147 results on '"McDonough MA"'
Search Results
2. An LNA-amide modification that enhances the cell uptake and activity of phosphorothioate exon-skipping oligonucleotides
- Author
-
Baker, YR, Thorpe, C, Chen, J, Poller, LM, Cox, L, Kumar, P, Lim, WF, Lie, L, McClorey, G, Epple, S, Singleton, D, McDonough, MA, Hardwick, JS, Christensen, KE, Wood, MJA, Hall, JP, El-Sagheer, AH, and Brown, T
- Subjects
Multidisciplinary ,General Physics and Astronomy ,Phosphorothioate Oligonucleotides ,RNA ,ddc:500 ,General Chemistry ,Exons ,Oligonucleotides, Antisense ,Amides ,General Biochemistry, Genetics and Molecular Biology - Abstract
Nature Communications 13(1), 4036 (2022). doi:10.1038/s41467-022-31636-2, Oligonucleotides that target mRNA have great promise as therapeutic agents for life-threatening conditions but suffer from poor bioavailability, hence high cost. As currently untreatable diseases come within the reach of oligonucleotide therapies, new analogues are urgently needed to address this. With this in mind we describe reduced-charge oligonucleotides containing artificial LNA-amide linkages with improved gymnotic cell uptake, RNA affinity, stability and potency. To construct such oligonucleotides, five LNA-amide monomers (A, T, C, 5mC and G), where the 3′-OH is replaced by an ethanoic acid group, are synthesised in good yield and used in solid-phase oligonucleotide synthesis to form amide linkages with high efficiency. The artificial backbone causes minimal structural deviation to the DNA:RNA duplex. These studies indicate that splice-switching oligonucleotides containing LNA-amide linkages and phosphorothioates display improved activity relative to oligonucleotides lacking amides, highlighting the therapeutic potential of this technology., Published by Nature Publishing Group UK, [London]
- Published
- 2022
- Full Text
- View/download PDF
3. Structure-Based Design of Selective Fat Mass and Obesity Associated Protein (FTO) Inhibitors
- Author
-
Shishodia, S, Demetriades, M, Zhang, D, Tam, NY, Maheswaran, P, Clunie-O'Connor, C, Tumber, A, Leung, IKH, Ng, YM, Leissing, TM, El-Sagheer, AH, Salah, E, Brown, T, Aik, WS, McDonough, MA, Schofield, CJ, Shishodia, S, Demetriades, M, Zhang, D, Tam, NY, Maheswaran, P, Clunie-O'Connor, C, Tumber, A, Leung, IKH, Ng, YM, Leissing, TM, El-Sagheer, AH, Salah, E, Brown, T, Aik, WS, McDonough, MA, and Schofield, CJ
- Abstract
FTO catalyzes the Fe(II) and 2-oxoglutarate (2OG)-dependent modification of nucleic acids, including the demethylation of N6-methyladenosine (m6A) in mRNA. FTO is a proposed target for anti-cancer therapy. Using information from crystal structures of FTO in complex with 2OG and substrate mimics, we designed and synthesized two series of FTO inhibitors, which were characterized by turnover and binding assays, and by X-ray crystallography with FTO and the related bacterial enzyme AlkB. A potent inhibitor employing binding interactions spanning the FTO 2OG and substrate binding sites was identified. Selectivity over other clinically targeted 2OG oxygenases was demonstrated, including with respect to the hypoxia-inducible factor prolyl and asparaginyl hydroxylases (PHD2 and FIH) and selected JmjC histone demethylases (KDMs). The results illustrate how structure-based design can enable the identification of potent and selective 2OG oxygenase inhibitors and will be useful for the development of FTO inhibitors for use in vivo.
- Published
- 2021
4. Broad Spectrum β-Lactamase Inhibition by a Thioether Substituted Bicyclic Boronate
- Author
-
Parkova, A, Lucic, A, Krajnc, A, Brem, J, Calvopiña, K, Langley, GW, McDonough, MA, Trapencieris, P, and Schofield, CJ
- Subjects
0303 health sciences ,03 medical and health sciences ,Infectious Diseases ,030306 microbiology ,Sulfides ,beta-Lactamase Inhibitors ,beta-Lactamases ,030304 developmental biology ,Anti-Bacterial Agents - Abstract
β-Lactamases comprise the most widely used mode of resistance to β-lactam antibiotics. Cyclic boronates have shown promise as a new class of β-lactamase inhibitor, with pioneering potential to potently inhibit both metallo- and serine-β-lactamases. We report studies concerning a bicyclic boronate ester with a thioether rather than the more typical β-lactam antibiotic “C-6/C-7” acylamino type side chain, which is present in the penicillin/cephalosporin antibiotics. The thioether bicyclic boronate ester was tested for activity against representative serine- and metallo-β-lactamases. The results support the broad inhibition potential of bicyclic boronate based inhibitors with different side chains, including against metallo-β-lactamases from B1, B2, and B3 subclasses. Combined with previous crystallographic studies, analysis of a crystal structure of the thioether inhibitor with the clinically relevant VIM-2 metallo-β-lactamase implies that further SAR work will expand the already broad scope of β-lactamase inhibition by bicyclic boronates.
- Published
- 2019
5. Biochemical and structural investigations clarify the substrate selectivity of the 2-oxoglutarate oxygenase JMJD6
- Author
-
Islam, MS, McDonough, MA, Chowdhury, R, Gault, J, Khan, A, Pires, E, Schofield, CJ, Islam, Md Saiful [0000-0001-9842-0676], McDonough, Michael A [0000-0003-4664-6942], Schofield, Christopher J [0000-0002-0290-6565], and Apollo - University of Cambridge Repository
- Subjects
Jumonji Domain-Containing Histone Demethylases ,RNA splicing ,Protein Conformation ,hypoxia ,hydroxylase ,metalloenzyme ,substrate specificity ,Lysine ,Estrogen Receptor alpha ,2-oxoglutarate and iron dependent dioxygenase ,Crystallography, X-Ray ,Hydroxylation ,enzyme catalysis ,C-5 hydroxylysine ,Catalysis ,enzyme structure ,Kinetics ,JMJD6 ,Enzymology ,JmjC domain-containing protein 6 ,Humans ,dioxygenase ,hydroxylysine (Hyl) ,X-ray crystallography - Abstract
JmjC domain-containing protein 6 (JMJD6) is a 2-oxoglutarate (2OG)-dependent oxygenase linked to various cellular processes, including splicing regulation, histone modification, transcriptional pause release, hypoxia sensing, and cancer. JMJD6 is reported to catalyze hydroxylation of lysine residue(s) of histones, the tumor-suppressor protein p53, and splicing regulatory proteins, including u2 small nuclear ribonucleoprotein auxiliary factor 65-kDa subunit (U2AF65). JMJD6 is also reported to catalyze N-demethylation of N-methylated (both mono- and di-methylated) arginine residues of histones and other proteins, including HSP70 (heat-shock protein 70), estrogen receptor α, and RNA helicase A. Here, we report MS- and NMR-based kinetic assays employing purified JMJD6 and multiple substrate fragment sequences, the results of which support the assignment of purified JMJD6 as a lysyl hydroxylase. By contrast, we did not observe N-methyl arginyl N-demethylation with purified JMJD6. Biophysical analyses, including crystallographic analyses of JMJD6Δ344-403 in complex with iron and 2OG, supported its assignment as a lysyl hydroxylase rather than an N-methyl arginyl-demethylase. The screening results supported some, but not all, of the assigned JMJD6 substrates and identified other potential JMJD6 substrates. We envision these results will be useful in cellular and biological work on the substrates and functions of JMJD6 and in the development of selective inhibitors of human 2OG oxygenases.
- Published
- 2019
6. Structure activity relationship studies on rhodanines and derived enethiol inhibitors of metallo-β-lactamases
- Author
-
Zhang, D, Markoulides, MS, Stepanovs, D, Rydzik, AM, El-Hussein, A, Bon, C, Kamps, JJAG, Umland, K-D, Collins, PM, Cahill, ST, Wang, DY, von Delft, F, Brem, J, McDonough, MA, and Schofield, CJ
- Subjects
Molecular Structure ,Rhodanine ,Antibiotic resistance ,Inhibitors ,Metallo β-lactamase ,Article ,beta-Lactamases ,Carbapenemase ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Sulfhydryl Compounds ,Enediynes ,beta-Lactamase Inhibitors ,ComputingMethodologies_COMPUTERGRAPHICS ,Structure activity relationships - Abstract
Graphical abstract, Metallo-β-lactamases (MBLs) enable bacterial resistance to almost all classes of β-lactam antibiotics. We report studies on enethiol containing MBL inhibitors, which were prepared by rhodanine hydrolysis. The enethiols inhibit MBLs from different subclasses. Crystallographic analyses reveal that the enethiol sulphur displaces the di-Zn(II) ion bridging ‘hydrolytic’ water. In some, but not all, cases biophysical analyses provide evidence that rhodanine/enethiol inhibition involves formation of a ternary MBL enethiol rhodanine complex. The results demonstrate how low molecular weight active site Zn(II) chelating compounds can inhibit a range of clinically relevant MBLs and provide additional evidence for the potential of rhodanines to be hydrolysed to potent inhibitors of MBL protein fold and, maybe, other metallo-enzymes, perhaps contributing to the complex biological effects of rhodanines. The results imply that any medicinal chemistry studies employing rhodanines (and related scaffolds) as inhibitors should as a matter of course include testing of their hydrolysis products.
- Published
- 2018
7. Cyclic Boronates Inhibit All Classes of β-Lactamase
- Author
-
Cahill, ST, Cain, R, Wang, DY, Lohans, CT, Wareham, DW, Oswin, HP, Mohammed, J, Spencer, J, Fishwick, CWG, McDonough, MA, Schofield, CJ, and Brem, J
- Subjects
polycyclic compounds ,biochemical phenomena, metabolism, and nutrition - Abstract
β-Lactamase-mediated resistance is a growing threat to the continued use of β-lactam antibiotics. The use of the β-lactam-based serine-β-lactamase (SBL) inhibitors clavulanic acid, sulbactam, tazobactam, and, more recently, the non-β-lactam inhibitor avibactam has extended the utility of β-lactams against bacterial infections demonstrating resistance via these enzymes. These molecules are, however, ineffective against the metallo-β-lactamases (MBLs), which catalyse their hydrolysis. To date, there are no clinically available metallo-β-lactamase inhibitors. Co-production of MBLs and SBLs in resistant infections is, thus, of major clinical concern. The development of 'dual-action' inhibitors, targeting both SBLs and MBLs, is of interest, but these are considered difficult to achieve due to the structural and mechanistic differences between the two enzyme classes. We recently reported evidence that cyclic boronates can inhibit both serine- and metallo-β-lactmases. Here we report that cyclic boronates are able to inhibit all four classes of β-lactamase, including the class A extended spectrum β-lactamase, CTX-M-15, the class C enzyme, AmpC from Pseudomonas aeruginosa, and class D OXA enzymes with carbapenem-hydrolysing capabilities. We demonstrate that cyclic boronates can potentiate the use of β-lactams against Gram-negative clinical isolates expressing a variety of β-lactamases. Comparison of a crystal structure of a CTX-M-15:cyclic boronate complex with structures of cyclic boronates complexed with other β-lactamases reveals remarkable conservation of the small molecule binding mode, supporting our proposal that these molecules work by mimicking the common tetrahedral anionic intermediate present in both serine- and metallo-β-lactamase catalysis.
- Published
- 2017
8. Structural basis of metallo-β-lactamase, serine-β-lactamase and penicillin-binding protein inhibition by cyclic boronates
- Author
-
Brem, J, Cain, R, Cahill, S, McDonough, MA, Clifton, IJ, Jimenez-Castellanos, JC, Avison, MB, Spencer, J, Fishwick, CWG, and Schofield, CJ
- Subjects
inorganic chemicals ,Science ,Drug Evaluation, Preclinical ,Microbial Sensitivity Tests ,biochemical phenomena, metabolism, and nutrition ,Boronic Acids ,Article ,beta-Lactamases ,Enzymes ,Structure-Activity Relationship ,HEK293 Cells ,Cyclization ,polycyclic compounds ,Serine ,Humans ,Penicillin-Binding Proteins ,Enzyme Inhibitors ,Structural biology - Abstract
β-Lactamases enable resistance to almost all β-lactam antibiotics. Pioneering work revealed that acyclic boronic acids can act as ‘transition state analogue' inhibitors of nucleophilic serine enzymes, including serine-β-lactamases. Here we report biochemical and biophysical analyses revealing that cyclic boronates potently inhibit both nucleophilic serine and zinc-dependent β-lactamases by a mechanism involving mimicking of the common tetrahedral intermediate. Cyclic boronates also potently inhibit the non-essential penicillin-binding protein PBP 5 by the same mechanism of action. The results open the way for development of dual action inhibitors effective against both serine- and metallo-β-lactamases, and which could also have antimicrobial activity through inhibition of PBPs., Bacterial beta-lactamases are responsible for resistance to beta-lactams, the most important family of antibiotics. Here, the authors reveal cyclic boronate inhibitors that are active against both serine- and metallo-beta-lactamases and represent a promising strategy for combined antimicrobial treatments.
- Published
- 2016
- Full Text
- View/download PDF
9. Structure of human RNA N⁶-methyladenine demethylase ALKBH5 provides insights into its mechanisms of nucleic acid recognition and demethylation
- Author
-
Aik, W, Scotti, JS, Choi, H, Gong, L, Demetriades, M, Schofield, CJ, and McDonough, MA
- Abstract
ALKBH5 is a 2-oxoglutarate (2OG) and ferrous iron-dependent nucleic acid oxygenase (NAOX) that catalyzes the demethylation of N(6)-methyladenine in RNA. ALKBH5 is upregulated under hypoxia and plays a role in spermatogenesis. We describe a crystal structure of human ALKBH5 (residues 66-292) to 2.0 Å resolution. ALKBH5₆₆₋₂₉₂ has a double-stranded β-helix core fold as observed in other 2OG and iron-dependent oxygenase family members. The active site metal is octahedrally coordinated by an HXD…H motif (comprising residues His204, Asp206 and His266) and three water molecules. ALKBH5 shares a nucleotide recognition lid and conserved active site residues with other NAOXs. A large loop (βIV-V) in ALKBH5 occupies a similar region as the L1 loop of the fat mass and obesity-associated protein that is proposed to confer single-stranded RNA selectivity. Unexpectedly, a small molecule inhibitor, IOX3, was observed covalently attached to the side chain of Cys200 located outside of the active site. Modelling substrate into the active site based on other NAOX-nucleic acid complexes reveals conserved residues important for recognition and demethylation mechanisms. The structural insights will aid in the development of inhibitors selective for NAOXs, for use as functional probes and for therapeutic benefit.
- Published
- 2016
- Full Text
- View/download PDF
10. OS-9: another piece in the HIF complex story
- Author
-
Flashman, E, McDonough, MA, and Schofield, CJ
- Subjects
Cell Biology ,Molecular Biology - Abstract
In this issue of Molecular Cell, the Semenza group reports that OS-9, a common protein of unassigned function, promotes the O2-dependent degradation of hypoxia inducible factor (HIF) via binding to both HIF and the HIF prolyl-hydroxylases, implying that OS-9 is part of a multiprotein complex involved in the hypoxic response (Baek et al., 2005).
- Published
- 2016
11. Crystallographic and mass spectrometric analyses of a tandem GNAT protein from the clavulanic acid biosynthesis pathway
- Author
-
Iqbal, A, Arunlanantham, H, Brown, T, Chowdhury, R, Clifton, IJ, Kershaw, NJ, Hewitson, KS, McDonough, MA, and Schofield, CJ
- Abstract
(3R,5R)-Clavulanic acid (CA) is a clinically important inhibitor of Class A beta-lactamases. Sequence comparisons suggest that orf14 of the clavulanic acid biosynthesis gene cluster encodes for an acetyl transferase (CBG). Crystallographic studies reveal CBG to be a member of the emerging structural subfamily of tandem Gcn5-related acetyl transferase (GNAT) proteins. Two crystal forms (C2 and P2(1) space groups) of CBG were obtained; in both forms one molecule of acetyl-CoA (AcCoA) was bound to the N-terminal GNAT domain, with the C-terminal domain being unoccupied by a ligand. Mass spectrometric analyzes on CBG demonstrate that, in addition to one strongly bound AcCoA molecule, a second acyl-CoA molecule can bind to CBG. Succinyl-CoA and myristoyl-CoA displayed the strongest binding to the "second" CoA binding site, which is likely in the C-terminal GNAT domain. Analysis of the CBG structures, together with those of other tandem GNAT proteins, suggest that the AcCoA in the N-terminal GNAT domain plays a structural role whereas the C-terminal domain is more likely to be directly involved in acetyl transfer. The available crystallographic and mass spectrometric evidence suggests that binding of the second acyl-CoA occurs preferentially to monomeric rather than dimeric CBG. The N-terminal AcCoA binding site and the proposed C-terminal acyl-CoA binding site of CBG are compared with acyl-CoA binding sites of other tandem and single domain GNAT proteins.
- Published
- 2016
- Full Text
- View/download PDF
12. 5-Carboxy-8-hydroxyquinoline is a broad spectrum 2-oxoglutarate oxygenase inhibitor which causes iron translocation
- Author
-
Hopkinson, RJ, Tumber, A, Yapp, C, Chowdhury, R, Aik, W, Che, KH, Li, XS, Kristensen, JBL, King, ONF, Chan, MC, Yeoh, KK, Choi, H, Walport, LJ, Thinnes, CC, Bush, JT, Lejeune, C, Rydzik, AM, Rose, NR, Bagg, EA, McDonough, MA, Krojer, TJ, Yue, WW, Ng, SS, Olsen, Lars, Brennan, PE, Oppermann, U, Muller, S, Klose, RJ, Ratcliffe, PJ, Schofield, CJ, Kawamura, A, Hopkinson, RJ, Tumber, A, Yapp, C, Chowdhury, R, Aik, W, Che, KH, Li, XS, Kristensen, JBL, King, ONF, Chan, MC, Yeoh, KK, Choi, H, Walport, LJ, Thinnes, CC, Bush, JT, Lejeune, C, Rydzik, AM, Rose, NR, Bagg, EA, McDonough, MA, Krojer, TJ, Yue, WW, Ng, SS, Olsen, Lars, Brennan, PE, Oppermann, U, Muller, S, Klose, RJ, Ratcliffe, PJ, Schofield, CJ, and Kawamura, A
- Published
- 2013
13. Poppy tea and the baker' first seizure
- Author
-
Drummer, OH, primary, King, MA, additional, McDonough, MA, additional, and Berkovic, SF, additional
- Published
- 1997
- Full Text
- View/download PDF
14. Mutate and Conjugate: A Method to Enable Rapid In-Cell Target Validation.
- Author
-
Thomas AM, Serafini M, Grant EK, Coombs EAJ, Bluck JP, Schiedel M, McDonough MA, Reynolds JK, Lee B, Platt M, Sharlandjieva V, Biggin PC, Duarte F, Milne TA, Bush JT, and Conway SJ
- Subjects
- Humans, Ligands, HEK293 Cells, Mutant Proteins, Cell Cycle Proteins genetics, Nuclear Proteins genetics, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
Target validation remains a challenge in drug discovery, which leads to a high attrition rate in the drug discovery process, particularly in Phase II clinical trials. Consequently, new approaches to enhance target validation are valuable tools to improve the drug discovery process. Here, we report the combination of site-directed mutagenesis and electrophilic fragments to enable the rapid identification of small molecules that selectively inhibit the mutant protein. Using the bromodomain-containing protein BRD4 as an example, we employed a structure-based approach to identify the L94C mutation in the first bromodomain of BRD4 [BRD4(1)] as having a minimal effect on BRD4(1) function. We then screened a focused, KAc mimic-containing fragment set and a diverse fragment library against the mutant and wild-type proteins and identified a series of fragments that showed high selectivity for the mutant protein. These compounds were elaborated to include an alkyne click tag to enable the attachment of a fluorescent dye. These clickable compounds were then assessed in HEK293T cells, transiently expressing BRD4(1)
WT or BRD4(1)L94C , to determine their selectivity for BRD4(1)L94C over other possible cellular targets. One compound was identified that shows very high selectivity for BRD4(1)L94C over all other proteins. This work provides a proof-of-concept that the combination of site-directed mutagenesis and electrophilic fragments, in a mutate and conjugate approach, can enable rapid identification of small molecule inhibitors for an appropriately mutated protein of interest. This technology can be used to assess the cellular phenotype of inhibiting the protein of interest, and the electrophilic ligand provides a starting point for noncovalent ligand development.- Published
- 2023
- Full Text
- View/download PDF
15. Structural basis for binding of the renal carcinoma target hypoxia-inducible factor 2α to prolyl hydroxylase domain 2.
- Author
-
Figg WD Jr, Fiorini G, Chowdhury R, Nakashima Y, Tumber A, McDonough MA, and Schofield CJ
- Subjects
- Humans, Hypoxia metabolism, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor-Proline Dioxygenases genetics, Hypoxia-Inducible Factor-Proline Dioxygenases chemistry, Hypoxia-Inducible Factor-Proline Dioxygenases metabolism, Oxygen metabolism, Procollagen-Proline Dioxygenase chemistry, Procollagen-Proline Dioxygenase genetics, Procollagen-Proline Dioxygenase metabolism, Prolyl Hydroxylases, Protein Isoforms, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell genetics, Kidney Neoplasms drug therapy, Kidney Neoplasms genetics
- Abstract
The hypoxia-inducible factor (HIF) prolyl-hydroxylases (human PHD1-3) catalyze prolyl hydroxylation in oxygen-dependent degradation (ODD) domains of HIFα isoforms, modifications that signal for HIFα proteasomal degradation in an oxygen-dependent manner. PHD inhibitors are used for treatment of anemia in kidney disease. Increased erythropoietin (EPO) in patients with familial/idiopathic erythrocytosis and pulmonary hypertension is associated with mutations in EGLN1 (PHD2) and EPAS1 (HIF2α); a drug inhibiting HIF2α activity is used for clear cell renal cell carcinoma (ccRCC) treatment. We report crystal structures of PHD2 complexed with the C-terminal HIF2α-ODD in the presence of its 2-oxoglutarate cosubstrate or N-oxalylglycine inhibitor. Combined with the reported PHD2.HIFα-ODD structures and biochemical studies, the results inform on the different PHD.HIFα-ODD binding modes and the potential effects of clinically observed mutations in HIFα and PHD2 genes. They may help enable new therapeutic avenues, including PHD isoform-selective inhibitors and sequestration of HIF2α by the PHDs for ccRCC treatment., (© 2023 The Authors. Proteins: Structure, Function, and Bioinformatics published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
16. In vitro selection of macrocyclic peptide inhibitors containing cyclic γ 2,4 -amino acids targeting the SARS-CoV-2 main protease.
- Author
-
Miura T, Malla TR, Owen CD, Tumber A, Brewitz L, McDonough MA, Salah E, Terasaka N, Katoh T, Lukacik P, Strain-Damerell C, Mikolajek H, Walsh MA, Kawamura A, Schofield CJ, and Suga H
- Subjects
- Antiviral Agents chemistry, Carboxylic Acids, Peptides chemistry, Protease Inhibitors chemistry, Protease Inhibitors pharmacology, Protein Conformation, SARS-CoV-2 metabolism, Amino Acids chemistry, COVID-19
- Abstract
γ-Amino acids can play important roles in the biological activities of natural products; however, the ribosomal incorporation of γ-amino acids into peptides is challenging. Here we report how a selection campaign employing a non-canonical peptide library containing cyclic γ
2,4 -amino acids resulted in the discovery of very potent inhibitors of the SARS-CoV-2 main protease (Mpro ). Two kinds of cyclic γ2,4 -amino acids, cis-3-aminocyclobutane carboxylic acid (γ1 ) and (1R,3S)-3-aminocyclopentane carboxylic acid (γ2 ), were ribosomally introduced into a library of thioether-macrocyclic peptides. One resultant potent Mpro inhibitor (half-maximal inhibitory concentration = 50 nM), GM4, comprising 13 residues with γ1 at the fourth position, manifests a 5.2 nM dissociation constant. An Mpro :GM4 complex crystal structure reveals the intact inhibitor spans the substrate binding cleft. The γ1 interacts with the S1' catalytic subsite and contributes to a 12-fold increase in proteolytic stability compared to its alanine-substituted variant. Knowledge of interactions between GM4 and Mpro enabled production of a variant with a 5-fold increase in potency., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
17. In Situ Inhibitor Synthesis and Screening by Fluorescence Polarization: An Efficient Approach for Accelerating Drug Discovery.
- Author
-
Li Z, Wu Y, Zhen S, Su K, Zhang L, Yang F, McDonough MA, Schofield CJ, and Zhang X
- Abstract
Target-directed dynamic combinatorial chemistry has emerged as a useful tool for hit identification, but has not been widely used, in part due to challenges associated with analyses involving complex mixtures. We describe an operationally simple alternative: in situ inhibitor synthesis and screening (ISISS), which links high-throughput bioorthogonal synthesis with screening for target binding by fluorescence. We exemplify the ISISS method by showing how coupling screening for target binding by fluorescence polarization with the reaction of acyl-hydrazides and aldehydes led to the efficient discovery of a potent and novel acylhydrazone-based inhibitor of human prolyl hydroxylase 2 (PHD2), a target for anemia treatment, with equivalent in vivo potency to an approved medicine., Competing Interests: The authors declare no conflict of interest., (© 2022 The Authors. Angewandte Chemie published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
18. An LNA-amide modification that enhances the cell uptake and activity of phosphorothioate exon-skipping oligonucleotides.
- Author
-
Baker YR, Thorpe C, Chen J, Poller LM, Cox L, Kumar P, Lim WF, Lie L, McClorey G, Epple S, Singleton D, McDonough MA, Hardwick JS, Christensen KE, Wood MJA, Hall JP, El-Sagheer AH, and Brown T
- Subjects
- Amides, Exons, RNA chemistry, Oligonucleotides, Antisense genetics, Phosphorothioate Oligonucleotides
- Abstract
Oligonucleotides that target mRNA have great promise as therapeutic agents for life-threatening conditions but suffer from poor bioavailability, hence high cost. As currently untreatable diseases come within the reach of oligonucleotide therapies, new analogues are urgently needed to address this. With this in mind we describe reduced-charge oligonucleotides containing artificial LNA-amide linkages with improved gymnotic cell uptake, RNA affinity, stability and potency. To construct such oligonucleotides, five LNA-amide monomers (A, T, C, 5mC and G), where the 3'-OH is replaced by an ethanoic acid group, are synthesised in good yield and used in solid-phase oligonucleotide synthesis to form amide linkages with high efficiency. The artificial backbone causes minimal structural deviation to the DNA:RNA duplex. These studies indicate that splice-switching oligonucleotides containing LNA-amide linkages and phosphorothioates display improved activity relative to oligonucleotides lacking amides, highlighting the therapeutic potential of this technology., (© 2022. Crown.)
- Published
- 2022
- Full Text
- View/download PDF
19. Mechanisms of substrate recognition and N6-methyladenosine demethylation revealed by crystal structures of ALKBH5-RNA complexes.
- Author
-
Kaur S, Tam NY, McDonough MA, Schofield CJ, and Aik WS
- Subjects
- Adenosine analogs & derivatives, Adenosine metabolism, Alpha-Ketoglutarate-Dependent Dioxygenase FTO genetics, Alpha-Ketoglutarate-Dependent Dioxygenase FTO metabolism, Demethylation, Humans, AlkB Homolog 5, RNA Demethylase chemistry, AlkB Homolog 5, RNA Demethylase metabolism, RNA chemistry
- Abstract
AlkB homologue 5 (ALKBH5) is a ferrous iron and 2-oxoglutarate dependent oxygenase that demethylates RNA N6-methyladenosine (m6A), a post-transcriptional RNA modification with an emerging set of regulatory roles. Along with the fat mass and obesity-associated protein (FTO), ALKBH5 is one of only two identified human m6A RNA oxidizing enzymes and is a potential target for cancer treatment. Unlike FTO, ALKBH5 efficiently catalyzes fragmentation of its proposed nascent hemiaminal intermediate to give formaldehyde and a demethylated nucleoside. A detailed analysis of the molecular mechanisms used by ALKBH5 for substrate recognition and m6A demethylation is lacking. We report three crystal structures of ALKBH5 in complex with an m6A-ssRNA 8-mer substrate and supporting biochemical analyses. Strikingly, the single-stranded RNA substrate binds to the active site of ALKBH5 in a 5'-3' orientation that is opposite to single-stranded or double-stranded DNA substrates observed for other AlkB subfamily members, including single-stranded DNA bound to FTO. The combined structural and biochemical results provide insight into the preference of ALKBH5 for substrates containing a (A/G)m6AC consensus sequence motif. The results support a mechanism involving formation of an m6A hemiaminal intermediate, followed by efficient ALKBH5 catalyzed demethylation, enabled by a proton shuttle network involving Lys132 and Tyr139., (© The Author(s) 2022. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
20. Studies on the Reactions of Biapenem with VIM Metallo β-Lactamases and the Serine β-Lactamase KPC-2.
- Author
-
Lucic A, Malla TR, Calvopiña K, Tooke CL, Brem J, McDonough MA, Spencer J, and Schofield CJ
- Abstract
Carbapenems are important antibacterials and are both substrates and inhibitors of some β-lactamases. We report studies on the reaction of the unusual carbapenem biapenem, with the subclass B1 metallo-β-lactamases VIM-1 and VIM-2 and the class A serine-β-lactamase KPC-2. X-ray diffraction studies with VIM-2 crystals treated with biapenem reveal the opening of the β-lactam ring to form a mixture of the (2 S )-imine and enamine complexed at the active site. NMR studies on the reactions of biapenem with VIM-1, VIM-2, and KPC-2 reveal the formation of hydrolysed enamine and (2 R )- and (2 S )-imine products. The combined results support the proposal that SBL/MBL-mediated carbapenem hydrolysis results in a mixture of tautomerizing enamine and (2 R )- and (2 S )-imine products, with the thermodynamically favoured (2 S )-imine being the major observed species over a relatively long-time scale. The results suggest that prolonging the lifetimes of β-lactamase carbapenem complexes by optimising tautomerisation of the nascently formed enamine to the (2 R )-imine and likely more stable (2 S )-imine tautomer is of interest in developing improved carbapenems.
- Published
- 2022
- Full Text
- View/download PDF
21. Inhibition of JMJD6 by 2-Oxoglutarate Mimics.
- Author
-
Islam MS, Thinnes CC, Holt-Martyn JP, Chowdhury R, McDonough MA, and Schofield CJ
- Subjects
- Dose-Response Relationship, Drug, Humans, Jumonji Domain-Containing Histone Demethylases metabolism, Ketoglutaric Acids chemistry, Molecular Structure, Prolyl-Hydroxylase Inhibitors chemistry, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Structure-Activity Relationship, Jumonji Domain-Containing Histone Demethylases antagonists & inhibitors, Ketoglutaric Acids pharmacology, Prolyl-Hydroxylase Inhibitors pharmacology
- Abstract
Studies on the inhibition of the human 2-oxoglutarate dependent oxygenase JMJD6, which is a cancer target, by 2-oxoglutarate mimics / competitors, including human drugs, drug candidates, and metabolites relevant to cancer are described. JMJD6 assays employed NMR to monitor inhibitor binding and use of mass spectrometry to monitor JMJD6-catalysed lysine hydroxylation. Notably, some clinically applied prolyl hydroxylase inhibitors also inhibit JMJD6. The results will help enable the development of inhibitors selective for human oxygenases, including JMJD6., (© 2021 The Authors. ChemMedChem published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
22. Imitation of β-lactam binding enables broad-spectrum metallo-β-lactamase inhibitors.
- Author
-
Brem J, Panduwawala T, Hansen JU, Hewitt J, Liepins E, Donets P, Espina L, Farley AJM, Shubin K, Campillos GG, Kiuru P, Shishodia S, Krahn D, Leśniak RK, Schmidt Adrian J, Calvopiña K, Turrientes MC, Kavanagh ME, Lubriks D, Hinchliffe P, Langley GW, Aboklaish AF, Eneroth A, Backlund M, Baran AG, Nielsen EI, Speake M, Kuka J, Robinson J, Grinberga S, Robinson L, McDonough MA, Rydzik AM, Leissing TM, Jimenez-Castellanos JC, Avison MB, Da Silva Pinto S, Pannifer AD, Martjuga M, Widlake E, Priede M, Hopkins Navratilova I, Gniadkowski M, Belfrage AK, Brandt P, Yli-Kauhaluoma J, Bacque E, Page MGP, Björkling F, Tyrrell JM, Spencer J, Lang PA, Baranczewski P, Cantón R, McElroy SP, Jones PS, Baquero F, Suna E, Morrison A, Walsh TR, and Schofield CJ
- Subjects
- Animals, Gram-Negative Bacteria drug effects, Humans, Mice, Microbial Sensitivity Tests, Protein Binding, Structure-Activity Relationship, beta-Lactamase Inhibitors chemistry, beta-Lactamase Inhibitors metabolism, beta-Lactamase Inhibitors pharmacology, beta-Lactams metabolism
- Abstract
Carbapenems are vital antibiotics, but their efficacy is increasingly compromised by metallo-β-lactamases (MBLs). Here we report the discovery and optimization of potent broad-spectrum MBL inhibitors. A high-throughput screen for NDM-1 inhibitors identified indole-2-carboxylates (InCs) as potential β-lactamase stable β-lactam mimics. Subsequent structure-activity relationship studies revealed InCs as a new class of potent MBL inhibitor, active against all MBL classes of major clinical relevance. Crystallographic studies revealed a binding mode of the InCs to MBLs that, in some regards, mimics that predicted for intact carbapenems, including with respect to maintenance of the Zn(II)-bound hydroxyl, and in other regards mimics binding observed in MBL-carbapenem product complexes. InCs restore carbapenem activity against multiple drug-resistant Gram-negative bacteria and have a low frequency of resistance. InCs also have a good in vivo safety profile, and when combined with meropenem show a strong in vivo efficacy in peritonitis and thigh mouse infection models., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
23. Structure-Based Design of Selective Fat Mass and Obesity Associated Protein (FTO) Inhibitors.
- Author
-
Shishodia S, Demetriades M, Zhang D, Tam NY, Maheswaran P, Clunie-O'Connor C, Tumber A, Leung IKH, Ng YM, Leissing TM, El-Sagheer AH, Salah E, Brown T, Aik WS, McDonough MA, and Schofield CJ
- Subjects
- Antineoplastic Agents chemistry, Crystallography, X-Ray, Histone Demethylases metabolism, Humans, Mixed Function Oxygenases metabolism, Structure-Activity Relationship, Alpha-Ketoglutarate-Dependent Dioxygenase FTO antagonists & inhibitors, Antineoplastic Agents pharmacology, Drug Design
- Abstract
FTO catalyzes the Fe(II) and 2-oxoglutarate (2OG)-dependent modification of nucleic acids, including the demethylation of N
6 -methyladenosine (m6 A) in mRNA. FTO is a proposed target for anti-cancer therapy. Using information from crystal structures of FTO in complex with 2OG and substrate mimics, we designed and synthesized two series of FTO inhibitors, which were characterized by turnover and binding assays, and by X-ray crystallography with FTO and the related bacterial enzyme AlkB. A potent inhibitor employing binding interactions spanning the FTO 2OG and substrate binding sites was identified. Selectivity over other clinically targeted 2OG oxygenases was demonstrated, including with respect to the hypoxia-inducible factor prolyl and asparaginyl hydroxylases (PHD2 and FIH) and selected JmjC histone demethylases (KDMs). The results illustrate how structure-based design can enable the identification of potent and selective 2OG oxygenase inhibitors and will be useful for the development of FTO inhibitors for use in vivo .- Published
- 2021
- Full Text
- View/download PDF
24. X-ray free-electron laser studies reveal correlated motion during isopenicillin N synthase catalysis.
- Author
-
Rabe P, Kamps JJAG, Sutherlin KD, Linyard JDS, Aller P, Pham CC, Makita H, Clifton I, McDonough MA, Leissing TM, Shutin D, Lang PA, Butryn A, Brem J, Gul S, Fuller FD, Kim IS, Cheah MH, Fransson T, Bhowmick A, Young ID, O'Riordan L, Brewster AS, Pettinati I, Doyle M, Joti Y, Owada S, Tono K, Batyuk A, Hunter MS, Alonso-Mori R, Bergmann U, Owen RL, Sauter NK, Claridge TDW, Robinson CV, Yachandra VK, Yano J, Kern JF, Orville AM, and Schofield CJ
- Subjects
- Catalysis, Catalytic Domain, Crystallography, X-Ray, Ferric Compounds, Humans, Lasers, Oxygen chemistry, Penicillins chemistry, Penicillins metabolism, Substrate Specificity, Electrons, Oxidoreductases chemistry
- Abstract
Isopenicillin N synthase (IPNS) catalyzes the unique reaction of l-δ-(α-aminoadipoyl)-l-cysteinyl-d-valine (ACV) with dioxygen giving isopenicillin N (IPN), the precursor of all natural penicillins and cephalosporins. X-ray free-electron laser studies including time-resolved crystallography and emission spectroscopy reveal how reaction of IPNS:Fe(II):ACV with dioxygen to yield an Fe(III) superoxide causes differences in active site volume and unexpected conformational changes that propagate to structurally remote regions. Combined with solution studies, the results reveal the importance of protein dynamics in regulating intermediate conformations during conversion of ACV to IPN. The results have implications for catalysis by multiple IPNS-related oxygenases, including those involved in the human hypoxic response, and highlight the power of serial femtosecond crystallography to provide insight into long-range enzyme dynamics during reactions presently impossible for nonprotein catalysts., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)
- Published
- 2021
- Full Text
- View/download PDF
25. Structural Basis of Prolyl Hydroxylase Domain Inhibition by Molidustat.
- Author
-
Figg WD Jr, McDonough MA, Chowdhury R, Nakashima Y, Zhang Z, Holt-Martyn JP, Krajnc A, and Schofield CJ
- Subjects
- Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Prolyl-Hydroxylase Inhibitors chemistry, Pyrazoles chemistry, Structure-Activity Relationship, Triazoles chemistry, Prolyl Hydroxylases metabolism, Prolyl-Hydroxylase Inhibitors pharmacology, Pyrazoles pharmacology, Triazoles pharmacology
- Abstract
Human prolyl-hydroxylases (PHDs) are hypoxia-sensing 2-oxoglutarate (2OG) oxygenases, catalysis by which suppresses the transcription of hypoxia-inducible factor target genes. PHD inhibition enables the treatment of anaemia/ischaemia-related disease. The PHD inhibitor Molidustat is approved for the treatment of renal anaemia; it differs from other approved/late-stage PHD inhibitors in lacking a glycinamide side chain. The first reported crystal structures of Molidustat and IOX4 (a brain-penetrating derivative) complexed with PHD2 reveal how their contiguous triazole, pyrazolone and pyrimidine/pyridine rings bind at the active site. The inhibitors bind to the active-site metal in a bidentate manner through their pyrazolone and pyrimidine nitrogens, with the triazole π-π-stacking with Tyr303 in the 2OG binding pocket. Comparison of the new structures with other PHD inhibitor complexes reveals differences in the conformations of Tyr303, Tyr310, and a mobile loop linking β2-β3, which are involved in dynamic substrate binding/product release., (© 2021 The Authors. ChemMedChem published by Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
26. Human Oxygenase Variants Employing a Single Protein Fe II Ligand Are Catalytically Active.
- Author
-
Brasnett A, Pfeffer I, Brewitz L, Chowdhury R, Nakashima Y, Tumber A, McDonough MA, and Schofield CJ
- Abstract
Aspartate/asparagine-β-hydroxylase (AspH) is a human 2-oxoglutarate (2OG) and Fe
II oxygenase that catalyses C3 hydroxylations of aspartate/asparagine residues of epidermal growth factor-like domains (EGFDs). Unusually, AspH employs two histidine residues to chelate FeII rather than the typical triad of two histidine and one glutamate/aspartate residue. We report kinetic, inhibition, and crystallographic studies concerning human AspH variants in which either of its FeII binding histidine residues are substituted for alanine. Both the H725A and, in particular, the H679A AspH variants retain substantial catalytic activity. Crystal structures clearly reveal metal-ligation by only a single protein histidine ligand. The results have implications for the functional assignment of 2OG oxygenases and for the design of non-protein biomimetic catalysts., Competing Interests: The authors declare no conflict of interest., (© 2021 The Authors. Angewandte Chemie published by Wiley-VCH GmbH.)- Published
- 2021
- Full Text
- View/download PDF
27. Inhibition of the Oxygen-Sensing Asparaginyl Hydroxylase Factor Inhibiting Hypoxia-Inducible Factor: A Potential Hypoxia Response Modulating Strategy.
- Author
-
Wu Y, Li Z, McDonough MA, Schofield CJ, and Zhang X
- Subjects
- Asparagine metabolism, Humans, Hydroxylation, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Hypoxia-Inducible Factor-Proline Dioxygenases chemistry, Hypoxia-Inducible Factor-Proline Dioxygenases metabolism, I-kappa B Proteins metabolism, Mixed Function Oxygenases antagonists & inhibitors, Oxygen chemistry, Repressor Proteins antagonists & inhibitors, Substrate Specificity, p300-CBP Transcription Factors chemistry, p300-CBP Transcription Factors metabolism, Mixed Function Oxygenases metabolism, Oxygen metabolism, Repressor Proteins metabolism
- Abstract
Factor inhibiting hypoxia-inducible factor (FIH) is a JmjC domain 2-oxogluarate and Fe(II)-dependent oxygenase that catalyzes hydroxylation of specific asparagines in the C-terminal transcriptional activation domain of hypoxia-inducible factor alpha (HIF-α) isoforms. This modification suppresses the transcriptional activity of HIF by reducing its interaction with the transcriptional coactivators p300/CBP. By contrast with inhibition of the HIF prolyl hydroxylases (PHDs), inhibitors of FIH, which accepts multiple non-HIF substrates, are less studied; they are of interest due to their potential ability to alter metabolism (either in a HIF-dependent and/or -independent manner) and, provided HIF is upregulated, to modulate the course of the HIF-mediated hypoxic response. Here we review studies on the mechanism and inhibition of FIH. We discuss proposed biological roles of FIH including its regulation of HIF activity and potential roles of FIH-catalyzed oxidation of non-HIF substrates. We highlight potential therapeutic applications of FIH inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
28. Faropenem reacts with serine and metallo-β-lactamases to give multiple products.
- Author
-
Lucic A, Hinchliffe P, Malla TR, Tooke CL, Brem J, Calvopiña K, Lohans CT, Rabe P, McDonough MA, Armistead T, Orville AM, Spencer J, and Schofield CJ
- Subjects
- Anti-Bacterial Agents metabolism, Catalytic Domain, Crystallography, X-Ray, Hydrolysis, Klebsiella pneumoniae enzymology, Protein Binding, Pseudomonas aeruginosa enzymology, Stenotrophomonas maltophilia enzymology, beta-Lactamase Inhibitors metabolism, beta-Lactamases metabolism, beta-Lactams metabolism, Anti-Bacterial Agents chemistry, beta-Lactamase Inhibitors chemistry, beta-Lactamases chemistry, beta-Lactams chemistry
- Abstract
Penems have demonstrated potential as antibacterials and β-lactamase inhibitors; however, their clinical use has been limited, especially in comparison with the structurally related carbapenems. Faropenem is an orally active antibiotic with a C-2 tetrahydrofuran (THF) ring, which is resistant to hydrolysis by some β-lactamases. We report studies on the reactions of faropenem with carbapenem-hydrolysing β-lactamases, focusing on the class A serine β-lactamase KPC-2 and the metallo β-lactamases (MBLs) VIM-2 (a subclass B1 MBL) and L1 (a B3 MBL). Kinetic studies show that faropenem is a substrate for all three β-lactamases, though it is less efficiently hydrolysed by KPC-2. Crystallographic analyses on faropenem-derived complexes reveal opening of the β-lactam ring with formation of an imine with KPC-2, VIM-2, and L1. In the cases of the KPC-2 and VIM-2 structures, the THF ring is opened to give an alkene, but with L1 the THF ring remains intact. Solution state studies, employing NMR, were performed on L1, KPC-2, VIM-2, VIM-1, NDM-1, OXA-23, OXA-10, and OXA-48. The solution results reveal, in all cases, formation of imine products in which the THF ring is opened; formation of a THF ring-closed imine product was only observed with VIM-1 and VIM-2. An enamine product with a closed THF ring was also observed in all cases, at varying levels. Combined with previous reports, the results exemplify the potential for different outcomes in the reactions of penems with MBLs and SBLs and imply further structure-activity relationship studies are worthwhile to optimise the interactions of penems with β-lactamases. They also exemplify how crystal structures of β-lactamase substrate/inhibitor complexes do not always reflect reaction outcomes in solution., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021. Published by Elsevier Masson SAS.)
- Published
- 2021
- Full Text
- View/download PDF
29. The methyltransferase METTL9 mediates pervasive 1-methylhistidine modification in mammalian proteomes.
- Author
-
Davydova E, Shimazu T, Schuhmacher MK, Jakobsson ME, Willemen HLDM, Liu T, Moen A, Ho AYY, Małecki J, Schroer L, Pinto R, Suzuki T, Grønsberg IA, Sohtome Y, Akakabe M, Weirich S, Kikuchi M, Olsen JV, Dohmae N, Umehara T, Sodeoka M, Siino V, McDonough MA, Eijkelkamp N, Schofield CJ, Jeltsch A, Shinkai Y, and Falnes PØ
- Subjects
- Amino Acid Motifs, Animals, Cells, Cultured, Histidine metabolism, Humans, Mammals classification, Mammals genetics, Mammals metabolism, Methylation, Methyltransferases genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitochondria metabolism, Mutation, Protein Processing, Post-Translational, Proteome chemistry, Substrate Specificity, Zinc metabolism, Methylhistidines metabolism, Methyltransferases metabolism, Proteome metabolism
- Abstract
Post-translational methylation plays a crucial role in regulating and optimizing protein function. Protein histidine methylation, occurring as the two isomers 1- and 3-methylhistidine (1MH and 3MH), was first reported five decades ago, but remains largely unexplored. Here we report that METTL9 is a broad-specificity methyltransferase that mediates the formation of the majority of 1MH present in mouse and human proteomes. METTL9-catalyzed methylation requires a His-x-His (HxH) motif, where "x" is preferably a small amino acid, allowing METTL9 to methylate a number of HxH-containing proteins, including the immunomodulatory protein S100A9 and the NDUFB3 subunit of mitochondrial respiratory Complex I. Notably, METTL9-mediated methylation enhances respiration via Complex I, and the presence of 1MH in an HxH-containing peptide reduced its zinc binding affinity. Our results establish METTL9-mediated 1MH as a pervasive protein modification, thus setting the stage for further functional studies on protein histidine methylation.
- Published
- 2021
- Full Text
- View/download PDF
30. Author Correction: Aspartate/asparagine-β-hydroxylase: a high-throughput mass spectrometric assay for discovery of small molecule inhibitors.
- Author
-
Brewitz L, Tumber A, Pfeffer I, McDonough MA, and Schofield CJ
- Published
- 2020
- Full Text
- View/download PDF
31. Biochemical and biophysical analyses of hypoxia sensing prolyl hydroxylases from Dictyostelium discoideum and Toxoplasma gondii .
- Author
-
Liu T, Abboud MI, Chowdhury R, Tumber A, Hardy AP, Lippl K, Lohans CT, Pires E, Wickens J, McDonough MA, West CM, and Schofield CJ
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Biocatalysis, Crystallography, X-Ray, Humans, Hydroxylation, Hypoxia-Inducible Factor 1, alpha Subunit chemistry, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kinetics, Molecular Dynamics Simulation, Oxygen metabolism, Prolyl Hydroxylases chemistry, Prolyl Hydroxylases genetics, Protein Structure, Tertiary, Protozoan Proteins chemistry, Protozoan Proteins genetics, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, S-Phase Kinase-Associated Proteins chemistry, S-Phase Kinase-Associated Proteins metabolism, Sequence Alignment, Substrate Specificity, Dictyostelium enzymology, Prolyl Hydroxylases metabolism, Protozoan Proteins metabolism, Toxoplasma enzymology
- Abstract
In animals, the response to chronic hypoxia is mediated by prolyl hydroxylases (PHDs) that regulate the levels of hypoxia-inducible transcription factor α (HIFα). PHD homologues exist in other types of eukaryotes and prokaryotes where they act on non HIF substrates. To gain insight into the factors underlying different PHD substrates and properties, we carried out biochemical and biophysical studies on PHD homologues from the cellular slime mold, Dictyostelium discoideum, and the protozoan parasite, Toxoplasma gondii , both lacking HIF. The respective prolyl-hydroxylases (DdPhyA and TgPhyA) catalyze prolyl-hydroxylation of S-phase kinase-associated protein 1 (Skp1), a reaction enabling adaptation to different dioxygen availability. Assays with full-length Skp1 substrates reveal substantial differences in the kinetic properties of DdPhyA and TgPhyA, both with respect to each other and compared with human PHD2; consistent with cellular studies, TgPhyA is more active at low dioxygen concentrations than DdPhyA. TgSkp1 is a DdPhyA substrate and DdSkp1 is a TgPhyA substrate. No cross-reactivity was detected between DdPhyA/TgPhyA substrates and human PHD2. The human Skp1 E147P variant is a DdPhyA and TgPhyA substrate, suggesting some retention of ancestral interactions. Crystallographic analysis of DdPhyA enables comparisons with homologues from humans, Trichoplax adhaerens , and prokaryotes, informing on differences in mobile elements involved in substrate binding and catalysis. In DdPhyA, two mobile loops that enclose substrates in the PHDs are conserved, but the C-terminal helix of the PHDs is strikingly absent. The combined results support the proposal that PHD homologues have evolved kinetic and structural features suited to their specific sensing roles., Competing Interests: Conflict of interest—The authors declare no competing financial interests., (© 2020 Liu et al.)
- Published
- 2020
- Full Text
- View/download PDF
32. Structures of Arabidopsis thaliana oxygen-sensing plant cysteine oxidases 4 and 5 enable targeted manipulation of their activity.
- Author
-
White MD, Dalle Carbonare L, Lavilla Puerta M, Iacopino S, Edwards M, Dunne K, Pires E, Levy C, McDonough MA, Licausi F, and Flashman E
- Subjects
- Cysteine Dioxygenase metabolism, Gene Expression Regulation, Plant physiology, Oxidation-Reduction, Signal Transduction physiology, Transcription Factors, Arabidopsis metabolism, Arabidopsis Proteins metabolism, Oxygen metabolism
- Abstract
In higher plants, molecular responses to exogenous hypoxia are driven by group VII ethylene response factors (ERF-VIIs). These transcriptional regulators accumulate in the nucleus under hypoxia to activate anaerobic genes but are destabilized in normoxic conditions through the action of oxygen-sensing plant cysteine oxidases (PCOs). The PCOs catalyze the reaction of oxygen with the conserved N-terminal cysteine of ERF-VIIs to form cysteine sulfinic acid, triggering degradation via the Cys/Arg branch of the N-degron pathway. The PCOs are therefore a vital component of the plant oxygen signaling system, connecting environmental stimulus with cellular and physiological response. Rational manipulation of PCO activity could regulate ERF-VII levels and improve flood tolerance, but requires detailed structural information. We report crystal structures of the constitutively expressed PCO4 and PCO5 from Arabidopsis thaliana to 1.24 and 1.91 Å resolution, respectively. The structures reveal that the PCOs comprise a cupin-like scaffold, which supports a central metal cofactor coordinated by three histidines. While this overall structure is consistent with other thiol dioxygenases, closer inspection of the active site indicates that other catalytic features are not conserved, suggesting that the PCOs may use divergent mechanisms to oxidize their substrates. Conservative substitution of two active site residues had dramatic effects on PCO4 function both in vitro and in vivo, through yeast and plant complementation assays. Collectively, our data identify key structural elements that are required for PCO activity and provide a platform for engineering crops with improved hypoxia tolerance., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
33. Anaerobic fixed-target serial crystallography.
- Author
-
Rabe P, Beale JH, Butryn A, Aller P, Dirr A, Lang PA, Axford DN, Carr SB, Leissing TM, McDonough MA, Davy B, Ebrahim A, Orlans J, Storm SLS, Orville AM, Schofield CJ, and Owen RL
- Abstract
Cryogenic X-ray diffraction is a powerful tool for crystallographic studies on enzymes including oxygenases and oxidases. Amongst the benefits that cryo-conditions (usually employing a nitro-gen cryo-stream at 100 K) enable, is data collection of di-oxy-gen-sensitive samples. Although not strictly anaerobic, at low temperatures the vitreous ice conditions severely restrict O
2 diffusion into and/or through the protein crystal. Cryo-conditions limit chemical reactivity, including reactions that require significant conformational changes. By contrast, data collection at room temperature imposes fewer restrictions on diffusion and reactivity; room-temperature serial methods are thus becoming common at synchrotrons and XFELs. However, maintaining an anaerobic environment for di-oxy-gen-dependent enzymes has not been explored for serial room-temperature data collection at synchrotron light sources. This work describes a methodology that employs an adaptation of the 'sheet-on-sheet' sample mount, which is suitable for the low-dose room-temperature data collection of anaerobic samples at synchrotron light sources. The method is characterized by easy sample preparation in an anaerobic glovebox, gentle handling of crystals, low sample consumption and preservation of a localized anaerobic environment over the timescale of the experiment (<5 min). The utility of the method is highlighted by studies with three X-ray-radiation-sensitive Fe(II)-containing model enzymes: the 2-oxoglutarate-dependent l-arginine hy-droxy-lase VioC and the DNA repair enzyme AlkB, as well as the oxidase isopenicillin N synthase (IPNS), which is involved in the biosynthesis of all penicillin and cephalosporin antibiotics., (© Patrick Rabe et al. 2020.)- Published
- 2020
- Full Text
- View/download PDF
34. Broad Spectrum β-Lactamase Inhibition by a Thioether Substituted Bicyclic Boronate.
- Author
-
Parkova A, Lucic A, Krajnc A, Brem J, Calvopiña K, Langley GW, McDonough MA, Trapencieris P, and Schofield CJ
- Subjects
- beta-Lactamase Inhibitors pharmacology, beta-Lactamases, Anti-Bacterial Agents pharmacology, Sulfides
- Abstract
β-Lactamases comprise the most widely used mode of resistance to β-lactam antibiotics. Cyclic boronates have shown promise as a new class of β-lactamase inhibitor, with pioneering potential to potently inhibit both metallo- and serine-β-lactamases. We report studies concerning a bicyclic boronate ester with a thioether rather than the more typical β-lactam antibiotic "C-6/C-7" acylamino type side chain, which is present in the penicillin/cephalosporin antibiotics. The thioether bicyclic boronate ester was tested for activity against representative serine- and metallo-β-lactamases. The results support the broad inhibition potential of bicyclic boronate based inhibitors with different side chains, including against metallo-β-lactamases from B1, B2, and B3 subclasses. Combined with previous crystallographic studies, analysis of a crystal structure of the thioether inhibitor with the clinically relevant VIM-2 metallo-β-lactamase implies that further SAR work will expand the already broad scope of β-lactamase inhibition by bicyclic boronates.
- Published
- 2020
- Full Text
- View/download PDF
35. Aspartate/asparagine-β-hydroxylase: a high-throughput mass spectrometric assay for discovery of small molecule inhibitors.
- Author
-
Brewitz L, Tumber A, Pfeffer I, McDonough MA, and Schofield CJ
- Subjects
- Antineoplastic Agents chemistry, Cell Line, Tumor, Crystallography, X-Ray, Enzyme Inhibitors chemistry, High-Throughput Screening Assays, Humans, Hydroxylation drug effects, Mass Spectrometry, Mixed Function Oxygenases chemistry, Neoplasms enzymology, Neoplasms pathology, Pyridines chemistry, Pyridines pharmacology, Antineoplastic Agents pharmacology, Drug Discovery methods, Drug Screening Assays, Antitumor methods, Enzyme Inhibitors pharmacology, Mixed Function Oxygenases antagonists & inhibitors, Neoplasms drug therapy
- Abstract
The human 2-oxoglutarate dependent oxygenase aspartate/asparagine-β-hydroxylase (AspH) catalyses the hydroxylation of Asp/Asn-residues in epidermal growth factor-like domains (EGFDs). AspH is upregulated on the surface of malign cancer cells; increased AspH levels correlate with tumour invasiveness. Due to a lack of efficient assays to monitor the activity of isolated AspH, there are few reports of studies aimed at identifying small-molecule AspH inhibitors. Recently, it was reported that AspH substrates have a non-canonical EGFD disulfide pattern. Here we report that a stable synthetic thioether mimic of AspH substrates can be employed in solid phase extraction mass spectrometry based high-throughput AspH inhibition assays which are of excellent robustness, as indicated by high Z'-factors and good signal-to-noise/background ratios. The AspH inhibition assay was applied to screen approximately 1500 bioactive small-molecules, including natural products and active pharmaceutical ingredients of approved human therapeutics. Potent AspH inhibitors were identified from both compound classes. Our AspH inhibition assay should enable the development of potent and selective small-molecule AspH inhibitors and contribute towards the development of safer inhibitors for other 2OG oxygenases, e.g. screens of the hypoxia-inducible factor prolyl-hydroxylase inhibitors revealed that vadadustat inhibits AspH with moderate potency.
- Published
- 2020
- Full Text
- View/download PDF
36. A human protein hydroxylase that accepts D-residues.
- Author
-
Choi H, Hardy AP, Leissing TM, Chowdhury R, Nakashima Y, Ge W, Markoulides M, Scotti JS, Gerken PA, Thorbjornsrud H, Kang D, Hong S, Lee J, McDonough MA, Park H, and Schofield CJ
- Abstract
Factor inhibiting hypoxia-inducible factor (FIH) is a 2-oxoglutarate-dependent protein hydroxylase that catalyses C3 hydroxylations of protein residues. We report FIH can accept (D)- and (L)-residues for hydroxylation. The substrate selectivity of FIH differs for (D) and (L) epimers, e.g., (D)- but not (L)-allylglycine, and conversely (L)- but not (D)-aspartate, undergo monohydroxylation, in the tested sequence context. The (L)-Leu-containing substrate undergoes FIH-catalysed monohydroxylation, whereas (D)-Leu unexpectedly undergoes dihydroxylation. Crystallographic, mass spectrometric, and DFT studies provide insights into the selectivity of FIH towards (L)- and (D)-residues. The results of this work expand the potential range of known substrates hydroxylated by isolated FIH and imply that it will be possible to generate FIH variants with altered selectivities., (© 2020. The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
37. Structure-Activity Relationship and Crystallographic Studies on 4-Hydroxypyrimidine HIF Prolyl Hydroxylase Domain Inhibitors.
- Author
-
Holt-Martyn JP, Chowdhury R, Tumber A, Yeh TL, Abboud MI, Lippl K, Lohans CT, Langley GW, Figg W Jr, McDonough MA, Pugh CW, Ratcliffe PJ, and Schofield CJ
- Subjects
- Crystallography, X-Ray, Dose-Response Relationship, Drug, Humans, Hypoxia-Inducible Factor-Proline Dioxygenases metabolism, Models, Molecular, Molecular Structure, Prolyl-Hydroxylase Inhibitors chemistry, Pyrimidinones chemistry, Structure-Activity Relationship, Hypoxia-Inducible Factor-Proline Dioxygenases antagonists & inhibitors, Prolyl-Hydroxylase Inhibitors pharmacology, Pyrimidinones pharmacology
- Abstract
The 2-oxoglutarate-dependent hypoxia inducible factor prolyl hydroxylases (PHDs) are targets for treatment of a variety of diseases including anaemia. One PHD inhibitor is approved for use for the treatment of renal anaemia and others are in late stage clinical trials. The number of reported templates for PHD inhibition is limited. We report structure-activity relationship and crystallographic studies on a promising class of 4-hydroxypyrimidine-containing PHD inhibitors., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
38. MeLAD: an integrated resource for metalloenzyme-ligand associations.
- Author
-
Li G, Su Y, Yan YH, Peng JY, Dai QQ, Ning XL, Zhu CL, Fu C, McDonough MA, Schofield CJ, Huang C, and Li GB
- Subjects
- Catalytic Domain, Drug Discovery, Ligands, Metals, Metalloproteins
- Abstract
Motivation: Metalloenzymes are attractive targets for therapeutic intervention owing to their central roles in various biological processes and pathological situations. The fast-growing body of structural data on metalloenzyme-ligand interactions is facilitating efficient drug discovery targeting metalloenzymes. However, there remains a shortage of specific databases that can provide centralized, interconnected information exclusive to metalloenzyme-ligand associations., Results: We created a Metalloenzyme-Ligand Association Database (MeLAD), which is designed to provide curated structural data and information exclusive to metalloenzyme-ligand interactions, and more uniquely, present expanded associations that are represented by metal-binding pharmacophores (MBPs), metalloenzyme structural similarity (MeSIM) and ligand chemical similarity (LigSIM). MeLAD currently contains 6086 structurally resolved interactions of 1416 metalloenzymes with 3564 ligands, of which classical metal-binding, non-classical metal-binding, non-metal-binding and metal water-bridging interactions account for 63.0%, 2.3%, 34.4% and 0.3%, respectively. A total of 263 monodentate, 191 bidentate and 15 tridentate MBP chemotypes were included in MeLAD, which are linked to different active site metal ions and coordination modes. 3726 and 52 740 deductive metalloenzyme-ligand associations by MeSIM and LigSIM analyses, respectively, were included in MeLAD. An online server is provided for users to conduct metalloenzyme profiling prediction for small molecules of interest. MeLAD is searchable by multiple criteria, e.g. metalloenzyme name, ligand identifier, functional class, bioinorganic class, metal ion and metal-containing cofactor, which will serve as a valuable, integrative data source to foster metalloenzyme related research, particularly involved in drug discovery targeting metalloenzymes., Availability and Implementation: MeLAD is accessible at https://melad.ddtmlab.org., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
39. Aspartate/asparagine-β-hydroxylase crystal structures reveal an unexpected epidermal growth factor-like domain substrate disulfide pattern.
- Author
-
Pfeffer I, Brewitz L, Krojer T, Jensen SA, Kochan GT, Kershaw NJ, Hewitson KS, McNeill LA, Kramer H, Münzel M, Hopkinson RJ, Oppermann U, Handford PA, McDonough MA, and Schofield CJ
- Subjects
- Amino Acid Sequence, Asparagine metabolism, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Catalytic Domain, Crystallography, Disulfides chemistry, Disulfides metabolism, Epidermal Growth Factor metabolism, Ferrous Compounds chemistry, Ferrous Compounds metabolism, Humans, Membrane Proteins genetics, Membrane Proteins metabolism, Mixed Function Oxygenases genetics, Mixed Function Oxygenases metabolism, Muscle Proteins genetics, Muscle Proteins metabolism, Protein Conformation, Calcium-Binding Proteins chemistry, Membrane Proteins chemistry, Mixed Function Oxygenases chemistry, Muscle Proteins chemistry
- Abstract
AspH is an endoplasmic reticulum (ER) membrane-anchored 2-oxoglutarate oxygenase whose C-terminal oxygenase and tetratricopeptide repeat (TPR) domains present in the ER lumen. AspH catalyses hydroxylation of asparaginyl- and aspartyl-residues in epidermal growth factor-like domains (EGFDs). Here we report crystal structures of human AspH, with and without substrate, that reveal substantial conformational changes of the oxygenase and TPR domains during substrate binding. Fe(II)-binding by AspH is unusual, employing only two Fe(II)-binding ligands (His679/His725). Most EGFD structures adopt an established fold with a conserved Cys1-3, 2-4, 5-6 disulfide bonding pattern; an unexpected Cys3-4 disulfide bonding pattern is observed in AspH-EGFD substrate complexes, the catalytic relevance of which is supported by studies involving stable cyclic peptide substrate analogues and by effects of Ca(II) ions on activity. The results have implications for EGFD disulfide pattern processing in the ER and will enable medicinal chemistry efforts targeting human 2OG oxygenases.
- Published
- 2019
- Full Text
- View/download PDF
40. Studies on the inhibition of AmpC and other β-lactamases by cyclic boronates.
- Author
-
Cahill ST, Tyrrell JM, Navratilova IH, Calvopiña K, Robinson SW, Lohans CT, McDonough MA, Cain R, Fishwick CWG, Avison MB, Walsh TR, Schofield CJ, and Brem J
- Subjects
- Anti-Bacterial Agents chemistry, Boronic Acids chemistry, Crystallography, X-Ray, Models, Molecular, Molecular Structure, Pseudomonas aeruginosa enzymology, beta-Lactamase Inhibitors chemistry, Anti-Bacterial Agents pharmacology, Boronic Acids pharmacology, beta-Lactamase Inhibitors pharmacology, beta-Lactamases metabolism
- Abstract
Background: The β-lactam antibiotics represent the most successful drug class for treatment of bacterial infections. Resistance to them, importantly via production of β-lactamases, which collectively are able to hydrolyse all classes of β-lactams, threatens their continued widespread use. Bicyclic boronates show potential as broad spectrum inhibitors of the mechanistically distinct serine- (SBL) and metallo- (MBL) β-lactamase families., Methods: Using biophysical methods, including crystallographic analysis, we have investigated the binding mode of bicyclic boronates to clinically important β-lactamases. Induction experiments and agar-based MIC screening against MDR-Enterobacteriaceae (n = 132) were used to evaluate induction properties and the in vitro efficacy of a bicyclic boronate in combination with meropenem., Results: Crystallographic analysis of a bicyclic boronate in complex with AmpC from Pseudomonas aeruginosa reveals it binds to form a tetrahedral boronate species. Microbiological studies on the clinical coverage (in combination with meropenem) and induction of β-lactamases by bicyclic boronates further support the promise of such compounds as broad spectrum β-lactamase inhibitors., Conclusions: Together with reported studies on the structural basis of their inhibition of class A, B and D β-lactamases, biophysical studies, including crystallographic analysis, support the proposal that bicyclic boronates mimic tetrahedral intermediates common to SBL and MBL catalysis., General Significance: Bicyclic boronates are a new generation of broad spectrum inhibitors of both SBLs and MBLs., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
41. Studies on spiro[4.5]decanone prolyl hydroxylase domain inhibitors.
- Author
-
Holt-Martyn JP, Tumber A, Rahman MZ, Lippl K, Figg W Jr, McDonough MA, Chowdhury R, and Schofield CJ
- Abstract
The 2-oxoglutarate (2OG) dependent hypoxia inducible factor (HIF) prolyl hydroxylases (PHDs) are targets for treatment of anaemia and other ischaemia related diseases. PHD inhibitors are in clinical trials; however, the number of reported templates for PHD inhibition is limited. We report structure-activity relationship and crystallographic studies on spiro[4.5]decanone containing PHD inhibitors. Together with other studies, our results reveal spiro[4.5]decanones as useful templates for generation of potent and selective 2OG oxygenase inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
42. Crystal structures of VIM-1 complexes explain active site heterogeneity in VIM-class metallo-β-lactamases.
- Author
-
Salimraj R, Hinchliffe P, Kosmopoulou M, Tyrrell JM, Brem J, van Berkel SS, Verma A, Owens RJ, McDonough MA, Walsh TR, Schofield CJ, and Spencer J
- Subjects
- Binding Sites, Catalytic Domain, Crystallography, X-Ray, Protein Binding, Protein Conformation, Substrate Specificity, beta-Lactamases chemistry, beta-Lactamases metabolism, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Enterobacteriaceae enzymology, Meropenem chemistry, Meropenem metabolism
- Abstract
Metallo-β-Lactamases (MBLs) protect bacteria from almost all β-lactam antibiotics. Verona integron-encoded MBL (VIM) enzymes are among the most clinically important MBLs, with VIM-1 increasing in carbapenem-resistant Enterobacteriaceae (Escherichia coli, Klebsiella pneumoniae) that are among the hardest bacterial pathogens to treat. VIM enzymes display sequence variation at residues (224 and 228) that in related MBLs are conserved and participate in substrate binding. How they accommodate this variability, while retaining catalytic efficiency against a broad substrate range, has remained unclear. Here, we present crystal structures of VIM-1 and its complexes with a substrate-mimicking thioenolate inhibitor, ML302F, that restores meropenem activity against a range of VIM-1 producing clinical strains, and the hydrolysed product of the carbapenem meropenem. Comparison of these two structures identifies a water-mediated hydrogen bond, between the carboxylate group of substrate/inhibitor and the backbone carbonyl of the active site zinc ligand Cys221, that is common to both complexes. Structural comparisons show that the responsible Cys221-bound water is observed in all known VIM structures, participates in carboxylate binding with other inhibitor classes, and thus effectively replicates the role of the conserved Lys224 in analogous complexes with other MBLs. These results provide a mechanism for substrate binding that permits the variation at positions 224 and 228 that is a hallmark of VIM MBLs. ENZYMES: EC 3.5.2.6 DATABASES: Co-ordinates and structure factors for protein structures described in this manuscript have been deposited in the Protein Data Bank (www.rcsb.org/pdb) with accession codes 5N5G (VIM-1), 5N5H (VIM-1:ML302F complex) and 5N5I (VIM-1-hydrolysed meropenem complex)., (© 2018 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2019
- Full Text
- View/download PDF
43. Biosynthesis of histone messenger RNA employs a specific 3' end endonuclease.
- Author
-
Pettinati I, Grzechnik P, Ribeiro de Almeida C, Brem J, McDonough MA, Dhir S, Proudfoot NJ, and Schofield CJ
- Subjects
- Amino Acid Sequence, Binding Sites, Cloning, Molecular, Crystallography, X-Ray, Endoribonucleases genetics, Endoribonucleases metabolism, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, HEK293 Cells, HeLa Cells, Histones genetics, Humans, Hydrolases, Kinetics, Models, Molecular, Mutagenesis, Site-Directed, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, RNA, Messenger genetics, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, S Phase Cell Cycle Checkpoints, Sequence Alignment, Sequence Homology, Amino Acid, Substrate Specificity, beta-Lactamases chemistry, beta-Lactamases genetics, beta-Lactamases metabolism, Endoribonucleases chemistry, Histones biosynthesis, RNA, Messenger biosynthesis
- Abstract
Replication-dependent (RD) core histone mRNA produced during S-phase is the only known metazoan protein-coding mRNA presenting a 3' stem-loop instead of the otherwise universal polyA tail. A metallo β-lactamase (MBL) fold enzyme, cleavage and polyadenylation specificity factor 73 (CPSF73), is proposed to be the sole endonuclease responsible for 3' end processing of both mRNA classes. We report cellular, genetic, biochemical, substrate selectivity, and crystallographic studies providing evidence that an additional endoribonuclease, MBL domain containing protein 1 (MBLAC1), is selective for 3' processing of RD histone pre-mRNA during the S-phase of the cell cycle. Depletion of MBLAC1 in cells significantly affects cell cycle progression thus identifying MBLAC1 as a new type of S-phase-specific cancer target., Competing Interests: IP, PG, CR, JB, MM, SD, CS No competing interests declared, NP Reviewing editor, eLife, (© 2018, Pettinati et al.)
- Published
- 2018
- Full Text
- View/download PDF
44. Born to sense: biophysical analyses of the oxygen sensing prolyl hydroxylase from the simplest animal Trichoplax adhaerens .
- Author
-
Lippl K, Boleininger A, McDonough MA, Abboud MI, Tarhonskaya H, Chowdhury R, Loenarz C, and Schofield CJ
- Abstract
Background: In humans and other animals, the chronic hypoxic response is mediated by hypoxia inducible transcription factors (HIFs) which regulate the expression of genes that counteract the effects of limiting oxygen. Prolyl hydroxylases (PHDs) act as hypoxia sensors for the HIF system in organisms ranging from humans to the simplest animal Trichoplax adhaerens ., Methods: We report structural and biochemical studies on the T. adhaerens HIF prolyl hydroxylase ( Ta PHD) that inform about the evolution of hypoxia sensing in animals., Results: High resolution crystal structures (≤1.3 Å) of Ta PHD, with and without its HIFα substrate, reveal remarkable conservation of key active site elements between T. adhaerens and human PHDs, which also manifest in kinetic comparisons., Conclusion: Conserved structural features of Ta PHD and human PHDs include those apparently enabling the slow binding/reaction of oxygen with the active site Fe(II), the formation of a stable 2-oxoglutarate complex, and a stereoelectronically promoted change in conformation of the hydroxylated proline-residue. Comparison of substrate selectivity between the human PHDs and Ta PHD provides insights into the selectivity determinants of HIF binding by the PHDs, and into the evolution of the multiple HIFs and PHDs present in higher animals., Competing Interests: Disclosure The authors report no conflicts of interest in this work.
- Published
- 2018
- Full Text
- View/download PDF
45. Structure activity relationship studies on rhodanines and derived enethiol inhibitors of metallo-β-lactamases.
- Author
-
Zhang D, Markoulides MS, Stepanovs D, Rydzik AM, El-Hussein A, Bon C, Kamps JJAG, Umland KD, Collins PM, Cahill ST, Wang DY, von Delft F, Brem J, McDonough MA, and Schofield CJ
- Subjects
- Enediynes chemistry, Inhibitory Concentration 50, Molecular Structure, Rhodanine chemical synthesis, Rhodanine pharmacology, Structure-Activity Relationship, Sulfhydryl Compounds pharmacology, beta-Lactamase Inhibitors pharmacology, beta-Lactamases drug effects, Rhodanine chemistry, Sulfhydryl Compounds chemistry, beta-Lactamase Inhibitors chemical synthesis, beta-Lactamases chemistry
- Abstract
Metallo-β-lactamases (MBLs) enable bacterial resistance to almost all classes of β-lactam antibiotics. We report studies on enethiol containing MBL inhibitors, which were prepared by rhodanine hydrolysis. The enethiols inhibit MBLs from different subclasses. Crystallographic analyses reveal that the enethiol sulphur displaces the di-Zn(II) ion bridging 'hydrolytic' water. In some, but not all, cases biophysical analyses provide evidence that rhodanine/enethiol inhibition involves formation of a ternary MBL enethiol rhodanine complex. The results demonstrate how low molecular weight active site Zn(II) chelating compounds can inhibit a range of clinically relevant MBLs and provide additional evidence for the potential of rhodanines to be hydrolysed to potent inhibitors of MBL protein fold and, maybe, other metallo-enzymes, perhaps contributing to the complex biological effects of rhodanines. The results imply that any medicinal chemistry studies employing rhodanines (and related scaffolds) as inhibitors should as a matter of course include testing of their hydrolysis products., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. YcfD RM is a thermophilic oxygen-dependent ribosomal protein uL16 oxygenase.
- Author
-
Sekirnik R, Wilkins SE, Bush J, Tarhonskaya H, Münzel M, Hussein A, Flashman E, Mohammed S, McDonough MA, Loenarz C, and Schofield CJ
- Subjects
- Enzyme Stability, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Hydroxylation, Mixed Function Oxygenases chemistry, Mixed Function Oxygenases genetics, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Rhodothermus genetics, Ribosomal Proteins chemistry, Ribosomal Proteins genetics, Sequence Homology, Escherichia coli Proteins metabolism, Mixed Function Oxygenases metabolism, Rhodothermus enzymology, Ribosomal Proteins metabolism
- Abstract
YcfD from Escherichia coli is a homologue of the human ribosomal oxygenases NO66 and MINA53, which catalyse histidyl-hydroxylation of the 60S subunit and affect cellular proliferation (Ge et al., Nat Chem Biol 12:960-962, 2012). Bioinformatic analysis identified a potential homologue of ycfD in the thermophilic bacterium Rhodothermus marinus (ycfD
RM ). We describe studies on the characterization of ycfDRM , which is a functional 2OG oxygenase catalysing (2S,3R)-hydroxylation of the ribosomal protein uL16 at R82, and which is active at significantly higher temperatures than previously reported for any other 2OG oxygenase. Recombinant ycfDRM manifests high thermostability (Tm 84 °C) and activity at higher temperatures (Topt 55 °C) than ycfDEC (Tm 50.6 °C, Topt 40 °C). Mass spectrometric studies on purified R. marinus ribosomal proteins demonstrate a temperature-dependent variation in uL16 hydroxylation. Kinetic studies of oxygen dependence suggest that dioxygen availability can be a limiting factor for ycfDRM catalysis at high temperatures, consistent with incomplete uL16 hydroxylation observed in R. marinus cells. Overall, the results that extend the known range of ribosomal hydroxylation, reveal the potential for ycfD-catalysed hydroxylation to be regulated by temperature/dioxygen availability, and that thermophilic 2OG oxygenases are of interest from a biocatalytic perspective.- Published
- 2018
- Full Text
- View/download PDF
47. In Silico Fragment-Based Design Identifies Subfamily B1 Metallo-β-lactamase Inhibitors.
- Author
-
Cain R, Brem J, Zollman D, McDonough MA, Johnson RM, Spencer J, Makena A, Abboud MI, Cahill S, Lee SY, McHugh PJ, Schofield CJ, and Fishwick CWG
- Subjects
- Drug Evaluation, Preclinical, Models, Molecular, Protein Conformation, Structure-Activity Relationship, beta-Lactamases chemistry, Anti-Bacterial Agents chemistry, Anti-Bacterial Agents pharmacology, Computer Simulation, Drug Design, beta-Lactamase Inhibitors chemistry, beta-Lactamase Inhibitors pharmacology, beta-Lactamases metabolism
- Abstract
Zinc ion-dependent β-lactamases (MBLs) catalyze the hydrolysis of almost all β-lactam antibiotics and resist the action of clinically available β-lactamase inhibitors. We report how application of in silico fragment-based molecular design employing thiol-mediated metal anchorage leads to potent MBL inhibitors. The new inhibitors manifest potent inhibition of clinically important B1 subfamily MBLs, including the widespread NDM-1, IMP-1, and VIM-2 enzymes; with lower potency, some of them also inhibit clinically relevant Class A and D serine-β-lactamases. The inhibitors show selectivity for bacterial MBL enzymes compared to that for human MBL fold nucleases. Cocrystallization of one inhibitor, which shows potentiation of Meropenem activity against MBL-expressing Enterobacteriaceae, with VIM-2 reveals an unexpected binding mode, involving interactions with residues from conserved active site bordering loops.
- Published
- 2018
- Full Text
- View/download PDF
48. 13 C-Carbamylation as a mechanistic probe for the inhibition of class D β-lactamases by avibactam and halide ions.
- Author
-
Lohans CT, Wang DY, Jorgensen C, Cahill ST, Clifton IJ, McDonough MA, Oswin HP, Spencer J, Domene C, Claridge TDW, Brem J, and Schofield CJ
- Subjects
- Azabicyclo Compounds chemistry, Carbon Isotopes, Crystallography, X-Ray, Halogens chemistry, Ions chemistry, Ions pharmacology, Models, Molecular, Molecular Conformation, beta-Lactamase Inhibitors chemistry, Azabicyclo Compounds pharmacology, Halogens pharmacology, beta-Lactamase Inhibitors pharmacology, beta-Lactamases metabolism
- Abstract
The class D (OXA) serine β-lactamases are a major cause of resistance to β-lactam antibiotics. The class D enzymes are unique amongst β-lactamases because they have a carbamylated lysine that acts as a general acid/base in catalysis. Previous crystallographic studies led to the proposal that β-lactamase inhibitor avibactam targets OXA enzymes in part by promoting decarbamylation. Similarly, halide ions are proposed to inhibit OXA enzymes via decarbamylation. NMR analyses, in which the carbamylated lysines of OXA-10, -23 and -48 were
13 C-labelled, indicate that reaction with avibactam does not ablate lysine carbamylation in solution. While halide ions did not decarbamylate the13 C-labelled OXA enzymes in the absence of substrate or inhibitor, avibactam-treated OXA enzymes were susceptible to decarbamylation mediated by halide ions, suggesting halide ions may inhibit OXA enzymes by promoting decarbamylation of acyl-enzyme complex. Crystal structures of the OXA-10 avibactam complex were obtained with bromide, iodide, and sodium ions bound between Trp-154 and Lys-70. Structures were also obtained wherein bromide and iodide ions occupy the position expected for the 'hydrolytic water' molecule. In contrast with some solution studies, Lys-70 was decarbamylated in these structures. These results reveal clear differences between crystallographic and solution studies on the interaction of class D β-lactamases with avibactam and halides, and demonstrate the utility of13 C-NMR for studying lysine carbamylation in solution.- Published
- 2017
- Full Text
- View/download PDF
49. Crystallographic analyses of isoquinoline complexes reveal a new mode of metallo-β-lactamase inhibition.
- Author
-
Li GB, Brem J, Lesniak R, Abboud MI, Lohans CT, Clifton IJ, Yang SY, Jiménez-Castellanos JC, Avison MB, Spencer J, McDonough MA, and Schofield CJ
- Subjects
- Crystallography, X-Ray, Dose-Response Relationship, Drug, Enterobacter aerogenes enzymology, Escherichia coli enzymology, Isoquinolines analysis, Klebsiella pneumoniae enzymology, Models, Molecular, Molecular Conformation, Structure-Activity Relationship, beta-Lactamase Inhibitors analysis, Isoquinolines pharmacology, beta-Lactamase Inhibitors pharmacology, beta-Lactamases metabolism
- Abstract
Crystallographic analyses of the VIM-5 metallo-β-lactamase (MBL) with isoquinoline inhibitors reveal non zinc ion binding modes. Comparison with other MBL-inhibitor structures directed addition of a zinc-binding thiol enabling identification of potent B1 MBL inhibitors. The inhibitors potentiate meropenem activity against clinical isolates harboring MBLs.
- Published
- 2017
- Full Text
- View/download PDF
50. Structural and stereoelectronic insights into oxygenase-catalyzed formation of ethylene from 2-oxoglutarate.
- Author
-
Zhang Z, Smart TJ, Choi H, Hardy F, Lohans CT, Abboud MI, Richardson MSW, Paton RS, McDonough MA, and Schofield CJ
- Subjects
- Bacterial Proteins metabolism, Catalysis, Crystallography, X-Ray, Ethylenes metabolism, Ketoglutaric Acids metabolism, Lyases metabolism, Bacterial Proteins chemistry, Ethylenes chemistry, Ketoglutaric Acids chemistry, Lyases chemistry, Models, Chemical, Pseudomonas syringae enzymology
- Abstract
Ethylene is important in industry and biological signaling. In plants, ethylene is produced by oxidation of 1-aminocyclopropane-1-carboxylic acid, as catalyzed by 1-aminocyclopropane-1-carboxylic acid oxidase. Bacteria catalyze ethylene production, but via the four-electron oxidation of 2-oxoglutarate to give ethylene in an arginine-dependent reaction. Crystallographic and biochemical studies on the Pseudomonas syringae ethylene-forming enzyme reveal a branched mechanism. In one branch, an apparently typical 2-oxoglutarate oxygenase reaction to give succinate, carbon dioxide, and sometimes pyrroline-5-carboxylate occurs. Alternatively, Grob-type oxidative fragmentation of a 2-oxoglutarate-derived intermediate occurs to give ethylene and carbon dioxide. Crystallographic and quantum chemical studies reveal that fragmentation to give ethylene is promoted by binding of l-arginine in a nonoxidized conformation and of 2-oxoglutarate in an unprecedented high-energy conformation that favors ethylene, relative to succinate formation., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.