Novel antimycobacterial compounds suppress ... - ACS Publications

Apr 10, 2019 - Andrei L Osterman , Irina Rodionova , Xiaoqing Li , Edward Sergienko , Chen-Ting Ma , Antonino Catanzaro , Mark E. Pettigrove , Robert ...
0 downloads 0 Views 3MB Size
Subscriber access provided by CLARKSON UNIV

Article

Novel antimycobacterial compounds suppress NAD biogenesis by targeting a unique pocket of NaMN adenylyltransferase Andrei L Osterman, Irina Rodionova, Xiaoqing Li, Edward Sergienko, ChenTing Ma, Antonino Catanzaro, Mark E. Pettigrove, Robert W Reed, Rashmi Gupta, Kyle H. Rohde, Konstantin V Korotkov, and LEONARDO SORCI ACS Chem. Biol., Just Accepted Manuscript • DOI: 10.1021/acschembio.9b00124 • Publication Date (Web): 10 Apr 2019 Downloaded from http://pubs.acs.org on April 11, 2019

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 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 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.

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 33 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 Chemical Biology

1

Novel antimycobacterial compounds suppress NAD biogenesis by targeting

2

a unique pocket of NaMN adenylyltransferase

3 4

Andrei L. Osterman1, Irina Rodionova1,†, Xiaoqing Li1, Eduard Sergienko2, Chen-

5

Ting Ma2, Antonino Catanzaro3, Mark E. Pettigrove3, Robert W. Reed4,‡, Rashmi

6

Gupta5, Kyle H. Rohde5, Konstantin V. Korotkov4,*, and Leonardo Sorci6,*

7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30

1

Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States 2 NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States 3 Department of Medicine, University of California San Diego, La Jolla, CA, United States 4 Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, United States 5 Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States 6 Department of Materials, Environmental Sciences and Urban Planning, Division of Bioinformatics and Biochemistry, Polytechnic University of Marche, Italy †

present address: Department of Molecular Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States ‡ present address: Division of Regulatory Services, College of Agriculture, Food and Environment, University of Kentucky, Lexington, Kentucky, United States * Correspondence to: Leonardo Sorci, [email protected]; Konstantin V. Korotkov, [email protected].

1 ACS Paragon Plus Environment

ACS Chemical Biology 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 2 of 33

31

ABSTRACT

32

Conventional treatments to combat the tuberculosis (TB) epidemic are falling short,

33

thus encouraging the search for novel antitubercular drugs acting on unexplored

34

molecular targets.

35

bioactive compounds with potent antitubercular activity. However, their cellular

36

target and mechanism of action remain largely unknown. Further evaluation of

37

these compounds may include their screening in search for known antitubercular

38

drug targets hits. Here, a collection of nearly 1,400 mycobactericidal compounds

39

was screened against Mycobacterium tuberculosis NaMN adenylyltransferase

40

(MtNadD), a key enzyme in the biogenesis of NAD cofactor that was recently

41

validated as a new drug target for dormant and active tuberculosis. We found three

42

chemotypes that efficiently inhibit MtNadD in the low micromolar range in vitro.

43

SAR and cheminformatics studies of commercially available analogs point to a

44

series of benzimidazolium derivatives, here named N2, with bactericidal activity on

45

different

46

tuberculosis, and dormant M. smegmatis. The on-target activity was supported by

47

the increased resistance of an M. smegmatis strain overexpressing the target and

48

by a rapid decline in NAD(H) levels. A co-crystal structure of MtNadD with N2-8

49

inhibitor reveals that the binding of the inhibitor induced the formation of a new

50

quaternary structure, a dimer-of-dimers where two copies of the inhibitor occupy

51

symmetrical positions in the dimer interface, thus paving the way for the

52

development of a new generation of selective MtNadD bioactive inhibitors. All

Several whole cell phenotypic screenings have delivered

mycobacteria,

including

M.

abscessus,

2 ACS Paragon Plus Environment

multidrug-resistant

M.

Page 3 of 33 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 Chemical Biology

53

these results strongly suggest that pharmacological inhibition of MtNadD is an

54

effective strategy to combat dormant and resistant Mtb strains.

55 56

INTRODUCTION

57 58

Despite extensive prevention and control measures in the last two decades,

59

Tuberculosis yet represents a dramatic health issue with global impact. The World

60

Health Organization (WHO) estimates over ten million active disease cases in

61

2016

62

immunocompromised HIV-positive individuals1. Mycobacterium tuberculosis

63

(Mtb), which causes TB, undergoes a metabolic shift in the human host into a

64

dormant state resulting in long-term persistent infection and phenotypic resistance

65

to many otherwise successful drugs2-4. Drug-susceptible strains can be effectively

66

treated with a 6 to 9-month regimen of multiple antibiotics, but the non-adherence

67

to the therapeutic guidelines on a global level has exacerbated the selection and

68

spreading of resistant strains in the last two decades5. Particularly alarming is the

69

increase of multidrug-resistant tuberculosis (MDR-TB), resistant to isoniazid and

70

rifampicin, two of the most powerful TB drugs, and extensively drug-resistant

71

(XDR-TB), which are additionally resistant to any fluoroquinolone, and to at least

72

one of the second line drugs (amikacin, capreomycin or kanamycin)6.

73

Consensus is growing that novel, much-needed antibiotics, should efficiently kill

74

drug-resistant strains and dormant mycobacteria as well as shorten therapy via

75

novel targets2, 3.

and

nearly

1.7

million

deaths,

22%

3 ACS Paragon Plus Environment

of

which

were

among

ACS Chemical Biology 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

76

Two novel classes of antibiotics, diarylquinolines and nitroimidazoles, have

77

reached stage III of clinical trials, proving to be effective alone in treating latent

78

tuberculosis (LTB)

79

diarylquinolines, is the first new drug in decades to be clinically approved for TB

80

treatment. In particular, due to its unique mechanism of action on mycobacterial

81

ATP synthase, bedaquiline opened a new door for discovering anti-TB/LTB drugs

82

using oxidative phosphorylation as a target system11. However, resistance to BDQ

83

has already been observed. Besides the selection of mutations of the atpE subunit

84

of target ATP synthase, drug-responsive mechanisms involve activation of

85

dormancy and metabolic remodeling

86

from substrate-level phosphorylation appears to enable transient bacterial survival,

87

as further confirmed by BDQ-enhanced killing of mycobacteria on non-fermentable

88

energy sources13. Glycolysis not only produces ATP faster, albeit less efficiently,

89

than oxidative phosphorylation but also supplies metabolic precursors required for

90

macromolecular biosynthesis. Strikingly, glycolytic enzymes are upregulated in

91

BDQ-treated mycobacteria, including lactate dehydrogenase that recycles NADH

92

to NAD13.

93

aspects of energy metabolism that are essential for both actively replicating and

94

dormant forms of mycobacteria.

95

Biogenesis and homeostasis of redox cofactors, most notably of NAD pool

96

(NAD(H) and NADP(H)), is another crucial aspect of respiratory energy

97

metabolism in both, replicating and dormant forms of mycobacteria 14, 15.

7-10.

Bedaquiline (BDQ), the first-in-class compound of

12, 13.

Notably, alternative ATP production

These observations emphasize the significance of targeting other

4 ACS Paragon Plus Environment

Page 4 of 33

Page 5 of 33 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 Chemical Biology

98

NAD(H) is the entry electron donor in the respiratory chain and the key oxidant

99

driving the glycolysis. Thus, depletion of NAD(H) pool generates a glycolytic

100

slowdown

101

synthesis from both sources.

102

In this view, NAD starvation, which represents alone a validated strategy to kill

103

mycobacteria recalcitrant to current therapies17, 18, may potentially show synergy

104

with drugs targeting ATP synthesis such as bedaquiline.

105

The universally conserved and essential enzymes, NadD, NadE, and NadF, which

106

drive the last non-redundant steps of NAD(P) biosynthesis, were implicated by our

107

previous genomics-driven studies as potential drug targets in numerous bacterial

108

pathogens19-22. Of these three enzymes, NadD is the most divergent from its

109

human counterparts (NMNAT1-323), offering the opportunity of developing

110

selective small-molecule inhibitors22, 24-27. Our recent studies showed that induced

111

degradation of NadD and NadE enzymes in a model system of M. smegmatis

112

(Msm) led to rapid depletion of the NAD cofactor pool followed by cell death, even

113

under non-replicative conditions17. Another group used a similar approach to show

114

that induction of NadE degradation also suppressed acute and chronic Mtb

115

infection in mice18. These findings provided the ultimate validation of NAD

116

metabolism as a target pathway for the development of new antimycobacterial

117

therapies. Structure-function and inhibition studies of the key NAD(P) biosynthetic

118

enzymes 22, 24, 25, 28, 29 including our recently published work on MtNadD26 support

119

their druggability.

16

and rapid shutdown of electron transfer chain thereby affecting ATP

5 ACS Paragon Plus Environment

ACS Chemical Biology 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 6 of 33

120

We set out to identify inhibitors of MtNadD through a target-based screen

121

of orphan compounds with mycobactericidal activity. Here, we report the

122

identification as well as the biochemical and biological characterization of several

123

potent inhibitors of NadD with bactericidal activity on mycobacteria, including M.

124

abscessus, multidrug-resistant M. tuberculosis, as well as a prominent bactericidal

125

effect under non-replicating conditions in M. smegmatis. To clarify in atomic detail

126

the mode of action of these compounds and their structure-activity relationships,

127

we also determined the crystal structure of MtNadD in complex with one of these

128

inhibitors.

129 130

RESULTS AND DISCUSSION

131 132

Screening of bioactive antimycobacterial compounds affords novel MtNadD

133

inhibitors. Previous whole cell phenotypic screening has identified potent anti-

134

mycobacterial compounds in a variety of tested conditions on replicating, non-

135

replicating and intracellular forms of Mtb. These compounds were selected via

136

several cell-based HTS campaigns as reported in PubChem30,

137

mechanisms and molecular targets remain unknown. We speculated that, given a

138

limited space of potential targets (~200–300 essential genes in M. tuberculosis),

139

at least some of these compounds could target the essential enzymes of NAD

140

metabolism. For this reason, a collection of 1389 bioactive compounds, kindly

141

provided by Global Alliance for TB Drug Development (TB Alliance) was tested

142

against the recombinant M. tuberculosis NadD enzyme using colorimetric 6 ACS Paragon Plus Environment

31,

but their

Page 7 of 33 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 Chemical Biology

143

detection assay of released PPi (Figure 1). The primary assays, performed at a

144

fixed final concentration of 25 µM for each compound, and at subsaturating

145

concentrations of ATP and NaMN, identified 16 compounds (~1% hit rate)

146

inhibiting more than 30% of enzymatic activity, with three of these demonstrating

147

>50% inhibition. Notably, this hit rate is comparable to our previous structure-

148

based approach on NadD from Escherichia coli and Staphylococcus aureus

149

which, however, used a more stringent concentration of the inhibitor (100 µM).

150

These primary hits were repurchased from different vendors, and their

151

concentration-response profiling yielded 5 compounds with IC50 values in the

152

range of 6–22 µM (Supplementary Table 1). The three top-ranked compounds with

153

significant structural diversity, termed here as N1, N2, and N3, were selected for

154

further analysis, including cheminformatics and SAR studies by analogs’ testing,

155

cell-based assays, and co-crystallization trials (Figure 1).

24

156 157

SAR for chemotypes N1, N2, and N3. Before any further investigation on these

158

chemotypes was pursued, we wanted to confirm their bioactivity as initially

159

displayed in whole cell phenotypic assay. For example, we determined a minimal

160

inhibitory concentration of 28 µM of the compound N2-11 for M. tuberculosis

161

H37Rv. This result is consistent with the inhibition value reported in PubChem

162

Bioassay 1626 (MIC of 16 µM). Overall, around 30 analogs of the three selected

163

chemotypes were purchased from different vendors and tested in vitro against

164

MtNadD (Figure 2 and Tables 1–3). Despite a limited chemical search exploration,

165

four analogs show improved MtNadD inhibition relative to their parent compound, 7 ACS Paragon Plus Environment

ACS Chemical Biology 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

166

with up to 5-fold lower IC50 (2.5 µM) for N1-11 analog, which represents the most

167

potent inhibitor for MtNadD described so far 26.

168

In class N1, the replacement of a 6-fluoro benzyl group with the more

169

hydrophobic phenylethyl (N1-11) or benzyl (N2-11) moieties yielded the best

170

improvement. More polar moieties such as five or six-membered rings with two or

171

more heteroatoms were poorly tolerated (IC50>50–100 µM), while linear or

172

branched hydrocarbon chains (N1-6 through N1-8), or the smaller cyclopropyl

173

moieties (N1-9) were well-tolerated substitutions (15 µM 100 >50 >50 >50

54.81 54.28 79.9 97.0 22.14 11.37 14.68 – – 20.43 – – 38.2 – – 41.76 – – – 14.6 – – – – –

Mycobacterium Growth inhibition (%) at MIC (µM) 50 µM tuberculosis smegmatisc e d

20 ACS Paragon Plus Environment

– – – – – nd nd ~50 ~20 nd ~50 nd nd nd nd ~20 nd ~50 ~100 ~10 ~20 nd nd nd nd

– – – – – – – – – 100 – – – – – – – – –

>25 50 25 25 >25 nd >25 – – 25 – – 25 – – nd – – – nd – – – – –

Page 21 of 33 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 Chemical Biology

N1-26 N1-27 N1-28 N1-29 N1-30 N1-31

1423254 1897037 3774654 658395 16682092 3333039

>50 >100 >100 – >100 –

– – – 19.8 22.17 54.21

nd nd nd – ~20 –

– – – >10 >100 –

– – – 25

447 448

a

449

Standard errors are reported. b in vitro inhibition of M. tuberculosis H37Rv fructose-

450

bisphosphate aldolase (FBA), as reported in PubChem Bioassay AID 588726.

451

Antibacterial activity against M. smegmatis is expressed as a percentage of growth

452

suppression at fixed 50 µM concentration, except for N1-18 that was tested at 20

453

µM.

454

obtained from PubChem Bioassay AID 449762 data. “ – “ , not assayed; nd, no

455

detected effect. In red, compounds identified in the primary screening. In blue,

456

analogs identified via cheminformatics search for which bioactivity is reported, but

457

not tested in our study.

Assays performed in duplicate in at least five different inhibitors concentration.

d

values are deduced from PubChem Bioassay AID 1626 data.

e

c

values

458 459

Table 2. Inhibitory properties of N2 class MtNadD inhibitors evaluated

460 ID

PubChem CID

MtNadDa IC50 (µM)

N2-1 N2-2 N2-3 N2-4 N2-5 N2-6 N2-7 N2-8 N2-9

2834410 2834412 2834423 2834427 2834425 2834414 2834416 2834419 16192954

5±1 111±16 – – – 46±16 13±3 18±2 18±3

Mycobacterium Growth inhibition, MIC (µM) tuberculosis smegmatis abscessus b c 5 25 – – – 25 5 5 5

46.7 – – – – I – 32.7 – I – I – 16.6 21.2 I 38.5 10.4

21 ACS Paragon Plus Environment

– – I 28 I I – I I

61.4 – – – – – – 26.7 26.2

ACS Chemical Biology 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

461

N2-10 N2-11

3908383 16195070

20±3 6±1

5 5

Page 22 of 33

19.7 – 28.2 15.9

– I

25.6 26

462

aAssays

463

Standard errors are reported. “–“ , not assayed; nd, no detected effect; b,c, values

464

deduced from PubChem Bioassay AID 1626 and AID 449762 data, respectively.

465

For consistency, these values represent the minimal inhibitory concentration

466

yielding >99% growth suppression and were obtained by fitting the reported EC50

467

and Hill Slope values into ECanything built-in equation in Prism 7.0. I, compounds

468

tagged as “Inconclusive” and which showed >98% inhibition at a single

469

concentration of 25 µM”. No dose-response analysis was carried out. ID color-

470

coding is as in Table 1.

performed in duplicate in at least five different inhibitors concentration.

471 472

Table 3. Inhibitory properties of N3 class MtNadD inhibitors. ID

473

N3-1 N3-2 N3-3

MtNadD IC50 (µM) 11±1 96±26 >100

Growth inhibition, MIC (µM) M. smegmatis

M. tuberculosis

M. abscessus

10 >50 nd

3.1 >100 –

20.9 >100 –

474

Assays performed in duplicate in at least five different inhibitors concentration.

475

Standard errors are reported. “ – “ , not assayed; nd, no detected effect (at 50

476

µM). ID color- coding is as in Table 1 and 2.

477

Table 4. Data collection and refinement statistics. M. tuberculosis NadD in complex with N2-8 (PDB 6BUV) Data collection Wavelength (Å)

1.0000 22 ACS Paragon Plus Environment

Page 23 of 33 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 Chemical Biology

Space group Cell dimensions: a, b, c (Å)    () Resolution (Å) Rsym CC1/2b I / I Completeness (%) Multiplicity Refinement Resolution (Å) No. reflections (total / free) Rwork / Rfree Number of atoms: Protein Ligand/ion Water B-factors: Protein Ligand/ion Water All atoms Wilson B R.m.s. deviations: Bond lengths (Å) Bond angles () Ramachandran distributionc (%): Favored Allowed Outliers Rotamer outliersc (%) Clashscored MolProbity scoree

H32 163.73, 163.73, 153.65 90, 90, 120 52.1–1.86 (1.91–1.86)a 0.077 (1.146) 99.6 (54.7) 11.08 (1.72) 99.3 (100.0) 3.8 (3.8) 52.1–1.86 65711 / 5923 0.172 / 0.192 2976 38 296 43.7 60.7 49.6 44.4 33.4 0.008 1.008 98.9 1.1 0 0 2.53 1.04

478 479

aValues

480

bCC

481

by XSCALE 44.

482

cCalculated

483

dClashscore

1/2

in parentheses are for the highest-resolution shell.

correlation coefficient as defined in Karplus & Diederichs 43 and calculated

using the MolProbity server (http://molprobity.biochem.duke.edu) 45. is the number of serious steric overlaps (> 0.4 Å) per 1000 atoms. 23 ACS Paragon Plus Environment

ACS Chemical Biology 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

484

eMolProbity

485

evaluations into a single score, normalized to be on the same scale as X-ray

486

resolution 45.

score combines the clashscore, rotamer, and Ramachandran

487 488

FIGURE LEGENDS

489

Figure 1. Flowchart of the whole-cell and target screening approach for

490

delivering novel antitubercular NaMN adenylyltransferase inhibitors.

491 492

Figure 2. Chemical diversity of the three series of novel MtNadD inhibitors

493

evaluated.

494

In red, molecules that were identified in our primary screen of the TB Alliance

495

bioactive library against MtNadD (IC50 < 20 µM). In black, novel analogs for each

496

chemotype tested in this study. In blue, analogs that were not tested in our study,

497

but for which a relevant activity against M. tuberculosis or an Mtb target enzyme

498

has been reported in PubChem (see Tables 1–2 and main text for details).

499 500

Figure 3. Bactericidal, on-target effects of N2 inhibitors in replicating and

501

non-replicating M. smegmatis.

502

(A–C) Growth suppression curves at different N2 series inhibitors’ concentrations

503

underline their killing activity on replicating Msm at above 5 µM. DMSO at 1 %

504

represents the “no inhibitor” control. Linezolid at 3 µM was used as a positive

505

control. (D) Addition of the inhibitor at ~ 5xMIC concentration at log-phase (after

506

12.5 hours of growth) stops the growth of Msm and (E) rapidly deplete NAD pool.

24 ACS Paragon Plus Environment

Page 24 of 33

Page 25 of 33 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 Chemical Biology

507

Residual levels of NAD were measured by a colorimetric detection kit and

508

normalized by total protein in samples taken for 6 hours, in 2 hours’ time interval.

509

(F) Cell viability by CFU assay after 1–4 days of incubation of Msm with 5xMIC

510

inhibitor (25 µM) under carbon starvation conditions. In this case, Linezolid was

511

used at 15 µM.

512 513

Figure 4. Structure of MtNadD in complex with inhibitor N2-8.

514

(A) N2-8 inhibitor-driven dimer-of-dimers of MtNadD with two molecules of the

515

ligand bridging the dimers formed by chains B and A’, and chains B’ and A,

516

respectively. (B) A close-up view of the N2-8 binding site. The hydrophobic

517

residues lining up the inhibitor binding site are shown in sticks representation. A-

518

weighted 2FO–FC electron density map contoured at 1 is shown as blue mesh.

519

(C) Comparison of the closed active-site conformation of MtNadD•N2-8 complex

520

(light blue) with MtNadD apo structure (yellow) (PDB ID, 4X0E) and

521

MaNadD•NaAD complex structure (purple) (PDB ID, 5DEO). H-bonding between

522

D109 and the adenosine moiety of the MaNadD•NaAD complex is shown as

523

dashed line. The steric clashes of L164 with adenine ring, I113 with nicotinate ring,

524

and G106 with the AMP ribose are marked by a dotted circle.

525 526 527 528 529 530 25 ACS Paragon Plus Environment

ACS Chemical Biology 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

531

REFERENCES

532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585

[1] WHO. (2017) Global tuberculosis report. [2] Fattorini, L., Piccaro, G., Mustazzolu, A., and Giannoni, F. (2013) Targeting dormant bacilli to fight tuberculosis, Mediterr J Hematol Infect Dis 5, e2013072. [3] Gengenbacher, M., and Kaufmann, S. H. (2012) Mycobacterium tuberculosis: success through dormancy, FEMS Microbiol Rev 36, 514-532. [4] Rohde, K., Yates, R. M., Purdy, G. E., and Russell, D. G. (2007) Mycobacterium tuberculosis and the environment within the phagosome, Immunol Rev 219, 37-54. [5] Trauner, A., Borrell, S., Reither, K., and Gagneux, S. (2014) Evolution of drug resistance in tuberculosis: recent progress and implications for diagnosis and therapy, Drugs 74, 10631072. [6] Gandhi, N. R., Nunn, P., Dheda, K., Schaaf, H. S., Zignol, M., van Soolingen, D., Jensen, P., and Bayona, J. (2010) Multidrug-resistant and extensively drug-resistant tuberculosis: a threat to global control of tuberculosis, Lancet 375, 1830-1843. [7] Andries, K., Verhasselt, P., Guillemont, J., Gohlmann, H. W., Neefs, J. M., Winkler, H., Van Gestel, J., Timmerman, P., Zhu, M., Lee, E., Williams, P., de Chaffoy, D., Huitric, E., Hoffner, S., Cambau, E., Truffot-Pernot, C., Lounis, N., and Jarlier, V. (2005) A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis, Science 307, 223-227. [8] Koul, A., Dendouga, N., Vergauwen, K., Molenberghs, B., Vranckx, L., Willebrords, R., Ristic, Z., Lill, H., Dorange, I., Guillemont, J., Bald, D., and Andries, K. (2007) Diarylquinolines target subunit c of mycobacterial ATP synthase, Nat Chem Biol 3, 323-324. [9] Chen, X., Hashizume, H., Tomishige, T., Nakamura, I., Matsuba, M., Fujiwara, M., Kitamoto, R., Hanaki, E., Ohba, Y., and Matsumoto, M. (2017) Delamanid Kills Dormant Mycobacteria In Vitro and in a Guinea Pig Model of Tuberculosis, Antimicrob Agents Chemother 61. [10] Xavier, A. S., and Lakshmanan, M. (2014) Delamanid: A new armor in combating drugresistant tuberculosis, J Pharmacol Pharmacother 5, 222-224. [11] Cook, G. M., Hards, K., Dunn, E., Heikal, A., Nakatani, Y., Greening, C., Crick, D. C., Fontes, F. L., Pethe, K., Hasenoehrl, E., and Berney, M. (2017) Oxidative Phosphorylation as a Target Space for Tuberculosis: Success, Caution, and Future Directions, Microbiol Spectr 5. [12] Huitric, E., Verhasselt, P., Koul, A., Andries, K., Hoffner, S., and Andersson, D. I. (2010) Rates and mechanisms of resistance development in Mycobacterium tuberculosis to a novel diarylquinoline ATP synthase inhibitor, Antimicrob Agents Chemother 54, 10221028. [13] Koul, A., Vranckx, L., Dhar, N., Gohlmann, H. W., Ozdemir, E., Neefs, J. M., Schulz, M., Lu, P., Mortz, E., McKinney, J. D., Andries, K., and Bald, D. (2014) Delayed bactericidal response of Mycobacterium tuberculosis to bedaquiline involves remodelling of bacterial metabolism, Nat Commun 5, 3369. [14] Boshoff, H. I., Xu, X., Tahlan, K., Dowd, C. S., Pethe, K., Camacho, L. R., Park, T. H., Yun, C. S., Schnappinger, D., Ehrt, S., Williams, K. J., and Barry, C. E., 3rd. (2008) Biosynthesis and recycling of nicotinamide cofactors in mycobacterium tuberculosis. An essential role for NAD in nonreplicating bacilli, J Biol Chem 283, 19329-19341. [15] Vilcheze, C., Weinrick, B., Wong, K. W., Chen, B., and Jacobs, W. R., Jr. (2010) NAD+ auxotrophy is bacteriocidal for the tubercle bacilli, Mol Microbiol 76, 365-377. [16] Ying, W., Chen, Y., Alano, C. C., and Swanson, R. A. (2002) Tricarboxylic acid cycle substrates prevent PARP-mediated death of neurons and astrocytes, J Cereb Blood Flow Metab 22, 774-779. [17] Rodionova, I. A., Schuster, B. M., Guinn, K. M., Sorci, L., Scott, D. A., Li, X., Kheterpal, I., Shoen, C., Cynamon, M., Locher, C., Rubin, E. J., and Osterman, A. L. (2014) Metabolic and bactericidal effects of targeted suppression of NadD and NadE enzymes in mycobacteria, MBio 5, e00747-00713. [18] Kim, J. H., O'Brien, K. M., Sharma, R., Boshoff, H. I., Rehren, G., Chakraborty, S., Wallach, J. B., Monteleone, M., Wilson, D. J., Aldrich, C. C., Barry, C. E., 3rd, Rhee, K. Y., Ehrt,

26 ACS Paragon Plus Environment

Page 26 of 33

Page 27 of 33 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

586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640

ACS Chemical Biology

S., and Schnappinger, D. (2013) A genetic strategy to identify targets for the development of drugs that prevent bacterial persistence, Proc Natl Acad Sci U S A 110, 19095-19100. [19] Gerdes, S. Y., Scholle, M. D., D'Souza, M., Bernal, A., Baev, M. V., Farrell, M., Kurnasov, O. V., Daugherty, M. D., Mseeh, F., Polanuyer, B. M., Campbell, J. W., Anantha, S., Shatalin, K. Y., Chowdhury, S. A., Fonstein, M. Y., and Osterman, A. L. (2002) From genetic footprinting to antimicrobial drug targets: examples in cofactor biosynthetic pathways, J Bacteriol 184, 4555-4572. [20] Osterman, A. L., and Begley, T. P. (2007) A subsystems-based approach to the identification of drug targets in bacterial pathogens, Prog Drug Res 64, 131, 133-170. [21] Sorci, L., Blaby, I., De Ingeniis, J., Gerdes, S., Raffaelli, N., de Crecy Lagard, V., and Osterman, A. (2010) Genomics-driven reconstruction of acinetobacter NAD metabolism: insights for antibacterial target selection, J Biol Chem 285, 39490-39499. [22] Sorci, L., Blaby, I. K., Rodionova, I. A., De Ingeniis, J., Tkachenko, S., de Crecy-Lagard, V., and Osterman, A. L. (2013) Quinolinate salvage and insights for targeting NAD biosynthesis in group A streptococci, J Bacteriol 195, 726-732. [23] Sorci, L., Kurnasov, O., Rodionov, D. A., and Osterman, A. L. (2010) Genomics and Enzymology of NAD Biosynthesis, In Comprehensive Natural Products II (Lew, M., and Hung-Wen, L., Eds.), pp 213-257, Elsevier, Oxford. [24] Sorci, L., Pan, Y., Eyobo, Y., Rodionova, I., Huang, N., Kurnasov, O., Zhong, S., MacKerell, A. D., Jr., Zhang, H., and Osterman, A. L. (2009) Targeting NAD biosynthesis in bacterial pathogens: Structure-based development of inhibitors of nicotinate mononucleotide adenylyltransferase NadD, Chem Biol 16, 849-861. [25] Huang, N., Kolhatkar, R., Eyobo, Y., Sorci, L., Rodionova, I., Osterman, A. L., Mackerell, A. D., and Zhang, H. (2010) Complexes of bacterial nicotinate mononucleotide adenylyltransferase with inhibitors: implication for structure-based drug design and improvement, J Med Chem 53, 5229-5239. [26] Rodionova, I. A., Zuccola, H. J., Sorci, L., Aleshin, A. E., Kazanov, M. D., Ma, C. T., Sergienko, E., Rubin, E. J., Locher, C. P., and Osterman, A. L. (2015) Mycobacterial nicotinate mononucleotide adenylyltransferase: structure, mechanism, and implications for drug discovery, J Biol Chem 290, 7693-7706. [27] Orsomando, G., Agostinelli, S., Bramucci, M., Cappellacci, L., Damiano, S., Lupidi, G., Maggi, F., Ngahang Kamte, S. L., Biapa Nya, P. C., Papa, F., Petrelli, D., Quassinti, L., Sorci, L., Vitali, L. A., and Petrelli, R. (2016) Mexican sunflower (Tithonia diversifolia, Asteraceae) volatile oil as a selective inhibitor of Staphylococcus aureus nicotinate mononucleotide adenylyltransferase (NadD), Articolo in rivista 85, 181-189. [28] Moro, W. B., Yang, Z., Kane, T. A., Brouillette, C. G., and Brouillette, W. J. (2009) Virtual screening to identify lead inhibitors for bacterial NAD synthetase (NADs), Bioorg Med Chem Lett 19, 2001-2005. [29] Petrelli, R., Sham, Y. Y., Chen, L., Felczak, K., Bennett, E., Wilson, D., Aldrich, C., Yu, J. S., Cappellacci, L., Franchetti, P., Grifantini, M., Mazzola, F., Di Stefano, M., Magni, G., and Pankiewicz, K. W. (2009) Selective inhibition of nicotinamide adenine dinucleotide kinases by dinucleoside disulfide mimics of nicotinamide adenine dinucleotide analogues, Bioorg Med Chem 17, 5656-5664. [30] Ananthan, S., Faaleolea, E. R., Goldman, R. C., Hobrath, J. V., Kwong, C. D., Laughon, B. E., Maddry, J. A., Mehta, A., Rasmussen, L., Reynolds, R. C., Secrist, J. A., 3rd, Shindo, N., Showe, D. N., Sosa, M. I., Suling, W. J., and White, E. L. (2009) High-throughput screening for inhibitors of Mycobacterium tuberculosis H37Rv, Tuberculosis (Edinb) 89, 334-353. [31] Maddry, J. A., Ananthan, S., Goldman, R. C., Hobrath, J. V., Kwong, C. D., Maddox, C., Rasmussen, L., Reynolds, R. C., Secrist, J. A., 3rd, Sosa, M. I., White, E. L., and Zhang, W. (2009) Antituberculosis activity of the molecular libraries screening center network library, Tuberculosis (Edinb) 89, 354-363. [32] Chaturvedi, V., Dwivedi, N., Tripathi, R. P., and Sinha, S. (2007) Evaluation of Mycobacterium smegmatis as a possible surrogate screen for selecting molecules active

27 ACS Paragon Plus Environment

ACS Chemical Biology 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

641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681

against multi-drug resistant Mycobacterium tuberculosis, J Gen Appl Microbiol 53, 333337. [33] de la Paz Santangelo, M., Gest, P. M., Guerin, M. E., Coincon, M., Pham, H., Ryan, G., Puckett, S. E., Spencer, J. S., Gonzalez-Juarrero, M., Daher, R., Lenaerts, A. J., Schnappinger, D., Therisod, M., Ehrt, S., Sygusch, J., and Jackson, M. (2011) Glycolytic and non-glycolytic functions of Mycobacterium tuberculosis fructose-1,6-bisphosphate aldolase, an essential enzyme produced by replicating and non-replicating bacilli, J Biol Chem 286, 40219-40231. [34] Mani, N., Gross, C. H., Parsons, J. D., Hanzelka, B., Muh, U., Mullin, S., Liao, Y., Grillot, A. L., Stamos, D., Charifson, P. S., and Grossman, T. H. (2006) In vitro characterization of the antibacterial spectrum of novel bacterial type II topoisomerase inhibitors of the aminobenzimidazole class, Antimicrob Agents Chemother 50, 1228-1237. [35] Lee, M. R., Sheng, W. H., Hung, C. C., Yu, C. J., Lee, L. N., and Hsueh, P. R. (2015) Mycobacterium abscessus Complex Infections in Humans, Emerg Infect Dis 21, 16381646. [36] Singla, D., Tewari, R., Kumar, A., Raghava, G. P., and Open Source Drug Discovery, C. (2013) Designing of inhibitors against drug tolerant Mycobacterium tuberculosis (H37Rv), Chem Cent J 7, 49. [37] Zhang, H., Zhou, T., Kurnasov, O., Cheek, S., Grishin, N. V., and Osterman, A. (2002) Crystal structures of E. coli nicotinate mononucleotide adenylyltransferase and its complex with deamido-NAD, Structure 10, 69-79. [38] D'Angelo, I., Raffaelli, N., Dabusti, V., Lorenzi, T., Magni, G., and Rizzi, M. (2000) Structure of nicotinamide mononucleotide adenylyltransferase: a key enzyme in NAD(+) biosynthesis, Structure 8, 993-1004. [39] O'Hara, J. K., Kerwin, L. J., Cobbold, S. A., Tai, J., Bedell, T. A., Reider, P. J., and Llinas, M. (2014) Targeting NAD+ metabolism in the human malaria parasite Plasmodium falciparum, PLoS One 9, e94061. [40] Silver, L. L. (2011) Challenges of antibacterial discovery, Clin Microbiol Rev 24, 71-109. [41] Thangavelu, K., Pan, C. Q., Karlberg, T., Balaji, G., Uttamchandani, M., Suresh, V., Schuler, H., Low, B. C., and Sivaraman, J. (2012) Structural basis for the allosteric inhibitory mechanism of human kidney-type glutaminase (KGA) and its regulation by Raf-Mek-Erk signaling in cancer cell metabolism, Proc Natl Acad Sci U S A 109, 7705-7710. [42] Keri, R. S., Rajappa, C. K., Patil, S. A., and Nagaraja, B. M. (2016) Benzimidazole-core as an antimycobacterial agent, Pharmacol Rep 68, 1254-1265. [43] Karplus, P. A., and Diederichs, K. (2012) Linking crystallographic model and data quality, Science 336, 1030-1033. [44] Kabsch, W. (2010) Xds, Acta Crystallogr D Biol Crystallogr 66, 125-132. [45] Chen, V. B., Arendall, W. B., 3rd, Headd, J. J., Keedy, D. A., Immormino, R. M., Kapral, G. J., Murray, L. W., Richardson, J. S., and Richardson, D. C. (2010) MolProbity: all-atom structure validation for macromolecular crystallography, Acta Crystallogr D Biol Crystallogr 66, 12-21.

28 ACS Paragon Plus Environment

Page 28 of 33

Page 29 of 33 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 Chemical Biology

Figure 1. Flowchart of the whole-cell and target screening approach for delivering novel antitubercular NaMN adenylyltransferase inhibitors. 121x160mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Chemical Biology 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. Chemical diversity of the 3 series of novel MtNadD inhibitors evaluated. In red, molecules that were identified in our primary screen of the TB Alliance bioactive library against MtNadD (IC50< 20 μM). In black, novel analogues for each chemotype tested in this study. In blue, analogs that were not tested in our study, but for which a relevant activity against M. tuberculosis or a Mtb target enzyme has been reported in PubChem (see Tables 1-2 and main text for details). 134x163mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 30 of 33

Page 31 of 33 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 Chemical Biology

Figure 3. Bactericidal, on-target effects of N2 inhibitors in replicating and non-replicating M. smegmatis. (A-C) Growth suppression curves at different N2 series inhibitors’ concentrations underline their killing activity on replicating Msm at above 5 μM. DMSO at 1 % represents the “no inhibitor” control. Linezolid at 3 μM was used as a positive control. (D) Addition of the inhibitor at ~ 5xMIC concentration at log-phase (after 12.5 hours of growth) stops the growth of Msm and (E) rapidly deplete NAD pool. Residual levels of NAD were measured by a colorimetric detection kit and normalized by total protein in samples taken for 6 hours, in 2 hours’ time interval. (F) Cell viability by CFU assay after 1-4 days of incubation of Msm with 5xMIC inhibitor (25 μM) under carbon starvation conditions. In this case, Linezolid was used at 15 μM. 134x116mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Chemical Biology 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. Structure of MtNadD in complex with inhibitor N2-8. (A) N2-8 inhibitor-driven dimer-ofdimers of MtNadD with two molecules of the ligand bridging the dimers formed by chains B and A’, and chains B’ and A, respectively. (B) A close-up view of the N2-8 binding site. The hydrophobic residues lining up the inhibitor binding site are shown in sticks representation. σA-weighted 2FO–FC electron density map contoured at 1σ is shown as blue mesh. (C) Comparison of the closed active-site conformation of MtNadD•N2-8 complex (light blue) with MtNadD apo structure (yellow) (PDB ID, 4X0E) and MaNadD•NaAD complex structure (purple) (PDB ID, 5DEO). H-bonding between D109 and the adenosine moiety of the MaNadD•NaAD complex is shown as dashed line. The steric clashes of L164 with adenine ring, I113 with nicotinate ring, and G106 with the AMP ribose are marked by a dotted circle. 177x195mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 32 of 33

Page 33 of 33 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 Chemical Biology

80x39mm (300 x 300 DPI)

ACS Paragon Plus Environment