human coronavirus diseaseSugiyama, Michael G.; Cui, Haotian; Redka, Dar'ya S.; Karimzadeh, Mehran; Rujas, Edurne; Maan, Hassaan; Hayat, Sikander; Cheung, Kyle; Misra, Rahul; McPhee, Joseph B.; Viirre, Russell D.; Haller, Andrew; Botelho, Roberto J.; Karshafian, Raffi; Sabatinos, Sarah A.; Fairn, Gregory D.; Madani Tonekaboni, Seyed Ali; Windemuth, Andreas; Julien, Jean-Philippe; Shahani, Vijay; MacKinnon, Stephen S.; Wang, Bo; Antonescu, Costin N.Scientific Reports (2021), 11 (1), 23315CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)Abstr.: The COVID-19 pandemic has highlighted the urgent need for the identification of new antiviral drug therapies for a variety of diseases. COVID-19 is caused by infection with the human coronavirus SARS-CoV-2, while other related human coronaviruses cause diseases ranging from severe respiratory infections to the common cold. We developed a computational approach to identify new antiviral drug targets and repurpose clin.-relevant drug compds. for the treatment of a range of human coronavirus diseases. Our approach is based on graph convolutional networks (GCN) and involves multiscale host-virus interactome anal. coupled to off-target drug predictions. Cell-based exptl. assessment reveals several clin.-relevant drug repurposing candidates predicted by the in silico analyses to have antiviral activity against human coronavirus infection. In particular, we identify the MET inhibitor capmatinib as having potent and broad antiviral activity against several coronaviruses in a MET-independent manner, as well as novel roles for host cell proteins such as IRAK1/4 in supporting human coronavirus infection, which can inform further drug discovery studies.
- 20He, Y.; Yang, C.; Wang, Y.; Sacher, J. R.; Sims, M. M.; Pfeffer, L. M.; Miller, D. D. Novel Structural-Related Analogs of PFI-3 (SRAPs) That Target the BRG1 Catalytic Subunit of the SWI/SNF Complex Increase the Activity of Temozolomide in Glioblastoma Cells. Bioorg. Med. Chem. 2022, 53, 116533 DOI: 10.1016/j.bmc.2021.11653320Novel structural-related analogs of PFI-3 (SRAPs) that target the BRG1 catalytic subunit of the SWI/SNF complex increase the activity of temozolomide in glioblastoma cellsHe, Yali; Yang, Chuanhe; Wang, Yinan; Sacher, Joshua R.; Sims, Michelle M.; Pfeffer, Lawrence M.; Miller, Duane D.Bioorganic & Medicinal Chemistry (2022), 53 (), 116533CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Glioblastoma (GBM) is the most aggressive and treatment-refractory malignant adult brain cancer. After std. of care therapy, the overall median survival for GBM is only ∼6 mo with a 5-yr survival <10%. Although some patients initially respond to the DNA alkylating agent temozolomide (TMZ), unfortunately most patients become resistant to therapy and brain tumors eventually recur. We previously found that knockout of BRG1 or treatment with PFI-3, a small mol. inhibitor of the BRG1 bromodomain, enhances sensitivity of GBM cells to temozolomide in vitro and in vivo GBM animal models. Those results demonstrated that the BRG1 catalytic subunit of the SWI/SNF chromatin remodeling complex appears to play a crit. role in regulating TMZ-sensitivity. In the present study we designed and synthesized Structurally Related Analogs of PFI-3 (SRAPs) and tested their bioactivity in vitro. Among of the SRAPs, 9f and 11d show better efficacy than PFI-3 in sensitizing GBM cells to the antiproliferative and cell death inducing effects of temozolomide in vitro, as well as enhancing the inhibitor effect of temozolomide on the growth of s.c. GBM tumors.
- 21Krivák, R.; Hoksza, D. P2Rank: Machine Learning Based Tool for Rapid and Accurate Prediction of Ligand Binding Sites from Protein Structure. Aust. J. Chem. 2018, 10, 39, DOI: 10.1186/s13321-018-0285-8There is no corresponding record for this reference.
- 22Schwefel, D.; Groom, H. C. T.; Boucherit, V. C.; Christodoulou, E.; Walker, P. A.; Stoye, J. P.; Bishop, K. N.; Taylor, I. A. Structural Basis of Lentiviral Subversion of a Cellular Protein Degradation Pathway. Nature 2014, 505, 234– 238, DOI: 10.1038/nature1281522Structural basis of lentiviral subversion of a cellular protein degradation pathwaySchwefel, David; Groom, Harriet C. T.; Boucherit, Virginie C.; Christodoulou, Evangelos; Walker, Philip A.; Stoye, Jonathan P.; Bishop, Kate N.; Taylor, Ian A.Nature (London, United Kingdom) (2014), 505 (7482), 234-238CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Lentiviruses contain accessory genes that have evolved to counteract the effects of host cellular defense proteins that inhibit productive infection. One such restriction factor, SAMHD1, inhibits human immunodeficiency virus (HIV)-1 infection of myeloid-lineage cells as well as resting CD4+ T cells by reducing the cellular deoxynucleoside 5'-triphosphate (dNTP) concn. to a level at which the viral reverse transcriptase cannot function. In other lentiviruses, including HIV-2 and related simian immunodeficiency viruses (SIVs), SAMHD1 restriction is overcome by the action of viral accessory protein x (Vpx) or the related viral protein r (Vpr) that target and recruit SAMHD1 for proteasomal degrdn. The mol. mechanism by which these viral proteins are able to usurp the host cell's ubiquitination machinery to destroy the cell's protection against these viruses has not been defined. Here we present the crystal structure of a ternary complex of Vpx with the human E3 ligase substrate adaptor DCAF1 and the carboxy-terminal region of human SAMHD1. Vpx is made up of a three-helical bundle stabilized by a zinc finger motif, and wraps tightly around the disk-shaped DCAF1 mol. to present a new mol. surface. This adapted surface is then able to recruit SAMHD1 via its C terminus, making it a competent substrate for the E3 ligase to mark for proteasomal degrdn. The structure reported here provides a mol. description of how a lentiviral accessory protein is able to subvert the cell's normal protein degrdn. pathway to inactivate the cellular viral defense system.
- 23The UniProt Consortium UniProt: The Universal Protein Knowledgebase in 2023. Nucleic Acids Res. 2023, 51, D523– D531, DOI: 10.1093/nar/gkac1052There is no corresponding record for this reference.
- 24Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235– 242, DOI: 10.1093/nar/28.1.23524The Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 25Hutchinson, A.; Seitova, A. Production of Recombinant PRMT Proteins Using the Baculovirus Expression Vector System. J. Vis. Exp. JoVE 2021, No. 173. DOI: 10.3791/62510 .There is no corresponding record for this reference.
- 26Minor, W.; Cymborowski, M.; Otwinowski, Z.; Chruszcz, M. HKL-3000: The Integration of Data Reduction and Structure Solution--from Diffraction Images to an Initial Model in Minutes. Acta Crystallogr. D Biol. Crystallogr. 2006, 62, 859– 866, DOI: 10.1107/S090744490601994926HKL-3000: the integration of data reduction and structure solution - from diffraction images to an initial model in minutesMinor, Wladek; Cymborowski, Marcin; Otwinowski, Zbyszek; Chruszcz, MaksymilianActa Crystallographica, Section D: Biological Crystallography (2006), D62 (8), 859-866CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)A new approach that integrates data collection, data redn., phasing and model building significantly accelerates the process of structure detn. and on av. minimizes the no. of data sets and synchrotron time required for structure soln. Initial testing of the HKL-3000 system (the beta version was named HKL-2000_ph) with more than 140 novel structure detns. has proven its high value for MAD/SAD expts. The heuristics for choosing the best computational strategy at different data resoln. limits of phasing signal and crystal diffraction are being optimized. The typical end result is an interpretable electron-d. map with a partially built structure and, in some cases, an almost complete refined model. The current development is oriented towards very fast structure soln. in order to provide feedback during the diffraction expt. Work is also proceeding towards improving the quality of phasing calcn. and model building.
- 27McCoy, A. J.; Grosse-Kunstleve, R. W.; Adams, P. D.; Winn, M. D.; Storoni, L. C.; Read, R. J. Phaser Crystallographic Software. J. Appl. Crystallogr. 2007, 40, 658– 674, DOI: 10.1107/S002188980702120627Phaser crystallographic softwareMcCoy, Airlie J.; Grosse-Kunstleve, Ralf W.; Adams, Paul D.; Winn, Martyn D.; Storoni, Laurent C.; Read, Randy J.Journal of Applied Crystallography (2007), 40 (4), 658-674CODEN: JACGAR; ISSN:0021-8898. (International Union of Crystallography)Phaser is a program for phasing macromol. crystal structures by both mol. replacement and exptl. phasing methods. The novel phasing algorithms implemented in Phaser have been developed using max. likelihood and multivariate statistics. For mol. replacement, the new algorithms have proved to be significantly better than traditional methods in discriminating correct solns. from noise, and for single-wavelength anomalous dispersion exptl. phasing, the new algorithms, which account for correlations between F+ and F-, give better phases (lower mean phase error with respect to the phases given by the refined structure) than those that use mean F and anomalous differences ΔF. One of the design concepts of Phaser was that it be capable of a high degree of automation. To this end, Phaser (written in C++) can be called directly from Python, although it can also be called using traditional CCP4 keyword-style input. Phaser is a platform for future development of improved phasing methods and their release, including source code, to the crystallog. community.
- 28Emsley, P.; Cowtan, K. Coot: Model-Building Tools for Molecular Graphics. Acta Crystallogr. D Biol. Crystallogr. 2004, 60, 2126– 2132, DOI: 10.1107/S090744490401915828Coot: model-building tools for molecular graphicsEmsley, Paul; Cowtan, KevinActa Crystallographica, Section D: Biological Crystallography (2004), D60 (12, Pt. 1), 2126-2132CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)CCP4mg is a project that aims to provide a general-purpose tool for structural biologists, providing tools for x-ray structure soln., structure comparison and anal., and publication-quality graphics. The map-fitting tools are available as a stand-alone package, distributed as 'Coot'.
- 29Murshudov, G. N.; Vagin, A. A.; Dodson, E. J. Refinement of Macromolecular Structures by the Maximum-Likelihood Method. Acta Crystallogr. D Biol. Crystallogr. 1997, 53, 240– 255, DOI: 10.1107/S090744499601225529Refinement of macromolecular structures by the maximum-likelihood methodMurshudov, Garib N.; Vagin, Alexei A.; Dodson, Eleanor J.Acta Crystallographica, Section D: Biological Crystallography (1997), D53 (3), 240-255CODEN: ABCRE6; ISSN:0907-4449. (Munksgaard)A review with many refs. on the math. basis of max. likelihood. The likelihood function for macromol. structures is extended to include prior phase information and exptl. std. uncertainties. The assumption that different parts of a structure might have different errors is considered. A method for estg. σA using "free" reflections is described and its effects analyzed. The derived equations have been implemented in the program REFMAC. This has been tested on several proteins at different stages of refinement (bacterial α-amylase, cytochrome c', cross-linked insulin and oligopeptide binding protein). The results derived using the max.-likelihood residual are consistently better than those obtained from least-squares refinement.
- 30Winn, M. D.; Ballard, C. C.; Cowtan, K. D.; Dodson, E. J.; Emsley, P.; Evans, P. R.; Keegan, R. M.; Krissinel, E. B.; Leslie, A. G. W.; McCoy, A.; McNicholas, S. J.; Murshudov, G. N.; Pannu, N. S.; Potterton, E. A.; Powell, H. R.; Read, R. J.; Vagin, A.; Wilson, K. S. Overview of the CCP4 Suite and Current Developments. Acta Crystallogr. D Biol. Crystallogr. 2011, 67, 235– 242, DOI: 10.1107/S090744491004574930Overview of the CCP4 suite and current developmentsWinn, Martyn D.; Ballard, Charles C.; Cowtan, Kevin D.; Dodson, Eleanor J.; Emsley, Paul; Evans, Phil R.; Keegan, Ronan M.; Krissinel, Eugene B.; Leslie, Andrew G. W.; McCoy, Airlie; McNicholas, Stuart J.; Murshudov, Garib N.; Pannu, Navraj S.; Potterton, Elizabeth A.; Powell, Harold R.; Read, Randy J.; Vagin, Alexei; Wilson, Keith S.Acta Crystallographica, Section D: Biological Crystallography (2011), 67 (4), 235-242CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)A review. The CCP4 (Collaborative Computational Project, No. 4) software suite is a collection of programs and assocd. data and software libraries which can be used for macromol. structure detn. by X-ray crystallog. The suite is designed to be flexible, allowing users a no. of methods of achieving their aims. The programs are from a wide variety of sources but are connected by a common infrastructure provided by std. file formats, data objects and graphical interfaces. Structure soln. by macromol. crystallog. is becoming increasingly automated and the CCP4 suite includes several automation pipelines. After giving a brief description of the evolution of CCP4 over the last 30 years, an overview of the current suite is given. While detailed descriptions are given in the accompanying articles, here it is shown how the individual programs contribute to a complete software package.
- 31Chen, V. B.; Arendall, W. B.; Headd, J. J.; Keedy, D. A.; Immormino, R. M.; Kapral, G. J.; Murray, L. W.; Richardson, J. S.; Richardson, D. C. MolProbity: All-Atom Structure Validation for Macromolecular Crystallography. Acta Crystallogr. D Biol. Crystallogr. 2010, 66, 12– 21, DOI: 10.1107/S090744490904207331MolProbity: all-atom structure validation for macromolecular crystallographyChen, Vincent B.; Arendall, W. Bryan, III; Headd, Jeffrey J.; Keedy, Daniel A.; Immormino, Robert M.; Kapral, Gary J.; Murray, Laura W.; Richardson, Jane S.; Richardson, David C.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 12-21CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MolProbity is a structure-validation web service that provides broad-spectrum solidly based evaluation of model quality at both the global and local levels for both proteins and nucleic acids. It relies heavily on the power and sensitivity provided by optimized hydrogen placement and all-atom contact anal., complemented by updated versions of covalent-geometry and torsion-angle criteria. Some of the local corrections can be performed automatically in MolProbity and all of the diagnostics are presented in chart and graphical forms that help guide manual rebuilding. X-ray crystallog. provides a wealth of biol. important mol. data in the form of at. three-dimensional structures of proteins, nucleic acids and increasingly large complexes in multiple forms and states. Advances in automation, in everything from crystn. to data collection to phasing to model building to refinement, have made solving a structure using crystallog. easier than ever. However, despite these improvements, local errors that can affect biol. interpretation are widespread at low resoln. and even high-resoln. structures nearly all contain at least a few local errors such as Ramachandran outliers, flipped branched protein side chains and incorrect sugar puckers. It is crit. both for the crystallographer and for the end user that there are easy and reliable methods to diagnose and correct these sorts of errors in structures. MolProbity is the authors' contribution to helping solve this problem and this article reviews its general capabilities, reports on recent enhancements and usage, and presents evidence that the resulting improvements are now beneficially affecting the global database.
- 32Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF Chimera--a Visualization System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25, 1605– 1612, DOI: 10.1002/jcc.2008432UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 33DeLano, W.; Schrödinger, L. PyMOL. Retrieved Httpwwwpymolorgpymol.There is no corresponding record for this reference.
- 34Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand Efficiency: A Useful Metric for Lead Selection. Drug Discovery Today 2004, 9, 430– 431, DOI: 10.1016/S1359-6446(04)03069-734Ligand efficiency: a useful metric for lead selectionHopkins Andrew L; Groom Colin R; Alex AlexanderDrug discovery today (2004), 9 (10), 430-1 ISSN:1359-6446.There is no expanded citation for this reference.
- 35Li, A. S. M.; Kimani, S.; Wilson, B.; Noureldin, M.; González-Álvarez, H.; Mamai, A.; Hoffer, L.; Guilinger, J. P.; Zhang, Y.; von Rechenberg, M.; Disch, J. S.; Mulhern, C. J.; Slakman, B. L.; Cuozzo, J. W.; Dong, A.; Poda, G.; Mohammed, M.; Saraon, P.; Mittal, M.; Modh, P.; Rathod, V.; Patel, B.; Ackloo, S.; Santhakumar, V.; Szewczyk, M. M.; Barsyte-Lovejoy, D.; Arrowsmith, C. H.; Marcellus, R.; Guié, M.-A.; Keefe, A. D.; Brown, P. J.; Halabelian, L.; Al-Awar, R.; Vedadi, M. Discovery of Nanomolar DCAF1 Small Molecule Ligands. J. Med. Chem. 2023, 66, 5041– 5060, DOI: 10.1021/acs.jmedchem.2c0213235Discovery of Nanomolar DCAF1 Small Molecule LigandsLi, Alice Shi Ming; Kimani, Serah; Wilson, Brian; Noureldin, Mahmoud; Gonzalez-Alvarez, Hector; Mamai, Ahmed; Hoffer, Laurent; Guilinger, John P.; Zhang, Ying; von Rechenberg, Moritz; Disch, Jeremy S.; Mulhern, Christopher J.; Slakman, Belinda L.; Cuozzo, John W.; Dong, Aiping; Poda, Gennady; Mohammed, Mohammed; Saraon, Punit; Mittal, Manish; Modh, Pratik; Rathod, Vaibhavi; Patel, Bhashant; Ackloo, Suzanne; Santhakumar, Vijayaratnam; Szewczyk, Magdalena M.; Barsyte-Lovejoy, Dalia; Arrowsmith, Cheryl H.; Marcellus, Richard; Guie, Marie-Aude; Keefe, Anthony D.; Brown, Peter J.; Halabelian, Levon; Al-awar, Rima; Vedadi, MasoudJournal of Medicinal Chemistry (2023), 66 (7), 5041-5060CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)DCAF1 is a substrate receptor of two distinct E3 ligases (CRL4DCAF1 and EDVP), plays a crit. physiol. role in protein degrdn., and is considered a drug target for various cancers. Antagonists of DCAF1 could be used toward the development of therapeutics for cancers and viral treatments. We used the WDR domain of DCAF1 to screen a 114-billion-compd. DNA encoded library (DEL) and identified candidate compds. using similarity search and machine learning. This led to the discovery of a compd. (Z1391232269) with an SPR KD of 11μM. Structure-guided hit optimization led to the discovery of OICR-8268 (26e) with an SPR KD of 38 nM and cellular target engagement with EC50 of 10μM as measured by cellular thermal shift assay (CETSA). OICR-8268 is an excellent tool compd. to enable the development of next-generation DCAF1 ligands toward cancer therapeutics, further investigation of DCAF1 functions in cells, and the development of DCAF1-based PROTACs.
- 36Schröder, M.; Renatus, M.; Liang, X.; Meili, F.; Zoller, T.; Ferrand, S.; Gauter, F.; Li, X.; Sigoillot, F.; Gleim, S.; Stachyra, M.-T.; Thomas, J.; Begue, D.; Lefeuvre, P.; Andraos-Rey, R.; Chung, B. Y.; Ma, R.; Carbonneau, S.; Pinch, B.; Hofmann, A.; Schirle, M.; Schmiedberg, N.; Imbach, P.; Gorses, D.; Calkins, K.; Bauer-Probst, B.; Maschlej, M.; Niederst, M.; Maher, R.; Henault, M.; Alford, J.; Ahrne, E.; Hollingworth, G.; Thomä, N. H.; Vulpetti, A.; Radimerski, T.; Holzer, P.; Thoma, C. R. Reinstating Targeted Protein Degradation with DCAF1 PROTACs in CRBN PROTAC Resistant Settings. bioRxiv 2023, DOI: 10.1101/2023.04.09.536153There is no corresponding record for this reference.
- 37Gironda-Martínez, A.; Donckele, E. J.; Samain, F.; Neri, D. DNA-Encoded Chemical Libraries: A Comprehensive Review with Succesful Stories and Future Challenges. ACS Pharmacol. Transl. Sci. 2021, 4, 1265– 1279, DOI: 10.1021/acsptsci.1c0011837DNA-encoded chemical libraries: A comprehensive review with successful stories and future challengesGironda-Martinez, Adrian; Donckele, Etienne J.; Samain, Florent; Neri, DarioACS Pharmacology & Translational Science (2021), 4 (4), 1265-1279CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)A review. DNA-encoded chem. libraries (DELs) represent a versatile and powerful technol. platform for the discovery of small-mol. ligands to protein targets of biol. and pharmaceutical interest. DELs are collections of mols., individually coupled to distinctive DNA tags serving as amplifiable identification barcodes. Thanks to advances in DNA-compatible reactions, selection methodologies, next-generation sequencing, and data anal., DEL technol. allows the construction and screening of libraries of unprecedented size, which has led to the discovery of highly potent ligands, some of which have progressed to clin. trials. In this Review, we present an overview of diverse approaches for the generation and screening of DEL mol. repertoires. Recent success stories are described, detailing how novel ligands were isolated from DEL screening campaigns and were further optimized by medicinal chem. The goal of the Review is to capture some of the most recent developments in the field, while also elaborating on future challenges to further improve DEL technol. as a therapeutic discovery platform.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00082.
Figure showing the domain architecture of human DCAF1 and the WDR fold, figure of the electron density of CYCA-117-70 and its binding site, figure depicting the comparison of CYCA-117-70 with other recently reported DCAF1 ligands (PDF)
SMILES string of the 101 compounds computationally selected for experimental testing (XLSX)
SPR raw data for CYCA-117-70 (XLSX)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.