Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

664

Quinolone Antibiotics Induce Shiga Toxin–Encoding Bacteriophages,


Toxin Production, and Death in Mice
Xiaoping Zhang,a Aaron D. McDaniel,a Lucas E. Wolf,a Division of Geographic Medicine and Infectious Diseases, Tupper

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


Gerald T. Keusch, Matthew K. Waldor, Research Institute, New England Medical Center,
and David W. K. Acheson Boston, Massachusetts

Shiga toxin–producing Escherichia coli (STEC) cause significant disease; treatment is sup-
portive and antibiotic use is controversial. Ciprofloxacin but not fosfomycin causes Shiga
toxin–encoding bacteriophage induction and enhanced Shiga toxin (Stx) production from
E. coli O157:H7 in vitro. The potential clinical relevance of this was examined in mice colonized
with E. coli O157:H7 and given either ciprofloxacin or fosfomycin. Both antibiotics caused a
reduction in fecal STEC. However, animals treated with ciprofloxacin had a marked increase
in free fecal Stx, associated with death in two-thirds of the mice, whereas fosfomycin did not.
Experiments that used a kanamycin-marked Stx2 prophage demonstrated that ciprofloxacin,
but not fosfomycin, caused enhanced intraintestinal transfer of Stx2 prophage from one
E. coli to another. These observations suggest that treatment of human STEC infection with
bacteriophage-inducing antibiotics, such as fluoroquinolones, may have significant adverse
clinical consequences and that fluoroquinolone antibiotics may enhance the movement of
virulence factors in vivo.

Shiga toxin–producing Escherichia coli (STEC), such as ferences in response to STEC. It is clear that the production
E. coli O157:H7, are emerging foodborne pathogens in the of Shiga toxins by STEC is one of the most critical factors in
United States and other parts of the world [1, 2]. Ingestion of the development of HUS. It is also probable, but not proven
STEC can result in gastrointestinal symptoms, such as diarrhea in humans, that the more toxin that is made, the more likely
and hemorrhagic colitis, and may also progress to hemolytic- a patient will develop significant medical problems. However,
uremic syndrome (HUS), a severe sequela of this infection [3]. the factors that control toxin production in the intestine and
STEC have become the most frequent cause of acute renal the processes that are critical in the movement of toxin from
failure in children in the United States in recent years and are the intestinal lumen to their final target in the kidney, brain,
currently one of the most devastating foodborne pathogens in and other organs are as yet undetermined.
many parts of the world. Little is known about the molecular The Shiga toxin family is made up of 2 main groups, Shiga
mechanisms leading to the development of the most serious toxin 1 (Stx1) and Shiga toxin 2 (Stx2), produced by E. coli
complications, such as HUS, but it is likely that different in- O157:H7 and many other STEC serotypes. Both are usually
dividuals respond to exposure to STEC in different ways. It is encoded in the genomes of lysogenic lambdoid phages, thereby
not clear why some people develop a mild gastrointestinal ill- allowing a mechanism for toxin gene dissemination via transfer
ness, whereas others develop hemorrhagic colitis and still others
of bacteriophages [4, 5]. Recent studies suggest that the life
develop full-blown HUS. Many factors, including both host
cycles of these Stx phages regulate toxin production and thereby
and bacterial determinants, may be responsible for these dif-
may influence pathogenesis [6]. In particular, Stx prophage in-
duction has been associated with increased Stx production in
Received 27 May 1999; revised 20 September 1999; electronically pub- vitro [7]. Stx prophages, like l prophages, are inducible with
lished 8 February 2000. agents that provoke the bacterial “SOS” response to DNA
Presented in part: IX International Congress of Bacteriology and Applied
Microbiology, Sydney, Australia, August 1999. damage and/or to the inhibition of DNA replication. Thus, the
Financial support: NIH (AI-39067, AI-07329, and DK-34928). M.K.W. induction of Stx prophages by the alkylating agent mitomycin
is a Pew Scholar in the Biomedical Sciences and a Tupper Research Fellow.
a
Present affiliations: Department of Pharmaceutics, College of Pharmacy,
C is associated with significant increases in Stx production in
Rutgers, State University of New Jersey, Piscataway (X.Z.); University of vitro [7]. In recA2 STEC strains, this SOS-triggering agent does
Southern California School of Medicine, Los Angeles (A.D.M.); Depart- not induce Stx prophages or augment Stx production [7]. Be-
ment of Medicine, Northeast Health Systems, Beverly, Massachusetts
(L.E.W.). cause RecA is known to be required for the initiation of the
Reprints or correspondence: Dr. David W. K. Acheson, Division of Geo- SOS response, but is not thought to directly regulate Stx pro-
graphic Medicine and Infectious Diseases, New England Medical Center,
duction, these findings indicate that phage induction is a re-
750 Washington St., Boston, MA 02111 (Dacheson@Lifespan.org).
quired step in the augmentation of toxin production that occurs
The Journal of Infectious Diseases 2000; 181:664–70
q 2000 by the Infectious Diseases Society of America. All rights reserved.
following exposure of STEC to SOS-inducing agents.
0022-1899/2000/18102-0034$02.00 The clinical relevance of prophage-mediated Stx production
JID 2000;181 (February) Quinolone Antibiotics Induce Shiga Toxins In Vivo 665

is suggested by the observation that patients treated with mi- thi) [21]; both were from our laboratory collection of bacterial
tomycin C have been reported to develop HUS [8]. Several strains. MC4100 lysogens of Stx2 phage 933W [4] and 933W-Kn
clinically useful antibiotics known to induce the SOS response, (see below), MC4100 (933W) and MC4100 (933W-Kn), respec-
including trimethoprim-sulfamethoxazole (commonly used to tively, were isolated as described elsewhere [7]. C600 (933W) is an
E. coli C600 lysogen of Stx2 phage 933W [4].
treat diarrheal disease in children) and ciprofloxacin (commonly

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


Construction of the 933W-Kn phage. 933W-Kn phage was con-
used to treat diarrheal disease in adults), have also been shown
structed from the Stx2 phage 933W [4] by replacing most of the
to enhance Stx production in vitro [9–11]. There have been no Stx2 toxin operon promoter and the toxin A subunit gene (stx2A)
large randomized controlled prospective studies to evaluate the with the KnR gene from Tn903. A 524-bp Stx2 fragment was cloned
utility of antimicrobial treatment of STEC infection; however, by use of the forward primer Stx2-1 (ACAG CTGCAGCG-
in 1 small retrospective study, treatment with sulfonamides, TTTCTGAACAG; PstI site underscored) and reverse primer Stx2-
which is known to induce bacteriophages, was associated with 2 (GCAG GGTACCACCGAACCCGTGT; KpnI site under-
an increased likelihood of developing HUS [12]. In contrast, a scored). This fragment begins 50 bp upstream of the stop codon
retrospective analysis of the large 1996 O157:H7 outbreak in of stx2A and ends 185 bp downstream of the stx2B gene stop
Japan suggested that the use of the antibiotic fosfomycin, an codon. This 524-bp fragment was cloned into a PstI/KpnI–cut de-
inhibitor of bacterial cell wall synthesis that is not known to rivative of pBluescript SK2 that had previously had its 2 BspHI
sites converted to AvrII sites, yielding pSK/933W-1. Forward
induce bacteriophages, was correlated with reduced likelihood
primer Stx2-10 (GGCA GGATCCGAATGTAGTCAGTCAGA-
of developing HUS [13].
ACGGATG; BamHI site underscored) and reverse primer Stx2-7
A variety of animal models have been developed for the study (GCTG CTGCAG-TCATGACCAGACCCGGCGCAGA; PstI
of STEC [14], among which mouse models with orally delivered and BspHI sites underscored) were used to clone a 364-bp region
STEC are widely used and relatively well characterized [15–18]. from 933W phage. This 364-bp region spans from 65 bp upstream
In the mouse models, animals given STEC orally frequently of the ileX start codon [22] to a point 50 bp upstream of the stxA
develop neurologic and renal symptoms, usually culminating start codon, including the Stx2 operon promoter [23]. The 364-bp
in death. Coadministration of anti-Stx antibody prevents death fragment was cloned into the BamHI/PstI–cut pSK/933W-1, yield-
of the mice, thereby suggesting that Stx is the principal cause ing pSK/933W-2. pSK/933W-2 was cut with BspHI and treated
of murine morbidity and mortality [18]. In mice, the principal with Klenow fragment, yielding a blunt-ended pSK/933W-2 back-
targets of Stx appear to be the central nervous system and bone. A third pair of primers, forward primer DA101 (GCGC
endothelial cells [16]. We recently demonstrated that E. coli Stx AGTACTCTGTGTGGCCACGTTGTG; ScaI site underscored)
and reverse primer DA102 (GCGC AGTACTGTTCTAGAGG-
phage lysogens produce infectious virions within the murine
CCTGTGTG; ScaI site underscored), were used in a polymerase
gastrointestinal tract [19]. We hypothesized that antibiotic-
chain reaction to clone the KnR gene (originally derived from
induced induction of Stx phage from intestinal O157:H7 in mice
Tn903) with its associated promoter from plasmid p216 (obtained
may augment Stx production and lead to adverse consequences. from A. Camilli, Tufts University, Medford, MA). The KnR frag-
In this study, we used an STEC mouse model to investigate the ment (1044 bp) was ligated into the blunted BspHI sites of the
effects of 2 antibiotics on fecal STEC colony-forming units, pSK/933W-2 to yield pSK/933W-2-Kn. pSK/933W-2-Kn contained
fecal toxin levels, intraintestinal phage production, and mouse the KnR gene flanked by the Stx2 operon promoter region (50) and
mortality. We sought to elucidate the potential risks associated the stx2B region (30). This entire Stx2 promoter–KnR gene–stx2B
with the use of antibiotics that result in DNA damage and gene region insert was ligated into the partially digested suicide
phage induction in treating patients infected with STEC. vector pCVD442 [24] to yield pCVD442/933W-2-Kn. pCVD442/
933W-2-Kn was electroporated into C600 (933W). One of the C600
KnR isolates chosen randomly was induced with mitomycin C (500
ng/mL) in Luria-Bertani (LB) broth. Phage-containing supernatant
Materials and Methods
was then used to lysogenize C600. In C600 (933W-Kn), the re-
Chemicals and reagents. Ciprofloxacin (2 mg/mL ciprofloxacin placement of stx2A by the KnR gene in the 933W-Kn prophage
in 5% dextrose) was purchased from Bayer (West Haven, CT). was confirmed by polymerase chain reaction with the primers used
Kanamycin monosulfate, fosfomycin disodium salt, streptomycin in the original cloning described above. The replacement of stx2A
sulfate, and tetracycline hydrochloride were purchased from Sigma by the KnR gene did not significantly alter the genome size of the
(St. Louis), PBS from Gibco BRL (Grand Island, NY), Vent DNA resulting phage (net 98-bp gain). As expected, E. coli cells lyso-
polymerase and restriction enzymes from New England Biolabs genized by this recombinant phage produced no Stx2 A subunit
(Beverly, MA), and pBluescript SK2 plasmid DNA from Stratagene by Western blot, and sonicated extracts of overnight cultures were
(La Jolla, CA). not cytotoxic to Vero cells [25] (data not shown). The ability of
Bacterial strains. E. coli O157:H7 strain 1:361 encodes Stx2 933W-Kn phage to form plaques, to be induced from an E. coli
only and was isolated during a nationwide surveillance study [20]. lysogen (e.g., C600 or MC4100), and to lysogenize a new E. coli
1:361R, an StrR derivative of 1:361, was isolated by selecting for host (e.g., C600 or MC4100) remained intact and were no different
a spontaneous streptomycin-resistant mutant. 1:361R was used for from the original 933W phage (data not shown). C600 cells lyso-
all in vitro and in vivo studies described below. MC4100, argG: genized with 933W and 933W-Kn phages had identical growth
Tn10, is a derivative of MC4100 (araD139, delta[lac]U169, strA, curves in LB medium (data not shown).
666 Zhang et al. JID 2000;181 (February)

Quantitation of colony-forming units and plaque-forming units. follows. Previous in vitro experiments suggested that subinhibitory
Samples from both in vitro and in vivo experiments were serially concentrations of ciprofloxacin caused maximal increases in Stx
diluted in PBS and plated onto LB agar plates containing strep- (data not shown). In initial experiments, we determined a dose of
tomycin at 500 mg/mL. Colony-forming units were counted after ciprofloxacin that caused a significant drop in fecal 1:361R colony-
overnight culture at 377C. Samples collected in the mouse lysogenic forming units (∼3 logs) but did not kill all of the organisms. The

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


conversion experiments were processed similarly and plated on ap- dose of fosfomycin was adjusted to cause an equivalent drop in
propriate selective plates. Plaque-forming units were measured as fecal colony-forming units (∼3 logs). In all experiments, mice were
described elsewhere [19]. observed twice daily from day 0 to the end of each experiment on
Stx2 concentration. Stx2 concentrations in bacterial cultures day 18. On days 0–8 and on day 11, a fresh mouse stool sample
were determined by EIA [26] by use of purified Stx2 as the standard. (2–3 pellets) was collected from each mouse and weighed. Each
Stx2 levels from murine fecal extracts were measured by means of stool sample was suspended in PBS at a ratio of 1 mL to 0.1 g of
Premier EHEC (Meridian Diagnostics, Cincinnati). In the in vitro stool. After stool debris were allowed to settle, the supernatant was
experiments, 1.0-mL samples of various LB cultures of 1:361R were sampled for determination of colony-forming units. The remainder
spun for 5 min in a microcentrifuge. After centrifugation, each was spun in a microcentrifuge for 5 min, and the second super-
supernatant was used for extracellular Stx2 determination, and natant was used for Stx2 determination as described above.
each pellet was resuspended in 1 mL of LB followed by sonication Effect of antibiotics on intraintestinal phage production. These
(3 cycles of 30 pulses at 67% dutycycle and 4.5 output) on a Branson experiments were designed to measure intestinal 933W-Kn phage
Sonifier 450 (Branson Sonic Power, Danbury, CT). The sonicated production after ciprofloxacin and fosfomycin administration. A
sample was then spun for 5 min, and the supernatants were used mixture of overnight cultures of the donor strain, MC4100 (933W-
for cell-associated Stx2 determination. In the in vivo experiments, Kn) (∼ 2 3 10 9/mouse), and the recipient strain, MC4100, argG:
mouse fecal samples were suspended in PBS (see below) and spun Tn10 (∼ 7 3 10 9/mouse), was administered together via a feeding
in a microcentrifuge for 5 min, and the supernatants were then needle to CD-1 mice as described above. Unlike the 1:361R strain,
used for determination of the Stx2 concentration. Plating of these these E. coli K12 strains colonized the mouse gut poorly. Stool
fecal supernatants on LB-streptomycin plates showed no or neg- colony-forming unit counts decreased rapidly, compared with those
ligible numbers of streptomycin-resistant cells, indicating that the of the 1:361R strain. Consequently, these experiments were com-
Stx2 levels measured represented primarily extracellular Stx2 in the pressed to 8 days. Ciprofloxacin and fosfomycin were given on
feces. days 2–4 at the same dosages as described above. From day 0 to
Effect of ciprofloxacin or fosfomycin on toxin expression in the end of the experiments on day 8, colony-forming units of the
vitro. Overnight cultures in LB medium were diluted in the same donor, the recipient, and the in vivo–derived transductants were
medium to an optical density at 600 nm of ∼0.08 and then were determined by plating the stool samples onto 3 sets of LB agar
split into 3 25-mL cultures in flasks. No antibiotic was added to plates containing, respectively, streptomycin (500 mg/mL)
the control culture, whereas ciprofloxacin and fosfomycin were –kanamycin (50 mg/mL), streptomycin (500 mg/mL)–tetracycline
added to the second and third flasks, respectively. The colony- (7.5 mg/mL), and streptomycin (500 mg/mL)–kanamycin (50 mg/
forming units, plaque-forming units, and Stx2 concentrations at mL)–tetracycline (7.5 mg/mL).
time 0 were determined in the control flask. The 3 flasks were
incubated at 377C with shaking. At 3 and 6 h, samples were taken
from each of the 3 flasks, and optical density at 600 nm, colony- Results
forming units, plaque-forming units, and Stx2 concentration were
Effect of ciprofloxacin or fosfomycin on Stx production in
determined.
Mouse infection with E. coli 1:361R. The procedure used was vitro. Our first goal was to demonstrate the effect of cipro-
a modification of that described in our previous studies [19]. Sets floxacin, an inhibitor of DNA gyrase, and fosfomycin, an in-
of 14 male CD-1 mice aged 5 weeks (Charles River Laboratories, hibitor of peptidoglycan synthesis, on Stx2 production and bac-
Wilmington, MA) were given drinking water containing strepto- teriophage induction in vitro from the clinical E. coli O157:H7
mycin (2 g/L) throughout each experiment. One day after strep- isolate, 1:361R. Three hours after the addition of ciprofloxacin
tomycin was begun, the mice were starved for food for 24 h; then (30 ng/mL) to the 1:361R culture, we noted a 7-fold increase
each mouse was inoculated intragastrically with ∼ 3.0 3 10 9 1:361R in the number of plaque-forming units per milliliter of culture
cells. Each inoculum comprised ∼0.15 mL of overnight culture that supernatant and a 17-fold increase in Stx concentration com-
had been washed once in PBS and resuspended in the same volume pared with that of the control culture (table 1). At the 6-h time
of 20% sucrose. The day of inoculation was designated day 0. In point, the ciprofloxacin-mediated induction of the Stx prophage
initial experiments, we found that on days 1 and 2 after 1:361R
and Stx production were even more marked, with a 11000-fold
inoculation there was a dramatic increase in the number of fecal
increase in the number of plaque-forming units per milliliter
1:361R cells and fecal Stx2. One or 2 mice of each group of 14
and an ∼60-fold increase in the total Stx2 concentration in the
died by day 4, after which no further deaths were observed. There-
fore, in subsequent antibiotic treatment experiments, 4 days after culture (table 1). The relative increase in both plaque-forming
inoculation with 1:361R, the surviving mice were divided into equal units and toxin compared with control or fosfomycin treatment
groups. Mice received an intraperitoneal dose of either 30 mg of were reproducible in subsequent experiments and at higher
ciprofloxacin or 15 mg of fosfomycin on days 4, 5, and 6. Control doses of ciprofloxacin (40 ng/mL; data not shown).
mice received only PBS. The rationale behind drug dosage is as This dramatic increase in Stx production occurred despite a
JID 2000;181 (February) Quinolone Antibiotics Induce Shiga Toxins In Vivo 667

Table 1. Ciprofloxacin induces Shiga toxin (Stx) prophage and augments Stx production
from Escherichia coli O157:H7 strain 1:361R.
Phage titer Stx concentration Cell no.
(pfu/mL 3103) (ng/mL) (cfu/mL 3106)
Time (h) Control Cpfx Fm Control Cpfx Fm Control Cpfx Fm

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


0 0.02 0.02 0.02 30 30 30 114.0 114.0 114.0
3 7.3 50.0 10.4 301 5325 231 1930.0 4.5 34.0
6 12.5 13,000 44.0 1406 81,800 522 4600.0 0.3 4200.0
NOTE. Overnight culture of 1:361R was diluted to optical density at 600 nm of 0.08 and split into
3 equal-volume cultures (time 0). At time 0, either ciprofloxacin (Cpfx, 25 ng/mL), fosfomycin (Fm, 800
ng/mL), or nothing (control) was added. Serial dilutions of chloroform-treated supernatants were plated
on E. coli strain C600. Polymerase chain reaction analysis was used to confirm that all 19 randomly picked
plaques were positive for stxA2 sequences. Stx2 concentration was determined in both culture supernatants
and sonicated pellets by EIA [22] with purified Stx2 as standard [23]. Total Stx2 is shown.

110,000-fold reduction in recoverable 1:361R colony-forming of 1:361R-colonized mice was reproduced in 2 subsequent ex-
units in the culture containing ciprofloxacin, probably a result periments. In the first of the repeat experiments, we observed
of cell killing by both the antibiotic and phage-mediated cell death in 4 of 7 ciprofloxacin-treated mice and in 0 of 6 PBS-
lysis. Thus, ciprofloxacin resulted in marked Stx prophage in- treated mice, and in the second experiment, 7 of 8 ciprofloxacin-
duction and increases in Stx production from Stx phage lyso- treated mice died. The mortality observed in the ciprofloxacin-
gens, similar to our previously reported results with mitomycin treated mice is not attributable to the antibiotic treatment alone,
C [7]. Stx prophage induction and increase in Stx production because there were no deaths in a separate control group of 6
are not nonspecific responses to antimicrobial agents. Despite mice that were treated with streptomycin and then with cip-
the fact that the fosfomycin-treated cultures had recovered by rofloxacin as above but that were not colonized with 1:361R
6 h, the drug resulted in a decline in 1:361R colony-forming (data not shown). There was no significant difference in the
units at 3 h but, unlike ciprofloxacin, had no effect on the amounts of fecal Stx2 detected in the fosfomycin-treated mice
induction of 1:361R Stx prophages or toxin production, com- compared with control (PBS-treated) mice (figure 2C vs. fig-
pared with controls (table 1). ure 2A).
Effect of ciprofloxacin or fosfomycin treatment in mice colo- Effect of ciprofloxacin on intraintestinal phage production in
nized with E. coli O157:H7. To address the potential clinical mice. To explore whether treatment of mice with ciproflox-
ramifications of antibiotic induction of Stx prophages and toxin acin leads to increased intraintestinal Stx phage production
production, we then compared the effects of ciprofloxacin and from a lysogen as seen in vitro, we used an intraintestinal Stx
fosfomycin on intestinal toxin production and mortality in mice phage lysogenic conversion assay that we recently described
colonized with 1:361R. To facilitate intestinal colonization with elsewhere [19], which involves intraintestinal transduction of a
this O157:H7 strain, CD-1 mice were given streptomycin [15] marked E. coli recipient strain (a TcR derivative of E. coli K12
for 2 days and then orally inoculated with 1:361R. In each set strain MC4100 [19]) to KnR by a Kn-marked Stx phage. This
of experiments, the mice were divided into equal groups on day phage, 933W-Kn, is harbored in an E. coli K12 strain, MC4100
4. One group received an antibiotic (either ciprofloxacin or (933W-Kn), the donor strain. After intragastric inoculation of
fosfomycin) and the other group received PBS. In control mice, the donor and recipient strains to 5 mice, the number of donor
the pattern of 1:361R intestinal colonization and toxin pro- cells (SmR KnR), recipient cells (SmR TcR), and “transductant”
duction in individual mice was consistent (figures 1A and 2A). cells (those recipient cells lysogenically converted to kanamycin
Fecal Stx was detectable 1 day after inoculation and declined resistance [SmR TcR KnR]) in fecal samples were determined
thereafter as the 1:361R excretion dropped (figures 1A and 2A). daily. Two days after the inoculation of the donor and recipient
After treatment with either antibiotic, there was a more rapid strains, mice were injected intraperitoneally with either cipro-
fall in fecal STEC colony-forming units than was observed in floxacin, fosfomycin, or PBS at the same doses used in our
the PBS control group (figure 1), with values differing by 3 previous experiments. Because there is no biologically active
orders of magnitude. Despite causing similar decreases in col- Stx-producing strain present in these experiments (replacement
ony-forming units, the antibiotics differed in their effect on fecal of stxA by the KnR gene inactivates the toxin), no deaths were
Stx. Ciprofloxacin treatment resulted in an increase in fecal Stx observed in these experiments. As in the experiments with 1:
concentration in all 6 mice, whereas fosfomycin did not (figure 361R in mice, both ciprofloxacin and fosfomycin treatments
2). This increase in intraintestinal toxin production was accom- resulted in 100- to 1000-fold reductions in the number of donor
panied by a dramatic clinical effect. Four of the 6 ciprofloxacin- and recipient cells detected in fecal samples, compared with the
treated mice (those with the highest stool Stx concentrations) PBS-treated control group (both of these strains are sensitive
died, whereas none of the fosfomycin-treated mice nor control to the 2 antibiotics; table 2). However, significant numbers of
mice died. The mortality resulting from ciprofloxacin treatment transductants were detected in all 5 of the mice treated with
668 Zhang et al. JID 2000;181 (February)

after induction of the Stx-encoding bacteriophages [7]. A num-


ber of reports have suggested that antibiotics are associated
with the development of HUS in patients infected with STEC
[12, 28]. Other in vitro studies have demonstrated that some
antibiotics, especially fluoroquinolones, will increase Stx pro-

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


duction [9, 10, 11, 29]. The purpose of this work was to take
these in vitro observations to a more critical level and to de-
termine whether fluoroquinolones induced toxin expression in
vivo and, if so, whether any potential clinical complications
might occur after therapy with fluoroquinolones in animals
infected with STEC.

Figure 1. Intestinal colonization with Escherichia coli O157:H7


strain 1:361R. After inoculation of 1:361R on day 0, colony-forming
units (cfu) per gram of stool were determined daily. Each line corre-
sponds to individual mouse. Treatments with either PBS (A), cipro-
floxacin (B), or fosfomycin (C) were given intraperitoneally on days
4–6 (shaded area). Because results for 2 control groups were virtually
identical, data from only 1 are shown (A).

ciprofloxacin and in none of the mice treated with fosfomycin


(table 2). One of the control mice also had a low level of trans-
ductants, suggesting that spontaneous Stx2 phage induction can
occur intraintestinally, similar to our previous results with Stx1
phage lysogens [19]. Spot checks of putative transductant cells
grown from fecal samples (SmR TcR KnR) with a polymerase
chain reaction assay for 933W-Kn phage DNA sequences con-
firmed that all of the colonies tested contained this phage.

Figure 2. Fecal Shiga toxin (Stx) concentrations after intragastric


Discussion inoculation of Escherichia coli O157:H7 strain 1:361R. Levels of Stx2
in stool samples (expressed as nanograms of Stx2 per gram of stool)
Little is known about the regulation of Stx expression in were determined daily after inoculation of 1:361R on day 0. Each line
vitro; nothing is known about its regulation in vivo. Stx1, like corresponds to individual mouse. Treatments with either intraperito-
neal PBS (A), ciprofloxacin (B), or fosfomycin (C) were given intra-
Shiga toxin from Shigella dysenteriae, is iron-regulated in vitro peritoneally on days 4–6 (shaded area). †, mice that died within 24 h
[27], but it is not clear whether this regulation is critical in vivo. of stool Stx determinations. Because results for 2 control groups were
Production of both Stx1 and Stx2 has been shown to increase virtually identical, data from only 1 are shown (A).
JID 2000;181 (February) Quinolone Antibiotics Induce Shiga Toxins In Vivo 669

Table 2. Ciprofloxacin enhances intraintestinal transduction of Kn-marked Shiga toxin (Stx) phage.
Ciprofloxacin treatment Fosfomycin treatment PBS treatment
Mouse Donor Recipient Transductant Donor Recipient Transductant Donor Recipient Transductant
1 4.0 3 105
8.0 3 105
1.1 3 104
9.2 3 105
2.8 3 104
UD 2.6 3 107
1.3 3 107
UD

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


2 1.6 3 105 5 3 104 1.7 3 104 1.0 3 103 1.0 3 104 UD 3.0 3 107 1.0 3 108 1.8 3 103
3 2.0 3 104 3 3 104 7.0 3 103 1.7 3 104 1.5 3 104 UD 2.0 3 107 2.0 3 107 UD
4 1.3 3 105 1.2 3 104 1.2 3 104 6.0 3 105 1.8 3 105 UD 1.2 3 108 6.0 3 106 UD
5 1.5 3 103 6.0 3 103 2.0 3 102 1.0 3 105 1.5 3 105 UD 4.9 3 107 1.2 3 108 UD
NOTE. On day 0, each mouse was orally inoculated with 2 3 109 cells of potential phage donor strain (MC4100 933W-Kn, SmR KnR)
and 5 3 109 cells of potential recipient strain (MC4100 argG:Tn10, SmR TcR); on days 2–4, mice were inoculated intraperitoneally with either
ciprofloxacin (30 mg/mouse), fosfomycin (15 mg/mouse), or PBS. Fecal samples were collected daily, weighed, resuspended in PBS, and
plated on media to allow determination of nos. of donor cells (SmR KnR), recipient cells (SmR TcR), and transductants (SmR KnR TcR). Data
are given as colony-forming units per gram of stool. Values shown are from day 3 after inoculation, time point representative of data from
entire week of study. Threshold level of detection is 102 cfu/g of stool; UD, undetectable at any time during experiment.

Ciprofloxacin and other fluoroquinolones are the antibiotics be deleterious in patients, even if the organisms are subse-
most frequently prescribed for the treatment of diarrheal dis- quently killed by the antibiotic. These pharmacologic consid-
eases in adults. Such therapy is often empirical and prescribed erations suggest that it may be very difficult to determine a safe
before results of stool cultures are available. We found that dose of fluoroquinolones for the treatment of STEC infection.
ciprofloxacin treatment of mice colonized with E. coli O157: The cause of a diarrheal illness is usually not known when
H7 induced Stx prophages. The consequences of intestinal Stx patients first seek medical attention. Because STEC are asso-
prophage induction were enhanced intraintestinal Stx produc- ciated with ∼1% of stools submitted for analysis to clinical
tion, increased Stx phage transmission, and, most dramatically, microbiology laboratories in the United States [20], our data
death of the ciprofloxacin-treated mice. Because fosfomycin and suggest that empirical quinolone treatment of diarrheal disease
ciprofloxacin treatments both reduced intestinal 1:361R excre- or, indeed, therapy with any other antibiotic known to cause
tion similarly and because fosfomycin causes bacterial lysis (by bacteriophage induction, should be reconsidered. If antimicro-
inhibiting cell wall synthesis), the absence of augmented intes- bial treatment of STEC infection is indicated, classes of anti-
tinal toxin levels and death in the mice treated with fosfomycin biotics that do not induce Stx prophages (e.g., cell wall synthesis
indicates that the effects observed with ciprofloxacin cannot be inhibitors such as fosfomycin or protein synthesis inhibitors
attributed just to antibiotic-related bacterial cell lysis and re- such as macrolides) should be used. Similar caution should be
lease of preformed toxin. Thus, expanding the scope of our in
exercised in the choice of antimicrobial agents in the treatment
vitro observations, ciprofloxacin, but not fosfomycin, enhanced
of other infections, in which prophage-encoded toxins are crit-
intraintestinal Stx2 production in mice colonized with E. coli
ical virulence factors [30, 31]. Clearly, many patients infected
O157:H7. In this murine model, these ciprofloxacin-mediated
with STEC go on to develop HUS without having received
increases in intraintestinal toxin levels were sufficient to kill the
antibiotic therapy, so there must be other important factors
mice.
relating to the control of toxin expression or its absorption
These findings suggest that quinolones, as well as other an-
from the intestine. However, our observations do suggest that
tibiotics that can induce the bacterial SOS response with re-
caution should be exercised in relation to empirical treatment
sulting Stx prophage induction, should not be used as therapy
of diarrheal disease in both children and adults.
for STEC infection. Although the dose of ciprofloxacin that
Our data raise not only questions about the safety of the use
we administered to mice colonized with E. coli O157:H7 was
only 20% of the dose (on a milligram per kilogram of body of fluoroquinolones in the treatment of diarrheal disease for
weight basis) commonly given to patients with diarrhea, the individual patients but also broader issues regarding the role
concentrations of ciprofloxacin in intestinal microenvironments of antimicrobials in the dissemination of bacteriophage-en-
probably vary with the nature of intestinal contents, intestinal coded virulence factors in the environment. We found that Stx-
mucus, bacterial flora, epithelial cell secretions, and gut motility. encoding phages are able to move from 1 strain of E. coli to
When antibiotic therapy is initiated, concentrations required another in the intestines of mice and that antibiotic-mediated
for antibiotic-mediated Stx prophage and toxin induction may prophage induction enhances this process. It is possible that
be achieved in critical intestinal microenvironments well before the use of phage-inducing antimicrobials in farm animals, es-
bactericidal concentrations can accumulate. Our in vitro ex- pecially at subtherapeutic levels, may inadvertently enhance the
periments demonstrated that even a 3-h exposure to ciproflox- dissemination of bacteriophage-encoded toxins. This could con-
acin was enough to induce phage and elevate toxin expression. tribute to the emergence of new pathogens and the spread of
Thus, we speculate that the use of higher doses of ciprofloxacin Stx genes to other members of the Enterobacteriaceae family,
will, in the initial phase, cause an elevation of toxin that may such as Citrobacter and Enterobacter species, both of which
670 Zhang et al. JID 2000;181 (February)

have been associated with Shiga toxin–related HUS in humans and disease caused by enterohemorrhagic Escherichia coli O157:H7. Infect
Immun 1990; 58:2438–45.
[32, 33].
16. Karpman D, Connell H, Svensson M. The role of lipopolysaccharide and
Shiga-like toxin in a mouse model of Escherichia coli O157:H7 infection.
Acknowledgments
J Infect Dis 1997; 175:611–20.

Downloaded from https://academic.oup.com/jid/article-abstract/181/2/664/824154 by Macau Polytechnic Institute user on 18 September 2019


17. Kurioka T, Yunou Y, Kita E. Enhancement of susceptibility to Shiga
We thank our colleagues A. Plaut, B. Davis, A. Camilli, and A. Kane toxin–producing Escherichia coli O157:H7 by protein calorie malnutrition
for their helpful comments. in mice. Infect Immun 1998; 66:1726–34.
18. Wadolkowski EA, Sung LM, Burris JA, Samuel JE, O’Brien AD. Acute renal
tubular necrosis and death of mice orally infected with Escherichia coli
References strains that produce Shiga-like toxin type II. Infect Immun 1990; 58:
3959–65.
1. Acheson DWK, Keusch GT. Which Shiga toxin–producing E. coli are im- 19. Acheson DWK, Reidl J, Zhang X, Keusch GT, Mekalanos JJ, Waldor MK.
portant? ASM News 1996; 62:302–6. In vivo transduction with Shiga toxin 1–encoding phage. Infect Immun
2. Kaper JB, O’Brien AD. Escherichia coli O157:H7 and other Shiga 1998; 66:4496–8.
toxin–producing E. coli strains. Washington, DC: American Society for 20. Acheson DWK, Frankson K, Willis D, and the STEC Prevalence Study
Microbiology Press, 1998. Group. Multicenter prevalence study of Shiga toxin–producing Escheri-
3. Griffin PM. Escherichia coli O157:H7 and other enterohemorrhagic Escher- chia coli [abstract C-205]. In: Proceedings of the 98th general meeting of
ichia coli. In: Blaser MJ, Smith PD, Ravdin JI, Greenberg HB, Guerrant the American Society for Microbiology. Washington, DC: American So-
RL, eds. Infections of the gastrointestinal tract. New York: Raven Press, ciety for Microbiology, 1998.
1995:739–61. 21. Tesh VL, O’Brien AD. The pathogenic mechanisms of Shiga toxin and the
4. O’Brien AD, Newland JW, Miller SF, Holmes RK, Williams-Smith H, Formal Shiga-like toxins. Mol Microbiol 1991; 5:1817–22.
SB. Shiga-like toxin converting phages from Escherichia coli strains that 22. Schmidt H, Scheef J, Janetzki-Mittmann C, Datz M, Karch H. An ileX tRNA
cause hemorrhagic colitis or infantile diarrhea. Science 1984; 226:694–6. gene is located close to the Shiga toxin II operon in enterohemorrhagic
5. Huang A, Frieman J, Brunton JL. Characterization of a bacteriophage that Escherichia coli O157 and non-O157 strains. FEMS Microbiol Lett
carries the genes for production of Shiga-like toxin 1 in Escherichia coli. 1997; 149:39–44.
J Bacteriol 1987; 169:4308–12. 23. Sung LM, Jackson MP, O’Brien AD, Holmes RK. Transcription of the Shiga-
6. Neely MN, Friedman DI. Functional and genetic analysis of regulatory
like toxin type II and Shiga-like toxin type II variant operons of Escher-
regions of coliphage H-19B: location of Shiga-like toxin and lysis genes
ichia coli. J Bacteriol 1990; 172:6386–95.
suggest a role for phage functions in toxin release. Mol Microbiol 1998;
24. Donnenberg MS, Kaper JB. Construction of an eae deletion mutant of en-
28:1255–67.
teropathogenic Escherichia coli by using a positive-selection suicide vector.
7. Muhldorfer I, Hacker J, Keusch GT, et al. Regulation of the Shiga-like toxin
Infect Immun 1991; 59:4310–7.
II operon in Escherichia coli. Infect Immun 1996; 64:495–502.
25. Jacewicz M, Feldman HA, Donohue-Rolfe A, Balasubramanian KA, Keusch
8. Acheson DWK, Donohue-Rolfe A. Cancer-associated hemolytic uremic syn-
GT. Pathogenesis of Shigella diarrhea. XIV. Analysis of Shiga toxin re-
drome: a possible role of mitomycin C in relation to Shiga-like toxins. J
ceptors on cloned Hela cells. J Infect Dis 1989; 159:881–9.
Clin Oncol 1989; 7:1943.
26. Donohue-Rolfe A, Acheson DWK, Kane AV, Keusch GT. Purification of
9. Karch H, Goroncy-Bermes H, Opferkuch W, Droll HP, O’Brien AD. Sub-
Shiga toxin and Shiga-like toxins I and II by receptor analog affinity
inhibitory concentrations of antibiotics modulate amount of Shiga-like
chromatography with immobilized P1 glycoprotein and production of
toxin produced by Escherichia coli. In: Adam D, Hahr H, Opterkuch W,
cross-reactive monoclonal antibodies. Infect Immun 1989; 57:3888–93.
eds. The influence of antibiotics on the host-parasite relationship. Berlin:
27. Calderwood SB, Mekalanos JJ. Iron regulation of Shiga-like toxin expression
Springer Verlag, 1985:239–45.
in Escherichia coli is mediated by the fur locus. J Bacteriol 1987; 169:
10. Walterspiel JN, Ashkenazi S, Morrow AL, Cleary TG. Effect of subinhibitory
4759–64.
concentrations of antibiotics on extracellular Shiga-like toxin 1. Infection
28. Carter AO, Borczyk AA, Carlson JAK, et al. A severe outbreak of Escherichia
1992; 20:25–9.
coli O157:H7–associated hemorrhagic colitis in a nursing home. N Engl
11. Matsushiro A, Sato K, Miyamoto H, Yamamura T, Honda T. Induction of
J Med 1987; 317:1496–500.
prophages of enterohemorrhagic Escherichia coli O157:H7 with norflox-
29. Karch H, Strockbine NA, O’Brien AD. Growth of Escherichia coli in the
acin. J Bacteriol 1999; 181:2257–60.
presence of trimethoprim-sulfamethoxazole facilitates detection of Shiga-
12. Pavia AT, Nichols CR, Green DP, et al. Hemolytic-uremic syndrome during
like toxin producing strains by colony blot assay. FEMS Microbiol Lett
an outbreak of Escherichia coli O157:H7 infections in institutions for
mentally retarded persons: clinical and epidemiological observations. J 1986; 35:141–5.
Pediatr 1990; 116:544–51. 30. Waldor M. Bacteriophage biology and bacterial virulence. Trends Microbiol
13. Takeda T, Yoshino YY, Uchida H, Ikeda N, Tanimura M. Early use of 1998; 6:295–7.
fosfomycin for Shiga toxin–producing Escherichia coli O157:H7 infection 31. Kehl KS, Havens P, Behnke CE, Acheson DWK. Evaluation of the Premier
reduces the risk of hemolytic-uremic syndrome. In: Kaper JB, O’Brien EHEC assay for detection of Shiga toxin–producing Escherichia coli. J
AD, eds. Escherichia coli O157:H7 and other Shiga toxin–producing E. Clin Microbiol 1997; 35:2051–4.
coli strains. Washington, DC: American Society for Microbiology Press, 32. Tschape H, Prager R, Streckel W, Fruth A, Tietze E, Bohme G. Verotoxi-
1998:385–7. nogenic Citrobacter freundii associated with severe gastroenteritis and
14. Moxley RA, Francis DH. Overview of animal models. In: Kaper JB, O’Brien cases of haemolytic uraemic syndrome in a nursery school: green butter
AD, eds. Escherichia coli O157:H7 and other Shiga toxin–producing E. as the infection source. Epidemiol Infect 1995; 114:441–50.
coli strains. Washington, DC: American Society for Microbiology Press, 33. Paton AW, Paton JC. Enterobacter cloacae producing a Shiga-like toxin
1998:249–60. II–related cytotoxin associated with a case of hemolytic-uremic syndrome.
15. Wadolkowski EA, Burris JA, O’Brien AD. Mouse model for colonization J Clin Microbiol 1996; 34:463–5.

You might also like