Download Novel Antibacterials: A Genomics Approach to Drug Discovery

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Oncogenomics wikipedia , lookup

Human genome wikipedia , lookup

No-SCAR (Scarless Cas9 Assisted Recombineering) Genome Editing wikipedia , lookup

Gene expression programming wikipedia , lookup

Biology and consumer behaviour wikipedia , lookup

Genomic imprinting wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

Ridge (biology) wikipedia , lookup

Essential gene wikipedia , lookup

Polycomb Group Proteins and Cancer wikipedia , lookup

Nutriepigenomics wikipedia , lookup

Genetic engineering wikipedia , lookup

Genomics wikipedia , lookup

Pharmacogenomics wikipedia , lookup

Genome (book) wikipedia , lookup

NEDD9 wikipedia , lookup

Metagenomics wikipedia , lookup

Gene wikipedia , lookup

Microevolution wikipedia , lookup

Epigenetics of human development wikipedia , lookup

Designer baby wikipedia , lookup

Pathogenomics wikipedia , lookup

Site-specific recombinase technology wikipedia , lookup

Therapeutic gene modulation wikipedia , lookup

RNA-Seq wikipedia , lookup

Public health genomics wikipedia , lookup

Gene expression profiling wikipedia , lookup

Genome evolution wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

History of genetic engineering wikipedia , lookup

Minimal genome wikipedia , lookup

Transcript
Current Drug Targets - Infectious Disorders, 2002, 2, 291-308
291
Novel Antibacterials: A Genomics Approach to Drug Discovery
Pan F. Chan1, Ricardo Macarron2, David J. Payne1, Magdalena Zalacain1 and
David J. Holmes1*
1Department
of Microbiology. Microbial, Musculoskeletal and Proliferative Diseases CEDD,
GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426-0989, USA,
2Department of Molecular Screening, GlaxoSmithKline, King of Prussia, PA, USA
Abstract: The appearance of antibiotic resistant pathogens, including vancomycin
resistant Staphylococcus aureus, in the clinic has necessitated the development of new
antibiotics. The golden age of antibiotic discovery, in which potent selective
compounds were readily extracted from natural product extracts is over and novel
approaches need to be implemented to cover the therapeutic shortfall. The generation of
huge quantities of bacterial sequence data has allowed the identification of all the
possible targets for therapeutic intervention and allowed the development of screens to identify inhibitors.
Here, we described a number of target classes in which genomics has contributed to its identification. As a
result of analyzing sequence data, all of the tRNA synthetases and all of the two-component signal
transduction systems were readily isolated; which would not have been easily identified if whole genome
sequences were not available. Fatty acid biosynthesis is a known antibacterial target, but genomics showed
which genes in that pathway had the appropriate spectrum to be considered as therapeutic targets. Genes of
unknown function may seem untractable targets, but if those that are broad spectrum and essential are
identified, it becomes valuable to invest time and effort to determine their cellular role. In addition, we discuss
the role of genomics in developing technologies that assist in the discovery of new antibiotics including
microarray gridding technology. Genomics can also increase the chemical diversity against which the novel
targets can be screened.
Key words: genomics, bioinformatics, target validation, tRNA synthetases, two-component signal transduction, genes of
unknown function, enzyme based screening, whole cell screening, microarray gridding, inducible promoters, antisense.
1. INTRODUCTION
In recent years there has been an explosion in the amount
of available genome data in all of the kingdoms. These
include sequences from the first complete genome, that of
the bacterium Haemophilus influenzae, to the recent
publication of the human genome sequence [1,2]. Moreover,
there has been an explosion in the number of genome
sequencing projects covering the three phylogenetic domains
including those for the yeast chromosomes, and plant as
well as bacterial and archael genomes. In fact over 100
complete bacterial genome sequences are readily available
(Table 1) with hundreds more currently in progress. The data
generated to date is only the beginning since bioinformatic,
proteomic, genetic and, especially, biochemical approaches
now need to be brought to bear. In this "post genomics era"
efforts are now focused towards determining the function of
many of these genes. Until recently, the bacterium was a
black box of poorly understood physiology. With the
availability of genome sequence data, researchers have access
to all of the genes that are involved in the biochemical
reactions of the bacterial cell, and which contribute to its
physiological and structural components. Unraveling the
*Address correspondence to this author at the Department of
Microbiology. Microbial, Musculoskeletal and Proliferative Diseases
CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA
19426-0989, USA; TEL: 610 917 6464, FAX: 610 917 7901, email:
[email protected]
1568-0053/02 $35.00+.00
specific function of each gene is not facile (see article by C.
Volker & J. R. Brown) and in most cases at least one-third
of the genes identified cannot be assigned even a putative
function.
The benefits of genome sequence data to pharmaceutical
companies were obvious as it could provide targets for
therapeutic intervention. The traditional method for
discovering antibiotics involves screening for inhibition of
bacterial cell growth by compound banks or, more
commonly, natural product extracts. Subsequently, the
antibacterial mode of action of the compounds had to be
determined. This difficult task was simplified by the fact
that most inhibitors discovered by this method affect
relatively few cellular processes including transcription,
translation, DNA replication and cell wall biosynthesis.
However, while this approach was highly successful during
the 1950's-1970's, very few novel antibiotics have been
discovered in the last 25 years, Fig. (1).
While antibiotic drug discovery has been on the decline,
resistance to the therapeutic agents employed has steadily
increased. The emergence of common pathogens that are
resistant to multiple antibiotics coupled with the failure of
traditional methods to yield novel anti-infective agents has
required a creative new approach to drug discovery.
Genome sequence data has led to the identification of
new classes of broad spectrum targets, and through
© 2002 Bentham Science Publishers Ltd.
292
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
combinatorial genomic techniques, novel sources of
compounds to screen. In addition, novel technologies
derived from genome sequence data have allowed improved
Holmes et al.
methods for determining the mode of action of compounds
identified by their ability to inhibit the growth of whole
cells.
Table 1. Publically Available Genome Sequences
Organism
Domain
Genes
Total Bases
Source
Actinobacillus actinomycetemcomitans HK1651
Bacteria
1,988
2,095,439
University of Oklahoma
Aeropyrum pernix K1
Archaea
1,840
1,669,695
NCBI
Agrobacterium tumefaciens C58
Bacteria
5,299
5,673,563
NCBI
Aquifex aeolicus VF5
Bacteria
1,553
1,590,791
NCBI
Archaeoglobus fulgidus DSM4304
Archaea
2,420
2,178,400
NCBI
Bacillus anthracis Ames
Bacteria
5,287
5,227,297
TIGR
Bacillus halodurans C-125
Bacteria
4,066
4,202,353
NCBI
Bacillus stearothermophilus 10
Bacteria
3,342
3,269,999
University of Oklahoma
Bacillus subtilis 168
Bacteria
4,112
4,214,814
NCBI
Bordetella pertussis Tohama I
Bacteria
3,892
4,086,186
Sanger Center
Borrelia burgdorferi B31
Bacteria
1,637
1,519,856
NCBI
Brucella melitensis 16M
Bacteria
3,198
3,294,931
NCBI
Buchnera sp. APS
Bacteria
574
655,725
NCBI
Campylobacter jejuni NCTC 11168
Bacteria
1,633
1,641,481
Sanger Center
Caulobacter crescentus
Bacteria
3,737
4,016,947
TIGR
Chlamydia muridarum
Bacteria
916
1,076,912
NCBI
Chlamydia trachomatis D/UW-3/Cx
Bacteria
921
1,042,519
NCBI
Chlamydophila pneumoniae AR39
Bacteria
1,109
1,229,853
NCBI
Chlamydophila pneumoniae CWL-029
Bacteria
1,052
1,230,230
Incyte
Chlamydophila pneumoniae J138
Bacteria
1,069
1,226,565
NCBI
Chlamydophila pneumoniae L2
Bacteria
1,042
1,234,390
TIGR
Chlorobium tepidum TLS
Bacteria
3,178
2,196,918
TIGR
Clostridium acetobutylicum ATCC824
Bacteria
3,672
3,940,880
TIGR
Clostridium perfringens 13
Bacteria
2,723
3,085,740
NCBI
Corynebacterium diphtheriae NCTC13129
Bacteria
2,128
2,488,600
Sanger Center
Corynebacterium glutamicum
Bacteria
2,989
3,309,400
EBI
Deinococcus radiodurans R1
Bacteria
3,103
3,284,156
TIGR
Desulfovibrio vulgaris
Bacteria
3,571,425
TIGR
Enterococcus faecalis V583
Bacteria
3,148
3,359,973
TIGR
Escherichia coli K-12
Bacteria
4,279
4,639,221
NCBI
Escherichia coli O157:H7
Bacteria
5,361
5,498,450
NCBI
Fusobacterium nucleatum ATCC 25586
Bacteria
2,067
2,174,500
NCBI
Haemophilus influenzae KW20
Bacteria
1,714
1,830,138
NCBI
Halobacterium sp. NRC-1
Archaea
2,605
2,571,010
NCBI
Helicobacter pylori 26695
Bacteria
1,576
1,667,867
NCBI
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 293
(Table 1). contd.....
Organism
Domain
Genes
Total Bases
Source
Helicobacter pylori J99
Bacteria
1,491
1,643,831
NCBI
Lactococcus lactis subsp.
Bacteria
2,266
2,365,589
NCBI
Listeria innocua Clip11262
Bacteria
2,968
3,011,208
NCBI
Listeria monocytogenes EGD
Bacteria
2,846
2,944,528
NCBI
Mesorhizobium loti
Bacteria
7,281
7,596,300
NCBI
Methanobacterium thermoautotrophicum delta H
Archaea
1,869
1,751,379
NCBI
Methanococcus jannaschii DSM 2661
Archaea
1,770
1,739,927
NCBI
Methanopyrus kandleri AV19
Archaea
1,687
1,694,969
NCBI
Methanosarcina acetivorans
Archaea
4,540
5,751,492
NCBI
Mycobacterium leprae
Bacteria
2,157
3,268,203
Sanger Center
Mycobacterium tuberculosis CDC1551
Bacteria
4,187
4,403,836
NCBI
Mycoplasma genitalium G-37
Bacteria
484
580,074
NCBI
Mycoplasma pneumoniae M129
Bacteria
688
816,394
NCBI
Mycoplasma pulmonis UAB CTIP
Bacteria
782
963,879
NCBI
Neisseria gonorrhoeae FA 1090
Bacteria
2,129
2,146,879
University of Oklahoma
Neisseria meningitidis Z2491
Bacteria
2,065
2,184,406
Sanger Center
Pasteurella multocida PM70
Bacteria
2,014
2,257,487
NCBI
Porphyromonas gingivalis W83
Bacteria
1,777
2,343,478
TIGR
Prochlorococcus marinus MED4
Bacteria
1,716
1,674,813
DOE Joint Genome
Pseudomonas aeruginosa PAO1
Bacteria
5,565
6,264,403
University of Washington
Pyrobaculum aerophilum
Archaea
2,275
2,222,890
UCLA Dept. Micr
Pyrococcus abyssi
Archaea
1,765
1,765,118
NCBI
Pyrococcus furiosus
Archaea
2,208
1,908,253
Utah
Pyrococcus horikoshii OT3
Archaea
2,058
1,738,505
NCBI
Ralstonia solanacearum
Bacteria
5,116
5,810,922
NCBI
Rhizobium sp. NGR234
Bacteria
417
536,165
NCBI
Rickettsia conorii
Bacteria
1,374
1,268,755
NCBI
Rickettsia prowazekii Madrid E
Bacteria
834
1,111,523
NCBI
Saccharomyces cerevisiae S288C
Eukaryota
6,261
12,057,849
NCBI
Salmonella typhi CT18
Bacteria
4,633
5,133,712
Sanger Center
Salmonella typhimurium LT2(strain AZ1516)
Bacteria
4,553
4,951,371
Washington Univ
Shewanella putrefaciens
Bacteria
4,221
5,131,063
TIGR
Sinorhizobium meliloti 1021
Bacteria
6,205
6,691,694
NCBI
Staphylococcus aureus EMRSA-16
Bacteria
2,679
2,902,619
Sanger Center
Staphylococcus aureus MW2, Mu50, N315
Bacteria
2,632
2,820,462
NCBI
Staphylococcus aureus NCTC 8325
Bacteria
2,631
2,836,373
University of Oklahoma
Staphylococcus epidermidis RP62A
Bacteria
2,444
2,646,310
TIGR
Streptococcus mutans UA159
Bacteria
1,871
2,030,921
University of Oklahoma
Streptococcus pneumoniae R6 hex
Bacteria
2,043
2,038,615
NCBI
Streptococcus pneumoniae type4
Bacteria
2,094
2,160,837
TIGR
294
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Holmes et al.
(Table 1). contd.....
Organism
Domain
Genes
Total Bases
Source
Streptococcus pyogenes M1 GAS
Bacteria
1,697
1,852,441
University of Oklahoma
Streptococcus pyogenes MGAS8232
Bacteria
1,845
1,895,017
NCBI
Sulfolobus solfataricus P2
Archaea
2,977
2,992,245
NCBI
Sulfolobus tokodaii
Archaea
2,826
2,694,765
NCBI
Synechocystis sp. PCC6803
Bacteria
3,169
3,573,470
NCBI
Thermoanaerobacter tengcongensis MB4T
Bacteria
2,588
2,689,445
NCBI
Thermoplasma volcanium
Archaea
1,499
1,584,804
NCBI
Thermotoga maritima MSB8
Bacteria
1,846
1,860,725
TIGR
Treponema pallidum Nichols
Bacteria
1,031
1,138,011
NCBI
Vibrio cholerae N16961
Bacteria
3,835
4,033,464
TIGR
Xanthomonas axonopodis citri str. 306
Bacteria
4,312
5,175,554
NCBI
Xanthomonas campestris ATCC 33913
Bacteria
4,181
5,076,188
NCBI
Xylella fastidiosa
Bacteria
2,831
2,731,750
NCBI
Yersinia pestis CO-92 Biovar Orientalis
Bacteria
4,083
4,829,855
Sanger Center
NCBI = National Center for Biotechnology Information, TIGR = The Institute for Genomic Research.
In this article we will discuss the criteria that define an
antibacterial target; their selection by bioinformatic analysis,
validation by genetic methods and improvements in
screening methods using specific examples. In addition,
alternative sources of compound diversity will be reviewed
along with methods for determining the cellular target of
compounds derived from a bacterial whole cell screen.
2. CRITERIA FOR A VALID ANTIBACTERIAL
TARGET
2.1 Novelty
Current antimicrobial agents target a relatively small
number of cellular processes. Fluoroquinolones and
Fig. (1). Antibiotics under threat. While few new chemical classes of antibiotics have been discovered in the past 20 years, the number
of resistant strains in the clinic continues to increase.
A Genomics Approach to Drug Discovery
novobiocin are examples of inhibitors of DNA replication;
protein synthesis is prevented by aminoglycosides,
macrolides, and tetracyclines while β -lactams and
carbapenems affect cell wall biosynthesis. These compounds
have been synthetically modified to overcome the resistance
issues that have developed over the period of their clinical
administration. In order to avoid being compromised by any
of the current resistance mechanisms it is desirable to
develop compounds that act against hitherto unexploited
targets including new biosynthetic enzymes, transcription
factors, or structural proteins etc. [3].
2.2 Spectrum/Selectivity
Since rapid, precise diagnosis of bacterial infection is
still not extensively accessible, the present unmet clinical
need is for a broad spectrum antibiotic to complement and/or
replace current chemotherapies such as methicillin,
amoxycillin and vancomycin which are compromised by
resistance mechanisms found in clinical isolates.
Comparison of bacterial genome sequences allows the
identification of targets that are present in all clinically
relevant pathogens and can be expected to be selective
against humans due to their absence or significant difference
in higher eukaryotes [4-6]. However, one can also imagine
directed therapies that would target genes restricted to one or
a few pathogens, thus affecting single or specific groups of
bacteria. Consequently, genes specific to Gram positive or
Gram negative bacteria could be targeted or in some cases,
such as Helicobacter pylori it may be desirable to screen a
species specific protein [7] (see article by Noonan B. and
Alm R.A.). The result would be that specific disease states
could be treated without necessarily affecting the entire
bacterial flora. Bioinformatic analysis can show the
anticipated spectrum of a compound aimed at any given
target if the genome sequence is available.
2.3 Essential for Cell Viability
Obviously, an antibacterial target must be essential for
cell viability either under laboratory conditions or during
infection. In order to focus the search for putative
antibacterial targets the essential genes within a given
bacterium should be identified. This could prove a daunting
task given the number of genes (about 2000) in each
pathogen that causes human disease. However, once
bioinformatic analysis described above, identifies the broad
spectrum genes in all organisms relevant to a particular
disease, only a few hundred genes remain to be examined.
Traditionally, essential genes were identified randomly
using a variety of genetic techniques. For example,
temperature sensitive mutants can be generated and the
determinant identified following complementation with the
corresponding wild-type gene. Transposon mutagenesis
involves the random insertion of a mobile element into the
chromosome. This method can be used to identify nonessential genes or genes essential under certain conditions.
Lately, transposon mutagenesis has been revolutionized by
the advent of genomics. Now, specific lengths of
chromosome from naturally transformable bacteria can
undergo saturation transposon mutagenesis in vitro and this
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 295
DNA can then be introduced back into the host organism.
The bank of mutants generated can be analysed to determine
which genes in the chromosome fragment contain insertions
[8,9]. Those genes not containing insertions are probably
essential for survival. However, it is not always possible to
demonstrate that every gene in the DNA fragment suffered at
least one insertion event.
Systematic gene-by-gene analysis of essentiality is also
possible. Methods are available for knocking out genes in a
directed fashion. The simplest form is plasmid insertion
mutagenesis [3]. Here a small internal fragment of the gene
of interest is cloned in a plasmid tagged with an antibiotic
resistance determinant that is expressed in the target
organism. This plasmid, which cannot replicate in that
bacterial species, is introduced into the organism and drugresistant colonies identified. The plasmid DNA is only
maintained following homologous recombination into the
target gene, and will therefore disrupt it. The method is
efficient, but has caveats: in many cases significant portions
of the "disrupted" gene remain intact resulting in partial gene
activity being expressed. If partial activity is sufficient for
cell survival, single crossover insertional inactivations may
miss essential genes giving rise to false negatives. Since the
insertion event introduces the entire vector into the
chromosome, this will almost certainly effect the expression
of genes downstream of the target (otherwise known as
polarity effects). As a result, insertion in a non-essential
gene may be lethal due to the effect on some distal gene
vital for cell growth and would therefore be incorrectly
assigned as essential.
In order to minimise the effects of partial activity and
polarity, allelic replacement can be employed. In this case, a
gene is replaced by an antibiotic resistance marker following
a double recombination event using homologous sequences
that flank the target gene [10-13]. If the resistance
determinant is carefully selected to ensure expression of
downstream genes, polarity effects are avoided. Indeed there
are examples where non-essential genes, upstream of known
essential genes, have been successfully deleted using this
method (Fig. (2)), Lunsford RD personal communication).
The procedure only identifies genes that are likely to be
essential, but very effectively defines those genes which are
not, allowing attention to focus on potential antibacterial
targets.
In this manner, we have tested >400 Streptococcus
pneumoniae and Staphylococcus aureus broad spectrum
genes that were possible targets for antibiotic development.
In over 70% of experiments it was possible to delete the
targeted gene, thereby demonstrating that it was not essential
for cell viability in vitro. The remaining genes, for which we
were unable to isolate allelic-replacement mutants despite
repeated attempts, are all potential targets for antibiotic
action (Fig. (3a)), personal communication, D. Lunsford,
GlaxoSmithKline). Over 100 mutants were tested for
virulence attenuation in a relevant animal infection model
and 72% were found to be attenuated by at least two logs,
Fig. (3 b ). Whether this was due to disruption of the
virulence process or simply effects on growth rate remain
unclear. The remaining 28% of mutants were not
significantly (< 2 logs) attenuated.
296
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Holmes et al.
Fig. (2). Example where non-essential genes, upstream of a known essential gene, have been successfully deleted demonstrating that
the allelic replacement technology used does not have polarity effects. The genes exoA and exoB encode subunits of an exonuclease
and have been shown to be non-essential for cell viability. On the other hand, ispA encodes geranyltranstransferase, an essential gene
involved isoprenyl pyrophosphate synthesis.
Fig. (3). Results of essentiality testing. (a) Proportion of genes that could be disrupted by allelic replacement in vitro (non
attenuated, dotted box; attenuated, oblique line box; in RTI model) and those that appeared essential; solid box. (b) Effects on
pathogenicity of selected mutants in RTI model. Mutants were considered in vivo essential if attenuated by >5 logs in an RTI model,
while those affected by <2 logs were considered non-attenuated.
Perhaps the best approach to confirm that a gene is
indeed essential for cell viability is by generating a directed
conditional mutant by placing the target gene under the
control of an inducible promoter. In contrast to knock-out
methods that give an all or nothing response, promoter
control systems allow the level of expression to be regulated
and will result in reduced cell growth or death when
expression is repressed. This method and the contribution of
genomics to defining appropriate promoter systems will be
discussed in more detail later.
2.4 Amenable to High-Throughput Screening
The purpose of these efforts is to identify suitable targets
which can be screened in order to discover new chemical
entities that will inhibit bacterial growth. Therefore the
novel target protein must have an activity that can be
assayed in high throughput (although knowledge of the exact
cellular function is not necessarily required) and reagents
must be readily available. This will be discussed later in
section 4.
Below, four kinds of targets are described which
exemplify the ways in which genomics has effected antiinfective drug discovery.
3. GENOMIC DERIVED TARGETS
3.1 Aminoacyl-tRNA Synthetases
Translation of messenger RNA into protein involves
precise recognition of the codons by the appropriately
charged tRNA. Aminoacyl-tRNA synthetases are responsible
for accurately combining the correct amino acid with its
cognate tRNA [14].
While in Gram negative bacteria there is a specific
enzyme for every tRNA molecule, Gram positive bacteria
possess only 19 aminoacyl-tRNA synthetases since
misacylated Glu-tRNAGln is the substrate for a transamidase
which converts the glutamyl adduct tRNAGln [15]. Each of
the tRNA synthetases are essential for cell viability.
Until recently, little was known about these enzymes in
Gram positive bacteria. Only two, isoleucyl-tRNA
synthetase [16] and lysyl-tRNA synthetase [17] had been
identified in S. aureus and none had been isolated from S.
pneumoniae. Indeed one of them, isoleucyl-tRNA synthetase
is the target of mupirocin, a valuable topical antibiotic with
potent Gram positive activity, including against methicillin
resistant S. aureus.
Isolating each aminoacyl-tRNA synthetase by standard
biochemical and genetic methods using appropriate DNA
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 297
probes is a plausible, albeit time-consuming and labor
intensive, task since the number of enzymes is known.
Clearly, this process is hugely simplified by the availability
of entire genome sequences. Identification of all 19 of the
aminoacyl-tRNA synthetase genes from both S. aureus and
S. pneumoniae in silico by comparison with those from E.
coli is facile. Moreover, this analysis can determine whether
a specific enzyme may be more appropriate for broad
spectrum or narrow spectrum utility as described above. For
example, as there are distinct differences between the Gram
positive and Gram negative methionyl-tRNA synthetases
(MRS), this enzyme may be more appropriate if a selective
Gram positive or Gram negative agent was required [18] and
glutaminyl-tRNA synthetase would obviously be a Gram
negative only target. Conversely, as the glutamyl-tRNA
synthetases from Gram positive and Gram negatives are
more similar this would be a more appropriate target if a
broad spectrum agent was desired [18].
3.2 Two-Component Signal Transduction Systems
Two-component signal transduction systems (TCSTSs),
more accurately defined as histidine-aspartate phosphorelay
Table 2.
systems, are perhaps the most widespread means of signal
transduction in bacteria. They regulate many cellular
responses, including osmoregulation, chemotaxis,
sporulation, antibiotic production and pathogenicity, in a
number of different bacteria [19]. TCSTSs are typically
composed of two signaling proteins: a sensor kinase and its
cognate response regulator. Specific environmental stimuli
activate the sensor kinase protein, resulting in the
autophosphorylation of a conserved histidine residue. This
high-energy phosphate group is then transferred to a
conserved aspartate residue in the cognate response regulator
resulting in structural changes in the protein that mediates
regulation of gene expression or protein function [20].
Histidine kinases are generally composed of a
transmembrane-spanning, sensor kinase at the aminoterminal and a carboxyl-terminus containing the transmitter
and kinase domains [21]. Since these systems are thought to
be the bacteria's way of recognizing and responding to its
environment, two-component signal transduction has been
considered as a potential antimicrobial target [22,23].
Pathogenic bacteria are confronted with dramatic changes in
environmental conditions (oxygen/pH/nutrient stress) when
they infect a host, as well as being challenged in most cases
by the host defence systems. Disruption of the TCST
Analysis of S. pneumoniae Two-Component Signal Transduction Systems. Allelic Replacement Mutants were
Generated (Y) in the Sensor Histidine Kinase (HK), the Response Regulator (RR), or the Gene Pair (HK & RR). 12 of
these (Marked with an Asterisk) were Tested in a Murine RTI Model. ND: Not done. WT: No Attenuation
Accession #
Histidine Kinase
Response Reg.
HK
RR
HK & RR
Attenuation in
RTI model
Homolog
AAK99148
AAK99147
Y
Y
Y*
WT
YvqCE
AAL00799
AAL00800
Y
Y
Y*
103-104
AAL00618
AAL00617
ND
Y*
ND
101
AAL00277
AAL00278
Y
Y*
Y
103-104
AAL00696
AAL00695
ND
Y*
ND
104
AAK98881
AAK98880
Y*
ND
ND
101
AAK99268
AAK99267
Y*
ND
ND
104
AAK99382
AAK99383
Y
Y
Y*
101-102
AAK99333
AAK99332
ND
ND
Y*
101
VncRS
AAK99909
AAK99910
Y*
Essential
ND
WT
YycFG
AAK98957
AAK98958
Y
Y
Y
-
AAK99140
-
Y*
-
104-105
AAL00844
AAL00843
ND
ND
ND
AAK99512
AAK99511
ND
ND
Y*
YvfTU
PnpSR
PhoPR
ComDE
103
CiaRH
298
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
signalling systems should impair both the viability of the
cell and its ability to establish and maintain infection.
However, His-Asp phosphorelays have not been linked to
the regulation of viability or pathogenicity. Prior to the
advent of genomics, four probable TCSTSs had been
identified in S. pneumoniae. The ciaRH, pnpSR and comDE
gene pairs appear to play a role in the regulation of
competence [24-28], and a fourth TCSTS encoded by the
vncSR gene pair has been associated with the development of
vancomycin tolerance [29]. In the absence of genome
sequence data it would be impossible to know whether all of
the TCSTSs had been identified from a particular bacterial
species. Unlike aminoacyl-tRNA synthetases, it is difficult
to estimate the number of TCSTS required by any particular
microorganism. H. influenzae has a total of only 5 TCSTSs,
Bacillus subtilis may have as many as 35 while
Pseudomonas aeruginosa has the highest number of all
bacterial genomes sequenced to date, with at least 63
TCSTSs [30]. Examination of the entire genome sequence of
S. pneumoniae identified 14 TCSTSs. All have been
investigated for their role for in vitro cell viability as well as
their role in the establishment and maintenance of infection
(Table 2).
Systematic mutagenesis studies and complementation
experiments demonstrated that one response regulator is
essential for cell growth in S. pneumoniae. Surprisingly,
inactivation of its cognate histidine kinase had little or no
effect on growth either in vivo or in vitro [31]. This system
is homologous to the YycFG TCSTS pair identified in S.
aureus and B. subtilis [32,33]. Unlike the situation in S.
pneumoniae, both the histidine kinase, YycG, and the
response regulator, YycF, were found to be vital for growth
in these bacteria. Characterization of the S. pneumoniae
mutant deleted for YycG showed no apparent phenotypic
alteration, indicating a functional YycF. This implies that
the activity of the YycF response regulator protein is
phosphorylation independent or is recognized by other
apparently "non-cognate" sensor kinase proteins. Analysis of
Streptococcus pyogenes, Enterococcus faecalis and
Lactococcus lactis genome sequence data shows that they
also contain YycFG homologues, suggesting that this
TCSTS is a broad spectrum target. The staphylococcal
yycFG locus may have a role in maintaining membrane
integrity and, it remains possible that YycFG co-ordinates
membrane growth with the cellular redox potential [33].
Of the 14 TCSTSs, identified in S. pneumoniae, seven
gene pairs appear to be important for the establishment
and/or maintenance of respiratory tract infections, while
having little effect on in vitro bacterial growth under normal
laboratory conditions [31].
Using a genomic-based approach, it has been possible to
identify and systematically disrupt the entire TCSTS gene
complement of S. pneumoniae and thereby define those
important for both pathogenicity and viability in a
respiratory tract infection. These studies provide support for
the contention that two-component signal transduction
systems are appropriate targets for the development of
antibacterial drugs.
Holmes et al.
3.3 Fatty Acid Biosynthesis
Cerulenin, thiolactomycin, diazaborines and triclosan are
antibiotics that selectively inhibit one or more of the
enzymes involved in fatty acid biosynthesis (FAB) [34]
validating this cyclic pathway as a therapeutic target. Access
to bacterial genomes has enabled all the components of the
FAB pathway to be identified in a variety of clinically
important pathogens (Table 3). This particular example
demonstrates the importance of genomics for predicting the
potential spectrum of each of the target enzymes.
Table 3.
Occurrence of Fab Genes in Key Pathogens
Target
S. aureus
S. pneumoniae
E. faecalis
H. influenzae
FabA
X
X
X
FabB
X
X
X
FabD
FabF
X
FabG
FabH
FabI
X
FabK
X
X
FabZ
= homologue present in genome; X = homologue absent from genome
In the pre-genomic era FabI was thought to be the unique
enoyl-acyl carrier protein (ACP) reductase in all bacteria as
data obtained from E. coli was assumed to pertain to all
prokaryotes. However, following the completion of several
additional genomes, it was obvious that fabI was absent
from a number of organisms, including S. pneumoniae and
E. faecalis. Since fatty acid biosynthesis is a cyclic process,
enoyl-ACP reductase activity had to exist in these bacteria.
Once again, genome sequence data provided the answer. In
S. pneumoniae the fatty acid biosynthetic genes are clustered
in one region of the chromosome, Fig. (4). Sequence
analysis identified many of the genes in the cluster and
while fabI seemed to be absent, one of the genes in the
cluster remained unannotated. Biochemical characterization
showed that the remaining ORF encoded an alternative
enoyl-ACP reductase (FabK), a homolog of which was later
also identified in E. faecalis. Consequently, inhibitors of
FabK could deliver selective streptococcal and enterococcal
agents whereas compounds that inhibited both FabK and
FabI could have broad spectrum utility.
3.4 Genes of Unknown Function
Clearly, essential genes encoding proteins of unknown
biochemical function constitute the most novel of all
possible antibiotic targets. More than 30% of the genes in a
bacterial genome are annotated as encoding proteins of
unknown function and at least 20% of these are broad
spectrum.
The difficulty in working with this type of target in drug
discovery is intrinsic in its nature. If their function and
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 299
Fig. (4). The fatty acid biosynthetic gene cluster in S. pneumoniae. The gene ncd-2 is similar to a 2-nitropropane dioxygenase gene
from Neurospora crassa. In fact, it encodes an enoyl-ACP reductase (FabK) that is distinct from orthologs encoded by fabI in other
bacteria.
enzymatic activity are unknown, they cannot be used in
high-throughput screening. A compromise can be achieved
by identifying essential genes encoding proteins whose
function can be predicted and here we discuss proteins
involved in stable RNA modification.
Table 4.
All living cells contain modified nucleosides in their
stable RNA (mostly ribosomal RNA and transfer RNA). To
date, a total of 96 modified nucleosides for which structures
have been assigned have been reported in RNA
(http://medlib.med.utah.edu/RNAmods). The distribution of
Stable RNA Modifying Enzymes. Genes Encoding tRNA or rRNA Modifying Enzymes were Tested for Essentiality in S.
pneumoniae. Those Required for Cell Viability could be Targets for Antibiotics
tRNA Modifying Enzymes
Accession #
Gene Name
Function
Essentiality data
AAL00255.1
hisT/truA
pseudouridine (ψ 38,39,40) synthetase
Non-essential
AAK99736.1
yerS
tRNA methyltransferase
Non-essential
AAL00520.1
yfjO
tRNA methyltransferase
Non-essential
AAK99491.1
trmD
tRNA (m1G37) methyltransferase
Essential
AAL00077.1
queA
SAM tRNA ribosyltransferase
Non-essential
AAL00671.1
tgt
queuine tRNA ribosyltransferase
NT
AAK99547.2
asuE/ trmU
tRNA (s2U34) thioltransferase
Essential
AAK99724.1
trmE
tRNA (cmnm5H34) methyltransferase
Essential
AAK99895.1
truB
pseudouridine (ψ 55) synthase
Non-essential
AAK99392.1
miaA
tRNA isopentenylpyrophosphate transferase
Non-essential
AAK99587.1
iscS
sulfurtransferase (s4U8 biosynthesis)
Non-essential
Accession #
Gene Name
Function
Essentiality data
AAL00382.1
fmu
16S rRNA methyltransferase (m5C907)
Non-essential
AAL00063.1
yloM
putative rRNA methyltransferase
Non-essential
AAK99280.1
ytmQ
putative rRNA methyltransferase
Non-essential
AAL00591.1
ysgA
rRNA methyltransferase
Non-essential
AAL00115.1
yacO
rRNA methyltransferase
Non-essential
AAK99236.1
cspR
rRNA methyltransferase
Non-essential
AAK99634.1
ylyB (rluD)
pseudouridine synthase
Non-essential
AAK99810.1
yjbO
pseudouridine synthase
Non-essential
AAL00492.1
ypuL1
pseudouridine 516 synthase
Non-essential
AAK99060.1
ypuL2
putative pseudouridine synthase
Non-essential
AAK99401.1
ypuL3
pseudouridine synthase
Essential
AAL00627.1
yhcT
putative pseudouridine synthase
Essential
rRNA Modifying Enzymes
NT=Not tested
300
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
these modifications is varied but a subset of them are present
in all three phylogenetic domains (Archaea, Bacteria and
Eukarya) sometimes even at comparative positions within
the RNA structure. On the other hand, there are also species
specific modifications (for reviews see, [35,36]).
In E. coli, there are 11 modified nucleosides in 16S
rRNA, 23 in 23S rRNA and 31 in tRNA. The formation of
these residues is catalyzed by highly specific enzymes
which, with the exception of the synthesis of quenosine,
operate during the maturation of the RNA after the primary
transcript is synthesized. It can be estimated that nearly 2%
of the genetic information in a bacteria is dedicated to the
synthesis of these modified residues and about half of the
enzymes involved in this process have not been identified.
This is clearly an under exploited target.
Bioinformatic analysis of completed genomes, have
identified 23 broad spectrum RNA modifying enzymes
(present in S. pneumoniae, S. aureus and H. influenzae),
nine of which have orthologs characterized in other
organisms (mostly E. coli). There is no biochemical
evidence for the function that the other 14 enzymes
performed, other than they modify RNA based on their
similarity to proteins of known function (Table 4).
Essentiality studies on 22 of these genes suggest that five of
them, TrmD, TrmE, TrmU, YhcT and ypuL3, are
indispensable for growth of S. pneumoniae. E. coli TrmD
and TrmE modify positions 37 and 34 respectively in certain
subsets of tRNAs and have been previously characterized
[37-39]. Phylogenetic analysis indicates that YhcT and
ypuL3 belong to the family of rRNA pseudouridine
synthetases but the position they modify is unknown. TrmU
has been annotated in the literature as a tRNA
thioltransferase [40] although there is no biochemical
evidence for that assertion and this needs to be confirmed.
Further studies on the essentiality of these proteins in other
organisms has shown that TrmE and ypuL3 are only
essential in S. pneumoniae and therefore constitute novel,
species specific targets. YhcT is also essential in S. aureus
but it has three distinct paralogs in H. influenzae and none
of them is essential individually. This would be a typical
Gram positive only target. On the other hand, TrmD and
TrmU are essential in both Gram positive and Gram negative
organisms and are, therefore, broad spectrum targets.
Consequently, genome sequence data can greatly expand
the number of therapeutic targets within a given class by
identifying putative orthologs.
4 TARGET SCREENING
4.1 Screening for Inhibitors
Despite the myriad of targets for antibacterial
intervention unveiled from genomic sequence analysis
through the process described above, and the fact that many
High-Throughput Screening (HTS) campaigns have been run
against dozens of these targets both at GlaxoSmithKline and
many other pharmaceutical companies, experience shows that
finding leads to these antimicrobial targets is more difficult
than for other therapeutic areas.
Holmes et al.
At GSK, a HTS success is defined as a screening
campaign that affords at least one lead compound of
sufficient quality to start a chemistry program, based on
structure, potency, selectivity and initial SAR trends. These
HTS campaigns are run according to well-established
standards of quality [41].
Possible causes for the lower success rate of obtaining
tractable hits in an antimicrobial HTS include the
"drugability" of the target and/or the diversity and size of the
screening collection. Target "drugability" refers to the
feasibility of finding drugs to any given target (reviewed in
[42]). In the last few years, the definition of a drug-like
molecule has become increasingly restricted to synthetic
small molecules that comply with the "rule-of-five" as
defined by Lipinski et al. [43]. The latter are a compendium
of guidelines derived from statistical observation of chemical
properties (molecular weight, lipophilicity as measured by
clogP, number of H bond donors and acceptors) of marketed
drugs. Their intent is to guide lead optimization programs
away from series or molecules with high probability of
failure due to poor absorption and/or poor permeability.
When comparing the success rate of in-house HTS
campaigns across different target classes some trends are
clearly observed. Most antibacterial targets screened are
enzymes other than protein kinases and proteases. When this
broad family of proteins is further analyzed, the success rates
within this group vary considerably between subfamilies.
Generally, HTS success correlates with the hydrophobicity
of the substrate binding pocket of the target. For example,
many chemically viable leads have been described for
enzymes involved in fatty acid biosynthesis [44,45], whereas
only a handful of less tractable ones have been discovered for
sugar binding enzymes [46].
One simplistic explanation is that enzymes that utilize
hydrophilic substrates (e.g. sugars) offer fewer opportunities
for interaction with small synthetic compounds because the
majority of these tend to be hydrophobic. This is due to the
nature of organic building blocks and their synthetic routes,
generally engineered in a water-free environment. On the
other hand, there is some evidence to suggest that
hydrophobic rather than hydrophilic ligands will form higher
affinity interactions because the water solvation of a
hydrophilic moiety imposes an energetic barrier to binding
[47]. As a result according to Lipinski et al., "one of the
most reliable methods in medicinal chemistry to improve in
vitro activity is to incorporate properly positioned lipophilic
groups".
In addition to the biochemical perspective described
above, most collections of synthetic molecules within
pharmaceutical and biotechnology companies contain a
significant proportion of compounds derived from specific
chemistry programs around the initial leads obtained after
successful screen campaigns. Accordingly, the majority of
molecules in such compound collections interact with certain
classes of G-protein coupled receptors, ion channels,
proteases, and more recently kinases, [48]. Consequently,
the probability of a new HTS target encountering high
affinity ligands in a synthetic compound screening collection
are higher if it is similar to a target that resulted in a
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 301
successful screening campaign and therefore contributed to
the growth of the collection. Nevertheless, serendipity plays
a role in HTS success, and novel targets can show high
affinity for compounds designed to interact with unrelated
proteins. For example, the series of methionyl-tRNA
synthetase inhibitors described by [49], evolved from an
HTS hit originally synthesized in an optimization program
for antagonists of histamine H1 receptor.
Given the low success of genomics-derived, isolated,
molecular targets from bacteria in HTS, another approach
[53] is to screen whole pathways or whole cells. The burden
of these approaches is that if interesting leads are found, a
labor intensive target identification effort must follow with
uncertain results. However, the advantage remains that the
intractable issue of cell penetrability is addressed up front
[54,55].
As discussed above, synthetic molecules tend to be
lipophilic. One might speculate on the degree to which the
Lipinski rules are a result of this general hydrophobia – i.e.
drug-like properties are so defined because molecules with
different properties are not progressed and therefore not
tested sufficiently – and how much they reflect the many
characteristics a drug must display within a physiological
environment: specific and potent binding, access to its
target, low adsorption to cellular and extracellular milieu
components, selectivity against many related and unrelated
proteins, in summary good ADME (absorption, distribution,
metabolism, excretion) and low toxicology.
There are a number of avenues, that are just starting to be
explored, which could potentially fill the current gap in the
global antibacterials pipeline:
The fact that many marketed antibiotics, and other
natural products, are apparently the main exception to the
rule-of-five seems to support this line of thought: a
historically biased view of what constitutes a drug-like
molecule may preclude finding antibiotics through the
current Drug Discovery paradigm. In the past few years,
HTS of natural products has been losing support in many
pharmaceutical companies because of a cost-benefit analysis
that some believe is short-sighted [50]. The fact remains that
the return in value from traditional approaches to natural
products screening has been in decline for the last two
decades, mainly because of the labor intensive nature of this
work and the fact that many of the metabolites found were
either already known, and thus unexploitable from an
intellectual property perspective, or too complex to enter a
chemical optimization program. However, many marketed
drugs are natural products that did not require any
derivatization to be effective and safe (e.g. lovastatin,
cyclosporin, clavulanic acid, erythromycin, caffeine, taxol).
Many of these compounds are complex chiral molecules that
contain several polar substituents – ideal for inhibiting less
tractable targets such as protein-protein interactions and
enzymes that act on polar substrates. These compounds may
not be “drug-like” – but they are drugs! A biased view of the
basic criteria in current drug discovery (a lead has to be
amenable to chemical modification to drive optimization
throughout drug development) may have diminished the
chances of this proven source of effective antibiotics to add
to our arsenal of weapons against bacterial infections.
Interestingly, and despite the exception to the rule-of-five
observed for some drugs of natural origin, recent studies
have shown that collections of plant and microbial
metabolites are, on average, more similar to synthetic
compound collections than previously thought and most of
them lie within the limits suggested by Lipinski et al.
[51,52]. Thus, there is scope for the addition of these kinds
of compounds to current collections to enrich the diversity,
broaden the physico-chemical space and maximize the
chances of finding leads for less tractable but highly
validated targets.
•
exploitation of untapped sources of microbial
metabolites (expression of biosynthetic pathways
from unculturable microorganisms; see section 4.2)
•
reduce the risks and cost of the traditional extract
screening by screening purified or semipurified
samples [56].
•
incorporate scaffolds from natural products into
combinatorial chemistry approaches [57].
•
derivatize natural products by biotransformation [58].
•
structure-based screening and design [59,60].
Finally, when analyzing the success of HTS in this or
any other area, it is fair to consider that the drug discovery
process has been reshaped in the last decade and we are only
now starting to see the fruits of this work entering the
development pipelines. The load of new knowledge
generated by high-throughput technologies applied in the
last 10 years to molecular biology, biochemistry and
chemistry and the increased help from computational tools
will hopefully start to pay off in the next few years and truly
new antibiotics – those with novel modes of action and
enhanced properties - will flow to the market.
4.2 Combinatorial Genomics
Identification of targets and development of robust highthroughput assays allows screening of millions of
compounds for inhibitory activity. Most of the currently
commercialized antibiotics are derived from natural product
extracts and at least four, the macrolides azithromycin and
clarithromycin, the cephalosporin ceftriaxone and the betalactam amoxicilin in conjunction with another natural
product, the beta-lactamase inhibitor, clavulanic acid, each
have annual sales in excess of $1B. In order to discover new
antibacterial compounds it is imperative that the novel
targets unraveled by genomics are screened against a wide
diversity of compounds which will come from new organic
syntheses or from the identification of novel natural products
produced by soil and marine organisms as well as plants.
Bacterial diversity is being exploited by searching extreme
habitats for secondary metabolite producers. However,
genomics is also being applied to the problem.
Natural products are complex molecules largely (though
not exclusively) produced by soil microorganisms such as
302
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Streptomyces. However, the vast majority of microbial
species cannot be cultivated under laboratory conditions.
Indeed, it is estimated that only 0.1-1% of microorganisms
can be isolated [61]. Several biotechnology companies are
now exploiting the ability of heterologous hosts to express
DNA containing secondary metabolic pathways extracted
from soil and thereby identify novel compounds [62]. In fact
they have already been able to demonstrate the production of
previously unidentified secondary metabolites using this
methodology [63] thereby extending the spectrum of
potential pharmaceuticals. This approach allows the rapid
isolation of the biosynthetic gene cluster. Subsequently, it
can be manipulated to provide intermediates and derivatives
of complex chiral molecules that would be refractory to
chemical synthesis and may be vital to determine SAR
[64,65].
Finally, the streptomycetes are one of the major
producers of secondary metabolites and the complete genome
sequence of Streptomyces coelicolor was recently published
[66]. Following decades of study, S. coelicolor was
demonstrated to produce four secondary metabolites under a
number of laboratory conditions. The genome sequence
reveals that S. coelicolor may well be capable of producing
as many as 20 secondary metabolites, all of which probably
make specific interactions with macromolecules and some of
which may prove to have some therapeutic use.
5. DETERMINATION OF MECHANISM OF ACTION
OF ANTIBACTERIALS
If an inhibitory compound has been identified by a target
based screen, it is important to confirm that the antibacterial
activity against whole bacteria is mediated via the suspected
target. On the other hand, if the antimicrobial compound
was isolated from a whole cell screen, then a concerted effort
must be made to identify the molecular mode of action.
Holmes et al.
Genomics can contribute to both of these processes and
examples are discussed below.
5.1 Regulated Gene Expression
Tightly-regulated, inducible promoters have been used in
antimicrobial drug discovery for demonstrating gene
essentiality, and hence, the validation of drug targets
[67,68]. Moreover, titratible promoter systems that are able
to modulate the levels of the protein target have proven to be
invaluable tools for tracking the mechanism of antibacterial
activity of novel inhibitors. The Pspac promoter (previously
characterized in B. subtilis) has been used in S. aureus to
demonstrate that methionyl-tRNA synthetase (MRS) and
peptide deformylase (PDF) are essential for cell viability
[67]. Moreover, the MICs of inhibitors of these enzymes
were directly related to their level of expression, correlating
whole cell activity with inhibition of the specific target.
Since such appropriate promoter systems were
unavailable in S. pneumoniae, we have applied genomics to
the problem (Chan P. et al., manuscript submitted). The
entire genome of S. pneumoniae was scanned for putative
novel sugar operons which are typically highly regulated. At
least 14 putative carbohydrate utilization operons were
identified, including those for putative cellobiose, fucose,
fructose, galactose, glucose, lactose, maltose, mannitol,
mannose, sucrose, trehalose and raffinose, and other
unknown sugar utilization operons. Most gene clusters
contained putative regulatory, uptake and sugar utilization
determinants. Bioinformatic analysis of the regulatory
regions in these operons identified a promoter within the
fucose gene cluster that had the requisite properties for
development of an inducible promoter system in S .
pneumoniae. The fucose operon contains 10 genes, many of
which show homology to the fucose catabolism genes of
E. coli and H. influenzae. The first gene, fcsK, encodes a
Fig. (5). (a) Genes under the control of an inducible promoter. By replacing the native promoter with an inducible promoter the level
of expression can be controlled. (b) Inducer-dependent growth of S. pneumoniae (PfcsK::defI) regulated strain confirms PDF is essential
for growth and a suitable antibacterial drug target.
A Genomics Approach to Drug Discovery
putative fuculose kinase enzyme in S. pneumoniae. fcsK is
divergently transcribed from a gene whose product shows
homology to the LacR family of transcriptional repressors
and therefore indicating that the fcsK promoter is subject to
down-regulation, and hence, an excellent candidate for
further study. Transcript analysis revealed a near canonical
bacterial promoter with a transcriptional start site located 24
bp upstream of fcsK. As predicted, the addition of fucose
induced expression of fcsK (>23-fold). While galactose (a
closely related sugar) was also able to induce f c s K
expression; raffinose, lactose, trehalose and mannose had no
significant effect on fcsK transcript levels. Moreover,
transcription of fcsK was tightly repressed by sucrose or
glucose (at least 10-fold). Hence the PfcsK promoter is
inducible by fucose and repressible by sucrose giving a
titratible dynamic range.
Several S. pneumoniae genes of interest have been placed
under the control of the fcsK promoter including def1 which
encodes PDF. The strategy for construction of the regulated
strain is outlined in Fig. (5a) and involves replacing the
wild-type promoter with P f c s K by allelic exchange
mutagenesis. The regulated S. pneumoniae PfcsK::def1 strain
in Fig (5b) shows an absolute dependence on inducer fucose
for normal growth confirming the essential nature of PDF
for growth of S. pneumoniae.
The availability of genomics data has allowed us to
successfully identify a fucose-regulated, endogenous
promoter from S. pneumoniae and its exploit it as a tool for
target validation and antimicrobial mode of action studies.
5.2 Antisense
In addition to the types of experiments described in the
previous section, regulatable promoters can also be used to
quantitatively express anti-sense RNA to specific transcripts
for essential gene products [69]. Fragments of essential
genes are inserted into a plasmid, downstream of the Pxyl-tet
controllable promoter, in their reverse orientation. While
cells are viable in the absence of inducer (since the wild-type
gene is transcribed and translated), in the presence of inducer
tetracycline, anti-sense RNA is generated, leading to
degradation of the native mRNA and inhibition of bacterial
growth. This system is particularly elaborate because by
feeding animals tetracycline, it is possible to induce antisense in experimental animal models of infection and
demonstrate that genes not required for growth on agar
plates, are essential for pathogenesis [70].
5.3 Microarray Gridding
Microarray gridding is a powerful and quantitative
genomic tool for analyzing the genome-wide expression of
genes in bacteria. Since gene expression is primarily
regulated at the level of transcription, the messenger RNA
(mRNA) profile of completed genomes can provide a global,
genomic snapshot of the transcriptome. Gene expression
patterns generated are reflective of the physiological state of
the cell [71]. Microarrays have been used to determine the
mode of action of drugs by comparing the expressions in
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 303
drug-resistant and wild type strains, or in cells treated with
and without drug. Using this approach the molecular target
or cellular pathway being affected may be identified or
predicted. Another powerful application of microarrays is the
identification of the role of genes of unknown function in
completed genomes, also called functional genomics.
Details of DNA microarray technology, and its various
applications in microbial systems have been extensively
reviewed elsewhere [72-74]. Typically, a microarray consists
of DNA representing every ORF of a genome, stamped onto
a glass slide or nylon membrane. In mRNA expression
profiling experiments, total RNA is commonly isolated
from bacteria exposed to drugs, labeled, and hybridized to a
microarray grid. Genes differentially expressed i.e. up- or
down-regulated following compound treatment are
identified. Microarray gridding technology has been applied
to study bacterial regulatory networks, genetic alterations,
host-microbe interactions, strain genotypes and validate drug
targets, as well as determine drug-resistance mechanisms and
antibacterial compound modes of action in many human
pathogens. Microbial microarrays are available for many of
the genomes sequenced to date including those for E. coli
[75], B. subtilis [76], Saccharomyces cerevisiae [77,78],
Mycobacterium tuberculosis [79,80], H. pylori [81], P.
aeruginosa [82], H. influenzae [71], S. pneumoniae [83,84],
and S. aureus1 [85,86].
Here, we focus on the impact of microarrays in antibiotic
drug discovery and give our experiences of using microarrays
to study the antibacterial mode of action of candidate lead
compounds. A S. aureus array platform covering about 80%
of the ORFs in the genome was constructed and used to
profile1 four antibacterial compounds of known modes of
action . Following two-color microarray hybridization, the
response of each gene was compared in drug-treated and
untreated samples, Fig. (6 ). mRNA profiling of
gemifloxacin and ciprofloxacin, both quinolone class of
DNA replication inhibitors, resulted in generally similar upregulation of genes associated with the SOS response and
DNA repair mechanisms including recA, lexA, and uvrBA
(excinuclease subunits). Triclosan, an inhibitor of FabI (see
section 3.3) induced a strong up-regulation of heat shock
proteins such as dnaK, ctsR, clpB, and groEL and some
genes involved in fatty acid biosynthesis, and caused a
down-regulation of purine biosynthetic genes and ribosomal
proteins. In contrast, mupirocin an inhibitor of protein
biosynthesis (see section 3.1) most strongly induced genes
involved in branched chain amino acid biosynthesis1. Genes
affected were generally consistent with inhibition of the
metabolic pathway targeted by the different antibiotics. In
addition, the expressions of many genes of unknown
function were altered by drug treatments implying putative
roles in the response of S. aureus to antibiotic stress and
survival. The co-regulation of genes organized in the same
operon in response to drug treatment further validated the
data. Other examples of microarray drug profiling studies
include isoniazid, a fatty acid pathway inhibitor in M .
1
Chan, P.F.; Lonetto, M.; Clark, S.M.; Gagnon, R.; Palmer, L.M.; Woodnutt, G.;
Warren, P.; Jaworski, D.D. 2001, Abstract A-26. 101st American Society of
Microbiology General Meeting, Orlando, FL
304
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Holmes et al.
Fig. (6). Example of a microarray following gene expression profiling of an antibiotic in S. aureus. Test (drug treated or untreated)
and reference RNA isolated from S. aureus were reverse transcribed to cDNA, labelled with fluorescence dyes Cy3 and Cy5
respectively, and hybridized onto a 1974 ORF microarray grid. Genes up- and down-regulated or unaffected in the test sample
compared to the reference control, are shown by green, red and orange spots respectively. Antibiotic-responsive genes are identified
by comparison of intensity ratios of each gene in the drug treated and the untreated samples.
tuberculosis [79] and ciprofloxacin and novobiocin in H.
influenzae [71]. In both cases, drug-specific gene expression
signatures were identified consistent with drug mechanisms
of action.
We have profiled more than 12 known antibiotics of
different modes of action in S. aureus including sub-classes
of inhibitors, and in most cases, identified distinct 2drug
signature expression patterns for these compounds . By
establishing a data bank of expression profiles, the mode of
action of antibacterial compounds identified from a bacterial
whole cell screen may be classified according to the their
transcriptome response. Microarray gene expression profiling
though a powerful tool will not always identify the precise
molecular target of the inhibitor but often predicts the
metabolic pathway being affected.
Genome-wide profiling using microarrays clearly has a
role, in conjunction with other molecular mode of action
tools, to support SAR efforts during lead optimization.
Since expression of most of the genes on the grid is
unaltered following drug treatment, once a cluster of the 300
most drug-responsive genes has been identified, a mini-array
may then be constructed to facilitate throughput during a
2
Chan, P.F.; Gagnon, R.; Clark, S.M.; O'Brien, S.; Boyle, R.; Javed, R.; Au, J.;
Lonetto, M.; Jaworski, D.D. 2002, Abstract A-43. 102nd American Society of
Microbiology.
lead validation and optimization project. Antibiotic gene
signatures identified by microarray profiling may also be
developed into a panel of cellular reporter strains as an
alternative novel screening method for predicting mechanism
of action or as a secondary assay for chemical optimization2.
Though still a relatively new technology, microarray
gridding has already become a useful tool in antimicrobial
drug discovery 2 [71,85]. Furthermore, microarrays are
invaluable for studying the function of genes of completed
genomes. In functional genomics, the role of essential
proteins of unknown biochemical function (see section 3.4)
might be elucidated using microarrays. Expression profiling
of regulated strains may lead to the identification of new
drug targets and development of possible assays. Other
current applications of microarrays include the study of
bacterial host interactions whereby total RNA is isolated
from infected tissue samples and hybridized to both
microbial and human/rat arrays [87,88]. In summary, we
have applied microarrays to both predict the mechanism of
action of novel antibacterial agents identified from a whole
cell screen and for validating the target of lead hits from a
HTS and prioritize these compounds for development.
Microarray gridding is a powerful tool for genome-wide
gene expression profiling, which is becoming widely
accepted while supplementing other established methods to
accelerate the antibacterial drug discovery process.
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 305
Dew, I.; Fasulo, D.; Flanigan, M.; Florea, L.; Halpern, A.;
Hannenhalli, S.; Kravitz, S.; Levy, S.; Mobarry, C.;
Reinert, K.; Remington, K.; Abu-Threideh, J.; Beasley, E.;
Biddick, K.; Bonazzi, V.; Brandon, R.; Cargill, M.;
Chandramouliswaran, I.; Charlab, R.; Chaturvedi, K.;
Deng, Z.; Di, F.V.; Dunn, P.; Eilbeck, K.; Evangelista, C.;
Gabrielian, A.E.; Gan, W.; Ge, W.; Gong, F.; Gu, Z.; Guan,
P.; Heiman, T.J.; Higgins, M.E.; Ji, R.R.; Ke, Z.; Ketchum,
K.A.; Lai, Z.; Lei, Y.; Li, Z.; Li, J.; Liang, Y.; Lin, X.; Lu,
F.; Merkulov, G.V.; Milshina, N.; Moore, H.M.; Naik,
A.K.; Narayan, V.A.; Neelam, B.; Nusskern, D.; Rusch,
D.B.; Salzberg, S.; Shao, W.; Shue, B.; Sun, J.; Wang, Z.;
Wang, A.; Wang, X.; Wang, J.; Wei, M.; Wides, R.; Xiao,
C.; Yan, C.; Yao, A.; Ye, J.; Zhan, M.; Zhang, W.; Zhang,
H.; Zhao, Q.; Zheng, L.; Zhong, F.; Zhong, W.; Zhu, S.;
Zhao, S.; Gilbert, D.; Baumhueter, S.; Spier, G.; Carter, C.;
Cravchik, A.; Woodage, T.; Ali, F.; An, H.; Awe, A.;
Baldwin, D.; Baden, H.; Barnstead, M.; Barrow, I.;
Beeson, K.; Busam, D.; Carver, A.; Center, A.; Cheng,
M.L.; Curry, L.; Danaher, S.; Davenport, L.; Desilets, R.;
Dietz, S.; Dodson, K.; Doup, L.; Ferriera, S.; Garg, N.;
Gluecksmann, A.; Hart, B.; Haynes, J.; Haynes, C.; Heiner,
C.; Hladun, S.; Hostin, D.; Houck, J.; Howland, T.;
Ibegwam, C.; Johnson, J.; Kalush, F.; Kline, L.; Koduru,
S.; Love, A.; Mann, F.; May, D.; McCawley, S.; McIntosh,
T.; McMullen, I.; Moy, M.; Moy, L.; Murphy, B.; Nelson,
K.; Pfannkoch, C.; Pratts, E.; Puri, V.; Qureshi, H.;
Reardon, M.; Rodriguez, R.; Rogers, Y.H.; Romblad, D.;
Ruhfel, B.; Scott, R.; Sitter, C.; Smallwood, M.; Stewart,
E.; Strong, R.; Suh, E.; Thomas, R.; Tint, N.N.; Tse, S.;
Vech, C.; Wang, G.; Wetter, J.; Williams, S.; Williams,
M.; Windsor, S.; Winn-Deen, E.; Wolfe, K.; Zaveri, J.;
Zaveri, K.; Abril, J.F.; Guigo, R.; Campbell, M.J.;
Sjolander, K.V.; Karlak, B.; Kejariwal, A.; Mi, H.;
Lazareva, B.; Hatton, T.; Narechania, A.; Diemer, K.;
Muruganujan, A.; Guo, N.; Sato, S.; Bafna, V.; Istrail, S.;
Lippert, R.; Schwartz, R.; Walenz, B.; Yooseph, S.; Allen,
D.; Basu, A.; Baxendale, J.; Blick, L.; Caminha, M.;
Carnes-Stine, J.; Caulk, P.; Chiang, Y.H.; Coyne, M.;
Dahlke, C.; Mays, A.; Dombroski, M.; Donnelly, M.; Ely,
D.; Esparham, S.; Fosler, C.; Gire, H.; Glanowski, S.;
Glasser, K.; Glodek, A.; Gorokhov, M.; Graham, K.;
Gropman, B.; Harris, M.; Heil, J.; Henderson, S.; Hoover,
J.; Jennings, D.; Jordan, C.; Jordan, J.; Kasha, J.; Kagan,
L.; Kraft, C.; Levitsky, A.; Lewis, M.; Liu, X.; Lopez, J.;
Ma, D.; Majoros, W.; McDaniel, J.; Murphy, S.; Newman,
M.; Nguyen, T.; Nguyen, N.; Nodell, M. Science, 2001,
291, 1304.
6. CONCLUDING REMARKS
Genomics has supplied us with every target in the
bacterial cell. Imaginative science is now required to exploit
this embarrassment of riches. It is now feasible to target
therapy at particular pathogens, whether they be individual
organisms such as H. pylori or specific groups such as Gram
positive bacteria, leaving the remaining flora unaffected.
Alternatively, inhibitors of broad spectrum targets, present
in all bacteria can be used when rapid diagnosis is not
possible. Certainly, these genes have been identified and, in
many cases, their essentiality determined. Given this
situation, why are we not inundated with novel antibacterial
agents? It is likely that the targets have not been screened
against sufficient chemical diversity to allow the
identification of adequate lead compounds. However, novel
technologies are now addressing this problem and genomics
will again play a pivotal role. Without doubt, novel
antibiotics that inhibit new bacterial targets are essential if
the current resistance problems are to be overcome.
ABBREVIATIONS
RNA
=
Ribonucleic acid
tRNA
=
Transfer ribonucleic acid
mRNA
=
Messenger ribonucleic acid
MRS
=
Methionyl-tRNA synthetase
TCSTS
=
Two-component signal transduction system
FAB
=
Fatty acid biosynthesis
ACP
=
Acyl carrier protein
HTS
=
High-throughput screening
SAR
=
Structure activity relationship
PDF
=
Peptide deformylasse
RTI
=
Respiratory tract infection
ORF
=
Open reading frame
[3]
Apfel, C.M.; Takacs, B.; Fountoulakis, M.; Stieger, M.;
Keck, W. J. Bacteriol., 1999, 181, 483.
[4]
Tatusov, R.L.; Koonin, E.V.; Lipman, D.J. Science, 1997,
278, 631.
REFERENCES
[1]
Fleischmann, R.D.; Adams, M.D.; White, O.; Clayton,
R.A.; Kirkness, E.F.; Kerlavage, A.R.; Bult, C.J.; Tomb,
J.F.; Dougherty, B.A.; Merrick, J.M. Science, 1995, 269,
496.
[5]
Tatusov, R.L.; Natale, D.A.; Garkavtsev, I.V.; Tatusova,
T.A.; Shankavaram, U.T.; Rao, B.S.; Kiryutin, B.;
Galperin, M.Y.; Fedorova, N.D.; Koonin, E.V. Nucleic
Acids Res., 2001, 29, 22.
[2]
Venter, J.C.; Adams, M.D.; Myers, E.W.; Li, P.W.; Mural,
R.J.; Sutton, G.G.; Smith, H.O.; Yandell, M.; Evans, C.A.;
Holt, R.A.; Gocayne, J.D.; Amanatides, P.; Ballew, R.M.;
Huson, D.H.; Wortman, J.R.; Zhang, Q.; Kodira, C.D.;
Zheng, X.H.; Chen, L.; Skupski, M.; Subramanian, G.;
Thomas, P.D.; Zhang, J.; Gabor Miklos, G.L.; Nelson, C.;
Broder, S.; Clark, A.G.; Nadeau, J.; McKusick, V.A.;
Zinder, N.; Levine, A.J.; Roberts, R.J.; Simon, M.;
Slayman, C.; Hunkapiller, M.; Bolanos, R.; Delcher, A.;
[6]
Arigoni, F.; Talabot, F.; Peitsch, M.; Edgerton, M.D.;
Meldrum, E.; Allet, E.; Fish, R.; Jamotte, T.; Curchod,
M.L.; Loferer, H. Nat. Biotechnol., 1998, 16, 851.
[7]
Tomb, J.F.; White, O.; Kerlavage, A.R.; Clayton, R.A.;
Sutton, G.G.; Fleischmann, R.D.; Ketchum, K.A.; Klenk,
H.P.; Gill, S.; Dougherty, B.A.; Nelson, K.; Quackenbush,
J.; Zhou, L.; Kirkness, E.F.; Peterson, S.; Loftus, B.;
Richardson, D.; Dodson, R.; Khalak, H.G.; Glodek, A.;
306
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Holmes et al.
McKenney, K.; Fitzegerald, L.M.; Lee, N.; Adams, M.D.;
Venter, J.C. .Nature, 1997, 388, 539.
[27]
Cheng, Q.; Campbell, E.A.; Naughton, A.M.; Johnson, S.;
Masure, H.R. Mol. Microbiol., 1997, 23, 683.
[8]
Reich, K.A.; Chovan, L.; Hessler, P. J. Bacteriol., 1999,
181, 4961.
[28]
Novak, R.; Cauwels, A.; Charpentier, E.; Tuomanen, E. J.
Bacteriol., 1999, 181, 1126.
[9]
Akerley, B.J.; Rubin, E.J.; Novick, V.L.; Amaya, K.;
Judson, N.; Mekalanos, J.J. Proc. Natl. Acad. Sci. USA,
2002, 99, 966.
[29]
Novak, R.; Henriques, B.; Charpentier, E.; Normark, S.;
Tuomanen, E. Nature, 1999, 399, 590.
[30]
[10]
Norgren, M.; Caparon, M.G.; Scott, J.R. Infect. Immun.,
1989, 57, 3846.
Rodrigue, A.; Quentin, Y.; Lazdunski, A.; Mejean, V.;
Foglino, M. Trends Microbiol., 2000, 8, 498.
[31]
[11]
Hynes, W.L.; Hancock, L.; Ferretti, J.J. Infect. Immun.,
1995, 63, 3015.
Throup, J.P.; Koretke, K.K.; Bryant, A.P.; Ingraham, K.A.;
Chalker, A.F.; Ge, Y.; Marra, A.; Wallis, N.G.; Brown, J.R.;
Holmes, D.J.; Rosenberg, M.; Burnham, M.K. M o l .
Microbiol., 2000, 35, 566.
[12]
Buckley, N.D.; Lee, L.N.; LeBlanc, D.J. J. Bacteriol., 1995,
177, 5028.
[32]
Fabret, C.; Hoch, J.A. J. Bacteriol., 1998, 180, 6375.
[13]
O'Connell, C.; Pattee, P.A.; Foster, T.J. J. Gen. Microbiol.,
1993, 139, (Pt 7), 1449.
[33]
Martin, P.K.; Li, T.; Sun, D.; Biek, D.P.; Schmid, M.B. J.
Bacteriol., 1999, 181, 3666.
[14]
Meinnel, T.; Mechulam, Y.; Blanquet, S. In t R N A :
Structure, Biosynthesis, and Function; Soll, D.;
RajBhandary, U. Eds. AMS Press, Washington, D.C.
1995, 251.
[34]
Payne, D.J.; Warren, P.V.; Holmes, D.J.; Ji, Y.; Lonsdale,
J.T. Drug Discov. Today, 2001, 6, 537.
[35]
Bjork, G.R. In tRNA: Structure, Biosynthesis, and
Function; Soll, D., RajBhandary, U. Eds. AMS Press,
Washington, D.C. 1995, 165.
[36]
Bjork, G.R. In Escherichia coli and Salmonella. Cellular
and molecular biology; Neidhardt, F.C. ed. AMS Press,
Washington, D.C. 1996, 861.
[37]
Hjalmarsson, K.J.; Bystrom, A.S.; Bjork, G.R. J. Biol.
Chem., 1983, 258, 1343.
[38]
Redlak, M.; Andraos-Selim, C.; Giege, R.; Florentz, C.;
Holmes, W.M. Biochemistry, 1997, 36, 8699.
[39]
Cabedo, H.; Macian, F.; Villarroya, M.; Escudero, J.C.;
Martinez-Vicente, M.; Knecht, E.; Armengod, M.E. EMBO
J., 1999, 18, 7063.
[40]
Green, S.M.; Malik, T.; Giles, I.G.; Drabble, W.T.
Microbiology, 1996, 142, 3219.
[41]
Macarron, R.; Hertzberg, R.P. In High Throughput
Screening Methods and Protocols; Janzen, W.P. ed.
Humana Press, Totowa, NJ. 2002, 1.
[15]
Schon, A.; Kannangara, C.G.; Gough, S.; Soll, D. Nature,
1988, 331, 187.
[16]
Grundy, F.J.; Haldeman, M.T.; Hornblow, G.M.; Ward,
J.M.; Chalker, A.F.; Henkin, T.M. J. Bacteriol., 1997, 179,
3767.
[17]
Green, C.J.; Vold, B.S. GenBank Accession number
L36472, 1994.
[18]
Payne, D.J.; Wallis, N.G.; Gentry, D.R.; Rosenberg, M.
Current Opinion in Drug Discovery and Development,
2000, 3, 177.
[19]
Appleby, J.L.; Parkinson, J.S.; Bourret, R.B. Cell, 1996,
86, 845.
[20]
Egger, L.A.; Park, H.; Inouye, M. Genes Cells, 1997, 2,
167.
[21]
Stock, J.B.; Ninfa, A.J.; Stock, A.M. Microbiol. Rev.,
1989, 53, 450.
[22]
Barrett, J.F.; Hoch, J.A. Antimicrob. Agents Chemother.,
1998, 42, 1529.
[42]
Viswanadhan, V.N.; Balan, C.; Hulme, C.; Cheetham, J.C.;
Sun, Y. Curr. Opin. Drug Discov. Devel., 2002, 5, 400.
[23]
Barrett, J.F.; Goldschmidt, R.M.; Lawrence, L.E.; Foleno,
B.; Chen, R.; Demers, J.P.; Johnson, S.; Kanojia, R.;
Fernandez, J.; Bernstein, J.; Licata, L.; Donetz, A.; Huang,
S.; Hlasta, D.J.; Macielag, M.J.; Ohemeng, K.; Frechette,
R.; Frosco, M.B.; Klaubert, D.H.; Whiteley, J.M.; Wang,
L.; Hoch, J.A. Proc. Natl. Acad. Sci. USA, 1998, 95, 5317.
[43]
Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J.
Advanced Drug Delivery Reviews, 1997, 23, 3.
[44]
Miller, W.H.; Seefeld, M.A.; Newlander, K.A.; Uzinskas,
I.N.; Burgess, W.J.; Heerding, D.A.; Yuan, C.C.; Head,
M.S.; Payne, D.J.; Rittenhouse, S.F.; Moore, T.D.;
Pearson, S.C.; Berry, V.; DeWolf, W.E. Jr.; Keller, P.M.;
Polizzi, B.J.; Qiu, X.; Janson, C.A.; Huffman, W.F. J. Med.
Chem., 2002, 45, 3246.
[45]
Payne, D.J.; Miller, W.H.; Berry, V.; Brosky, J.; Burgess,
W.J.; Chen, E.; DeWolf, W.E. Jr.; Fosberry, A.P.;
Greenwood, R.; Head, M.S.; Heerding, D.A.; Janson, C.A.;
Jaworski, D.D.; Keller, P.M.; Manley, P.J.; Moore, T.D.;
Newlander, K.A.; Pearson, S.; Polizzi, B.J.; Qiu, X.;
Rittenhouse, S.F.; Slater-Radosti, C.; Salyers, K.L.;
Seefeld, M.A.; Smyth, M.G.; Takata, D.T.; Uzinskas, I.N.;
Vaidya, K.; Wallis, N.G.; Winram, S.B.; Yuan, C.C.;
[24]
Guenzi, E.; Gasc, A.M.; Sicard, M.A.; Hakenbeck, R. Mol.
Microbiol., 1994, 12, 505.
[25]
Zahner, D.; Grebe, T.; Guenzi, E.; Krauss, J.; van der, L.M.;
Terhune, K.; Stock, J.B.; Hakenbeck, R. Microb. Drug
Resist., 1996, 2, 187.
[26]
Pestova, E.V.; Havarstein, L.S.; Morrison, D.A. Mol.
Microbiol., 1996, 21, 853.
A Genomics Approach to Drug Discovery
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4 307
S.; Huang, C.H.; Kieser, T.; Larke, L.; Murphy, L.; Oliver,
K.; O'Neil, S.; Rabbinowitsch, E.; Rajandream, M.A.;
Rutherford, K.; Rutter, S.; Seeger, K.; Saunders, D.; Sharp,
S.; Squares, R.; Squares, S.; Taylor, K.; Warren, T.;
Wietzorrek, A.; Woodward, J.; Barrell, B.G.; Parkhill, J.;
Hopwood, D.A. Nature, 2002, 417, 141.
Huffman, W.F. Antimicrob. Agents Chemother., 2002, 46,
3118.
[46]
Baum, E.Z.; Montenegro, D.A.; Licata, L.; Turchi, I.;
Webb, G.C.; Foleno, B.D.; Bush, K. Antimicrob. Agents
Chemother., 2001, 45, 3182.
[47]
Morgan, B.P.; Scholtz, J.M.; Ballinger, M.D.; Zipkin, I.D.;
Bartlett, P.A. Journal of the American Chemical Society,
1991, 113, 297.
[67]
Zhang, L.; Fan, F.; Palmer, L.M.; Lonetto, M.A.; Petit, C.;
Voelker, L.L.; St John, A.; Bankosky, B.; Rosenberg, M.;
McDevitt, D. Gene, 2000, 255, 297.
[48]
Hopkins, A.L.; Groom, C.R. Nat. Rev. Drug Discov., 2002,
1, 727.
[68]
[49]
Jarvest, R.L.; Berge, J.M.; Berry, V.; Boyd, H.F.; Brown,
M.J.; Elder, J.S.; Forrest, A.K.; Fosberry, A.P.; Gentry,
D.R.; Hibbs, M.J.; Jaworski, D.D.; O'Hanlon, P.J.; Pope,
A.J.; Rittenhouse, S.; Sheppard, R.J.; Slater-Radosti, C.;
Worby, A. J. Med. Chem., 2002, 45, 1959.
Fan, F.; Lunsford, R.D.; Sylvester, D.; Fan, J.; Celesnik,
H.; Iordanescu, S.; Rosenberg, M.; McDevitt, D. Plasmid,
2001, 46, 71.
[69]
Ji, Y.; Zhang, B.; Van Horn, S.F.; Warren, P.; Woodnutt,
G.; Burnham, M.K.; Rosenberg, M. Science, 2001, 293,
2266.
[70]
Ji, Y.; Marra, A.; Rosenberg, M.; Woodnutt, G. J .
Bacteriol., 1999, 181 , 6585.
[50]
Demain, A.L. Nat. Biotechnol., 2002, 20, 331.
[51]
Henkel, T.; Brunne, R.M.; Muller, H.; Reichel, F. Angew.
Chem. Int. Ed., 1999, 38, 643.
[71]
Gmuender, H.; Kuratli, K.; Di Padova, K.; Gray, C.P.;
Keck, W.; Evers, S. Genome Res., 2001, 11, 28.
[52]
Lee, M.L.; Schneider, G. J. Comb. Chem., 2001, 3, 284.
[72]
[53]
Wijkmans, J.C.; Beckett, R.P. Drug Discov. Today, 2002,
7, 126.
Lucchini, S.; Thompson, A.; Hinton, J.C. Microbiology,
2001, 147, 1403.
[73]
Ye, R.W.; Wang, T.; Bedzyk, L.; Croker, K.M. J .
Microbiol. Methods, 2001, 47, 257.
[74]
Freiberg, C.; Brunner, N.A. Targets, 2002, 1, 20.
[75]
Tao, H.; Bausch, C.; Richmond, C.; Blattner, F.R.;
Conway, T. J. Bacteriol., 1999, 181, 6425.
An, H.; Haly, B.D.; Cook, P.D. J. Med. Chem., 1998, 41,
706.
[76]
Ye, R.W.; Tao, W.; Bedzyk, L.; Young, T.; Chen, M.; Li, L.
J. Bacteriol., 2000, 182, 4458.
Manly, S.P.; Padmanabha, R.; Lowe, S.E. Methods Mol.
Biol., 2002, 190, 153.
[77]
Bammert, G.F.; Fostel, J.M. Antimicrob.
Chemother., 2000, 44, 1255.
[78]
Schena, M.; Shalon, D.; Davis, R.W.; Brown, P.O. Science,
1995, 270, 467.
[79]
Wilson, M.; DeRisi, J.; Kristensen, H.H.; Imboden, P.;
Rane, S.; Brown, P.O.; Schoolnik, G.K. Proc. Natl. Acad.
Sci. USA, 1999, 96, 12833.
[54]
[55]
[56]
Kung, P.P.; Casper, M.D.; Cook, K.L.; Wilson-Lingardo,
L.; Risen, L.M.; Vickers, T.A.; Ranken, R.; Blyn, L.B.;
Wyatt, J.R.; Cook, P.D.; Ecker, D.J. J. Med. Chem., 1999,
42, 4705.
Agents
[57]
Ganesan, A. Drug Discov. Today, 2002, 7, 47.
[58]
Rich, J.O.; Michels, P.C.; Khmelnitsky, Y.L. Current
Opinion in Chemical Biology, 2002, 6, 161.
[59]
van Dongen, M.; Weigelt, J.; Uppenberg, J.; Schultz, J.;
Wikstrom, M. Drug Discov. Today, 2002, 7, 471.
[60]
Carr, R.; Jhoti, H. Drug Discov. Today, 2002, 7, 522.
[80]
[61]
Goksoyr, T.V.J.; Daae, F.L. Appl. Environ. Microbiol.,
1990, 56, 782.
Betts, J.C.; Lukey, P.T.; Robb, L.C.; McAdam, R.A.;
Duncan, K. Mol. Microbiol., 2002, 43, 717.
[81]
Salama, N.; Guillemin, K.; McDaniel, T.K.; Sherlock, G.;
Tompkins, L.; Falkow, S. Proc. Natl. Acad. Sci. USA,
2000, 97, 14668.
[82]
Whiteley, M.; Bangera, M.G.; Bumgarner, R.E.; Parsek,
M.R.; Teitzel, G.M.; Lory, S.; Greenberg, E.P. Nature,
2001, 413, 860.
[83]
de Saizieu, A.; Certa, U.; Warrington, J.; Gray, C.; Keck,
W.; Mous, J. Nat. Biotechnol., 1998, 16, 45.
[84]
de Saizieu, A.; Gardes, C.; Flint, N.; Wagner, C.; Kamber,
M.; Mitchell, T.J.; Keck, W.; Amrein, K.E.; Lange, R. J.
Bacteriol., 2000, 182, 4696.
[85]
Dunman, P.M.; Murphy, E.; Haney, S.; Palacios, D.;
Tucker-Kellogg, G.; Wu, S.; Brown, E.L.; Zagursky, R.J.;
Shlaes, D.; Projan, S.J. J. Bacteriol., 2001, 183 , 7341.
[62]
Yap, W.H.; Li, X.; Soong, T.W.; Davies, J.E. J. Ind.
Microbiol., 1996, 17, 179.
[63]
Wang, G.Y.; Graziani, E.; Waters, B.; Pan, W.; Li, X.;
McDermott, J.; Meurer, G.; Saxena, G.; Andersen, R.J.;
Davies, J. Org. Lett., 2000, 2, 2401.
[64]
Kao, C.M.; Katz, L.; Khosla, C. Science, 1994, 265, 509.
[65]
Gokhale, R.S.; Tsuji, S.Y.; Cane, D.E.; Khosla, C. Science,
1999, 284, 482.
[66]
Bentley, S.D.; Chater, K.F.; Cerdeno-Tarraga, A.M.;
Challis, G.L.; Thomson, N.R.; James, K.D.; Harris, D.E.;
Quail, M.A.; Kieser, H.; Harper, D.; Bateman, A.; Brown,
S.; Chandra, G.; Chen, C.W.; Collins, M.; Cronin, A.;
Fraser, A.; Goble, A.; Hidalgo, J.; Hornsby, T.; Howarth,
308
[86]
Current Drug Targets - Infectious Disorders, 2002, Vol. 2, No. 4
Fitzgerald, J.R.; Sturdevant, D.E.; Mackie, S.M.; Gill,
S.R.; Musser, J.M. Proc. Natl. Acad. Sci. USA, 2001, 98,
8821.
Holmes et al.
[87]
Cummings, C.A.; Relman, D.A. Emerg. Infect. Dis., 2000,
6, 513.
[88]
Kato-Maeda, M.; Gao, Q.; Small, P.M. Cell Microbiol.,
2001, 3, 713.