chemical biology applied to the study of bacterial pathogens · chemical biology applied to the...

14
Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics & Molecular Biology of Intestinal Pathogens, Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA In recent years, chemical biology and chemical genomics have been increasingly applied to the field of microbiology to uncover new potential therapeutics as well as to probe virulence mechanisms in pathogens. The approach offers some clear advantages, as identified compounds (i) can serve as a proof of principle for the applicability of drugs to specific targets; (ii) can serve as condi- tional effectors to explore the function of their targets in vitro and in vivo; (iii) can be used to modulate gene expression in oth- erwise genetically intractable organisms; and (iv) can be tailored to a narrow or broad range of bacteria. This review highlights recent examples from the literature to illustrate how the use of small molecules has advanced discovery of novel potential treat- ments and has been applied to explore biological mechanisms underlying pathogenicity. We also use these examples to discuss practical considerations that are key to establishing a screening or discovery program. Finally, we discuss the advantages and challenges of different approaches and the methods that are emerging to address these challenges. R esearchers have taken increasing interest in small-molecule screening programs because of their dual use in discovering new potential therapeutics and in generating molecular probes useful for studying virulence pathways in vitro and in vivo. The fields of drug discovery and microbial biology have both benefited from findings made using small-molecule screening programs. The Centers for Disease Control and Prevention recently esti- mated that over 2,000,000 illnesses are caused by antibiotic-resis- tant bacteria and fungi annually, resulting in over 23,000 deaths, though this is likely an underestimate (1). Despite the huge initial success of antibiotic therapy, overuse and misuse have led to their increasingly limited effectiveness, and today, at least one resistant strain of bacteria exists for every known antibiotic (2). Equally troubling is that antibiotic therapy kills a large portion of the host microbiota in addition to the targeted pathogen. The resulting dysbiosis can lead to acute and chronic intestinal problems (3, 4) and is a leading cause of hospital-acquired infections by Clostrid- ium difficile (5). As more and more pathogens become resistant to antibiotics, and with increasing awareness of the protective effects of the microbiota, researchers have started to look for alternative therapies for treating bacterial infections. Anti-infective drugs, also known as antivirulence drugs, are attractive alternatives because they disarm pathogens rather than killing them, providing significant advantages over antibiotic treatment. First, resistance developed against anti-infective drugs may be driven by a weaker selective pressure; thus, resistance would take longer to develop, if it develops at all. Second, by tar- geting a virulence trait, anti-infectives affect only the bacteria that possess that pathogenic trait—ideally leaving the microbiota rel- atively unaffected. In addition to their use in drug discovery, small molecules have played important roles in biomedical research because of their use in uncovering new virulence requirements. There are several ad- vantages to probing pathogenesis with small molecules at the bench: (i) they act quickly, (ii) they may act reversibly, (iii) they do not require manipulation of the genome, a quality that is espe- cially advantageous in studying genetically intractable organisms, (iv) the dose can be adjusted to fine-tune the effects, and (v) they can be used across multiple bacterial species to determine how conserved a pathway is between different species or strains. Pathogenic bacteria have evolved numerous strategies for es- tablishing infection and causing disease in various hosts. For any given pathogen, there are multiple points in the infection process that can be inhibited to reduce virulence potential. Figure 1 de- picts small-molecule inhibitors (shown in red) discussed in this review in a generalized model of pathogenesis. Some of these vir- ulence mechanisms are restricted to only one or a few organisms (for example, actin-mediated cell-to-cell spread is used promi- nently by Listeria monocytogenes, Shigella spp., and Rickettsia spp.), while others are more broadly conserved. Such an “Achilles’ heel” of pathogenesis can be exploited to develop new treatment therapies. We begin this review with a brief introduction of the screening process and then discuss examples of successful screens (summarized in Table 1) to demonstrate how they have influ- enced our understanding of pathogenesis and uncovered new po- tential drug targets. We explore the advantages and limitations of small-molecule research and conclude with some ways in which small-molecule screens could further our understanding of pathogenesis. THE SCREENING PROCESS To understand the impact of using small molecules to study and target bacterial pathogens and pathogenicity, it is important to appreciate the process involved in their identification. A brief comment is warranted regarding the sort of chemical libraries that may be used in the basic screening process outlined in Fig. 2.A diverse library enables researchers to probe more chemical space, increasing the likelihood of finding a novel inhibitor, but less is Accepted manuscript posted online 17 November 2014 Citation Anthouard R, DiRita VJ. 2015. Chemical biology applied to the study of bacterial pathogens. Infect Immun 83:456 – 469. doi:10.1128/IAI.02021-14. Editor: H. L. Andrews-Polymenis Address correspondence to Victor J. DiRita, [email protected]. * Present address: Rebecca Anthouard, Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. Copyright © 2015, American Society for Microbiology. All Rights Reserved. doi:10.1128/IAI.02021-14 MINIREVIEW 456 iai.asm.org February 2015 Volume 83 Number 2 Infection and Immunity on May 22, 2020 by guest http://iai.asm.org/ Downloaded from

Upload: others

Post on 22-May-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

Chemical Biology Applied to the Study of Bacterial Pathogens

Rebecca Anthouard,* Victor J. DiRita

Laboratory of Genetics & Molecular Biology of Intestinal Pathogens, Department of Microbiology & Immunology, University of Michigan Medical School, AnnArbor, Michigan, USA

In recent years, chemical biology and chemical genomics have been increasingly applied to the field of microbiology to uncovernew potential therapeutics as well as to probe virulence mechanisms in pathogens. The approach offers some clear advantages, asidentified compounds (i) can serve as a proof of principle for the applicability of drugs to specific targets; (ii) can serve as condi-tional effectors to explore the function of their targets in vitro and in vivo; (iii) can be used to modulate gene expression in oth-erwise genetically intractable organisms; and (iv) can be tailored to a narrow or broad range of bacteria. This review highlightsrecent examples from the literature to illustrate how the use of small molecules has advanced discovery of novel potential treat-ments and has been applied to explore biological mechanisms underlying pathogenicity. We also use these examples to discusspractical considerations that are key to establishing a screening or discovery program. Finally, we discuss the advantages andchallenges of different approaches and the methods that are emerging to address these challenges.

Researchers have taken increasing interest in small-moleculescreening programs because of their dual use in discovering

new potential therapeutics and in generating molecular probesuseful for studying virulence pathways in vitro and in vivo. Thefields of drug discovery and microbial biology have both benefitedfrom findings made using small-molecule screening programs.

The Centers for Disease Control and Prevention recently esti-mated that over 2,000,000 illnesses are caused by antibiotic-resis-tant bacteria and fungi annually, resulting in over 23,000 deaths,though this is likely an underestimate (1). Despite the huge initialsuccess of antibiotic therapy, overuse and misuse have led to theirincreasingly limited effectiveness, and today, at least one resistantstrain of bacteria exists for every known antibiotic (2). Equallytroubling is that antibiotic therapy kills a large portion of the hostmicrobiota in addition to the targeted pathogen. The resultingdysbiosis can lead to acute and chronic intestinal problems (3, 4)and is a leading cause of hospital-acquired infections by Clostrid-ium difficile (5). As more and more pathogens become resistant toantibiotics, and with increasing awareness of the protective effectsof the microbiota, researchers have started to look for alternativetherapies for treating bacterial infections.

Anti-infective drugs, also known as antivirulence drugs, areattractive alternatives because they disarm pathogens rather thankilling them, providing significant advantages over antibiotictreatment. First, resistance developed against anti-infective drugsmay be driven by a weaker selective pressure; thus, resistancewould take longer to develop, if it develops at all. Second, by tar-geting a virulence trait, anti-infectives affect only the bacteria thatpossess that pathogenic trait—ideally leaving the microbiota rel-atively unaffected.

In addition to their use in drug discovery, small molecules haveplayed important roles in biomedical research because of their usein uncovering new virulence requirements. There are several ad-vantages to probing pathogenesis with small molecules at thebench: (i) they act quickly, (ii) they may act reversibly, (iii) they donot require manipulation of the genome, a quality that is espe-cially advantageous in studying genetically intractable organisms,(iv) the dose can be adjusted to fine-tune the effects, and (v) theycan be used across multiple bacterial species to determine howconserved a pathway is between different species or strains.

Pathogenic bacteria have evolved numerous strategies for es-tablishing infection and causing disease in various hosts. For anygiven pathogen, there are multiple points in the infection processthat can be inhibited to reduce virulence potential. Figure 1 de-picts small-molecule inhibitors (shown in red) discussed in thisreview in a generalized model of pathogenesis. Some of these vir-ulence mechanisms are restricted to only one or a few organisms(for example, actin-mediated cell-to-cell spread is used promi-nently by Listeria monocytogenes, Shigella spp., and Rickettsiaspp.), while others are more broadly conserved. Such an “Achilles’heel” of pathogenesis can be exploited to develop new treatmenttherapies. We begin this review with a brief introduction of thescreening process and then discuss examples of successful screens(summarized in Table 1) to demonstrate how they have influ-enced our understanding of pathogenesis and uncovered new po-tential drug targets. We explore the advantages and limitations ofsmall-molecule research and conclude with some ways in whichsmall-molecule screens could further our understanding ofpathogenesis.

THE SCREENING PROCESS

To understand the impact of using small molecules to study andtarget bacterial pathogens and pathogenicity, it is important toappreciate the process involved in their identification. A briefcomment is warranted regarding the sort of chemical libraries thatmay be used in the basic screening process outlined in Fig. 2. Adiverse library enables researchers to probe more chemical space,increasing the likelihood of finding a novel inhibitor, but less is

Accepted manuscript posted online 17 November 2014

Citation Anthouard R, DiRita VJ. 2015. Chemical biology applied to the study ofbacterial pathogens. Infect Immun 83:456 – 469. doi:10.1128/IAI.02021-14.

Editor: H. L. Andrews-Polymenis

Address correspondence to Victor J. DiRita, [email protected].

* Present address: Rebecca Anthouard, Department of Microbiology &Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NorthCarolina, USA.

Copyright © 2015, American Society for Microbiology. All Rights Reserved.

doi:10.1128/IAI.02021-14

MINIREVIEW

456 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 2: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

known regarding the molecules in these libraries. Some of theexamples we provide below arose through screening diverse li-braries. Probing more-defined libraries of molecules with knownbiological function, on the other hand, has the advantage that themechanism of action for these molecules has been previously de-scribed in at least some settings. However, such libraries lack thebroad chemical space of the diverse libraries. We also providesome examples where these more-defined libraries have beenscreened successfully.

For screening and postscreening evaluation, the process (out-lined in Fig. 2) is essentially as follows. First, a robust assay isdeveloped. Second, a series of high-throughput screens are per-formed to identify small-molecule inhibitors. A primary screen isperformed with selected libraries of compounds to identify thosethat affect the assay. A secondary screen is performed to confirmgenuine hits and determine whether they behave in a dose-depen-dent manner. A counterscreen may also be performed, in parallelto or subsequent to the primary and secondary screens, to provideadditional information for triage of hit compounds. Counter-screens may, for example, report on the specificity or general tox-icity of a compound. After triage of hits from the screen, a smallmolecule is selected for further characterization. There are manyways to characterize a small molecule, and the choice largely de-pends on the aim of the study. If, for example, the principal goal isto develop a novel therapeutic, emphasis will be placed on opti-mizing in vivo efficacy. However, if the compound is primarilyintended as a biological probe, determining its target and mecha-

nism of action will be prioritized. Several approaches to com-pound characterization are often performed in parallel becauseinformation gleaned from one approach can impact the design orinterpretation of data obtained by other approaches. As we discusssmall molecules that target bacterial pathogenesis in this review,we also point out how different strategies used in the assay, screen-ing, and characterization steps affect the type of small moleculeidentified.

SMALL MOLECULES THAT AFFECT VIRULENCE FACTORSAND THEIR REGULATION

Many chemical screens have targeted bacterial virulence factors ortheir regulation, an approach that boasts numerous advantages.From a drug discovery standpoint, antivirulence compounds havethe advantages that (i) they presumably would have no (or per-haps a very limited) effect on the host microbiota in comparisonto traditional antibiotics, which act more indiscriminately, and(ii) they disarm the pathogen rather than kill it, theoretically im-posing a lower selective pressure with respect to resistance thantraditional antibiotics, reducing the risk for the emergence of re-sistant strains. Also, targeting virulence factors enables drugging arange of species, depending on how conserved the target is. Fi-nally, antivirulence molecules are valuable as research tools, asthey deepen our understanding of the molecular mechanisms re-quired for virulence gene activation.

Virstatin. One early example of such a molecule is virstatin(see Table 1). Virstatin targets Vibrio cholerae specifically by inhib-

FIG 1 Critical steps in the pathogenicity of microbes and the small molecules that inhibit each step. Inhibitors discussed in this review (shown in red) target manyaspects of pathogenesis (shown in black), including specific virulence factors and their regulation (steps 1 to 5, 10, and 13) and broader aspects of host-pathogeninteractions (steps 6 to 9, 11 to 12, and 14).

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 457Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 3: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

TA

BLE

1N

ames

,str

uct

ure

s,an

dpe

rtin

ent

info

rmat

ion

rega

rdin

gco

mpo

un

dsdi

scu

ssed

inth

isre

view

a

Com

pou

nd(

s)St

ruct

ure

Ass

ay

No.

ofco

mpo

un

dssc

reen

edor

desc

ript

ion

IC5

0(�

M)

Tar

get

MO

AR

efer

ence

(s)

Org

anis

mra

nge

CC

CP

Cl

H NN H

N

N

Live

-cel

l,Le

pA-B

laM

repo

rter

assa

y48

0�

1T

4SS

tran

sloc

atio

n

Kn

own

toaf

fect

the

prot

onm

otiv

efo

rce

(PM

F)6

Legi

onel

lapn

eum

ophi

la

CC

G-2

979

N

N

S

H3C

H2C

O

Live

-cel

l,sk

a-ka

nre

port

er�

55,0

00�

31St

rept

okin

ase

prom

oter

Un

know

n7-

8G

rou

pA

stre

ptoc

occu

s

FPSS

F

H N

S

O

O

Live

-cel

l,op

uCA

-gus

repo

rter

�57

,000

3.0–

3.5

�B

Inh

ibit

sR

sbV

phos

phor

ylat

ion

,fr

eein

gR

sbW

tose

ques

ter

�B

9,10

List

eria

mon

ocyt

ogen

es,

Bac

illus

subt

ilis

LED

209

N HN H

SS

N HOO

Live

-cel

l,LE

E1-

lacZ

repo

rter

�15

0,00

0�

10Q

seC

Inh

ibit

ion

ofQ

seC

auto

phos

phor

ylat

ion

11Sa

lmon

ella

Typ

him

uri

um

,Fr

anci

sella

tula

rens

is

Pili

cide

sN

S

R1

R2

R3

OC

O2

Li

Rat

ion

alde

sign

usi

ng

Pap

D-P

apG

stru

ctu

re

Des

ign

edas

Pap

Gm

imet

ics

Var

ies

Pilu

sbi

ogen

esis

Inte

rfer

esw

ith

the

chap

eron

e-u

sher

inte

ract

ion

12,1

3E

sche

rich

iaco

li

Pim

ozid

e

F

N

N

NH

F

O

Inte

rnal

izat

ion

ofG

FP-

labe

led

bact

eria

asa

read

out

for

infe

ctio

n48

0N

otre

port

ed

L.m

onoc

ytog

enes

infe

ctio

nof

bon

em

arro

w-

deri

ved

mac

roph

ages

Un

know

n14

L.m

onoc

ytog

enes

RW

J-60

475

Br

O

NH

O

O–

PH

OO

H

O

HO

Live

-cel

l,Le

pA-B

laM

repo

rter

assa

y48

0N

otre

port

edT

4SS

tran

sloc

atio

n

Kn

own

toin

hib

itth

ere

cept

orty

rosi

ne

phos

phat

eph

osph

atas

eC

D45

6L.

pneu

mop

hila

Minireview

458 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 4: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

Tox

tazi

nA

OH

3C

N

N

F

FF

SC

H3

O

O

Live

-cel

l,to

xT-g

fpre

port

er�

63,0

0024

.5to

xT tran

scri

ptio

nU

nkn

own

15V

.cho

lera

e

Tox

tazi

nB

Br

N

HN

N

O

N

OH

3C

Live

-cel

l,to

xT-g

fpre

port

er�

63,0

002.

7tc

pP tran

scri

ptio

nU

nkn

own

15V

.cho

lera

e

TT

S29

N

SN

OH

O

CH

3

O

CH

3

O

Live

cell,

phos

phol

ipas

ese

cret

ion

�92

,000

Not

repo

rted

T3S

Sse

cret

ion

Inh

ibit

sfo

rmat

ion

orst

abili

tyof

the

T3S

Sn

eedl

eco

mpl

ex

16

Yer

sini

aen

tero

colit

ica,

Salm

onel

laT

yph

imu

riu

m,

Pse

udom

onas

aeru

gino

sa,

Fran

cise

llano

vici

da,

Pse

udom

onas

syri

ngae

pv.t

omat

o

Vir

stat

inNO O

OH

O

Liv

ece

ll,fl

uor

esce

nce

�50

,000

3–40

,dep

endi

ng

onst

rain

Tox

TIn

hib

its

Tox

Tdi

mer

izat

ion

17,1

8,19

V.c

hole

rae

aIC

50,5

0%in

hib

itor

yco

nce

ntr

atio

n;M

OA

,mod

eof

acti

on.

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 459Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 5: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

iting its ability to produce cholera toxin (CT) (step 1 in Fig. 1)(17). Virstatin was identified using a live-cell-based reporter assayin which the promoter for the genes encoding cholera toxin(ctxAB) directed expression of the tetracycline resistance genetetA. In this screen, compounds that inhibited expression of thecholera toxin genes resulted in poor growth of the reporter strainin media containing tetracycline. Such cell-based assays are par-ticularly useful to basic biology because the identified compoundsmay inhibit a protein or pathway not yet known to play a role invirulence, thus allowing new discoveries about biological systemsto be made.

In contrast to screens against purified proteins, a cell-basedscreen requires postscreen target identification. This process canbe extremely challenging because the target (i) may have an un-specified role in virulence, (ii) may be expressed only under cer-tain conditions, and/or (iii) may not be a single protein but mayrather be a protein complex or (iv) not a protein at all but ratherDNA, RNA, lipids, or the redox state of the cell, to name a few ofthe possibilities.

In the case of virstatin, target identification was accomplishedby taking advantage of the fact that the virulence regulatory path-way in V. cholerae is well defined (20–26). Expression profiles of

genes expressed upstream of ctxAB transcription in the regulatorycascade were examined by quantitative reverse transcription-PCR(qRT-PCR), and none was affected by virstatin, suggesting that itstarget is late in the transcription cascade leading to ctxAB expres-sion. Virstatin could still inhibit ctxAB transcription even whentoxT, encoding the direct activator of ctxAB transcription, wasexpressed ectopically, indicating that virstatin inhibits ToxT ac-tivity (17).

To examine further whether ToxT is the likely target of virsta-tin, a library of toxT mutant alleles was generated and screened forresistance to the compound; one allele, toxTL113P, was identified(17). The protein expressed from this mutant allele resembledwild-type ToxT in that it was found predominantly in the multi-meric form; however, in the presence of virstatin, wild-type ToxTis largely in the monomeric state, while ToxTL113P remains pre-dominantly multimeric (18, 19). This supported a long-held hy-pothesis, based on the arrangement of the binding sites on ToxT-activated promoters known as toxboxes (27), that ToxT is active asa dimer. Virstatin was then used to probe ToxT activity at otherToxT-dependent promoters, which led to the discovery that somepromoters favor the monomeric form whereas others favordimerized ToxT (28).

FIG 2 Identifying and characterizing small-molecule inhibitors of pathogenesis require many considerations at each step of the process. The major strategies areoutlined here and are discussed further in the text.

Minireview

460 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 6: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

To assess its in vivo efficacy, virstatin was tested in an infantmouse model of V. cholerae colonization (17). Mice inoculatedwith wild-type V. cholerae and given virstatin had a 4-log decreasein colonization relative to mice given the dimethyl sulfoxide(DMSO) control, suggesting that virstatin may have therapeuticpotential. To determine whether the decreased colonization wasToxT dependent, an atypical strain of V. cholerae that colonizesthe mouse in a ToxT-independent manner was used. This straincolonized mice treated with the DMSO control and mice treatedwith virstatin equally well, indicating that virstatin reduces colo-nization of the typical endemic strains of V. cholerae by inhibitingToxT activity in vivo. Although it remains to be directly tested, thatcolonization by ToxT-independent strains of V. cholerae is notaffected by virstatin suggests that other bacteria lacking ToxT,including the host microbiota, would be unaffected by virstatintreatment, making virstatin an attractive therapeutic lead.

The cell-based screening approach leading to the discovery ofvirstatin and the subsequent work characterizing its mechanism ofaction advanced the field of V. cholerae research in several ways.First, this approach provided further strong evidence for the im-portance of the dimerization state of ToxT. Second, it proveduseful for determining whether a V. cholerae isolate colonizes in aToxT-dependent or -independent manner. Finally, as virstatinwas shown to be a compound that significantly reduces coloniza-tion of a pathogen by specifically inhibiting its virulence factors invivo, it provided a proof of principle for the concept of antiviru-lence drugs.

Toxtazins. The toxtazins (step 1 in Fig. 1) constitute anotherclass of antivirulence compounds that inhibit expression of viru-lence factors in V. cholerae (15). Toxtazins A and B were identifiedin a cell-based screen using a toxT-gfp reporter strain to probe theToxT pathway directly. As indicated in Fig. 2, assays can be de-signed to target a phenotype or a molecular pathway, and eachapproach has unique advantages. Targeting a pathway restrictshits to those that affect only that pathway, while targeting a phe-notype, CT production, for example, would include hits that tar-get the ToxT pathway as well as inhibitors of CT folding and se-cretion.

The toxtazins significantly reduce expression of both choleratoxin and the toxin-coregulated pilus (15). Furthermore, toxtazinB reduced the level of V. cholerae colonization of infant mice by 2logs, suggesting that it may have therapeutic potential in the treat-ment or prevention of cholera. Further analysis demonstrated thattoxtazin A does not affect expression of ToxR or TcpP, the tran-scriptional activators of toxT, while toxtazin B reduced expressionof both tcpP transcript and TcpP protein levels. TcpP is transcrip-tionally activated by AphA and AphB, neither of which is affectedby toxtazin B treatment. In fact, ectopic expression of ToxT in cellstreated with either toxtazin A or B restored expression of choleratoxin, confirming that both of these compounds act upstream ofToxT expression. From these results, it was determined that tox-tazin A targets toxT transcription whereas toxtazin B targets tcpPtranscription (15).

The exact mechanisms of action for these compounds havenot been identified, but preliminary evidence showing that cellstreated with toxtazin A mount a stress response indicates the pres-ence of a pathway which was not previously known to influenceregulation of toxT expression but which affects the virulence po-tential of V. cholerae.

FPSS. Unlike virstatin and the toxtazins, which target species-

specific virulence regulators, fluoro-phenyl-styrene-sulfonamide(FPSS; see Table 1) targets a general microbial regulatory path-way—the sigma B (�B) regulon (step 2 in Fig. 1) (9). Sigma factorsare dissociable subunits of RNA polymerase (RNAP) that associ-ate with RNAP in response to certain environmental signals anddirectly activate or repress subsets of genes, resulting in a rapidchange in global transcription that is appropriate for the givenenvironment. Different sigma factors respond to different sets ofenvironmental signals, allowing bacteria to quickly respond tospecific environments.

Different sigma factors can respond to the same signal, anddifferent sigma factors can regulate the same genes in response todistinct signals, creating a complex web of regulation (29). In ad-dition, some sigma factors are essential in some or all growthconditions, making them difficult to study using classical geneticapproaches. Small-molecule inhibitors of specific sigma factorsenable the study of individual sigma factors in isolation, so thecontributions of individual sigma factors and of different environ-mental signals can be explored. This class of inhibitor may alsohave therapeutic potential because sigma factors regulate viru-lence gene expression in many pathogens (30–32), as reviewed inreference 33.

In Listeria monocytogenes, �B responds to environmental stress(i.e., acidic conditions, ethanol, or high salt concentrations), en-ergy stress, and growth at low temperatures (34). In response tothese signals, �B activates or represses genes involved in centralmetabolism and activates PrfA, the master regulator of virulence(35). FPSS was identified as an inhibitor of �B by screening adiverse library of synthetic compounds using a cell-based reporterassay (9).

FPSS was discovered using a screening strain containing a �B-dependent promoter, opuCA, fused to the gene for glucuronidase,gus, such that glucuronidase activity could be used as a readout for�B activity. Inhibition of �B activity by FPSS was confirmed byqRT-PCR. Transcription of �B-dependent promoters was at thelevel of a �B mutant after FPSS treatment, indicating that FPSScompletely inhibits �B activity at these promoters. Microarrayanalysis showed that L. monocytogenes cells treated with FPSS phe-nocopy an L. monocytogenes �B mutant, affecting 91% of previ-ously defined �B-regulated genes (9). FPSS also affected 83 othergenes, which, excluding side effects, could potentially expand the�B regulon. Gene-set enrichment analysis (GSEA) determinedthat genes specifically regulated by �H or by �L are not signifi-cantly enriched among genes differentially transcribed in FPSS-treated cells, indicating that FPSS inhibits �B specifically.

Bacillus subtilis was used to determine the mechanism by whichFPSS inhibits �B activity, because its �B activity is also inhibitedby FPSS (9) and the components of its well-characterized �B regu-lon are highly conserved with the components of the insufficientlystudied �B regulon of L. monocytogenes. In B. subtilis, �B activity isregulated by three distinct branches (shown in Fig. 3 and reviewedin reference 36) that convey conditions of environmental stress(i.e., acidic conditions, ethanol, or high salt concentrations), en-ergy stress (i.e., limitation of glucose, ATP, GTP, or phosphate),and growth at low temperatures.

In B. subtilis, �B is kept in the off state by an anti-sigma factor,RsbW. The ability of RsbW to sequester �B is controlled by thephosphorylation state of another protein, RsbV; phosphorylatedRsbV cannot bind RsbW, leaving RsbW free to sequester �B,thereby keeping the regulon from being expressed. In contrast,

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 461Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 7: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

unphosphorylated RsbV binds RsbW, liberating �B and leading toactivation of the �B regulon (37). RsbV phosphorylation is con-trolled by energy and environmental stresses (see Fig. 3) (38), andFPSS inhibited �B activation by several such stresses, indicatingthat it works on a factor common to these pathways (9). Thepossibility that FPSS could bind or interact with �B was ruled outby in vitro and in vivo experiments (10). Taken together, theseresults support a model whereby FPSS targets the partner-switch-ing mechanism between RNA polymerase, �B, and its anti-sigmafactor, RsbW.

FPSS provides a unique tool to study �B activity in multipleorganisms. Microarray analysis of FPSS-treated cultures (9) sug-gests that the �B regulon is broader than currently understood,showing another value of using this compound to study �B. Whileanimal studies with FPSS have not been reported, �B is an attrac-tive therapeutic target because it is conserved and important in theactivation of virulence genes in several pathogenic bacteria (39,40). One potential drawback to the therapeutic potential of thismolecule, however, is that FPSS may also inhibit �B in commensalstrains of the microbiota, which could lead to dysbiosis, althoughthis has not been experimentally assessed.

TSS29. Secretion mechanisms have been targeted in small-molecule screens for antivirulence drugs. In particular, the type 3secretion system (T3SS) injectosome has been attractive becauseof its broad conservation in Gram-negative pathogens (step 3 inFig. 1). Inhibitors of the Salmonella enterica serovar TyphimuriumT3SS were identified using a reporter strain that secretes phospho-lipase in a T3SS-dependent manner (16). This phenotypic ap-proach to cell-based screening can result in hits that affect any of

several steps in production of a functional phospholipase, includ-ing transcription, translation, protein folding, protein-protein in-teraction, and translocation. Eighty-nine compounds remainedafter the primary and secondary screens. Tertiary screens aresometimes performed to select compounds with particular char-acteristics (Fig. 2). In that study, the authors performed two ter-tiary screens to eliminate compounds that affect bacterial growthand those that are not specific (i.e., that inhibit bacterial transla-tion, sec-dependent secretion, or disulfide bond isomerization).Seven compounds passed the tertiary screens, and one of these, a2-imino-5-arylidene thiazolidinone termed TTS29, was investi-gated further (16). Cultures grown with TTS29 displayed an over-all reduction in the levels of type 3 secretion system needle com-plexes. Levels of needle complex protein constituents were notreduced in whole-cell lysates, indicating that the proteins werebeing produced but that their assembly into the needle complexwas inhibited by TTS29 (16).

Because components of the T3SS are conserved in other bac-teria, TTS29 has the potential to work against other T3S-encodingbacteria. Yersinia species express two types of T3SS: (i) the plas-mid-encoded Ysc system in Yersinia pestis, Yersinia enterocolitica,and Yersinia pseudotuberculosis, which secretes Yop (Yersiniaouter protein) into the cytosol of target cells, and (ii) the chromo-somally encoded Ysa system in Y. enterocolitica, which secrets Ysp(Yersinia secreted protein) (41, 42). TSS29 inhibited secretion ofboth Yop and Ysp into Y. enterocolitica culture supernatants, in-dicating its potential utility as a broad inhibitor of T3SS (16). Incontrast, TSS29 did not alter flagellar motility or decrease the lev-els of flagellar components in either S. Typhimurium or Pseu-

FIG 3 FPSS targets sigma B. Multiple stresses activate �B activity in B. subtilis. Energy stress is relayed via RsbPQ, environmental stress is relayed via the“stressosome” and RsbU, and low-temperature stress is relayed via an as-yet-unknown mechanism. Sensing of stress results in dephosphorylation of the RsbVanti-anti-sigma factor, allowing it to bind the RsbW anti-sigma factor, which in turn releases �B, to interact with RNAP and activate the �B regulon. Phosphor-ylated RsbV interacts poorly with RsbW, which is then free to bind �B, leading to low-level expression of the regulon. FPSS inhibits �B activity by drivingequilibrium toward the state in which �B is bound to RsbW. SNP, single nucleotide polymorphism.

Minireview

462 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 8: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

domonas aeruginosa, bacteria that depend on a flagellum-specificT3SS for motility (16). This suggests that TSS29 targets a compo-nent of the T3SS that is not conserved with the evolutionarilyrelated flagellum-specific T3SS (42).

Because one component of the T3SS, secretin, is conserved inthe type 2 secretion system (T2SS) that delivers enzymes and otherproteins across the Gram-negative envelope (43), TSS29 wastested for its ability to inhibit such systems. Secretion of elastase(44) by the T2SS in P. aeruginosa was inhibited by TSS29, as wastwitching motility, which is controlled by a pilus whose assemblyincludes components similar to those of the T2SS (16). Theseresults demonstrate that TSS29 is a broad inhibitor of secretionaffecting different secretion systems in multiple bacterial patho-gens.

The assessment of the in vivo effectiveness of TSS29 was sup-ported by demonstrating its ability to reduce killing of bone mar-row-derived macrophages (BMMs) in a tissue culture model ofinfection (16). It also inhibited Pseudomonas syringae pv. tomatoDC3000 from inducing a hypersensitivity response in tobaccoplants (16). Thus, TSS29 has broad therapeutic potential.

Pilicides. Pili are recognized as important virulence factorsbecause of their role in colonization of many pathogens in their

respective hosts (step 4 in Fig. 1). While some microbes secretepilus adherence structures through type 2-like secretion systemsthat can be inhibited by TSS29 as described above, others assemblepili using pathways that rely on a periplasmic chaperone (45–47)and an outer membrane usher (48, 49) (see references 50 and 51for reviews). The chaperone mediates the folding, stabilization,and transport of pilus subunits, while the usher aids in incorpo-rating subunits into the growing pilus (51).

Because the chaperone-usher pathway of pilus assembly is con-served in a wide range of pathogens (51), inhibitors of these sys-tems would theoretically be effective against a broad range of bac-terial species, making them attractive targets for therapeuticdevelopment. Substituted bicyclic 2-pyridones, called pilicides(pharmacophore shown in Table 1), are a well-studied group ofsynthetic small-molecule inhibitors that prevent formation of piliin uropathogenic Escherichia coli (UPEC) (51).

UPEC bacteria produce different pathogenicity-associated pili,the most representative of which are P pili and type I pili; theseadhesion structures and others like them are generally termedchaperone-usher pili for the key proteins associated with theirassembly (Fig. 4) (51). P pili are made up of subunits called PapA(the major subunit), PapE, PapF, PapG, PapH, and PapK. These

FIG 4 Pilicides affect both P pili and the type 1 pili, which have similar structures. Pili consist of several repeating subunits arranged in a helical structure.Subunits are translocated from the cytoplasm to the periplasm, where a chaperone (PapD or FimC) folds the protein, stabilizes it, and transfers it to the usherprotein (PapC or FimD), which secretes the protein and incorporates it into the pilus structure. Pilicides inhibit formation of pili by preventing the chaperonefrom passing the subunit to the usher protein.

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 463Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 9: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

are assembled by the PapD chaperone and the usher PapC. Type 1pili are made up of FimA (the major subunit), FimG, and FimHand are assembled by the chaperone FimC and the usher FimD(51).

In contrast to inhibitors discovered by screening compoundlibraries, pilicides were developed using a rational design ap-proach (12). Pilus biogenesis requires the chaperone protein tobind its natural ligands (52), so the solved crystal structure of thePapD-PapG complex was used to chemically design molecularmimetics that would bind within the active site of periplasmicchaperones PapD and FimC (12). A library of pilicides was syn-thesized and demonstrated to be effective at inhibiting pilus for-mation (13).

PiIlicides do not affect cell growth or viability but inhibit man-nose-sensitive hemagglutination (MSHA) and hemagglutination(HA), traits of type 1 pili and P pili, respectively (13). Pilicides alsoreduce biofilm formation and bacterial attachment to host cells,both of which require type 1 pili (13). Pilicide 2c in particularreduced the ability of E. coli strains to adhere to cultured bladdercells by 90% (13).

Cocrystallization of pilicide 2c with the PapD chaperone (13)demonstrated close contact with a hydrophobic patch that runsacross the back of the F1-C1-D1 beta sheet on PapD, a regionhighly conserved in all pilus periplasmic chaperones, which maymediate interactions between the chaperone and the N-terminaldomain of the usher protein (13, 53). Surface plasmon resonanceshowed that pilicide 2c also prevents FimC-FimH (chaperone-adhesin) from binding to FimD (usher) (13), suggesting that pili-cide 2c inhibits the biogenesis of pili by preventing the chaperonefrom passing the subunit to the usher.

Studies of pilicides have uncovered knowledge about molecu-lar mechanisms involved in the biogenesis of pili, and their dis-covery and characterization demonstrate the power of crystallog-raphy to guide chemists to create a small molecule with apredictable activity. This technique, called structure-based drugdesign (SBDD), can be extended by cocrystallizing the compoundand target to provide information useful for improving com-pound activity.

SMALL MOLECULES THAT AFFECT HOST-PATHOGENINTERACTIONS

Small molecules can provide valuable insight into underlying bi-ological mechanisms in pathogenicity and a relevant system forstudying pathogens in the context of their hosts. While the previ-ously discussed compounds targeted the pathogen, the followingexamples are of small molecules that work by disrupting impor-tant host-pathogen interactions and demonstrate that this is alsoan effective strategy for generating compounds with therapeuticpotential.

LED209. Virulence gene expression is often induced by signalsfrom the host environment, making sensory transduction mech-anisms potential therapeutic targets (step 5 in Fig. 1). For exam-ple, E. coli, S. Typhimurium, and Francisella tularensis express asensor histidine kinase called QseC that detects host-derived ad-renergic signals (epinephrine and norepinephrine) as well as quo-rum-sensing autoinducer-3 (AI-3) (54, 55). In response to eitherof these signals, QseC autophosphorylates and then transfers thephosphate to transcription factor QseB, leading to transcription ofkey virulence genes, including the LEE1 operon in enterohemor-rhagic E. coli (EHEC) (56).

A diverse chemical library of 150,000 compounds was screenedfor those that block expression of a LEE1-lacZ reporter strain ofEHEC (11). Compounds were screened in an assay using spentmedium (which contains AI-3) to activate QseC and induce re-porter gene expression. The most potent inhibitor identified wasimproved by structure-activity relationship (SAR) studies andnamed LED209 (see Table 1 for structure) (11).

LED209 did not inhibit bacterial growth but rather selectivelyinhibited virulence gene expression by inhibiting QseC autophos-phorylation (11). While LED209 was ineffective at reducing colo-nization loads in an infant rabbit model of EHEC infection (per-haps due to rapid absorption from the gastrointestinal tract), itsignificantly reduced the mouse pathogenicity of both S. Typhi-murium and F. tularensis, which express QseC homologs of 87%and 57% similarity to EHEC QseC, respectively (11). QseC is im-portant for motility in S. Typhimurium (41) and for systemicinfection in F. tularensis (57).

Similar to the ToxT inhibitors virstatin and the toxtazins,LED209 inhibits virulence by targeting a specific virulence regu-lator without affecting growth. Its target, QseC, is broader, how-ever, being conserved in over 25 pathogenic bacteria but absent inmammals (11), giving this molecule a bacterium-specific broadspectrum of activity and making it an attractive lead compound.

CCG-2979. In addition to targeting sensory transduction path-ways, screens can target the ability of pathogens to manipulate thehost. Such a strategy identified CCG-2979 (see Table 1 for struc-ture), which reduces production of streptokinase (SK) by Strepto-coccus pyogenes (see step 6 in Fig. 1). CCG-2979 targets the pro-moter of SK (7), which is secreted by S. pyogenes and other groupA streptococci (GAS) and activates the host zymogen plasmino-gen to form plasmin, the central protease of the fibrinolytic systemcritical for regulating blood clots (58, 59). CCG-2979 was identi-fied using a cell-based assay in which ska, the SK gene promoter,controlled expression of the kanamycin resistance gene; positivehits were those that decreased the growth of the reporter strain butnot growth of a constitutive kanamycin-resistant strain (7).

CCG-2979 reduces SK activity in a dose-dependent mannerwithout inhibiting bacterial growth. Furthermore, GAS treatedwith 5 or 50 �M CCG-2979 were more susceptible to phagocytosisby host cells. A series of structural analogs of CCG-2979 was gen-erated, and structure-activity relationship (SAR) studies were per-formed (8, 60). The effect of one chemical analog, CCG-102487,on global gene expression was examined by microarray analysis,which demonstrated that expression of 29% of GAS genes in ad-dition to that of ska was altered, with the vast majority of thembeing reduced in expression. Included among these were othervirulence-associated genes coding for adhesins and toxins, alongwith genes encoding some metabolic functions (7).

Transgenic mice expressing the human plasminogen gene wereused to determine the therapeutic potential of CCG-2979. Usingthis well-established model (59), mice were subcutaneously in-jected with GAS, given a day to establish an infection, and thenintraperitoneally treated with compound daily for 5 days. Micetreated with CCG-2979 showed a statistically significant improve-ment in survival, while those treated with the CCG-102487 analogwere not protected from GAS-induced mortality (7).

Another analog, CCG-203592, inhibited SK at a level 35-foldgreater than that seen with CCG-2979 (8). Subsequent work led tothe observation that this and other analogs are also effectiveagainst another Gram-positive pathogen, Staphylococcus aureus.

Minireview

464 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 10: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

S. aureus is a major public health problem due in part to its abilityto form biofilms on implantable devices (61). Biofilm formationof three different biofilm-producing S. aureus strains was reducedin the presence of 50 �M CCG-203592, both in laboratory micro-titer plates and on silicone, the most common surface used forimplantable medical devices (8). Thus, this class of compoundsmay have therapeutic applications against different traits ex-pressed by two classes of important human pathogens, GAS and S.aureus, making them interesting therapeutic leads.

Pimozide. In contrast to screens using large, diverse chemicallibraries whose functions are completely unknown, screens usingcompounds with known biological activities result in more readilyidentified mechanisms of action in pathogenic processes. Pimoz-ide (see Table 1 for structure) was identified as an inhibitor ofListeria monocytogenes infection by screening a library of FDA-approved drugs (14).

L. monocytogenes infects macrophages (step 7 in Fig. 1) (62)and escapes the phagocytic vacuole by producing a hemolysin,listeriolysin O (LLO) (step 8 in Fig. 1). It replicates in the cytosolbefore spreading to neighboring cells by polymerizing host cellactin to propel itself into adjacent cells (step 9 in Fig. 1) (13, 63).While genetic and cellular biological studies have uncoveredmuch about L. monocytogenes pathogenicity, the mechanisms reg-ulating infection remain incompletely defined.

A novel chemical genetics approach was taken in which bothbacterial cells and host cells were exposed to a library of com-pounds (14). Primary bone marrow-derived macrophages(BMMs) were infected with L. monocytogenes constitutively ex-pressing green fluorescent protein (GFP) to enable visualizationof internalized bacteria. A total of 480 compounds with diverse,known biological activities were screened; 21 of these altered L.monocytogenes infection in one of three ways (14). The majority ofinhibitory compounds inhibited BMM infection, as observed by adecrease in GFP fluorescence within BMMs (i.e., internalized bac-teria) relative to the DMSO control results. Others enhanced bac-terial uptake or intracellular replication, causing an increase inGFP fluorescence per BMM cell. Lastly, some compounds inhib-ited cell-to-cell spread, observed as an increase in GFP fluores-cence per BMM, but only in very few cells.

Compounds that answered the screen fell into four categoriesbased on their previously known activities, and each category en-genders testable hypotheses about the conditions required for L.monocytogenes infection. The first category of inhibitors includescompounds that disrupt actin, which is required for phagocytosisby BMMs and for cell-to-cell spread of L. monocytogenes (64). Asecond group targets kinases and phosphatases, important for ac-tin rearrangement. These may provide new insights into the roleof specific kinases and phosphatases in L. monocytogenes infectionbut were not further analyzed. The members of a third group ofcompounds were categorized together on the basis of lackingshared activities with other compounds from the screen; furtherstudies on these may also yield new insights into L. monocytogenesinfection. The fourth category, composed of nine compounds(43% of the hits), includes molecules that affect calcium pathways.Calcium is not only important in many cellular signaling path-ways such as phagocytosis (65), it is also released in response toprotein kinase C (PKC) activation, which occurs during L. mono-cytogenes infection and modulates bacterial uptake and escapefrom the vacuole (66). Pimozide falls into this fourth category andwas chosen for further study; it should be emphasized, however,

that whether the action of pimozide on calcium pathways is alsowhat disrupts any given aspect of L. monocytogenes infection hasyet to be resolved. This raises an issue that confounds design of anysmall-molecule inhibitor, whether used as a chemical probe forbiological effects or as an actual therapeutic: inhibitors effective inspecific screens may have other off-target effects.

Pimozide inhibited intracellular infection of BMMs by L.monocytogenes by an order of magnitude after a 10-h treatment(14). While the exact mechanism for this is unclear, pimozide wasfound to inhibit infection at three distinct steps. The most potenteffect of pimozide was inhibition of macrophage phagocytosis,which was not limited to phagocytosis of L. monocytogenes but wasseen with three other bacteria as well: B. subtilis, S. Typhimurium,and E. coli K-12. Pimozide inhibited internalization of bacteria bymacrophages by 99% in a calcium-independent manner (14).Pimozide also reduced vacuolar escape of L. monocytogenes by26%, and this was not due to inhibition of LLO. Finally, pimozidetreatment decreased cell-to-cell spread by approximately 50%(14).

This screen generated many testable hypotheses regarding thecellular requirements of L. monocytogenes infection. Additionally,pimozide can be used to probe the molecular mechanisms under-lying BMM phagocytosis of bacteria in general and of L. monocy-togenes specifically. Pimozide blocks postsynaptic dopamine re-ceptors and is an FDA-approved antipsychotic molecule used totreat severe Tourette’s syndrome and schizophrenia (67). The mo-lecular mechanism by which pimozide inhibits macrophage func-tion leading to alterations in Listeria infection is not clear, but,given the class of compound into which it fell in this screen, itsmechanism may involve calcium signaling. In addition, this com-pound was shown to affect this particular host-pathogen system inmultiple and yet synergistic ways and highlights the connectionbetween what otherwise appear to be different pathogenic mech-anisms. Molecules such as pimozide have increased therapeuticappeal because of the lowered probability of bacteria overcomingtheir multiple effects by mutation. Additionally, an added benefitof an FDA-approved compound such as pimozide is that there is awealth of information regarding safety, pharmacokinetics, andpharmacodynamics.

Type 4 secretion inhibitors. Compounds with known biolog-ical activities have also been used to probe the type 4 secretionsystem (T4SS; also called the Icm/Dot type IVB system) (step 10 inFig. 1) in Legionella pneumophila, which infects and replicates inlung alveolar macrophages and causes Legionnaires’ disease (68,69). L. pneumophila avoids phagosome-lysosome fusion by usingits T4SS to secrete effectors that interfere with vesicular traffick-ing, the host innate immune response, phosphoinositide metab-olism, and ubiquitination (reviewed in reference 70). Given itsimportance in intracellular survival and replication (71), the T4SSis an attractive target for drug development. The T4SS can beactivated by contact with the host cell (72), but the signals thattrigger secretion of effectors are not well understood.

Compounds with known biological targets were screened toprobe the mechanisms of type 4 secretion in L. pneumophila (6). AT4SS-secreted effector protein (LepA) was fused to the TEM-1�-lactamase (BlaM), which cleaves a green substrate, CCF4, toproduce a blue product. Host cells were incubated with com-pounds for 24 h and infected with the L. pneumophila lepA-blaMreporter strain, and host-bacterial cell contact was initiated by alow-speed centrifugation step. CCF4 was added 1 h later, and flu-

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 465Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 11: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

orescence was measured 2 h later to quantify the ratio of cleavedCCF4 to uncleaved CCF4 (6).

Of 2,640 compounds screened, 22 inhibited translocation withefficiencies ranging from 63% to 100% and were categorized intogroups based on their previously known targets: ionophore orprotonophores, calmodulin, cytoskeleton dynamics, NF-�B, ser-ine proteases, kinases or phosphatases, and others (6). The major-ity of the inhibitors are molecules previously known to affect hostcytoskeleton dynamics, including those affecting tubulin (step 11in Fig. 1), actin (step 12 in Fig. 1), and phosphoinositol 3-kinase(PI3K). Ionophores and protonophores discharge membraneelectric potential (�) and collapse the proton gradient (�pH),both components of the proton motive force (PMF) (step 13 inFig. 1) (73, 74). All three identified ionophores and protono-phores inhibited Icm/Dot-dependent lysis of red blood cells by L.pneumophila (6), indicating that translocation of effectors is atleast partially dependent on the PMF. Furthermore, one iono-phore, carbonyl cyanide m-chlorophenylhydrazone (CCCP)(shown in Table 1), inhibited LepA translocation in L. pneumo-phila in a dose-dependent and reversible manner (6). The PMFwas not previously known to play a role in T4SS-mediated trans-location.

Nineteen of the 22 identified inhibitors significantly affectedthe ability of macrophages to phagocytose other bacteria (step 7 inFig. 1) (6). To test whether phagocytosis is required for transloca-tion of L. pneumophila Icm/Dot effector proteins, cytoskeletoninhibitors were used to inhibit coiling phagocytosis, the type ofphagocytosis used by L. pneumophila to enter macrophages (75).Host-bacterium contact was instead initiated by opsonizationwith L. pneumophila-specific antibodies (76). Opsonization re-stored effector translocation in the absence of phagocytosis (6),indicating that host-cell binding, and not phagocytosis, is re-quired for translocation by the Icm/Dot system. These results sup-port a model where the T4SS in L. pneumophila is in a “locked andloaded” state, ready to inject effector proteins upon contact with ahost cell (step 10 in Fig. 1).

Another important finding from this study came by furtheranalyzing the RWJ-60475 translocation inhibitor (shown in Table1), previously known to inhibit a receptor tyrosine phosphatephosphatase (step 14 in Fig. 1) (77). Work with this compoundrevealed that CD45 and CD148 are required to phagocytose L.pneumophila (6).

In this Legionella work, screening just 2,500 small moleculeseventually generated many new testable hypotheses from the hitsthat arose. Additionally, new information was uncovered relatingphagocytosis and effector translocation to the PMF, host cell con-tact, and CD45 or CD148, providing a great example of howchemical genetics deepens our understanding of pathogenicitymechanisms.

ADVANCES IN TARGET IDENTIFICATION

Several thorough reviews (78–82) have been published discussingadvances made in improved target identification, and some ofthese approaches are briefly noted here. Target identification canbe the most challenging, time-consuming aspect of small-mole-cule discovery programs. As noted in the discussion of pimocide,one of the major challenges is that small molecules may alter tar-gets other than the one of interest. The potential for this must beconsidered during target identification.

In this review, we have noted several approaches that have been

used to characterize the molecules discussed. Generally, these canbe classified into three categories: genetic, proteomic, and chem-ical. Genetic approaches include comparing transcriptomes oftreated and untreated samples using microarray analysis (83, 84)or transcriptome sequencing (RNA-seq) (85), sequencing a resis-tant mutant (86–88), and error-prone PCR-based target identifi-cation (89). Proteomic methods include comparing the pro-teomes of treated and untreated samples using affinity pulldownanalysis (90), stable isotope labeling by amino acids in cell culture(SILAC) (91, 92), or isobaric tags for relative and absolute quan-tification (iTRAQ) (93, 94). Finally, chemical approaches, includ-ing click chemistry (95–97) and synergy (98–100), are also becom-ing fruitful for identifying molecular targets, as is in silico chemicalmodeling (101, 102).

CONCLUSIONS

Chemical genetics can uncover small-molecule inhibitors affect-ing a range of targets important for pathogenesis, including fac-tors involved in virulence regulation and host-pathogen interac-tions (Fig. 1). This review has covered molecules that are excellentchemical probes for studying molecular mechanisms in living cellsand that have potential as therapeutic leads, with various degreesof pathogen specificity and minimal effects on the microbiota. Wehave also tried to illustrate common strategies used in small-mol-ecule screening (outlined in Fig. 2), for example, the use of large,diverse chemical libraries or of smaller libraries of compoundswhose biological targets and mechanisms of action are unknown.Each approach can lead to important new discoveries. For thera-peutic lead development, however, the use of screens of previouslystudied compounds (sometimes termed “repurposing” [103]) hasthe advantage of reducing the costs, risks, and time associated withdrug development. We have covered how secondary and tertiaryscreens can help eliminate false hits and improve the specificity,efficacy, and toxicity of a given compound.

The power and appeal of using chemical genetics—such as inthe examples we have covered—are in how it supports both basicand translational research, answering questions about host-pathogen biology and providing potential therapeutic lead com-pounds to combat the increasing threat of pathogens resistant totraditional antibiotics.

REFERENCES1. Centers for Disease Control and Prevention. 2013. Antibiotic resis-

tance threats in the United States, 2013. Centers for Disease Controland Prevention, Atlanta, GA. http://www.cdc.gov/drugresistance/threat-report-2013/.

2. D’Costa VM, McGrann KM, Hughes DW, Wright GD. 2006. Samplingthe antibiotic resistome. Science 311:374 –377. http://dx.doi.org/10.1126/science.1120800.

3. McFarland LV. 2008. Antibiotic-associated diarrhea: epidemiology,trends and treatment. Future Microbiol 3:563–578. http://dx.doi.org/10.2217/17460913.3.5.563.

4. Sekirov I, Russell SL, Antunes LC, Finlay BB. 2010. Gut microbiota inhealth and disease. Physiol Rev 90:859 –904. http://dx.doi.org/10.1152/physrev.00045.2009.

5. Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC,Pepin J, Wilcox MH; Society for Healthcare Epidemiology of America;Infectious Diseases Society of America. 2010. Clinical practice guide-lines for Clostridium difficile infection in adults: 2010 update by theSociety for Healthcare Epidemiology of America (SHEA) and the Infec-tious Diseases Society of America (IDSA). Infect Control Hosp Epide-miol 31:431– 455. http://dx.doi.org/10.1086/651706.

6. Charpentier X, Gabay JE, Reyes M, Zhu JW, Weiss A, Shuman HA.2009. Chemical genetics reveals bacterial and host cell functions critical

Minireview

466 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 12: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

for type IV effector translocation by Legionella pneumophila. PLoS Pat-hog 5:e1000501. http://dx.doi.org/10.1371/journal.ppat.1000501.

7. Sun H, Xu Y, Sitkiewicz I, Ma Y, Wang X, Yestrepsky BD, Huang Y,Lapadatescu MC, Larsen MJ, Larsen SD, Musser JM, Ginsburg D.2012. Inhibitor of streptokinase gene expression improves survival aftergroup A streptococcus infection in mice. Proc Natl Acad Sci U S A 109:3469 –3474. http://dx.doi.org/10.1073/pnas.1201031109.

8. Ma Y, Xu Y, Yestrepsky BD, Sorenson RJ, Chen M, Larsen SD, Sun H.2012. Novel inhibitors of Staphylococcus aureus virulence gene expres-sion and biofilm formation. PLoS One 7:e47255. http://dx.doi.org/10.1371/journal.pone.0047255.

9. Palmer ME, Chaturongakul S, Wiedmann M, Boor KJ. 2011. TheListeria monocytogenes sigmaB regulon and its virulence-associatedfunctions are inhibited by a small molecule. mBio 2:e00241–11. http://dx.doi.org/10.1128/mBio.00241-11.

10. Ringus DL, Gaballa A, Helmann JD, Wiedmann M, Boor KJ. 2013.Fluoro-phenyl-styrene-sulfonamide, a novel inhibitor of sigmaB activ-ity, prevents the activation of sigmaB by environmental and energystresses in Bacillus subtilis. J Bacteriol 195:2509 –2517. http://dx.doi.org/10.1128/JB.00107-13.

11. Rasko DA, Moreira CG, Li DR, Reading NC, Ritchie JM, Waldor MK,Williams N, Taussig R, Wei S, Roth M, Hughes DT, Huntley JF, FinaMW, Falck JR, Sperandio V. 2008. Targeting QseC signaling and viru-lence for antibiotic development. Science 321:1078 –1080. http://dx.doi.org/10.1126/science.1160354.

12. Svensson A, Fex T, Kihlberg J. 2000. Preparation of fluorinated linkers:use of 19F NMR spectroscopy to establish conditions for solid-phasesynthesis of pilicide libraries. J Comb Chem 2:736 –748. http://dx.doi.org/10.1021/cc0000646.

13. Pinkner JS, Remaut H, Buelens F, Miller E, Aberg V, Pemberton N,Hedenstrom M, Larsson A, Seed P, Waksman G, Hultgren SJ,Almqvist F. 2006. Rationally designed small compounds inhibit pilusbiogenesis in uropathogenic bacteria. Proc Natl Acad Sci U S A 103:17897–17902. http://dx.doi.org/10.1073/pnas.0606795103.

14. Lieberman LA, Higgins DE. 2009. A small-molecule screen identifies theantipsychotic drug pimozide as an inhibitor of Listeria monocytogenesinfection. Antimicrob Agents Chemother 53:756 –764. http://dx.doi.org/10.1128/AAC.00607-08.

15. Anthouard R, DiRita VJ. 2013. Small-molecule inhibitors of toxT ex-pression in Vibrio cholerae. mBio 4:e00403-13. http://dx.doi.org/10.1128/mBio.00403-13.

16. Felise HB, Nguyen HV, Pfuetzner RA, Barry KC, Jackson SR, BlancMP, Bronstein PA, Kline T, Miller SI. 2008. An inhibitor of gram-negative bacterial virulence protein secretion. Cell Host Microbe 4:325–336. http://dx.doi.org/10.1016/j.chom.2008.08.001.

17. Hung DT, Shakhnovich EA, Pierson E, Mekalanos JJ. 2005. Small-molecule inhibitor of Vibrio cholerae virulence and intestinal coloniza-tion. Science 310:670 – 674. http://dx.doi.org/10.1126/science.1116739.

18. Shakhnovich EA, Sturtevant D, Mekalanos JJ. 2007. Molecular mech-anisms of virstatin resistance by non-O1/non-O139 strains of Vibriocholerae. Mol Microbiol 66:1331–1341.

19. Shakhnovich EA, Hung DT, Pierson E, Lee K, Mekalanos JJ. 2007.Virstatin inhibits dimerization of the transcriptional activator ToxT.Proc Natl Acad Sci U S A 104:2372–2377. http://dx.doi.org/10.1073/pnas.0611643104.

20. Beck NA, Krukonis ES, DiRita VJ. 2004. TcpH influences virulencegene expression in Vibrio cholerae by inhibiting degradation of the tran-scription activator TcpP. J Bacteriol 186:8309 – 8316. http://dx.doi.org/10.1128/JB.186.24.8309-8316.2004.

21. Crawford JA, Krukonis ES, DiRita VJ. 2003. Membrane localization ofthe ToxR winged-helix domain is required for TcpP-mediated virulencegene activation in Vibrio cholerae. Mol Microbiol 47:1459 –1473. http://dx.doi.org/10.1046/j.1365-2958.2003.03398.x.

22. DiRita VJ. 1992. Co-ordinate expression of virulence genes by ToxR inVibrio cholerae. Mol Microbiol 6:451– 458. http://dx.doi.org/10.1111/j.1365-2958.1992.tb01489.x.

23. DiRita VJ, Parsot C, Jander G, Mekalanos JJ. 1991. Regulatory cascadecontrols virulence in Vibrio cholerae. Proc Natl Acad Sci U S A 88:5403–5407. http://dx.doi.org/10.1073/pnas.88.12.5403.

24. Krukonis ES, DiRita VJ. 2003. DNA binding and ToxR responsivenessby the wing domain of TcpP, an activator of virulence gene expression inVibrio cholerae. Mol Cell 12:157–165. http://dx.doi.org/10.1016/S1097-2765(03)00222-3.

25. Matson JS, Withey JH, DiRita VJ. 2007. Regulatory networks control-ling Vibrio cholerae virulence gene expression. Infect Immun 75:5542–5549. http://dx.doi.org/10.1128/IAI.01094-07.

26. Yu RR, DiRita VJ. 2002. Regulation of gene expression in Vibrio chol-erae by ToxT involves both antirepression and RNA polymerase stimu-lation. Mol Microbiol 43:119 –134. http://dx.doi.org/10.1046/j.1365-2958.2002.02721.x.

27. Withey JH, DiRita VJ. 2006. The toxbox: specific DNA sequence re-quirements for activation of Vibrio cholerae virulence genes by ToxT.Mol Microbiol 59:1779 –1789. http://dx.doi.org/10.1111/j.1365-2958.2006.05053.x.

28. Hsiao A, Xu X, Kan B, Kulkarni RV, Zhu J. 2009. Direct regulation bythe Vibrio cholerae regulator ToxT to modulate colonization and anti-colonization pilus expression. Infect Immun 77:1383–1388. http://dx.doi.org/10.1128/IAI.01156-08.

29. Chaturongakul S, Raengpradub S, Palmer ME, Bergholz TM, Orsi RH,Hu Y, Ollinger J, Wiedmann M, Boor KJ. 2011. Transcriptomic andphenotypic analyses identify coregulated, overlapping regulons amongPrfA, CtsR, HrcA, and the alternative sigma factors sigmaB, sigmaC,sigmaH, and sigmaL in Listeria monocytogenes. Appl Environ Microbiol77:187–200. http://dx.doi.org/10.1128/AEM.00952-10.

30. Llamas MA, van der Sar A, Chu BC, Sparrius M, Vogel HJ, Bitter W.2009. A novel extracytoplasmic function (ECF) sigma factor regulatesvirulence in Pseudomonas aeruginosa. PLoS Pathog 5:e1000572. http://dx.doi.org/10.1371/journal.ppat.1000572.

31. Raman S, Hazra R, Dascher CC, Husson RN. 2004. Transcriptionregulation by the Mycobacterium tuberculosis alternative sigma factorSigD and its role in virulence. J Bacteriol 186:6605– 6616. http://dx.doi.org/10.1128/JB.186.19.6605-6616.2004.

32. Yin Y, Withers TR, Wang X, Yu HD. 2013. Evidence for sigma factorcompetition in the regulation of alginate production by Pseudomonasaeruginosa. PLoS One 8:e72329. http://dx.doi.org/10.1371/journal.pone.0072329.

33. Kazmierczak MJ, Wiedmann M, Boor KJ. 2005. Alternative sigmafactors and their roles in bacterial virulence. Microbiol Mol Biol Rev69:527–543. http://dx.doi.org/10.1128/MMBR.69.4.527-543.2005.

34. Ferreira A, O’Byrne CP, Boor KJ. 2001. Role of sigma(B) in heat,ethanol, acid, and oxidative stress resistance and during carbon starva-tion in Listeria monocytogenes. Appl Environ Microbiol 67:4454 – 4457.http://dx.doi.org/10.1128/AEM.67.10.4454-4457.2001.

35. Xayarath B, Freitag NE. 2012. Optimizing the balance between hostand environmental survival skills: lessons learned from Listeriamonocytogenes. Future Microbiol 7:839 – 852. http://dx.doi.org/10.2217/fmb.12.57.

36. Hecker M, Pane-Farre J, Volker U. 2007. SigB-dependent general stressresponse in Bacillus subtilis and related gram-positive bacteria. AnnuRev Microbiol 61:215–236. http://dx.doi.org/10.1146/annurev.micro.61.080706.093445.

37. Benson AK, Haldenwang WG. 1993. Bacillus subtilis sigma B is regu-lated by a binding protein (RsbW) that blocks its association with coreRNA polymerase. Proc Natl Acad Sci U S A 90:2330 –2334. http://dx.doi.org/10.1073/pnas.90.6.2330.

38. Hecker M, Volker U. 2001. General stress response of Bacillus subtilisand other bacteria. Adv Microb Physiol 44:35–91.

39. Depke M, Burian M, Schafer T, Broker BM, Ohlsen K, Volker U. 2012.The alternative sigma factor B modulates virulence gene expression in amurine Staphylococcus aureus infection model but does not influencekidney gene expression pattern of the host. Int J Med Microbiol 302:33–39. http://dx.doi.org/10.1016/j.ijmm.2011.09.013.

40. Fouet A, Namy O, Lambert G. 2000. Characterization of the operonencoding the alternative sigma(B) factor from Bacillus anthracis and itsrole in virulence. J Bacteriol 182:5036 –5045. http://dx.doi.org/10.1128/JB.182.18.5036-5045.2000.

41. Cornelis GR. 2002. The Yersinia Ysc-Yop virulence apparatus. Int J MedMicrobiol 291:455– 462.

42. Cornelis GR. 2006. The type III secretion injectisome. Nat Rev Microbiol4:811– 825. http://dx.doi.org/10.1038/nrmicro1526.

43. Cianciotto NP. 2005. Type II secretion: a protein secretion system for allseasons. Trends Microbiol 13:581–588. http://dx.doi.org/10.1016/j.tim.2005.09.005.

44. Wretlind B, Pavlovskis OR. 1983. Pseudomonas aeruginosa elastase andits role in pseudomonas infections. Rev Infect Dis 5(Suppl 5):S998 –S1004. http://dx.doi.org/10.1093/clinids/5.Supplement_5.S998.

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 467Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 13: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

45. Hultgren SJ, Abraham S, Caparon M, Falk P, St Geme JW, III,Normark S. 1993. Pilus and nonpilus bacterial adhesins: assembly andfunction in cell recognition. Cell 73:887–901. http://dx.doi.org/10.1016/0092-8674(93)90269-V.

46. Klemm P. 1992. FimC, a chaperone-like periplasmic protein of Esche-richia coli involved in biogenesis of type 1 fimbriae. Res Microbiol 143:831– 838. http://dx.doi.org/10.1016/0923-2508(92)90070-5.

47. Lindberg F, Tennent JM, Hultgren SJ, Lund B, Normark S. 1989.PapD, a periplasmic transport protein in P-pilus biogenesis. J Bacteriol171:6052– 6058.

48. Nishiyama M, Vetsch M, Puorger C, Jelesarov I, Glockshuber R. 2003.Identification and characterization of the chaperone-subunit complex-binding domain from the type 1 pilus assembly platform FimD. J MolBiol 330:513–525. http://dx.doi.org/10.1016/S0022-2836(03)00591-6.

49. Thanassi DG, Stathopoulos C, Dodson K, Geiger D, Hultgren SJ. 2002.Bacterial outer membrane ushers contain distinct targeting and assemblydomains for pilus biogenesis. J Bacteriol 184:6260 – 6269. http://dx.doi.org/10.1128/JB.184.22.6260-6269.2002.

50. Geibel S, Waksman G. 16 October 2013, posting date. The moleculardissection of the chaperone-usher pathway. Biochim Biophys Acta http://dx.doi.org/10.1016/j.bbamcr.2013.09.023.

51. Sauer FG, Remaut H, Hultgren SJ, Waksman G. 2004. Fiber assemblyby the chaperone-usher pathway. Biochim Biophys Acta 1694:259 –267.http://dx.doi.org/10.1016/j.bbamcr.2004.02.010.

52. Sauer FG, Futterer K, Pinkner JS, Dodson KW, Hultgren SJ, Waksman G.1999. Structural basis of chaperone function and pilus biogenesis. Science285:1058–1061. http://dx.doi.org/10.1126/science.285.5430.1058.

53. Nishiyama M, Horst R, Eidam O, Herrmann T, Ignatov O, Vetsch M,Bettendorff P, Jelesarov I, Grutter MG, Wuthrich K, Glockshuber R,Capitani G. 2005. Structural basis of chaperone-subunit complex recog-nition by the type 1 pilus assembly platform FimD. EMBO J 24:2075–2086. http://dx.doi.org/10.1038/sj.emboj.7600693.

54. Hughes DT, Sperandio V. 2008. Inter-kingdom signalling: communi-cation between bacteria and their hosts. Nat Rev Microbiol 6:111–120.http://dx.doi.org/10.1038/nrmicro1836.

55. Sperandio V, Torres AG, Jarvis B, Nataro JP, Kaper JB. 2003. Bacteria-host communication: the language of hormones. Proc Natl Acad SciU S A 100:8951– 8956. http://dx.doi.org/10.1073/pnas.1537100100.

56. Clarke MB, Hughes DT, Zhu C, Boedeker EC, Sperandio V. 2006. TheQseC sensor kinase: a bacterial adrenergic receptor. Proc Natl Acad SciU S A 103:10420 –10425. http://dx.doi.org/10.1073/pnas.0604343103.

57. Weiss DS, Brotcke A, Henry T, Margolis JJ, Chan K, Monack DM.2007. In vivo negative selection screen identifies genes required for Fran-cisella virulence. Proc Natl Acad Sci U S A 104:6037– 6042. http://dx.doi.org/10.1073/pnas.0609675104.

58. Boxrud PD, Fay WP, Bock PE. 2000. Streptokinase binds to humanplasmin with high affinity, perturbs the plasmin active site, and inducesexpression of a substrate recognition exosite for plasminogen. J BiolChem 275:14579 –14589. http://dx.doi.org/10.1074/jbc.275.19.14579.

59. Sun H, Ringdahl U, Homeister JW, Fay WP, Engleberg NC, Yang AY,Rozek LS, Wang X, Sjobring U, Ginsburg D. 2004. Plasminogen is acritical host pathogenicity factor for group A streptococcal infection.Science 305:1283–1286. http://dx.doi.org/10.1126/science.1101245.

60. Yestrepsky BD, Xu Y, Breen ME, Li X, Rajeswaran WG, Ryu JG,Sorenson RJ, Tsume Y, Wilson MW, Zhang W, Sun D, Sun H, LarsenSD. 2013. Novel inhibitors of bacterial virulence: development of 5,6-dihydrobenzo[h]quinazolin-4(3H)-ones for the inhibition of group Astreptococcal streptokinase expression. Bioorg Med Chem 21:1880 –1897. http://dx.doi.org/10.1016/j.bmc.2013.01.046.

61. Kokai-Kun JF, Chanturiya T, Mond JJ. 2009. Lysostaphin eradicatesestablished Staphylococcus aureus biofilms in jugular vein catheterizedmice. J Antimicrob Chemother 64:94 –100. http://dx.doi.org/10.1093/jac/dkp145.

62. Gaillard JL, Berche P, Mounier J, Richard S, Sansonetti P. 1987. Invitro model of penetration and intracellular growth of Listeria monocy-togenes in the human enterocyte-like cell line Caco-2. Infect Immun55:2822–2829.

63. Portnoy DA, Jacks PS, Hinrichs DJ. 1988. Role of hemolysin for theintracellular growth of Listeria monocytogenes. J Exp Med 167:1459 –1471. http://dx.doi.org/10.1084/jem.167.4.1459.

64. Kocks C, Gouin E, Tabouret M, Berche P, Ohayon H, Cossart P. 1992.L. monocytogenes-induced actin assembly requires the actA gene prod-uct, a surface protein. Cell 68:521–531.

65. TranVan Nhieu G, Clair C, Grompone G, Sansonetti P. 2004. Calciumsignalling during cell interactions with bacterial pathogens. Biol Cell 96:93–101. http://dx.doi.org/10.1016/j.biolcel.2003.10.006.

66. Wadsworth SJ, Goldfine H. 2002. Mobilization of protein kinase C inmacrophages induced by Listeria monocytogenes affects its internaliza-tion and escape from the phagosome. Infect Immun 70:4650 – 4660. http://dx.doi.org/10.1128/IAI.70.8.4650-4660.2002.

67. Tueth MJ, Cheong JA. 1993. Clinical uses of pimozide. South Med J86:344 –349. http://dx.doi.org/10.1097/00007611-199303000-00019.

68. Fraser DW, Tsai TR, Orenstein W, Parkin WE, Beecham HJ, SharrarRG, Harris J, Mallison GF, Martin SM, McDade JE, Shepard CC,Brachman PS. 1977. Legionnaires’ disease: description of an epidemic ofpneumonia. N Engl J Med 297:1189 –1197. http://dx.doi.org/10.1056/NEJM197712012972201.

69. McDade JE, Shepard CC, Fraser DW, Tsai TR, Redus MA, DowdleWR. 1977. Legionnaires’ disease: isolation of a bacterium and demon-stration of its role in other respiratory disease. N Engl J Med 297:1197–1203. http://dx.doi.org/10.1056/NEJM197712012972202.

70. Horwitz MA. 1983. The Legionnaires’ disease bacterium (Legionellapneumophila) inhibits phagosome-lysosome fusion in human mono-cytes. J Exp Med 158:2108 –2126. http://dx.doi.org/10.1084/jem.158.6.2108.

71. Vogel JP, Andrews HL, Wong SK, Isberg RR. 1998. Conjugative trans-fer by the virulence system of Legionella pneumophila. Science 279:873–876. http://dx.doi.org/10.1126/science.279.5352.873.

72. Chen J, Reyes M, Clarke M, Shuman HA. 2007. Host cell-dependentsecretion and translocation of the LepA and LepB effectors of Legionellapneumophila. Cell Microbiol 9:1660 –1671. http://dx.doi.org/10.1111/j.1462-5822.2007.00899.x.

73. Jana B, Panja S, Saha S, Basu T. 2009. Mechanism of protonophores-mediated induction of heat-shock response in Escherichia coli. BMCMicrobiol 9:20. http://dx.doi.org/10.1186/1471-2180-9-20.

74. Bakker EP, Mangerich WE. 1981. Interconversion of components of thebacterial proton motive force by electrogenic potassium transport. J Bac-teriol 147:820 – 826.

75. Horwitz MA. 1984. Phagocytosis of the Legionnaires’ disease bacterium(Legionella pneumophila) occurs by a novel mechanism: engulfmentwithin a pseudopod coil. Cell 36:27–33. http://dx.doi.org/10.1016/0092-8674(84)90070-9.

76. Husmann LK, Johnson W. 1992. Adherence of Legionella pneumophilato guinea pig peritoneal macrophages, J774 mouse macrophages, andundifferentiated U937 human monocytes: role of Fc and complementreceptors. Infect Immun 60:5212–5218.

77. Beers SA, Malloy EA, Wu W, Wachter MP, Gunnia U, Cavender D,Harris C, Davis J, Brosius R, Pellegrino-Gensey JL, Siekierka J.1997. Nitroarylhydroxymethylphosphonic acids as inhibitors ofCD45. Bioorg Med Chem 5:2203–2211. http://dx.doi.org/10.1016/S0968-0896(97)00174-0.

78. Burdine L, Kodadek T. 2004. Target identification in chemical genetics:the (often) missing link. Chem Biol 11:593–597. http://dx.doi.org/10.1016/j.chembiol.2004.05.001.

79. Zheng XS, Chan TF, Zhou HH. 2004. Genetic and genomic approachesto identify and study the targets of bioactive small molecules. Chem Biol11:609 – 618. http://dx.doi.org/10.1016/j.chembiol.2003.08.011.

80. Chan JN, Nislow C, Emili A. 2010. Recent advances and method devel-opment for drug target identification. Trends Pharmacol Sci 31:82– 88.http://dx.doi.org/10.1016/j.tips.2009.11.002.

81. Veenstra TD. 2007. Global and targeted quantitative proteomics forbiomarker discovery. J Chromatogr B Analyt Technol Biomed Life Sci847:3–11. http://dx.doi.org/10.1016/j.jchromb.2006.09.004.

82. Schenone M, Dancik V, Wagner BK, Clemons PA. 2013. Target iden-tification and mechanism of action in chemical biology and drug discov-ery. Nat Chem Biol 9:232–240. http://dx.doi.org/10.1038/nchembio.1199.

83. Lee JH, Cho MH, Lee J. 2011. 3-Indolylacetonitrile decreases Esche-richia coli O157:H7 biofilm formation and Pseudomonas aeruginosavirulence. Environ Microbiol 13:62–73. http://dx.doi.org/10.1111/j.1462-2920.2010.02308.x.

84. O’Loughlin CT, Miller LC, Siryaporn A, Drescher K, Semmelhack MF,Bassler BL. 2013. A quorum-sensing inhibitor blocks Pseudomonasaeruginosa virulence and biofilm formation. Proc Natl Acad Sci U S A110:17981–17986. http://dx.doi.org/10.1073/pnas.1316981110.

85. Wacker SA, Houghtaling BR, Elemento O, Kapoor TM. 2012. Using

Minireview

468 iai.asm.org February 2015 Volume 83 Number 2Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from

Page 14: Chemical Biology Applied to the Study of Bacterial Pathogens · Chemical Biology Applied to the Study of Bacterial Pathogens Rebecca Anthouard,* Victor J. DiRita Laboratory of Genetics

transcriptome sequencing to identify mechanisms of drug action andresistance. Nat Chem Biol 8:235–237. http://dx.doi.org/10.1038/nchembio.779.

86. Comas I, Borrell S, Roetzer A, Rose G, Malla B, Kato-Maeda M,Galagan J, Niemann S, Gagneux S. 2012. Whole-genome sequencing ofrifampicin-resistant Mycobacterium tuberculosis strains identifies com-pensatory mutations in RNA polymerase genes. Nat Genet 44:106 –110.http://dx.doi.org/10.1038/ng.1038.

87. Smurnyy Y, Cai M, Wu H, McWhinnie E, Tallarico JA, Yang Y, FengY. 2014. DNA sequencing and CRISPR-Cas9 gene editing for target val-idation in mammalian cells. Nat Chem Biol 10:623– 625. http://dx.doi.org/10.1038/nchembio.1550.

88. Ioerger TR, O’Malley T, Liao R, Guinn KM, Hickey MJ, Mohaideen N,Murphy KC, Boshoff HI, Mizrahi V, Rubin EJ, Sassetti CM, Barry CE,III, Sherman DR, Parish T, Sacchettini JC. 2013. Identification of newdrug targets and resistance mechanisms in Mycobacterium tuberculosis.PLoS One 8:e75245. http://dx.doi.org/10.1371/journal.pone.0075245.

89. Belanger AE, Lai A, Brackman MA, LeBlanc DJ. 2002. PCR-basedordered genomic libraries: a new approach to drug target identificationfor Streptococcus pneumoniae. Antimicrob Agents Chemother 46:2507–2512. http://dx.doi.org/10.1128/AAC.46.8.2507-2512.2002.

90. Dadvar P, O’Flaherty M, Scholten A, Rumpel K, Heck AJ. 2009. Achemical proteomics based enrichment technique targeting the interac-tome of the PDE5 inhibitor PF-4540124. Mol Biosyst 5:472– 482. http://dx.doi.org/10.1039/b815709j.

91. Ong SE, Schenone M, Margolin AA, Li X, Do K, Doud MK, Mani DR,Kuai L, Wang X, Wood JL, Tolliday NJ, Koehler AN, Marcaurelle LA,Golub TR, Gould RJ, Schreiber SL, Carr SA. 2009. Identifying theproteins to which small-molecule probes and drugs bind in cells. ProcNatl Acad Sci U S A 106:4617– 4622. http://dx.doi.org/10.1073/pnas.0900191106.

92. Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, PandeyA, Mann M. 2002. Stable isotope labeling by amino acids in cell culture,SILAC, as a simple and accurate approach to expression proteomics. MolCell Proteomics 1:376 –386. http://dx.doi.org/10.1074/mcp.M200025-MCP200.

93. Bantscheff M, Eberhard D, Abraham Y, Bastuck S, Boesche M, Hob-son S, Mathieson T, Perrin J, Raida M, Rau C, Reader V, Sweetman G,Bauer A, Bouwmeester T, Hopf C, Kruse U, Neubauer G, Ramsden N,Rick J, Kuster B, Drewes G. 2007. Quantitative chemical proteomicsreveals mechanisms of action of clinical ABL kinase inhibitors. Nat Bio-technol 25:1035–1044. http://dx.doi.org/10.1038/nbt1328.

94. Ross PL, Huang YN, Marchese JN, Williamson B, Parker K, Hattan S,Khainovski N, Pillai S, Dey S, Daniels S, Purkayastha S, Juhasz P,Martin S, Bartlet-Jones M, He F, Jacobson A, Pappin DJ. 2004.Multiplexed protein quantitation in Saccharomyces cerevisiae usingamine-reactive isobaric tagging reagents. Mol Cell Proteomics 3:1154 –1169. http://dx.doi.org/10.1074/mcp.M400129-MCP200.

95. Willand N, Desroses M, Toto P, Dirie B, Lens Z, Villeret V, RucktooaP, Locht C, Baulard A, Deprez B. 2010. Exploring drug target flexibilityusing in situ click chemistry: application to a mycobacterial transcrip-tional regulator. ACS Chem Biol 5:1007–1013. http://dx.doi.org/10.1021/cb100177g.

96. Vila A, Tallman KA, Jacobs AT, Liebler DC, Porter NA, Marnett LJ.2008. Identification of protein targets of 4-hydroxynonenal using clickchemistry for ex vivo biotinylation of azido and alkynyl derivatives.Chem Res Toxicol 21:432– 444. http://dx.doi.org/10.1021/tx700347w.

97. Wang K, Yang T, Wu Q, Zhao X, Nice EC, Huang C. 2012. Chemistry-based functional proteomics for drug target deconvolution. Expert RevProteomics 9:293–310. http://dx.doi.org/10.1586/epr.12.19.

98. Farha MA, Brown ED. 2010. Chemical probes of Escherichia coli un-covered through chemical-chemical interaction profiling with com-pounds of known biological activity. Chem Biol 17:852– 862. http://dx.doi.org/10.1016/j.chembiol.2010.06.008.

99. Miller ML, Molinelli EJ, Nair JS, Sheikh T, Samy R, Jing X, He Q,Korkut A, Crago AM, Singer S, Schwartz GK, Sander C. 2013. Drugsynergy screen and network modeling in dedifferentiated liposarcomaidentifies CDK4 and IGF1R as synergistic drug targets. Sci Signal 6:ra85.http://dx.doi.org/10.1126/scisignal.2004014.

100. Wolpaw AJ, Shimada K, Skouta R, Welsch ME, Akavia UD, Pe’er D,Shaik F, Bulinski JC, Stockwell BR. 2011. Modulatory profiling identi-fies mechanisms of small molecule-induced cell death. Proc Natl AcadSci U S A 108:E771–E780. http://dx.doi.org/10.1073/pnas.1106149108.

101. Kolb P, Ferreira RS, Irwin JJ, Shoichet BK. 2009. Docking and che-moinformatic screens for new ligands and targets. Curr Opin Biotechnol20:429 – 436. http://dx.doi.org/10.1016/j.copbio.2009.08.003.

102. Boyce SE, Mobley DL, Rocklin GJ, Graves AP, Dill KA, Shoichet BK.2009. Predicting ligand binding affinity with alchemical free energymethods in a polar model binding site. J Mol Biol 394:747–763. http://dx.doi.org/10.1016/j.jmb.2009.09.049.

103. Swamidass SJ. 2011. Mining small-molecule screens to repurpose drugs.Brief Bioinform 12:327–335. http://dx.doi.org/10.1093/bib/bbr028.

Minireview

February 2015 Volume 83 Number 2 iai.asm.org 469Infection and Immunity

on May 22, 2020 by guest

http://iai.asm.org/

Dow

nloaded from