31 results on '"Mem Noble"'
Search Results
2. 276 An X-ray crystal structure-based understanding of the inhibition of the MDM2–p53 protein–protein interaction by isoindolinones
- Author
-
Timothy J. Blackburn, SJ Cully, C.J. Drummond, Mem Noble, J. Liu, B. Anil, Bernard T. Golding, C. Riedinger, K. Haggerty, C.H. Revill, E. Blackburn, Jane A. Endicott, A.F. Watson, John Lunec, Q. Xu, Yan Zhao, David R. Newell, Roger J. Griffin, and Ian R. Hardcastle
- Subjects
Cancer Research ,Crystallography ,Oncology ,Chemistry ,X-ray ,Mdm2 p53 ,Crystal structure ,Protein–protein interaction - Published
- 2014
- Full Text
- View/download PDF
3. 325 Identification and characterization of an irreversible inhibitor of CDK2
- Author
-
Mathew P. Martin, E. Anscombe, R. Mora Vidal, Mem Noble, Bernard T. Golding, U.H. Danielson, Roger J. Griffin, Jane A. Endicott, David R. Newell, Stephen R. Wedge, Matthis Geitmann, Lan-Zhen Wang, E. Meschini, David Staunton, and Tristan Reuillon
- Subjects
Cancer Research ,Oncology ,Chemistry ,Identification (biology) ,Computational biology ,Characterization (materials science) - Published
- 2014
- Full Text
- View/download PDF
4. 445 Structure-based design of C8-substituted O6-alkylguanine CDK1 and 2 inhibitors
- Author
-
Mem Noble, Bernard T. Golding, David R. Newell, IR Hardcastle, Roger J. Griffin, Benoit Carbain, Celine Cano, C. Roche, L. Zhen-Wang, and Jane A. Endicott
- Subjects
Cancer Research ,Oncology ,Chemistry ,Structure based ,Combinatorial chemistry - Published
- 2010
- Full Text
- View/download PDF
5. The crystal structure of cyclin A
- Author
-
Soichi Wakatsuki, Edward P. Mitchell, N.R. Brown, Mem Noble, Jane A. Endicott, Tim Hunt, Elspeth F. Garman, B. Rasmussen, and LN Johnson
- Subjects
Models, Molecular ,Protein Folding ,Cyclin E ,Transcription, Genetic ,Protein Conformation ,Cyclin D ,Recombinant Fusion Proteins ,Cyclin A ,Molecular Sequence Data ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Protein Structure, Secondary ,Cyclin D1 ,Species Specificity ,cyclin ,Cyclin-dependent kinase ,Structural Biology ,Cyclins ,CDC2-CDC28 Kinases ,Animals ,Humans ,Computer Simulation ,Amino Acid Sequence ,Molecular Biology ,Cyclin ,biology ,Sequence Homology, Amino Acid ,Cyclin-Dependent Kinase 2 ,protein kinase ,regulation ,Cyclin-Dependent Kinases ,Peptide Fragments ,Protein Structure, Tertiary ,Biochemistry ,biology.protein ,Cyclin-dependent kinase complex ,Transcription Factor TFIIB ,Cattle ,cell cycle ,X-ray structure ,Sequence Alignment ,Cyclin A2 ,Algorithms ,Protein Binding ,Transcription Factors - Abstract
Background: Eukaryotic cell cycle progression is regulated by cyclin dependent protein kinases (CDKs) whose activity is regulated by association with cyclins and by reversible phosphorylation. Cyclins also determine the subcellular location and substrate specificity of CDKs. Cyclins exhibit diverse sequences but all share homology over a region of approximately 100 amino acids, termed the cyclin box. From the determination of the structure of cyclin A, together with results from biochemical and genetic analyses, we can identify which parts of the cyclin molecule may contribute to cyclin A structure and function. Results We have solved the crystal structure, at 2.0 a resolution, of an active recombinant fragment of bovine cyclin A, cyclin A-3, corresponding to residues 171–432 of human cyclin A. The cyclin box has an α -helical fold comprising five α helices. This fold is repeated in the C-terminal region, although this region shares negligible sequence similarity with the cyclin box. Conclusion Analysis of residues that are conserved throughout the A, B, and E cyclins identifies two exposed clusters of residues, one of which has recently been shown to be involved in the association with human CDK2. The second cluster may identify another site of cyclin A–protein interaction. Comparison of the structure of the unbound cyclin with the structure of cyclin A complexed with CDK2 reveals that cyclin A does not undergo any significant conformational changes on complex formation. Threading analysis shows that the cyclin-box fold is consistent with the sequences of the transcription factor TFIIB and other functionally related proteins. The structural results indicate a role for the cyclin-box fold both as a template for the cyclin family and as a generalised adaptor molecule in the regulation of transcription.
- Full Text
- View/download PDF
6. Different applications and differentiated libraries for crystallographic fragment screening.
- Author
-
Watt J, Martin MP, Endicott JA, and Noble MEM
- Abstract
Macromolecular X-ray crystallography allows detection and characterisation of the binding of small, low-affinity chemical fragments. Here we review the utility of fragment screening for drug discovery, its potential for use in discovery science, as well as some of the distinct types of fragments that have been compiled into libraries., Competing Interests: Declaration of competing interest The authors declare no competing interests. Some work in the authors’ laboratory is supported by a research grant from Astex Pharmaceuticals., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
7. Structural requirements for the specific binding of CRABP2 to cyclin D3.
- Author
-
Pastok MW, Tomlinson CWE, Turberville S, Butler AM, Baslé A, Noble MEM, Endicott JA, Pohl E, and Tatum NJ
- Subjects
- Humans, Binding Sites, Protein Conformation, alpha-Helical, Tretinoin metabolism, Mutation, Helix-Loop-Helix Motifs, Crystallography, X-Ray, Amino Acid Sequence, Cyclin D3 metabolism, Cyclin D3 chemistry, Cyclin D3 genetics, Protein Binding, Receptors, Retinoic Acid metabolism, Receptors, Retinoic Acid chemistry, Receptors, Retinoic Acid genetics, Models, Molecular
- Abstract
Cellular retinoic acid binding protein 2 (CRABP2) transports retinoic acid from the cytoplasm to the nucleus where it then transfers its cargo to retinoic acid receptor-containing complexes leading to activation of gene transcription. We demonstrate using purified proteins that CRABP2 is also a cyclin D3-specific binding protein and that the CRABP2 cyclin D3 binding site and the proposed CRABP2 nuclear localization sequence overlap. Both sequences are within the helix-loop-helix motif that forms a lid to the retinoic acid binding pocket. Mutations within this sequence that block both cyclin D3 and retinoic acid binding promote formation of a CRABP2 structure in which the retinoic acid binding pocket is occupied by an alternative lid conformation. Structural and functional analysis of CRABP2 and cyclin D3 mutants combined with AlphaFold models of the ternary CDK4/6-cyclin D3-CRABP2 complex supports the identification of an α-helical protein binding site on the cyclin D3 C-terminal cyclin box fold., Competing Interests: Declaration of interests Some work in the authors’ laboratory is supported by a research grant from Astex Pharmaceuticals., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Cryo-EM structure of the CDK2-cyclin A-CDC25A complex.
- Author
-
Rowland RJ, Korolchuk S, Salamina M, Tatum NJ, Ault JR, Hart S, Turkenburg JP, Blaza JN, Noble MEM, and Endicott JA
- Subjects
- Humans, Protein Binding, Models, Molecular, Amino Acid Sequence, cdc25 Phosphatases metabolism, cdc25 Phosphatases chemistry, cdc25 Phosphatases ultrastructure, cdc25 Phosphatases genetics, Cyclin-Dependent Kinase 2 metabolism, Cyclin-Dependent Kinase 2 chemistry, Cyclin-Dependent Kinase 2 ultrastructure, Cryoelectron Microscopy, Cyclin A metabolism, Cyclin A chemistry
- Abstract
The cell division cycle 25 phosphatases CDC25A, B and C regulate cell cycle transitions by dephosphorylating residues in the conserved glycine-rich loop of CDKs to activate their activity. Here, we present the cryo-EM structure of CDK2-cyclin A in complex with CDC25A at 2.7 Å resolution, providing a detailed structural analysis of the overall complex architecture and key protein-protein interactions that underpin this 86 kDa complex. We further identify a CDC25A C-terminal helix that is critical for complex formation. Sequence conservation analysis suggests CDK1/2-cyclin A, CDK1-cyclin B and CDK2/3-cyclin E are suitable binding partners for CDC25A, whilst CDK4/6-cyclin D complexes appear unlikely substrates. A comparative structural analysis of CDK-containing complexes also confirms the functional importance of the conserved CDK1/2 GDSEID motif. This structure improves our understanding of the roles of CDC25 phosphatases in CDK regulation and may inform the development of CDC25-targeting anticancer strategies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Targeting Cytotoxic Agents through EGFR-Mediated Covalent Binding and Release.
- Author
-
Morese PA, Anthony N, Bodnarchuk M, Jennings C, Martin MP, Noble RA, Phillips N, Thomas HD, Wang LZ, Lister A, Noble MEM, Ward RA, Wedge SR, Stewart HL, and Waring MJ
- Subjects
- ErbB Receptors, Esters, Mass Spectrometry, Cytotoxins, Fluorouracil pharmacology
- Abstract
A major drawback of cytotoxic chemotherapy is the lack of selectivity toward noncancerous cells. The targeted delivery of cytotoxic drugs to tumor cells is a longstanding goal in cancer research. We proposed that covalent inhibitors could be adapted to deliver cytotoxic agents, conjugated to the β-position of the Michael acceptor, via an addition-elimination mechanism promoted by covalent binding. Studies on model systems showed that conjugated 5-fluorouracil (5FU) could be released upon thiol addition in relevant time scales. A series of covalent epidermal growth factor receptor (EGFR) inhibitors were synthesized as their 5FU derivatives. Achieving the desired release of 5FU was demonstrated to depend on the electronics and geometry of the compounds. Mass spectrometry and NMR studies demonstrated an anilinoquinazoline acrylate ester conjugate bound to EGFR with the release of 5FU. This work establishes that acrylates can be used to release conjugated molecules upon covalent binding to proteins and could be used to develop targeted therapeutics.
- Published
- 2023
- Full Text
- View/download PDF
10. Fragment expansion with NUDELs - poised DNA-encoded libraries.
- Author
-
Salvini CLA, Darlot B, Davison J, Martin MP, Tudhope SJ, Turberville S, Kawamura A, Noble MEM, Wedge SR, Crawford JJ, and Waring MJ
- Abstract
Optimisation of the affinity of lead compounds is a critical challenge in the identification of drug candidates and chemical probes and is a process that takes many years. Fragment-based drug discovery has become established as one of the methods of choice for drug discovery starting with small, low affinity compounds. Due to their low affinity, the evolution of fragments to desirable levels of affinity is often a key challenge. The accepted best method for increasing the potency of fragments is by iterative fragment growing, which can be very time consuming and complex. Here, we introduce a paradigm for fragment hit optimisation using poised DNA-encoded chemical libraries (DELs). The synthesis of a poised DEL, a partially constructed library that retains a reactive handle, allows the coupling of any active fragment for a specific target protein, allowing rapid discovery of potent ligands. This is illustrated for bromodomain-containing protein 4 (BRD4), in which a weakly binding fragment was coupled to a 42-member poised DEL via Suzuki-Miyaura cross coupling resulting in the identification of an inhibitor with 51 nM affinity in a single step, representing an increase in potency of several orders of magnitude from an original fragment. The potency of the compound was shown to arise from the synergistic combination of substructures, which would have been very difficult to discover by any other method and was rationalised by X-ray crystallography. The compound showed attractive lead-like properties suitable for further optimisation and demonstrated BRD4-dependent cellular pharmacology. This work demonstrates the power of poised DELs to rapidly optimise fragments, representing an attractive generic approach to drug discovery., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)
- Published
- 2023
- Full Text
- View/download PDF
11. Cryo-EM structure of SKP1-SKP2-CKS1 in complex with CDK2-cyclin A-p27KIP1.
- Author
-
Rowland RJ, Heath R, Maskell D, Thompson RF, Ranson NA, Blaza JN, Endicott JA, Noble MEM, and Salamina M
- Subjects
- Cyclin-Dependent Kinase Inhibitor p27 metabolism, Cyclin A metabolism, Cryoelectron Microscopy, Cyclin-Dependent Kinases metabolism, S-Phase Kinase-Associated Proteins metabolism, CDC2-CDC28 Kinases
- Abstract
p27KIP1 (cyclin-dependent kinase inhibitor 1B, p27) is a member of the CIP/KIP family of CDK (cyclin dependent kinase) regulators that inhibit cell cycle CDKs. p27 phosphorylation by CDK1/2, signals its recruitment to the SCF
SKP2 (S-phase kinase associated protein 1 (SKP1)-cullin-SKP2) E3 ubiquitin ligase complex for proteasomal degradation. The nature of p27 binding to SKP2 and CKS1 was revealed by the SKP1-SKP2-CKS1-p27 phosphopeptide crystal structure. Subsequently, a model for the hexameric CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex was proposed by overlaying an independently determined CDK2-cyclin A-p27 structure. Here we describe the experimentally determined structure of the isolated CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex at 3.4 Å global resolution using cryogenic electron microscopy. This structure supports previous analysis in which p27 was found to be structurally dynamic, transitioning from disordered to nascent secondary structure on target binding. We employed 3D variability analysis to further explore the conformational space of the hexameric complex and uncovered a previously unidentified hinge motion centred on CKS1. This flexibility gives rise to open and closed conformations of the hexameric complex that we propose may contribute to p27 regulation by facilitating recognition with SCFSKP2 . This 3D variability analysis further informed particle subtraction and local refinement approaches to enhance the local resolution of the complex., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
12. The CCP4 suite: integrative software for macromolecular crystallography.
- Author
-
Agirre J, Atanasova M, Bagdonas H, Ballard CB, Baslé A, Beilsten-Edmands J, Borges RJ, Brown DG, Burgos-Mármol JJ, Berrisford JM, Bond PS, Caballero I, Catapano L, Chojnowski G, Cook AG, Cowtan KD, Croll TI, Debreczeni JÉ, Devenish NE, Dodson EJ, Drevon TR, Emsley P, Evans G, Evans PR, Fando M, Foadi J, Fuentes-Montero L, Garman EF, Gerstel M, Gildea RJ, Hatti K, Hekkelman ML, Heuser P, Hoh SW, Hough MA, Jenkins HT, Jiménez E, Joosten RP, Keegan RM, Keep N, Krissinel EB, Kolenko P, Kovalevskiy O, Lamzin VS, Lawson DM, Lebedev AA, Leslie AGW, Lohkamp B, Long F, Malý M, McCoy AJ, McNicholas SJ, Medina A, Millán C, Murray JW, Murshudov GN, Nicholls RA, Noble MEM, Oeffner R, Pannu NS, Parkhurst JM, Pearce N, Pereira J, Perrakis A, Powell HR, Read RJ, Rigden DJ, Rochira W, Sammito M, Sánchez Rodríguez F, Sheldrick GM, Shelley KL, Simkovic F, Simpkin AJ, Skubak P, Sobolev E, Steiner RA, Stevenson K, Tews I, Thomas JMH, Thorn A, Valls JT, Uski V, Usón I, Vagin A, Velankar S, Vollmar M, Walden H, Waterman D, Wilson KS, Winn MD, Winter G, Wojdyr M, and Yamashita K
- Subjects
- Crystallography, X-Ray, Macromolecular Substances, Proteins chemistry, Software
- Abstract
The Collaborative Computational Project No. 4 (CCP4) is a UK-led international collective with a mission to develop, test, distribute and promote software for macromolecular crystallography. The CCP4 suite is a multiplatform collection of programs brought together by familiar execution routines, a set of common libraries and graphical interfaces. The CCP4 suite has experienced several considerable changes since its last reference article, involving new infrastructure, original programs and graphical interfaces. This article, which is intended as a general literature citation for the use of the CCP4 software suite in structure determination, will guide the reader through such transformations, offering a general overview of the new features and outlining future developments. As such, it aims to highlight the individual programs that comprise the suite and to provide the latest references to them for perusal by crystallographers around the world., (open access.)
- Published
- 2023
- Full Text
- View/download PDF
13. Crystallographic fragment screening in academic cancer drug discovery.
- Author
-
Martin MP, Endicott JA, Noble MEM, and Tatum NJ
- Subjects
- Crystallography, X-Ray, Drug Discovery methods, Proteins, Drug Evaluation, Preclinical methods, Early Detection of Cancer, Neoplasms
- Abstract
Fragment-based drug discovery (FBDD) has brought several drugs to the clinic, notably to target proteins once considered to be challenging, or even undruggable. Screening in FBDD relies upon observing and/or measuring weak (millimolar-scale) binding events using biophysical techniques or crystallographic fragment screening. This latter structural approach provides no information about binding affinity but can reveal binding mode and atomic detail on protein-fragment interactions to accelerate hit-to-lead development. In recent years, high-throughput platforms have been developed at synchrotron facilities to screen thousands of fragment-soaked crystals. However, using accessible manual techniques it is possible to run informative, smaller-scale screens within an academic lab setting. This chapter describes general protocols for home laboratory-scale fragment screening, from fragment soaking through to structure solution and, where appropriate, signposts to background, protocols or alternatives elsewhere., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
14. Mapping Ligand Interactions of Bromodomains BRD4 and ATAD2 with FragLites and PepLites─Halogenated Probes of Druglike and Peptide-like Molecular Interactions.
- Author
-
Davison G, Martin MP, Turberville S, Dormen S, Heath R, Heptinstall AB, Lawson M, Miller DC, Ng YM, Sanderson JN, Hope I, Wood DJ, Cano C, Endicott JA, Hardcastle IR, Noble MEM, and Waring MJ
- Subjects
- Ligands, Protein Domains, Binding Sites, Crystallography, X-Ray, Peptides metabolism, Protein Binding, Cell Cycle Proteins metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
The development of ligands for biological targets is critically dependent on the identification of sites on proteins that bind molecules with high affinity. A set of compounds, called FragLites, can identify such sites, along with the interactions required to gain affinity, by X-ray crystallography. We demonstrate the utility of FragLites in mapping the binding sites of bromodomain proteins BRD4 and ATAD2 and demonstrate that FragLite mapping is comparable to a full fragment screen in identifying ligand binding sites and key interactions. We extend the FragLite set with analogous compounds derived from amino acids (termed PepLites) that mimic the interactions of peptides. The output of the FragLite maps is shown to enable the development of ligands with leadlike potency. This work establishes the use of FragLite and PepLite screening at an early stage in ligand discovery allowing the rapid assessment of tractability of protein targets and informing downstream hit-finding.
- Published
- 2022
- Full Text
- View/download PDF
15. Exiting the tunnel of uncertainty: crystal soak to validated hit.
- Author
-
Martin MP and Noble MEM
- Subjects
- Ligands, Uncertainty, Retrospective Studies, Small Molecule Libraries chemistry, Drug Discovery methods
- Abstract
Crystallographic fragment screens provide an efficient and effective way to identify small-molecule ligands of a crystallized protein. Due to their low molecular weight, such hits tend to have low, often unquantifiable, affinity for their target, complicating the twin challenges of validating the hits as authentic solution-phase ligands of the target and identifying the `best' hit(s) for further elaboration. In this article, approaches that address these challenges are assessed. Using retrospective analysis of a recent ATAD2 hit-identification campaign, alongside other examples of successful fragment-screening campaigns, it is suggested that hit validation and prioritization are best achieved by a `triangulation' approach in which the results of multiple available biochemical and biophysical techniques are correlated to develop qualitative structure-activity relationships (SARs). Such qualitative SARs may indeed be the only means by which to navigate a project through the tunnel of uncertainty that prevails before on-scale biophysical, biochemical and/or biological measurements become possible., (open access.)
- Published
- 2022
- Full Text
- View/download PDF
16. Build-Couple-Transform: A Paradigm for Lead-like Library Synthesis with Scaffold Diversity.
- Author
-
Uguen M, Davison G, Sprenger LJ, Hunter JH, Martin MP, Turberville S, Watt JE, Golding BT, Noble MEM, Stewart HL, and Waring MJ
- Subjects
- High-Throughput Screening Assays, Small Molecule Libraries chemistry
- Abstract
High-throughput screening provides one of the most common ways of finding hit compounds. Lead-like libraries, in particular, provide hits with compatible functional groups and vectors for structural elaboration and physical properties suitable for optimization. Library synthesis approaches can lead to a lack of chemical diversity because they employ parallel derivatization of common building blocks using single reaction types. We address this problem through a "build-couple-transform" paradigm for the generation of lead-like libraries with scaffold diversity. Nineteen transformations of a 4-oxo-2-butenamide scaffold template were optimized, including 1,4-cyclizations, 3,4-cyclizations, reductions, and 1,4-additions. A pool-transformation approach efficiently explored the scope of these transformations for nine different building blocks and synthesized a >170-member library with enhanced chemical space coverage and favorable drug-like properties. Screening revealed hits against CDK2. This work establishes the build-couple-transform concept for the synthesis of lead-like libraries and provides a differentiated approach to libraries with significantly enhanced scaffold diversity.
- Published
- 2022
- Full Text
- View/download PDF
17. Parallel Optimization of Potency and Pharmacokinetics Leading to the Discovery of a Pyrrole Carboxamide ERK5 Kinase Domain Inhibitor.
- Author
-
Miller DC, Reuillon T, Molyneux L, Blackburn T, Cook SJ, Edwards N, Endicott JA, Golding BT, Griffin RJ, Hardcastle I, Harnor SJ, Heptinstall A, Lochhead P, Martin MP, Martin NC, Myers S, Newell DR, Noble RA, Phillips N, Rigoreau L, Thomas H, Tucker JA, Wang LZ, Waring MJ, Wong AC, Wedge SR, Noble MEM, and Cano C
- Subjects
- Cell Proliferation, Mitogen-Activated Protein Kinase 7, Pyrroles pharmacology
- Abstract
The nonclassical extracellular signal-related kinase 5 (ERK5) mitogen-activated protein kinase pathway has been implicated in increased cellular proliferation, migration, survival, and angiogenesis; hence, ERK5 inhibition may be an attractive approach for cancer treatment. However, the development of selective ERK5 inhibitors has been challenging. Previously, we described the development of a pyrrole carboxamide high-throughput screening hit into a selective, submicromolar inhibitor of ERK5 kinase activity. Improvement in the ERK5 potency was necessary for the identification of a tool ERK5 inhibitor for target validation studies. Herein, we describe the optimization of this series to identify nanomolar pyrrole carboxamide inhibitors of ERK5 incorporating a basic center, which suffered from poor oral bioavailability. Parallel optimization of potency and in vitro pharmacokinetic parameters led to the identification of a nonbasic pyrazole analogue with an optimal balance of ERK5 inhibition and oral exposure.
- Published
- 2022
- Full Text
- View/download PDF
18. An Alkynylpyrimidine-Based Covalent Inhibitor That Targets a Unique Cysteine in NF-κB-Inducing Kinase.
- Author
-
Al-Khawaldeh I, Al Yasiri MJ, Aldred GG, Basmadjian C, Bordoni C, Harnor SJ, Heptinstall AB, Hobson SJ, Jennings CE, Khalifa S, Lebraud H, Martin MP, Miller DC, Shrives HJ, de Souza JV, Stewart HL, Temple M, Thomas HD, Totobenazara J, Tucker JA, Tudhope SJ, Wang LZ, Bronowska AK, Cano C, Endicott JA, Golding BT, Hardcastle IR, Hickson I, Wedge SR, Willmore E, Noble MEM, and Waring MJ
- Subjects
- Alkynes chemical synthesis, Alkynes chemistry, Cysteine chemistry, Dose-Response Relationship, Drug, Humans, Molecular Structure, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases metabolism, Pyrimidines chemical synthesis, Pyrimidines chemistry, Structure-Activity Relationship, NF-kappaB-Inducing Kinase, Alkynes pharmacology, Cysteine pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
NF-κB-inducing kinase (NIK) is a key enzyme in the noncanonical NF-κB pathway, of interest in the treatment of a variety of diseases including cancer. Validation of NIK as a drug target requires potent and selective inhibitors. The protein contains a cysteine residue at position 444 in the back pocket of the active site, unique within the kinome. Analysis of existing inhibitor scaffolds and early structure-activity relationships (SARs) led to the design of C444-targeting covalent inhibitors based on alkynyl heterocycle warheads. Mass spectrometry provided proof of the covalent mechanism, and the SAR was rationalized by computational modeling. Profiling of more potent analogues in tumor cell lines with constitutively activated NIK signaling induced a weak antiproliferative effect, suggesting that kinase inhibition may have limited impact on cancer cell growth. This study shows that alkynyl heterocycles are potential cysteine traps, which may be employed where common Michael acceptors, such as acrylamides, are not tolerated.
- Published
- 2021
- Full Text
- View/download PDF
19. Structure-Based Design of Potent and Orally Active Isoindolinone Inhibitors of MDM2-p53 Protein-Protein Interaction.
- Author
-
Chessari G, Hardcastle IR, Ahn JS, Anil B, Anscombe E, Bawn RH, Bevan LD, Blackburn TJ, Buck I, Cano C, Carbain B, Castro J, Cons B, Cully SJ, Endicott JA, Fazal L, Golding BT, Griffin RJ, Haggerty K, Harnor SJ, Hearn K, Hobson S, Holvey RS, Howard S, Jennings CE, Johnson CN, Lunec J, Miller DC, Newell DR, Noble MEM, Reeks J, Revill CH, Riedinger C, St Denis JD, Tamanini E, Thomas H, Thompson NT, Vinković M, Wedge SR, Williams PA, Wilsher NE, Zhang B, and Zhao Y
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents metabolism, Bone Neoplasms drug therapy, Cell Line, Tumor, Cell Proliferation drug effects, Crystallography, X-Ray, Drug Stability, Female, Humans, Isoindoles chemical synthesis, Isoindoles metabolism, Macaca fascicularis, Male, Mice, Inbred BALB C, Mice, Nude, Microsomes, Liver metabolism, Molecular Structure, Protein Binding, Structure-Activity Relationship, Xenograft Model Antitumor Assays, Mice, Antineoplastic Agents pharmacology, Isoindoles pharmacology, Osteosarcoma drug therapy, Protein Multimerization drug effects, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Inhibition of murine double minute 2 (MDM2)-p53 protein-protein interaction with small molecules has been shown to reactivate p53 and inhibit tumor growth. Here, we describe rational, structure-guided, design of novel isoindolinone-based MDM2 inhibitors. MDM2 X-ray crystallography, quantum mechanics ligand-based design, and metabolite identification all contributed toward the discovery of potent in vitro and in vivo inhibitors of the MDM2-p53 interaction with representative compounds inducing cytostasis in an SJSA-1 osteosarcoma xenograft model following once-daily oral administration.
- Published
- 2021
- Full Text
- View/download PDF
20. Discriminative SKP2 Interactions with CDK-Cyclin Complexes Support a Cyclin A-Specific Role in p27KIP1 Degradation.
- Author
-
Salamina M, Montefiore BC, Liu M, Wood DJ, Heath R, Ault JR, Wang LZ, Korolchuk S, Baslé A, Pastok MW, Reeks J, Tatum NJ, Sobott F, Arold ST, Pagano M, Noble MEM, and Endicott JA
- Subjects
- Binding Sites, CDC2-CDC28 Kinases chemistry, CDC2-CDC28 Kinases genetics, CDC2-CDC28 Kinases metabolism, Cyclin A genetics, Cyclin A metabolism, Cyclin E chemistry, Cyclin E genetics, Cyclin E metabolism, Cyclin-Dependent Kinase 2 genetics, Cyclin-Dependent Kinase 2 metabolism, Cyclin-Dependent Kinase Inhibitor p27 genetics, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Gene Expression Regulation, HEK293 Cells, Humans, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Proteolysis, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, S-Phase Kinase-Associated Proteins genetics, S-Phase Kinase-Associated Proteins metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Signal Transduction, Cyclin A chemistry, Cyclin-Dependent Kinase 2 chemistry, Cyclin-Dependent Kinase Inhibitor p27 chemistry, G1 Phase Cell Cycle Checkpoints, S-Phase Kinase-Associated Proteins chemistry
- Abstract
The SCF
SKP2 ubiquitin ligase relieves G1 checkpoint control of CDK-cyclin complexes by promoting p27KIP1 degradation. We describe reconstitution of stable complexes containing SKP1-SKP2 and CDK1-cyclin B or CDK2-cyclin A/E, mediated by the CDK regulatory subunit CKS1. We further show that a direct interaction between a SKP2 N-terminal motif and cyclin A can stabilize SKP1-SKP2-CDK2-cyclin A complexes in the absence of CKS1. We identify the SKP2 binding site on cyclin A and demonstrate the site is not present in cyclin B or cyclin E. This site is distinct from but overlapping with features that mediate binding of p27KIP1 and other G1 cyclin regulators to cyclin A. We propose that the capacity of SKP2 to engage with CDK2-cyclin A by more than one structural mechanism provides a way to fine tune the degradation of p27KIP1 and distinguishes cyclin A from other G1 cyclins to ensure orderly cell cycle progression., (Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
21. Crystallographic and electrophilic fragment screening of the SARS-CoV-2 main protease.
- Author
-
Douangamath A, Fearon D, Gehrtz P, Krojer T, Lukacik P, Owen CD, Resnick E, Strain-Damerell C, Aimon A, Ábrányi-Balogh P, Brandão-Neto J, Carbery A, Davison G, Dias A, Downes TD, Dunnett L, Fairhead M, Firth JD, Jones SP, Keeley A, Keserü GM, Klein HF, Martin MP, Noble MEM, O'Brien P, Powell A, Reddi RN, Skyner R, Snee M, Waring MJ, Wild C, London N, von Delft F, and Walsh MA
- Subjects
- Betacoronavirus enzymology, Binding Sites, Catalytic Domain, Coronavirus 3C Proteases, Crystallography, X-Ray, Cysteine Endopeptidases metabolism, Drug Design, Mass Spectrometry, Models, Molecular, Peptide Fragments metabolism, Protein Conformation, SARS-CoV-2, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Static Electricity, Viral Nonstructural Proteins metabolism, Betacoronavirus chemistry, Cysteine Endopeptidases chemistry, Peptide Fragments chemistry, Viral Nonstructural Proteins chemistry
- Abstract
COVID-19, caused by SARS-CoV-2, lacks effective therapeutics. Additionally, no antiviral drugs or vaccines were developed against the closely related coronavirus, SARS-CoV-1 or MERS-CoV, despite previous zoonotic outbreaks. To identify starting points for such therapeutics, we performed a large-scale screen of electrophile and non-covalent fragments through a combined mass spectrometry and X-ray approach against the SARS-CoV-2 main protease, one of two cysteine viral proteases essential for viral replication. Our crystallographic screen identified 71 hits that span the entire active site, as well as 3 hits at the dimer interface. These structures reveal routes to rapidly develop more potent inhibitors through merging of covalent and non-covalent fragment hits; one series of low-reactivity, tractable covalent fragments were progressed to discover improved binders. These combined hits offer unprecedented structural and reactivity information for on-going structure-based drug design against SARS-CoV-2 main protease.
- Published
- 2020
- Full Text
- View/download PDF
22. Paradoxical activation of the protein kinase-transcription factor ERK5 by ERK5 kinase inhibitors.
- Author
-
Lochhead PA, Tucker JA, Tatum NJ, Wang J, Oxley D, Kidger AM, Johnson VP, Cassidy MA, Gray NS, Noble MEM, and Cook SJ
- Subjects
- Gene Expression Regulation, HEK293 Cells, HeLa Cells, Humans, Inflammation metabolism, Mitogen-Activated Protein Kinase 7 genetics, Models, Molecular, Mutation, Protein Conformation, Protein Domains, Protein Kinase Inhibitors pharmacology, Transcription, Genetic, Mitogen-Activated Protein Kinase 7 metabolism, Protein Kinase Inhibitors metabolism, Transcription Factors metabolism
- Abstract
The dual protein kinase-transcription factor, ERK5, is an emerging drug target in cancer and inflammation, and small-molecule ERK5 kinase inhibitors have been developed. However, selective ERK5 kinase inhibitors fail to recapitulate ERK5 genetic ablation phenotypes, suggesting kinase-independent functions for ERK5. Here we show that ERK5 kinase inhibitors cause paradoxical activation of ERK5 transcriptional activity mediated through its unique C-terminal transcriptional activation domain (TAD). Using the ERK5 kinase inhibitor, Compound 26 (ERK5-IN-1), as a paradigm, we have developed kinase-active, drug-resistant mutants of ERK5. With these mutants, we show that induction of ERK5 transcriptional activity requires direct binding of the inhibitor to the kinase domain. This in turn promotes conformational changes in the kinase domain that result in nuclear translocation of ERK5 and stimulation of gene transcription. This shows that both the ERK5 kinase and TAD must be considered when assessing the role of ERK5 and the effectiveness of anti-ERK5 therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
23. Identification of a novel orally bioavailable ERK5 inhibitor with selectivity over p38α and BRD4.
- Author
-
Myers SM, Miller DC, Molyneux L, Arasta M, Bawn RH, Blackburn TJ, Cook SJ, Edwards N, Endicott JA, Golding BT, Griffin RJ, Hammonds T, Hardcastle IR, Harnor SJ, Heptinstall AB, Lochhead PA, Martin MP, Martin NC, Newell DR, Owen PJ, Pang LC, Reuillon T, Rigoreau LJM, Thomas HD, Tucker JA, Wang LZ, Wong AC, Noble MEM, Wedge SR, and Cano C
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Biological Availability, Cell Cycle Proteins, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Female, Humans, Mice, Mice, Nude, Mitogen-Activated Protein Kinase 14 metabolism, Mitogen-Activated Protein Kinase 7 metabolism, Molecular Structure, Neoplasms, Experimental drug therapy, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Nuclear Proteins metabolism, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors chemistry, Structure-Activity Relationship, Transcription Factors metabolism, Antineoplastic Agents pharmacology, Mitogen-Activated Protein Kinase 14 antagonists & inhibitors, Mitogen-Activated Protein Kinase 7 antagonists & inhibitors, Nuclear Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Transcription Factors antagonists & inhibitors
- Abstract
Extracellular regulated kinase 5 (ERK5) signalling has been implicated in driving a number of cellular phenotypes including endothelial cell angiogenesis and tumour cell motility. Novel ERK5 inhibitors were identified using high throughput screening, with a series of pyrrole-2-carboxamides substituted at the 4-position with an aroyl group being found to exhibit IC
50 values in the micromolar range, but having no selectivity against p38α MAP kinase. Truncation of the N-substituent marginally enhanced potency (∼3-fold) against ERK5, but importantly attenuated inhibition of p38α. Systematic variation of the substituents on the aroyl group led to the selective inhibitor 4-(2-bromo-6-fluorobenzoyl)-N-(pyridin-3-yl)-1H-pyrrole-2-carboxamide (IC50 0.82 μM for ERK5; IC50 > 120 μM for p38α). The crystal structure (PDB 5O7I) of this compound in complex with ERK5 has been solved. This compound was orally bioavailable and inhibited bFGF-driven Matrigel plug angiogenesis and tumour xenograft growth. The selective ERK5 inhibitor described herein provides a lead for further development into a tool compound for more extensive studies seeking to examine the role of ERK5 signalling in cancer and other diseases., (Copyright © 2019 Elsevier Masson SAS. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
24. FragLites-Minimal, Halogenated Fragments Displaying Pharmacophore Doublets. An Efficient Approach to Druggability Assessment and Hit Generation.
- Author
-
Wood DJ, Lopez-Fernandez JD, Knight LE, Al-Khawaldeh I, Gai C, Lin S, Martin MP, Miller DC, Cano C, Endicott JA, Hardcastle IR, Noble MEM, and Waring MJ
- Subjects
- Binding Sites, Crystallography, X-Ray, Drug Discovery methods, Drug Evaluation, Preclinical, Ligands, Small Molecule Libraries chemistry, Halogenation
- Abstract
Identifying ligand binding sites on proteins is a critical step in target-based drug discovery. Current approaches to this require resource-intensive screening of large libraries of lead-like or fragment molecules. Here, we describe an efficient and effective experimental approach to mapping interaction sites using a set of halogenated compounds expressing paired hydrogen-bonding motifs, termed FragLites. The FragLites identify productive drug-like interactions, which are identified sensitively and unambiguously by X-ray crystallography, exploiting the anomalous scattering of the halogen substituent. This mapping of protein interaction surfaces provides an assessment of druggability and can identify efficient start points for the de novo design of hit molecules incorporating the interacting motifs. The approach is illustrated by mapping cyclin-dependent kinase 2, which successfully identifies orthosteric and allosteric sites. The hits were rapidly elaborated to develop efficient lead-like molecules. Hence, the approach provides a new method of identifying ligand sites, assessing tractability and discovering new leads.
- Published
- 2019
- Full Text
- View/download PDF
25. Differences in the Conformational Energy Landscape of CDK1 and CDK2 Suggest a Mechanism for Achieving Selective CDK Inhibition.
- Author
-
Wood DJ, Korolchuk S, Tatum NJ, Wang LZ, Endicott JA, Noble MEM, and Martin MP
- Subjects
- CDC2 Protein Kinase isolation & purification, CDC2 Protein Kinase metabolism, Cyclin-Dependent Kinase 2 isolation & purification, Cyclin-Dependent Kinase 2 metabolism, Humans, Molecular Conformation, Molecular Dynamics Simulation, Protein Kinase Inhibitors chemistry, Surface Plasmon Resonance, CDC2 Protein Kinase antagonists & inhibitors, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Entropy, Protein Kinase Inhibitors pharmacology
- Abstract
Dysregulation of the cell cycle characterizes many cancer subtypes, providing a rationale for developing cyclin-dependent kinase (CDK) inhibitors. Potent CDK2 inhibitors might target certain cancers in which CCNE1 is amplified. However, current CDK2 inhibitors also inhibit CDK1, generating a toxicity liability. We have used biophysical measurements and X-ray crystallography to investigate the ATP-competitive inhibitor binding properties of cyclin-free and cyclin-bound CDK1 and CDK2. We show that these kinases can readily be distinguished by such inhibitors when cyclin-free, but not when cyclin-bound. The basis for this discrimination is unclear from either inspection or molecular dynamics simulation of ligand-bound CDKs, but is reflected in the contacts made between the kinase N- and C-lobes. We conclude that there is a subtle but profound difference between the conformational energy landscapes of cyclin-free CDK1 and CDK2. The unusual properties of CDK1 might be exploited to differentiate CDK1 from other CDKs in future cancer therapeutic design., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Tuning the Binding Affinity and Selectivity of Perfluoroaryl-Stapled Peptides by Cysteine-Editing.
- Author
-
Verhoork SJM, Jennings CE, Rozatian N, Reeks J, Meng J, Corlett EK, Bunglawala F, Noble MEM, Leach AG, and Coxon CR
- Subjects
- Amino Acid Sequence, Cysteine metabolism, Humans, Peptides chemical synthesis, Peptides metabolism, Protein Binding, Protein Conformation, Proto-Oncogene Proteins c-mdm2 chemistry, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Tumor Suppressor Protein p53 chemistry, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cysteine chemistry, Fluorocarbons chemistry, Peptides chemistry
- Abstract
A growing number of approaches to "staple" α-helical peptides into a bioactive conformation using cysteine cross-linking are emerging. Here, the replacement of l-cysteine with "cysteine analogues" in combinations of different stereochemistry, side chain length and beta-carbon substitution, is explored to examine the influence that the thiol-containing residue(s) has on target protein binding affinity in a well-explored model system, p53-MDM2/MDMX, which is constituted by the interaction of the tumour suppressor protein p53 and proteins MDM2 and MDMX, which regulate p53 activity. In some cases, replacement of one or more l-cysteine residues afforded significant changes in the measured binding affinity and target selectivity of the peptide. Computationally constructed homology models indicate that some modifications, such as incorporating two d-cysteine residues, favourably alter the positions of key functional amino acid side chains, which is likely to cause changes in binding affinity, in agreement with measured surface plasmon resonance data., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
27. Identification of a novel ligand for the ATAD2 bromodomain with selectivity over BRD4 through a fragment growing approach.
- Author
-
Miller DC, Martin MP, Adhikari S, Brennan A, Endicott JA, Golding BT, Hardcastle IR, Heptinstall A, Hobson S, Jennings C, Molyneux L, Ng Y, Wedge SR, Noble MEM, and Cano C
- Subjects
- ATPases Associated with Diverse Cellular Activities chemistry, Cell Cycle Proteins, Computer-Aided Design, DNA-Binding Proteins chemistry, Humans, Ligands, Molecular Docking Simulation, Neoplasms drug therapy, Neoplasms metabolism, Nuclear Proteins chemistry, Protein Binding, Protein Domains drug effects, Proto-Oncogene Mas, Transcription Factors chemistry, ATPases Associated with Diverse Cellular Activities antagonists & inhibitors, ATPases Associated with Diverse Cellular Activities metabolism, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins metabolism, Drug Design, Nuclear Proteins metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Transcription Factors metabolism
- Abstract
ATAD2 is an ATPase that is overexpressed in a variety of cancers and associated with a poor patient prognosis. This protein has been suggested to function as a cofactor for a range of transcription factors, including the proto-oncogene MYC and the androgen receptor. ATAD2 comprises an ATPase domain, implicated in chromatin remodelling, and a bromodomain which allows it to interact with acetylated histone tails. Dissection of the functional roles of these two domains would benefit from the availability of selective, cell-permeable pharmacological probes. An in silico evaluation of the 3D structures of various bromodomains suggested that developing small molecule ligands for the bromodomain of ATAD2 is likely to be challenging, although recent reports have shown that ATAD2 bromodomain ligands can be identified. We report a structure-guided fragment-based approach to identify lead compounds for ATAD2 bromodomain inhibitor development. Our findings indicate that the ATAD2 bromodomain can accommodate fragment hits (M
r < 200) that yield productive structure-activity relationships, and structure-guided design enabled the introduction of selectivity over BRD4.- Published
- 2018
- Full Text
- View/download PDF
28. Structure-based discovery of cyclin-dependent protein kinase inhibitors.
- Author
-
Martin MP, Endicott JA, and Noble MEM
- Subjects
- Antineoplastic Agents chemical synthesis, Catalytic Domain, Cyclin-Dependent Kinases chemistry, Cyclin-Dependent Kinases metabolism, Humans, Molecular Targeted Therapy, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism, Neoplasms drug therapy, Neoplasms enzymology, Neoplasms genetics, Neoplasms pathology, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Protein Kinase Inhibitors chemical synthesis, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Cyclin-Dependent Kinases antagonists & inhibitors, Drug Discovery methods, Molecular Docking Simulation, Neoplasm Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
The cell fate-determining roles played by members of the cyclin-dependent protein kinase (CDK) family explain why their dysregulation can promote proliferative diseases, and identify them as potential targets for drug discovery in oncology and beyond. After many years of research, the first efficacious CDK inhibitors have now been registered for clinical use in a defined segment of breast cancer. Research is underway to identify inhibitors with appropriate CDK-inhibitory profiles to recapitulate this success in other disease settings. Here, we review the structural data that illustrate the interactions and properties that confer upon inhibitors affinity and/or selectivity toward different CDK family members. We conclude that where CDK inhibitors display selectivity, that selectivity derives from exploiting active site sequence peculiarities and/or from the capacity of the target CDK(s) to access conformations compatible with optimizing inhibitor-target interactions., (© 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2017
- Full Text
- View/download PDF
29. Differential Regulation of G1 CDK Complexes by the Hsp90-Cdc37 Chaperone System.
- Author
-
Hallett ST, Pastok MW, Morgan RML, Wittner A, Blundell KLIM, Felletar I, Wedge SR, Prodromou C, Noble MEM, Pearl LH, and Endicott JA
- Subjects
- Adenosine Triphosphate chemistry, Adenosine Triphosphate metabolism, Aminopyridines chemistry, Aminopyridines metabolism, Benzimidazoles metabolism, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins genetics, Chaperonins antagonists & inhibitors, Chaperonins genetics, Cyclin D metabolism, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Fluorescence Resonance Energy Transfer, HSP90 Heat-Shock Proteins genetics, Humans, Inhibitory Concentration 50, Kinetics, Piperazines chemistry, Piperazines metabolism, Protein Binding, Purines chemistry, Purines metabolism, Pyridines chemistry, Pyridines metabolism, Surface Plasmon Resonance, Cell Cycle Proteins metabolism, Chaperonins metabolism, Cyclin-Dependent Kinase 4 metabolism, Cyclin-Dependent Kinase 6 metabolism, HSP90 Heat-Shock Proteins metabolism
- Abstract
Selective recruitment of protein kinases to the Hsp90 system is mediated by the adaptor co-chaperone Cdc37. We show that assembly of CDK4 and CDK6 into protein complexes is differentially regulated by the Cdc37-Hsp90 system. Like other Hsp90 kinase clients, binding of CDK4/6 to Cdc37 is blocked by ATP-competitive inhibitors. Cdc37-Hsp90 relinquishes CDK6 to D3- and virus-type cyclins and to INK family CDK inhibitors, whereas CDK4 is relinquished to INKs but less readily to cyclins. p21CIP1 and p27KIP1 CDK inhibitors are less potent than the INKs at displacing CDK4 and CDK6 from Cdc37. However, they cooperate with the D-type cyclins to generate CDK4/6-containing ternary complexes that are resistant to cyclin D displacement by Cdc37, suggesting a molecular mechanism to explain the assembly factor activity ascribed to CIP/KIP family members. Overall, our data reveal multiple mechanisms whereby the Hsp90 system may control formation of CDK4- and CDK6-cyclin complexes under different cellular conditions., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Validating and enabling phosphoglycerate dehydrogenase (PHGDH) as a target for fragment-based drug discovery in PHGDH-amplified breast cancer.
- Author
-
Unterlass JE, Baslé A, Blackburn TJ, Tucker J, Cano C, Noble MEM, and Curtin NJ
- Abstract
3-Phosphoglycerate dehydrogenase (PHGDH) has recently been identified as an attractive target in cancer therapy as it links upregulated glycolytic flux to increased biomass production in cancer cells. PHGDH catalyses the first step in the serine synthesis pathway and thus diverts glycolytic flux into serine synthesis. We have used siRNA-mediated suppression of PHGDH expression to show that PHGDH is a potential therapeutic target in PHGDH -amplified breast cancer. Knockdown caused reduced proliferation in the PHGDH -amplified cell line MDA-MB-468, whereas breast cancer cells with low PHGDH expression or with elevated PHGDH expression in the absence of genomic amplification were not affected. As a first step towards design of a chemical probe for PHGDH, we report a fragment-based drug discovery approach for the identification of PHGDH inhibitors. We designed a truncated PHGDH construct that gave crystals which diffracted to high resolution, and could be used for fragment soaking. 15 fragments stabilising PHGDH were identified using a thermal shift assay and validated by X-ray crystallography and ITC competition experiments to exhibit 1.5-26.2 mM affinity for PHGDH. A structure-guided fragment growing approach was applied to the PHGDH binders from the initial screen, yielding greater understanding of the binding site and suggesting routes to achieve higher affinity NAD-competitive inhibitors., Competing Interests: CONFLICTS OF INTEREST None.
- Published
- 2016
- Full Text
- View/download PDF
31. CDK1 structures reveal conserved and unique features of the essential cell cycle CDK.
- Author
-
Brown NR, Korolchuk S, Martin MP, Stanley WA, Moukhametzianov R, Noble MEM, and Endicott JA
- Subjects
- Adenosine Triphosphate chemistry, Animals, Binding, Competitive, CDC2 Protein Kinase, CDC2-CDC28 Kinases genetics, Carrier Proteins genetics, Cattle, Cell Cycle Proteins genetics, Conserved Sequence, Crystallography, X-Ray, Cyclin A chemistry, Cyclin A genetics, Cyclin B genetics, Cyclin-Dependent Kinase 2 chemistry, Cyclin-Dependent Kinase 2 genetics, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases genetics, Gene Expression, Humans, Kinetics, Models, Molecular, Peptides chemical synthesis, Peptides chemistry, Protein Binding, Protein Kinase Inhibitors chemistry, Protein Stability, Protein Structure, Secondary, Protein Structure, Tertiary, Recombinant Fusion Proteins genetics, Substrate Specificity, CDC2-CDC28 Kinases chemistry, Carrier Proteins chemistry, Cell Cycle Proteins chemistry, Cyclin B chemistry, Cyclin-Dependent Kinases chemistry, Recombinant Fusion Proteins chemistry
- Abstract
CDK1 is the only essential cell cycle CDK in human cells and is required for successful completion of M-phase. It is the founding member of the CDK family and is conserved across all eukaryotes. Here we report the crystal structures of complexes of CDK1-Cks1 and CDK1-cyclin B-Cks2. These structures confirm the conserved nature of the inactive monomeric CDK fold and its ability to be remodelled by cyclin binding. Relative to CDK2-cyclin A, CDK1-cyclin B is less thermally stable, has a smaller interfacial surface, is more susceptible to activation segment dephosphorylation and shows differences in the substrate sequence features that determine activity. Both CDK1 and CDK2 are potential cancer targets for which selective compounds are required. We also describe the first structure of CDK1 bound to a potent ATP-competitive inhibitor and identify aspects of CDK1 structure and plasticity that might be exploited to develop CDK1-selective inhibitors.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.