Open Access Article
Mariska
de Munnik
a,
Pauline A.
Lang
a,
Francisco
De Dios Anton
b,
Mónica
Cacho
b,
Robert H.
Bates
b,
Jürgen
Brem‡
a,
Beatriz
Rodríguez Miquel
*b and
Christopher J.
Schofield
*a
aChemistry Research Laboratory, Department of Chemistry, the Ineos Oxford Institute of Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK. E-mail: christopher.schofield@chem.ox.ac.uk
bTres Cantos Medicines Development Campus, GlaxoSmithKline, Calle Severo Ochoa 2, Tres Cantos, Madrid, Spain. E-mail: beatriz.rodriguez@gsk.com
First published on 30th May 2023
Disruption of bacterial cell wall biosynthesis in Mycobacterium tuberculosis is a promising target for treating tuberculosis. The L,D-transpeptidase LdtMt2, which is responsible for the formation of 3 → 3 cross-links in the cell wall peptidoglycan, has been identified as essential for M. tuberculosis virulence. We optimised a high-throughput assay for LdtMt2, and screened a targeted library of ∼10
000 electrophilic compounds. Potent inhibitor classes were identified, including established (e.g., β-lactams) and unexplored covalently reacting electrophilic groups (e.g., cyanamides). Protein-observed mass spectrometric studies reveal most classes to react covalently and irreversibly with the LdtMt2 catalytic cysteine (Cys354). Crystallographic analyses of seven representative inhibitors reveal induced fit involving a loop enclosing the LdtMt2 active site. Several of the identified compounds have a bactericidal effect on M. tuberculosis within macrophages, one with an MIC50 value of ∼1 μM. The results provide leads for the development of new covalently reaction inhibitors of LdtMt2 and other nucleophilic cysteine enzymes.
β-Lactams are the most widely used antibacterials, though historically they have not been deemed to be generally useful for TB treatment, in part due to resistance mediated by the chromosomally encoded M. tuberculosis serine β-lactamase BlaC.4,5 β-Lactams are covalently reacting inhibitors of the transpeptidases (penicillin binding proteins, PBPs) that catalyse the essential meso-Dap-D-Ala (4 → 3) cross-linking transpeptidation step in peptidoglycan biosynthesis.6 A mechanistically related class of enzymes, the L,D-transpeptidases (Ldts), also catalyses the formation of cross-links, but rather than 4 → 3 cross-links the Ldts produce meso-Dap-meso-Dap (3 → 3) cross-links.7 By contrast with PBPs, which employ a nucleophilic serine-residue in catalysis, the Ldts employ a nucleophilic cysteine-residue.7,8 While only low levels of 3 → 3 cross-linking are evident in the exponential growth phase of M. tuberculosis, they comprise ∼80% of all cross-links in the stationary phase.9 LdtMt2 in particular has been identified as being essential for M. tuberculosis virulence, making it an attractive target for TB treatment.10
The extramembrane section of LdtMt2 consists of three domains, two of which have an immunoglobulin-related fold, and the catalytic domain which has an ErfK/YbiS/YhnG fold (Fig. 1A).11 The LdtMt2 active site within the catalytic domain is bordered by a flexible loop (residues 300–323), sometimes referred to as the active site “lid”, which creates two entrances to the active site: the inner and outer cavities.11 The Ldt mechanism is proposed to be analogous to that proposed for the PBPs, although employing a catalytic triad analogous to that observed in some cysteine-proteases, involving Cys354, His336 and Ser337, for the formation of the cross-link between the donor and acceptor stem substrates (Fig. 1A and B).12 The formation of a negatively charged tetrahedral enzyme-substrate intermediate is proposed to be stabilised by an oxyanion hole, consisting of the backbone NH groups of His352, Gly353 and Cys354 (Fig. 1A).13
![]() | ||
| Fig. 1 The structure and function of LdtMt2. (A) Views from a crystal structure of LdtMt2 (PDB 6RLG),19 showing entrances to the active site, the catalytic triad (consisting of residues Cys354, His336 and Ser337) and the oxyanion hole (consisting of the backbone NHs of His352, Gly353 and Cys354). The two LdtMt2 immunoglobulin-like domains are in teal and cyan, the catalytic domain is in grey, the active site “lid” is in green, the catalytic triad is in yellow, and the oxyanion hole is in red. (B) Outline mechanism for reaction of LdtMt2 and the disaccharide tetrapeptide monomer of the M. tuberculosis peptidoglycan. The LdtMt2 catalytic triad residues are in black, the meso-Dap residue of the donor substrate in green, and the meso-Dap residue of the acceptor substrate in red. | ||
In general, the Ldts are not reported to be efficiently inhibited by β-lactams, however, their nucleophilic cysteine reacts with the β-lactam rings of penems and carbapenems to form acyl–enzyme complexes.14–16 Efforts have been made to identify β-lactams which more efficiently inhibit LdtMt2, though to date with limited success.17,18 In an alternative strategy for LdtMt2 inhibition, we have reported on the inhibition of LdtMt2 by cysteine-reactive reagents such as ebselen.19 The covalent modification of nucleophilic cysteine residues is a validated method for clinically useful inhibition of various cysteine proteases, including the main protease (Mpro) of SARS-CoV-2, highlighting a need for new functional groups targeting nucleophilic cysteine residues.20–22
Here we report on the development of a fluorescence based high-throughput screen (HTS) for LdtMt2 inhibition and its application to screen a library of potential nucleophilic cysteine reacting compounds. The results reveal the discovery of new mechanism-based nucleophilic cysteine enzyme targeting inhibitors.
The assay manifested good tolerance for DMSO concentrations of ≤3% (v/v) (Fig. S2F†). Stock solutions of LdtMt2 and probe 1 in assay buffer were apparently stable over 7 hours when the reagents were kept on ice (Table S2†). No evidence for nonspecific plate patterns was observed during the first hour after reaction initiation under the optimised conditions (Fig. S3†). The assay was validated in dose–response analyses of independent repeats in quadruplicate using a set of 24 tool compounds with known inhibitory activity for LdtMt2 (Table S3†); the results showed high reproducibility and good correlation with reported values.19,23
A previously optimised and validated β-lactamase assay26 was optimised for our assay set-up with BlaC, which we envisioned as a secondary assay for the HTS output. Cross titrations showed the optimal BlaC concentration to be 2.5 nM and the optimal probe concentration to be 2.5 μM in an assay volume of 10 μL (Fig. S4†), leading to a robust Z′ of 0.84. A dose–response assay with the set of 24 tool compounds was performed in quadruplicate independent repeats to validate the assay, showing high reproducibility (Table S3†).
000 electrophilic compounds was constructed from the GlaxoSmithKline high-throughput screening (GSK HTS) collection, including compounds with a β-lactam core, cysteine protease inhibitors, serine protease inhibitors, and compounds containing known nucleophilic cysteine warheads (Fig. 2). These compounds were assessed for inhibition of LdtMt2 with two independent repeats at a single concentration of 100 μM. The results identified 733 potentially active compounds manifesting >78% apparent inhibition (an overall hit rate of 7.7%). The identified compounds were then subjected to dose–response analyses with LdtMt2 and BlaC (two independent repeats at concentrations 1.7 nM to 100 μM). In an effort to reduce false positives, an interference assay was performed, wherein the inhibitors were added after the completion of the reaction between LdtMt2 and probe 1 (5 hours at room temperature). Compounds with a pIC50 value >4.0 in the interference assay were excluded from further analyses. Compounds were then prioritised and selected for further studies based on their observed inhibitory potency for LdtMt2 and structural diversity, employing protein observed MS. The results for 39 selected compounds (1–39) are summarised in Table S5.†
![]() | ||
| Fig. 3 Representative examples of hits for the eight identified LdtMt2 inhibitor classes. The core electrophilic motif is shown in teal. | ||
Notably, two of the identified LdtMt2 inhibitors belong to the cephalosporin subclass of β-lactams (19, 20); this observation is interesting because cephalosporins have previously been deemed not to be of use for inhibition of LdtMt2, potentially because their stereochemistry mimicks the D-Ala-D-Ala substrate of PBPs rather than that of Ldts, which accept a substrate with C-terminal L,D-chirality.23,27,28 However, the LdtMt2 inhibition observed with cephalosporins 19–20 (pIC50 5.55–5.62) is comparable to that observed for the carbapenems biapenem, doripenem and meropenem under the same assay conditions (pIC50 5.54–5.87, Table S4†). The results thus imply that the cephalosporin scaffold is of considerable interest for inhibition of LdtMt2 and hence TB treatment. While most compounds showed no significant inhibition of BlaC, cephalosporin 20 was a potent inhibitor (pIC50 7.72, Fig. S9†).
The SPE-MS results suggest that α-chloro ketones 1–2 react with LdtMt2 with loss of a chloride ion, consistent with a nucleophilic substitution involving Cys354 (Fig. S10† entries 1–2). The maleimide derivatives 3–7 reacted with LdtMt2 without fragmentation, resulting in adducts comprising the intact inhibitor mass (Fig. S10† entries 3–7). Over the course of 24 hours, partial hydrolysis of the ester sidechain of 7 was observed, as manifested by a mass decrease of 106 Da (relative to the modified LdtMt2, Fig. S10† entry 7).
Acrylamide 8 (MW 436 Da), a trans-dichloro substituted Michael acceptor, reacted to give covalently modified LdtMt2 (+399 Da), corresponding to the addition of a single molecule accompanied with loss of one chloride ion (Fig. S10† entry 8). 9 (MW 306 Da) covalently modified LdtMt2 (+322 Da, Fig. S10† entry 9), corresponding to the mass of the inhibitor plus an additional 16 Da. A further +16 Da adduct was observed to accumulate over 21 hours, potentially reflecting slow oxidation of the Cys354 sulphur derived thioester following reaction with the inhibitor. While acrylamides 10–12 are structurally closely related, only 10 and 11 were observed to manifest substantial modification of LdtMt2 (+186 and +298 for 10 and +286 Da for 11, Fig. S10† entries 10–11). By contrast, reaction with 12 (MW 268 Da) manifested only low amounts of a +268 Da LdtMt2 modification (Fig. S10† entry 12).
Although the fumaryl amides 13 and 14 (MW 225 and 204 Da, respectively) contain a potential Michael acceptor, the SPE-MS experiments solely showed +207 Da and +189 Da adducts on reaction of LdtMt2 with 13 and 14, respectively, in agreement with substitution of the terminal amide resulting in loss of NH3 (Fig. S10† entries 13–14). Ebsulfur analogue 15 (MW 264 Da) reacted without apparent fragmentation to give a +264 Da adduct (Fig. S10† entry 15). Isatin derivatives 16 and 17 (MW 404 and 390 Da, respectively) showed only low levels of modification of LdtMt2 manifesting +404 and +390 Da mass increments, respectively, but addition of 2 molecules of 17 was observed (Fig. S10† entries 16, 17). 18 (MW 366 Da) was not observed to modify LdtMt2 (Fig. S10† entry 18). β-Lactams 19–21 exhibited complex fragmentation patterns, which were observed to vary with time and inhibitor concentration (Fig. S10† entries 19–21). However, a +626 Da adduct, corresponding to the addition of a non-fragmented molecule of 20 (MW 626 Da), dominated at lower inhibitor to enzyme ratios.
In thermal shift assays32 stabilisation by isatins 16–18 was observed, increasing the melting temperature of LdtMt2 by 2.4–3 °C (Fig. 4D and S12B†). By contrast, the other hits did not show a substantial change in the melting temperatures of LdtMt2, apart from nitriles 26 and 36, which lowered the melting temperature by 2.1 and 3.0 °C, respectively (Table S5†). The reversibility of isatin binding was assessed by jump dilution studies.33 While inhibition of the inhibitors 1–11, 13–15 and 19–39 was found to be irreversible, inhibition by isatins 16–18 was reversible with koff values of 103 s−1, 84.6 s−1, and 160 s−1, respectively, resulting in half-lives of the LdtMt2 inhibitor complexes of 24.6 s, 29.4 s, and 15.6 s, respectively (Fig. S12C, Table S7†). Dose–response assays at varying inhibitor pre-incubation times (ranging between 0 min and 2 h) manifested no time-dependency for potency of inhibition by the isatins 16–18 (Fig. S12D, Table S8†). The reversible binding mode of the isatins, either via reversible noncovalent binding, or reversible covalent binding involving reaction of the nucleophilic Cys354 with the C3 isatin ketone, differ from most of the classes identified, which involve irreversible inhibition via alkylation, substitution, or acylation mechanisms.
Most of the identified cyanamide inhibitors apparently reacted once with LdtMt2, predominantly with Cys354; however, 24, 33, 34, 35 and 37 were observed to react with LdtMt2 multiple times (Fig. S13 and S14†). The latter observation likely relates to reaction with non-cysteine residues, as evidenced by SPE-MS experiments with the LdtMt2-ebselen complex (Fig. 5B and S14†). The precise reason for the differences in reactivity of the cyanamides is unknown, but it is notable that 33, 34 and 35 all contain a pyridine ring, and 24 has an analogous pyridazine ring. The extent of cyanation increased with increasing pH (as shown with 22, 27, 31 and 36 at pH 5.5–7.5, Fig. S15†). Cyanation appeared to be stable for ∼6 hours, after which time backbone cleavage of LdtMt2 at Cys354 was observed, as previously reported for the known cysteine cyanating agents 2-nitro-5-thiocyanobenzoic acid (NTCB) and 1-cyano-4-dimethylaminopyridinium tetrafluoroborate (CDAP) (Fig. 5).34,35 However, β-elimination, which is reported as a side reaction to protein cleavage by NTCB and CDAP,35,36 was not observed. Additionally, cleavage of methionine residues, as reported for treatment of proteins with cyanogen bromine (Fig. 5D),37 was not observed.
The cyanating activity of 22–36 was compared to a selection of known electrophilic cyanation agents, including N-based cyanating reagents 40–43 (including CDAP) and S-based cyanating reagents NTCB (44) and CDTP (45) (Table S10†).38,39 SPE-MS studies on LdtMt2 reaction with 40–44 showed comparable results to those for cyanamides 22–36, with predominantly single- or double-cyanation being observed, while 45 did not modify LdtMt2 (Fig. S16†). The amount of cleavage of LdtMt2 at Cys354 was comparable for 22–36 and 40–44 (Fig. S17†). While most of the cyanamides 22–36 potently inhibited LdtMt2 (pIC50 values >7.0), cyanating agents 40–44 were apparently less potent inhibitors (pIC50 values 5.3–6.5), while 45 was inactive (Table S10†). The intrinsic thiol reactivity of 40–42 was generally lower (kchem 0.90 M−1 s−1, 0.90 M−1 s−1, and 3.69 M−1 s−1, respectively, Table S10†) than observed with 22–36 (exceptions being 28 and 32). Due to assay interference, the intrinsic thiol reactivities of for 43–45 could not be determined.
The intrinsic reactivity (kchem) with a thiol group was assessed for all the selected compounds 1–39 (Table S5†). Following the same principle as used for the assay determining kinact/KI, a modified form of a previously reported high-throughput endpoint assay,42 based on competition for reaction with glutathione between an irreversibly reacting covalent probe and test compounds, was used employing probe 2 (Fig. S20A†).43 The assay was validated using a small number of thiol-reactive compounds (Fig. S20B†), which show good correlation with reported kchem values (Fig. S20C, Table S11†).42 The irreversibility of the reaction of glutathione with probe 2 was shown over the duration of the measurements (>24 hours) using a displacement assay with N-ethylmaleimide (Fig. S20D and E†), as previously reported.42
The majority of the compounds showed low to moderate intrinsic thiol (glutathione) reactivity under the tested conditions (kchem < 0.08–13.0 M−1 s−1). The maleimide derivatives (3–7) exhibited relatively higher kchem values (34.2–129.9 M−1 s−1), as anticipated based on their known reactivity as alkylating agents and application in thiol-coupling reactions.44,45 A selection of the nitrile derivatives, i.e. those containing a cyanamide functional group (22–36), also displayed relatively high kchem values (2.12–61.6 M−1 s−1). Comparison of the obtained kinact/KI values with the kchem values, implies an increased reactivity for LdtMt2 compared to the intrinsic thiol reactivity for the majority of inhibitors (Fig. 4C), implying that with appropriate derivatisation, selective inhibition of LdtMt2 should be possible.46
Overall, the LdtMt2 fold in the structures of the complexes strongly resembles that of unmodified LdtMt2 (PDB 6RLG;19 main chain RMSD: 0.54 Å, 0.37 Å, 0.45 Å, 0.86 Å, 0.7103 Å, and 0.77 Å, for unmodified LdtMt2 compared to LdtMt2 reacted with 2, 3, 4, 8, 13, and 15, respectively). As a notable exception, substantial variation of the position of the active site lid (residues 300–323) was observed, resulting in apparent reorientation of various active site residues involved in the binding of the inhibitors, in particular of Tyr308, Met303 and Tyr318 (Fig. 6).
![]() | ||
| Fig. 6 Inhibitor binding induces conformational changes in the active site “lid” (residues 300–323) of LdtMt2, resulting in reorientation of certain active site residues, in particular Met303, Tyr308, and Tyr318. Superimposition of unmodified LdtMt2 (dark grey, PDB 6RLG)19 and LdtMt2 reacted with 2 (yellow, PDB 8A1L), 4 (teal, PDB 8A1M), 8 (green, PDB 8A1O) and 13 (red, PDB 8A1N). Note that the conformation of Cys354 (shown only for unmodified LdtMt2) is very similar in each complex. | ||
While the overall folds of the two LdtMt2 molecules in the structure of the LdtMt2-2 complex overlay well (main chain RMDS: 0.72 Å), the reacted 2 is observed in two distinct binding modes (Fig. 7). In the conformation observed in chain B, the ester group of 2 is positioned to engage in polar interactions with the sidechains of Tyr308 (3.5 Å) and Tyr318 (2.7 Å). These residues are disordered in chain A, an observation which can be rationalised by a potential steric clash with the phenyl ring of 2 in chain A. In both binding modes, the carbonyl group of 2 is positioned in the oxyanion hole (the respective distances to the backbone nitrogens of His352, Gly353 and Cys354 in chain A are 3.9 Å, 3.4 Å and 3.4 Å and in chain B are 3.6 Å, 3.0 Å and 3.1 Å). The sec-butyl- and phenyl-groups of the inhibitor are oriented towards a hydrophobic pocket made up of the active site residues: Met303, Thr320, Val333, Phe334, His336, Trp340, and His352.
![]() | ||
| Fig. 7 Views from a crystal structure derived by reaction of LdtMt2 with α-chloromethyl ketone 2 (yellow, PDB 8A1L). The immunoglobulin-like domains are in teal and cyan. The catalytic domain is grey, with the active site lid in green. Two different conformations of LdtMt2-2 were observed in chains A and B. mF0 − DFc polder OMIT maps59 are contoured at 3.0σ, carved around Cys354 bound 2 (shown in grey mesh). Polar interactions are shown in dark grey dashes. The crystallographically assigned product is consistent with that observed by mass spectrometry (Fig. S10†). | ||
The maleimides 3 and 4 are structurally related, but their binding modes differ (Fig. 8). While the sulfone group on the N-phenyl ring of 4 engages in polar interactions with Tyr318 (3.5 Å, Fig. 8B), the N-phenyl ring of 3 is positioned for hydrophobic interactions with Met303, Tyr318, Thr320, and Trp340 (Fig. 8A). In both binding modes, one of the maleimide carbonyls is positioned in the oxyanion hole (the respective distances to the backbone nitrogens of His352, Gly353 and Cys354 for 3 are 3.4 Å, 3.5 Å and 3.0 Å and for 4 are 2.9 Å, 3.4 Å and 3.4 Å). Additional polar interactions were observed between the second maleimide carbonyl of 4 and the phenolic OH of Tyr318 (2.9 Å). Despite differences in their binding modes, both maleimides bind to LdtMt2 with the same stereochemical outcome, that is forming the (R)-enantiomer.
![]() | ||
| Fig. 8 Views from crystal structures of the complex derived by reaction of LdtMt2 with maleimides 3 and 4. (A) Active site view of LdtMt2 reacted with 3 (teal; PDB 8A1J). (B) Active site view of LdtMt2 reacted with 4 (cyan; PDB 8A1M). (C) Superimposition of LdtMt2 complexes with 3 (teal) and 4 (cyan). Despite close structural similarity between 3 and 4, different conformations were observed, with variations in the positions of active site “lid” residues Met303, Tyr308 and Tyr318 (dark green for the LdtMt2-3 complex, light green for the LdtMt2-4 complex). Active site views from (A), (B) and (C) are shown as observed in chain A. mF0 − DFc polder OMIT maps59 are contoured at 3.0σ, and carved around Cys354 bound 3 and 4. (shown in grey mesh). Polar interactions are shown in dark grey dashes. The crystallographically assigned products are consistent with those observed by mass spectrometry (Fig. S10†). | ||
By contrast with the other LdtMt2 structures reported here, a crystal structure of LdtMt2 in complex with acrylamide 8 was solved in the P21212 space group with a single molecule in the ASU. Inhibitor 8 contains an asymmetric Michael acceptor warhead and has the potential to covalently react with LdtMt2 either via Michael addition to its acrylamide or via reaction of its vinyl sulphone group. The obtained structure implies that LdtMt2 reacts regioselectively by Michael reaction with the vinyl sulphone of 8 (Fig. 9A). Polar interactions were observed between the amide carbonyl and backbone nitrogen of His352 (2.8 Å) and the amide nitrogen and the side chain of His336 (3.4 Å). The amide carbonyl of 8 is positioned in the oxyanion hole (the respective distances to the backbone nitrogens of His352, Gly353 and Cys354: 2.8 Å, 3.5 Å and 3.5 Å).
![]() | ||
| Fig. 9 Views from crystal structures of the complex derived by reaction of LdtMt2 with inhibitors 8, 13, 15 and 31. (A) Active site view of LdtMt2 reacted with 8 (cyan, PDB 8A1O). (B) Active site view of LdtMt2 reacted with 13 (teal, PDB 8A1N). (C) Active site view of LdtMt2 reacted with 15 (cyan; PDB 8A1K). (D) Superimposition view from crystal structures of LdtMt2 in complex with 15 (LdtMt2-bound 15 in cyan, active site “lid” in dark green; PDB 8A1K) and in complex with ebselen (LdtMt2-bound ebselen in teal, active site “lid” in light green; PDB 6RRM).19. (E) Active site view of LdtMt2 reacted with 31 (cyan; PDB 8AHO). Active site views in (A–E) are shown as observed in chain A. mF0 − DFc polder OMIT maps59 contoured at 3.0σ, carved around Cys354 bound 8, 13, 15 and 31 are shown in grey mesh. Polar interactions are shown in grey dashes. The crystallographically assigned products are consistent with those observed by mass spectrometry (Fig. S10 and S13†). | ||
A structure of LdtMt2 reacted with 13 manifests clear additional electron density at the LdtMt2 active site in chain A and partial density in chain B. 13 was thus only modelled in chain A. Notably, analysis of the electron density in chain A showed that, in accord with the SPE-MS experiments, 13 covalently modifies LdtMt2 through reaction of Cys354 with its terminal amide, rather than by Michael reaction with its fumaryl group (Fig. 9B). Whilst in solution 13 exists as a mixture of E/Z isomers, the obtained structure implies (at least predominantly) selective reaction of LdtMt2 with the (Z)-stereoisomer of 13. The nitrogen of 13 is positioned for polar interaction with the Cys354-13 acyl (2.4 Å), and the Cys354-13 acyl carbonyl group is positioned in the oxyanion hole (the respective distances of the inhibitor carbonyl oxygen to the backbone nitrogens of His352, Gly353 and Cys354: 3.7 Å, 3.7 Å and 3.0 Å).
A structure of LdtMt2 reacted with the ebsulfur analogue 15 revealed the inhibitor covalently bonded to Cys354 via reaction of the N–S bond, analogous to the reaction of LdtMt2 with ebselen.19 The inhibitor hydroxy group of the species is positioned to engage in hydrogen-boding interactions with the backbone carbonyl of His352 (3.4 Å) and the inhibitor amide with the sidechain of Tyr318 (2.8 Å, Fig. 9C). Comparison of the binding mode of 15 with that of ebselen (PDB 6RRM),19 implies that while both compounds are positioned similarly in the active site, the phenyl ring of ebselen is positioned for π-interactions with Tyr318, while the hydroxy group of 15 is positioned for polar interactions with the backbone carbonyl of His352 (Fig. 9D), accompanied by reorientation of the active site lid.
A crystal structure of LdtMt2 reacted with cyanamide 31 manifested covalent modification of Cys354 with formation of a carbamoyl group, likely resulting from initial formation of cyanated cysteine followed by hydrolysis (Fig. 9E). The carbamoyl group was directed towards the oxyanion hole (the respective distances of the carbonyl oxygen to the backbone nitrogens of His352, Gly353 and Cys354: 3.6 Å, 3.5 Å and 3.1 Å). Further polar interactions were observed between the nitrogen of the carbamoyl group and the sidechain of Tyr318 (3.4 Å in chain A) and the backbone carbonyl of Gly332 (3.4 Å in chain A).
The results of our HTS for inhibitors of LdtMt2 with ∼10
000 electrophilic compounds identified multiple classes of compounds that react covalently with LdtMt2, that is: α-chloro ketones, maleimides, acrylamides, fumaryl amides, an ebsulfur analogue, isatins, nitriles (among which a subset of cyanamides), and β-lactams. Several of these functional groups are present in human drugs.48 The results show that unexpected reaction modes can occur as observed, amongst other compounds, for the fumaryl amides 13 and 14. Among the most potent identified LdtMt2 inhibitors were the ebsulfur analogue 15 (pIC50 7.99) and the cyanamides 22–36 (pIC50 6.39–7.53).
In most cases inhibition can be related to irreversible covalent reaction with the nucleophilic Cys354, as evidenced by protein observed MS analyses, with an exception being the isatins, which likely react via a reversible covalent mechanism. X-ray crystallographic results suggests that, in at least some cases, induced fit occurs during inhibitor binding. In particular, differences in the conformation of the loop that borders the active site (residues 300–323) are observed, leading to variations in the positions of some active site residues (in particular Tyr308, Tyr318 and Met303) that modulate both polar and hydrophobic interactions with the inhibitors. These observations are in accord with previous observations regarding changes in the conformation of the flexible loop upon binding of β-lactams and ebselen.13,14,19 At present, the exact relationship between inhibitor reactions and binding and changes in the conformation of the flexible loop and related residues remain unclear. However, the previous reported structural data13,14,19 combined with data reported here forms a basis for detailed biophysical and modelling studies to understand the role of the flexible loop in LdtMt2 catalysis and inhibition.
Efforts to inhibit LdtMt2 have thus far focussed on inhibition by β-lactams, in particular by the carbapenem subclass.14–18 In our HTS campaign we aimed to build on the observation that LdtMt2 can be inhibited by alternative types of cysteine-reactive compounds.19 Indeed, the majority of hit compounds that we identified are non-β-lactam covalent inhibitors of LdtMt2. Importantly, we identified LdtMt2 inhibitors from the cephalosporin subclass of β-lactams, which have previously been deemed to be ineffective in the inhibition of LdtMt2.23,27 The identified cephalosporins manifested pIC50 values comparable to the observed potencies of carbapenems such as meropenem, doripenem and biapenem (which were included in the HTS as control compounds). This is a notable observation, given that cephalosporins are very widely used antibacterials that are, unlike the intravenously used carbapenems, orally active.49
Of the LdtMt2 inhibitor classes identified here, the cyanamides 22–36 are of particular mechanistic interest, as they are electrophilic agents that cyanate Cys354 with high inhibitory potency. The cyanamide series exhibit increased reactivity over the reported cyanating agents 40–45,34,35,38,39 whilst most derivatives remain selective for reaction with Cys354. Interestingly, in some cases we observed hydrolysis of the S-nitrile group to give an S-carbamoyl group, as validated by crystallography in the case of 31. Additionally, we observed cleavage of LdtMt2 following S-cyanation. Such cleavage likely occurs via 5-exo-dig cyclisation of the Cys354 amide nitrogen onto the carbon of the S-nitrile group, followed by hydrolysis on the N-terminal side of the cysteine residue.50 Although not the focus of our work, the results presented here suggest that developing cyanamide type reagents for cleavage at cysteine residues in proteins should be possible. Although it may be challenging to develop cyanamides as drugs due to their reactivity, we also identified nitrile inhibitors (38, 39). The potential of nitriles for the inhibition of nucleophilic cysteine enzymes is highlighted by the recent development of cysteine reacting inhibitor of the main protease (Mpro) of SARS-CoV-2.21
The HTS approach is validated by the observation that out of the 39 selected hit compounds, 11 were found to have a bactericidal effect on M. tuberculosis in macrophages. The most potent identified compound was the cyanamide 30 (MIC50 of 0.98 μM). Due to environmental factors, such as phagosomal acidification and presence of reactive nitrogen species, M. tuberculosis residing in macrophages is typically in the nonreplicating or replication-inhibited phase.51–53 The observed bactericidal effect of the selected hit compounds on M. tuberculosis within macrophages, but not M. tuberculosis under replicating conditions, is thus in accord with the anticipated behaviour of LdtMt2 inhibition, which is proposed to be essential during the nonreplicating stage.9,10 However, further research is necessary to confirm the mechanism of action of these compounds.
000 electrophilic compounds was constructed from the GSK compound collection of ∼4 million compounds, by means of 21 different substructure searches using Smart Queries, and a molecular weight filter (MW < 450). Substructures consisting of known, or potential cysteine, binding warheads were identified from literature searches, and datasets from other cysteine and serine protease programmes within GSK.
| Y = (Y∞ − Y0)[1 − e(−kobs×t)] + Y0 | (1) |
The (kinact/KI)probe was derived from kobs using eqn (2), yielding a value of 39.8 M−1 s−1.
| kobs = (kinact/KI)probe[probe 2] | (2) |
The half-life (t1/2) of probe 2 (10 μM), was determined using eqn (3) to be 28.7 min. Therefore, an incubation time of 2.4 h should ensure <97% receptor occupancy. With this knowledge, an incubation time of 3 h was selected for the determination of (kinact/KI)inhibitor values.
t1/2 = ln 2/kobs | (3) |
LdtMt2 (100 nM) added to a mixture of varying concentrations of inhibitor (400 μM to 20.3 nM) for 10 min in assay buffer (50 mM HEPES, pH 7.2, 0.01% (v/v) Triton X-100) and probe 2 (10 μM), and incubated for 3 h prior to determination of the fluorescence intensity using a PHERAstar plate reader (BMG Labtech) with λex = 480 nm and λem = 520 nm. Dose–response analyses were performed using a variable slope nonlinear regression model in GraphPad Prism 9 (GraphPad Software). The (kinact/KI)inhibitor values were derived from the IC50 values using eqn (4).
![]() | (4) |
| kobs = kprobe[probe 2] | (5) |
Inhibitors were serially diluted (by 1/3) across columns 2–10 of a 96-well plate (starting concentration 10 mM). The final two columns of the microplate (11 and 12) were used as inhibitor-free controls and contained only DMSO. To a 384-well plate was added assay buffer (14 μL) using a MultiDrop Combi dispenser (ThermoFisher Scientific). The inhibitor or blank (1 μL) solutions were added using a CyBio liquid handling system (Analytik Jena AG), with 4 replicates per inhibitor concentration. To each well was added probe 2 (5 μL, final concentration 10 μM) using a MultiDrop Combi dispenser (ThermoFisher Scientific). L-Glutathione (5 μL, final concentration 500 nM, final volume 25 μL) was added and the plate was sealed and incubated for 15 h at room temperature. The fluorescence signal was measured using a PHERAstar plate reader (BMG Labtech) with λex = 480 nm and λem = 520 nm and normalised against the mean average of no inhibitor controls and the mean average of no enzyme controls. Nonlinear regression analyses of dose–response were conducted using GraphPad Prism 9 (GraphPad Software) with a variable slope model. The kchem of inhibitors was calculated from the obtained IC50 value using eqn (6).
![]() | (6) |
![]() | (7) |
The half-life of the enzyme–inhibitor complex (t1/2) was calculated using eqn (8).
![]() | (8) |
:
1 with a single cell suspension of Mtb H37Rv-Luc cells. After incubation, infected cells were washed four times to remove extracellular bacilli and resuspended (2 × 105 cells per mL) in RPMI medium supplemented with 10% FBS (Hyclone), 2 mM L-glutamine and pyruvate and dispensed in white, flat bottom 384-well plates (Greiner) in a final volume of 50 μL containing test compounds (11 serial dilutions of 1/3 with starting concentration 100 μM, max. 0.5% DMSO). Plates were incubated for 5 days under 5% CO2 atmosphere, 37 °C, 80% relative humidity before growth assessment. The Bright-Glo™ Luciferase Assay System (Promega, Madison, WI) was used as cell growth indicator for the Mtb H37Rv-Luc strain. Luminescence was measured in an Envision Multilabel Plate Reader (PerkinElmer) using the opaque 384-plate Ultra-Sensitive luminescence mode, with a measurement time of 50 ms. A reduction in light production was considered growth inhibition and the IC50 or IC90 value was interpolated from the dose response curve.
Footnotes |
| † Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d2sc06858c |
| ‡ Present address: Janssen Research & Development, Janssen Pharmaceutica, B-2340 Beerse, Belgium. |
| This journal is © The Royal Society of Chemistry 2023 |