Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 17.
Published in final edited form as: ACS Infect Dis. 2021 Sep 30;7(11):3062–3076. doi: 10.1021/acsinfecdis.1c00342

The enzymatic activity of inosine 5’-monophosphate dehydrogenase may not be a vulnerable target for Staphylococcus aureus infections

Gyan Modi 1, Gary M Marqus 2, Mohana Rao Vippila 3, Deviprasad R Gollapalli 1, Youngchang Kim 4, Adhar C Manna 5, Shibin Chacko 1, Natalia Maltseva 4, Xingyou Wang 2, Ryan T Cullinane 6, Yubo Zhang 6, Judy L M Kotler 7, Petr Kuzmic 8, Minjia Zhang 1, Ann P Lawson 1, Andrzej Joachimiak 4,10, Ambrose Cheung 5, Barry B Snider 9, David M Rothstein 11, Gregory D Cuny 3, Lizbeth Hedstrom 1,9,*
PMCID: PMC9575405  NIHMSID: NIHMS1839774  PMID: 34590817

Abstract

Many bacterial pathogens, including Staphylococcus aureus, require inosine 5’-monophosphate dehydrogenase (IMPDH) for infection, making this enzyme a promising new target for antibiotics. Although potent selective inhibitors of bacterial IMPDHs have been reported, relatively few have displayed antibacterial activity. Here we use structure-informed design to obtain inhibitors of S. aureus IMPDH (SaIMPDH) that have potent antibacterial activity (minimal inhibitory concentrations less than 2 μM) and low cytotoxicity in mammalian cells. The physicochemical properties of the most active compounds were within typical Lipinski/Veber space, suggesting that polarity is not a general requirement for achieving antibacterial activity. Five compounds failed to display activity in mouse models of septicemia and abscess infection. Inhibitor-resistant S. aureus strains readily emerged in vitro. Resistance resulted from substitutions in the cofactor/inhibitor binding site of SaIMPDH, confirming on-target antibacterial activity. These mutations decreased the binding of all inhibitors tested, but also decreased catalytic activity. Nonetheless, the resistant strains had comparable virulence to wild-type bacteria. Surprisingly, strains expressing catalytically inactive SaIMPDH displayed only a mild virulence defect. Collectively these observations question the vulnerability of the enzymatic activity of SaIMPDH as a target for the treatment of S. aureus infections, suggesting other functions of this protein may be responsible for its role in infection.

Keywords: IMPDH, antibiotic space, guaB, guanine nucleotide biosynthesis, target vulnerability, virulence

Graphical Abstract

graphic file with name nihms-1839774-f0001.jpg


The impending obsolescence of clinical antibiotics demands the development of new therapies1. Treatment options are already severely limited for many pathogens, including methicillin resistant Staphylococcus aureus (MRSA)2. The few new antibiotics developed recently are largely re-tooled β-lactams, tetracyclines, quinolones and aminoglycosides and there is legitimate concern that these too will quickly become compromised. New antimicrobial drugs are urgently needed, ideally with new targets.

Inosine monophosphate dehydrogenase (IMPDH) is a promising potential antibiotic target3. This enzyme catalyzes the rate limiting step in guanine nucleotide biosynthesis, the conversion of IMP to XMP with concomitant oxidation of NAD+. The gene encoding IMPDH (guaB) has been found to be essential for infection by many pathogens in genome-wide screens313. IMPDH is a particularly alluring target for S. aureus infections. Both genome-wide transposon and single gene knockout experiments indicate that guaB is essential for growth in whole blood and ocular fluids and for infections in mice8, 14. Nonetheless, these observations are somewhat surprising given that S. aureus possesses salvage pathways that should provide guanine nucleotides in the absence of IMPDH.

The cofactor binding site of S. aureus IMPDH (SaIMPDH) is very different from those found in the human enzymes15, 16. Importantly, selective inhibitors, derived from several scaffolds, have already been reported for IMPDHs with similar cofactor binding sites, including IMPDHs from the intracellular protozoan pathogen Cryptosporidium parvum (Cp)1722, Bacillus anthracis (Ba)15, 23, Clostridium perfringens15, Campylobacter jejuni15, Francisella tularensis24 and Mycobacterium tuberculosis (Mtb)2531. However, relatively few compounds display antibacterial activity. Moreover, when antibacterial activity is observed, the spectrum is usually limited to a few pathogens, most likely reflecting differences in the structure activity relationships (SAR) for both enzyme inhibition and compound uptake.

The urea and triazole scaffolds, designated P and A in our previous reports, respectively, displayed the greatest antibacterial activity against B. anthracis, as exemplified by P150 and A98 (Figure 1A)23. These compounds were originally designed for uptake into mammalian and protozoan cells in our CpIMPDH inhibitor program, and are consequently more lipophilic than typical for antibiotics3234. For example, P32 and P37 are moderately potent inhibitors of BaIMPDH with weak antibacterial activity (Figure 1A)23, while the more polar P146 displays superior antibacterial activity despite being a considerably weaker enzyme inhibitor. These observations suggested that antibacterial activity might be improved if the compounds were redesigned to increase polarity while maintaining potency of enzyme inhibition.

Figure 1. Bacterial IMPDH inhibitors.

Figure 1.

A. Inhibitors with antibacterial activity against B. anthracis23. B. The residues within 5 Å of P32 in BaIMPDH are shown (PDB 4MYX, A-C’ active site). The subunit listed first contains the IMP site while the subunit denoted with a prime contains the adenosine subsite. Val25’ (light green) is Ile in SaIMPDH. The aromatic rings of P32 (pink) are denoted “R” and “L”. IMP is gray. Hydrogen and halogen bonds are shown in cyan. Water molecules were removed for clarity. C. Inhibitor SAR of BaIMPDH and SaIMPDH are similar. The black line denotes equal values of Ki,app for both enzymes. The dotted lines denote 2-fold differences. D. The P32 complex of BaIMPDH (PDB id 4MYX, chains A-C’) is overlayed with the cofactor complex of MtbIMPDH2 (PDB id 4ZQM, chains D-C’). Residues within 5 Å of P32 and NAD+ are shown. Water molecules removed for clarity. BaIMPDH, tan; MtbIMPDH2, sky blue, XMP light green (MtbIMPDH), IMP gray (BaIMPDH) and light green (MtbIMPDH); P32 (pink); NAD, dark cyan. BaIMPDH residues are labeled15.

Here we report second generation bacterial IMPDH inhibitors that display potent antibacterial activity against S. aureus in vitro and low cytotoxicity in mammalian cells, but do not display in vivo efficacy. Moreover, resistant strains of S. aureus readily emerged that have comparable virulence to wild-type strains. Lastly, bacteria expressing catalytically inactive IMPDH displayed only a mild virulence defect. Collectively these observations question the value of targeting the enzymatic activity of IMPDH for the treatment of S. aureus infections.

RESULTS

Characterization of SaIMPDH.

We previously characterized five bacteria IMPDHs predicted to be sensitive to CpIMPDH inhibitors15, 23, 25, 35. The SAR for enzyme inhibition was surprisingly varied despite the close similarity of the inhibitor binding sites in these enzymes. Therefore, we characterized IMPDH from S. aureus (SaIMPDH) to better enable the development of inhibitors for the treatment of S. aureus infections. We expressed and purified N-terminal His-tagged SaIMPDH (sequence NC_007622.1) in E. coli BL21ΔguaB as described previously for other bacterial IMPDHs15. The Michaelis-Menten parameters were similar to those of BaIMPDH (Table 1). The value of Km(NAD+) for SaIMPDH was smaller than that previously reported by another laboratory (literature value 2.3 mM; other parameters were not reported36), but these assays were performed at different temperature and pH.

Table 1.

Steady state kinetic parameters for bacterial IMPDHs.

Parameter BaIMPDHa SaIMPDHb Sa2IMPDHb,c G445Sb,d Y446Cb,e
K m (IMP), μM 18 ± 2 35 ± 2 25 ± 2 88 ± 3 96 ± 5f
K m (NAD + ), μM 550 ± 50 390 ± 20 228 ± 8 200 ± 10 5800 ± 400
K ii (NAD + ), mM 3.9 ± 0.5 8.0 ± 0.4 7.3 ± 0.6 12.0 ± 0.5 n.a.
k cat , s −1 1.4 ± 0.2 2.3 ± 0.0 2.0 ± 0.0 0.50 ± 0.01 0.90 ± 0.01d

Buffer 50 mM Tris, pH 8.0, 150 mM KCl, 3 mM EDTA and 1 mM DTT.

a.

BaIMPDH, recombinant IMPDH from B. anthracis, values from35.

b.

SaIMPDH, recombinant IMPDH from S. aureus as reported in NC_007622.1. This enzyme was used routinely for inhibitor characterization. Sa2IMPDH, recombinant IMPDH from S. aureus Smith. Sa2IMPDH-G445S and Sa2IMPDH-Y446C, recombinant IMPDH from S. aureus Smith containing the eponymous mutations associated with resistance to 25.

c.

[E] = 20 nM;

d.

[E] = 20 nM;

e.

[E] = 200 nM;

f.

[NAD+] = 8 mM. n.a., not applicable.

Many x-ray crystal structures of IMPDH inhibitor complexes have been reported, including ten with BaIMPDH15, 35. We compared the structure-activity relationship (SAR) of enzyme inhibition for SaIMPDH and BaIMPDH in order to determine the relevance of these structures for inhibitor design. SaIMPDH is 73% identical to BaIMPDH overall; only one residue within 5 Å of inhibitor P32 is different in the two enzymes (Val25 is Ile in SaIMPDH; Figure 1B). We surveyed a set of 34 compounds drawn from the A (15 compounds) and P (19 compounds) series, including A98, A110, P146 and P150 (Figure 1C, Table S1). The values of Ki,app for SaIMPDH and BaIMPDH were very similar for all of the A compounds. However, while the P compounds followed the same trend, the values of Ki,app were lower for SaIMPDH by an average factor of 2.5. The structural features that account for the different SAR of the A and P compounds are not readily apparent. Nonetheless, these observations suggested that the structures of BaIMPDH inhibitor complexes would be a useful guide for further medicinal chemistry optimization.

Redesign of the P series to increase t-PSA.

We initially focused on further optimization of the P series because these compounds include the most potent inhibitors of SaIMPDH and are more readily synthesized18, 23. We retained the 4-Cl on the R-ring because this moiety forms a halogen bond with the main chain carbonyl C of Gly444’ (Figure 1B). We first increased the polarity at the 3-position on the R-ring with the addition of sulfonamides and sulfoxides (1, 2 and 3) (Table 2). We also substituted the 3-position with small heterocycles that improved antibacterial activity in topoisomerase inhibitors (4, 5, 6)37, 38. All of these substitutions had a deleterious effect on enzyme inhibition relative to P37, ranging from a factor of ~2 (1 and 5) to a factor of ~20 (2). Nonetheless, several compounds displayed better antibacterial activity than P37, most notably the trifluoromethylpyrazole 4 and the trifluoromethylthiazole 5, where antibacterial activity improved by factors of 40–60.

Table 2.

Optimization of the P series.

ID Cmpd t-PSA
2)
cLogP Kiapp (nM) MIC (μM)
−Guo +Guo
P37 a graphic file with name nihms-1839774-t0002.jpg 73.72 3.58 5.5 ± 0.4 60a n.d.
P146 a graphic file with name nihms-1839774-t0003.jpg 103.26 2.67 26 ± 9 2 a >30a
P150 a graphic file with name nihms-1839774-t0004.jpg 94.03 3.77 15 ± 2 4 a >30a
1 graphic file with name nihms-1839774-t0005.jpg 128.27 2.55 10 ± 5 13 ± 5 b >40c
2 graphic file with name nihms-1839774-t0006.jpg 139.79 2.10 110 ± 50 n.d. n.d.
3 graphic file with name nihms-1839774-t0007.jpg 136.55 2.19 37 ± 3 13 ± 5 >64
4 graphic file with name nihms-1839774-t0008.jpg 91.54 4.47 29 ± 10 2.5 ± 1.5 32
5 graphic file with name nihms-1839774-t0009.jpg 97.38 4.47 13 ± 2 1 ± 0 >64
6 graphic file with name nihms-1839774-t0010.jpg 117.56 3.66 28 ± 7 8 ± 0 >64
7 graphic file with name nihms-1839774-t0011.jpg 112.75 2.29 51 ± 9 >30c >30c
8 graphic file with name nihms-1839774-t0012.jpg 124.27 1.84 830 ± 50c n.d. n.d.
9 graphic file with name nihms-1839774-t0013.jpg 121.03 1.93 250 ± 90 > 30c >30c
10 graphic file with name nihms-1839774-t0014.jpg 76.02 4.21 118 ± 2 >30c >30c
11 graphic file with name nihms-1839774-t0015.jpg 99.33 4.94 25 ± 1 >30b >30b
12 graphic file with name nihms-1839774-t0016.jpg 119.47 2.32 220 ± 20 n.d. n.d.
13 graphic file with name nihms-1839774-t0017.jpg 108.32 1.97 2800 ± 300c n.d. n.d.
14 a graphic file with name nihms-1839774-t0018.jpg 41.13 5.04 9.5 ± 1.0 60 n.d.
15 graphic file with name nihms-1839774-t0019.jpg 64.92 3.36 560 ± 150 n.d. n.d.
16 graphic file with name nihms-1839774-t0020.jpg 81.26 3.17 1600 ± 200c n.d. n.d.
17 graphic file with name nihms-1839774-t0021.jpg 70.23 3.20 500 ± 130c n.d. n.d.
18 graphic file with name nihms-1839774-t0022.jpg 61.44 3.10 960 ± 160b n.d. n.d.
19 graphic file with name nihms-1839774-t0023.jpg 99.74 2.77 21.5 ± 0.2 4 ± 2 28 ± 4
20 graphic file with name nihms-1839774-t0024.jpg 99.74 3.40 12.9 ± 0.1 1.2 ± 0.1 >32
21 graphic file with name nihms-1839774-t0025.jpg 67.15 4.36 12.8 ± 0.7 1.7 ± 0.3 >32
22 graphic file with name nihms-1839774-t0026.jpg 61.36 4.68 9 ± 4 3 ± 1 >50
23 graphic file with name nihms-1839774-t0027.jpg 90.49 4.60 4 ± 1 >40 >40
24 graphic file with name nihms-1839774-t0028.jpg 76.66 5.30 19 ± 1 >64 >64 c
25 graphic file with name nihms-1839774-t0029.jpg 93.45 3.95 6 ± 1 1.1 ± 0.1 >32
26 graphic file with name nihms-1839774-t0030.jpg 99.69 3.87 6 ± 2 4 ± 0 b ≥64
27 graphic file with name nihms-1839774-t0031.jpg 109.25 3.84 16 ± 1 1.7 ± 0.5 >64
28 graphic file with name nihms-1839774-t0032.jpg 126.04 2.50 2.4 ± 0.3 1 ± 0 >50
29 d graphic file with name nihms-1839774-t0033.jpg 123.08 3.51 44 ± 11 11 ± 5 >64
30 graphic file with name nihms-1839774-t0034.jpg 120.28 3.50 20 ± 7 6 ± 2 >30
31 d graphic file with name nihms-1839774-t0035.jpg 123.08 3.51 20 ± 5 2 ± 2 >64
32 graphic file with name nihms-1839774-t0036.jpg 154.29 1.74 13 ± 2 >64 >64 c
33 graphic file with name nihms-1839774-t0037.jpg 162.57 1.38 52 ± 19 >64 n.d.
34 graphic file with name nihms-1839774-t0038.jpg 140.87 4.39 10 ± 2 0.8 ± 0.3 >40
35 graphic file with name nihms-1839774-t0039.jpg 117.56 3.66 26 ± 5 1.5 ± 0.5 >40

The values of Ki,app for SaIMPDH are averages and standard deviations of at least 3 independent determinations unless otherwise noted. Note that a single determination is derived from at least 30 rate measurements, triplicates of at least 10 different inhibitor concentrations. Conditions: 20 nM enzyme, 1000 μM IMP and 1000 μM NAD+. Minimum inhibitory concentration (MIC) for S. aureus Smith in RPMI media are the average and range of at least two independent determinations.

a.

Synthesis/data from23.

b.

Average and range of 2 independent determinations.

c.

Value and error of the fit for 1 determination. n.d., not determined. cLogP and t-PSA calculated on the FAFDrugs4 website39.

d.

Compound is likely a mixture of both pyranoses (major) and both furanoses (minor)4042.

In general, replacement of the oxime on the L-ring with a ketone was deleterious for enzyme inhibition, increasing the values of Kiapp by factors of 4–7 (compare 1 vs. 7, 2 vs. 8, 3 vs. 9 and 4 vs. 10). The ketone analog of 5 (11) was also a good inhibitor of SaIMPDH, but displayed little antibacterial activity. Replacement of the oxime with cyano was more deleterious, increasing the value of Kiapp by factors of 22–75 (1 vs. 12 and 3 vs. 13). To further explore the requirements of the L-ring oxime, we compared analogs of P37 where the oxime was replaced with alkene (14), cyano (15), carboxylate (16), methylamide (17), dimethylamide (18) and hydroxyamidine (19) (Table 2). All of these substitutions were deleterious with respect to enzyme inhibition. Most of these substitutions increased Ki,app by factors of 100 or more. However, the alkene and hydroxyamidine substitutions had increased Ki,app by only factors of 2 and 4, respectively. Remarkably, the hydroxyamidine 19 displayed a 10-fold improvement in antibacterial activity despite the loss of potency in enzyme inhibition. We also evaluated the di-Cl derivative 20 since prior work indicated that this substitution improved antibacterial activity23. This substitution improved both enzyme inhibition relative to 19 by a factor of 18 and antibacterial activity by a factor of 3.3 (20 MIC = 1.2 μM).

Redesign to interact with the adenosine ribose binding site.

Recent x-ray crystal structures of bacterial IMPDHs revealed that the A and P compounds bind in the cofactor binding site15, 25. Overlay of the structures of the BaIMPDH•P32 complex and MtbIMPDH2•cofactor complex indicates that the R-ring of the P compounds binds in a subsite occupied by the adenine base, but does not extend into the ribose binding subsite (Figure 1D). Further inspection suggested that modifications of the 3-position on the R-ring would extend into the ribose binding site, providing another attractive strategy for increasing potency and polarity.

We turned to a starting scaffold that contains an alkene group on the L-ring because it is more easily synthesized than the oximes and hydroxyamidines (e.g. 14, Table 2). Although 14 was a potent inhibitor of SaIMPDH, it displayed little antibacterial activity. We first added 3-NH2 (21) and 3-OH (22) substituents to enable further elaboration of the 3-position. These compounds were potent inhibitors of SaIMPDH, equivalent to 14 (Table 2). Both 21 and 22 displayed improved antibacterial activity relative to 14 (MIC = 1.7 and 3 μM, respectively).

We next added hydrogen bonding moieties to the 3-position of the R-ring with the intent of introducing an interaction with Ser257 (23, 24, 25 and 26). Addition of an acetate group (23) increased enzyme affinity, but eliminated antibacterial activity. Such substitutions have been similarly debilitating in the antibacterial activity of topoisomerase inhibitors37. The corresponding ethyl ester derivative (24) had a deleterious effect on enzyme affinity, and also failed to display antibacterial activity. In contrast, the amide (25) and hydroxamic acid (26) derivatives remained good enzyme inhibitors and displayed superior antibacterial activity (MIC = 1.1 and 4 μM, respectively). Since the oxime substituted L-ring was more polar and soluble, we synthesized the oxime versions of 24 (e.g. 27) and 25 (e.g. 28). Compound 27 displayed similar enzyme inhibition as 24. Nonetheless, antibacterial activity improved by more than a factor of 30. In contrast, no improvement in antibacterial activity was observed for 28 relative to 25, although enzyme inhibition improved by a factor of approximately 2.5. These observations reveal that the structure-activity relationship (SAR) for antibacterial activity is very context dependent.

Further inspection of the crystal structure of BaIMPDH•IMP•P32 suggested that a sugar attached to the 3-position of the R-ring would bind in the site occupied by the nicotinamide ribose of the cofactor (Figure 1D). Therefore, ribose (29 and 30) and arabinose (31) were appended to this position. These compounds exist as a mixture of anomers. In addition, the reaction of aniline with ribose gives a mixture containing 50% of the β-pyranose, 35% of the α-pyranose, 10% of the β-furanose and 5% of the α-furanose, that can equilibrate readily under neutral conditions by ring opening to the hydroxy imine intermediate. Thus, 29 and 31 are likely mixtures of both pyranoses (major) and both furanoses (minor)4042. These substitutions decreased the potency of enzyme inhibition by 2 to 4-fold relative to 21 and 22. The antibacterial activity of 29 was similarly decreased, while 30 and 31 maintained antibacterial activity.

Combination of L- and R-ring substitutions.

We combined the L-ring hydroxyamidine with the successful R-ring substituents to produce 32, 33, 34 and 35, with the goal of further improving antibacterial activity. Hydroxyamidine substitution had little effect on enzyme inhibition relative to the oxime and alkene analogs (e.g., 1 vs 32, 3 vs. 33, 4 vs. 35 and 5 vs. 34) (Table 2). Disappointingly, the hydroxamidine substitution did not improve antibacterial activity relative to the oxime analogs. No antibacterial activity was observed for 32 and 33. The antibacterial activity of 34 and 35 was similar to their oxime analogs with MIC values of ~1 μM. These observations further underscore the seemingly capricious nature of antibacterial SAR.

A-series SaIMPDH inhibition and antibacterial activity.

Two compounds in the A series, A98 and A110, previously demonstrated potent antibacterial activity against S. aureus23. Given the improved antibacterial activity imparted by the thiazole in the P series, we decided to introduce this heterocycle into the 3-position of the phenyl of the A series (Table 3). In the four compounds evaluated (36-39), which contained various quinoline and quinoline N-oxide ethers, SaIMPDH inhibition was maintained with Ki,app values ≤ 30 nM. Antibacterial activity was also retained with MIC values of 3 – 4 μM, although this was not an improvement relative to A98.

Table 3.

A series compounds.

ID Cmpd t-PSAa2) cLogPa Ki,appb (nM) MIC (μM) c
−Guo +Guo
A98 d graphic file with name nihms-1839774-t0040.jpg 52.83 4.15 20 ± 4 2a 16a
A110 d graphic file with name nihms-1839774-t0041.jpg 65.4 3.15 50 ± 20 8a >30a
36 graphic file with name nihms-1839774-t0042.jpg 93.96 5.76 7 ± 2 3 ± 1 > 64
37 graphic file with name nihms-1839774-t0043.jpg 93.96 5.76 8 ± 2 3 ± 1 >64b
38 graphic file with name nihms-1839774-t0044.jpg 106.53 4.76 30 ± 10 4 ± 0 > 64
39 graphic file with name nihms-1839774-t0045.jpg 106.53 4.76 16 ± 2 3 ± 1 >64

See legend Table 2 for conditions.

a.

cLogP and t-PSA as calculated by FAFDrugs4 website39.

b.

Average and standard deviation from at least 3 independent determinations.

d.

Average and range from at least two independent determinations.

d.

Synthesis described in23.

Antibacterial activity is due to inhibition of SaIMPDH.

Figure 2 shows the correlation of antibacterial activity and inhibition of SaIMPDH for the P and A compounds described above. Greater antibacterial activity was associated with more potent enzyme inhibition, as expected if antibacterial activity derived from inhibition of SaIMPDH. Poor enzyme inhibitors displayed at best weak antibacterial activity. Some potent enzyme inhibitors did not display antibacterial activity, perhaps due to poor uptake or transformation into an inactive metabolite by the bacteria. To further confirm on-target antibacterial activity, we determined the values of MIC in the presence of guanosine, which allows cells to grow in the absence of IMPDH activity. None of the inhibitors displayed antibacterial activity in the presence of guanosine (MIC >30 μM), with the exception of 19, where the value of MIC increased by a factor of 7 (Tables 2 and 3). These observations indicate that antibacterial activity derives from inhibition of SaIMPDH as designed.

Figure 2. Physicochemical properties of enzyme inhibition and antibacterial activity.

Figure 2.

P compounds are denoted with circles, A compounds with squares. The dotted line denotes MIC = 2 μM. Compounds with no antibacterial activity were arbitrarily assigned MIC = 60 μM. Symbols are colored according to cLogP and t-PSA as calculated by FAFDrugs4 website39.

The physicochemical properties of antibacterial activity.

Although some studies have suggested that antibiotics are smaller and more polar than Lipinski/Veber guidelines3234, 43, these trends were not observed for the SaIMPDH inhibitors (Figure 2 and Figure S1). The average values of cLogP and t-PSA are 3.7 and 107 Å2, respectively, for the 11 P compounds with values of MIC ≤ 2 μM. The best antibacterial compounds, 34, 20 and 28, vary in cLogP from 2.50 to 4.39, while 32 does not display antibacterial activity despite being a potent inhibitor (Ki,app = 13 nM) with low lipophillicity (cLogP = 1.74). All of the A compounds have comparable antibacterial activity despite large differences in lipophilicity (cLogP varies from 3.15 to 5.76) and polar surface area (t-PSA varies from 52.83 to 106.53 Å2). These observations suggest that increasing polarity may not be a general strategy for antibiotic design.

Structures of enzyme-inhibitor complexes.

To determine how the thiazole, ribose, arabinose and hydroxyamidine groups interacted with the enzyme, we solved the x-ray crystal structures of five inhibitors (5, 19, 29, 30 and 31) with the catalytic domain of BaIMPDH (BaIMPDHΔL). This protein is missing the regulatory CBS subdomain, yet retains catalytic activity equivalent to the wild-type enzyme15. All complexes included IMP in addition to inhibitor. The crystal, data collection and refinement statistics can be found in Table S2.

The five structures were very similar overall (Figure 3). The unit cell of the complex with inhibitor 5 contained one tetramer, while two tetramers were found in the unit cells of the other complexes. All five inhibitors bound in the same site as P32. The L and R rings occupied the same position in all active sites. As expected, the L-ring interacted with IMP and the R-ring interacted with Tyr445’ in the adjacent subunit while the urea nitrogens formed hydrogen bonds with Glu41615. All of the inhibitors also interacted with residues Ala253, His254 and Pro27’, and all formed a halogen bond between the 4-Cl on the R-ring and the main chain carbonyl carbon of Gly444’. The 3-substituents of the R ring extended into the adenosine ribose subsite in all cases. In contrast, the structure of the “flap” (residues 390–418) displayed varying amounts of disorder in different complexes as well as in different active sites of the same complex (Figure 3). At least 4 different flap conformations were observed among the five structures, suggesting that the flap was dynamic.

Figure 3. The flap has multiple conformations in inhibitor complexes of BaIMPDHΔL.

Figure 3.

Representative active sites from E•IMP•inhibitor complexes are shown. Hydrogen bonds are indicated by solid cyan lines. The subunit containing the IMP binding site is listed first and the subunit containing the adenosine subsite of the cofactor binding site is denoted with a prime. The disordered regions are denoted by dashed lines. The structure of BaIMPDH•IMP•5 contains a single tetramer (PDB id 5UUV). Two flap conformations are exemplified in active sites A-D’ (light green) and D-B’ (lime green). The structure of BaIMPDH•IMP•19 contains two tetramers (PDB id 5UUZ). Two flap conformations were observed in the 8 active sites, exemplified by C-D’ (coral) and E-G’ (orange). The structure of BaIMPDH•IMP•29 complex contains two tetramers (PDB id 7MTU). Three flap conformations were observed in the 8 active sites, exemplified by A-C’ (tan), G-H’ (sienna), and H-F’ (brown). The structure of BaIMPDH•IMP•30 contains two tetramers of BaIMPDH (PDB id 5URS). Two flap conformations were observed in the 8 active sites, exemplified by D-B’ (plum) and G-H’ (medium purple). The structure of BaIMPDH•IMP•31 contains two tetramers (PDB id 7MTX). The flap conformations varied among the active sites, with B-A’ (light blue) and E-G’ (dark sky blue) representing the most divergent positions.

Distinct interactions were observed with the other substituents of the L and R rings in each inhibitor complex (Figures 4 and 5). For 5, L-ring thiazole ring formed a hydrogen bond with the side chain of Ser257 and the R-ring oxime OH formed a hydrogen bonds with the side chains of Thr310, Glu416, and Tyr445’ (Figure 4A). These hydrogen bonds were not observed with the oxime of P32 (Figure 1B). The L-ring hydroxyamidine N2 of 19 also formed a hydrogen bond with the carboxylate of Glu416 while the OH made a water mediated hydrogen bond with the carboxylate of Asp27 (Figure 4B). Thus, the L-ring substituents interact with BaIMPDH in a context-dependent manner that may explain the unpredictable SAR.

Figure 4. Interactions of the oxime and hydroxyamidine substitutions on the A ring.

Figure 4.

Hydrogen bonds are shown in cyan, Halogen bond in gold. A. Structure BaIMPDH•IMP•5 (PDB id 5UUV). 5 is magenta. Contact residues are shown for the D-B’ active site. B. Structure of BaIMPDH•IMP•19 (PDB id 5UUZ). 19 is green. Contact residues are shown for the active site formed by chains A-C’).

Figure 5. Interactions of sugar containing inhibitors with BaIMPDH.

Figure 5.

Contact residues in representative active sites are shown. Hydrogen bonds are shown in cyan and halogen bonds are gold. A. E•IMP•29 (PDB id 7mtx). D-B’ protein is plum, 29 is purple. B-A’ protein is light green, 29 is dark green. E-G’ protein is sky blue, 29 is dark sky blue. B. E•IMP•30 (PDB id 5urs). G-H’ protein is plum, 30 is purple; H-F’ protein is light green, 30 is dark green. C. E•IMP•31 (PDB id 7mtu). A-C’ protein is plum, 31 is purple. H-F’ protein is light green, 31 is dark green.

The sugar moieties of 29, 30 and 31 bound in the adenosine ribose site as designed (Figure 5AC). The structures were refined with the sugar conformer that best fit the electron density in each active site. However, the resolution was not sufficient to unambiguously distinguish tautomeric and anomeric configurations and it is possible that each active site contained a dynamic mixture of species. The ribose of 29 was modeled as pyranose, the favored tautomer of aniline-linked pentoses4042. The sugar moiety appeared to form hydrogen bonds to the side chain OHs of Thr252 and Ser257 and the main chain carbonyl Os of Val229 and His254 (Figure 5A). These interactions variously involved the ring O, 2’-OH, 3’-OH and/or 4’-OH depending on the active site. The sugar moiety of 30 was modeled as a furanose (Figure 5B). Depending on the active site, the 2’-OH variously formed hydrogen bonds with Thr252 side chain OH and the main chain NH and/or carbonyl O of His254. The 3’-OH also formed hydrogen bonds with the OH of Thr252 while the 5’-OH formed hydrogen bonds with Ser257. The arabinose moiety of 31 was modeled as a pyranose. The sugar appeared to make hydrogen bonds with the side chain hydroxyls of Thr252 and Ser257 and main chain carbonyl of His254. These interactions involved the 2’-OH, 3’-OH and 4’-OH depending on the sugar conformation modeled into a given active site (Figure 5C).

Inhibitor resistant S. aureus strains have mutations in SaIMPDH.

We selected strains of S. aureus Smith resistant to inhibitor 25 to gain further insight into the antibacterial activity of the SaIMPDH inhibitors. We chose this compound because it displayed potent antibacterial activity (MIC = 1 μM) and was easily synthesized. Twenty-six independent resistant clones were isolated that could grow in the presence of 25 (62 μM, 25 μg/mL) after multiple passages on agar plates. All 26 mutants displayed MICs ≥ 320 μM (128 μg/mL). The gene encoding IMPDH (guaB) gene was cloned from each strain and sequenced (note that the promoter was not included in sequencing because it is approximately 2.4 kb upstream from guaB, with two intervening genes). The wild-type guaB gene contained 21 substitutions relative to the sequence of SaIMPDH (NC_007622.1). We will denote the S. aureus Smith enzyme as Sa2IMPDH. Twenty-one resistant strains, exemplified by strain Sa26, contained a G->A mutation at nt1333 of guaB, causing the substitution of Ser for Gly445’. This residue is analogous to Gly444 in BaIMPDH, which interacts with the R-ring of the inhibitors (Figure 1D). Four resistant clones, exemplified by strain Sa12, contained a single A->G mutation at nt1337, causing the substitution of Cys for Tyr446’. The corresponding residue in BaIMPDH, Tyr445, also interacts with the R-ring. Intriguingly, the analogous Cys for Tyr substitution was also observed in an Mtb strain selected for resistance to another bacterial IMPDH inhibitor26. No mutations were discovered in the guaB coding sequence of the remaining resistant clone. The observation that 25/26 resistant clones contain mutations in guaB further confirms that antibacterial activity derives from inhibition of SaIMPDH as designed.

Resistance mutations impair enzyme activity.

Both Gly445’ and Tyr446’ interact with the R-ring of the P inhibitors, so substitutions at these residues can explain resistance to 25. However, these residues are also part of the cofactor binding site, which suggested that the substitutions might also impair cofactor binding and enzyme activity. Sa12 (expressing Y446C) displayed a slow growth phenotype (Figure S1), further suggesting that the enzymatic activity might be compromised in the resistant variants.

We characterized purified recombinant wild-type Sa2IMPDH and mutant enzymes Sa2IMPDH-G445S and Sa2IMPDH-Y446C to assess the functional consequences of the substitutions (Table 1). The kinetic parameters of Sa2IMPDH are very similar to SaIMPDH used in the SAR experiments described above. The catalytic activity of Sa2IMPDH-G445S was slightly compromised relative to the wild-type enzyme, with a 3.5-fold increase in the value of Km for IMP and a decrease in the value of kcat by a factor of 4. The catalytic activity of Sa2IMPDH-Y446C was also impaired. The values of Km for both IMP and NAD+ were increased, by 4-fold and 15-fold, respectively, and the value of kcat was reduced by a factor of 2-fold. Assuming cellular concentrations of 270 μM IMP and 2.5 mM NAD+ 44, the relative activities of Sa2IMPDH, Sa2IMPDH-G445S and Sa2IMPDH-Y446C would be approximately 1, 0.6 and 0.2, respectively.

Resistant Sa2IMPDH mutants are likely resistant to all inhibitors.

As expected, G445S and Y446C displayed lower affinity for 25, with values of Ki,app approximately 200-fold greater than the wild-type enzyme (Table 4). All bacterial IMPDH-specific inhibitors bind in the same site and interact with Gly445’ and Tyr446’15, 17, 22, 2527, 35, 45, 46, which suggested that Sa2IMPDH-G445S and Sa2IMPDH-Y446C would display decreased affinity to all bacterial IMPDH inhibitors. Indeed, Sa2IMPDH-G445S and Sa2IMPDH-Y446C were resistant to all twelve inhibitors tested, which included examples of 5 different scaffolds (Table 4). The two mutations had similar effects on the binding of most inhibitors, with Ki values typically 100–300-fold higher than those of the wild-type enzyme. Therefore, these mutations will likely provide resistance against any inhibitor targeting the cofactor site.

Table 4.

Inhibition of wild-type and mutant S. aureus IMPDHs.

Compound Sa2IMPDHa G445Sb Y446Cc
Kiapp (nM) Kiapp (nM) Ratio Kiapp (nM) Ratio
25 2.9 ± 0.5 570 ± 70 190 680 ± 40 230
29 39 ± 7 6200 ± 700 160 12000 ± 1500 310
30 15 ± 2 3800 ± 100 250 6200 ± 40 410
5 4.2 ± 1.7 1330 ± 30 320 920 ± 30 220
19 12.7 ± 0.4 4200 ± 100 330 3800 ± 400 300
34 4.8 ± 1.5 930 ± 30 190 1170 ± 5 240
35 20.5 ± 0.1 2500 ± 100 125 2800 ± 20 140
31 15 ± 5 3350 ± 150 220 5270 ± 90 350
A110 d 52 ± 5 8600 ± 100 160 >5000 94
36 2.7 ± 0.7 460 ± 10 170 10000 ± 1000 3700
graphic file with name nihms-1839774-t0046.jpg
Q21 e
8 ± 1 113 ± 5 14 3700 ± 2000 450
graphic file with name nihms-1839774-t0047.jpg
C91 f
77 ± 4 >5000 >65 >5000 >65
graphic file with name nihms-1839774-t0048.jpg
D67 g
320 ± 50 >5000 >16 >5000 >16
P32 h 29 ± 4 7100 ± 500 240 5600 ± 200 190

Sa2IMPDH, recombinant IMPDH from S. aureus Smith. G445S and Y446C, recombinant IMPDH from S. aureus Smith containing the eponymous mutations associated with resistance to 25. Buffer 50 mM Tris, pH 8.0, 150 mM KCl, 3 mM EDTA and 1 mM DTT. [IMP] = 1.0 mM.

a.

[E] = 20 nM, [NAD+] = 500 μM;

b.

[E] = 20 nM, [NAD+] = 500 μM.

c.

[E] = 200 nM, [NAD+] = 8 mM. n.a., not applicable.

d.

21;

e.

17;

f.

20;

g.

19;

h.

18.

Resistant mutants are virulent.

The above observations demonstrated that the mutations impaired the activity of Sa2IMPDH, suggesting that the resistant strains might have a reduced virulence. Initial testing of the wild-type strain established that infecting mice with an inoculum of 5×106 CFUs caused mortality reproducibly within 24 to 48 h (Table S3). Survival stabilized after 48 h and no additional mortality was observed over a total of 7 days. Survival increased as the inoculum was decreased by factors of 10. Two wild-type samples were used, one freshly revived from storage (designated “A”) and the second passaged to approximate the isolation of the resistant strains (designated “B”). Comparable survival was observed for mice infected with the two wild type samples, indicating that culture history had no effect on virulence. The mutant strains Sa12 and Sa26 were infected at higher numbers than the wild-type because reduced virulence was anticipated (Figure 6A and Table S3). However, survival was similar for mice infected with wild type and resistant strains, suggesting that there was little fitness cost associated with resistant mutations. These observations suggest that resistance will develop readily if IMPDH inhibitors are used as monotherapy. Moreover, the uncompromised virulence of Sa12, despite expressing an enzyme with only 20% of wild-type activity, suggests that in vivo efficacy will require inhibition exceeding 80%.

Figure 6. Virulence of S. aureus strains.

Figure 6.

A. Virulence of 25-resistant strains. WT-A, S. aureus Smith wild type strain freshly revived from storage; WT-B, denotes the wild-type strain passaged to approximate the isolation of the resistant strains. Sa12 carries Y446C substitution in IMPDH; Sa26 carries the G445S mutation. Survival was monitored over 7 days. B. Virulence of S. aureus JE2 expressing catalytically inactive IMPDH containing Ala for the essential catalytic residue Cys307.

SaIMPDH inhibitors are not cytotoxic.

Ten compounds with potent antibacterial activity, 8 from the P series and 2 from the A series, were tested for cytotoxicity in HepG2 cells by monitoring release of lactate dehydrogenase (Figure S3). Significant cytotoxicity (>10% release) was only observed in cells treated with 25 μM 34 and 35. Approximately 6% LDH release was observed when cells were treated with 25 μM 20 and 27. No cytotoxicity (<2% release) was observed in HepG2 cells treated with P146, A98, 5, 21, 31 and 39.

No in vivo efficacy is observed in mouse septicemia.

We tested the efficacy of five compounds, 20, 27, 34, 35 and 39 in a mouse septicemia model to further assess the antibiotic potential of SaIMPDH inhibitors. In the infection control group, only one mouse survived of the ten inoculated with 1.6 × 106 colony forming units (CFUs) of S. aureus Smith (Table S4). Survival increased markedly when mice were infected with fewer bacteria: 40% survival was observed when mice were inoculated with 1.6 × 105 CFUs while 90% survival was observed with 1.6 × 104 CFUs. As expected, all five mice treated with vancomycin (10 mg/kg i.v.) survived, further validating the model. These observations demonstrate that the experiment would be able to reveal in vivo efficacy. Disappointingly, none of the compounds displayed significant in vivo activity (Table S4). Only one mouse survived of the five treated with 20 (75 mg/kg i.v.) or 27 (25 mg/kg i.v.). These results are statistically indistinguishable from the untreated controls. No mice survived when treated with 34 (50 mg/kg i.v.), 35 (50 mg/kg i.v.) or 39 (30 mg/kg i.v.).

Preliminary pharmacokinetic evaluation.

We evaluated the pharmacokinetics of 20 and 39 to determine if sufficient exposure was achieved in the septicemia model. For 20 (75 mg/kg i.v.), C0 = 195 μM and τ1/2 = 1.1 h. For 39 (30 mg/kg i.v.), C0 = 150 μM and τ1/2 = 6.4 h. In both cases, the values of C0 greatly exceed MIC (1.2 μM and 2.1 μM for 20 and 39, respectively). However, both compounds display high plasma protein binding, with free fractions of 0.0043 and 0.014 for 20 and 39, respectively. These observations suggest that the concentration of free drug was at most 0.8 μM and 2.1 μM for 20 and 39, respectively, which would be approximately the value of MIC for each compound. Since survival improved when the inoculum decreased by a factor of 10, MIC concentrations of free 20 or 39 for even a few hours would be expected to display in vivo efficacy.

No in vivo efficacy is observed in mouse abscess infection model.

We also tested the efficacy of four compounds, 20, 34, 35 and 39, in S. aureus thigh abscess infection of mice, where SaIMPDH has been shown to be essential8, 14. To avoid potential issues with metabolic stability and plasma protein binding, compounds were injected directly into the abscess, once 2 h after inoculation and again after 12 h. A 2.5 log10 increase in colony forming units (CFUs) was observed after 26 h in the control abscesses treated with vehicle, while only a 1 log10 increase in CFUs was observed in mice treated with the positive control linezolid (Table S3). Despite the high local concentrations of compound, no statistically significant decreases in CFUs were observed relative to the vehicle control (Table S3). These results cast further doubt on the in vivo vulnerability of SaIMPDH.

Bacteria expressing catalytically inactive SaIMPDH have modestly reduced virulence.

Both genome-wide transposon analysis and single gene knockout experiments indicate that guaB, the gene encoding SaIMPDH, is essential for S. aureus infection in murine models of infection8, 14. These experiments suggest that loss of SaIMPDH reduces virulence by a factor of 105-106. However, these experiments did not address whether virulence requires the catalytic activity of IMPDH or some other function. Therefore, we constructed S. aureus JE2 strains expressing catalytically inactive IMPDH containing an Ala substitution at the catalytic Cys307 (guaB::C307A). This strain is a MRSA USA300 isolate comparable to NRS384, the strain used in previous work14. As expected, guaB::C307A cannot grow in absence of guanine (Figure S4). Growth is restored by plasmids expressing wild-type SaIMPDH.

We tested the virulence of wild-type and two guaB::C307A clones in the mouse septicemia model (Figure 6B and Tables S5 and S6). No survival was observed when mice were inoculated with 2.3×108 CFUs of wild-type bacteria, while 60% survival was observed with a 2.3×107 CFU inoculum. Approximately 20-fold more guaB::C307A bacteria were required to observe comparable mortality. Therefore, virulence appears to be only modestly dependent on the catalytic activity of IMPDH, suggesting that another function of this protein may be responsible for the essentiality associated with ΔguaB strains. Collectively these observations suggest that inhibitors of SaIMPDH may not be effective in treating S. aureus infections.

DISCUSSION

Gene knockout experiments have suggested that SaIMPDH is a promising target for the development of antibiotics to treat S. aureus infections8, 14 and structure-guided optimization of SaIMPDH inhibitors reported above yielded potent inhibitors with excellent antibacterial activity in vitro (MIC ≤ 2 μM). Some themes emerge from the SAR for enzyme inhibition that can be rationalized from the crystal structure. For example, the L-ring oxime and hydroxyamidine moieties form hydrogen bonds with the enzyme that may account for their increased affinity relative to the alkene and ketone groups. Nonetheless, different constellations of hydrogen bonds are observed depending on the R-ring, accounting for the varying effect of the oxime/hydroxyamidine substitutions. In contrast, the SAR for antibacterial activity appears almost capricious in comparison. For example, the addition of small heterocycles to the R-ring improves antibacterial activity by 60-fold in the context of the L-ring oxime, but has no effect in an L-ring hydroxyamidine (5 versus P37 and 34). The processes responsible for the unpredictable SAR of antibacterial activity are unclear, but could include inhibitor uptake and metabolism as well as regulation of enzyme function by post translational modification or other interactions. Moreover, it appears that these inhibitors bind to two different enzyme complexes, E•IMP and the covalent intermediate E-XMP*, and the preference for each complex varies between compounds. Therefore, potency can vary depending upon whether substrates are saturating (causing E-XMP* to accumulate) or sub-saturating (causing E•IMP to accumulate). We do not know which complex predominates in cells.

Since antibiotics appear to be characterized by low lipophilicity and high t-PSA32, 34, 43, the optimization strategy involved decreasing lipophilicity and increasing polarity. Nonetheless, the physicochemical properties of the 11 most active compounds were indistinguishable from inactive compounds with comparable potency in enzyme inhibition (MIC ≥ 30 μM): average cLogP = 3.7 and 3.5 while average t-PSA = 103 and 101 Å2 for active and inactive compounds, respectively. Thus decreasing lipophilicity/increasing t-PSA may not be a general strategy for antibiotic design. A survey of antibiotic discovery programs at AstraZeneca reached a similar conclusion33.

While we succeeded in identifying SaIMPDH inhibitors with potent in vitro antibacterial activity and low cytotoxicity, we were unable to demonstrate in vivo antibacterial activity. The pharmacokinetic characteristics of the tested compounds were not ideal, nonetheless our experiments suggest that inhibiting the enzymatic activity of SaIMPDH may not be an effective strategy to treat S. aureus infection in vivo. Moreover, resistance develops easily, and the resulting strains have similar virulence as the wild-type. While the emergence of resistance can be mitigated with combination therapy, this observation is particularly troubling because one strain expressed an enzyme with approximately 20% of wild-type activity. This observation suggests that inhibition must exceed 80% to block infection. Achieving and sustaining such high levels of inhibition is likely to be challenging in vivo. Worryingly, incomplete inhibition could conceivably promote infection: CodY, the master regulator of metabolism and repressor of virulence in S. aureus, is activated by GTP47. PurR, the master regulator of purine biosynthesis, also represses toxin expression48. Therefore reduction of the guanine nucleotide pools could conceivably increase virulence by relieving repression of toxin production.

The essentiality of SaIMPDH was surprising given that S. aureus also has the pathways required to salvage xanthine, xanthosine and guanosine, which would enable the production of guanine nucleotides in the absence of SaIMPDH. Xanthine is present at ~0.8 μM in mouse plasma49, and concentrations as high as 4.9 μM have been reported in human plasma50. Xanthosine (5 μM) and guanosine (0.8 μM) are also present in plasma, while guanine is undetectable. Therefore, we became concerned that the essentiality of SaIMPDH might not derive from enzymatic activity and examined the virulence of strains expressing catalytically inactive enzyme. These strains displayed a modest virulence defect, casting further doubt on the in vivo vulnerability of S. aureus to SaIMPDH inhibitors. Salvage pathways vary widely among bacterial pathogens, so IMPDH inhibitors may be effective treatment for other infections. For example, Mtb does not salvage xanthine, xanthosine or guanosine. Mtb does salvage guanine, but requires concentrations at the limit of solubility51.

Our experiments suggest that the essentiality of SaIMPDH may derive from extra-catalytic functions of IMPDH. SaIMPDH, like most IMPDHs, contains a regulatory domain of uncertain function in addition to the catalytic domain52. Deletion of CBS subdomain dysregulates the purine nucleotide pools in E. coli, leading to the toxic accumulation of ATP and other adenine nucleotides53, 54. Intriguingly, the antiproliferative effects of sanglifehrin A in mammalian cells derives from interactions with the CBS subdomain that do not affect catalytic activity55. Similar moonlighting functions of the CBS domain may be responsible for the essentiality of SaIMPDH. In addition, SaIMPDH has been reported to interact with 62 proteins, including the ribosome, tRNA synthetases, penicillin binding proteins, uridylate kinase, PRPP synthetase, adenylosuccinate synthetase and DNA helicase56. Perhaps the most intriguing SaIMPDH interactor is the transcription repressor PurR. The physiological consequences of these interactions are unknown. Given these myriad interactions, the failure of SaIMPDH inhibitors to phenocopy knockouts should perhaps not be surprising.

METHODS

Materials.

IMP, Tris, and common chemicals were purchased from Sigma-Aldrich (St. Louis, MO). NAD+ was purchased from Roche. The synthesis of the inhibitors is described in the Supporting Material. Values of clogP and t-PSA were calculated by FAFDrugs4 website39.

Expression and purification of SaIMPDH.

SaIMPDH was expressed as a N-terminal 6-His tagged protein using pET28b in BL21(DE3)ΔguaB bacteria. The cells were grown in LB media with 50 μg/ml of kanamycin at 37°C to an OD600 0.5–0.8. Protein expression was initiated by 0.25 mM isopropyl β-D-thiogalactoside (IPTG) and the culture was maintained at 30°C overnight. Cells were harvested, resuspended in lysis buffer [50 mM phosphate (pH 8.0), 500 mM KCl, 10 mM imidazole, 0.5 mM (tris(2-carboxyethyl)phosphine) (TCEP) and 10% glycerol], and stored at −80 °C. SaIMPDH was purified according to a standard protocol for His tagged protein. Protease inhibitor cocktail (Roche, Indianapolis, IN; 50 mL/g of wet cells) were added to the thawed cell suspension. The cells lysed on ice by sonication. The lysate was clarified by centrifugation at 18000g for 1 h and filtered through a 0.44 μm membrane. Clarified lysate was applied to a 5 mL Ni-NTA column (McLab, San Francisco CA). The column was washed with 10 column volume (CV) lysis buffer, 10 CV of lysis buffer containing 30 mM imidazole, and the protein was eluted with the same buffer containing 250 mM imidazole. SaIMPDH was dialyzed against 20 mM Hepes (pH 8.0), 150 mM KCl, 3 mM EDTA and 2 mM DTT, concentrated, flash-frozen, and stored in liquid nitrogen.

Enzyme assays.

IMPDH assays were performed in 50 mM Tris-HCl, pH 8.0, 100 mM KCl, 3 mM EDTA and 1 mM DTT. Activity was routinely assayed in the presence of 20–50 nM IMPDH at 25 °C. NADH production was monitored either by following absorbance change at wavelength 340 nm using a Hitachi U-2000 spectrophotometer (ε = 6.2 mM–1 cm–1) or by following fluorescence on a Biotek plate reader (excitation wavelength = 340 nm, emission wavelength = 460 nm). Each determination of Ki,app was derived from duplicate measurements of enzyme activity in the presence of twelve different inhibitor concentrations. Reported values are averages of at least two independent determinations. Enzyme was incubated with inhibitor (50 pM - 100 μM) for 10 min at room temperature prior to addition of substrates. The concentrations of IMP and NAD+ were fixed at 1.0 mM (saturating) and 1.0 mM (~2.5 × Km), respectively, for SaIMPDH. Ki,app values were calculated for each inhibitor according to Equation 1 (when Ki,app >> [E]) or Equation 2 (when Ki,app ~ [E]) using the SigmaPlot program (SPSS, Inc.):

vi=v0/(1 + [I]/Ki,app) (1)

where vi is initial velocity in the presence of inhibitor (I) and v0 is the initial velocity in the absence of inhibitor. The Morrison equation (Equation 2) was used to evaluate tight-binding inhibitors57.

vi/v0= {E0I0K*+ [(E0I0Ki,app)2+ 4E0Ki,app]0.5}/2E0 (2)

where vi is the initial velocity in the presence of inhibitor, v0 is the initial velocity in the absence of inhibitor, E0 is the total concentration of enzyme, I0 is the total concentration of inhibitor, and Ki,app is the apparent inhibition constant. Alternatively Dynafit was used to determine Ki,app58.

Antibacterial activity.

Broth micro-dilution MICs were performed following CLSI standard procedures against S. aureus (ATCC 13709, Smith). Stock compound solutions were prepared from powders at a concentration of 0.64 mg/mL (0.32 mg/mL for vancomycin) in 100% DMSO. Two-fold serial dilutions were performed in 96 well plates. The bacterial strain was prepared from an overnight culture plate by re-suspending in sterile saline and adjusting to a 0.5 McFarland standard confirmed by OD. The bacterial suspension was then diluted 1:100 in RPMI without phenol red or RPMI without phenol red containing 1 mM guanosine media. The prepared bacterial inoculum was then added to each corresponding 96 well plate at 190 uL per well, resulting in the top concentration of test article to be 32 μg/mL. The inoculated plates were incubated at 35°C for 18–20 h. The MICs were identified as the first well with no growth by visual inspection of the 96 well plates. Bacterial input inoculum was verified by plating the prepared suspension on TSA plates and incubating overnight resulting in 5.2×105 CFU/well.

Cytotoxicity.

All compounds were dissolved in DMSO and further diluted with culture medium before use in tissue culture assays (final DMSO concentrations were ≤ 0.1%). To determine cytotoxicity, LDH release was measured with the LDH Cytotoxicity Assay Kit (Pierce) according to manufacturer’s protocol. Briefly, 96 well plates were seeded with 13,000 HepG2 cells (all other cell lines seeded at 6,000 to 8,000 cells per well) and the cells were cultured for 24 h prior to drug treatment. The cells were incubated in 110 μL of EMEM containing compound or DMSO (vehicle only, control) for 24 h at 37°C. At least four wells from each plate were used as either ‘spontaneous’ LDH controls or as ‘maximum’ LDH controls per manufacturer’s instructions. Cytotoxicity was determined by measuring absorbance on a microplate reader. Data represent two independent experiments each performed in quadruplicate (n =8).

Plasma protein binding.

Compound (10 μM) was incubated in mouse plasma (BIOIVT, Westbury NY). The free fraction was collected by filtration through a 3K Microcon® centrifugal filter. The concentration of free inhibitor was determined by measuring inhibition of BaIMPDH.

Resistant strains.

Resistant strains were selected for growth on RPMI agar medium in the presence of 25 (25 μg/mL, 25xMIC). The S. aureus guaB gene is the third gene in a four gene operon, so sequencing focused on the coding region and did not include the promoter. Oligonucleotides SaF1 and SaR1 were used to amplify the coding sequence of guaB. DNA sequencing was performed with primers SaF1, SaR1 and internal primers SaF2 and SaR2. Sequences were deposited in GenBank with the following accession numbers: guaBwt, MW436410; guaB-G445S, MW436411 and guaB-Y446C, MW436412.

Name sequence purpose
SaF1 AATTGTTTTAGGTGCAATCTCTGC Outside gene
SaR1 TGGCTACCAAAGTCTAAGACAAGG Outside gene
SaF2 TTTTAACAAACCGCGACTTACG Inside gene
SaR2v2 TACCATGTTTGCGTGCTTCA Inside gene

Construction of guaB::C307A.

The C307A mutation was inserted guaB using overlapping PCR to construct a fragment with ~ 1 kb flanking sequence to facilitate homologous recombination. The PCR fragment was cloned into temperature-sensitive shuttle plasmid pMAD59 in IM08B, an E. coli DC10B derivative that methylates DNA according to the CC8 hsdMS methylation complex60. Plasmid DNA from this E. coli strain was directly transformed into NRS384. Transformants were selected by erythromycin (Erm, 5 μg/ml) resistance and blue color on X-gal at 30°C. Bacteria were grown in Erm-containing medium at 30°C for the isolation of plasmid to confirm the presence of right construct. Cells with right construct from 30°C cultures was diluted to grow at 43°C for overnight to promote single crossover (ErmR and light blue), and subsequently diluted to grow at 30°C without erythromycin containing medium to promote homologous recombination to yield white and ErmS colonies. Mutants were verified by chromosomal PCR followed by DNA sequencing to confirm the right mutation and the junction of the recombination. The guaB::C307A strains could not grow in the absence of a guanosine. This growth defect was complemented by pEPSA5 carrying xylose-inducible wild-type guaB.

In vivo studies.

All experiments were performed in accordance with institutional guidelines as defined by Institutional Animal Care and Use Committees of Brandeis University, Avastus Preclinical Services (Cambridge, MA) and NeoSome Life Sciences, LLC (Lexington, MA), as well as OLAW standards. Pharmacokinetic studies were performed by GVK Biosciences (Hyderbad India). Compounds were administered in a formulation of 20% DMSO/ 80% diluted liquid fill61. Liquid fill was prepared by combining 375 g of Etocas 35NF, 4.4 g of pluronic acid F68, 50.8 g of polyethylene glycol 400, and 10.8 ml of water and stirring overnight. Aliquots of 15 ml were stored at −80°C. Liquid fill was thawed as needed, and mixed with 33 ml of water to make a diluted liquid fill stock. Compounds were dissolved in DMSO and mixed 1 part to 4 parts diluted liquid fill to give a final dosage volume of 0.2 ml for a 20 g mouse.

Efficacy and virulence studies were performed by Avastus Preclinical Services (Cambridge, MA) and NeoSome Life Sciences, LLC (Lexington, MA). CD-1 female mice (18–20 g) were obtained from Charles River Laboratories and allowed to acclimate for 5 days prior to start of study. Animals were housed 5 per cage with free access to food and water. Bacteria were prepared from an overnight liquid culture. The overnight culture was diluted 1:10 and grown for another 5 h while shaking. The 5 h culture was serially diluted in 5% hog gastric mucin (pH=7.4) to a concentration to achieve at least 90% mortality within 48 hours after infection. Mice were infected with 0.5 ml of bacterial suspension via intraperitoneal injection, 5 mice per test article group (10 mice for the infection control groups). The test articles were delivered 1 h post infection as a slow bolus IV administration at a volume of 10 mL/kg. Infection control mice were dosed with test article vehicle.

Abscess experiments were performed at NeoSome Life Sciences, LLC (Waltham MA). Female CD-1 mice S. aureus ATCC 13709 (Smith), was prepared for infection from an overnight plate culture. A portion of the plate was resuspended in sterile saline and adjusted to an OD of 0.1 at 625 nm. The adjusted bacterial suspension was further diluted to target an infecting inoculum of 1.0×106 CFU/mouse, the actual inoculum size was 1.4×106 CFU/mouse. Plate counts of the inoculum were performed to confirm inoculum concentration. Under anesthesia, mice were injected subcutaneously in the right flank with 0.25 mL of the prepared bacterial inoculum mixed with cyclodextrin beads. Animals were returned to their home cages and allowed to recover from the anesthesia. Beginning at two hours post infection mice were dosed with either test article or positive control antibiotic. Mice receiving test compounds were dosed directly into the subcutaneous abscess in a 100 uL volume, a second dose of test agent were delivered 12 hours later. Linezolid served as the positive control agent and was delivered orally at 20 mg/kg. Five animals were dosed per group. One group of five mice were euthanized at initiation of therapy (T = 0) and CFUs determined. All remaining mice were euthanized at 26 h post infection. Abscesses were aseptically removed, homogenized to a uniform consistency, serially diluted and plated on bacterial growth media. The CFUs were enumerated after overnight incubation. The average and standard deviations for each group were determined.

Supplementary Material

Modi supporting

Table S1. Comparison of inhibition of BaIMPDH and SaIMPDH

Table S2. Crystal and Data Collection and Refinement Statistics.

Table S3. Virulence of Sa12 and Sa26.

Table S4. In vivo activity of SaIMPDH inhibitors.

Table S5. Virulence of guaB::C307A strains.

Table S6. List of strains related to guaB C307A mutations.

Table S7. Compound naming convention.

Figure S1. Physicochemical properties of antibacterial activity.

Figure S2. P226-resistant strain Sa12 displays a growth phenotype.

Figure S3. Cytotoxicity of select compounds.

Figure S4. S. aureus JE2 strains guaB::C307A require guanine or xanthine for growth.

Methods, including enzyme purification and crystallization, data collection and structure solution and refinement and virulence studies

Compound characterization

Acknowledgment:

The authors thank Dr. Susan Pochapsky (Brandeis University) for collecting 800 MHz spectra. MICs were determined at Vivisource (Cambridge, MA), Avastus Preclinical Services (Cambridge, MA) and NeoSome Life Sciences, LLC (Lexington, MA). Michael Cameron (Scripps Florida) performed the ADMET and PK experiments. This work was funded in part by the National Institute of Health (NIH)/National Institute of Allergy and Infectious Diseases (NIAID) grants 1R21AI138048 (L.H.) and R01AI125362 (G.D.C.) and contract HHSN272201700060C (AJ), by the U.S. Department of Energy (DOE) Office of Science under Contract No. DE-AC02-06CH11357operated for the DOE Office of Science by Argonne National Laboratory (AJ) and a Brandeis Sprout Grant (D.R.G.)

Abbreviations Used:

Ba

Bacillus anthracis

IMPDH

inosine 5’-monophosphate dehydrogenase

Mtb

Mycobacterium tuberculosis

Sa

Staphylococcus aureus

SAR

structure activity relationship

t-PSA

topological polar surface area

Footnotes

Supporting Information: The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.xxx.

PDB ID Codes: The atomic coordinates and structure factors are available in the Protein Data Bank (PDB). The accession codes are 5UUV, 5UUZ, 7MTX, 5URS, 7MTU, for BaIMPDHΔL•IMP•5, BaIMPDHΔL•IMP•19, BaIMPDHΔL•IMP•29, BaIMPDHΔL•IMP•30, and BaIMPDHΔL•IMP•31, respectively.

REFERENCES

  • (1).O’Neill J (2016) Tackling drug-resistant infections globally: final report and recommendations, Government of the United Kingdom. [Google Scholar]
  • (2).US Centers for Disease Control and Prevention, Antibiotic resistance threats in the United States, 2019, (US Centers for Disease Control and Prevention, 2019). [Google Scholar]
  • (3).Hedstrom L, Liechti G, Goldberg JB, and Gollapalli DR (2011) The antibiotic potential of prokaryotic IMP dehydrogenase inhibitors, Curr Med Chemistry 18, 1909–1918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (4).Griffin JE, Gawronski JD, Dejesus MA, Ioerger TR, Akerley BJ, and Sassetti CM (2011) High-resolution phenotypic profiling defines genes essential for mycobacterial growth and cholesterol catabolism, PLoS Pathog 7, e1002251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (5).van Opijnen T, and Camilli A (2012) A fine scale phenotype-genotype virulence map of a bacterial pathogen, Genome Res 22, 2541–2551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (6).Le Breton Y, Mistry P, Valdes KM, Quigley J, Kumar N, Tettelin H, and McIver KS (2013) Genome-wide identification of genes required for fitness of group A Streptococcus in human blood, Infect Immun 81, 862–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (7).Wang N, Ozer EA, Mandel MJ, and Hauser AR (2014) Genome-wide identification of Acinetobacter baumannii genes necessary for persistence in the lung, mBio 5, e01163–01114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (8).Valentino MD, Foulston L, Sadaka A, Kos VN, Villet RA, Santa Maria J Jr., Lazinski DW, Camilli A, Walker S, Hooper DC, and Gilmore MS (2014) Genes contributing to Staphylococcus aureus fitness in abscess- and infection-related ecologies, mBio 5, e01729–01714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (9).Truong QL, Cho Y, Barate AK, Kim S, Watarai M, and Hahn TW (2015) Mutation of purD and purF genes further attenuates Brucella abortus strain RB51, Microb Pathog 79, 1–7. [DOI] [PubMed] [Google Scholar]
  • (10).Santiago AE, Mann BJ, Qin A, Cunningham AL, Cole LE, Grassel C, Vogel SN, Levine MM, and Barry EM (2015) Characterization of Francisella tularensis Schu S4 defined mutants as live-attenuated vaccine candidates, Pathog Dis 73, ftv036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (11).Bachman MA, Breen P, Deornellas V, Mu Q, Zhao L, Wu W, Cavalcoli JD, and Mobley HL (2015) Genome-wide identification of Klebsiella pneumoniae fitness genes during lung infection, mBio 6, e00775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (12).Lee SA, Gallagher LA, Thongdee M, Staudinger BJ, Lippman S, Singh PK, and Manoil C (2015) General and condition-specific essential functions of Pseudomonas aeruginosa, Proc Natl Acad Sci U S A 112, 5189–5194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (13).DeJesus MA, Gerrick ER, Xu W, Park SW, Long JE, Boutte CC, Rubin EJ, Schnappinger D, Ehrt S, Fortune SM, Sassetti CM, and Ioerger TR (2017) Comprehensive essentiality analysis of the Mycobacterium tuberculosis genome via saturating transposon mutagenesis, mBio 8, e02133–02116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (14).Kofoed EM, Yan D, Katakam AK, Reichelt M, Lin B, Kim J, Park S, Date SV, Monk IR, Xu M, Austin CD, Maurer T, and Tan MW (2016) De novo guanine biosynthesis but not the riboswitch-regulated purine salvage pathway Is required for Staphylococcus aureus infection in vivo, J Bacteriol 198, 2001–2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (15).Makowska-Grzyska M, Kim Y, Maltseva N, Osipiuk J, Gu M, Zhang M, Mandapati K, Gollapalli DR, Gorla SK, Hedstrom L, and Joachimiak A (2015) A novel cofactor-binding mode in bacterial IMP dehydrogenases explains inhibitor selectivity, J Biol Chem 290, 5893–5911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (16).Gollapalli DR, Macpherson IS, Liechti G, Gorla SK, Goldberg JB, and Hedstrom L (2010) Structural determinants of inhibitor selectivity in prokaryotic IMP dehydrogenases, Chem Biol 17, 1084–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (17).Gorla SK, Kavitha M, Zhang M, Chin JE, Liu X, Striepen B, Makowska-Grzyska M, Kim Y, Joachimiak A, Hedstrom L, and Cuny GD (2013) Optimization of benzoxazole-based inhibitors of Cryptosporidium parvum inosine 5’-monophosphate dehydrogenase, J Med Chem 56, 4028–4043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (18).Gorla SK, Kavitha M, Zhang M, Liu X, Sharling L, Gollapalli DR, Striepen B, Hedstrom L, and Cuny GD (2012) Selective and potent urea inhibitors of Cryptosporidium parvum inosine 5’-monophosphate dehydrogenase, J Med Chem 55, 7759–7771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (19).Johnson CR, Gorla SK, Kavitha M, Zhang M, Liu X, Striepen B, Mead JR, Cuny GD, and Hedstrom L (2013) Phthalazinone inhibitors of inosine-5’-monophosphate dehydrogenase from Cryptosporidium parvum, Bioorg Med Chem Lett 23, 1004–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (20).Kirubakaran S, Gorla SK, Sharling L, Zhang M, Liu X, Ray SS, Macpherson IS, Striepen B, Hedstrom L, and Cuny GD (2012) Structure-activity relationship study of selective benzimidazole-based inhibitors of Cryptosporidium parvum IMPDH, Bioorg Med Chem Lett 22, 1985–1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (21).Maurya SK, Gollapalli DR, Kirubakaran S, Zhang M, Johnson CR, Benjamin NN, Hedstrom L, and Cuny GD (2009) Triazole inhibitors of Cryptosporidium parvum inosine 5’-monophosphate dehydrogenase, J Med Chem 52, 4623–4630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (22).Sun Z, Khan J, Makowska-Grzyska M, Zhang M, Cho JH, Suebsuwong C, Vo P, Gollapalli DR, Kim Y, Joachimiak A, Hedstrom L, and Cuny GD (2014) Synthesis, in vitro evaluation and cocrystal structure of 4-oxo-[1]benzopyrano[4,3-c]pyrazole Cryptosporidium parvum inosine 5’-monophosphate dehydrogenase (CpIMPDH) inhibitors, J Med Chem 57, 10544–10550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (23).Mandapati K, Gorla SK, House AL, McKenney ES, Zhang M, Rao SN, Gollapalli DR, Mann BJ, Goldberg JB, Cuny GD, Glomski IJ, and Hedstrom L (2014) Repurposing cryptosporidium inosine 5’-monophosphate dehydrogenase inhibitors as potential antibacterial agents, ACS Med Chem Lett 5, 846–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (24).Gorla SK, Zhang Y, Rabideau MM, Qin A, Chacko S, House AL, Johnson CR, Mandapati K, Bernstein HM, McKenney ES, Boshoff H, Zhang M, Glomski IJ, Goldberg JB, Cuny GD, Mann BJ, and Hedstrom L (2017) Benzoxazoles, Phthalazinones, and arylurea-based compounds with IMP dehydrogenase-independent antibacterial activity against Francisella tularensis, Antimicrob Agents Chemother 61, e00939–00917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (25).Makowska-Grzyska M, Kim Y, Gorla SK, Wei Y, Mandapati K, Zhang M, Maltseva N, Modi G, Boshoff HI, Gu M, Aldrich C, Cuny GD, Hedstrom L, and Joachimiak A (2015) Mycobacterium tuberculosis IMPDH in complexes with substrates, products and antitubercular compounds, PLoS One 10, e0138976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (26).Singh V, Donini S, Pacitto A, Sala C, Hartkoorn RC, Dhar N, Keri G, Ascher DB, Mondesert G, Vocat A, Lupien A, Sommer R, Vermet H, Lagrange S, Buechler J, Warner DF, McKinney JD, Pato J, Cole ST, Blundell TL, Rizzi M, and Mizrahi V (2017) The inosine monophosphate dehydrogenase, GuaB2, is a vulnerable new bactericidal drug target for tuberculosis, ACS Infect Dis 3, 5–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (27).Park Y, Pacitto A, Bayliss T, Cleghorn LA, Wang Z, Hartman T, Arora K, Ioerger TR, Sacchettini J, Rizzi M, Donini S, Blundell TL, Ascher DB, Rhee K, Breda A, Zhou N, Dartois V, Jonnala SR, Via LE, Mizrahi V, Epemolu O, Stojanovski L, Simeons F, Osuna-Cabello M, Ellis L, MacKenzie CJ, Smith AR, Davis SH, Murugesan D, Buchanan KI, Turner PA, Huggett M, Zuccotto F, Rebollo-Lopez MJ, Lafuente-Monasterio MJ, Sanz O, Diaz GS, Lelievre J, Ballell L, Selenski C, Axtman M, Ghidelli-Disse S, Pflaumer H, Bosche M, Drewes G, Freiberg GM, Kurnick MD, Srikumaran M, Kempf DJ, Green SR, Ray PC, Read K, Wyatt P, Barry CE 3rd, and Boshoff HI (2017) Essential but not vulnerable: indazole sulfonamides targeting inosine monophosphate dehydrogenase as potential leads against Mycobacterium tuberculosis, ACS Infect Dis 3, 18–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (28).Singh V, Pacitto A, Donini S, Ferraris DM, Boros S, Illyes E, Szokol B, Rizzi M, Blundell TL, Ascher DB, Pato J, and Mizrahi V (2019) Synthesis and structure-activity relationship of 1-(5-isoquinolinesulfonyl)piperazine analogues as inhibitors of Mycobacterium tuberculosis IMPDH, Eur J Med Chem 174, 309–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (29).Chacko S, Boshoff HIM, Singh V, Ferraris DM, Gollapalli DR, Zhang M, Lawson AP, Pepi MJ, Joachimiak A, Rizzi M, Mizrahi V, Cuny GD, and Hedstrom L (2018) Expanding benzoxazole-based inosine 5’-monophosphate dehydrogenase (IMPDH) inhibitor structure-activity as potential antituberculosis agents, J Med Chem 61, 4739–4756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (30).Trapero A, Pacitto A, Singh V, Sabbah M, Coyne AG, Mizrahi V, Blundell TL, Ascher DB, and Abell C (2018) Fragment-based approach to targeting inosine-5’-monophosphate dehydrogenase (IMPDH) from Mycobacterium tuberculosis, J Med Chem 61, 2806–2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (31).Usha V, Gurcha SS, Lovering AL, Lloyd AJ, Papaemmanouil A, Reynolds RC, and Besra GS (2011) Identification of novel diphenyl urea inhibitors of Mt-GuaB2 active against Mycobacterium tuberculosis, Microbiology (Reading, England) 157, 290–299. [DOI] [PubMed] [Google Scholar]
  • (32).O’Shea R, and Moser HE (2008) Physicochemical properties of antibacterial compounds: implications for drug discovery, J Med Chem 51, 2871–2878. [DOI] [PubMed] [Google Scholar]
  • (33).Brown DG, May-Dracka TL, Gagnon MM, and Tommasi R (2014) Trends and exceptions of physical properties on antibacterial activity for Gram-positive and Gram-negative pathogens, J Med Chem 57, 10144–10161. [DOI] [PubMed] [Google Scholar]
  • (34).Pawlowski AC, Johnson JW, and Wright GD (2016) Evolving medicinal chemistry strategies in antibiotic discovery, Curr Opin Biotechnol 42, 108–117. [DOI] [PubMed] [Google Scholar]
  • (35).Makowska-Grzyska M, Kim Y, Wu R, Wilton R, Gollapalli DR, Wang XK, Zhang R, Jedrzejczak R, Mack JC, Maltseva N, Mulligan R, Binkowski TA, Gornicki P, Kuhn ML, Anderson WF, Hedstrom L, and Joachimiak A (2012) Bacillus anthracis inosine 5’-monophosphate dehydrogenase in action: the first bacterial series of structures of phosphate ion-, substrate-, and product-bound complexes, Biochemistry 51, 6148–6163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (36).Alexandre T, Raynal B, and Munier-Lehmann H (2015) Two classes of bacterial IMPDHs according to their quaternary structures and catalytic properties, PLoS One 10, e0116578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (37).Basarab GS, Manchester JI, Bist S, Boriack-Sjodin PA, Dangel B, Illingworth R, Sherer BA, Sriram S, Uria-Nickelsen M, and Eakin AE (2013) Fragment-to-hit-to-lead discovery of a novel pyridylurea scaffold of ATP competitive dual targeting type II topoisomerase inhibiting antibacterial agents, J Med Chem 56, 8712–8735. [DOI] [PubMed] [Google Scholar]
  • (38).Manchester JI, Dussault DD, Rose JA, Boriack-Sjodin PA, Uria-Nickelsen M, Ioannidis G, Bist S, Fleming P, and Hull KG (2012) Discovery of a novel azaindole class of antibacterial agents targeting the ATPase domains of DNA gyrase and topoisomerase IV, Bioorg Med Chem Lett 22, 5150–5156. [DOI] [PubMed] [Google Scholar]
  • (39).Lagorce D, Bouslama L, Becot J, Miteva MA, and Villoutreix BO (2017) FAF-Drugs4: free ADME-tox filtering computations for chemical biology and early stages drug discovery, Bioinformatics 33, 3658–3660. [DOI] [PubMed] [Google Scholar]
  • (40).Chavis C, de Gourcy C, Dumont F, and Imbach J-L (1983) N. M. R. Studies of D-Ribosylamines in Solution: Derivatives of Primary Amines, Carbohydrate Research 113, 1–20. [Google Scholar]
  • (41).Ellis GP, and Williams JM (1981) The identification of the two crystalline N-phenyl-D-ribosylamines (“ribose anilides”) by n. m. r. spectroscopy, Carbohydr Res 95, 304–307. [Google Scholar]
  • (42).Ndombera FT, VanHecke GC, Nagi S, and Ahn YH (2016) Carbohydrate-based inducers of cellular stress for targeting cancer cells, Bioorg Med Chem Lett 26, 1452–1456. [DOI] [PubMed] [Google Scholar]
  • (43).Macielag MJ (2012) Chemical properties of antimicrobials and their uniqueness, In Antibiotic Discovery and Development (Dougherty TJ, and Pucci MJ, Eds.), pp 793–820, Springer US. [Google Scholar]
  • (44).Park JO, Rubin SA, Xu YF, Amador-Noguez D, Fan J, Shlomi T, and Rabinowitz JD (2016) Metabolite concentrations, fluxes and free energies imply efficient enzyme usage, Nat Chem Biol 12, 482–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (45).Kim Y, Makowska-Grzyska M, Gorla SK, Gollapalli DR, Cuny GD, Joachimiak A, and Hedstrom L (2015) Structure of Cryptosporidium IMP dehydrogenase bound to an inhibitor with in vivo antiparasitic activity, Acta Crystallogr F Struct Biol Commun 71, 531–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (46).MacPherson IS, Kirubakaran S, Gorla SK, Riera TV, D’Aquino JA, Zhang M, Cuny GD, and Hedstrom L (2010) The structural basis of Cryptosporidium-specific IMP dehydrogenase inhibitor selectivity, J Am Chem Soc 132, 1230–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (47).Richardson AR, Somerville GA, and Sonenshein AL (2015) Regulating the intersection of metabolism and pathogenesis in Gram-positive bacteria, Microbiol Spectr 3, MBP-0004–2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (48).Sause WE, Balasubramanian D, Irnov I, Copin R, Sullivan MJ, Sommerfield A, Chan R, Dhabaria A, Askenazi M, Ueberheide B, Shopsin B, van Bakel H, and Torres VJ (2019) The purine biosynthesis regulator PurR moonlights as a virulence regulator in Staphylococcus aureus, Proc Natl Acad Sci U S A 116, 13563–13572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (49).Simmonds RJ, and Harkness RA (1981) High-performance liquid chromatographic methods for base and nucleoside analysis in extracellular fluids and in cells, J Chromatogr 226, 369–381. [DOI] [PubMed] [Google Scholar]
  • (50).Traut TW (1994) Physiological concentrations of purines and pyrimidines, Mol Cell Biochem 140, 1–22. [DOI] [PubMed] [Google Scholar]
  • (51).Hedstrom L (2017) The Bare Essentials of Antibiotic Target Validation, ACS Infect Dis 3, 2–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (52).Hedstrom L (1999) IMP dehydrogenase: mechanism of action and inhibition, Curr. Med. Chem 6, 545–560. [PubMed] [Google Scholar]
  • (53).Pimkin M, and Markham GD (2008) The CBS subdomain of inosine 5’-monophosphate dehydrogenase regulates purine nucleotide turnover, Mol Microbiol 69, 342–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (54).Pimkin M, Pimkina J, and Markham GD (2009) A regulatory role of the Bateman domain of IMP dehydrogenase in adenylate nucleotide biosynthesis, J Biol Chem 284, 7960–7969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (55).Pua KH, Stiles DT, Sowa ME, and Verdine GL (2017) IMPDH2 Is an intracellular target of the cyclophilin A and sanglifehrin A complex, Cell Rep 18, 432–442. [DOI] [PubMed] [Google Scholar]
  • (56).Cherkasov A, Hsing M, Zoraghi R, Foster LJ, See RH, Stoynov N, Jiang J, Kaur S, Lian T, Jackson L, Gong H, Swayze R, Amandoron E, Hormozdiari F, Dao P, Sahinalp C, Santos-Filho O, Axerio-Cilies P, Byler K, McMaster WR, Brunham RC, Finlay BB, and Reiner NE (2011) Mapping the protein interaction network in methicillin-resistant Staphylococcus aureus, J Proteome Res 10, 1139–1150. [DOI] [PubMed] [Google Scholar]
  • (57).Morrison JF (1969) Kinetics of reversible inhibition of enzyme catalyzed reactions by tight binding inhibitors., Biochim. Biophys. Acta 185, 269–286. [DOI] [PubMed] [Google Scholar]
  • (58).Kuzmic P (2009) DynaFit--a software package for enzymology, Methods Enzymol 467, 247–280. [DOI] [PubMed] [Google Scholar]
  • (59).Arnaud M, Chastanet A, and Debarbouille M (2004) New vector for efficient allelic replacement in naturally nontransformable, low-GC-content, Gram-positive bacteria, Appl Environ Microbiol 70, 6887–6891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (60).Monk IR, Tree JJ, Howden BP, Stinear TP, and Foster TJ (2015) Complete bypass of restriction systems for major Staphylococcus aureus lineages, mBio 6, e00308–00315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • (61).Rothstein DM, Farquhar RS, Sirokman K, Sondergaard KL, Hazlett C, Doye AA, Gwathmey JK, Mullin S, van Duzer J, and Murphy CK (2006) Efficacy of novel rifamycin derivatives against rifamycin-sensitive and -resistant Staphylococcus aureus isolates in murine models of infection, Antimicrob Agents Chemother 50, 3658–3664. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Modi supporting

Table S1. Comparison of inhibition of BaIMPDH and SaIMPDH

Table S2. Crystal and Data Collection and Refinement Statistics.

Table S3. Virulence of Sa12 and Sa26.

Table S4. In vivo activity of SaIMPDH inhibitors.

Table S5. Virulence of guaB::C307A strains.

Table S6. List of strains related to guaB C307A mutations.

Table S7. Compound naming convention.

Figure S1. Physicochemical properties of antibacterial activity.

Figure S2. P226-resistant strain Sa12 displays a growth phenotype.

Figure S3. Cytotoxicity of select compounds.

Figure S4. S. aureus JE2 strains guaB::C307A require guanine or xanthine for growth.

Methods, including enzyme purification and crystallization, data collection and structure solution and refinement and virulence studies

Compound characterization

RESOURCES