A Phenotypic Based Target Screening Approach ... - ACS Publications

May 5, 2017 - binding enzymes from different sources (Escherichia coli, rabbit muscle, and .... for competitive inhibition (eq 2)22 with Origin 8 soft...
0 downloads 0 Views 2MB Size
Subscriber access provided by Eastern Michigan University | Bruce T. Halle Library

Article

A phenotypic based target screening approach delivers new antitubercular CTP synthetase inhibitors Marta Esposito, Sára Szadocka, Giulia Degiacomi, Beatrice Silvia Orena, Giorgia Mori, Valentina Piano, Francesca Boldrin, Julia Zemanova, Stanislav Huszár, David Barros, Sean Ekins, Joël Lelièvre, Riccardo Manganelli, Andrea Mattevi, Maria Rosalia Pasca, Giovanna Riccardi, Lluis Ballell, Katarína Mikušová, and Laurent R. Chiarelli ACS Infect. Dis., Just Accepted Manuscript • Publication Date (Web): 05 May 2017 Downloaded from http://pubs.acs.org on May 6, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Infectious Diseases is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

A phenotypic based target screening approach delivers new antitubercular CTP synthetase inhibitors.

Marta Esposito†+, Sára Szadocka‡+, Giulia Degiacomi#, Beatrice S. Orena†, Giorgia Mori†, Valentina Piano†, Francesca Boldrin#, Júlia Zemanová‡, Stanislav Huszár‡, David Barros§, Sean Ekins∆⊥, Joel Lelièvre§, Riccardo Manganelli#, Andrea Mattevi†, Maria Rosalia Pasca†, Giovanna Riccardi†, Lluis Ballell§, Katarína Mikušová‡, Laurent R. Chiarelli†*.



Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, via

Ferrata 9, 27100 Pavia, Italy ‡

Department of Biochemistry, Faculty of Natural Sciences, Comenius University in

Bratislava, Mlynská dolina CH1, 84215 Bratislava, Slovakia. #

Department of Molecular Medicine, University of Padova, via Gabelli 63, 35121 Padova,

Italy. §

Diseases of the Developing World, GlaxoSmithKline, Calle Severo Ochoa 2, 28760 Tres

Cantos, Madrid, Spain ∆

Collaborative Drug Discovery Inc., 1633 Bayshore Highway, Suite 342, Burlingame, CA

94010, USA.

E-mail: [email protected]

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Despite its great potential the target-based approach has been mostly unsuccessful in tuberculosis drug discovery, while whole cell phenotypic screening has delivered several active compounds. However, for many of these hits the cellular target has not yet been identified, thus preventing further target-based optimization of the compounds. In this context, the newly validated drug target CTP synthetase PyrG was exploited to assess a target-based approach of already known, but untargeted, antimycobacterial compounds. To this purpose the publically available GlaxoSmithKline antimycobacterial compound set was assayed, uncovering a series of 4-(pyridin-2-yl)thiazole derivatives which efficiently inhibit the Mycobacterium tuberculosis PyrG enzyme activity, one of them showing low activity against the human CTP synthetase. The three best compounds were ATP binding site competitive inhibitors, with Ki values ranging from 3 to 20 µM, but did not show any activity against a small panel of different prokaryotic and eukaryotic kinases, thus demonstrating specificity for the CTP synthetases. Metabolic labelling experiments, demonstrated that the compounds directly interfere not only with CTP biosynthesis, but also with other CTP dependent biochemical pathways, such as lipid biosynthesis. Moreover, using a M. tuberculosis pyrG conditional knock-down strain, it was shown that the activity of two compounds is dependent on the intracellular concentration of the CTP synthetase. All these results strongly suggest a role of PyrG as a target of these compounds, thus strengthening the value of this kind of approach for the identification of new scaffolds for drug development.

Keywords: drug discovery; phenotypic screening; target-based screening; Mycobacterium tuberculosis; CTP synthetase; pyridine-thiazole.

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Although there has been extensive investment over the last few decades, tuberculosis (TB) still remains an urgent global health issue, with more than 10.4 million estimated new cases and about 1.8 million deaths each year1. Moreover, the increasing spread of Mycobacterium tuberculosis (Mtb) multidrug resistant (MDR) strains (with about 480,000 cases in 2015) of which 10% have been reported as extremely drug resistant (XDR), has led to an increased requirement for new drugs, with new mechanisms of action2. After the first complete sequencing of the Mtb genome, there were great expectations derived from target-based approach for TB drug discovery which proved to be mostly unsuccessful. High-throughput screening (HTS) has been the preferential method adopted for the identification of new hits from large compound libraries3. In the case of Mtb, wholecell screening using HTS can provide compounds able to cross the mycobacterial cell envelope, overcoming one of the main challenges for TB drugs. The large number of phenotypic screening efforts performed until now led to the identification of several active molecules, however, the mechanism of action of many of them is still not known4. The elucidation of the cellular target is an important component of the hit to lead optimization process, enabling the assistance of biochemistry and structural biology to characterize the mechanism of action and to better assess at molecular level potential host cell toxicities. However, for several reasons (e. g. difficulty in obtaining resistant mutants etc.) this task is not trivial4. A great improvement arose from the introduction of next generation sequencing platforms, allowing wide and intensive use of genomics in combination with chemical library screening, for identification of the cellular target of novel compounds4, 5. Furthermore, it is possible to combine target-based and phenotypic screening strategies6. In this context, GlaxoSmithKline (GSK) has identified and released the structures and activities of a large number of anti-mycobacterials (https://www.ebi.ac.uk/chembl/)7 verified to be active

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

against M. tuberculosis H37Rv cells and showing low toxicity against human cell-lines8, 9. Different screens of this set allowed the identification of the cellular targets of several hits, not only by the classical genomic-based target assignment10, but also through different strategies11. For instance, a new series of leads targeting DprE1 has been recently identified through the screening of this set against a dprE1 over-expressing strain of Mycobacterium bovis BCG12. Moreover, since new drug candidates should possess a novel mechanism of action to ensure effectiveness against Mtb MDR and XDR strains2,13, the drug discovery exploitation of the latest validated targets seems a reasonable starting point. In this respect, the novel TB drug target CTP synthetase PyrG14 could represent a good platform for target based screening of antitubercular compound libraries. This essential enzyme was demonstrated to be druggable, and, being involved in several biochemical pathways, its inhibition has proven to affect several aspects of mycobacterial physiology14. With this aim, we exploited the GSK antimycobacterial compound set (GSK TB-set) library to explore the possibility of finding new antitubercular CTP synthetase inhibitors.

Results and Discussion Screening of the GSK TB-set against PyrG affords new inhibitors A previous whole cell phenotypic screening of GSK compounds identified a set of 177 molecules (GSK TB-set) active against M. tuberculosis cell growth, whose targets are largely unknown8. For this reason, to identify compounds targeting PyrG, the GSK TB-set was assayed against the recombinant M. tuberculosis enzyme. The assays, performed at a final concentration of 100 µM for each compound, and at subsaturating concentrations of ATP, identified 16 compounds (9% hit rate) inhibiting more than 50% of enzymatic activity (Table 1). ACS Paragon Plus Environment

Page 4 of 36

Page 5 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Table 1: compounds of the GSK TB-set showing activity against M. tuberculosis PyrG PyrG

PyrG GSK ID

structure

inhibition GSK ID

structure

a

inhibition (%)a

(%) 1570606A

90.8

735816A

62.6

735826A

79.2

829969A

60.5

920684A

74.3

275984A

59.5

1611550A

66.9

345724A

53.8

1729177A

66.8

547543A

55.1

1668869A

66.7

921190A

52.2

445886A

68.1

636544A

51.1

920703A

60.0

2200160A

51.1

a

Enzyme assays were performed in the presence of 100 µM of each compound

About half of these active compounds, in particular the most active, were characterized by a 4-(pyridin-2-yl)thiazole group, suggesting a particular affinity of PyrG for this moiety. Among them, GSK1570606A (1), GSK920684A (2), and GSK735826A (3) (Table 1), (Mtb Minimal Inhibitory Concentration, MIC: 16 µM, 7.6 µM and 1.4 µM, respectively) showed ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

higher inhibitory effects against PyrG activity (IC50 values of 2.9 ± 0.6 µM, 23.3 ± 2.2 µM and 19.7 ± 2.2 µM, for compounds (1) (2) and (3), respectively), and were selected for further investigation. Firstly, in an attempt to gain insight into the binding between PyrG and the three inhibitors, a computational analysis of the possible poses of the compounds was performed by docking in the PyrG ATP binding site14 (Fig. 1) using Discovery Studio 4.1 (Biovia, San Diego, CA). The superimposition with the UTP molecule shows a partial overlap. The 4(pyridin-2-yl)thiazole moiety is in a comparable position for (2) and (3), while (1) has a flipped orientation in the protein. However, all 3 molecules have phenyl rings that are suggested to pi-stack with Arg223. This superimposition is very similar to that of the 11426026 compound, a previously described PyrG inhibitor, with a IC50 value (35 µM) similar to that of the three GSK compounds, which has been demonstrated to be competitive towards the ATP binding site14.

ACS Paragon Plus Environment

Page 6 of 36

Page 7 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 1. Docking GSK compounds in CTP synthetase. (A) compound (1) libdock score 88.16; (B) compound (2) libdock score 92.96; (C) compound (3) libdock score 87.23. All molecules (grey) are compared to UTP (Yellow); (D) compound 11426026, previously reported as a PyrG ATP binding site competitive inhibitor.

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

To confirm the hypothesis that the compounds bind to the ATP site, PyrG steady state kinetic analysis was performed in the presence of different concentrations of the compounds. The analysis revealed that all the compounds behave as competitive inhibitors towards the ATP binding site, showing apparent Ki values of 3.5±0.4 µM (1); 22.0±0.6 µM (2); and 16.3±0.5 µM (3) (Fig. 2). By contrast, all of them were uncompetitive toward the pyrimidine nucleotide binding site (Fig. S1).

Figure 2. Double reciprocal plots of PyrG kinetic analysis towards ATP, in the presence of different concentrations of compound (1), (2) and (3) (panel A, B and C, respectively).

Finally, as the CTP synthetases physiologically use glutamine as an ammonia donor to fulfill the catalysis, the effects of the three compounds against the glutaminase activity of PyrG were evaluated. As depicted in Fig. S2, the presence of the three compounds leads to a decrease in the Vmax values, but leaving unchanged the Km, thus excluding a direct interaction of the compounds with the glutamine binding site.

Effects of GSK PyrG inhibitors against human CTP synthetase Due to the relatively large similarity between Mtb and human CTP synthetases (about 44% sequence identity), the type 1 human CTP synthetase (Hu-CTPS1) was obtained, to ACS Paragon Plus Environment

Page 8 of 36

Page 9 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

assess the compounds’ specificity. The recombinant Hu-CTPS1 was produced using Pichia pastoris expression system and purified as described in Methods (Fig. S3), affording about 3 mg of protein from 1 liter of yeast culture, with a specific activity of 6.5 s-1, and Km values of 0.2 mM towards either UTP or ATP. The enzymatic activity assay of Hu-CTPS1 in the presence of the compounds, performed at sub-saturating substrate concentrations, revealed that all the three 4-(pyridin-2-yl)thiazole derivatives inhibited the human CTP synthetase (Fig. 3). By contrast, the 11426026 compound, a Mtb PyrG inhibitor previously identified14, displayed a significantly higher IC50 against Hu-CTPS1 (>1 mM for Hu-CTPS1 vs 13.5 ± 1.3 µM for the Mtb PyrG). However, although compounds (2) and (3) showed IC50 values against Hu-CTPS1 (38.0 ± 3.1 µM and 24.2 ± 3.7 µM, for (2) and (3), respectively), similar to that against PyrG, compound (1) was a weaker inhibitor of the human enzyme, with an IC50 10-fold higher (27.8 ± 2.3 µM).

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 3. Effects of PyrG inhibitors on human CTP synthetase 1 activity. IC50 determination of 11426026 compound (panel A) and GSK compounds (1) (panel B), (2) (panel C) and (3) (panel D) on the M. tuberculosis PyrG (filled symbols) and Hu-CTPS1 (open symbols).

The possibility to screen compound libraries against both Mtb and human CTPsynthetases could be a valuable “double tool” for identification of inhibitors affecting almost exclusively mycobacterial PyrG, such as 11426026. It is worth noting that, despite their activity against the human enzyme, the GSK compounds showed low toxicity against a human cell-line8. Moreover, the fact that compound (1) is tenfold less active toward the human enzyme, highlights the possibility to develop analogs that are more specific for the mycobacterial CTP synthetase. ACS Paragon Plus Environment

Page 10 of 36

Page 11 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Validation of PyrG as a target of the three GSK inhibitors As the GSK compounds have been shown to be ATP binding site inhibitors, their specificity towards the CTP synthetases was assessed. In order to do this, the compounds were assayed against a small panel of diverse kinases or ATP binding enzymes, from different sources (Escherichia coli, rabbit muscle, and human pyruvate kinases, human adenylate kinase, M. tuberculosis phosphoribosyl pyrophosphate synthetase, human phosphoglycerate kinase, yeast hexokinase). As shown in table S1, none of these enzymes is inhibited by the three compounds, thus excluding a broad specificity of these compounds. To confirm the role of PyrG as a target of the compounds, the isolation of Mtb resistant mutants was attempted, to check for the presence of possible mutations in the pyrG gene. Concentrations of compounds ranging from 5-fold to 20-fold the MIC were used, but despite several attempts no resistant colonies could be isolated. The inability to obtain spontaneous resistant mutants could be related to the strict essentiality of the CTP synthetase enzyme14,15, and clearly reflects a very low mutation frequency of the pyrG gene, reinforcing its significance as a drug target. However, as the three compounds were demonstrated to compete with the ATP binding site of PyrG, mutations that could hamper their binding, but without detrimental effects on the enzyme activity, could confer drug resistance. For this reason, the GSK compounds were also tested on a previously characterized M. tuberculosis mutant, harboring the V186G mutation in PyrG, and resistant to the already described 11426026 compound, an ATP binding site PyrG competitive inhibitor14. The Mtb mutant strain did not show cross resistance with compound (3) and was weakly resistant to the other two compounds (MIC values 2-fold higher than that of the wild type) (Table S2). The three compounds assayed against the recombinant PyrG V186G, were found to maintain activity against the mutant enzyme,

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

showing IC50 values slightly increased with respect to the wild type, thus explaining the sensitivity of the Mtb mutant strain (Fig. S4). To confirm that also the intracellular mechanism of action of the compounds effectively involves the CTP synthetase, the compounds were assayed against an Mtb conditional knock-down (cKD) mutant strain (TB456), constructed using the Pip-ON inducible system. In this system pyrG gene was under the control of the inducible promoter Pptr and its expression was allowed only in the presence of the inducer (Pristinamycin I). The conditional mutant TB456 was able to grow only on plates containing Pristinamycin I (100 ng/ml), confirming the essentiality of the gene14 (Fig. S5). Moreover, TB456 growth was shown to be induced in a dose-dependent manner in the presence of different amounts of Pristinamycin I in liquid media. Standing cultures of TB456 were grown using different concentrations of inducer (0 to 100 ng/ml) for 72 hours; then bacteria were diluted to OD540 0.05 in the same media conditions. After 72 hours it was clearly visible that the mutant growth depended on Pristinamycin concentration. After the second re-fresh the growth of bacteria exposed to the lowest concentrations of Pristinamycin I was arrested (Fig. S6) Then, TB456 cultures grown in different Pristinamycin I concentrations were exposed to the compounds object of the study, and to 11426026, which is known to target PyrG, as control (Table 2). Actually, the MIC for compound (2) did not show any variation in function of Pi amount in which the bacteria were grown. On the contrary the MICs for compounds (1), as well as to 11426026, were halved at lower concentration of the inducer, increasing only at the highest Pi concentration. Furthermore, in the presence of compound (3) the TB456 strain did not grow at minimal Pi concentrations, showing a four- fold lower MIC value even at the highest inducer. The dependence of the effects of compound (1) and particularly of compound (3) on the PyrG intracellular level demonstrate its role in the mechanism of their action, thus supporting the hypothesis that PyrG is an intracellular target of these two compounds. ACS Paragon Plus Environment

Page 12 of 36

Page 13 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Table 2: MIC values of Mtb wild-type and TB456 cKD strains.

Compound

H37Rv

M. tuberculosis MIC (µ µM) TB456 [pristinamycin] (ng/ml)

0 5 (1) 20 n.d. 10 (2) 38 n.d. 38 (3) 4.3 n.d. n.d. 11426026 42.5 n.d. 21.2 n.d.: not determined because of lack of cells growth

15 10 38 1.1 21.2

25 10 38 1.1 21.2

50 20 38 1.1 42.5

GSK PyrG inhibitors cause several metabolism alterations in M. tuberculosis The role of the CTP synthetase in the mechanism of action of these compounds was also examined by metabolic labeling of the nonpathogenic model strain M. tuberculosis H37Ra (MIC of 20.8 µM, 15.2 µM and 0.28 µM, for compound (1), (2) and (3), respectively) with [14C]-uracil. Within mycobacterial cells [14C]-uracil is incorporated into [14C]UMP by uracil phosphoribosyltransferase(Upp) from the pyrimidine salvage pathway16, being then further metabolized to the whole range of nucleotides. M. tuberculosis H37Ra cells were grown in the absence or presence of the compounds (8 x MIC) for one hour, then [14C]-uracil was added and radiolabeling continued for three hours. The radiolabeled nucleotides were then extracted from cell pellets with 9% formic acid17, and separated by thin layer chromatography. As depicted in Figure 4A, addition of the compounds to the media during cultivation of mycobacteria led to a significant increase in the ratios of radioactivity incorporated to [14C]UTP and [14C]CTP compared to the control, pointing to inhibition of PyrG. A similar trend was found in the positive control 11426026, while for benzothiazinone BTZ 043, the drug with an unrelated mode of action targeting cell wall enzyme DprE118, the [14C]UTP/[14C]CTP ratio was comparable to the untreated control cells (Table S3).

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Clearly, inhibition of CTP synthetase activity would affect numerous catabolic and anabolic reactions in mycobacteria, due to the requirement of CTP by bifunctional CoaBC enzyme involved in biosynthesis of coenzyme A. This enzyme was recently shown as a validated bactericidal target in M. tuberculosis19. CTP is critical for lipid metabolism20 also for producing of activated CDP-derivatives required for biosynthesis of phospholipids. We thus investigated the effects of the compounds on lipid biosynthesis by [14C]-acetate metabolic labeling in the same conditions as described for [14C]-uracil. Analysis of lipids extracted from the radiolabeled cells revealed that incorporation of [14C]-acetate was decreased, in the cells treated with compounds (1), (2), (3) and 11426026. On the contrary, the presence of BTZ043 resulted in an increase of the radiolabel incorporation due to accumulation of TDM, as expected (Fig. 4 B and C). These experiments confirm that the effects of the compounds are pleiotropic, as expected, but it is not possible to conclude, if they are the result of only PyrG inhibition, or due to targeting additional enzyme(s).

ACS Paragon Plus Environment

Page 14 of 36

Page 15 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 4. Examination of effects of (1), (2) and (3) on M. tuberculosis H37Ra cells by metabolic labeling. Bacterial cells were pre-treated with 8-fold MIC of the drugs for 1 h, followed by 3 h of radiolabeling with [14C]uracil (A) or [14C]-acetate (B, C). (A) Ratios of radioactivity incorporated to [14C]UTP and [14C]CTP extracted from [14C]uracil-labeled cells. Nucleotide extracts were separated by TLC on PEI-cellulose plate. Columns and error bars represent mean ± SD of triplicate measurement of radioactivity in the [14C]UTP and [14C]CTP spots cut from TLC plates corresponding to two separate experiments; from each experiment two plates were analyzed. The autoradiograph is representative image from three separate experiments. (B) TLC analysis of the lipids from [14C]-acetate-labeled cells. Lipids were extracted as in “Methods”, aliquots corresponding to the same amounts of extracted material (as shown in 5C) were loaded on a silica gel TLC plate and ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

separated in Solvent I: CHCl3/CH3OH/NH4OH/H2O (65:25:0.5:4); Solvent II CHCl3/CH3OH/H2O (20:4:0.5); and Solvent III: petroleum ether/ethyl acetate (98:2; 3 times).The plates were exposed to an autoradiography film at −80 °C for 6 days. TDM, trehalosedimycolates; TMM, trehalosemonomycolates; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PIM, phosphatidylinositol mannosides; CL, cardiolipin; TAG, triacylglycerol. Autoradiographs are representative images from three separate experiments. (C) Visualization of the cold lipids by cupric sulfate staining.

Conclusion The use of whole cell phenotypic screening in combination with target based assay has been recently demonstrated a useful strategy for the identification of new leads12. With this aim an antitubercular compound library was screened against CTP synthetase PyrG, identifying a new series of 4-(pyridin-2-yl)thiazole compounds, able to inhibit the enzyme activity. These compounds, which have been shown to be quite specific for the CTP synthetases, confirm the high druggability of PyrG. Indeed, several lines of evidence, which emerged from the different approaches used, point to PyrG as a main target of this series, although further additional targets could not be completely excluded, demonstrating the effectiveness of such a screen against newly validated targets, to identify novel scaffolds for drug development. Moreover, the parallel screen against the corresponding human enzyme CTPS-1, could help in defining the selectivity of the compounds. Indeed, among the three hit molecules identified, compound (1) was more selective for the mycobacterial enzyme and represents a useful scaffold for further target-assisted structure activity relationship studies in order to improve its efficacy.

ACS Paragon Plus Environment

Page 16 of 36

Page 17 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

METHODS Compound library screening against PyrG enzyme activity. Wild-type and mutant M. tuberculosis PyrG recombinant proteins were obtained in E. coli, as previously reported 14. The publically available GlaxoSmithKline antimycobacterial compound set (GSK TB-set), consisting of 177 compounds active against M. tuberculosis growth8, was screened against PyrG activity, at a final concentration of 100 µM for each compound. PyrG enzyme activity was determined in a final volume of 100 µl at 37°C by continuous spectrophotometric assay following the rate of increase in absorbance at 291 nm due to the conversion of UTP to CTP (ε= 1.34 mM-1 cm-1)21. The reaction mixture contained: 50 mM HEPES pH 8.0, 10 mM MgCl2, 1 mM UTP, 0.2 mM ATP, 0.5 µM PyrG, 1 µl of compound (10 mM in dimethylsulfoxide, DMSO), and the reaction was started by the addition of 100 mM NH4Cl. Blank reactions were performed by adding 1 µl of DMSO. All assays were performed in triplicate. The compounds inhibiting more than 75% of PyrG activity in these conditions, and selected

for

further

investigation,

were

re-purchased

from

MolPort

(Riga,

Latvia):GSK1570606A,(2-(4-fluorophenyl)-N-(4-(pyridin-2-yl)thiazol-2-yl)acetamide, #MolPort-003-158-205;

GSK735826A,

N-(4-(pyridin-2-yl)thiazol-2-yl)-

[1,3]dioxolo[4',5':4,5]benzo[1,2-d]thiazol-6-amine #MolPort-003-038-940; GSK920684A, 2(3-fluorophenoxy)-N-(4-(pyridin-2-yl)thiazol-2-yl)acetamide, #MolPort-004-106-239. PyrG inhibition was reconfirmed, then IC50 and Ki values were determined. For IC50 determinations, the enzyme activities were measured in the presence of compound and values were estimated according to the Equation 1, where A[I] is the enzyme activity at inhibitor concentration [I] and A[0] is the enzyme activity without inhibitor. A[I] =A[0] × ቀ1- ሾIሿ+IC ቁ [I]

equation 1

50

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The inhibition constant (Ki) values were determined by assaying the PyrG enzymatic activity at different substrates and compound concentrations. The assays were performed at a UTP final concentration of 2 mM when ATP was the variable substrate, and at 2 mM ATP when UTP was the variable substrate. Ki values were calculated using an adapted equation for competitive inhibition (Equation 2)22 with Origin 8 software. v=

Vmax [S]

ሾIሿ Ki

ሾSሿ+ Km ൬1+ ൰

equation 2

PyrG glutaminase activity was assayed by using a glutamate dehydrogenase (GDH, Sigma-Aldrich) coupled assay, which converts the produced glutamic acid into αketoglutaric acid through the reduction of NAD+. Thus the activity was determined measuring the increasing in absorbance at 340 nm of the produced NADH (ε = 6220 M-1 cm-1). Assays were performed at 37°C in 50 mM Hepes pH 8.0, 10 mM MgCl2, 1 mM NAD+, 100 µM GTP, 0.2 mM ATP, 2 mM UTP, 1 U GDH, PyrG 1 µM, and reaction started by adding the glutamine solution. Steady state kinetics toward Gln were performed at 3 and 6 µM for cpd (1) and 20 and 40 µM for cpds (2) and (3).

Screening of the compounds against different kinases. E. coli pyruvate kinase, human R type pyruvate kinase, human adenylate kinase, M. tuberculosis phosphoribosyl pyrophosphate synthetase and human phosphoglycerate kinase were prepared, and their activity assayed, as previously reported23-27. Rabbit muscle pyruvate kinase and yeast hexokinase were purchased from Sigma-Aldrich, and their activity was assayed according to the manufacturer. Enzyme activities were determined at their subsaturating ATP concentrations, in the presence and in the absence of 100 µM of compound (1), (2) or (3).

ACS Paragon Plus Environment

Page 18 of 36

Page 19 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Minimal inhibitory concentration determinations and isolation of M tuberculosis spontaneous resistant mutants. The MIC of the compounds was determined in solid medium. A single colony of each M. tuberculosis strain was inoculated in complete Middlebrook 7H9 supplemented with 10% OADC Middlebrook Enrichment, and grown at 37°C until exponential growth phase (~108 CFU/ml). Dilutions to the final concentration of ~106CFU/ml were performed and about 1 µl of cell culture was streaked onto plates containing two-fold serial dilutions of appropriate compound. MIC values were assigned as the lowest drug concentrations inhibiting bacterial cell growth. All experiments were performed in triplicate. The isolation of M. tuberculosis mutants was attempted by plating ~1010 cells from an exponential growth phase wild-type culture onto 7H11 medium containing different concentrations of compounds, ranging from 5 to 20-fold the MIC. MICs of the compounds for M. tuberculosis H37Ra were established by resazurine microplate assay (REMA)28.

Construction and characterization of a pyrG conditional knock down mutant. A Mtb pyrG conditional knockdown mutant was constructed using the PIP-ON system29. At this purpose the first 694bp nucleotides of the pyrG coding sequence were amplified with RP1609

(5’-ttttatgcatcgaaagcacccgcaaacc-3’)

and

RP1610

(5’-

ttttactagtcatcaacgcaatcttgtttttca-3’) primers and cloned in a suicide plasmid in frame with the pristinamycin-inducible promoter Pptr. The obtained plasmid pGi11 was than electroporated in an H37Rv-derivative containing the pip gene integrated at the L5 attB site obtaining TB456. To characterize the growth of the pyrG cKD mutant, TB456 was grown to mid-logarithmic phase and then diluted to a theoretical OD540of 0.05 in medium with different amounts of Pristinamycin I (0,5,7, 15, 25, 50, 75, 100 ng/ml). Serial dilutions

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

of standing culture of TB456 grown in Middlebrook 7H9 without Pristinamycin I for 24-48 hrs, were plated onto solid media with/ without Pristinamycin I (100 ng/ml).

MIC determinations in pyrG conditional knock down mutant. Compounds sensitivity of the Mtb pyrG knockdown mutant was determined in presence of different pristinamycin concentrations using the resazurine microplate assay (REMA) as previously described28.

Docking. The PyrG protein was prepared as described previously14 for docking using the default settings of the ‘prepare protein’ protocol in Discovery Studio 4.1 (Biovia, San Diego, CA). The PyrG protein (PDB ID: 4ZDJ)14 was used for docking using LibDock30. The protocol included 100 hotspots and docking tolerance (0.25). The FAST conformation method was also used along with steepest descent minimization with CHARMm.

Metabolic labeling of M. tuberculosis H37Ra. The effects of the PyrG inhibitors on the mycobacterial metabolism were assessed through metabolic labelling of M. tuberculosis H37Ra with [14C]-uracil, or [14C]-acetate. Briefly, M. tuberculosis H37Ra was grown statically in 7H9 medium supplemented with 10% ADC and 0.05% Tween 80 until OD600~0.25-0.35. The culture was then divided to 5 ml aliquots and the compounds (1), (2) and (3) dissolved in DMSO were added at 8x MIC; control drugs 11426026 and BTZ043 were added at 16 µg/ml or 0.4 µg/ml, respectively. Final concentration of DMSO in all cultures was 1%. After 1 hr drug treatment [14C]-uracil (American Radiolabeled Chemicals, specific activity 53 mCi/mmol) was added to a final concentration 0.5 or 1 µCi/ml, or [14C]acetate (American Radiolabeled Chemicals, specific activity 106 mCi/mmol) was added to a final concentration 0.5 µCi/ml, and the incubation continued for next three hours.

ACS Paragon Plus Environment

Page 20 of 36

Page 21 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Inhibition of mycobacterial growth by all tested compounds after 4 hr treatment was between 2 to 10 % based on OD600 measurement. Extraction of [14C]-uracil-labeled nucleotides with ice-cold 9% formic acid was carried out as previously reported14. For TLC analysis the extracts were evaporated under vacuum, mixed with 10 nmol each of UTP and CTP and loaded on PEI Cellulose F plate (Millipore). After 15 min soaking in CH3OH, the air-dried TLC plate was developed in 0.75 M KH2PO4, pH adjusted to 3.5 with 0.75M H3PO417 for 3 hrs. Spots corresponding to UTP and CTP were visualized under UV (λ=254 nm), then they were cut from the plate and placed to scintillation vials with EcoLite scintillation liquid for determination of radioactivity. Bacteria labeled with [14C]-acetate were subjected to extraction with 1.5 ml CHCl3/CH3OH (1:2), followed by extraction with 1.5 ml CHCl3/CH3OH (2:1). Each extraction was carried out for 2 hrs at 56°C; the extracts were combined, dried and subjected to fractionation in CHCl3/CH3OH/H2O (4:2:1). Upper (water) phase was discarded and bottom (organic) phase was dried and dissolved in Solvent I (see below) in the ratio 75 µl/OD600 0.5/2 ml of the culture.5 µl aliquots of the lipid extracts were analyzed by TLC on Silica gel plates (Merck) in Solvent I: CHCl3/CH3OH/NH4OH/H2O (65:25:0.5:4); Solvent II CHCl3/CH3OH/H2O (20:4:0.5); and Solvent III: petroleum ether/ethyl acetate (98:2; 3 times). After chromatography the plates were exposed to autoradiography film (BioMax MR) at -80°C. For visualization of the cold lipids the plates were sprayed with cupric sulfate (10% in 8% phosphoric acid).

Cloning, expression and purification of the human CTPS1. Recombinant human CTPsynthetase 1 (HuCTPS-1) was expressed in P. pastoris MutS strain KM71H (Invitrogen)31. The full length HuCTPS-1 cDNA was purchased from Dharmacon. The cDNA was inserted into BamHI/NotI restriction sites of pPICZ-B-eGFP-6His vector, affording the pPICZ-BHuCTPS-1-eGFP-6His, to produce the recombinant protein fused with C-terminal

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

enhanced Green Fluorescent Protein (eGFP) and 6Xhistidines tags. The pPICZ-B-eGFP plasmid is a modified pPICZ-B vector (Invitrogen), in which the PreScission protease recognition sequence and the eGFP-6His tag sequence were inserted at the 3’ of the multi-cloning site. P. pastoris KM71H transformation was performed according to Lin-Cereghino et al.32, using the pPICZB-HuCTPS-1-eGFP-6His plasmid linearized with SacI (Promega). Transformed cells were plated on YPD (2% Bacto-Yeast Extract, 2% dextrose,1% peptone) agar supplemented with 100 µg/ml zeocin, and incubated at 30 °C for 2-3 days, until colonies appeared. In order to identify the clone showing the best protein expression, each colony was inoculated in a 24-deep wells plate containing 2 ml of BMGY medium (100 mM HK2PO4 pH 6.0, 6.34 % Yeast Nitrogen Base with Ammonium Sulfate without amino acids, 1 % glycerol, 4× 10–5 % biotin), incubated al 30 °C shaking at 280 rpm for 60 hours. Successively, for protein expression induction, the medium was exchanged with BMM (100 mM HK2PO4pH 6, 1.34 %Yeast Nitrogen Base with Ammonium Sulfate without amino acids, 4 x 10-50 biotin, 0.5% methanol), and eGFP fluorescence was measured after 24, 48 and 72 hours, using the Clariostar plate reader (BMG Labtech; excitation 489 nm, emission 509 nm), thus identifying the clone having the highest fluorescence signal. For large scale protein production, a pre-inoculum of the selected colony was grown overnight at 30°C and diluted 80-fold in a 5 liter flask containing 1 liter of BMGY, and incubated at 30 °C in 200 rpm shaking incubator. After 72 hours, cells were harvested by centrifugation and resuspended in half-volume of BMM medium, and 0.5% methanol was added every 24 hours. After 48 hours, cells were collected and resuspended in Buffer A (50 mM sodium phosphate pH 7.5, 500 mM NaCl, 10% glycerol, 1 mM phenyl-methyl sulfonyl fluoride (PMSF; Sigma-Aldrich), protease inhibitors Complete EDTA-Free (Roche), 1 mg/ml DNAse). An equal volume of zirconia beads (0.5 µm, BioSpech) was ACS Paragon Plus Environment

Page 22 of 36

Page 23 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

added to the suspension, and yeast cells were mechanically disrupted with a BioSpec Mini Bead-Beater. The mixture was passed through a cloth mesh straines to remove zirconia beads, and centrifuged at 70000 x g for 30 minutes at 4 °C. The supernatant was applied on a HisTrap column (1 ml, GE Healthcare) previously equilibrated in Buffer A, the column was washed with 20 mM of imidazole, and then CTPS-1 was eluted with 500 mM imidazole in the same buffer. The purified enzyme was dialyzed against Buffer B (50 mM Tris-HCl pH 7.5, 1 mM EDTA, 150 mM NaCl, 10% glycerol, 1 mM DTT),digested with PreScission protease (GE Healthcare, 400 mU/ml), and further purified by a second affinity chromatography, followed by size exclusion chromatography on a HiLoad Superdex 200 column (GE Healthcare). Samples purity was checked by SDS-PAGE and protein concentration evaluated by Bradford reagent33.

ASSOCIATED CONTENTS Supporting Information Effects of GSK compounds against different ATP binding enzymes; MIC values of M. tuberculosis wild-type and pyrG mutant strains to GSK compounds; ratios of radioactivity incorporated to [14C]UTP and [14C]CTP of the control experiments; PyrG kinetic analysis towards UTP in the presence compounds; effects of the GSK compounds against PyrG glutaminase activity; SDS-PAGE of the purification steps of HuCTPS-1; IC50 values of compounds to the wild-type and the V186G mutant PyrG enzyme; characterization of the pyrG conditional mutant TB456. This information is available via the Internet at http://pubs.acs.org/.

AUTHOR INFORMATION Corresponding Authors *(L.R.C.) E-mail: [email protected] ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Current address ⊥

S.E.: Collaborations Pharmaceuticals Inc., Fuquay Varina, NC 27526, USA

Author Contributions +

M.E. and S.S. contributed equally and are considered as co-first authors to this work.

Author Contributions M.E. and L.R.C. performed the screening of the TB-set library, provided by D.B., J.L. and L.B.; M.E. and L.R.C. performed proteins expression, purification and characterization; S.E. performed docking experiments; B.S.O. and G.M. performed MIC determinations and isolation of resistant mutants; G.D. and F.B. constructed pyrG conditional knock down mutant and performed MIC determinations; S.S., S.H. and J.Z. performed metabolic labeling experiments; M.E. and V.P. cloned the human CTPS1; R.M., D.B., J.L., M.R.P., G.R., L.B., K.M. and L.R.C. analysed data; R.M., A.M., M.R.P., G.R., K.M., L.R.C. supervised and directed the work; S.E., R.M., K.M., L.R.C. wrote the paper. All authors discussed the results and commented on the manuscript.

Competing Financial Interests SE was a consultant for Collaborative Drug Discovery, Inc. All other authors declare no competing financial interest.

ACKNOWLEDGMENTS This work was supported by European Community’s Seventh Framework Program (Grant 260872); by University of Pavia, Italy (“Universitiamo–Tubercolosi: un killer riemergente” to G.R.); by the Slovak Research and Development Agency (Contract No. DO7RP-0015-11 to K.M.). ACS Paragon Plus Environment

Page 24 of 36

Page 25 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

ABBREVIATIONS HTS, high-throughput screening; Hu-CTPS1, human CTP synthetase 1; MDR multi drug resistant; MIC, Minimal Inhibitory Concentration; Mtb, M. tuberculosis; PyrG, Mtb CTP synthetase; TB, tuberculosis; XDR, extremely drug resistant.

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

REFERENCES [1] World. Health Organization, Global tuberculosis report 2016. [2] Wallis, R. S., Maeurer, M., Mwaba, P., Chakaya, J., Rustomjee, R., Migliori, G. B., Marais, B., Schito, M., Churchyard, G., Swaminathan, S., Hoelscher, M., and Zumla, A. (2016) Tuberculosis-advances in development of new drugs, treatment regimens, host-directed therapies, and biomarkers, Lancet Infect Dis 16, e34-46. doi: 10.1016/S1473-3099(16)00070-0. [3] Bajorath, J. (2002) Integration of virtual and high-throughput screening, Nat Rev Drug Discov 1, 882-894. doi:10.1038/nrd941. [4] Lechartier, B., Rybniker, J., Zumla, A., and Cole, S. T. (2014) Tuberculosis drug discovery in the post-post-genomic era, EMBO Mol Med 6, 158-168. doi: 10.1002/emmm.201201772. [5] Chiarelli, L. R., Mori, G., Esposito, M., Orena, B. S., and Pasca, M. R. (2016) New and old hot drug targets in tuberculosis, Curr Med Chem 23, 3813-3846. doi: 10.2174/1389557516666160831164925 [6] Zuniga, E. S., Early, J., and Parish, T. (2015) The future for early-stage tuberculosis drug discovery, Future Microbiol 10, 217-229. doi: 10.2217/fmb.14.125. [7] Bento, A. P., Gaulton, A., Hersey, A., Bellis, L. J., Chambers, J., Davies, M., Krüger, F. A., Light, Y., Mak, L., McGlinchey, S., Nowotka, M., Papadatos, G., Santos, R., and Overington, J. P. (2014) The ChEMBL bioactivity database: an update, Nucleic Acids Res 42, D1083-1090. doi: 10.1093/nar/gkt1031. [8] Ballell, L., Bates, R. H., Young, R. J., Alvarez-Gomez, D., Alvarez-Ruiz, E., Barroso, V., Blanco, D., Crespo, B., Escribano, J., González, R., Lozano, S., Huss, S., SantosACS Paragon Plus Environment

Page 26 of 36

Page 27 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Villarejo, A., Martín-Plaza, J. J., Mendoza, A., Rebollo-Lopez, M. J., Remuiñan-Blanco, M., Lavandera, J. L., Pérez-Herran, E., Gamo-Benito, F. J., García-Bustos, J. F., Barros, D., Castro, J. P., and Cammack, N. (2013) Fueling open-source drug discovery: 177 small-molecule leads against tuberculosis, ChemMedChem 8, 313-321. doi: 10.1002/cmdc.201200428. [9] Rebollo-Lopez, M. J., Lelièvre, J., Alvarez-Gomez, D., Castro-Pichel, J., MartínezJiménez, F., Papadatos, G., Kumar, V., Colmenarejo, G., Mugumbate, G., Hurle, M., Barroso, V., Young, R. J., Martinez-Hoyos, M., González del Río, R., Bates, R. H., Lopez-Roman, E. M., Mendoza-Losana, A., Brown, J. R., Alvarez-Ruiz, E., MartiRenom, M. A., Overington, J. P., Cammack, N., Ballell, L., and Barros-Aguire, D. (2015) Release of 50 new, drug-like compounds and their computational target predictions for open source anti-tubercular drug discovery, PLoS One 10, e0142293. doi: 10.1371/journal.pone.0142293. [10] Abrahams, K. A., Chung, C. W., Ghidelli-Disse, S., Rullas, J., Rebollo-López, M. J., Gurcha, S. S., Cox, J. A., Mendoza, A., Jiménez-Navarro, E., Martínez-Martínez, M. S., Neu, M., Shillings, A., Homes, P., Argyrou, A., Casanueva, R., Loman, N. J., Moynihan, P. J., Lelièvre, J., Selenski, C., Axtman, M., Kremer, L., Bantscheff, M., Angulo-Barturen, I., Izquierdo, M. C., Cammack, N. C., Drewes, G., Ballell, L., Barros, D., Besra, G. S., and Bates, R. H. (2016) Identification of KasA as the cellular target of an anti-tubercular scaffold, Nat Commun 7, 12581. doi: 10.1038/ncomms12581. [11] Cox, J. A., Abrahams, K. A., Alemparte, C., Ghidelli-Disse, S., Rullas, J., AnguloBarturen, I., Singh, A., Gurcha, S. S., Nataraj, V., Bethell, S., Remuiñán, M. J., Encinas, L., Jervis, P. J., Cammack, N. C., Bhatt, A., Kruse, U., Bantscheff, M., Fütterer, K., Barros, D., Ballell, L., Drewes, G., and Besra, G. S. (2016) THPP target

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 36

assignment reveals EchA6 as an essential fatty acid shuttle in mycobacteria, Nat Microbiol 1, 15006. doi: 10.1038/nmicrobiol.2015.6. [12] Batt, S. M., Cacho Izquierdo, M., Castro Pichel, J., Stubbs, C. J., Vela-Glez Del Peral, L., Pérez-Herrán, E., Dhar, N., Mouzon, B., Rees, M., Hutchinson, J. P., Young, R. J., McKinney, J. D., Barros Aguirre, D., Ballell, L., Besra, G. S., and Argyrou, A. (2015) Whole cell target engagement identifies novel inhibitors of Mycobacterium tuberculosis decaprenylphosphoryl-β-d-ribose

oxidase,

ACS

Infect

Dis

1,

615-626.

doi:

10.1021/acsinfecdis.5b00065. [13] Mdluli, K., Kaneko, T., and Upton, A. (2015) The tuberculosis drug discovery and development pipeline and emerging drug targets, Cold Spring Harb Perspect Med 5. doi: 10.1101/cshperspect.a021154. [14] Mori, G., Chiarelli, L. R., Esposito, M., Makarov, V., Bellinzoni, M., Hartkoorn, R. C., Degiacomi, G., Boldrin, F., Ekins, S., de Jesus Lopes Ribeiro, A. L., Marino, L. B., Centárová, I., Svetlíková, Z., Blaško, J., Kazakova, E., Lepioshkin, A., Barilone, N., Zanoni, G., Porta, A., Fondi, M., Fani, R., Baulard, A. R., Mikušová, K., Alzari, P. M., Manganelli, R., de Carvalho, L. P., Riccardi, G., Cole, S. T., and Pasca, M. R. (2015) Thiophenecarboxamide derivatives activated by EthA kill Mycobacterium tuberculosis by

inhibiting

the

CTP

synthetase

PyrG,

Chem

Biol

22,

917-927.

doi:

10.1016/j.chembiol.2015.05.016. [15] Turnbough, C. L., and Switzer, R. L. (2008) Regulation of pyrimidine biosynthetic gene expression in bacteria: repression without repressors, Microbiol Mol Biol Rev 72, 266300. doi: 10.1128/MMBR.00001-08.

ACS Paragon Plus Environment

Page 29 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

[16] Villela, A. D., Sánchez-Quitian, Z. A., Ducati, R. G., Santos, D. S., and Basso, L. A. (2011) Pyrimidine salvage pathway in Mycobacterium tuberculosis, Curr Med Chem 18, 1286-1298. doi: 10.2174/092986711795029555 [17] Bochner, B. R., and Ames, B. N. (1982) Complete analysis of cellular nucleotides by two-dimensional thin layer chromatography, J Biol Chem 257, 9759-9769. [18] Makarov, V., Manina, G., Mikusova, K., Möllmann, U., Ryabova, O., Saint-Joanis, B., Dhar, N., Pasca, M. R., Buroni, S., Lucarelli, A. P., Milano, A., De Rossi, E., Belanova, M., Bobovska, A., Dianiskova, P., Kordulakova, J., Sala, C., Fullam, E., Schneider, P., McKinney, J. D., Brodin, P., Christophe, T., Waddell, S., Butcher, P., Albrethsen, J., Rosenkrands, I., Brosch, R., Nandi, V., Bharath, S., Gaonkar, S., Shandil, R. K., Balasubramanian, V., Balganesh, T., Tyagi, S., Grosset, J., Riccardi, G., and Cole, S. T. (2009) Benzothiazinones kill Mycobacterium tuberculosis by blocking arabinan synthesis, Science 324, 801-804. doi: 10.1126/science.1171583. [19] Evans, J. C., Trujillo, C., Wang, Z., Eoh, H., Ehrt, S., Schnappinger, D., Boshoff, H. I., Rhee, K. Y., Barry, C. E., and Mizrahi, V. (2016) Validation of CoaBC as a bactericidal target in the coenzyme a pathway of Mycobacterium tuberculosis, ACS Infect Dis 2, 958-968. doi: 10.1021/acsinfecdis.6b00150 [20] Chang, Y. F., and Carman, G. M. (2008) CTP synthetase and its role in phospholipid synthesis in the yeast Saccharomyces cerevisiae, Prog Lipid Res 47, 333-339. doi: 10.1016/j.plipres.2008.03.004. [21] Lunn, F. A., MacDonnell, J. E., and Bearne, S. L. (2008) Structural requirements for the activation of Escherichia coli CTP synthase by the allosteric effector GTP are stringent, but requirements for inhibition are lax, J Biol Chem 283, 2010-2020. doi: 10.1074/jbc.M707803200 ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 36

[22] Copeland, A. (2000) Enzymes: A Practical Introduction to Structure, Mechanism, and Data Analysis., 2nd ed., John Wiley & Sons Inc., New York, NY. [23] Valentini, G., Chiarelli, L., Fortini, R., Speranza, M. L., Galizzi, A., and Mattevi, A. (2000) The allosteric regulation of pyruvate kinase: A site-directed mutagenesis study, J Biol Chem 275, 18145-18152. doi: 10.1074/jbc.M001870200 [24] Wang, C., Chiarelli, L. R., Bianchi, P., Abraham, D. J., Galizzi, A., Zanella, A. M. A., and Valentini, G. (2001) Human erythrocyte pyruvate kinase: characterization of the recombinant enzyme and a mutant form (R510Q) causing nonspherocytic hemolytic anemia, Blood 98, 3113-3120. doi: 10.1182/blood.V98.10.3113 [25] Abrusci, P., Chiarelli, L. R., Galizzi, A., Fermo, E., Bianchi, P., Zanella, A., and Valentini, G. (2007) Erythrocyte adenylate kinase deficiency: characterization of recombinant mutant forms and relationship with nonspherocytic hemolytic anemia, Exp Hematol 35, 1182-1189. doi: 10.1016/j.exphem.2007.05.004 [26] Lucarelli, A. P., Buroni, S., Pasca, M. R., Rizzi, M., Cavagnino, A., Valentini, G., Riccardi,

G.,

and

Chiarelli,

L.

R.

(2010)

Mycobacterium

tuberculosis

phosphoribosylpyrophosphate synthetase: biochemical features of a crucial enzyme for

mycobacterial

cell

wall

biosynthesis,

PLoS

One

5,

e15494.

doi:

10.1371/journal.pone.0015494. [27] Chiarelli, L. R., Morera, S. M., Bianchi, P., Fermo, E., Zanella, A., Galizzi, A., and Valentini, G. (2012) Molecular insights on pathogenic effects of mutations causing phosphoglycerate

kinase

deficiency,

PLoS

One

10.1371/journal.pone.0032065.

ACS Paragon Plus Environment

7,

e32065.

doi:

Page 31 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

[28] Palomino, J. C., Martin, A., Camacho, M., Guerra, H., Swings, J., and Portaels, F. (2002) Resazurin microtiter assay plate: simple and inexpensive method for detection of drug resistance in Mycobacterium tuberculosis, Antimicrob Agents Chemother 46, 2720-2722. doi: 10.1128/AAC.46.8.2720-2722.2002. [29] Forti, F., Crosta, A., and Ghisotti, D. (2009) Pristinamycin-inducible gene regulation in mycobacteria, J Biotechnol 140, 270-277. doi: 10.1016/j.jbiotec.2009.02.001. [30] Rao, S. N., Head, M. S., Kulkarni, A., and LaLonde, J. M. (2007) Validation studies of the site-directed docking program LibDock, J Chem Inf Model 47, 2159-2171. doi: 10.1021/ci6004299. [31] Krainer, F. W., Dietzsch, C., Hajek, T., Herwig, C., Spadiut, O., and Glieder, A. (2012) Recombinant protein expression in Pichia pastoris strains with an engineered methanol utilization pathway, Microb Cell Fact 11, 22. doi: 10.1186/1475-2859-11-22. [32] Lin-Cereghino, J., Wong, W. W., Xiong, S., Giang, W., Luong, L. T., Vu, J., Johnson, S. D., and Lin-Cereghino, G. P. (2005) Condensed protocol for competent cell preparation and transformation of the methylotrophic yeast Pichia pastoris, Biotechniques 38, 44-48. doi: 10.2144/05381BM04. [33] Bradford, M. M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Anal Biochem 72, 248-254. doi: 10.1016/0003-2697(76)90527-3.

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

For Table of Contents Use Only

A phenotypic based target screening approach delivers new antitubercular CTP synthetase inhibitors.

Marta Esposito, Sára Szadocka, Giulia Degiacomi, Beatrice S. Orena, Giorgia Mori, Valentina Piano, Francesca Boldrin, Júlia Zemanová, Stanislav Huszár, David Barros, Sean Ekins, Joel Lelièvre, Riccardo Manganelli, Andrea Mattevi, Maria Rosalia Pasca, Giovanna Riccardi, Lluis Ballell, Katarína Mikušová, Laurent R. Chiarelli.

The combination of whole cell phenotypic screening and biochemical target based assay can be fruitfully exploited for the identification and development of novel antitubercular leads, as demonstrated by our small screening against the recently validated target CTP synthetase.

ACS Paragon Plus Environment

Page 32 of 36

Page 33 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 1. Docking GSK compounds in CTP synthetase. (A) compound (1) libdock score 88.16; (B) compound (2) libdock score 92.96; (C) compound (3) libdock score 87.23. All molecules (grey) are compared to UTP (Yellow); (D) compound 11426026, previously reported as a PyrG ATP binding site competitive inhibitor. 156x202mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 2. Double reciprocal plots of PyrG kinetic analysis towards ATP, in the presence of different concentrations of compound (1), (2) and (3) (panel A, B and C, respectively). 69x27mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 34 of 36

Page 35 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 3. Effects of PyrG inhibitors on human CTP synthetase 1 activity. IC50 determination of 11426026 compound (panel A) and GSK compounds (1) (panel B), (2) (panel C) and (3) (panel D) on the M. tuberculosis PyrG (filled symbols) and Hu-CTPS1 (open symbols). 137x135mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Infectious Diseases

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 4. Examination of effects of (1), (2) and (3) on M. tuberculosis H37Ra cells by metabolic labeling. Bacterial cells were pre-treated with 8-fold MIC of the drugs for 1 h, followed by 3 h of radiolabeling with [14C]uracil (A) or [14C]-acetate (B, C). (A) Ratios of radioactivity incorporated to [14C]UTP and [14C]CTP extracted from [14C]uracil-labeled cells. Nucleotide extracts were separated by TLC on PEI-cellulose plate. Columns and error bars represent mean ± SD of triplicate measurement of radioactivity in the [14C]UTP and [14C]CTP spots cut from TLC plates corresponding to two separate experiments; from each experiment two plates were analyzed. The autoradiograph is representative image from three separate experiments. (B) TLC analysis of the lipids from [14C]-acetate-labeled cells. Lipids were extracted as in “Methods”, aliquots corresponding to the same amounts of extracted material (as shown in 5C) were loaded on a silica gel TLC plate and separated in Solvent I: CHCl3/CH3OH/NH4OH/H2O (65:25:0.5:4); Solvent II CHCl3/CH3OH/H2O (20:4:0.5); and Solvent III: petroleum ether/ethyl acetate (98:2; 3 times).The plates were exposed to an autoradiography film at −80 °C for 6 days. TDM, trehalosedimycolates; TMM, trehalosemonomycolates; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PIM, phosphatidylinositol mannosides; CL, cardiolipin;

ACS Paragon Plus Environment

Page 36 of 36

Page 37 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

TAG, triacylglycerol. Autoradiographs are representative images from three separate experiments. (C) Visualization of the cold lipids by cupric sulfate staining. 154x190mm (300 x 300 DPI)

ACS Paragon Plus Environment