You are on page 1of 4

Communication

pubs.acs.org/biochemistry

First-In-Class Inhibitor of Ribosomal RNA Synthesis with


Antimicrobial Activity against Staphylococcus aureus
Xiao Yang, Ming Jing Luo, Apple C. M. Yeung, Peter J. Lewis, Paul K. S. Chan,, Margaret Ip,*,
and Cong Ma*,,

Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong

School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia

Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong

Department of Applied Biology and Chemical Technology and State Key Laboratory of Chirosciences, The Hong Kong Polytechnic
University, Hung Hom, Hong Kong

The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
*
S Supporting Information

only one RNA polymerase in bacteria, which is associated with


ABSTRACT: We report the discovery of the rst a number of elongation factors for forming so-called rRNA
bacterial ribosomal RNA (rRNA) synthesis inhibitor that antitermination complexes, which ensure ecient transcription
has specic antimicrobial activity against methicillin- of the rRNA genes.6
resistant Staphylococcus aureus (MRSA). A pharmacophore NusB and NusE (also known as the ribosomal protein S10 of
model was constructed on the basis of the proteinprotein the 30S ribosomal subunit) are highly conserved essential small
interaction between essential bacterial rRNA transcription transcription factors involved in the formation of rRNA
factors NusB and NusE and employed for an in silico antitermination complexes (Figure S1).7 The proteinprotein
screen to identify potential leads. One compound, (E)-2- interaction between NusB and NusE represents the rst
{[(3-ethynylphenyl)imino]methyl}-4-nitrophenol (MC4), regulatory step in rRNA transcription antitermination complex
demonstrated antimicrobial activity against a panel of S. assembly.8 Once a NusBNusE heterodimer forms, it interacts
aureus strains, including MRSA, without signicant toxicity with a region of the rRNA leader sequence called boxA.9
to mammalian cells. MC4 resulted in a decrease in the Following binding of the NusBNusEboxA complex to RNA
rRNA level in bacteria, and the target specicity of MC4 polymerase, other factors (such as NusA, NusG, and others)
was conrmed at the molecular level. Results obtained will be recruited, among which only NusG has a eukaryotic
from this work validated the bacterial rRNA transcription homologue.10
machinery as a novel antimicrobial target. This approach Because NusB and NusE are essential for bacterial cell
may be extended to other factors in rRNA transcription, viability,11 we hypothesize that disruption of NusBNusE
and MC4 could be applied as a chemical probe to dissect heterodimer formation by small molecules will result in reduced
the relationship among MRSA infection, MRSA growth rates of rRNA synthesis and bacterial cell proliferation.
rate, and rRNA synthesis, in addition to its therapeutic Previously, by rational design and pharmacophore-based virtual
potential. screening, we identied small chemical molecule inhibitors with
antimicrobial activities, targeting the interaction between
bacterial RNA polymerase and the essential housekeeping
transcription initiation factor .12 Using a similar approach, we
I nfections caused by the Gram-positive pathogen methicillin-
resistant Staphylococcus aureus (MRSA) have become one of
the most serious public health concerns worldwide.1 The
have identied an inhibitor against bacterial rRNA synthesis
that has antimicrobial activities against S. aureus strains,
pharmaceutical arsenal available to control MRSA is limited to including MRSA.
vancomycin, daptomycin, and linezolid,2 for which resistance A bacterial rRNA transcription complex was modeled on the
has already evolved.3 Therefore, there is an urgent need to basis of the crystal structure of the RNA polymerase elongation
validate a new antibiotic target, to develop novel antimicrobials complex13 with a suite of Nus transcription factors NusA,
with potent and specic activities to combat MRSA-associated NusB, NusE, and NusG (Figure 1A). NusG binds to the central
infections. cleft of RNA polymerase via its N-terminal domain,14 and its C-
In bacterial cells, rRNA comprises up to 80% of total RNA terminal domain interacts with NusE,15 which anchors the
and transcription of rRNA has been shown to positively NusBNusEboxA subcomplex to the downstream face of
correlate with bacterial growth rate to meet the demand for RNA polymerase (Figure 1A). NusA binds to RNA polymerase
protein synthesis.4 Although rRNA synthesis is one of the most near the RNA exit channel (Figure 1A),16 consistent with its
fundamental requirements for living cells, there is a noticeable
discrepancy in this process. In eukaryotic cells, the ribosomal Received: April 18, 2017
genes are transcribed by dierent types of RNA polymerases, Revised: July 27, 2017
namely, RNA Pol I and Pol III.5 On the other hand, there is Published: August 7, 2017

2017 American Chemical Society 5049 DOI: 10.1021/acs.biochem.7b00349


Biochemistry 2017, 56, 50495052
Biochemistry Communication

Figure 1. (A) Model of the bacterial rRNA transcription complex. (B) NusBNusE interface.

binding to rRNA just downstream of the boxA sequence.17 The L22 and NusE V26. In addition to the interactions, a series of
interaction between RNA polymeraseNus factors and rRNA exclusion zones (gray) were added to minimize steric clashes
results in a constrained loop, facilitating rapid and proper within the shallow pocket that forms the binding site on NusB.
folding of the emerging transcript, which is consistent with The nal pharmacophore model was then created using Biovia
previous biochemical observations that the RNA polymerase DS4.5 to map on all the features required.24 As the
Nus factor complex would play the role of a chaperone in pharmacophore model was designed on the basis of the
rRNA synthesis.18 This assembly also has possible roles in properties of the important amino acid residues on the NusE
preventing the termination factor Rho from accessing the rRNA protein responsible for binding to NusB, theoretically, the ideal
transcript,19 ensuring complete transcription of the relatively small molecules capable of docking into this pharmacophore
large rRNA operons during rapid bacterial cell growth. Recently model should be able to bind to NusB and demonstrate
reported structural information about the phage protein N- inhibitory activity against the NusBNusE interaction accord-
dependent transcription antitermination complex also was ingly.
similar to that of our rRNA transcription complex model.20 On the basis of the pharmacophore model, an in silico screen
Examination of the published crystal structures of the was performed using a virtual compound library constructed by
Escherichia coli NusBNusE heterodimer [Protein Data Bank combining the mini-Maybridge library and the Enamine
(PDB) entry 3D3B] (Figure 1B)21 reveals that NusE contains antibacterial library.25 The top 50 hits from the initial virtual
only 18% -helix and binds with NusB mainly via interactions screen were re-mapped against the pharmacophore model and
with helix 2 (Figure 1B).22 The hydrogen bonding interactions the energy-minimized conformations of compounds visually
occur between NusB E81 and NusE H15, NusB Y18 and NusE inspected. The compounds that poorly t into the pharmaco-
D19, and NusB E75 and NusE R16 (Figure 1B, expanded view; phore were removed. As a result, we initially short-listed seven
E. coli amino acid residue numbering), which are highly compounds (Figure S2) for wet-laboratory testing.
conserved across prokaryotes (Figure S1, arrows). Additionally, We rst screened the antimicrobial activity of the seven
a nuclear magnetic resonance study of the Aquifex aeolicus compounds against community-acquired MRSA strain
NusBNusE interaction also conrmed similar interactions USA300. Of the analogues evaluated, MC4 (Figure 2B) was
exist in solution.23 found to demonstrate growth inhibition eects with a
The structural information about the NusBNusE hetero- minimum inhibitory concentration (MIC) of 64 g/mL
dimer co-crystal (PDB entry 3D3B)22 was used to develop a (Figure 3). With a molecular weight of 266.3, MC4 has been
pharmacophore model (Figure 2A). The pharmacophore reported to be of use only to form a metal complex dye in
model comprised two hydrogen donors (pink), one acceptor optical layers for optical data recording.26 We then tested the
(green) to mimic the major hydrogen bonds between NusB
and NusE as mentioned above, and one conserved hydrophobic
interaction (cyan, Figure 2A) between E. coli residues NusB

Figure 2. (A) Pharmacophore model with MC4 docked in. (B) Figure 3. Antimicrobial activity of MC4 against selected pathogenic
Chemical structure of (E)-2-{[(3-ethynylphenyl)imino]methyl}-4- bacteria. Abbreviations: MIC, minimum inhibitory concentration;
nitrophenol (MC4). MBC, minimum bactericidal concentration; ND, not determined.

5050 DOI: 10.1021/acs.biochem.7b00349


Biochemistry 2017, 56, 50495052
Biochemistry Communication

antimicrobial activities of MC4 against a panel of representative pharmacophore model and demonstrated that the reactivity of
strains of pathogens. MC4 demonstrated preferred antimicro- imine and p-nitrophenol did not contribute to the activity of
bial activity against S. aureus strains, including MRSA, over MC4.
other pathogens tested, with MICs as low as 8 g/mL against We have also biophysically quantied the interaction
control strain S. aureus 25923 and 16 g/mL against healthcare- between MC4 and NusB. A previous report demonstrated
acquired MRSA ST239 (Figure 3). Additionally, MC4 did not that NusB bound to NusE in a 1:1 ratio with a Kd of 1 M as
show signicant cytotoxicity against mammalian cell lines determined by isothermal titration calorimetry (ITC).21 In
compared to 5-uorouracil (Table S1). similar experiments, we found MC4 bound specically to NusB
We analyzed the level of macromolecules in S. aureus ATCC (Figure S3A) with a one-site binding mode with a Kd of 1.45
25923 cells during exponential growth due to MC4 treatment. 0.55 M. Binding of MC4 to NusE could not be detected by
MC4, rifampicin, and oxacillin were added at a one-quarter ITC (not shown), or between MC4 and NusB variants (Y18A,
MIC level, which did not interfere with the rate of growth of S. D76A, and D81A) with the three amino acid residues
aureus ATCC 25923 cells. As shown in Figure 4, none of the responsible for NusE binding altered to alanine (Figure S3B
D). These results together suggest the inhibition of NusB
NusE heterodimer formation is achieved via specic interaction
between MC4 and NusB as designed. Further experiments will
be performed to resolve the structure of NusB in complex with
MC4 for target validation, as well as structure-based lead
optimization.
The potential impact of untreatable antibiotic-resistant
infections on society is profound, and there is an urgent need
to identify new drug targets.28 Traditionally, the bacterial
ribosome itself (both 30S and 50S subunits) has been one of
the most commonly exploited targets for antibiotics inhibiting
protein synthesis.29 Recent drug discovery research had
validated the nding that inhibition of rescuing stalled
ribosomes at the end of mRNAs resulted in antibacterial
Figure 4. Eects of MC4, rifampicin (Rif), and oxacillin (Oxa) at one- activity.30 Given the ribosome is positively related to fast
quarter minimum inhibitory concentrations (MICs) on DNA, rRNA growth/proliferation and the large dierence between
(16S + 23S), and protein production in S. aureus 25923 cells. eukaryotic and prokaryotic rRNA transcription machinery, it
is tempting to hypothesize inhibition of rRNA synthesis would
treatment aected the DNA level, as previewed by the mode of be expected to have a major impact on cell growth and/or
action. In the control cells, the level of major rRNA (16S + viability. This hypothesis is strengthened by recent ndings
23S) was around 78% of total RNA (Figure 4).4 Rifampicin showing that many anticancer drugs inhibit rRNA synthesis or
resulted in reduction in the rRNA level consistent with previous maturation.31
observations (Figure 4).27 MC4 showed a signicant reduction In this work, we have used a pharmacophore-based in silico
in the rRNA level, which was lower than that of rifampicin- screen followed by biological conrmation to identify a
treated cells (Figure 4). Furthermore, MC4 treatment led to a potential new antibiotic lead. We have targeted an essential
signicant reduction in the protein level, while rifampicin did interaction between transcription factors NusB and NusE that
not show this eect, probably as a result of a decreased level of is required for the formation of highly processive complexes
ribosome production, aecting the protein synthesis ability. used for the synthesis of rRNA within bacterial cells. One of the
Oxacillin-treated cells displayed rRNA and protein production short-listed compounds (MC4) showed specic activity against
levels slightly higher than those of control cells. S. aureus strains, including MRSA, without signicant toxicity to
Finally, we intended to establish MC4s mode of action at the mammalian cell lines. It is, to the best of our knowledge, the
molecular level. An enzyme-linked immunosorbent assay-based rst compound designed to target bacterial rRNA synthesis that
inhibitory assay was performed to assess the in vitro inhibition has antimicrobial activities. The detailed eect of MC4 in rRNA
of NusBNusE heterodimer formation by MC4.24 Puried transcription/processing, ribosome biogenesis, and S. aureus
NusB was used to coat the 96-well plate, and GST-tagged NusE virulence is currently under investigation. Although MC4 has
was used as the probe. MC4 showed positive inhibition of the been shown to specically inhibit NusBNusE interaction at
NusBNusE interaction with an IC50 of 34.7 0.13 M. By the molecular level, any potential o-target eect on bacterial
further testing a series of MC4 analogues (Figure S3), we found cells remains to be elucidated. Because NusB and NusE are
that three functional groups on the molecule targeting highly conserved in bacteria, the reason that MC4 has preferred
interactions between NusB E81 and NusE H15, NusB Y18 antimicrobial activity against S. aureus over other pathogens
and NusE D19, and NusB E75 and NusE R16 were compulsory needs to be further investigated.
for inhibiting NusBNusE binding, as predicted by FitScore of We validated an essential proteinprotein interaction
Biovia DS4.5 (Table S3). With the change in the phenyl between transcription factors in the bacterial rRNA synthesis
acetylene group to phenols, the IC 50 values of the machinery as a novel antimicrobial target. Other important
corresponding MC4 analogues increased, while deletion of proteinprotein interactions involved in bacterial rRNA
this terminal triple bond or replacing by methyl or chloride transcription, e.g., between NusE and NusG, the NusERNA
caused a reduced IC50. When p-nitrophenol was modied to o- polymerase complex might also have the potential as novel
nitrophenol, the IC50 values increased, probably because of the antimicrobial targets.32 This work paves the way for the
involvement of the phenol group in the binding interaction structural optimization of MC4, and potentially other
with NusB Y81 (Table S3). These results conrmed our compounds from more comprehensive screens, for develop-
5051 DOI: 10.1021/acs.biochem.7b00349
Biochemistry 2017, 56, 50495052
Biochemistry Communication

ment as prospective new antimicrobial lead molecules targeting (13) Vassylyev, D. G., Vassylyeva, M. N., Perederina, A., Tahirov, T.
bacterial rRNA synthesis. H., and Artsimovitch, I. (2007) Nature 448, 157162.

(14) Liu, B., and Steitz, T. A. (2017) Nucleic Acids Res. 45, 968974.
ASSOCIATED CONTENT (15) Burmann, B. M., Schweimer, K., Luo, X., Wahl, M. C., Stitt, B.
L., Gottesman, M. E., and Rosch, P. (2010) Science 328, 501504.
*
S Supporting Information
(16) Yang, X., Molimau, S., Doherty, G. P., Johnston, E. B., Marles-
The Supporting Information is available free of charge on the Wright, J., Rothnagel, R., Hankamer, B., Lewis, R. J., and Lewis, P. J.
ACS Publications website at DOI: 10.1021/acs.bio- (2009) EMBO Rep. 10, 9971002.
chem.7b00349. (17) (a) Beuth, B., Pennell, S., Arnvig, K. B., Martin, S. R., and
Experimental details and additional data (PDF) Taylor, I. A. (2005) EMBO J. 24, 35763587. (b) Prasch, S., Jurk, M.,

Washburn, R. S., Gottesman, M. E., Wohrl, B. M., and Rosch, P.


(2009) Nucleic Acids Res. 37, 47364742.
AUTHOR INFORMATION (18) (a) Bubunenko, M., Court, D. L., Al Refaii, A., Saxena, S.,
Corresponding Authors Korepanov, A., Friedman, D. I., Gottesman, M. E., and Alix, J. H.
*E-mail: cong.ma@polyu.edu.hk. (2013) Mol. Microbiol. 87, 382393. (b) Wong, T., Sosnick, T. R., and
*E-mail: margaretip@cuhk.edu.hk. Pan, T. (2005) Biochemistry 44, 75357542.
(19) Anderson, A. C. (2003) Chem. Biol. 10, 787797.
ORCID (20) Said, N., Krupp, F., Anedchenko, E., Santos, K. F., Dybkov, O.,
Cong Ma: 0000-0001-9245-0356 Huang, Y. H., Lee, C. T., Loll, B., Behrmann, E., Burger, J., Mielke, T.,
Author Contributions Loerke, J., Urlaub, H., Spahn, C. M. T., Weber, G., and Wahl, M. C.
X.Y. and C.M. conceived the idea. X.Y., M.J.L., A.C.M.Y., and (2017) Nat. Microbiol. 2, 17062.
(21) Das, R., Loss, S., Li, J., Waugh, D. S., Tarasov, S., Wingfield, P.
C.M. performed experiments. X.Y., M.J.L., P.J.L., P.K.S.C., M.I.,
T., Byrd, R. A., and Altieri, A. S. (2008) J. Mol. Biol. 376, 705720.
and C.M. analyzed data. X.Y. and C.M. wrote the paper. (22) Luo, X., Hsiao, H. H., Bubunenko, M., Weber, G., Court, D. L.,
Funding Gottesman, M. E., Urlaub, H., and Wahl, M. C. (2008) Mol. Cell 32,
This work was funded by the Department of Applied Biology 791802.
and Chemical Technology Set-up Fund and the State Key (23) Stagno, J. R., Altieri, A. S., Bubunenko, M., Tarasov, S. G., Li, J.,
Laboratory of Chirosciences, HKPU (C.M.), a Shenzhen Court, D. L., Byrd, R. A., and Ji, X. (2011) Nucleic Acids Res. 39, 7803
Science and Technology Innovation Commission Research 7815.
Grant (JCYJ20170303155923684, to C.M.), the Department of (24) Yang, X., Ma, C., and Lewis, P. J. (2015) Methods 86, 4550.
(25) Lipinski, C. A., Lombardo, F., Dominy, B. W., and Feeney, P. J.
Microbiology Research Fund, CUHK (X.Y. and M.I.), and the (2001) Adv. Drug Delivery Rev. 46, 326.
National Health and Medical Research Council of Australia (26) (a) Luecke, L., Weiss, A., Graciet, J.-C., Steanut, P., Klein, C.,
(G1008014, to P.J.L.). and Winter, M. A. (2008) Eur. Pat. Appl. EP 1930378. (b) Luecke, L.,
Notes Weiss, A., Graciet, J.-C., Steanut, P., Klein, C., and Winter, M. A.
The authors declare no competing nancial interest. (2008) Eur. Pat. Appl. EP 1925643.

(27) (a) Cangelosi, G. A., and Brabant, W. H. (1997) J. Bacteriol. 179,


REFERENCES 44574463. (b) Halford, C., Gonzalez, R., Campuzano, S., Hu, B.,
Babbitt, J. T., Liu, J., Wang, J., Churchill, B. M., and Haake, D. A.
(1) (a) Jean, S. S., and Hsueh, P. R. (2011) Int. J. Antimicrob. Agents (2013) Antimicrob. Agents Chemother. 57, 936943.
37, 291295. (b) Frieden, T. R. (2013) N. Engl. J. Med. 368, 1857 (28) ONeil, J. (2014) Antimicrobial resistance: Tackling a crisis for the
1859. health and wealth of nations, Wellcome Trust and HM Government,
(2) Chambers, H. F., and Deleo, F. R. (2009) Nat. Rev. Microbiol. 7, London.
629641. (29) Chellat, M. F., Raguz, L., and Riedl, R. (2016) Angew. Chem., Int.
(3) (a) Gardete, S., and Tomasz, A. (2014) J. Clin. Invest. 124, 2836 Ed. 55, 66006626.
2840. (b) Nannini, E., Murray, B. E., and Arias, C. A. (2010) Curr. (30) Goralski, T. D., Dewan, K. K., Alumasa, J. N., Avanzato, V.,
Opin. Pharmacol. 10, 516521. Place, D. E., Markley, R. L., Katkere, B., Rabadi, S. M., Bakshi, C. S.,
(4) Jin, D. J., Cagliero, C., and Zhou, Y. N. (2012) FEMS Microbiol. Keiler, K. C., and Kirimanjeswara, G. S. (2016) Antimicrob. Agents
Rev. 36, 269287. Chemother. 60, 32763282.
(5) Viktorovskaya, O. V., and Schneider, D. A. (2015) Gene 556, 19 (31) Brighenti, E., Trere, D., and Derenzini, M. (2015) Oncotarget 6,
26. 3861738627.
(6) Santangelo, T. J., and Artsimovitch, I. (2011) Nat. Rev. Microbiol. (32) Ma, C., Yang, X., and Lewis, P. J. (2016) Microbiol. Mol. Biol.
9, 319329. Rev. 80, 139160.
(7) Richardson, J. P., and Greenblatt, J. (1996) in Escherichia coli and
Salmonella: Cellular and molecular biology, 2nd ed., pp 822848,
American Society for Microbiology, Washington, DC.
(8) Greive, S. J., Lins, A. F., and von Hippel, P. H. (2005) J. Biol.
Chem. 280, 3639736408.
(9) (a) Nodwell, J. R., and Greenblatt, J. (1993) Cell 72, 261268.
(b) Burmann, B. M., Luo, X., Rosch, P., Wahl, M. C., and Gottesman,
M. E. (2010) Nucleic Acids Res. 38, 314326.
(10) Werner, F., and Grohmann, D. (2011) Nat. Rev. Microbiol. 9,
8598.
(11) (a) Kato, J., and Hashimoto, M. (2007) Mol. Syst. Biol. 3, 132.
(b) Bubunenko, M., Baker, T., and Court, D. L. (2007) J. Bacteriol.
189, 28442853.
(12) (a) Ma, C., Yang, X., Kandemir, H., Mielczarek, M., Johnston, E.
B., Griffith, R., Kumar, N., and Lewis, P. J. (2013) ACS Chem. Biol. 8,
19721980. (b) Ma, C., Yang, X., and Lewis, P. J. (2016) ACS Infect.
Dis. 2, 3946.

5052 DOI: 10.1021/acs.biochem.7b00349


Biochemistry 2017, 56, 50495052

You might also like